Abstract
Objectives
The influence of N7-methylguanosine (m7G) on cancer prognosis and immune response has been well-reported. However, the role of m7G-related long non-coding RNAs (lncRNAs) in bladder cancer (BC) remains largely unexplored. This study wanted to explore the relationship between m7G-related lncRNAs and BC.
Methods
To construct the m7G-related lncRNA signature, we utilized data obtained from TCGA. The collected data was then analyzed using R (version 4.2.1, Bell Laboratories, Boston, USA) and relevant packages.
Results
The m7G-related lncRNA signature consisted of seven lncRNAs (including GATA3-AS1, LINC00930, LINC01341, MED14OS, MIR100HG, RUSC1-AS1, SNHG4). The prognostic and clinical relevance of the risk score was corroborated in both the TCGA and IMvigor210 datasets. Individuals characterized by a high-risk score displayed substantial enrichment in pathways associated with immunity, notably those pertaining to the innate immune response, cytokine-mediated signaling pathways, and the adaptive immune system. Additionally, the high-risk score group showed a positive correlation with many immune checkpoints, including CD274, CD40, CTLA4, PDCD1, PDCD1LG2, among others. Moreover, a significant difference in the TCIA score was observed between the high-risk and low-risk score groups, indicating the potential distinct immunotherapy response rates. Furthermore, patients with a high-risk score demonstrated increased sensitivity to cisplatin, docetaxel, doxorubicin, gemcitabine, and vinblastine.
Conclusions
This m7G-related lncRNA signature demonstrates considerable promise as a prognostic biomarker in BC, facilitating the anticipation of responses to both immunotherapy and chemotherapy. This study provides a solid foundation for future investigations into the role of m7G-related lncRNAs in BC.
Introduction
Urothelial carcinoma is most commonly localized in bladder, both in China and on a global scale [1, 2]. Bladder cancer (BC) manifests with age-specific and gender-specific incidence disparities; in incidence, with notable gender differences as well [3]. Despite lower incidence rates among females, their prognostic outcomes are less favorable compared to males [3, 4]. For individuals diagnosed with muscle-invasive BC, the standard therapeutic intervention is radical cystectomy (RC). This procedure imposes considerable economic, social, and psychological burdens. Importantly, suboptimal five years survival rates persist even post-RC [5]. Given the range of existing therapeutic options, BC remains susceptible to recurrence, progression, and increased mortality rates, emphasizing the need for innovative therapeutic strategies [4]. In light of these clinical imperatives, efforts are actively searching for potent underway to identify effective pharmaceutical agents [6]. Concurrent research endeavors utilize various investigative strategies, including but not limited to single-cell RNA sequencing, artificial intelligence, and analyses of clinical and lifestyle parameters, lifestyle determinants, and beyond, to unravel the fundamental underlying mechanisms governing responsible for BC initiation and progression [6, 7]. This multidisciplinary approach has contributed to the development of an array of treatment modalities, including surgery, chemotherapy, immunotherapy, and radiotherapy. Of these, emerging immunotherapies represent a significant advancement. To optimize the utility of these treatments, there is an urgent requirement for the identification and validation of reliable prognostic biomarkers [8]. Such biomarkers would enable clinicians to formulate personalized treatment plans, thereby facilitating informed decision-making regarding aggressive interventions, such as RC.
RNA methylations play a critical role in the regulation of posttranscriptional gene expression [9]. The dysregulation of such methylations is notably implicated in disease onset and prognostic outcomes. Specifically, RNA methylations are integral to both the initiation and progression of various cancers [9]. While recent investigations have considerably enriched our comprehension of the link between mRNA N6-methyladenosine (m6A) and cancer, a significant knowledge gap persists concerning N7-methylguanosine (m7G) and cancer, particularly in long non-coding RNAs (lncRNAs) [10]. M7G is a key catalytic agent for methylation at the N7 position of ribo-guanosine, thereby augmenting the efficacy of mRNA translation efficiency [11, 12]. The role of m7G in cancer pathogenesis has elicited substantial scientific scrutiny, as numerous studies underscore its salient role in carcinoma development [13]. In the realm of prostate cancer, for example, the m7G-associated gene eIF4E has been identified to elevate the translation rates of oncogenic mRNAs, hence fostering tumorigenicity [14]. In the specific context of BC, our prior work introduced an m7G-related mRNA molecular subtype with predictive capacity for patient prognosis and therapeutic response [15]. Furthermore, METTL1, another m7G-associated gene, has been demonstrated to be overexpressed in tumor samples, thereby significantly accelerating BC progression [16]. However, the role of m7G-related lncRNAs in BC has been comparatively underexplored. LncRNAs critically modulate gene expression through a variety of mechanisms, including chromatin remodeling, transcriptional and post-transcriptional regulation, and protein metabolism [17, 18]. Preliminary studies have begun to examine the correlation between m7G and lncRNAs in other malignancies. For instance, in colon cancer, a signature based on m7G-related lncRNAs has been formulated to assess patient prognosis effectively [19]. Similar signatures have been constructed in endometrial cancer for predicting drug sensitivity, and in renal cancer for anticipating immunotherapeutic responses [20, 21].
Despite these advancements, a comprehensive understanding of the interplay between BC and m7G-associated lncRNAs remains to be achieved. Consequently, our research aim is to delineate the influence of m7G-related lncRNAs on BC through the establishment of a bioinformatically derived signature.
Materials and methods
Data acquisition and clear
In alignment with previously established protocols, data were acquired based on predefined inclusion and exclusion criteria [15]. The m7G-related genes were selected through a comprehensive literature review [22] and vetted against the Gene Set Enrichment Analysis (GSEA) database (http://www.gsea-msigdb.org/gsea/index.jsp). This selection was further validated by two independent investigations [23, 24]. A total of 414 BC samples and 18 normal samples were analyzed using the ‘limma’ package to identify differentially expressed genes. The criteria for differential expression incorporated a p-value of less than 0.05 and an absolute |log2FoldChange| exceeding 1. Validation was executed using the IMvigor210 cohort, which consists of BC patients undergoing treatment with atezolizumab [25].
Constructing the signature
Pearson correlation analysis was employed to identify m7G-related lncRNAs, applying a criterion of |coefficients| greater than 0.4 and p-value less than 0.05. Following this initial selection, co-expressed lncRNAs were ascertained through Venn Diagram analysis. This involved differentially expressed lncRNAs from TCGA, IMvigor210 lncRNAs, and m7G-related lncRNAs. A lasso regression model was used to mitigate the risk of overfitting, incorporating all differentially co-expressed m7G-related lncRNAs. Based on these analytical procedures, a risk score involving seven lncRNAs was formulated with the following equation: signature=(GATA3-AS1*(−0.0832))+(LINC00930*(−0.157))+ (LINC01341*(−0.295))+(MED14OS*(−0.282))+(MIR100HG*0.484)+(RUSC1- AS1*(−0.187))+(SNHG4*0.099).
Validating the risk score
The prognostic utility of the established risk score was assessed through Kaplan–Meier analysis for both overall survival (OS) and cancer-specific survival (CSS) in internal and external cohorts, as well as their subgroups. Statistical comparisons of risk score distributions in different clinical contexts were conducted via one-way ANOVA or the Mann–Whitney U test, contingent upon data normality and variance quality.
The risk score’s prognostic significance was further scrutinized using univariable and multivariable Cox regression analyses in both the TCGA and IMvigor210 datasets. Subsequently, two nomograms were rigorously constructed: one based on the TCGA dataset incorporating risk score and clinically relevant parameters as determined by the multivariable Cox regression model, and another using the IMvigor210 dataset that included risk score, sex, and American Joint Committee on Cancer (AJCC) stage. The validity of these nomograms was assessed through multiple evaluative metrics, comprising the concordance index (C-index), multiparameter receiver operating characteristic (ROC) analysis, decision curve analysis (DCA), and calibration curves.
Functional enrichment analysis and immune-related explorations
Gene Ontology (GO) enrichment analysis and enriched Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were performed to elucidate the functional roles associated with the established risk score [26], [27], [28]. Inclusion criteria for the selection of GO and KEGG terms encompassed significance thresholds of p<0.05 and Q<0.05. Complementary to this, Gene Set Enrichment Analysis (GSEA) [29], [30], [31] was employed to rigorously select relevant REACTOME and KEGG pathways, adhering to significance criteria of p<0.05 and FDR<25 %.
To assess the impact of the risk score on immune checkpoint activity, expression levels of 47 checkpoints were compared using data derived from TCGA [32]. Subsequent analyses involved quantification of the proportions of 22 unique immune cell types present in TCGA bladder cancer samples, which were compared between groups using the CIBERSORTx website (https://cibersortx.stanford.edu) [33].
Immunotherapy and chemotherapy
To corroborate the initial findings, data were obtained from The Cancer Immunome Database (TCIA) website (https://tcia.at/home), consistent with prior methodology [34, 35]. Comparative analyses of TCIA scores were conducted between high and low-risk score cohorts. For chemotherapy response and drug sensitivity assessment, the primary endpoint was the half-maximal inhibitory concentration (IC50), calculated using the “pRRophetic” package in R (version 4.2.1, Bell Laboratories, Boston, USA). Subsequently, a comparative evaluation of IC50 values between groups was executed.
Statistical analysis
Data were analyzed using either one-way ANOVA or Mann–Whitney U tests, contingent on the properties of the variances, specifically for comparisons involving three or more continuous variables. For pairwise comparisons involving quantitative data, a Student’s t-test was utilized. All results are reported as mean±standard deviation (SD). A significance threshold of p<0.05 was deemed statistically relevant for all conducted analyses. Analyses were executed using R software (version 4.2.1, Bell Laboratories, Boston, USA) along with the appropriate packages.
Results
Constructing the risk score
The study workflow is outlined in Figure 1, and Supplementary Table 1 lists the 42 m7G-associated genes under investigation. Pearson correlation analysis yielded 271 m7G-related lncRNAs, as illustrated in Figure 2A. A Venn Diagram was subsequently utilized to filter 20 co-expressed, differentially regulated m7G-related lncRNAs. A lasso regression model was then applied to this subset, selecting seven lncRNAs (GATA3-AS1, LINC00930, LINC01341, MED14OS, MIR100HG, RUSC1-AS1, SNHG4) for the formulation of the m7G-related risk score, as detailed in Figure 2B. The prognostic value of these selected lncRNAs was evaluated via univariable Cox regression analysis. Figure 2C substantiates that GATA3-AS1, LINC00930, LINC01341, MED14OS, and MIR100HG serve as effective prognostic indicators of OS and BC patients within the TCGA dataset. Patients in both TCGA and IMvigor210 cohorts were stratified based on median risk score values into high and low-risk groups. Elevated risk scores were significantly correlated with poorer OS (Figure 2E, p<0.001) and CSS (Figure 2E, p<0.001) in the TCGA cohort. This trend was corroborated in the external IMvigor210 cohort, where a high-risk score was indicative of a worse prognosis (Figure 2F, p=0.022).

The workflow of this study.

Construction of the m7G-related risk score: the co-expressed differentially lncRNAs (A), the result of the LASSO regression model (B), the result of the univariable Cox regression model in TCGA (C). Validation of the risk score: the Kaplan–Meier method results of overall survival (D) and cancer-specific survival (E) in TCGA, and the Kaplan–Meier method result of overall survival in IMvigor210 (F). The correlation between the risk score and clinical parameters: TCGA: WHO grade (G), T stage (H), lymph node metastasis (I), age (J), sex (K); IMvigor210: sex (L).
Basic clinical parameters for both TCGA and IMvigor210 cohorts are tabulated in Table 1 and Supplementary Table 2, respectively. Within the TCGA dataset, Table 1 demonstrates a statistically significant relationship between the risk score and multiple clinical variables, including WHO grade, AJCC stage, T stage, lymph node metastasis, OS, and CSS. In the IMvigor210 cohort, the risk score was found to significantly discriminate differences in OS.
The clinicopathologic characteristics of the TCGA included patients.
Characteristic | High risk-score | Low risk-score | p |
---|---|---|---|
n | 201 | 197 | |
Age, mean±SD | 68.44±9.92 | 67.16±11.08 | 0.227 |
Follow-up time (median, month) | 48.67 | 27.73 | <0.001 |
Survival time (median, month) | 22.83 | 88.03 | <0.001 |
BMI, mean±SD | 27.32±5.71 | 26.61±5.61 | 0.244 |
|
|||
Gender, n, % | 0.914 | ||
|
|||
Female | 54 (13.6 %) | 51 (12.8 %) | |
Male | 147 (36.9 %) | 146 (36.7 %) | |
|
|||
Smoking history, n, % | 0.333 | ||
|
|||
No | 49 (12.7 %) | 56 (14.5 %) | |
Yes | 148 (38.4 %) | 132 (34.3 %) | |
|
|||
Lymphatic vessel invasion, n, % | 0.490 | ||
|
|||
No | 60 (22.2 %) | 63 (23.3 %) | |
YES | 79 (29.3 %) | 68 (25.2 %) | |
|
|||
WHO grade, n, % | <0.001 | ||
|
|||
High grade | 199 (50.4 %) | 178 (45.1 %) | |
Low grade | 1 (0.3 %) | 17 (4.3 %) | |
|
|||
AJCC stage, n, % | <0.001 | ||
|
|||
AJCC stage I–II | 38 (9.6 %) | 88 (22.2 %) | |
AJCC stage III–IV | 163 (41.2 %) | 107 (27 %) | |
|
|||
T stage, n, % | <0.001 | ||
|
|||
T stage 3_4 | 154 (42 %) | 95 (25.9 %) | |
T stage a_2 | 39 (10.6 %) | 79 (21.5 %) | |
|
|||
Lymph node metastasis, n, % | 0.015 | ||
|
|||
YES | 78 (21.8 %) | 49 (13.7 %) | |
NO | 109 (30.5 %) | 121 (33.9 %) | |
|
|||
Distant metastasis, n, % | 0.747 | ||
|
|||
YES | 5 (2.5 %) | 5 (2.5 %) | |
NO | 81 (40.1 %) | 111 (55 %) | |
|
|||
Overall survival, n, % | <0.001 | ||
|
|||
Alive | 88 (22.1 %) | 135 (33.9 %) | |
Dead | 113 (28.4 %) | 62 (15.6 %) | |
|
|||
Cancer-specific survival, n, % | <0.001 | ||
|
|||
Alive | 111 (28.9 %) | 153 (39.8 %) | |
Dead | 79 (20.6 %) | 41 (10.7 %) |
-
AJCC, American Joint Committe on Cancer; BMI, body mass index; SD, standard deviation; WHO, World Health Organization; n, number.
Validating the risk score
In the analysis of the TCGA dataset, we identified a statistically significant positive correlation between the risk score and pivotal clinical parameters, specifically WHO grade (Figure 2G, p=2.4e-06), T stage (Figure 2H, p=7.8e-07), and lymph node metastasis (Figure 2I, p=2.1e-04). Conversely, the data revealed no significant associations between the risk score and age (Figure 2J, p=0.15) or sex (Figure 2K, p=0.58). These observations were corroborated by the IMvigor210 dataset, which also exhibited an absence of significant correlation between the risk score and sex (Figure 2L, p=0.64).
To substantiate the prognostic utility of the risk score, Kaplan–Meier survival analyses were executed across multiple subgroups within the TCGA and IMvigor210 datasets. In the TCGA dataset, it was unequivocally observed that patients with elevated risk scores manifested significantly inferior OS in various demographic and clinical subgroups: females (Figure 3A, p=0.035), smokers (Figure 3B, p<0.001), individuals aged 70 years or below (Figure 3C, p=0.01), males (Figure 3D, p<0.001), patients with WHO high-grade tumors (Figure 3E, p<0.001), those devoid of lymphatic vessel invasion (Figure 3F, p<0.001), individuals at Ta_2 stage (Figure 3G, p=0.021), patients without lymph node metastasis (Figure 3H, p=0.002), those lacking distant metastasis (Figure 3I, p<0.001), those classified under AJCC stage III–IV (Figure 3J, p=0.026), and individuals with a body mass index of 25 or below (Figure 3K, p<0.001). Concomitant analysis of the IMvigor210 cohort corroborated the significance of the risk score, revealing a positive correlation with smoking status (Figure 3L, p=0.002), AJCC stage I–II (Figure 3M, p=0.015), and male gender (Figure 3N, p=0.032).

The prognostic ability of the risk score: Kaplan–Meier analysis results of subgroups in TCGA: Female (A), smoker (B), age ≤70 (C), male (D), WHO high grade (E), no lymphatic vessel invasion (F), T a_2 (G), no lymph node metastasis (H), no distant metastasis (I), AJCC stage III–IV (J), body mass index ≤25 (K); IMvigor210: smoker (L), AJCC stage I–II (M), male (N).
Constructing and validating a nomogram
In the TCGA dataset, univariable Cox regression analysis results (Figure 4A) guided the development of a multivariable Cox regression model incorporating both the risk score and pertinent clinical variables such as age, smoking history, AJCC stage, T stage, lymph node metastasis stage, and distant metastasis stage. The multivariable analysis underscored the independent prognostic utility of the risk score in OS (Figure 4B, p<0.001). Similarly, in the IMvigor210 cohort, the risk score demonstrated significant prognostic implications in both univariable (Figure 4C, p=0.008) and multivariable (Figure 4D, p=0.015) Cox regression models.

Construction and validation of a nomogram: the results of univariable (A) and multivariable (B) Cox regression model in TCGA, the results of univariable (C) and multivariable (D) Cox regression model in IMvigor210, the TCGA nomogram (E), the results of multiparameter ROC analysis without (F) and with (G) the risk score in TCGA nomogram, the calibration curves of TCGA nomogram (H), the DCA curve of TCGA nomogram (I).
A nomogram was constructed based on the multivariable Cox regression model from the TCGA dataset (Figure 4E). The C-index was calculated as 0.72 (0.689–0.754). For further nomogram validation, a multiparameter ROC analysis was conducted, revealing that the inclusion of the risk score elevated the area under the curve value from 0.73 (Figure 4F) to 0.8 (Figure 4G). Calibration curves for 1, 3, and 5 years exhibited strong alignment between predicted and observed outcomes (Figure 4H). Additionally, decision curve analysis (DCA) confirmed the favorable performance of the risk score (Figure 4I). Supplementary Figure 1 provides the IMvigor210 nomogram and corresponding validation data.
Enrichment analyses for the risk score
Utilizing TCGA data, we conducted GO, KEGG, and GSEA to elucidate functional insights. GO analysis indicated significant enrichment in biological processes (BP) encompassing neutrophil-mediated immunity, granulocyte chemotaxis, and neutrophil chemotaxis, as well as in cellular components including the external side of the plasma membrane and the cornified envelope. Molecular function (MF) was enriched in categories such as chemokine activity, immunoglobulin binding, and IgG binding (Figure 5A). Similarly, KEGG analysis highlighted immune-related pathways, including cytokine–cytokine receptor interaction, the PI3K-Akt signaling pathway, the B cell receptor signaling pathway, the chemokine signaling pathway, and the IL-17 signaling pathway (Figure 5B). To enhance the credibility of our GSEA results, we enriched pathways from both REACTOME and KEGG databases. As depicted in Figure 5C, increased REACTOME pathways were primarily related to immune processes, cytokine signaling in the immune system, adaptive immune system, signaling by interleukins, and neutrophil degranulation pathways. In contrast, downregulated REACTOME pathways were primarily involved in metabolic processes, encompassing cytochrome P450 – organized by substrate type, fatty acid metabolism, regulation of lipid metabolism via PPAR alpha, and lipid metabolism (Figure 5D). Similarly, enhanced KEGG pathways were predominantly immune-related (Figure 5E), while downregulated KEGG pathways were primarily associated with metabolism, notably drug metabolism (Figure 5F).

Function analysis: Gene ontology results (A), Kyoto Encyclopedia of Genes and Genomes results (B), Gene Set Enrichment Analysis results in REACTOME (C and D) and KEGG (E and F).
The risk score in immune checkpoints and immune infiltration analyses
Regarding immune checkpoints, samples with higher risk scores exhibited positive correlations with the expression levels of CD274, CD40, CTLA4, PDCD1, and PDCD1LG2, among others. Inversely, the risk score maintained a positive association with the expression of CD96, SIGLEC15, TNFRSF14, TNFRSF25, and TNFSF15 (Figure 6A). Regarding immune cell infiltration, high-risk score samples exhibited positive correlations with CD4+ T memory activated cells, T gamma delta cells, Macrophage M0 cells, Macrophage M1 cells, Macrophage M2 cells, and neutrophil cells. Conversely, these high-risk samples displayed negative associations with B plasma cells, NK resting cells, and monocytes (Figure 6B).

Immune checkpoints analysis results (A), immune infiltration (B). The TCIA score result (C). Chemotherapy response (D). IC50: The half-maximal inhibitory concentration; ns, p≥0.05; *, p<0.05; **, p<0.01; ***, p<0.001.
The correlation between the risk score and immunotherapy and chemotherapy
The TCIA outcomes indicated that individuals with a high-risk score displayed elevated TCIA scores primarily in the CTLA4_positive_PD1_positive subgroup (Figure 6C). The risk score displayed a positive correlation with CTLA4_positive_PD1_positive phenotypes, whereas it showed a negative association with both CTLA4_positive_PD1_negative and CTLA4_negative_PD1_negative phenotypes.
In the context of chemotherapy response, patients with high-risk scores displayed sensitivity to agents such as cisplatin, docetaxel, doxorubicin, gemcitabine, and vinblastine, with statistical significance observed in all cases. Conversely, patients with low-risk scores exhibited sensitivity to methotrexate (Figure 6D, all p<0.01).
Discussion
The m7G cap serves as an indispensable element for post-transcriptional modifications and is a key factor for efficient RNA processing [17]. Numerous m7G-associated lncRNAs have been implicated as prognostic indicators across a range of malignancies [36]. Moreover, m7G-related mRNA has proven to be of prognostic relevance in breast cancer patients [20]. Building on this foundation, the current study endeavors to delineate the relationship between m7G-related lncRNAs and BC. We have formulated a BC-specific signature, and our empirical data corroborate the prognostic utility of this m7G-related lncRNA signature in BC cohorts, a validation that extends to both internal and external samples. Additionally, we discerned a meaningful association between this signature and the likely effectiveness of both immunotherapeutic and chemotherapeutic interventions, thereby endorsing the prospective utility of these lncRNAs and the signature as valuable biomarkers in BC.
GATA3-AS1 is observed to be markedly upregulated in breast cancer and is implicated in both tumor progression and mechanisms of immune evasion [37]. It also has a contributory role in the carcinogenesis of endometrial carcinoma [38]. LINC00930 is upregulated and exhibits a relationship with tumorigenesis in nasopharyngeal carcinoma [39]. LINC01341 has been characterized as a 5-methylcytosine-associated lncRNA in lung squamous cell carcinoma, thereby offering insights into its plausible involvement in RNA methylation processes [40]. MIR100HG is elevated in BC and plays a role in disease progression [41], while concurrently influencing the formation of cancer stem cells and affecting the distant metastasis of lung cancer [42]. RUSC1-AS1 accelerates the pathogenesis of both osteosarcoma [43] and breast cancer [44, 45]. Finally, the expression of SNHG4 has been found to impact multiple cancer types [46], [47], [48], [49].
A formidable obstacle in harnessing lncRNA-related signatures for BC research lies in the paucity of external validation, largely due to limited lncRNA expression data in publicly available datasets. To address this, our study employed external validation using the IMvigor210 dataset, which incorporates lncRNA expression metrics, thereby fortifying the credibility of our conclusions. We found that the risk score not only demonstrated statistically significant variations across an array of clinical parameters but also sustained positive correlations with multiple clinical variables. Notably, no significant associations were discerned between the risk score and either age or smoking history across both the TCGA and IMvigor210 datasets. Within the TCGA dataset, the risk score was relatively consistent between female and male cohorts. Therefore, the applicability of the risk score appears to be expansive, unhindered by variables such as age, smoking history, or gender. In prognostic assessments, the risk score independently evinced substantial prognostic merit for BC patients in the TCGA dataset, a finding corroborated in the IMvigor210 cohort. Age is a salient risk factor for BC [50] and influences the effectiveness of BCG treatment, thereby informing therapeutic decisions [51, 52]. Fortunately, the risk score exhibited no significant differences between patients aged 70 years or less and those aged 70 years or older, implying that the risk score can be applied without age-related restrictions. Importantly, we observed no marked differences in the risk score between patients aged 70 years or younger and those older than 70 years, suggesting the score’s applicability across age groups. Furthermore, the risk score demonstrated prognostic significance across heterogeneous subgroups in both the TCGA and IMvigor210 datasets. This corroborates earlier studies highlighting the prognostic utility of m7G-related lncRNAs in a variety of malignancies, including lung adenocarcinoma [53], colon carcinoma [54], hepatocellular carcinoma [36], and endometrial cancer [20]. In urological malignant tumors, the m7G-related lncRNA risk score has also independently prognosticated patient outcomes in renal cancer [21]. Based on these findings, we postulate that our risk score could function as a viable biomarker for BC.
GO analysis revealed enrichment in neutrophil-mediated immunity, neutrophil chemotaxis, and chemokine activity. Concurrently, KEGG and GSEA identified enriched pathways pertinent to immune functions and metabolic processes, including cytokine–cytokine receptor interaction, B cell receptor signaling pathway, adaptive immune system, and neutrophil degranulation, among others. In the ensuing checkpoint analysis, a positive correlation was observed between the risk score and the expression levels of key immune markers such as CD274, CD40, CTLA4, PDCD1, and PDCD1LG2. In contrast, samples characterized by low-risk scores demonstrated a positive correlation with the expression levels of CD96, SIGLEC15, TNFRSF14, TNFRSF25, and TNFSF15. The current literature presents no consensus on the relationship between PD-L1 expression and response to immunotherapy [55]. However, findings from the phase 3 IMvigor130 trial indicated that patients with elevated PD-L1 expression experienced favorable outcomes with atezolizumab (anti-PD1/PD-L1) monotherapy [56]. These data corroborate the notion that higher risk scores may predict increased efficacy from anti-PD1/PD-L1 immunotherapy. M6A RNA modification has been implicated as a pivotal factor influencing the tumor immune microenvironment in BC [57, 58]. Our analysis of immune cell infiltration in TCGA samples, approached from the perspective of m7G modifications, revealed a positive association between risk scores and increased infiltration of CD4+ T memory activated cells, M2 macrophages, and neutrophils. Elevated infiltration of these specific immune cell types generally indicates a higher likelihood of positive response to immunotherapy [58], [59], [60]. In alignment with our prognostic outcomes, the presence of Macrophage M0 cells was associated with an unfavorable prognosis [61]. This result lends further credence to the hypothesis that individuals with higher risk scores are more likely to derive pronounced benefits from anti-PD1/PD-L1 immunotherapy. Finally, we employed data from the Tumor Cancer Immunome Atlas (TCIA) to project immunotherapy responses. The analysis further substantiated that high-risk score patients are more likely to demonstrate favorable responses to anti-PD1 immunotherapy.
The empirical data suggest that patients categorized with higher risk scores may exhibit an enhanced responsiveness to anti-PD1 immunotherapy. This supposition is congruent with the upregulated expression of PD-1 and PD-L1 observed in this high-risk cohort. Conjoint analyses of gene expression patterns, immune cell infiltration metrics, and data from the TCIA collectively fortify the proposition that patients with elevated risk scores are more likely to benefit from anti-PD1/PD-L1 immunotherapeutic interventions as opposed to those with lower scores. Moreover, extant research lends credence to the idea that patients with lower risk scores could potentially derive therapeutic advantages from anti-CTLA4 immunotherapy. Investigations led by Wang et al. demonstrate that the lncRNA GATA3-AS1 modulates immune evasion mechanisms in breast cancer via the regulation of GATA3 and PD-L1 expression [62]. Similarly, aberrant expression of MIR100HG has been implicated in immune escape phenomena in the context of gastric cancer [63]. The regulatory role of SNHG4 in immune evasion in colorectal cancer is enacted through its modulation of the miR-144-3p/MET signaling axis [64]. These molecular mechanisms serve to elucidate the utility of the risk score as a predictive metric for immunotherapy responsiveness. In pharmacological responsiveness, patients with elevated risk scores displayed heightened sensitivity to a panel of chemotherapeutic agents, including cisplatin, docetaxel, doxorubicin, gemcitabine, and vinblastine. Conversely, methotrexate was identified as a chemotherapeutic agent to which patients with lower risk scores exhibited sensitivity. Notably, GATA3-AS1 has been ascertained to possess predictive value in gauging the response of breast cancer to neoadjuvant chemotherapy [37]. MIR100HG, which is derived from cancer stem cells, has been substantially implicated in chemotherapeutic resistance mechanisms [42, 65].
Although the expression and prognostic value of the risk score were exhibited in TCGA and external cohorts. We still need to demonstrate these findings using basic experiments in our future work.
Conclusions
Our results validated the risk score had significant clinical value in BC patients. Additionally, a notable correlation was observed between the risk score and responses to treatments. The included m7G-related lncRNAs and the signature show promising potential for managing BC.
Acknowledgments
We appreciated the Figdraw (www.figdraw.com) and Chengdu Basebiotech Co, Ltd for their assistance in drawing and data process.
-
Research ethics: This study does not involve human participants and animals. Therefore, it does not require ethical review and approval.
-
Informed consent: All data of this study was extracted from online database. Thus, it does not need an informed consent.
-
Author contributions: The authors confirm their contribution to the paper as follows: study conception and design: Deng-xiong Li and Rui-cheng Wu; data collection: Wang Jie and Deng-xiong Li; analysis and interpretation of results: Deng-xiong Li, Rui-cheng Wu and De-chao Feng; draft manuscript preparation: Deng-xiong Li and Shi Deng. All authors reviewed the results and approved the final version of the manuscript.
-
Competing interests: The authors declare that they have no conflicts of interest to report regarding the present study.
-
Research funding: None.
-
Data availability: All data of this study was extracted from online database. Therefore, everyone can get the data online. Further inquiries can be directed to the corresponding author.
References
1. Sung, H, Ferlay, J, Siegel, RL, Laversanne, M, Soerjomataram, I, Jemal, A, et al.. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 2021;71:209–49. https://doi.org/10.3322/caac.21660.Search in Google Scholar PubMed
2. Zi, H, He, SH, Leng, XY, Xu, XF, Huang, Q, Weng, H, et al.. Global, regional, and national burden of kidney, bladder, and prostate cancers and their attributable risk factors, 1990–2019. Mil Med Res 2021;8:60. https://doi.org/10.1186/s40779-021-00354-z.Search in Google Scholar PubMed PubMed Central
3. Theodorescu, D, Li, Z, Li, X. Sex differences in bladder cancer: emerging data and call to action. Nat Rev Urol 2022;19:447–9. https://doi.org/10.1038/s41585-022-00591-4.Search in Google Scholar PubMed PubMed Central
4. Shih, KW, Chen, WC, Chang, CH, Tai, TE, Wu, JC, Huang, AC, et al.. Non-muscular invasive bladder cancer: re-envisioning therapeutic journey from traditional to regenerative interventions. Aging Dis 2021;12:868–85. https://doi.org/10.14336/ad.2020.1109.Search in Google Scholar PubMed PubMed Central
5. EAU Guidelines. edn. Presented at the EAU Annual Congress Milan 2023. Available from: EAU Guidelines Citing, Usage & Republication - Uroweb.Search in Google Scholar
6. Nishal, S, Jhawat, V, Gupta, S, Phaugat, P. Utilization of kinase inhibitors as novel therapeutic drug targets: a review. Oncol Res 2022;30:221–30. https://doi.org/10.32604/or.2022.027549.Search in Google Scholar PubMed PubMed Central
7. Li, DX, Yu, QX, Feng, DC, Zhang, FC, Wu, RC, Xu, S, et al.. Systemic immune-inflammation index (SII) during induction has higher predictive value than preoperative SII in non-muscle-invasive bladder cancer patients receiving intravesical Bacillus Calmette–Guerin. Clin Genitourin Cancer 2023;21:e145–e52. https://doi.org/10.1016/j.clgc.2022.11.013.Search in Google Scholar PubMed
8. Dai, B, Xu, Z, Li, H, Wang, B, Cai, J, Liu, X. Racial bias can confuse AI for genomic studies. Oncologie 2022;24:113–30. https://doi.org/10.32604/oncologie.2022.020259.Search in Google Scholar
9. Tang, Q, Li, L, Wang, Y, Wu, P, Hou, X, Ouyang, J, et al.. RNA modifications in cancer. Br J Cancer 2023;129:204–21. https://doi.org/10.1038/s41416-023-02275-1.Search in Google Scholar PubMed PubMed Central
10. Zeng, L, Huang, X, Zhang, J, Lin, D, Zheng, J. Roles and implications of mRNA N(6) – methyladenosine in cancer. Cancer Commun 2023;43:729–48. https://doi.org/10.1002/cac2.12458.Search in Google Scholar PubMed PubMed Central
11. Cowling, VH. Regulation of mRNA cap methylation. Biochem J 2009;425:295–302. https://doi.org/10.1042/bj20091352.Search in Google Scholar
12. Fresco, LD, Buratowski, S. Conditional mutants of the yeast mRNA capping enzyme show that the cap enhances, but is not required for, mRNA splicing. RNA 1996;2:584–96.Search in Google Scholar
13. Cheng, W, Gao, A, Lin, H, Zhang, W. Novel roles of METTL1/WDR4 in tumor via m(7)G methylation. Mol Ther Oncolytics 2022;26:27–34. https://doi.org/10.1016/j.omto.2022.05.009.Search in Google Scholar PubMed PubMed Central
14. D’Abronzo, LS, Ghosh, PM. eIF4E phosphorylation in prostate cancer. Neoplasia 2018;20:563–73. https://doi.org/10.1016/j.neo.2018.04.003.Search in Google Scholar PubMed PubMed Central
15. Li, DX, Feng, DC, Wang, XM, Wu, RC, Zhu, WZ, Chen, K, et al.. M7G-related molecular subtypes can predict the prognosis and correlate with immunotherapy and chemotherapy responses in bladder cancer patients. Eur J Med Res 2023;28:55. https://doi.org/10.1186/s40001-023-01012-x.Search in Google Scholar PubMed PubMed Central
16. Ying, X, Liu, B, Yuan, Z, Huang, Y, Chen, C, Jiang, X, et al.. METTL1-m(7) G-EGFR/EFEMP1 axis promotes the bladder cancer development. Clin Transl Med 2021;11:e675. https://doi.org/10.1002/ctm2.675.Search in Google Scholar PubMed PubMed Central
17. Derrien, T, Johnson, R, Bussotti, G, Tanzer, A, Djebali, S, Tilgner, H, et al.. The GENCODE v7 catalog of human long noncoding RNAs: analysis of their gene structure, evolution, and expression. Genome Res 2012;22:1775–89. https://doi.org/10.1101/gr.132159.111.Search in Google Scholar PubMed PubMed Central
18. Ulitsky, I, Bartel, DP. lincRNAs: genomics, evolution, and mechanisms. Cell 2013;154:26–46. https://doi.org/10.1016/j.cell.2013.06.020.Search in Google Scholar PubMed PubMed Central
19. Liu, L, Wu, Y, Chen, W, Li, Y, Yu, J, Zhang, G, et al.. The m7G-related long noncoding RNA signature predicts prognosis and indicates tumour immune infiltration in colon cancer. Front Genet 2022;13:892589. https://doi.org/10.3389/fgene.2022.892589.Search in Google Scholar PubMed PubMed Central
20. Sun, J, Li, L, Chen, H, Gan, L, Guo, X, Sun, J. Identification and validation of an m7G-related lncRNAs signature for prognostic prediction and immune function analysis in endometrial cancer. Genes 2022;13:1301. https://doi.org/10.3390/genes13081301.Search in Google Scholar PubMed PubMed Central
21. Ming, J, Wang, C. N7-methylguanosine-related lncRNAs: integrated analysis associated with prognosis and progression in clear cell renal cell carcinoma. Front Genet 2022;13:871899. https://doi.org/10.3389/fgene.2022.871899.Search in Google Scholar PubMed PubMed Central
22. Mei, W, Jia, X, Xin, S, Liu, X, Jin, L, Sun, X, et al.. A N(7)-methylguanine-related gene signature applicable for the prognosis and microenvironment of prostate cancer. J Oncol 2022;2022:8604216–31. https://doi.org/10.1155/2022/8604216.Search in Google Scholar PubMed PubMed Central
23. Liberzon, A, Birger, C, Thorvaldsdóttir, H, Ghandi, M, Mesirov, JP, Tamayo, P. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst 2015;1:417–25. https://doi.org/10.1016/j.cels.2015.12.004.Search in Google Scholar PubMed PubMed Central
24. Shaheen, R, Abdel-Salam, GM, Guy, MP, Alomar, R, Abdel-Hamid, MS, Afifi, HH, et al.. Mutation in WDR4 impairs tRNA m(7)G46 methylation and causes a distinct form of microcephalic primordial dwarfism. Genome Biol 2015;16:210. https://doi.org/10.1186/s13059-015-0779-x.Search in Google Scholar PubMed PubMed Central
25. Yuen, KC, Liu, LF, Gupta, V, Madireddi, S, Keerthivasan, S, Li, C, et al.. High systemic and tumor-associated IL-8 correlates with reduced clinical benefit of PD-L1 blockade. Nat Med 2020;26:693–8. https://doi.org/10.1038/s41591-020-0860-1.Search in Google Scholar PubMed PubMed Central
26. Ashburner, M, Ball, CA, Blake, JA, Botstein, D, Butler, H, Cherry, JM, et al.. Gene Ontology: tool for the unification of biology. The Gene Ontology Consortium. Nat Genet 2000;25:25–9. https://doi.org/10.1038/75556.Search in Google Scholar PubMed PubMed Central
27. Aleksander, SA, Balhoff, J, Carbon, S, Cherry, JM, Drabkin, HJ, Ebert, D, et al.. The Gene Ontology knowledgebase in 2023. Genetics 2023;224:iyad031. https://doi.org/10.1093/genetics/iyad031.Search in Google Scholar PubMed PubMed Central
28. Kanehisa, M, Furumichi, M, Sato, Y, Kawashima, M, Ishiguro-Watanabe, M. KEGG for taxonomy-based analysis of pathways and genomes. Nucleic Acids Res 2023;51:D587–d92. https://doi.org/10.1093/nar/gkac963.Search in Google Scholar PubMed PubMed Central
29. Subramanian, A, Tamayo, P, Mootha, VK, Mukherjee, S, Ebert, BL, Gillette, MA, et al.. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA 2005;102:15545–50. https://doi.org/10.1073/pnas.0506580102.Search in Google Scholar PubMed PubMed Central
30. Mootha, VK, Lindgren, CM, Eriksson, KF, Subramanian, A, Sihag, S, Lehar, J, et al.. PGC-1alpha-responsive genes involved in oxidative phosphorylation are coordinately downregulated in human diabetes. Nat Genet 2003;34:267–73. https://doi.org/10.1038/ng1180.Search in Google Scholar PubMed
31. Li, DX, Feng, DC, Shi, X, Wu, RC, Chen, K, Han, P. Identification of endothelial-related molecular subtypes for bladder cancer patients. Front Oncol 2023;13:1101055. https://doi.org/10.3389/fonc.2023.1101055.Search in Google Scholar PubMed PubMed Central
32. Yu, Q, Zhang, F, Feng, D, Li, D, Xia, Y, Gan, MF. An inflammation-related signature could predict the prognosis of patients with kidney renal clear cell carcinoma. Front Genet 2022;13:866696. https://doi.org/10.3389/fgene.2022.866696.Search in Google Scholar PubMed PubMed Central
33. Gentles, AJ, Newman, AM, Liu, CL, Bratman, SV, Feng, W, Kim, D, et al.. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat Med 2015;21:938–45. https://doi.org/10.1038/nm.3909.Search in Google Scholar PubMed PubMed Central
34. Charoentong, P, Finotello, F, Angelova, M, Mayer, C, Efremova, M, Rieder, D, et al.. Pan-cancer immunogenomic analyses reveal Genotype-immunophenotype relationships and predictors of response to checkpoint blockade. Cell Rep 2017;18:248–62. https://doi.org/10.1016/j.celrep.2016.12.019.Search in Google Scholar PubMed
35. Li, DX, Yu, QX, Zeng, CX, Ye, LX, Guo, YQ, Liu, JF, et al.. A novel endothelial-related prognostic index by integrating single-cell and bulk RNA sequencing data for patients with kidney renal clear cell carcinoma. Front Genet 2023;14:1096491. https://doi.org/10.3389/fgene.2023.1096491.Search in Google Scholar PubMed PubMed Central
36. Wei, W, Liu, C, Wang, M, Jiang, W, Wang, C, Zhang, S. Prognostic signature and tumor immune landscape of N7-methylguanosine-related lncRNAs in hepatocellular carcinoma. Front Genet 2022;13:906496. https://doi.org/10.3389/fgene.2022.906496.Search in Google Scholar PubMed PubMed Central
37. Contreras-Espinosa, L, Alcaraz, N, De La Rosa-Velázquez, IA, Díaz-Chávez, J, Cabrera-Galeana, P, Rebollar-Vega, R, et al.. Transcriptome analysis identifies GATA3-AS1 as a long noncoding RNA associated with resistance to neoadjuvant chemotherapy in locally advanced breast cancer patients. J Mol Diagn 2021;23:1306–23. https://doi.org/10.1016/j.jmoldx.2021.07.014.Search in Google Scholar PubMed
38. Liu, YX, Yuan, S, Liu, XJ, Huang, YX, Qiu, P, Gao, J, et al.. LncRNA GATA3-AS1 promoted invasion and migration in human endometrial carcinoma by regulating the miR-361/ARRB2 axis. J Mol Med 2022;100:1271–86. https://doi.org/10.1007/s00109-022-02222-2.Search in Google Scholar PubMed
39. He, B, Pan, H, Zheng, F, Chen, S, Bie, Q, Cao, J, et al.. Long noncoding RNA LINC00930 promotes PFKFB3-mediated tumor glycolysis and cell proliferation in nasopharyngeal carcinoma. J Exp Clin Cancer Res 2022;41:77. https://doi.org/10.1186/s13046-022-02282-9.Search in Google Scholar PubMed PubMed Central
40. Xu, R, Zhang, W. Prognostic value and immune landscapes of m5C-related lncRNAs in lung squamous cell carcinoma. Front Genet 2022;13:960229. https://doi.org/10.3389/fgene.2022.960229.Search in Google Scholar PubMed PubMed Central
41. Zhang, S, Wang, Q, Li, W, Chen, J. MIR100HG regulates CALD1 gene expression by targeting miR-142-5p to affect the progression of bladder cancer cells in vitro, as revealed by transcriptome sequencing. Front Mol Biosci 2021;8:793493. https://doi.org/10.3389/fmolb.2021.793493.Search in Google Scholar PubMed PubMed Central
42. Shi, L, Li, B, Zhang, Y, Chen, Y, Tan, J, Chen, Y, et al.. Exosomal lncRNA Mir100hg derived from cancer stem cells enhance glycolysis and promote metastasis of lung adenocarcinoma through mircroRNA-15a-5p/31-5p. Cell Commun Signal 2023;21:248. https://doi.org/10.1186/s12964-023-01281-3.Search in Google Scholar PubMed PubMed Central
43. Jiang, R, Zhang, Z, Zhong, Z, Zhang, C. Long-non-coding RNA RUSC1-AS1 accelerates osteosarcoma development by miR-101-3p-mediated Notch1 signalling pathway. J Bone Oncol 2021;30:100382. https://doi.org/10.1016/j.jbo.2021.100382.Search in Google Scholar PubMed PubMed Central
44. Hu, CC, Liang, YW, Hu, JL, Liu, LF, Liang, JW, Wang, R. LncRNA RUSC1-AS1 promotes the proliferation of breast cancer cells by epigenetic silence of KLF2 and CDKN1A. Eur Rev Med Pharmacol Sci 2019;23:6602–11. https://doi.org/10.26355/eurrev_201908_18548.Search in Google Scholar PubMed
45. Ayoufu, A, Paierhati, P, Qiao, L, Zhang, N, Abudukeremu, M. RUSC1-AS1 promotes the malignant progression of breast cancer depending on the regulation of the miR-326/XRCC5 pathway. Thoracic Cancer 2023;14:2504–14. https://doi.org/10.1111/1759-7714.15035.Search in Google Scholar PubMed PubMed Central
46. Chu, Q, Gu, X, Zheng, Q, Guo, Z, Shan, D, Wang, J, et al.. Long noncoding RNA SNHG4: a novel target in human diseases. Cancer Cell Int 2021;21:583. https://doi.org/10.1186/s12935-021-02292-1.Search in Google Scholar PubMed PubMed Central
47. Dong, Q, Qiu, H, Piao, C, Li, Z, Cui, X. LncRNA SNHG4 promotes prostate cancer cell survival and resistance to enzalutamide through a let-7a/RREB1 positive feedback loop and a ceRNA network. J Exp Clin Cancer Res CR 2023;42:209. https://doi.org/10.1186/s13046-023-02774-2.Search in Google Scholar PubMed PubMed Central
48. Khajehdehi, M, Khalaj-Kondori, M, Baradaran, B. The siRNA-mediated knockdown of SNHG4 efficiently induced pro-apoptotic signaling and suppressed metastasis in SW1116 colorectal cancer cell line. Mol Biol Rep 2023. https://doi.org/10.1007/s11033-023-08742-5. [Epub ahead of print]Search in Google Scholar PubMed
49. Pourghasem, N, Ghorbanzadeh, S, Nejatizadeh, AA. Expression and regulatory roles of Small nucleolar RNA host gene 4 in gastric cancer. Curr Protein Pept Sci 2023;24:767–79. https://doi.org/10.2174/1389203724666230810094548.Search in Google Scholar PubMed
50. Yang, F, Liu, G, Wei, J, Dong, Y, Zhang, X, Zheng, Y. Relationship between bladder cancer, nutritional supply, and treatment strategies: a comprehensive review. Nutrients 2023;15:3812. https://doi.org/10.3390/nu15173812.Search in Google Scholar PubMed PubMed Central
51. Ram, P, Mandal, S, Das, MK, Nayak, P. Association of increased age with decreased response to intravesical instillation of bacille Calmette–Guerin in patients with high-risk non-muscle invasive bladder cancer: retrospective multi-institute results from the Japanese Urological Oncology Research Group JUOG-UC-1901-BCG. Urology 2023;173:229. https://doi.org/10.1016/j.urology.2022.10.031.Search in Google Scholar PubMed
52. Noel, OD, Stewart, E, Cress, R, Dall’Era, MA, Shrestha, A. Underutilization of intravesical chemotherapy and immunotherapy for high grade non-muscle invasive bladder cancer in California between 2006-2018: effect of race, age and socioeconomic status on treatment disparities. Urol Oncol 2023;41:431.e7–14. https://doi.org/10.1016/j.urolonc.2023.05.019.Search in Google Scholar PubMed
53. Zhang, C, Zhou, D, Wang, Z, Ju, Z, He, J, Zhao, G, et al.. Risk model and immune signature of m7G-related lncRNA based on lung adenocarcinoma. Front Genet 2022;13:907754. https://doi.org/10.3389/fgene.2022.907754.Search in Google Scholar PubMed PubMed Central
54. Yang, S, Zhou, J, Chen, Z, Sun, Q, Zhang, D, Feng, Y, et al.. A novel m7G-related lncRNA risk model for predicting prognosis and evaluating the tumor immune microenvironment in colon carcinoma. Front Oncol 2022;12:934928. https://doi.org/10.3389/fonc.2022.934928.Search in Google Scholar PubMed PubMed Central
55. Labadie, BW, Balar, AV, Luke, JJ. Immune checkpoint inhibitors for Genitourinary cancers: treatment indications, investigational approaches and biomarkers. Cancers 2021;13:5415. https://doi.org/10.3390/cancers13215415.Search in Google Scholar PubMed PubMed Central
56. Powles, T, O’Donnell, H, Massard, C, Arkenau, T, Friedlander, W, Hoimes, C, et al.. Updated efficacy and tolerability of durvalumab in locally advanced or metastatic urothelial carcinoma. J Clin Oncol 2017;35:286. https://doi.org/10.1200/jco.2017.35.6_suppl.286.Search in Google Scholar
57. Deng, H, Tang, F, Zhou, M, Shan, D, Chen, X, Cao, K. Identification and validation of N6-methyladenosine-related biomarkers for bladder cancer: implications for immunotherapy. Front Oncol 2022;12:820242. https://doi.org/10.3389/fonc.2022.820242.Search in Google Scholar PubMed PubMed Central
58. Nguyen, S, Chevalier, MF, Benmerzoug, S, Cesson, V, Schneider, AK, Rodrigues-Dias, SC, et al.. Vδ2 T cells are associated with favorable clinical outcomes in patients with bladder cancer and their tumor reactivity can be boosted by BCG and zoledronate treatments. J Immunother Cancer 2022;10:e004880. https://doi.org/10.1136/jitc-2022-004880.Search in Google Scholar PubMed PubMed Central
59. Yang, W, Han, B, Chen, Y, Geng, F. SAAL1, a novel oncogene, is associated with prognosis and immunotherapy in multiple types of cancer. Aging 2022;14:6316–37. https://doi.org/10.18632/aging.204224.Search in Google Scholar PubMed PubMed Central
60. Wang, A, Bu, Z. Pan-cancer tumor-infiltrating T cells: a great hallmark in cancer immunology research. Chin J Cancer Res 2022;34:115–6. https://doi.org/10.21147/j.issn.1000-9604.2022.02.06.Search in Google Scholar PubMed PubMed Central
61. Wu, S, Yang, W, Zhang, H, Ren, Y, Fang, Z, Yuan, C, et al.. The prognostic landscape of tumor-infiltrating immune cells and immune checkpoints in glioblastoma. Technol Cancer Res Treat 2019;18:1533033819869949. https://doi.org/10.1177/1533033819869949.Search in Google Scholar PubMed PubMed Central
62. Zhang, M, Wang, N, Song, P, Fu, Y, Ren, Y, Li, Z, et al.. LncRNA GATA3-AS1 facilitates tumour progression and immune escape in triple-negative breast cancer through destabilization of GATA3 but stabilization of PD-L1. Cell Prolif 2020;53:e12855. https://doi.org/10.1111/cpr.12855.Search in Google Scholar PubMed PubMed Central
63. Li, P, Ge, D, Li, P, Hu, F, Chu, J, Chen, X, et al.. CXXC finger protein 4 inhibits the CDK18-ERK1/2 axis to suppress the immune escape of gastric cancer cells with involvement of ELK1/MIR100HG pathway. J Cell Mol Med 2020;24:10151–65. https://doi.org/10.1111/jcmm.15625.Search in Google Scholar PubMed PubMed Central
64. Zhou, N, Chen, Y, Yang, L, Xu, T, Wang, F, Chen, L, et al.. LncRNA SNHG4 promotes malignant biological behaviors and immune escape of colorectal cancer cells by regulating the miR-144-3p/MET axis. Am J Transl Res 2021;13:11144–61.10.18632/aging.202737Search in Google Scholar PubMed PubMed Central
65. Zhang, H, Wang, X, Yu, Y, Yang, Z. Progression of Exosome-mediated chemotherapy resistance in cancer. Oncologie 2022;24:247–59. https://doi.org/10.32604/oncologie.2022.020993.Search in Google Scholar
Supplementary Material
This article contains supplementary material (https://doi.org/10.1515/oncologie-2023-0334).
© 2023 the author(s), published by De Gruyter, Berlin/Boston
This work is licensed under the Creative Commons Attribution 4.0 International License.
Articles in the same Issue
- Frontmatter
- Review Articles
- Mitochondrial thermogenesis in cancer cells
- Application of indocyanine green in the management of oral cancer: a literature review
- Long non-coding RNA, FOXP4-AS1, acts as a novel biomarker of cancers
- The role of synthetic peptides derived from bovine lactoferricin against breast cancer cell lines: a mini-review
- Single cell RNA sequencing – a valuable tool for cancer immunotherapy: a mini review
- Research Articles
- Global patterns and temporal trends in ovarian cancer morbidity, mortality, and burden from 1990 to 2019
- The association between NRF2 transcriptional gene dysregulation and IDH mutation in Grade 4 astrocytoma
- More than just a KRAS inhibitor: DCAI abrogates the self-renewal of pancreatic cancer stem cells in vitro
- DUSP1 promotes pancreatic cancer cell proliferation and invasion by upregulating nephronectin expression
- IMMT promotes hepatocellular carcinoma formation via PI3K/AKT/mTOR pathway
- MiR-100-5p transfected MSCs-derived exosomes can suppress NSCLC progression via PI3K-AKT-mTOR
- Inhibitory function of CDK12i combined with WEE1i on castration-resistant prostate cancer cells in vitro and in vivo
- Prognostic potential of m7G-associated lncRNA signature in predicting bladder cancer response to immunotherapy and chemotherapy
- Case Reports
- A rare FBXO25–SEPT14 fusion in a patient with chronic myeloid leukemia treatment to tyrosine kinase inhibitors: a case report
- Stage I duodenal adenocarcinoma cured by a short treatment cycle of pembrolizumab: a case report
- Rapid Communication
- ROMO1 – a potential immunohistochemical prognostic marker for cancer development
- Article Commentary
- A commentary: Role of MTA1: a novel modulator reprogramming mitochondrial glucose metabolism
Articles in the same Issue
- Frontmatter
- Review Articles
- Mitochondrial thermogenesis in cancer cells
- Application of indocyanine green in the management of oral cancer: a literature review
- Long non-coding RNA, FOXP4-AS1, acts as a novel biomarker of cancers
- The role of synthetic peptides derived from bovine lactoferricin against breast cancer cell lines: a mini-review
- Single cell RNA sequencing – a valuable tool for cancer immunotherapy: a mini review
- Research Articles
- Global patterns and temporal trends in ovarian cancer morbidity, mortality, and burden from 1990 to 2019
- The association between NRF2 transcriptional gene dysregulation and IDH mutation in Grade 4 astrocytoma
- More than just a KRAS inhibitor: DCAI abrogates the self-renewal of pancreatic cancer stem cells in vitro
- DUSP1 promotes pancreatic cancer cell proliferation and invasion by upregulating nephronectin expression
- IMMT promotes hepatocellular carcinoma formation via PI3K/AKT/mTOR pathway
- MiR-100-5p transfected MSCs-derived exosomes can suppress NSCLC progression via PI3K-AKT-mTOR
- Inhibitory function of CDK12i combined with WEE1i on castration-resistant prostate cancer cells in vitro and in vivo
- Prognostic potential of m7G-associated lncRNA signature in predicting bladder cancer response to immunotherapy and chemotherapy
- Case Reports
- A rare FBXO25–SEPT14 fusion in a patient with chronic myeloid leukemia treatment to tyrosine kinase inhibitors: a case report
- Stage I duodenal adenocarcinoma cured by a short treatment cycle of pembrolizumab: a case report
- Rapid Communication
- ROMO1 – a potential immunohistochemical prognostic marker for cancer development
- Article Commentary
- A commentary: Role of MTA1: a novel modulator reprogramming mitochondrial glucose metabolism