Abstract Background Ferroptosis is a novel type of programmed cell death in various tumors; however, underlying mechanisms remain unclear. We aimed to develop ferroptosis-related long non-coding RNA (FRlncRNA) risk scores to predict lower-grade glioma (LGG) prognosis and to conduct functional analyses to explore potential mechanisms. Methods LGG-related RNA sequencing data were extracted from The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) databases. Pearson correlation analysis was used to identify the FRlncRNAs, univariate Cox regression analysis was for identify the prognostic FRlncRNAs, and then intersection FRlncRNAs were screened between TCGA and CGGA. Least absolute shrinkage and selection operator (LASSO) Cox regression was used to develop a risk score to predict LGG prognosis. Results A total of nine FRlncRNAs were screened to construct the novel prognostic risk score of LGG, and high-risk score patients had a worse overall survival than low-risk score patients both in TCGA and CGGA datasets. The risk score was quite correlated with clinicopathological characteristics (age, WHO grade, status of MGMT Methtlation, IDH mutation, 1p/19q codeletion, and TMB), and could promote current molecular subtyping systems. Comprehensive analyses revealed that signaling pathways of B-cell receptor and T-cell receptor, immune cells of macrophage cell and CD4+ T cell, tumor microenvironment of stroma score and immune score, and immune checkpoints of PD-1, PD-L1, and CTLA4 were all enriched in the high-risk score group. Conclusion The nine FRlncRNAs risk scores was a promising biomarker to predict the LGG’s prognosis and distinguish the characteristics of molecular and immune. Supplementary Information The online version contains supplementary material available at 10.1007/s12672-024-01587-9. Keywords: Ferroptosis, Long non-coding RNAs, Lower-grade glioma, Risk score, Immune checkpoints Introduction Approximately 30,000 patients were diagnosed with glioma in the United States from 2013 to 2017, accounting for approximately 81.1% of all malignant central nervous system tumors [[32]1]. Surgical excision followed by temozolomide-based adjuvant treatment is standard treatment for gliomas, but long-term prognosis remains far from satisfactory [[33]2]. The reasons for this poor prognosis may be (1) patients in the same subtype stratified by the existing molecular subtyping often have different outcomes [[34]3], (2) total removal of intracranial lesions is anatomically difficult to perform [[35]2], and (3) while adjuvant treatments based on the current molecular pathology have failed in several trials [[36]4–[37]6]. Therefore, the investigation of novel predictive biomarkers and therapeutic targets to treat gliomas is urgently needed. Ferroptosis is a newly discovered type of non-apoptotic programmed cell death that is characterized by iron-dependent lipid peroxidation [[38]7]. Previous findings suggest that ferroptosis can suppress tumor growth and progression, and survival models based on ferroptosis-related genes (FRGs) have been successfully constructed to predict the prognosis of various types of malignant tumors [[39]8–[40]10], including lower-grade glioma (LGG) [[41]11]. In addition, ferroptosis is also associated with therapy resistance to chemotherapy or radiotherapy, which provides insight into novel therapeutic alternatives for glioma treatment, such as using iron chelators [[42]12] and lipid peroxidation inhibitors [[43]13]. Although FRGs are reported to be associated with the prognosis of gliomas due to cell membrane destruction regulated by iron-dependent lipid peroxidation, the transcription and translation of FRGs remain unclear [[44]14]. Long non-coding RNAs (lncRNAs) are a subset of RNA molecules of approximately 200 nt that play significant roles in gene transcription, microRNA (miRNA) regulation, and protein folding [[45]15]. Preliminary studies have found that aberrant lncRNA expression is associated with glioma pathogenicity, such as that of lncRNAs HOXA11-AS [[46]16] and MALAT1 [[47]17], and previous studies identified the stability of lncRNA-based prognostic signatures in various kinds of cancers [[48]18, [49]19]. Meanwhile, lncRNAs also participate in the regulation of ferroptosis. Silencing of lncRNAs LINC00618 [[50]20], PVT1 [[51]21], and ZFAS1 [[52]22] has been found to attenuate ferroptosis through lipid peroxidation. However, there are currently no studies on ferroptosis-related lncRNAs in gliomas. In the current study, we developed a multi-ferroptosis-related lncRNAs (FRlncRNAs) risk score to predict overall survival (OS) of LGG patients using data from The Cancer Genome Atlas (TCGA), which was also validated using the Chinese Glioma Genome Atlas (CGGA) databases. Then comprehensive analysis was performed to explore the potential value of the risk score, and a competing endogenous RNA (ceRNA) network was developed to search for a potential target for LGGs. Methods Data sources LGG in this study was defined as grade II or grade III gliomas according to the World Health Organization (WHO) classification. RNA sequencing data related to LGG in the TCGA database were extracted using the Genomic Data Commons Data Portal ([53]https://portal.gdc.cancer.gov/). Related clinicopathological data were acquired from the cBioPortal website ([54]https://www.cbioportal.org/). RNA expression files in the CGGA database and their corresponding clinicopathological data were downloaded from the CGGA website ([55]http://www.cgga.org.cn/). The [56]GSE108474 and [57]GSE43378 datasets were retrieved from the Gene Expression Omnibus (GEO) database ([58]www.ncbi.nlm.nih.gov/geo). Clinical characteristics of the patients in the datasets are shown in Table [59]1 Table 1. Clinical characteristics of the LGG patients used in this study^a TCGA cohort CGGA cohort [60]GSE108474 [61]GSE43378 No. of patients 515 625 187 18 Age Median (range) 41 (14–87) 40 (10–74) 0 (0.0%) 51.5 (24–73) ≤ 40 (%) 254 (49.3%) 330 (52.8%) 0 (0.0%) 6 (33.3%) > 40 (%) 261 (50.7%) 294 (47.0%) 0 (0.0%) 12 (66.7%) Unknown 0 (0.0%) 1 (0.2%) 187 (100.0%) 0 (0.0%) Gender (%) Female 230 (44.7%) 263 (42.1%) 60 (32.1%) 4 (22.2%) Male 285 (55.3%) 362 (57.9%) 94 (50.3%) 14 (77.8%) Unknown 0 (0.0%) 0 (0.0%) 33 (17.6%) 0 (0.0%) Grade (%) WHO I 0 (0.0%) 0 (0.0%) 2 (1.1%) 0 (0.0%) WHO II 249 (48.3%) 291 (46.6%) 92 (49.2%) 5 (27.8%) WHO III 265 (51.5%) 334 (53.4%) 69 (36.9%) 13 (72.2%) Unknown 1 (0.2%) 0 (0.0%) 24 (12.8%) 0 (0.0%) MGMT methylation status (%) Methylated 93 (18.1%) 298 (47.7%) NA NA Unmethylated 422 (81.9%) 211 (33.8%) NA NA Unknown 0 (0.0%) 116 (18.5%) NA NA IDHMutant (%) Mutant 414 (80.4%) 439 (70.2%) NA NA Wild 92 (17.9%) 144 (23.1%) NA NA Unknown 9 (1.7%) 42 (6.7%) NA NA 1p/19q Coldel (%) Codel 173 (33.6%) 191 (30.6%) NA NA Non-codel 210 (40.8%) 393 (62.9%) NA NA Unknown 132 (25.6%) 41 (6.6%) NA NA [62]Open in a new tab a. Inclusion Criteria: 1. Gene expression profiles of patients with grade I-III glioma Exclusion Criteria: 1. Patients with grade IV glioma. 2. Duplicate reports from the same cohort. 3. Protocol, case reports, or reviews. 4. Data unavailable lncRNAs annotation The lncRNA annotation file (Genome Reference Consortium Human Build 38, GRCh38) was downloaded from the GENCODE website ([63]https://www.gencodegenes.org/human/) and used to annotate the lncRNA in the TCGA dataset. Accordingly, 14,247 lncRNAs were identified in the TCGA dataset and 4304 identified in the CGGA dataset. FRlncRNAs identification A list of recognized FRGs, including 150 driver genes, 109 suppressor genes, and 123 marker genes, was acquired from the FerrDb database (Table S1) [[64]23]. Pearson correlation analysis (|R|> 0.3, P < 0.0001) was used to identify FRlncRNAs in each dataset. The FRlncRNAs in the two datasets were screened using univariate Cox regression analysis based on OS (P < 0.001). The FRlncRNAs that intersected both the TCGA and CGGA datasets were identified as candidate FRlncRNAs for determining the risk scores. FRlncRNAs risk score construction Least absolute shrinkage and selection operator (Lasso)-penalized Cox regression was used to construct the FRlncRNAs risk scores following the minimum criteria with the penalty parameter (λ) based on “glmnet” R package using a tenfold cross-validation (Fig. S1). The risk score of each patient was calculated according to the following formula: [MATH: Riskscore=i=1 nCoefi×xi :MATH] Patients in each dataset were then divided into high-risk and low-risk subgroups according to their risk scores. The median risk score was used as the cut-off value. The risk score was evaluated using the receiver operating characteristic (ROC) curve and validated using univariate & multivariate Cox analyses based on OS in CGGA, [65]GSE108474, and [66]GSE43378 datasets. Function analysis Gene ontology (GO) function analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were preformed according to the differentially expressed genes (DEGs) between the high-risk and low-risk groups. The DEGs were defined according to an absolute log-twofold-change (|log2FC|) ≥ 1 and a false detection rate (FDR) < 0.05. Gene set enrichment analysis (GSEA) was used to identify potential biological processes and pathways that differed between the high-risk and low-risk subgroups [[67]24]. The correlation between estimated numbers of immune cells in the tumor environment and risk scores was evaluated using various tools available on the TIMER 2.0 website ([68]http://timer.cistrome.org), including XCELL, TIMER, QUANTISEQ, MCPcounter, EPIC, and CIBERSORT. Hierarchical clustering To examine the relationship between the newly identified molecular subtypes and the previously characterized LGG immune subtypes, we conducted unsupervised hierarchical clustering utilizing the CIBERSORT algorithm, which quantifies the immune cell composition of each sample. This analysis enabled us to categorize the patients into two distinct subgroups, cluster 1 (n = 771) and cluster 2 (n = 369). Analysis of tumor mutation burden (TMB) Mutation data were also downloaded from the TCGA database, including somatic mutations, insertion-deletion mutations, coding mutations, and base replacement per million bases data. The TMB score was calculated as the total number of somatic mutations. The LGG patients were then separated into low-TMB and high-TMB groups using the median TMB value as a cut-off value. Predictive nomogram construction Independent prognostic factors were identified using multivariate Cox regression analysis. Based on the analysis results, a nomogram was developed to predict the prognosis of patients with LGG. Ferroptosis-related ceRNA network construction To investigate the potential regulatory mechanism of FRlncRNAs in LGG on mRNA expression via the sponging of miRNAs, the ceRNA network was constructed. Briefly, FRlncRNA-related miRNAs were identified using the miRcode database ([69]http://www.mircode.org/) and mRNAs related to the miRNAs were identified using the miRDB ([70]http://mirdb.org/), miRTarBase ([71]http://mirtarbase.mbc.nctu.edu.tw/php/index.php), and TargetScan databases ([72]http://www.targetscan.org/). The ceRNA network was plotted using Cytoscape 3.7.2 software [[73]25]. Statistical analysis The Pearson correlation analysis used to evaluate the correlation between lncRNAs and FRGs was performed using a |R^2|< 0.3 and P-value of 0.0001. Kaplan–Meier analysis was used to compare OS between different groups using the log-rank test. Univariate and multivariate Cox regression analyses were performed to identify independent predictors of OS. The diagnostic value of the risk scores and nomogram model were evaluated using ROC curves. The Spearman test was used to determine the association between estimated immune cell numbers and risk scores. The Wilcoxon rank sum test was used to evaluate differences stratified by various clinicopathological characteristics. All statistical analyses were conducted using R software (version 4.0.3, [74]https://www.r-project.org). Unless specified otherwise, P with two tailed value of less than 0.05 was considered statistically significant. Results Identification of FRlncRNAs in LGG patients Totally, 515 patients and 625 patients were screened in the TCGA database, and CGGA database, respectively. A total of 1637 FRlncRNAs were identified in the TCGA dataset and 493 in the CGGA dataset. In addition, 667 prognostic lncRNAs in the TCGA dataset and 215 in the CGGA dataset were confirmed to be associated with OS in LGG patients (Table S2). Finally, 81 lncRNAs were identified as intersecting prognostic FRlncRNAs between the two datasets (Table S3). A flow chart of this study is depicted in Fig. [75]1a. Fig. 1. [76]Fig. 1 [77]Open in a new tab a Flow chart of the study. b The competing endogenous RNA (ceRNA) network of 31 ferroptosis-related lncRNAs (FRlncRNAs; red), 30 target microRNAs (miRNAs; yellow), and 49 mRNAs (blue) Ferroptosis-related ceRNA network construction The ceRNA network is a complex regulatory network between non-coding RNAs and coding RNAs in the cell via miRNAs. Of the 81 intersecting FRlncRNAs, 31 were found in the miRcode database. Thirty of the miRNAs were identified as being associated with FRlncRNAs with 280 pairs. Subsequently, 49 mRNAs were found to be associated with 80 pairs of the FRlncRNA-related miRNAs. The ceRNA network of the most critical 31 lncRNAs, 30 miRNAs, and 49 mRNAs is depicted in Fig. [78]1b. Construction of a prognostic risk score in the TCGA training set Using LASSO Cox regression analysis, nine FRlncRNAs, including AGAP2-AS1, FAM181A-AS1, CRNDE, PAXIP1-AS2, TMEM254-AS1, SLC25A21-AS1, DGCR9, LINC00844, and LINC00641, were screened to establish a risk score for predicting the prognosis of LGG patients in the TCGA training set, which is shown in Fig. [79]2a along with each coefficient value. The prognostic risk score of each patient was determined according to the following formula: Fig. 2. [80]Fig. 2 [81]Open in a new tab a Coefficient values of nine ferroptosis-related lncRNAs (FRlncRNAs) associated with a risk model for lower-grade glioma (LGG). b Sankey diagram showing the top five mRNAs and risk type related to each FRlncRNA. Kaplan–Meier curves (c), risk plot distribution and survival status (d), and time-dependent receiver operating characteristic (ROC) curves (e) of The Cancer Genome Atlas (TCGA) dataset. Kaplan–Meier curves (f), risk plot distribution and survival status (g), and time-dependent ROC curves (h) of the Chinese Glioma Genome Atlas (CGGA) dataset [MATH: Riskscore=AGAP2-< /mo>AS1×0.066+FAM181A -AS1×0.087+CRNDE×< /mo>0.264+PAXIP1< /mn>-AS2×0.330-TMEM254 -AS1×0.190-SLC25A2 1-AS1×0.109< /mrow>-DGCR9×< /mo>0.077-LINC00844×0.076-LINC00641×0.066 :MATH] Nine FRlncRNAs related mRNAs were identified based on |R^2|< 0.3 and a P-value of 0.0001 and the top five mRNAs related to each FRlncRNA are shown in Fig. [82]2b. Of note, TMEM254-AS1, SLC25A21-AS1, DGCR9, LINC00844, and LINC00641 were protective factors for OS, while AGAP2-AS1, FAM181A-AS1, CRNDE, and PAXIP1-AS2 were risk factors for OS. The median risk score of the patients in the TCGA dataset was − 0.754 (− 1.433–1.556). The patients were divided into low-risk and high-risk groups based on the median risk score values and the groups exhibited apparent differences in terms of OS (P < 0.001, Fig. [83]2c). The corresponding risk scores and survival status are plotted in Fig. [84]2d. The time-dependent ROC curves revealed the FRlncRNAs exhibited better discrimination than the other prognostic models in the TCGA dataset (age, gender, grade, and status of MGMT methtlation, IDH mutation, 1p/19q codeletion; Fig. [85]2e). Similar findings were observed for the CGGA, [86]GSE108474, and [87]GSE43378 (Fig. [88]2f–h, Fig. S2a–d). Univariate and multivariate Cox analyses of risk scores In the TCGA dataset, univariate analysis showed the risk score was significantly associated with OS with a hazard ratio (HR) = 4.075, 95% confidence interval (CI) 3.108–5.344, and P < 0.001 (Fig. [89]3a). It was also significantly associated with age (Fig. [90]3c), WHO tumor grade (Fig. [91]3e), isocitrate dehydrogenase (IDH) mutant status (Fig. [92]3g), and 1p/19q codeletion status (Fig. [93]3h) with each having a P-value < 0.001. Further multivariate Cox analysis showed the risk score was an independent factor of OS (HR = 3.330, 95% CI 1.773–6.254, P < 0.001; Fig. [94]3a). Similar to that of the TCGA dataset, risk score was also found to be associated with OS and an independent prognostic factor of the CGGA, [95]GSE108474, and [96]GSE43378 datasets based on both univariate and multivariate Cox analyses (Fig. [97]3b, Fig. S3a, b). Fig. 3. [98]Fig. 3 [99]Open in a new tab Univariate and multivariate Cox regression analysis of lower-grade glioma (LGG) prognosis in The Cancer Genome Atlas (TCGA) (a) and Chinese Glioma Genome Atlas (CGGA) (b) datasets. Evaluation of clinicopathological characteristics in the TCGA dataset, including age (c), gender (d), World Health Organization (WHO) grade (e), O-6-methylguanine-DNA methyltransferase (MGMT) methylation status (f), isocitrate dehydrogenase (IDH) mutation status (g), and 1p/19q codeletion status (h). *p < 0.05, **p < 0.01 and ***p < 0.001 Risk score stratified by clinicopathological characteristics We also wanted to determine whether the risk scores correlated with various clinicopathological characteristics of patients with LGG. Results showed that patient age > 40, WHO grade III, unmethylated O-6-methylguanine-DNA methyltransferase (MGMT), wild-type IDH, and non-1p/19q codeletion were found to have higher risk scores (Fig. [100]3c, e–h; all P < 0.001), but no significant difference was observed in terms of risk score between females and males (P = 0.36; Fig. [101]3d). Similar results were observed for the CGGA dataset (Fig. S4). Risk score benefit to current molecular classification According to the existing molecular classification systems, patients exhibited apparently different OS when stratified by WHO grading, IDH mutation status, and 1p/19q codeletion status (P < 0.001, Fig. [102]4a, c, d), but no significant difference was observed between subgroups stratified by MGMT methylation status (P = 0.091, Fig. [103]4b). Further analyses showed that patients in each subtype could also be divided into two different groups in terms of OS according to their risk score (all P < 0.05, Fig. S5a–d, f–i). Of note, patients with WHO grade II tumors and high-risk scores had a parallel prognosis to that of patients with WHO grade III tumors and low-risk scores (Fig. [104]4e). A similar finding was observed in the existing classification system of 1p/19 codeletion (Fig. [105]4h). Fig. 4. [106]Fig. 4 [107]Open in a new tab Kaplan–Meier curves of multiple subgroups, including World Health Organization (WHO) grading (a), O-6-methylguanine-DNA methyltransferase (MGMT) methylation status (b), isocitrate dehydrogenase (IDH) mutation status (c), and 1p/19q codeletion status (d). The risk score was further explored to supplement WHO grading (e) and molecular subtypes, including MGMT methylation status (f), IDH mutation status (g), and 1p/19q codeletion status (h). Association between tumor mutation burden (TMB) and ferroptosis-related lncRNA (FRlncRNA) low-risk and high-risk scores (i). Kaplan–Meier curves of TMB (j) and TMB with low-risk and high-risk score (k) TMB analysis The TMB score in the subgroup with a high-risk score was higher than that in the subgroup with a low-risk score (P < 0.001; Fig. [108]4i, Table S4) and patients with high TMB were found to have worse OS than those with low TMB (P < 0.001, Fig. [109]4j). K–M curves showed that patients with high-risk score had a worse OS than those with low-risk score both in the subgroup of high TMB and low TMB (both P < 0.05, Fig. S5e, j). However, further analysis showed that patients with high TMB and low-risk scores had a similar OS compared with that of patients with low TMB and high-risk scores (Fig. [110]4k). Functional analysis A total of 739 DEGs were identified between the low-risk and high-risk groups of the TCGA dataset and 63 DEGs between the low-risk and high-risk groups of the CGGA dataset. As shown in Fig. [111]5a, GO enrichment revealed many enriched terms clearly related to immunity, including humoral immune response (biological process), MHC protein complex (cellular component), and antigen binding (molecular function). KEGG analysis identified immune-related pathways, such as complement and coagulation cascades, antigen processing and presentation, and intestinal immune network for IgA production, and DEGs accompanied by an activated PI3K-Akt signaling pathway were also enriched (Fig. [112]5b). The results were confirmed with similar findings for the CGGA dataset (Fig. S6). Fig. 5. [113]Fig. 5 [114]Open in a new tab The most significant gene ontology (GO) enrichment (a) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways (b) in The Cancer Genome Atlas (TCGA) dataset. Multiple gene set enrichment analysis (GSEA) for metabolism-related (c) and immune-related (d) pathways of the same dataset Functional annotation was performed using GSEA. DEGs were only enriched in the high-risk group (P-value < 0.05, Table S5). Metabolism related signaling pathways including galactose, nicotinate, phenylalanine, starch, and sucrose were enriched in the high-risk score group (Fig. [115]5c), as well as immunity related signaling pathways such as antigen processing and presentation, B-cell receptor signaling pathway, and T-cell receptor signaling pathway, among others (Fig. [116]5d). Subsequent correlation analyses revealed that the PI3K-Akt signaling pathway exhibited a significant positive correlation with the risk score, ferroptosis, as well as the previously mentioned immune pathways (e.g. B-cell receptor signaling pathway, T-cell receptor signaling pathway, and), immune cell infiltration (e.g. macrophage cell), and immune checkpoint expression (e.g. PD-1, PD-L1, and CTLA4) within both the dataset of TCGA and CGGA (P < 0.05, Fig. S7, Fig. S8). Similar results were confirmed in the external validation dataset [117]GSE108474 (Fig. S9). Immune response and risk score As shown in Fig. [118]6a, many tumor-infiltrating immune cells were found to be associated with evaluated FRlncRNA risk scores using different infiltration estimating tools, including XCELL, TIMER, QUANTISEQ, MCPcounter, EPIC, and CIBERSORT. In particular, estimations of all macrophage subtypes in the high-risk group were higher than those in the low-risk group (P < 0.001, Fig. [119]6b). Estimations of most CD4^+ T cells in the high-risk group were also higher than those in the low-risk group (P < 0.001, Fig. [120]6c). However, all estimations of NK cells in the high-risk group were lower than those in the low-risk group (P < 0.001, Fig. [121]6e). Notably, no definite difference was observed in terms of CD8^+ T cells between the high-risk and low-risk groups (Fig. [122]6d). Fig. 6. [123]Fig. 6 [124]Open in a new tab Correlation coefficient between the tumor-infiltrating immune cells and ferroptosis-related lncRNA (FRlncRNA) risk score (a). The boxplots show the differences in infiltrating immune cell subtypes, including macrophages (b), CD4+ T cells (c), CD8+ T cells (d), and natural killer (NK) cells (e) between the low-risk score (yellow) and high-risk score (blue) groups. Association of microenvironment score (f) and PD-1, PD-L1, and CTLA4 expression (g) between the low-risk score (yellow) and high-risk score (blue) groups. *p < 0.05, **p < 0.01 and ***p < 0.001 The tumor environment and immune checkpoints between the high-risk and low-risk groups were also explored using expression data (ESTIMATE). Results for the tumor environment showed that stromal score, immune scores, and total score were each higher in the high-risk score group compared to those in the low-risk score group (P < 0.001, Fig. [125]6f). Immune checkpoints results indicated that PD-1, PD-L1, and CTLA4 were all overexpressed in the high-risk score group compared with those in the low-risk score group (P < 0.001, Fig. [126]6g). Relationship between immune subtypes and risk score The degree of immune infiltration in each sample was evaluated using CIBERSORT algorithm, which served as the foundation for implementing unsupervised clustering to identify two immune subtypes exhibiting significantly distinct immune characteristics (Fig. S10a). Further analysis revealed that cluster 1, despite exhibiting a higher risk score and poorer prognosis (P < 0.05, Fig. S10b, c), demonstrated elevated levels of immune checkpoint expression (P < 0.05, Fig. S10d–f). This finding indicates that cluster 1 exhibits a stronger correlation with the high risk score group, which comprises patients who may demonstrate increased sensitivity to immunotherapy. Nomogram construction based on FRlncRNAs risk scores A nomogram was developed based on FRlncRNA risk score to further predict OS of LGG patients (Fig. [127]7a). The concordance-index was as high as 0.848 (0.803–0.892). Good calibrations were observed in terms of the predicted vs. observed survival rates at 1, 3, and 5 year (Fig. [128]7b–d). Time-dependent ROC curves showed the current nomogram had a higher AUC for 1-, 3-, and 5-year OS than that of the other predictors (AUC at 1-year: 0.883, Fig. [129]7e; AUC at 3-year: 0.899, Fig. [130]7f; and AUC at 5-year: 0.826, Fig. [131]7g). Fig. 7. [132]Fig. 7 [133]Open in a new tab a Nomogram for predicting survival of lower-grade glioma (LGG) patients. b–d Calibration plots of the nomogram for predicting the probability of overall survival (OS) at 1, 3, and 5 years. e–g Receiver operating characteristic (ROC) curves for the ferroptosis-related lncRNA (FRlncRNA) risk score, patient age, patient gender, tumor grade, O-6-methylguanine-DNA methyltransferase (MGMT) methylation status, isocitrate dehydrogenase (IDH) mutation status, and 1p/19q codeletion status and a nomogram for predicting 1-, 3-, and 5-year OS Discussion Ferroptosis and lncRNAs have both been well-concerned topics over the past decade and LGG is a highly heterogeneous molecular malignant tumor. In the present study, we initially identified a novel prognostic signature comprising nine FRlncRNAs to predict the long-term outcomes of patients with LGG. Further analysis showed that the risk score was also correlated with clinicopathological characteristics of LGG patients. Comprehensive analyses revealed that the risk score was highly associated with anti-tumor immune. These findings revealed that the novel risk score could be taken as a promising prognostic biomarker of LGG patients and ferroptosi might be a potential therapeutic target. Overall, 1637 (10.3%) FRlncRNAs in the TCGA dataset and 493 (11.5%) in the CGGA dataset were found to be associated with OS, indicating FRlncRNAs might play an extensive and important role in the outcome of LGG. The risk score exhibited better discrimination than the other models (age, gender, grade, and status of MGMT Methtlation, IDH mutation, 1p/19q codeletion), allowed the patients to be divided into two subgroups that exhibited quite divergent OS both in datasets of TCGA and CGGA (both P < 0.01). In addition, the current risk score was also identified as an independent risk factor of OS using multivariate Cox regression analysis both in the two datasets (both P < 0.001). All these findings above indicated that the novel risk score could be taken as a promising prognostic biomarker of LGG patients. The nine FRlncRNAs included five protective ones (TMEM254-AS1, SLC25A21-AS1, DGCR9, LINC00844, and LINC00641) and four risk ones (AGAP2-AS1, FAM181A-AS1, CRNDE, and PAXIP1-AS2). Previous studies have found that SLC25A21-AS1 can induce multidrug resistance in nasopharyngeal carcinoma through miR-324-3p/IL-6 [[134]26]. Meanwhile, DGCR9 expression is upregulated in gastric cancer and is involved in cell proliferation and migration and affects glucose metabolism [[135]27]. However, in the current study, these two lncRNAs were determined to be protective factors and deserve further validation. LINC00844 has been found to inhibit prostate cancer and hepatocellular carcinoma proliferation by regulating the expression of GSTP1 and NDRG1 [[136]28, [137]29], respectively. Yang et al. found that LINC00641 can suppress glioma progression through the miR-4262/NRGN axis, which provides a novel therapeutic target [[138]30]. Meanwhile, Yan et al and Luo et al reported that AGAP2-AS1 can promote the proliferation, migration, and invasion of glioma cells by regulating PTN, EZH2, and LSD1 [[139]31]. FAM181A-AS1 and CRNDE have been found to promote glioma development by enhancing ZRANB2 expression and inhibiting Bcl-2 and Wnt2 expression, respectively [[140]32, [141]33]. Considering there are no relevant studies on PAXIP1-AS2 and its similar origin lncRNAs, lncRNA PAXIP1-AS1 was found to promote glioma cell invasion and angiogenesis by upregulating the ETS1/KIF14 axis, the prognostic value of PAXIP1-AS2 was reliable [[142]34]. In summary, the nine FRlncRNAs we identified may also be considered as potential novel targets for the management of LGG. It is well known that LGG is highly heterogeneous and the existing molecular subtyping is inadequate to guide clinical practice [[143]2]. In the current study, we found that the risk score was quite correlated with clinicopathological characteristics of LGG patients, including age, WHO grading, TMB, and status of MGMT methylation status, IDH mutation status, and 1p/19q codeletion (all P < 0.01). And then, we found that the current risk score could also re-categorize subgroup patients stratified by the current molecular/gene classification system. Furthermore, patients with WHO grade II tumors and high-risk scores were found to have a parallel prognosis with patients with WHO grade III tumors and low-risk score; similar findings were observed in the classification system of 1p/19q codeletion status. These findings might explain why LGG patients of the same subtype often have different outcomes, and the current risk score could be taken as a biomarker to supplement the existing molecular classification system. Ferroptosis is not only associated with tumorigenesis and aggressive progression, but also associated with treatment response including immune therapy [[144]4, [145]35–[146]37]. To the best of our knowledge, all trials of immune checkpoint inhibitors to treat glioma have failed, but the mechanism remains unclear. Similar to FRGs in previous reports [[147]8, [148]10], the current risk score was also associated with immunity. Immune cells including macrophages, CD8+ T cells, and NK cells were found to be enriched in subgroup with the high-risk score using seven different evaluation tools (all P < 0.05), which indicated that the conclusion was robust. On the other hand, immune-related pathways such as antigen processing and presentation, the B-cell receptor signaling pathway, and the T-cell receptor signaling pathway were also activated in patients with high-risk scores, which suggested that inhibitors of these pathways may be considered as potential therapeutic targets [[149]38]. In addition, PD-1, PD-L1, and CTLA4 were all found in this study to be over-expressed in patients with high-risk scores, which provides insight into immune therapy of LGG and the screening the potential patients that may benefit from a particular treatment, which are key for a successful trial. This study had several limitations. First, the definition of LGG remains controversial, even though it has been widely adopted in bioinformatics-related studies [[150]39–[151]41]. Second, although the data from the TCGA and CGGA databases were adjusted before subsequent analyses, they were obtained from different batches. Third, the current risk score exhibited good calibration for the TCGA and CGGA datasets, but it lacked validation using external real-world data. Fourth, the mechanism of tumor aggression is highly complex and a single hallmark to predict OS is unlikely. Fifth, additional interesting findings between the current risk score and the immune system were observed, but future experiments are warranted. Conclusion In summary, we determined a risk score based on nine FRlncRNAs, which could not only be taken as a promising biomarker of prognosis of LGG, but also supplement the existing molecular classification systems. Our findings also provide clues for future research regarding therapeutic targets, including immune checkpoint inhibitors. However, the underlying mechanisms require further validation using in vitro and in vivo experiments. Supplementary Information [152]12672_2024_1587_MOESM1_ESM.tif^ (1.8MB, tif) Supplementary Material 1. Fig. S1 (a) LASSO coefficient profiles of the expression of the candidate FRlncRNAs. (b) Selection of the penalty parameter (Lambda) in the LASSO model. [153]12672_2024_1587_MOESM2_ESM.tif^ (4.5MB, tif) Supplementary Material 2. Fig. S2 Kaplan–Meier curves (a), risk plot distribution and survival status (b) of GSE108474 dataset. Kaplan–Meier curves (c), risk plot distribution and survival status (d) of GSE43378 dataset. [154]12672_2024_1587_MOESM3_ESM.tif^ (3.4MB, tif) Supplementary Material 3. Fig. S3 Univariate and multivariate Cox regression analysis of lower-grade glioma (LGG) prognosis in GSE108474 dataset (a) and GSE43378 (b) datasets. [155]12672_2024_1587_MOESM4_ESM.tif^ (1.9MB, tif) Supplementary Material 4. Fig. S4 Clinicopathological characteristics evaluation including age (a), gender (b), WHO grade (c), MGMT methylation status (d), IDH mutation status (e) and 1p/19q codeletion status (f) in CGGA dataset. *p < 0.05, **p < 0.01 and ***p < 0.001. [156]12672_2024_1587_MOESM5_ESM.tif^ (1.5MB, tif) Supplementary Material 5. Fig. S5 Patients in each subtype could also be divided into two different groups in terms of OS according to their risk score. WHO grading (a, f), MGMT methylation status (b, g), IDH mutation status (c, h), 1p/19q codeletion status (d, i), and TMB (e, j). [157]12672_2024_1587_MOESM6_ESM.tif^ (2MB, tif) Supplementary Material 6. Fig. S6 GO enrichment (a) and KEGG pathways (b) in the CGGA set. [158]12672_2024_1587_MOESM7_ESM.tif^ (9.4MB, tif) Supplementary Material 7. Fig. S7 Scatter plot and heat map showed the correlations among PI3K-Akt signaling pathway, risk score, ferroptosis, as well as the previously mentioned immune pathways (e.g. B-cell receptor signaling pathway, T-cell receptor signaling pathway), immune cell infiltration (e.g. macrophage cell and T cell CD4+), and immune checkpoint expression (e.g. PD-1, PD-L1, and CTLA4) in TCGA cohort. [159]12672_2024_1587_MOESM8_ESM.tif^ (9.9MB, tif) Supplementary Material 8. Fig. S8 Scatter plot and heat map showed the correlations among PI3K-Akt signaling pathway, risk score, ferroptosis, as well as the previously mentioned immune pathways (e.g. B-cell receptor signaling pathway, T-cell receptor signaling pathway), immune cell infiltration (e.g. macrophage cell and T cell CD4+), and immune checkpoint expression (e.g. PD-1, PD-L1, and CTLA4) in CGGA cohort. [160]12672_2024_1587_MOESM9_ESM.tif^ (8.5MB, tif) Supplementary Material 9. Fig. S9 Scatter plot and heat map showed the correlations among PI3K-Akt signaling pathway, risk score, ferroptosis, as well as the previously mentioned immune pathways (e.g. B-cell receptor signaling pathway, T-cell receptor signaling pathway), immune cell infiltration (e.g. macrophage cell and T cell CD4+), and immune checkpoint expression (e.g. PD-1, PD-L1, and CTLA4) in GSE108474 cohort. [161]12672_2024_1587_MOESM10_ESM.tif^ (5.8MB, tif) Supplementary Material 10. Fig. S10 (a) Heatmap representation of unsupervised clustering. (b) Kaplan-Meier curves of OS with two clusters. (c)Boxplot shows the distribution of risk score between C1 and C2. (d)Boxplot shows the distribution of PD-1 expression between C1 and C2. (e)Boxplot shows the distribution of PD-L1 expression between C1 and C2. (f)Boxplot shows the distribution of CTLA4 expression between C1 and C2. [162]12672_2024_1587_MOESM11_ESM.xlsx^ (38.1KB, xlsx) Supplementary Material 11. Table S1 A total of 382 ferroptosis-related genes. [163]12672_2024_1587_MOESM12_ESM.xlsx^ (2.9MB, xlsx) Supplementary Material 12. Table S2 A total of 1637 ferroptosis-related lncRNAs were identified by co-expression analysis. [164]12672_2024_1587_MOESM13_ESM.xlsx^ (28.2KB, xlsx) Supplementary Material 13. Table S3 The 81 intersected ferroptosis-related prognostic lncRNAs. [165]12672_2024_1587_MOESM14_ESM.xlsx^ (31KB, xlsx) Supplementary Material 14. Table S4 The risk score and TMB information in TCGA cohort. [166]12672_2024_1587_MOESM15_ESM.xlsx^ (15.7KB, xlsx) Supplementary Material 15. Table S5 The enriched pathways of the high-risk score groups in TCGA cohort. [167]12672_2024_1587_MOESM16_ESM.xlsx^ (567.7KB, xlsx) Supplementary Material 16. Table S6 The infiltration estimation for TCGA cohort in LGG. Acknowledgements