Abstract The tumor immune microenvironment (TIME) significantly influences cancer progression and treatment. This study sought to uncover novel TIME-related glioma biomarkers to advance antitumor immunotherapies by integrating data from sequencing of bulk RNA as well as scRNA. Immunologic and epithelial-mesenchymal transition (EMT) characteristics were used to classify glioma patients into two immune subtypes (ISs) and two EMT subtypes (ESs). Patients in IS1 and ES1, characterized by high immune infiltration and low stemness scores, exhibited poor clinical outcomes and limited responsiveness to immunotherapy. A new risk signature was developed using 16 genes and validated in independent glioma cohorts. Among these, HAVCR2, IL18, LAGLS9, and PTPN6 emerged as hub genes, with IL18 identified as a potential independent indicator. The upregulation of IL18 in high-grade gliomas and U-251 MG cells aligned with bioinformatics analysis. These insights deepen the understanding of TIME-related mechanisms in glioma and highlight potential therapeutic targets, offering a theoretical foundation for effective antitumor immunotherapies in glioma. Keywords: Molecular subtypes, Immunologic and epithelial-mesenchymal transition gene sets, Tumor immune microenvironment, Immunotherapy 1. Introduction Gliomas exist as the most common brain tumor that arises from cells in the glia, significantly contributing to morbidity and mortality [[27]1]. Glioblastoma (GBM) stands as the most frequent variant of malignant glioma, comprising roughly 80 % of primary malignant brain tumors. It has a grim prognosis, with under two years of median overall survival (OS), significantly diminishing the quality of life [[28]2]. Despite advances in understanding glioma biology and the development of targeted therapies in preclinical and clinical trials, the incidence of recurrence, cognitive deficiencies, disability, and mortality associated with glioma remains high [[29]3]. Most glioma treatments with notable survival benefits, such as radiation and chemotherapy, rely on nonspecific targeting of proliferating cells and are often compromised by glioma cell invasion and immune evasion [[30]3,[31]4]. Therefore, it is crucial to identify unknown factors or further investigate known factors that influence the tumor microenvironment (TME) to promote glioma progression, malignant transformation, and drug resistance. This approach is vital for advancing the assessment and customization of therapeutic options for gliomas. The TME, which is the site of tumor emergence, is significantly implicated in its development [[32]5,[33]6]. TME primarily consists of the extracellular matrix, various cellular components such as fibroblasts and immune components, along with vascular networks that are sourced from adjoining tissues [[34][5], [35][6], [36][7]]. With the participation of immune-related genes (IRGs) and epithelial-mesenchymal transition (EMT)-related genes (ERGs), the TME functions crucially in tumor progression and treatment resistance, influenced by extracellular matrices, cytokines, growth signals, and other elements, thereby affecting cancer treatment [[37]8]. Compared to brain tissue in the normal state, which exhibits a low to moderate immune response, the tumor immune microenvironment (TIME) significantly contributes to the aggressive advancement and therapeutic resistance in glioma [[38]9]. A suppressive TIME can severely impair GBM treatment [[39]10]. Remodeling TIME using a liposomal honokiol and disulfiram/copper codelivery system (CDX-LIPO) specifically targeting the brain can trigger autophagy and immune-mediated cell death in tumor cells. This process activates macrophages and dendritic cells infiltrating tumors, as well as the priming of T and NK (natural killer) cells, leading to an immunity response against tumors and tumor shrinkage [[40]10]. Recently, various IRGs have been identified, which can be categorized as low-risk and high-risk [[41][11], [42][12], [43][13]], suggesting that IRGs play differential and complex roles in the emergence, advancement, and metastatic spread of gliomas. Increasing evidence suggests EMT facilitates glioma invasion and progression. EMT, a multifactor-regulated process, involves various induction agents, families of transcription factors, and an extensive range of genes involved in signaling pathways [[44]14]. Enhanced EMT is associated with poor survival, whereas the EMT in the initial stages iscomparatively less malignant in GBM [[45]15], suggesting that EMT assessment could predict GBM prognosis and aid in the advancement of innovative therapeutic strategies and prognostic markers [[46]15]. Additionally, the elevated expression of EMT genes in GBMs and pilocytic astrocytomas is indicative of mural cells proliferation within newly formed blood vessels [[47]16]. Six collagen genes have been identified as regulators of both the glioma immunosuppressive tumor microenvironment and the EMT process, namely COL1A1, COL1A2, COL3A1, COL4A1, COL4A2, and COL5A2 [[48]17]. This suggests that IRGs and ERGs may synergistically influence glioma behaviors such as invasion, migration, and metastasis. Cohort analysis of The Cancer Genome Atlas (TCGA) uncovered two immune subtypes (ISs) and two EMT subtypes (ESs) in patients with glioma based on IRG and ERG sets. This resulted in the elucidation of key immune- and EMT-related genes (IERGs), with sixteen chosen to construct a novel risk prediction model for glioma patients. Among these, four IERGs (HAVCR2, IL18, LAGLS9, and PTPN6) emerged as hub genes, showing high expression in M1 macrophages and monocytes. Furthermore, the upregulation of IL18 in GBM indicates its potential as an independent clinical marker for patients with high-grade glioma. This integrated analysis of IRGs and ERGs in gliomas establishes a theoretical framework aimed at developing and implementing more effective antitumor immunotherapies. 2. Results 2.1. Identification of novel ISs and ESs of gliomas and analysis of their survival and different immune features The study framework is detailed in [49]Supplementary Fig. 1. Consensus clustering without supervision (k = 2) was applied to 663 gliomas from the TCGA cohort, focusing on immunologic and EMT gene signatures. This analysis categorized patients with glioma into two ISs (IS1 and IS2) and two ESs (ES1 and ES2) ([50]Fig. 1A–H). Based on Kaplan-Meier survival analysis, individuals in the IS1 and ES1 subtypes had better OS compared to those in IS2 and ES2 ([51]Fig. 1B–I). Gene set enrichment analysis (GESA) of HALLMARK pathways revealed significant differential enrichment in five pathways—interferon-gamma response, IL2-STAT5 signaling, apoptosis, hypoxia, and glycolysis—in IS1 ([52]Fig. 1C) and ES1 ([53]Fig. 1J) based on NES, P-value, and FDR value. Fig. 1. [54]Fig. 1 [55]Open in a new tab Unsupervised consensus clustering of gliomas based on immunologic and EMT gene signatures in the TCGA cohort. (A and H) Heatmap depicting consensus clustering matrix (k = 2) for 1959 immunologic genes (A) and 1263 EMT genes (H) across 663 glioma samples. (B and I) Kaplan-Meier survival analysis of patients with gliomas exhibiting different ISs (B) and ESs (I). (C and J) GSEA for pathways in HALLMARK terms of different ISs (C) and ESs (J). (D and K) Potential ICB response predicted with TIDE algorithm in different ISs (D) and ESs (K). (E and L) Stemness score based on OCLR algorithm in different ISs (E) and ESs (L). (F-G and M-N) TMB and MSI score in different ISs (F-G) and ESs (M-N). The TIDE algorithm, used to predict potential immunotherapeutic responses, showed significantly higher TIDE scores in IS1 and ES1 compared to IS2 and ES2 ([56]Fig. 1D–K), indicating that IS1 and ES1 patients had poorer responses to immune checkpoint blockade (ICB) immunotherapy and experienced decreased survival following therapies. The OCLR algorithm assessed stemness scores, showing higher scores in IS2 and ES2 than in IS1 and ES1 ([57]Fig. 1E–L), which correlates with greater tumor differentiation in IS2 and ES2 patients. Tumor mutational burden (TMB) and microsatellite instability (MSI) scores, critical for predicting immune efficacy, were higher in IS1 and ES1 compared to IS2 and ES2 ([58]Fig. 1F–M) and lower in MSI score ([59]Fig. 1G–N). Interestingly, IS1 and ES1 exhibited similar survival prognoses and clinicopathological features. 2.2. TIME features of distinct TCGA cohort subtypes To evaluate the association of glioma subtypes within immune scores, the ESTIMATE algorithm evaluated stromal and immune cell infiltration, along with tumor purity, across different ISs and ESs. This analysis generated four scores: immune, stromal, ESTIMATE, and tumor purity. The results indicated IS1 and ES1 showed increased immune scores ([60]Supplemental Figs. 2A and E), stromal scores ([61]Supplemental Figs. 2B and F), and ESTIMATE scores ([62]Supplemental Figs. 2C and G), along with lower tumor purity ([63]Supplemental Figs. 2D and H) compared to IS2 and ES2. Higher tumor purity in IS2 and ES2 suggested fewer infiltrated immune and stromal cells in these gliomas. The infiltration profiles of 22 distinct immune cell types were assessed by the CIBERSORT algorithm across various ISs and ESs, and the results were displayed using heatmaps ([64]Supplemental Figs. 2I and K). Clear differences were noted in various populations of immune cells in the comparison of IS1 and IS2, and between ES1 and ES2. Notably, the composition of B cell plasma, resting T cells CD4^+ memory, monocytes, and M2 macrophages varied significantly between IS1 and IS2 and between ES1 and ES2 ([65]Supplemental Figs. 2J and L). 2.3. Linkage between ISs and ESs with ICPs, ICD modulators, and HLA family Considering the critical roles of immune checkpoint proteins (ICPs), immunogenic cell death (ICD) modulators, and HLA family genes in cancer immunity and the effectiveness of vaccines against mRNA, distinct expression of these elements was assessed in different ISs and ESs. The levels of expression for 25 ICD modulators, 47 ICPs, and 24 HLA family genes were investigated across different ISs and ESs. The analysis revealed that 19 ICD modulators, 39 ICPs, and 23 HLA-related genes were differentially expressed in these subtypes ([66]Supplemental Fig. 3). For instance, 18 ICD modulators were significantly upregulated in IS1 and ES1 gliomas. Additionally, 38 ICPs, excluding ADORA2A, CD27, IDO2, KIR3DL1, TIGIT, TNFRSF25, TNFSF18, TNFSF9, and VSIR, showed significant upregulation in IS1 and ES1 gliomas ([67]Supplemental Fig. 3). All HLA-related genes, except HLA.J, exhibited significantly higher expression levels in IS1 and ES1 gliomas ([68]Supplemental Fig. 3). These findings suggest that IS1 and ES1 gliomas share similar expression profiles of ICPs, ICD modulators, and HLA family genes, indicating that patients with IS1 and ES1 gliomas may benefit from similar mRNA vaccine therapies. 2.4. Exploring Time-related modules by WGCNA To recognize IERGs, WGCNA on the gliomas-TCGA cohort constructed co-expression networks and pinpointed gene modules linked with immune scores. Setting the soft threshold to 12 ([69]Fig. 2A), the analysis identified 20 key TIME-related gene modules, each color-coded ([70]Fig. 2B). The eigengene adjacency heatmap illustrated correlations among these modules ([71]Fig. 2C). Further exploration revealed their associations with clinical factors, such as grade, age, gender, and various immune-related parameters ([72]Fig. 2D). The brown module, showing the strongest links to stromal, immune and ESTIMATE score, was selected to conduct deeper analysis ([73]Fig. 2D). Scatter plots demonstrated significant positive associations of the brown module membership with immune score ([74]Fig. 2E), stromal score ([75]Fig. 2F), ESTIMATE score ([76]Fig. 2G), and tumor purity ([77]Fig. 2H). Fig. 2. [78]Fig. 2 [79]Open in a new tab WGCNA screening for TIME-related module in the TCGA cohort. (A) Analysis of the scale-free topology fit index and mean connectivity for various soft-threshold powers (β), selecting a power of 12. (B) Dendrogram of all genes clustered based on a dissimilarity measure (1-TOM). (C) Correlation heatmap of different modules. (D) Determination of module-trait relationships in patients with glioma. (E-H) Scatter plots of module eigengene significance for immune score (E), stromal score (F), ESTIMATE score (G), and tumor purity (H) in the brown module. (For interpretation of the references to color in this