Open Access

Preclinical analysis of novel prognostic transcription factors and immune‑related gene signatures for bladder cancer via TCGA‑based bioinformatic analysis

  • Authors:
    • Yuyou Deng
    • Xin Hong
    • Chengfan Yu
    • Hui Li
    • Qiang Wang
    • Yi Zhang
    • Tian Wang
    • Xiaofeng Wang
  • View Affiliations

  • Published online on: March 3, 2021     https://doi.org/10.3892/ol.2021.12605
  • Article Number: 344
  • Copyright: © Deng 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

Bladder cancer (BLCA) is a common malignancy of human urinary tract, whose prognosis is influenced by complex gene interactions. Immune response activity can act as a potential prognostic factor in BLCA. The present study established a prognostic model, based on the identification of tumor transcription factors (TFs) and immune‑related genes (IRGs), and further explored their therapeutic potential in BLCA. The enrichment scores of 29 IRG sets, identified in The Cancer Genome Atlas BLCA tumor samples, were quantified by single‑sample Gene Set Enrichment Analysis. The abundance of infiltrated immune cells in tumor tissues was determined using the Estimating Relative algorithm. Tumor‑related TFs and IRGs signatures were retrieved using Least Absolute Shrinkage and Selection Operator Cox regression analysis. A prognostic gene network was built using Pearson's correlation analysis as a means of predicting the regulatory relationship between prognostic TFs and IRGs. A nomogram was devised to also predict the overall survival (OS) rate of patients with BLCA. Based on the Genomics of Drug Sensitivity in Cancer data, potential therapeutic drugs were identified upon analyzing the relationship between the expression level of prognostic genes and respective IC50 values. In vitro experiments were implemented for further validation. Respective TF binding profiles were acquired from the JASPAR 2020 database. The elevated infiltration of CD8+ T Cells was correlated with an improved OS of patients with BLCA. An innovative prognostic model for BLCA was then constructed that composed of nine putative gene markers: CXCL13, prepronociceptin, microtubule‑associated protein tau, major histocompatibility class I polypeptide‑related sequence B, prostaglandin E2 receptor EP3 subtype, IL20RA, proepiregulin, early growth response protein 1 and FOS‑related antigen 1 (FOSL1). Furthermore, a theoretical basis for the correlation between the prognostic TFs and IRGs was reported. For this, 10 potentially effective drugs targeting the TFs in the present model for patients with BLCA were identified. It was then verified that downregulation of FOSL1 can lead to an enhanced sensitivity of the TW37 in T24 bladder cancer cells. Overall, the present prognostic model demonstrated a robust capability of predicting OS of patients with BLCA. Hence, the gene markers identified could be applied for targeted therapies against BLCA.

Introduction

Bladder cancer (BLCA) is a common malignancy that affects the urinary tract, with an estimated 81,400 new cases diagnosed in the United States in 2020 (1). BLCA represents a major cause of tumor-associated death that muscle-invasive bladder cancer outcomes remain <10% survival at 5 years despite aggressive treatment (2). Patients with BLCA are more likely to develop distant metastasis when their respective tumor cells invade the bladder's muscle membrane (3). Historically, the combination chemotherapy, including methotrexate, vinblastine, doxorubicin and cisplatin (MVAC), has been the first-line therapy for advanced BLCA. Nevertheless, the clinical benefits of this multi-chemotherapeutic approach have been restricted due to extensive drug toxicity, which includes neutropenia, cardiac and neurological conditions, thus making approximately half of patients with BLCA ineligible for this type of chemotherapy (4,5).

The suppression of programmed cell death ligand 1 (PD-L1) or programmed cell death protein 1 (PD-1) may recover immune cell cytotoxicity and also suppresses tumor progression (6). Hence, the application of PD-L1 and PD-1 inhibitors appear to have a positive impact on the overall survival (OS) of patients with advanced BLCA (7). Still, the immunotherapeutic efficiency of these drugs varies among patients, where some patients can achieve prolonged survival while others may show continuous tumor progression (8). Therefore, identifying novel prognostic genes that may predict patient survival and sensitivity to immunotherapy is important so that personalized treatments can be further implemented.

Previous studies, focused on the tumor microenvironment (TME) associated with BCLA, have indicated that the efficacy of certain immunotherapies is associated with host immune response and the composition of infiltrated immune cells in the TME (9,10). Indeed, a number of IRG signatures, associated with prognosis of patients with MIBC, have been reported (11). Although a number of genes and mechanisms have been shown to be closely associated with the immune response and prognosis of BLCA, a more comprehensive overview of all potential immune-related genes (IRGs) and their regulatory networks in BLCA is needed.

Upon analysis of The Cancer Genome Atlas (TCGA) database, the present study investigated the immune landscape associated with the transcriptome of patients with BLCA. Hence, the differentially expressed IRGs and transcription factors (TFs) associated with patients from high and low immune response groups were compared. A putative prognostic model, mainly consisting of nine IRGs and TFs, was constructed using a Least Absolute Shrinkage and Selection Operator (LASSO) Cox regression model. According to various statistical methods, the model was robust enough to predict the OS of patients with BLCA. Additionally, a gene-based network that could decipher the potential regulatory relationships between TFs and IRGs was constructed. Finally, a small molecule inhibitor named TW-37 was tested as a putative therapeutic drug for BLCA, using T24 cells as an in vitro model.

Materials and methods

Normalization of gene expression data and clinical information

Firstly, fragments per kilobase million (FPKM) data of RNA-sequencing profiles from BLCA samples (n=433) were downloaded from TCGA (https://portal.gdc.cancer.gov). Clinical information of patients with BLCA (n=391) was downloaded from TCGA (https://portal.gdc.cancer.gov/). Case records with unknown Tumor-Node-Metastasis stages or clinical stages were excluded (n=35). Before data analysis, FPKM values were transformed into transcripts per kilobase million (TPM) values using R software 3.6.3 (https://www.r-project.org).

Implementation of immune-related hierarchy clustering

Immune, stromal and tumor purity scores, and ESTIMATE scores were evaluated, based on normalized gene expression data matrix, using the ESTIMATE R package (version 3.6.3) (12). IRGs, originated from respective tumor tissues, were collected from the immunological signatures gene dataset from the Molecular Signatures Database (MSIGDB) and downloaded from the Broad Institute official website (https://www.gsea-msigdb.org/gsea/msigdb/collections). The quantified scores of 29 IRG sets were acquired from published signature gene lists across all BLCA samples using the single-sample Gene Set Enrichment Analysis (ssGSEA) method (13). The ssGSEA scores of each individual IRG set were respectively obtained and normalized. The quantified scores of each IRG set were grouped into high, medium and low immune response group according to the ssGSEA scores. This classification was performed using a hierarchical agglomerative clustering method, based on Euclidean distance accessible on the ‘sparcl’ R package (14).

Estimative of the immune cell proportions in the BLCA tumor microenvironment

The Cell Type Identification By Estimating Relative (CIBERSORT) R package was utilized to assess the proportion of tumor-infiltrating immune cells in the TME (15). BLCA gene expression profiles were normalized using the limma-voom method (16) and transformed into an infiltrating proportion of 22 immune cells. The relative proportions of these 22 immune cells for each sample was determined. Significant results with P<0.05 were selected for further analyses.

Differentially expressed genes (DEGs) from distinct immune response groups

The R package ‘limma’ was used to identify DEGs in the low versus high immune response BLCA groups, the empirical Bayesian approach was implemented to evaluate changes on gene expression (17). Genes with P<0.05 and a |log2 fold-change (FC) value|>1.0 were defined as DEGs. The list of IRGs was retrieved from the Immunology Database and Analysis Portal (ImmPort, http://immport.niaid.nih.gov) (18). Among the 2,498 IRGs retrieved from the ImmPort database, 401 differentially expressed IRGs were identified using a cut-off of |log2FC|>1.0 and adjusted P<0.05. Tumor-related TFs were obtained from the Cistrome project (http://cistrome.org/CistromeCancer/) for further analysis (19).

Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses

The R package ‘clusterProfiler’ was used to implement GO and KEGG evaluation as well as the Gene Set Enrichment Analysis (GSEA) (20). The GSVA R package was also used to perform the Gene Set Variation Analysis (GSVA) among the tumor samples with changes on evaluated pathway activity, upon comparing high versus low immune response groups (13). The P<0.05 and |log2FC|>0.20 was set as the cut-off threshold for the comparative analysis of the activated or suppressed signaling pathways.

Establishment of a prognostic gene signature using LASSO Cox regression modeling

Genes from the ‘immunological signatures gene set’, which was retrieved from the MSIGDB were selected as potential prognostic biomarkers. For this, DEG analysis was performed by comparing high versus low immune response groups to narrow down the screening scope. Univariate and multivariate Cox regression analyses were performed to build an appropriate prognostic signature for BLCA samples, based on the differentially expressed IRGs. The risk score of the prognostic immune gene model was calculated, for each patient tumor, according to the relative expression of each individual gene and its associated coefficient. The calculation formula was the following: Risk score = ∑i=1n(coefi×Expri),

where: ‘Expri’ represents the expression value of each specific IRG in the model of patient ‘I’, ‘coefi’ is the coefficient of gene ‘I’ obtained via multivariate Cox regression analysis.

Prediction ability of the prognostic gene signature

According to the patient clinical data obtained from TCGA database (n=391), patients with BLCA were classified in low- and high-risk groups, as determined by the risk scores, which were generated using our IRG based prognostic model. Thereafter, a time-dependent receiver operating characteristic (ROC) curve was used to assess the sensitivity and specificity of the model. In order to evaluate the prediction accuracy of the model, the area under the curve (AUC) for 1-, 3- and 5-year OS was further assessed, according to the ‘survival ROC’ R package (21).

Independence of the TF and IRG-based prognostic model

To determine whether our prognostic model was significantly distinct from standard clinical characteristics, all patient details featured on the TCGA-BLCA dataset, including age, sex, pathological tumor stage, histological subtype and TNM stage, were assessed. Patients were divided according to their age (≤ and ≥65 years). Patient subgroups were generated according to sex, I–IV pathological tumor stage, N0 and N1-3 stage, pathological T0-T2 and T3-4 stage. Low and high-risk subgroups were defined according to the mean risk score of OS assessed by the COX regression model for each patient. Forest plots were used to illustrate the hazard ratio (HR) of each subgroup.

TF binding site prediction and correlation analysis between IRGs and TFs

Boxplots were constructed to show the relationship between risk score and the distinct immune response subgroups. Heatmaps were constructed according to the expression of IRGs from each respective group (low, median and high immune response). Dot plots and corresponding fit curves were used to illustrate the correlation between model-related immune genes and prognostic transcription factors. According to the expression values of each IRG from the prognostic model and respective TFs, Pearson's correlation coefficient analysis was performed. The TF binding motifs (TFBMs) of related TFs, as well as the TF binding sites of related target genes, were identified using the JASPAR 2020 database (22).

Correlation analysis between TFs and drug sensitivity

Data were extracted from the Genomics of Drug Sensitivity of Cancer database (GDSC; http://www.cancerrxgene.org), in which numerous anticancer drugs were screened using various cancer cell lineages to determine their sensitivity. Gene expression profiles in 990 cancer cell lines and a drug screening IC50 dataset, covering 265 compounds across 19 BLCA cell lines, was downloaded from the GDSC database. The Pearson's correlation values between the gene expressions of prognostic TFs and IC50 values of each compound was investigated.

Reagents and kits

The small molecule inhibitor TW-37 (MCE; category no. HY-12020) was purchased from MedChemExpress and stored as 10 mM solution using DMSO as a solvent. Primary antibodies were against FOSL1 (Cell Signaling Technology, Inc; cat. no. 5281) and GAPDH (Wanlei Bio, http://www.wanleibio.cn; cat. no. WL01114) were diluted 1,000 times using Tris-HCL buffer solution (cat. no. WLA098, Wanlei Bio, http://www.wanleibio.cn). Secondary antibodies were anti-rabbit and anti-mouse IgGs (Cell Signaling Technology, Inc; cat. nos. 7074 and 7076) were diluted 3,000 times using Tris-HCL buffer solution. Negative control fluorescein (FAM) and FOSL1 siRNAs were designed by Shanghai GenePharma Co., Ltd.

Cell culture and viability analysis

T24 bladder cancer cells were obtained from The Cell Bank of Type Culture Collection of The Chinese Academy of Sciences. Cells were cultured using Dulbecco's modified Eagle medium (DMEM) supplemented with 10% fetal bovine serum (FBS) (both Gibco; Thermo Fisher Scientific, Inc.) and incubated at 37°C in an atmosphere of 5% CO2. Cell viability was assessed using an MTS Assay kit (cat. no. ab197010; Abcam). Absorbance at a wavelength of 450 nm was measured using a BioTek ELISA reader.

Immunoblotting

T24 cells were collected from 12-well plates and lysed with RIPA buffer (Cell Signaling Technology, Inc.) for 20 mins at 4°C. Whole cell lysates were then centrifuged at 12,000 × g at 4°C for 15 mins, and respective supernatants were denatured at 100°C for 15 mins after addition of loading buffer. Protein samples were quantified by protein electrophoresis method, using the SDS-PAGE kit (cat. no. WLA013, Wanlei Bio, http://www.wanleibio.cn). The bicinchoninic acid method was used to determine the protein content in each sample. Protein (50 ug/lane) were separated via 12% SDS-PAGE. The separated proteins were subsequently transferred onto nitrocellulose membranes (EMD Millipore) and blocked with 20% skimmed milk tris buffered saline (TBS) for 30 min at room temperature. The membranes were incubated with primary antibodies at 4°C for 8 h. Subsequently, the nitrocellulose membranes were washed with Tris-Hcl solution for 10 min, and ECL chemiluminescent substrate (Yeasen, Shanghai, China, http://www.yeasen.com) was used to visualize the protein bands.

Transfection protocol

In total, ~10,000 T24 cells were seeded into each well of the 12-well plate, and incubated with 10% fetal bovine serum in DMEM for 24 h. After removing the cell culturing medium, 300 ng siRNAs with 3 µl Lipofectamine® 2000 (Invitrogen; Thermo Fisher Scientific, Inc.) in 250 µl DMEM as the transfection mixture was added to each well. The transfection mixture and extra 250 µl DMEM were added into each well of the 12-well plate. The gene sequence of negative control siRNA was: 5′-UUCUUCGAACGUGUCACGUTT-3′, the gene sequence of FOSL1 targeted siRNA was: 5′-UUAUGAAUGAAAAGUUCUCGG-3′. After incubation at room temperature for 20 min, 500 µl pre-warmed 20% fetal bovine serum DMEM was added per well. After cultivating the transfected T24 cells at 37°C for 48 h, cells were collected for detection of the silencing efficiency.

Statistical analysis

Statistical analyses and data visualization were performed using GraphPad Prism 8.0 or R software 3.6.3. P<0.05 was considered to indicate a statistically significant difference. The Kruskal-Wallis followed by Dunn's or the Mann-Whitney U tests were performed to determine the significance of the median value between different groups. The two-tailed unpaired Student's t-test was applied to define the significance when comparing mean values of two unpaired groups with different characteristics. ANOVA and Tukey's post hoc test was used to analyze data with more than two groups. Kaplan-Meier survival curves were plotted to illustrate the distinct OS between the groups. A log-rank test was used to determine the statistical significance between survival curves. Each in vitro assay was performed three times and data are presented as the mean ± standard deviations.. For the cell viability assays, the two-tailed unpaired Student's t-test was used to compare differences between two groups.

Results

Immune response phenotype and enrichment score landscape of the TME from BLCA samples

Infiltrated immune cells in the TME control a series of immune-related processes that can frequently lead to antitumor activities (23). The ssGSEA of the BLCA gene expression data revealed quantified enrichment scores of the immunological signature gene set (obtained from the MSIGDB) that corresponded to 29 IRG subsets. Enrichment scores of the identified normalized gene set were higher for genes related to i) associate dendritic cells (aDCs), ii) antigen-presenting cells (APC) co-inhibition, iii) APC co-stimulation, iv) B cells, v) CCR, vi) CD8+ T Cells, vii) immune check-point, viii) cytolytic activity, ix) DCs (dendritic cells), x) HLA genes, xi) interdigitating (i) DCs, xii) inflammation-promoting, xiii) macrophages, xiv) masT Cells, xv) MHC class I, xvi) neutrophils, xvii) natural killer (NK) cells, xviii) para-inflammation, xix) pDCs, xx) T Cell co-inhibition, xxi) T Cell co-stimulation, xxii) T follicular helper cells (Tfh), xxiii) T helper 1 (Th1) cells, xxiv) Th2 cells, xxv) TIL, xxvi) Treg cells, xxvii) type I IFN response and xxviii) type II IFN response (Fig. 1E). According to the ssGSEA enrichment scores, BLCA samples were divided into high (154 samples), median (168 samples) and low (92 samples) immune response subgroups. Furthermore, the stromal, immune and ESTIMATE scores were calculated as well as the tumor purity of 414 BLCA samples. The violin plot indicated that BLCA samples clustered in the low immune response subgroup had the highest tumor purity but lower stromal, immune and ESTIMATE scores compared with those included into the medium or high immune response subgroups (Fig. 1A-D). The enrichment scores of each IRG set were also lower in the low immune response group when compared with the high immune response group (Fig. 1E). The BLCA samples were classified into three subgroups for further investigation using the ssGSEA method, and the clinical characteristics of patients with bladder cancer from TCGA database are listed in Table I.

Table I.

Clinical characteristics of 391 patients (239 alive and 152 dead) with bladder cancer acquired from The Cancer Genome Atlas database.

Table I.

Clinical characteristics of 391 patients (239 alive and 152 dead) with bladder cancer acquired from The Cancer Genome Atlas database.

Patient status

VariableAlive, n (%)Dead, n (%)Overall
Sex
  Female58 (24.3)42 (27.6)100 (25.6)
  Male181 (75.7)110 (72.4)291 (74.4)
Age, years
  ≥65131 (53.0)116 (47.0)247 (63.2)
  <65108 (75.0)36 (25.0)144 (36.8)
Grade
  High216 (90.4)152 (100)368 (94.1)
  Low21 (8.8)0 (0)21 (5.4)
  Unknown2 (0.8)0 (0)2 (0.5)
Clinical Stage
  I2 (0.8)0 (0)2 (0.5)
  II98 (41.0)26 (17.1)124 (31.7)
  III85 (35.6)47 (30.9)132 (33.8)
  IV54 (22.6)78 (51.3)132 (33.8)
  Unknown0 (0)1 (0.7)1 (0.3)
T stage
  01 (0.4)0 (0)1 (0.3)
  13 (1.3)0 (0)3 (0.8)
  230 (12.6)8 (5.3)38 (9.7)
  2a21 (8.8)2 (1.3)23 (5.9)
  2b35 (14.6)17 (11.2)52 (13.3)
  327 (11.3)13 (8.6)40 (10.2)
  3a36 (15.1)32 (21.1)68 (17.4)
  3b37 (15.5)41 (27.0)78 (19.9)
  45 (2.1)6 (3.9)11 (2.8)
  4a23 (9.6)18 (11.0)41 (10.5)
  4b1 (0.4)3 (2.0)4 (1.0)
  Unknown20 (8.4)11 (7.2)31 (7.9)
  X0 (0)1 (0.7)1 (0.3)
N stage
  0165 (69.0)60 (39.5)225 (57.5)
  120 (8.4)24 (15.8)44 (11.3)
  229 (12.1)45 (29.6)74 (18.9)
  33 (1.3)5 (3.3)8 (2.0)
  X19 (7.9)16 (10.5)35 (9.0)
  Unknown3 (1.3)2 (1.3)5 (1.3)
M stage
  0125 (52.3)59 (38.8)184 (47.1)
  14 (1.7)6 (3.9)10 (2.6)
  X109 (45.6)86 (56.6)195 (49.9)
  Unknown1 (0.4)1 (0.7)2 (0.5)

[i] T, tumor; N, node; M, metastasis; X, for the Tx, Nx or Mx stage.

Enrichment scores of gene subsets related to CD8+ T Cells may predict BLCA prognosis

The clinical data of patients with BLCA and their corresponding immunological signatures were combined. Kaplan-Meier survival analysis was performed to further search potential prognostic factors that could show that the survival curve of the high immune response group had a trend toward improved prognosis compared with the low immune response group. However, these changes were not statistically significant (P=0.50; Fig. 2A). Moreover, it was verified that higher enrichment scores (scores ≥ mean values) of CD8+ T Cell-related genes predicted an improved BLCA prognosis (Fig. 2B). The 5-year survival rate of the low score CD8+ T Cell group was 35.9±5.09%, with a 95% confidential interval from 0.272 to 0.474, while the rate was 55.8±5.65%, with a 95% confidential interval from 0.457 to 0.680, in the high-score CD8+ T Cell group (P=0.022). Thus, the increased infiltration rate of CD8+ T Cells may benefit the long-term OS of patients with BLCA.

Landscape of immune cell infiltration in the BLCA microenvironment

In order to estimate the immune cell infiltration in each individual BLCA sample, the ‘CIBERSORT’ R package was applied to the BLCA-specific gene expression profile data matrix. We found 22 types of infiltrated immune cells were identified (Fig. 3A). A higher abundance of resting DCs and M2 macrophages was observed in the high immune response group compared with the low response group. At the same time, activated DCs and naive CD4+ T Cells were decreased in the high immune response group (Fig. 3B). In order to validate the prognostic effect of the amount of CD8+ T Cell infiltration in patients with BLCA, Kaplan-Meier curves and log-rank tests were implemented according to the CIBERSORT results (Fig. 3C). Although the changes were not statistically significant (P=0.061), there was a trend toward an improved BLCA prognosis in the high CD8+ T Cell infiltration group compared with the low infiltration group. The 5-year survival rate of the low CD8+ T Cell infiltration group was 35.6±8.16%, with a 95% confidential interval from 0.227 to 0.558, while the rate was 58.4±7.03%, with a 95% confidential interval from 0.461 to 0.739, in the high infiltration group. Thus, according to the results of ssGSEA and CIBERSORT analyses, higher infiltration of CD8+ T Cells in the TME can improve the OS of patients with BLCA.

Assessment of signaling pathway activities in low and high immune response groups

To evaluate the distinct signaling pathway activities between the low and high immune response groups, a GSVA of hallmark gene sets was implemented based on the gene expression profiles of BLCA samples. Based on this comparative analysis, it was demonstrated that there were 46 activated signaling pathways in high immune response group. Both a volcano plot and heatmap were constructed to show the landscape of most activated and/or suppressed signaling pathways (Fig. 4A). In addition, biological process, cellular component, molecular function and KEGG enrichment analysis were performed using the GSEA approach. This analysis was performed according to gene expression profile of identified IRG sets that were positively enriched in the high immune response group (Fig. 4B-E). In the high immune response group, immune related signaling pathways, including JAK-STAT, NF-κB and PD-L1 expression, and PD-1 checkpoint pathway in cancer, antigen processing and presentation via MHC class Ib, and T Cell receptor signaling pathway were enriched according to GO and KEGG enrichment analyses.

Differentially expressed tumor-related IRGs and TFs in low and high immune response groups

Among the 2,498 IRGs retrieved from the ImmPort database, 50 downregulated and 351 upregulated genes, were identified as the differential expressed IRGs. A heatmap was designed to illustrate the expression level of 50 random immune-related DEGs, while a volcano plot shows the whole content of immune-related DEGs identified (n=401) (Fig. 5A). The top 20 most significant immune-related DEGs, identified in the comparison between low and high immune response groups, are listed in Table II. Moreover, 318 tumor-related TFs were obtained from the Cistrome database. In total, 44 differentially expressed TFs were detected, of which 14 were downregulated and 30 were upregulated in the high immune response group. A heatmap and a volcano plot were designed to illustrate the expression landscape of differentially expressed TFs by comparing the low and high immune response groups (Fig. 5B).

Table II.

Top 10 differentially expressed immune-related genes.

Table II.

Top 10 differentially expressed immune-related genes.

Gene IDlogFCAverage expressionP-valueAdjusted P-value
CRH−3.9090.3965 3.82×10−14 4.01×10−11
FAM3B−2.6582.9128 4.70×10−10 4.49×10−07
FAM3D−2.6261.7495 4.59×10−11 4.51×10−08
BMP3−2.6701.6009 5.20×10−10 4.96×10−07
CHP2−2.3701.0492 5.19×10−9 4.80×10−06
IDO14.61263.1194 3.63×10−41 4.48×10−38
CCL184.63533.8358 4.51×10−40 5.54×10−37
CXCL104.71125.5732 1.64×10−49 2.06×10−46
CXCL134.73143.5218 8.75×10−44 1.09×10−40
CXCL95.35254.2464 1.84×10−59 2.36×10−56

[i] FC, fold-change.

Construction of a BLCA prognostic model based on differential expressed IRGs and TFs

In order to identify the potential associations between immune-related DEGs, differentially expressed TFs and BLCA prognosis, univariate Cox regression analysis was implemented based on gene expression and clinical information. Results from this regression analysis indicated that 54 IRGs were associated with the OS of patients with BLCA. Moreover, the HR values of five distinct genes, including microtubule-associated protein tau (MAPT), prostaglandin E2 receptor EP3 subtype (PTGER3), β nerve growth factor, IL31RA and proepiregulin (EREG), were >1, while 49 prognosis-associated immune genes had HR values <1. Moreover, seven TFs were identified as putative prognostic factors (Fig. 6A). The LASSO method was applied to further streamline potential prognostic genes originated from the univariate Cox regression analysis. Hence, 15 genes were selected with an optimal penalty parameter log (λ) value=−3 (Fig. 6B and C). Thereafter, a multivariate Cox regression model was established based on the gene expression value of the candidate genes (n=15). As a result, an immune-related signature composed of nine candidate genes, was defined for the OS prediction of patients with BLCA. The risk score of this prognostic model was calculated as the following: Risk Score=CXCL13 expression × (−0.128) + prepronociceptin expression × (−0.129) + MAPT expression × (0.248) + major histocompatibility class (MHC) I polypeptide-related sequence B (MICB) expression × (−0.268) + PTGER3 expression × (0.192) + IL20RA expression × (−0.186) + EREG expression × (0.0734) + early growth response protein 1 (EGR1) expression × (0.162) + FOSL1 expression × (0.213). Patients with BLCA were divided into low and high-risk groups according to the median value of the risk score of each sample. The Kaplan-Meier curves indicated that the low-risk group had prolonged OS compared with the high-risk group (Fig. 6D). The 5-year OS for the high-risk group was 23.4±5.95%, with a 95% confidential interval from 0.142 to 0.385, while the 5-year OS for the low risk group was 71.7±4.27%, with a 95% confidential interval from 0.740 to 0.907 (P<0.05).

Prediction accuracy of the prognostic IRG model

Time-dependent receiver operating characteristic (ROC) curves were applied to examine the prediction accuracy of the OS prognostic model over the 1-, 3- and 5-year period. The values of area under the ROC (AUC) for the prognostic model were 0.73, 0.82, and 0.83 at 1-, 3- and 5-year period, respectively (Fig. 7A). Thereafter, the risk score of each patient was sorted into a descending order. The survival status of each patient was marked accordingly on the dot plots. It was observed that patients with low risk scores had an improved OS rate compared with patients with high risk scores (Fig. 7B and C). The heatmap shows the expression levels of nine genes from the prognostic model (Fig. 7D). According to the results of the present study, the differentially expressed IRGs and TFs based gene model exhibited good predictive performance.

A robust prognostic gene model for BLCA

Cox regression analyses were performed to evaluate whether our prognostic model acted as an independent prognostic factor among other clinical traits, including age, sex, and pathological and T/N stages. Univariate analysis indicated that risk score, pathological stage, MAPT, EGR1 and/or FOSL1 expression may affect the prognosis of patients with BLCA (Fig. 8A). Multivariate Cox regression results revealed that both pathological stage and risk score served as independent prognostic factors for BLCA (Fig. 8B). The AUCs of the time dependent ROC curves for pathological stage during the 1-, 3- and 5-year periods were 0.59, 0.67 and 0.62, respectively (Fig. 8C). The comparison between the AUCs for risk score and those for pathological stage indicated that the present model had improved predictive accuracy (P=0.0029) of patient OS compared with the pathological stage (Fig. 8D).

Clinical application of a novel nomogram incorporating the IRG model

According to the results of Cox regression analyses, which were based on the clinical traits and the risk score of the prognostic model, a nomogram was constructed that combined other factors, including age, sex, pathological and T/N stages, as well as the IRG model to provide a more quantitative method for clinicians to assess the 1-, 3- and 5-year OS of patients with BLCA (Fig. 9A). In this manner, every patient received a quantifiable point for each prognostic measuring parameter. The calibration plots illustrated that, in comparison with an ideal model, the current nomogram had a stable predictive performance for 1- and 3-year OS (Fig. 9B).

Construction of a prognostic TF- and IRG-based regulatory network

The correlation between the nine selected IRGs, used to implement the prognostic model, and corresponding risk scores was further established (Fig. 10A and B). Thus, Pearson's correlation analysis was performed between the prognostic TFs and IRGs. The cut-off thresholds presently defined were |Pearson correlation coefficient value|>0.5 and P<0.01. All prognostic IRGs (n=45) were positively regulated by prognostic TFs, where EREG acted as a high-risk IRG that was positively regulated by FOSL1 (Fig. 10C). A Sankey map illustrated the association between the expression level of the nine selected IRGs and the infiltration of immune cell subtypes (Fig. 10D). According to the correlation analysis results, FOSL1 and ERG1 were identified as two TFs capable of performing pivotal transcriptional regulation processes within tumor cells.

Analysis of TF binding motifs and binding sites in targeted IRGs

According to the prognostic gene signature and correlation analysis results, the specific TFBMs of FOSL1 and ERG1 were analyzed using the JASPAR 2020 database. The predicted matrix IDs of TFBMs for EGR1 and FOSL1 were MA0162.2 and MA0477.1, respectively (Fig. 11A and B). According to the results of gene expression correlation analysis, it was reported that MICB and EREG were positively regulated by EGR1 and FOSL1, respectively. Therefore, potential specific TFBSs for EGR1 and FOSL1 were identified within the TSSs of MICB and EREG genes (Table III). Thus, blocking these potential TFBSs may interfere the transcription process of related target genes by inhibiting TF binding to respective TSS along the gene sequences.

Table III.

Transcription factor binding site matching results of target genes.

Table III.

Transcription factor binding site matching results of target genes.

Matrix IDNameScoreRelative scoreStartEndPredicted sequence
MA0162.2EGR116.3420.9551315328TCCCCTCCCCCACC
MA0162.2EGR113.4240.9239277290AGCCCTCCCCCTCC
MA0162.2EGR111.5470.9038928941CCTCCTCCCCCATT
MA0477.1FOSL15.91750.850818091819AATGAGTTAGG
MA0477.1FOSL14.38210.829516771687TGTGAATAACC
MA0477.1FOSL14.19610.8269467477TGTGAGTCTGT

[i] EGR1, early growth response protein 1; FOSL1, FOS-related antigen 1.

Correlation between modeled gene expression and sensitivity to targeted therapeutics

To examine the correlation between all gene expressions integrated into our prognostic model and targeted drug sensitivity, experimental drug sensitivity data covering 265 therapeutic compounds across 19 BLCA cell lines were retrieved from the GDSC database. According to the expression profiles of the present model genes, the top 10 correlated drugs were identified, using a cut-off threshold of |Pearson correlation coefficient| >0.5 and a P<0.025 (Table IV). A Pearson correlation coefficient curve suggested that the IC50 values of ‘AS601245’ and ‘PI-103’ were proportional to EGR1 expression levels. At the same time, the IC50 values of ‘TW 37’ and ‘STF-62247’ were inversely correlated to the expression of FOSL1 (Fig. 11C).

Table IV.

Results of model gene and IC50 value of therapeutics using the Pearson's correlation analysis based on Genomics of Drug Sensitivity in Cancer bladder cancer cells data.

Table IV.

Results of model gene and IC50 value of therapeutics using the Pearson's correlation analysis based on Genomics of Drug Sensitivity in Cancer bladder cancer cells data.

GeneDrugCorrelationP-valueTargetPathway
EGR1AS6012450.5720.0171JNKJNK and p38 signaling
EGR1PI-1030.5500.0218PI3K α, DNAPKPI3K signaling
FOSL1QL-XI-92−0.5590.0201DDR1RTK signaling
FOSL1GSK269962A−0.560.0198ROCK1, ROCK2Cytoskeleton
FOSL1Bicalutamide−0.5810.0156Androgen receptorOther
FOSL1MLN4924−0.5840.0151NEDD8-activating enzymeOther
FOSL1Olaparib−0.5850.0149PARP1, PARP2Genome integrity
FOSL1T0901317−0.5960.0131LXROther
FOSL1STF-62247−0.5980.0128AutophagyOther
FOSL1TW 37−0.6050.0118BCL-2, BCL-XLApoptosis regulation

[i] EGR1, early growth response protein 1; FOSL1, FOS-related antigen 1.

Verification of the predicted correlation between TW-37 and FOSL1

To further validate the predicted relationship between the expression levels of FOSL1 and the IC50 value of potential drugs, in vitro studies were performed using T24 BLCA cells. Upon cell treatment, it was observed that the TW-37 significantly impaired the viability of T24 cells at the concentration of 600 nM (Fig. 11D). However, the cytotoxicity of TW-37 was attenuated after knockdown of FOSL1 (Fig. 11E). Cell viability was 61.76±3.66% when T24 cells without knockdown of FOSL1 were treated with 600 nM TW-37 for 36 h. After depletion of FOSL1 in T24 cells, cell viability increased to 83.06±3.57%. Thus, increased FOSL1 expression may improve the drug resistance of TW-37 in T24 cells.

Discussion

BLCA is considered a complex malignancy with high morbidity and mortality rates, which have caused a severe social-economic burden worldwide. So far, in 2020, a total of 17,980 estimated deaths in the USA have been related to BLCA (1). Advances in the trans-urethral resection of bladder tumor, as well as in the application of bladder instillation therapy, have been able to control tumor progression in patients with early BLCA (24). Still, approximately half of patients with MIBC suffer from recurrence or distant metastasis after radical cystectomy, which frequently results in a significant decrease on the survival rate and makes the management of advanced BLCA more challenging (25).

Recent progress on the field of immunotherapy have provided opportunities for precision management and treatment of BLCA. In the past decade, therapeutic approaches that target PD1 and PDL1 have already been applied to treat patients with BCLA (26). Since high-throughput sequencing technologies and bioinformatics analyses have become more conventionally applied into biological studies, oncologists can now implement such technologies to explore potential factors that may predict prognostic outcomes and, consequently, affect the efficacy of distinct cancer immunotherapies (27). Thus, identifying novel IRG signatures is important to guide decision-making on immune therapeutics.

Previous research on TME alterations has been mostly related to infiltrating immune cells, based on the deconvolution of bulk gene expression profiles (28,29). In the current study, a subset of BLCA samples were classified into three categories (high, intermediate and low immune response groups) according to the enrichment scores of 29 IRG sets via ssGSEA. Estimated results of tumor purity indicated that the low immune response group had a significantly higher tumor purity compared with the high immune response group, implying that the population of stromal and immune cells were decreased in samples originated from low immune response tumors. Notably, it was reported that high enrichment scores of genes related to CD8+ T Cell infiltrates were correlated with improved prognostic outcomes. Based on the CIBERSORT analysis, similar findings validated that a high infiltration of CD8+ T Cells could improve the 5-year OS of patients with BCLA. CD8+ T Cells are typically considered anticancer cells in the TME (27). Upon interaction with T Cell antigen receptors, these cells bind peptide MHCs, expressed on the antigen-presenting cell surface, to kill tumor cells (30). However, after infiltrating into tumor tissues, CD8+ T Cells gradually become dysfunctional, which may disable their antitumor activity (31). Identifying novel genes associated with T Cell exhaustion, as well as therapies that could restore the cell killing ability of CD8+ T Cells, should be focused on in future immunological studies.

In the high immune response group, GSVA results suggested that multiple classic pathways were activated, including those related to apoptosis, antigen processing and presentation, T Cell and B cell receptors, NK cell mediated cytotoxicity, JAK-STAT, RIG-I-like, NOD-like receptor and TOLL-like receptor (TLR) signaling cascades. Specifically, GSEA results based on the KEGG database indicated that the NF-κB, PD-L1 and PD-1 checkpoint pathways were enriched in the high immune response group. These signaling pathways have been reported to be related to human immune response process, and for decades they have been studied in the area of onco-immunology (6,32). Pathways related to apoptosis activation, antigen processing and presentation, cancer-associated macrophages, T and B cell receptors, as well as NK cell-mediated cytotoxicity, may suggest thaT Cellular immunity is activated in the high immune response group. RIG-I-like receptor, NOD-like receptor and TLR signaling pathways are pattern recognition receptors that play a pivotal role in initiating the innate immune response (33). TLR stimulation has been shown to rescue activated Th1 cells from exhaustion. Particularly, TLR2 engagement in CD8+ T Cells appears to be capable of augmenting antitumor activity in vivo (6). Relevant evidence has also revealed that the rescue of chronically activated CD8+ T Cells by blockage of checkpoint regulators (for example PD-1 and cytotoxic T lymphocyte protein 4) is a successful platform cancer immunotherapy (34). Accumulating evidence also supports the possibility of manipulating TLR signaling to further modulate the expression of immune checkpoint molecules (35).

In the current study, a prognostic model was constructed based on differentially expressed IRGs and TFs identified in high and low immune response groups of BLCA samples. EGR1 and FOSL1 are risk TFs, detected in the present model, which may decrease the OS of patients. Some reports have claimed that reactive oxygen species can induce the chromatin remodeling of effector T Cells and their conversion into exhausted T Cells, further attenuating host antitumor immunity (36). The FOSL1 transcription factor is a major effector of RAS-ERK1/2 pathway and FOSL1 can enhance pancreatic cancer metastasis by activating the EMT pathway (37,38). Such evidence suggests that the underlying mechanisms of EGR1 and FOSL1 as oncogenes may promote tumorigenesis and metastasis, resulting in impaired prognosis of patients with BLCA. Rodriguez-Aguayo et al (39) reported that silencing of PTGER3 expression in ovarian cancer cells is associated with decreased cell proliferation and attenuated invasiveness, while higher PTGER3 expression is associated with a shorter patient survival. The MAPT gene encodes the microtubule-associated protein Tau (40). Studies have suggested that increased levels of cytoplasmic and nuclear Tau, before intravesical chemotherapy, are significantly associated with decreased patients' recurrence-free survival (41). The EREG gene encodes epiregulin, an epidermal growth factor receptor ligand, which contributes to inflammation, angiogenesis, vascular remodeling and cell proliferation (42). In the present prognostic model, these aforementioned genes were recognized as risk factors for the decreased OS of patients with BLCA, serving as promising targets in a therapeutic intervention.

Previous studies have established gene models for prognosis prediction in patients with BLCA using bioinformatic analysis. Cao et al (43) identified seven EMT-related genes as a signature for the prognosis of MIBC. Jiang et al (11) constructed an IRG signature (TIM signature), which partially explains the complicated relationships between immune cell infiltration of TME and the survival. Furthermore, due to the wide use of the high throughput sequencing technologies and bioinformatic analysis, researchers have reported that differentially expressed non-coding RNAs also possess important roles in the regulation of gene expression, as well as putative predictors of patient survival in multiple types of cancer (44,45). The present study implemented comprehensive bioinformatics mining and then performed in vitro tests for further validation, thus providing novel insights for targeted therapies of BLCA in the near future. The time-dependent ROC curve of OS exhibited an improved predictive ability compared with pathological stage of patients with BLCA. A gene regulatory network was established to depict the relationship between the prognostic TFs and IRGs. The application of a nomogram, based on our model, provided a reference tool for clinical work. Hence, clinical oncologists may comprehensively assess the prognostic risk of each patient with BLCA. Of note, the present study proposed a prognostic model based on TFs and IRGs. Moreover, upon analysis of drug sensitivity datasets (GDSC database), it was observed that the expression of FOSL1 was negatively associated with the drug sensitivity of eight distinct therapeutic agents. In vitro experiments using T24 cells further demonstrated that silencing FOSL1 expression may enhance the drug resistance due to TW-37. These results provide supporting experimental evidence for our theoretical hypothesis based on bioinformatics analysis.

Inevitably, the current study had some limitations. Data from TCGA database was used to assess the immune response based on bioinformatics methods, the present study was unable to collect substantial clinical data from multiple medical centers to further confirm our current perspectives. Experimental assays using patient-derived tumor tissues are required to validate the present results. Additionally, the mechanisms of how non-coding RNAs could impact BLCA development should be further investigated.

In summary, the present study implemented a comprehensive way to assess the immune landscape of BLCA samples from TCGA database by constructing a novel prognostic model based on IRGs. The study also established a regulatory network of BLCA prognostic TFs and IRGs, thus identifying potential effective therapeutics which could target our model genes. The current study identified novel predictive gene signatures of BLCA, and it may also guide the development of drug combination strategies which may benefit patients with BLCA in the future.

Acknowledgements

Not applicable.

Funding

The present study was supported by the Peking University International Hospital Young and Middle-aged Research Startup Fund (grant no. YN2016QN04).

Availability of data and materials

The datasets generated and/or analyzed during the current study are available in The Cancer Genome Atlas (TCGA) repository, https://www.cancer.gov/tcga. The additional datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

YD and XW designed the study, performed the data analysis and drafted the initial manuscript. TW, QW, XH and HL collected data and participated in the analysis process of the gene profiles. CY and QW performed the in vitro experiments. CY and YZ provided academic urological advices and participated in clinical data analyzing and drafting of the initial manuscript. YD and XW confirmed the authenticity of all the raw data. All authors have 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:

BLCA

bladder cancer

MIBC

muscle invasive bladder cancer

TCGA

The Cancer Genome Atlas

DEGs

differentially expressed genes

ssGSEA

single-sample gene set enrichment analysis

CIBERSORT

Cell Type Identification By Estimating Relative

IRG

immune-related gene

TF

transcription factor

TFBM

transcription factor-binding motif

TFBS

transcription factor binding site

TSS

transcription starting site

GDSC

Genomics of Drug Sensitivity in Cancer

MHC

major histocompatibility complex

TME

tumor microenvironment

NK

natural killer cell

Treg

regulatory T Cell

GO

Gene Ontology

KEGG

Kyoto Encyclopedia of Genes and Genomes pathway

DCs

dendritic cells

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2020. CA Cancer J Clin. 70:7–30. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Babjuk M: Trends in bladder cancer incidence and mortality: Success or disappointment? Eur Urol. 71:109–110. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Knowles MA and Hurst CD: Molecular biology of bladder cancer: New insights into pathogenesis and clinical diversity. Nat Rev Cancer. 15:25–41. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Nadal R and Bellmunt J: Management of metastatic bladder cancer. Cancer Treat Rev. 76:10–21. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Loehrer PJ Sr, Einhorn LH, Elson PJ, Crawford ED, Kuebler P, Tannock I, Raghavan D, Stuart-Harris R, Sarosdy MF, Lowe BA, et al: A randomized comparison of cisplatin alone or in combination with methotrexate, vinblastine, and doxorubicin in patients with metastatic urothelial carcinoma: A cooperative group study. J Clin Oncol. 10:1066–1073. 1992. View Article : Google Scholar : PubMed/NCBI

6 

Zou W, Wolchok JD and Chen L: PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci Transl Med. 8:328rv3242016. View Article : Google Scholar

7 

Aggen DH and Drake CG: Biomarkers for immunotherapy in bladder cancer: A moving target. J Immunother Cancer. 5:942017. View Article : Google Scholar : PubMed/NCBI

8 

Carosella ED, Ploussard G, LeMaoult J and Desgrandchamps F: A systematic review of immunotherapy in urologic cancer: Evolving roles for targeting of CTLA-4, PD-1/PD-L1, and HLA-G. Eur Urol. 68:267–279. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Xue Y, Tong L, Liu F, Liu A, Liu A, Zeng S, Xiong Q, Yang Z, He X, Sun Y and Xu C: Tumor infiltrating M2 macrophages driven by specific genomic alterations are associated with prognosis in bladder cancer. Oncol Rep. 42:581–594. 2019.PubMed/NCBI

10 

Miyake M, Tatsumi Y, Gotoh D, Ohnishi S, Owari T, Iida K, Ohnishi K, Hori S, Morizawa Y, Itami Y, et al: Regulatory T Cells and tumor-associated macrophages in the tumor microenvironment in non-muscle invasive bladder cancer treated with intravesical bacille calmette-guerin: A long-term follow-up study of a Japanese cohort. Int J Mol Sci. 18:2017. View Article : Google Scholar : PubMed/NCBI

11 

Jiang W, Zhu D, Wang C and Zhu Y: An immune relevant signature for predicting prognoses and immunotherapeutic responses in patients with muscle-invasive bladder cancer (MIBC). Cancer Med. 9:2774–2790. 2020. View Article : Google Scholar : PubMed/NCBI

12 

Yoshihara K, Shahmoradgoli M, Martinez 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

13 

Hanzelmann S, Castelo R and Guinney J: GSVA: Gene set variation analysis for microarray and RNA-seq data. BMC Bioinformatics. 14:72013. View Article : Google Scholar : PubMed/NCBI

14 

Witten DM and Tibshirani R: A framework for feature selection in clustering. J Am Stat Assoc. 105:713–726. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, Hoang CD, Diehn M and Alizadeh AA: Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 12:453–457. 2015. View Article : Google Scholar : PubMed/NCBI

16 

da Silveira WA, Hazard ES, Chung D and Hardiman G: Molecular profiling of RNA tumors using high-throughput RNA sequencing: From raw data to systems level analyses. Methods Mol Biol. 1908:185–204. 2019. View Article : Google Scholar : PubMed/NCBI

17 

Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W and Smyth GK: Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43:e472015. View Article : Google Scholar : PubMed/NCBI

18 

Bhattacharya S, Andorf S, Gomes L, Dunn P, Schaefer H, Pontius J, Berger P, Desborough V, Smith T, Campbell J, et al: ImmPort: Disseminating data to the public for the future of immunology. Immunol Res. 58:234–239. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Wan B, Liu B, Huang Y, Yu G and Lv C: Prognostic value of immune-related genes in clear cell renal cell carcinoma. Aging (Albany NY). 11:11474–11489. 2019. View Article : Google Scholar : PubMed/NCBI

20 

Yu G, Wang LG, Han Y and He QY: ClusterProfiler: An R package for comparing biological themes among gene clusters. Omics. 16:284–287. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Heagerty PJ and Zheng Y: Survival model predictive accuracy and ROC curves. Biometrics. 61:92–105. 2005. View Article : Google Scholar : PubMed/NCBI

22 

Fornes O, Castro-Mondragon JA, Khan A, Khan A, Lee RVD, Zhang X, Richmond PA, Modi BP, Correard S, Gheorghe M, et al: JASPAR 2020: Update of the open-access database of transcription factor binding profiles. Nucleic Acids Res. 48:D87–d92. 2020.PubMed/NCBI

23 

Song J, Deng Z, Su J, Yuan D, Liu J and Zhu J: Patterns of immune infiltration in hnc and their clinical implications: A gene expression-based study. Front Oncol. 9:12852019. View Article : Google Scholar : PubMed/NCBI

24 

Ploeg M, Aben KK and Kiemeney LA: The present and future burden of urinary bladder cancer in the world. World J Urol. 27:289–293. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Babjuk M, Böhle A, Burger M, Capoun O, Cohen D, Compérat EM, Hernández V, Kaasinen E, Palou J, Rouprêt M, et al: EAU Guidelines on non-muscle-invasive urothelial carcinoma of the bladder: Update 2016. Eur Urol. 71:447–461. 2017. View Article : Google Scholar : PubMed/NCBI

26 

Felsenstein KM and Theodorescu D: Precision medicine for urothelial bladder cancer: Update on tumour genomics and immunotherapy. Nat Rev Urol. 15:92–111. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Shu B, Zhang J, Sethuraman V, Cui G, Yi X and Zhong G: Transcriptome analysis of Spodoptera frugiperda Sf9 cells reveals putative apoptosis-related genes and a preliminary apoptosis mechanism induced by azadirachtin. Sci Rep. 7:132312017. View Article : Google Scholar : PubMed/NCBI

28 

Rui X, Shao S, Wang L and Leng J: Identification of recurrence marker associated with immune infiltration in prostate cancer with radical resection and build prognostic nomogram. BMC Cancer. 19:11792019. View Article : Google Scholar : PubMed/NCBI

29 

Zhang S, Zhang E, Long J, Hu Z, Peng J, Liu L, Tang F, Li L, Ouyang Y and Zeng Z: Immune infiltration in renal cell carcinoma. Cancer Sci. 110:1564–1572. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Rosenberg J and Huang J: CD8(+) T Cells and NK cells: Parallel and complementary soldiers of immunotherapy. Curr Opin Chem Eng. 19:9–20. 2018. View Article : Google Scholar : PubMed/NCBI

31 

He QF, Xu Y, Li J, Huang ZM, Li XH and Wang X: CD8+ T-cell exhaustion in cancer: Mechanisms and new area for cancer immunotherapy. Brief Funct Genomics. 18:99–106. 2019. View Article : Google Scholar : PubMed/NCBI

32 

Mitchell S, Vargas J and Hoffmann A: Signaling via the NFκB system. Wiley Interdiscip Rev Syst Biol Med. 8:227–241. 2016. View Article : Google Scholar : PubMed/NCBI

33 

Imanishi T and Saito T: T Cell co-stimulation and functional modulation by innate signals. Trends Immunol. 41:200–212. 2020. View Article : Google Scholar : PubMed/NCBI

34 

Asprodites N, Zheng L, Geng D, Velasco-Gonzalez C, Sanchez-Perez L and Davila E: Engagement of Toll-like receptor-2 on cytotoxic T-lymphocytes occurs in vivo and augments antitumor activity. Faseb J. 22:3628–3637. 2008. View Article : Google Scholar : PubMed/NCBI

35 

Chi D, Xu W, Tao X, Zhang T and Cui Y: PD-L1 expression in colorectal cancer and its relationship with TLR-4 expression. J Buon. 25:1423–1429. 2020.PubMed/NCBI

36 

Dan L, Liu L, Sun Y, Song J, Yin Q, Zhang G, Qi F, Hu Z, Yang Z, Zhou Z, et al: The phosphatase PAC1 acts as a T cell suppressor and attenuates host antitumor immunity. Nat Immunol. 21:287–297. 2020. View Article : Google Scholar : PubMed/NCBI

37 

Elangovan IM, Vaz M, Tamatam CR, Potteti HR, Reddy NM and Reddy SP: FOSL1 promotes kras-induced lung cancer through amphiregulin and cell survival gene regulation. Am J Respir Cell Mol Biol. 58:625–635. 2018. View Article : Google Scholar : PubMed/NCBI

38 

Luo YZ, He P and Qiu MX: FOSL1 enhances growth and metastasis of human prostate cancer cells through epithelial mesenchymal transition pathway. Eur Rev Med Pharmacol Sci. 22:8609–8615. 2018.PubMed/NCBI

39 

Rodriguez-Aguayo C, Bayraktar E, Ivan C, Aslan B, Mai J, He G, Mangala L, Jiang D, Nagaraja A, Ozpolat B, et al: PTGER3 induces ovary tumorigenesis and confers resistance to cisplatin therapy through up-regulation Ras-MAPK/Erk-ETS1-ELK1/CFTR1 axis. EBioMedicine. 40:290–304. 2019. View Article : Google Scholar : PubMed/NCBI

40 

Coupland KG, Kim WS, Halliday GM, Hallupp M, Dobson-Stone C and Kwok JB: Role of the long non-coding RNA MAPT-AS1 in regulation of microtubule associated protein tau (MAPT) expression in parkinson's disease. PLoS One. 11:e01579242016. View Article : Google Scholar : PubMed/NCBI

41 

Wosnitzer MS, Domingo-Domenech J, Castillo-Martin M, Ritch C, Mansukhani M, Petrylack DP, Benson MC, McKiernan JM and Cordon-Cardo C: Predictive value of microtubule associated proteins tau and stathmin in patients with nonmuscle invasive bladder cancer receiving adjuvant intravesical taxane therapy. J Urol. 186:2094–2100. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Riese DJ 2nd and Cullum RL: Epiregulin: roles in normal physiology and cancer. Semin Cell Dev Biol. 28:49–56. 2014. View Article : Google Scholar : PubMed/NCBI

43 

Cao R, Yuan L, Ma B, Wang G, Qiu W and Tian Y: An EMT-related gene signature for the prognosis of human bladder cancer. J Cell Mol Med. 24:605–617. 2020. View Article : Google Scholar : PubMed/NCBI

44 

Chen Z, Liu G, Hossain A, Danilova IG, Bolkov MA, Liu G, Tuzankina IA and Tan W: A co-expression network for differentially expressed genes in bladder cancer and a risk score model for predicting survival. Hereditas. 156:242019. View Article : Google Scholar : PubMed/NCBI

45 

Liu S, Suo J, Wang C, Sun X, Wang D, He L, Zhang Y and Li W: Prognostic significance of low miR-144 expression in gastric cancer. Cancer Biomark. 20:547–552. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

May-2021
Volume 21 Issue 5

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
Deng Y, Hong X, Yu C, Li H, Wang Q, Zhang Y, Wang T and Wang X: Preclinical analysis of novel prognostic transcription factors and immune‑related gene signatures for bladder cancer via TCGA‑based bioinformatic analysis. Oncol Lett 21: 344, 2021
APA
Deng, Y., Hong, X., Yu, C., Li, H., Wang, Q., Zhang, Y. ... Wang, X. (2021). Preclinical analysis of novel prognostic transcription factors and immune‑related gene signatures for bladder cancer via TCGA‑based bioinformatic analysis. Oncology Letters, 21, 344. https://doi.org/10.3892/ol.2021.12605
MLA
Deng, Y., Hong, X., Yu, C., Li, H., Wang, Q., Zhang, Y., Wang, T., Wang, X."Preclinical analysis of novel prognostic transcription factors and immune‑related gene signatures for bladder cancer via TCGA‑based bioinformatic analysis". Oncology Letters 21.5 (2021): 344.
Chicago
Deng, Y., Hong, X., Yu, C., Li, H., Wang, Q., Zhang, Y., Wang, T., Wang, X."Preclinical analysis of novel prognostic transcription factors and immune‑related gene signatures for bladder cancer via TCGA‑based bioinformatic analysis". Oncology Letters 21, no. 5 (2021): 344. https://doi.org/10.3892/ol.2021.12605