Abstract Background: Tertiary lymphoid structures (TLSs), organized clusters of immune cells within non-lymphoid tissues, significantly influence tumor progression and therapeutic response. However, their prognostic relevance and underlying regulatory mechanisms in clear cell renal cell carcinoma (ccRCC) remain insufficiently characterized. Methods: We integrated transcriptomic and clinical data from 928 ccRCC patients to construct a TLS-related prognostic RiskScore using machine learning algorithms. TLS maturation heterogeneity was characterized via immunohistochemistry and multiplex immunofluorescence analyses. The functional role of interferon regulatory factor 4 (IRF4), a key regulator within the TLS gene network, was investigated using in vitro assays. Single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics were employed to dissect the involvement of IRF4 in TLS formation and maturation. Results: The derived TLS-associated signature RiskScore, comprising CCL22, LOXL1, LIPA, ADAM8, and SAA1, effectively stratified patients into distinct prognostic groups and showed robust associations with clinical parameters, tumor microenvironment (TME) features, and predicted immunotherapy responses. Functional assays demonstrated that IRF4 significantly enhanced the malignant phenotype of 786-O and 769-P ccRCC cells. Clinically, elevated IRF4 expression independently predicted worse patient outcomes, characterized by a predominance of immature TLS phenotypes, reduced TLS density, and diminished CD8⁺ T cell infiltration. Mechanistically, scRNA-seq analyses revealed that active IRF4 signaling was predominantly confined to immature B cell states and was inversely associated with TLS maturation trajectories. Spatial transcriptomics further confirmed IRF4 enrichment within TLS regions, notably spatially segregated from high endothelial venules (HEVs) and mature TLS compartments. Conclusion: In conclusion, this study establishes a robust TLS-related prognostic signature for ccRCC and elucidates the mechanistic role of IRF4 in promoting TLS immaturity and immune dysfunction. By potentially recruiting immature B cells while impairing their maturation, IRF4 contributes to an ineffective anti-tumor immune landscape, offering a promising target for therapeutic intervention. Keywords: tertiary lymphoid structures (TLSs), clear cell renal cell carcinoma (ccRCC), tumor microenvironment (TME), interferon regulatory factor 4 (IRF4), single-cell RNA sequencing (scRNA-seq), spatial transcriptomics analysis Introduction Renal cell carcinoma (RCC) represents a significant cause of cancer mortality worldwide [55]^1^-[56]^3. In 2022, it was estimated that there were over 7.37×10^4 novel cases and 2.40×10^4 fatalities due to RCC [57]^4. RCC prevalence in China has increased over the past thirty years, and the rate of new diagnoses per 100,000 individuals has nearly tripled, rising from 1.16 in 1990 to 3.21 in 2019[58]^5. Clear cell renal cell carcinoma (ccRCC), the most frequent histological subtype, accounts for approximately 70% of all RCC cases [59]^6. Genetic alterations in key driver genes such as VHL, SETD2, PBRM1, and BAP1 are characteristic of ccRCC, contributing to genomic instability and impaired DNA repair mechanisms, thereby potentially promoting ccRCC tumorigenesis [60]^7. ccRCC is characterized by significant heterogeneity, leading to variable patient prognoses and highlighting the critical need for improved biomarkers beyond traditional clinicopathological features. In recent years, considerable research effort has focused on developing molecular signatures derived from high-throughput data to better stratify patients and predict outcomes. These efforts have explored various biological signatures of ccRCC. For instance, researchers have investigated signatures linked to specific cell death pathways [61]^8^,[62]^9, cancer stemness and non-coding RNAs [63]^10, tumor microenvironment (TME) and immune components [64]^11^,[65]^12. This landscape of recently proposed biomarkers underscores the diverse strategies being employed and sets the stage for introducing novel signatures, like the TLS-based one in this study, which may offer unique insights into the tumor's immune landscape and patient prognosis. Despite identifying numerous potential genomic biomarkers, the translation of these findings into routinely used tissue or blood-based molecular biomarkers for guiding clinical decision-making in ccRCC remains a significant challenge [66]^13. TME is an intricate and dynamic system composed of diverse cell types, including endothelial cells, fibroblasts, and immune cells [67]^14. The TME plays a critical role in tumor initiation, progression, and metastasis, and modulates therapeutic response [68]^15^,[69]^16, being actively shaped by components such as cancer-associated fibroblasts [70]^17. Furthermore, systemic host factors, such as the gut microbiota, and intrinsic microenvironmental conditions, such as hypoxia, are increasingly recognized as modulators of the TME [71]^18^,[72]^19. Recent research indicates that tumor-associated immunity is evident in tertiary lymphoid structures (TLSs) [73]^20. TLSs form in nonlymphoid tissues under pathological conditions, including cancer, rather than during physiological states [74]^21^,[75]^22. Structurally, TLSs are featured by a central cluster of CD20^+ B cells surrounded by CD3^+ T cells, similar to the lymphoid follicles in secondary lymphoid organs (SLOs) [76]^23. The T cell region within TLSs consists of CD4^+ T follicular helper (Tfh) cells, CD8^+ cytotoxic T lymphocytes, CD4^+ T helper 1 (TH1) cells, and regulatory T cells (Tregs) [77]^24^,[78]^25. Unlike SLOs, TLSs typically lack a distinct capsule, potentially facilitating their cellular constituents to interact directly with adjacent tissue [79]^26. TLSs are implicated in initiating and enhancing adaptive immune responses [80]^27^,[81]^28. T cells within TLSs interact with mature dendritic cells (DCs) and B cells, inducing T cell differentiation and the development of germinal centers (GCs) [82]^21. After B cells achieve complete maturation within TLSs, they become plasma cells that secrete high-affinity IgG and IgA, potentially increasing the tumor's response to immunotherapy [83]^29. TLSs are increasingly recognized as crucial organizing centers for anti-tumor immunity, influencing prognosis and therapeutic response across numerous malignancies [84]^30. Accumulating evidence across diverse malignancies, such as melanoma [85]^31, ovarian cancer [86]^32, non-small cell lung cancer [87]^33, bladder cancer [88]^34, and pancreatic cancer [89]^35, consistently links the presence, density, and particularly the maturation state of TLSs with patient prognosis and efficacy of immune checkpoint inhibitor (ICI) therapies. Indeed, understanding the broader TME's impact is crucial, as studies highlight its pivotal role in regulating intercellular communication, shaping treatment responses, and driving resistance to chemoradiation, targeted therapy, and immunotherapy [90]^36. However, the presence of TLSs in various tumors can differ significantly, which may lead to variable outcomes [91]^37. Factors influencing TLS maturation, such as local metabolic constraints [92]^38, and the specific cellular interactions within them, including T-cell activation levels [93]^35, may contribute to this heterogeneity. In ccRCC, the different localization and maturity of TLSs can lead to different prognoses, with tumor-proximal TLSs being more mature and exhibiting better prognoses [94]^39. Therefore, prognostic models for ccRCC incorporating TLS molecular markers hold potential clinical significance. Interferon regulatory factor 4 (IRF4) serves as one of the genetic markers for identifying TLSs in cancers, and it fulfills a complex role in immune regulation [95]^23. Increased IRF4 levels enhance the differentiation of CD4^+ CD25^low effector T (Teff) cells and reduce T follicular helper (Tfh) cell numbers [96]^40. Additionally, IRF4 directs effector regulatory T (Treg) cell differentiation and promotes immune suppression [97]^41. Upregulated IRF4^+ Treg cells within tumors are significantly linked to early tumor recurrence as well as worse disease-free survival (DFS) and overall survival (OS) [98]^42. Although IRF4 fosters proliferation and continuous differentiation of CD8^+ T cells [99]^43, persistently high IRF4 can lead to CD8^+ T cell exhaustion [100]^44. Specifically, recent studies confirm that IRF4 upregulation following T cell activation impedes human CD8 T cell effector function, while promoting cell proliferation and PD-1 expression, contributing to an exhausted phenotype in tumor-infiltrating lymphocytes (TILs) rather than activation alone [101]^45. Moreover, IRF4 induces M2-type macrophages [102]^46, contributing to an immunosuppressive TME. This process can be influenced by upstream signaling [103]^47 and by post-translational modifications [104]^48. Previous studies have elucidated IRF4's involvement across various tumor types. Anaplastic large cell lymphoma shows a dependency on IRF4 signaling, with MYC as a key target of IRF4[105]^49. Furthermore, mutations affecting IRF4-DNA binding can upregulate genes specific to human lymphomas [106]^50. IRF4 mRNA escalates from normal tissues to oral submucous fibrosis and oral squamous cell carcinoma (OSCC), paralleling immune infiltration [107]^51. Moreover, IRF4 acts as an oncogenic factor in human non-small cell lung cancer, partly through triggering the Notch-Akt pathway [108]^52. In multiple myeloma (MM), IRF4 is a critical transcription factor whose activity and cellular growth effects can be deregulated by the loss of the inhibitory protein BCL7A, which normally limits its DNA binding activity [109]^53. Due to its specific overexpression and role in mediating MM progression and survival, IRF4 is actively being pursued as a therapeutic target, with novel direct small molecule inhibitors that bind its DNA-binding domain being designed and synthesized [110]^54 and antisense oligonucleotide-based approaches being explored to silence its expression [111]^55. Nevertheless, the precise functions of IRF4 in ccRCC are yet to be elucidated. This study employed an integrative approach to elucidate the complex role of TLSs in ccRCC. We developed and validated a robust TLS-related signature capable of predicting patient prognosis, TME characteristics, and immunotherapy response. Then, since IRF4 is a key TLS-associated gene with unclear functions in ccRCC, we investigated its specific contribution to tumor progression and TLS maturation. By combining in vitro functional assays with single-cell and spatial transcriptomics analyses, we specifically sought to determine how IRF4 expression influences B cell dynamics within the TME, contributes to TLS heterogeneity, and ultimately impacts ccRCC progression, thereby identifying potential mechanisms driving immune evasion and revealing novel therapeutic vulnerabilities. Materials and Methods Raw Data Collection and Standardization Process This study compiled transcriptomic and clinical data for 763 ccRCC patients from several online databases, including the Cancer Genome Atlas (TCGA) database, clinical proteomic tumor analysis consortium (CPTAC) database, European Molecular Biology Laboratory (EMBL) database, and International Cancer Genome Consortium (ICGC) database, using datasets TCGA-KIRC, CPTAC-3, E-MTAB-3267, and RECA-EU. To ensure comparability and standardization, RNA sequencing data initially presented as fragments per kilobase of transcript per million mapped reads (FPKM) were converted to transcripts per million (TPM), followed by log2(TPM+1) transformation. Additionally, we mitigated the batch effect using the “ComBat” algorithm of the “sva” package. The study also incorporated genetic alteration data from the TCGA database, covering somatic mutations, copy number variations (CNVs), and tumor mutational burden (TMB). All patient consent and ethical approvals were appropriately secured in the original studies. Baseline clinical data for the patients were presented in [112]Table S1. Consensus Clustering Analysis Our study utilized a consensus clustering approach to divide ccRCC patients into separate subgroups according to TLS-related genes and differentially expressed genes (DEGs), respectively. We executed consensus hierarchical clustering and confirmed the ideal cluster quantity and the distribution of patients using the “ConsensusClusterPlus” package in R, performing 1000 repetitions for credibility assurance [113]^56. Correlation of Clinical Traits and Immune Landscape with Molecular Profiles Several clinical variables were taken into consideration, including patient age, gender, tumor grade, and Stage. Utilizing the “survival” and “survminer” packages in R software, we performed the Kaplan-Meier (K-M) analysis to compare prognostic outcomes [114]^57. Then, we estimated the prevalence of immune cells within samples by implementing the CIBERSORT algorithm [115]^58, and we pinpointed the immune cells infiltrating proportion through the single-sample gene set enrichment analysis (ssGSEA) algorithm [116]^59. Additionally, we calculated the ESTIMATE score for each ccRCC specimen using the ESTIMATE algorithm [117]^60, and compared immune checkpoints (ICPs) expression across subgroups. Functional and Pathway Enrichment Analysis We performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses with the “clusterProfiler” package [118]^61. Additionally, we utilized the KEGG gene set (c2.cp.kegg.v7.5.1) to execute gene set variation analysis (GSVA) to underscore functional discrepancies among clusters, based on the adjusted P value < 0.001 and the threshold of |log2-fold change (FC)| > 0.2[119]^62. Distinctive Expression and Development of the TLS-Related Predictive Signature The “limma” package in R software was utilized to pinpoint DEGs among different TLS-gene-related clusters, setting the selection threshold at |log2-FC| > 1.25 and the adjusted P value < 0.001[120]^63. We then devised a prognostic scoring system (RiskScore). To recognize prognosis-related genes, univariate Cox regression analysis was performed, considering DEGs expression levels and survival data. Subsequently, we used the least absolute shrinkage and selection operator (LASSO) technique and multivariate Cox regression analysis to create an optimal predictive model. RiskScore was calculated as follows: RiskScore = h0(t) × exp (expression of CCL22 × corresponding coefficient + expression of LOXL1 × corresponding coefficient + expression of LIPA × corresponding coefficient + expression of ADAM8 × corresponding coefficient + expression of SAA1 × corresponding coefficient). Patients were then categorized into low or high RiskGroups based on the median RiskScore. Clinical Relevance of the Predictive Signature and Establishment of a Prognostic Outcome Prediction Nomogram We compared clinical outcomes between the two RiskGroups by employing K-M analysis. We confirmed the predictive accuracy of the RiskScore system using a receiver operating characteristic (ROC) curve, created with the “survival ROC” package in R. Moreover, the prognostic relevance of the signature was investigated by categorizing ccRCC patients according to their clinical features. Integrating the RiskScore with clinical parameters, we formulated a nomogram for forecasting the 1-year, 3-year, and 5-year overall survival (OS) rates of ccRCC patients, utilizing the “rms”, “regplot”, and “survival” packages in R. To determine the accuracy and robustness of the nomogram, we conducted calibration curves, time-dependent ROC curves, and decision curve analysis (DCA). Forecasting the Treatment Response to ICI Therapy To forecast the response to immune checkpoint inhibitor (ICI) therapy in ccRCC patients, we utilized the tumor immune dysfunction and exclusion (TIDE) analysis. TIDE assesses two principal immune escape mechanisms: impaired T cell functionality and the inhibition of T cell infiltration with low cytotoxic T lymphocyte (CTL) counts [121]^64. Low TIDE score patients are anticipated to have a favorable response to immunotherapy. Furthermore, we corroborated the response to immunotherapy of the TLS-related signature by analyzing the David Liu cohort, which involved 121 patients with metastatic melanoma who received anti-PD-1 blocking agent treatment (nivolumab or pembrolizumab) [122]^65. We also validated the result with CheckMate 009 and 025 (CheckMate cohort), and 136 advanced ccRCC patients treated with nivolumab (anti-PD-1) [123]^66. Specimen Collection and H&E Staining In our study, we included a cohort of 60 patients diagnosed with ccRCC who were treated at the Department of Urology, Fudan University Shanghai Cancer Center (FUSCC, Shanghai, China). We also included clinical data for 105 ccRCC cases from the Department of Urology, FUSCC, between 2010 and 2023, to examine the relationship between IRF4 expression and prognosis. These patients underwent surgical procedures during which tissue samples were collected for further analysis. The study design and the collection and use of these tissue samples were carried out with strict adherence to the Declaration of Helsinki. The study was approved by the FUSCC Ethics Committee, and all patients provided informed permission. To assess the extent of lymphocytic infiltration, histopathological examinations were conducted. Hematoxylin and eosin (H&E) staining was conducted on slides of both ccRCC and adjacent normal tissues following reported protocols [124]^67, and they were carefully reviewed by two experienced pathologists independently. TLS Maturation Analysis with Multiplex Immunofluorescence (mIF) Staining Assays To further investigate the tumor microenvironment [125]^68^-[126]^70, we employed independent IHC staining on adjacent slides and multiplex immunofluorescence staining assay, conducted in collaboration with Shanghai KR Pharmtech, Inc., Ltd. (Shanghai, China). Specifically, we used the 7-color multiplex immunofluorescence kit (KR Pharmtech, Inc., Ltd. (Shanghai, China) to analyze the maturation of TLSs. For the staining process, we selected a panel of specific antibodies, namely CD8 (Abcam, ab178089, 1:100), CD20 (Dako, L26, IR604), CD21 (Abcam, ab227662, 1:100), CD23 (Abcam, ab315289,1:100), CK (Abcam, ab7753, 1:100), and PD-L1 (CST, E1L3N, 13 684S, 1:400) [127]^71^,[128]^72. The tissue slides obtained were first treated with Antibody Diluent Block buffer. Following this, the slides were incubated with the primary antibody for 40-60 minutes. Then, the slides were treated with a polymer horseradish peroxidase (HRP)-conjugated secondary antibody for 10 minutes. Next, the Fluorophore Working Solution was applied to the slides for 10 minutes, visualizing the complexes. The nuclei within the tissue slides were stained with 4',6-diamidino-2-phenylindole (DAPI). Finally, whole slide scans were conducted at 20× magnification using the KR-HT5 system (KR Pharmtech, Inc., Ltd. (Shanghai, China)). The acquired images were then analyzed using inForm 2.4.0 software. Images were classified into TLS maturation stages as follows: early TLS (E-TLS) depicted dense lymphocytic aggregates of mixed CD8^+T and B cells (CD8 and CD20 positive) without follicular DCs (FDCs) and GC (CD21 and CD23 signals negative); primary follicle-like TLS (PFL-TLS) showed dense lymphocytic aggregates with FDCs and absence of a GC (CD21 positive but without CD23 signals); secondary follicle-like TLS (SFL-TLS) were identified by both FDCs and a GC (CD21 and CD23 positive)[129]^39. TLS-positive samples in the study were categorized as follows: SFL-TLS positive, containing at least one instance of SFL-TLS; PFL-TLS positive, with at least one PFL-TLS occurrence but no SFL-TLS; E-TLS positive, exhibiting neither PFL-TLS nor SFL-TLS [130]^39^,[131]^73. Immunohistochemistry and Scoring Assays After identifying at least one instance of TLSs in H&E-stained slides, adjacent slides were subjected to immunohistochemical (IHC) staining, performed as previously described [132]^67, and IRF4 (ab315394, Abcam) was identified. In order to quantify the intensity of IRF4 staining, an immunohistochemistry scoring system was employed, named IHCscore. The staining number score was determined based on the percentage of cells showing positive staining: a score of 0 was assigned for negative staining, 1 for 1-25% staining, 2 for 26-50% staining, 3 for 51-75% staining, and 4 for 76-100% staining. The staining color scored 0 for negative, 1 for weak, 2 for medium, and 3 for strong staining. The final IHCscore was calculated by multiplying the staining number score by the color score [133]^74. Based on the median IHCscore, the samples were further categorized into high and low groups. Cell Culture and siRNA Transfection The human ccRCC cell lines 786-O and 769-P were acquired from the Type Culture Collection Cell Bank, Chinese Academy of Sciences. These cells were nurtured in RPMI 1640 medium, supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin. Cells were maintained in a 37°C incubator with 5% CO[2]. Cells were seeded in 10-cm dishes at 50% confluency one day before transfection. Transfection commenced when cell density approached 70%. Hieff Trans^® Liposomal Transfection Reagent (Yeasen, Shanghai, China) was used to prepare complexes with the negative control and IRF4 siRNA, according to the protocol from the manufacturer. After a 15-minute room temperature incubation, these complexes were introduced to the cells. We harvested the cells at 72 hours post-transfection for later analysis. Western Blotting Assay Cells were lysed using Cell lysis buffer for Western and IP (P0013, Beyotime) supplemented with protease inhibitors. Protein concentrations were determined using the BCA assay (P0009, Beyotime). Equal amounts of protein were separated by 10% SDS-PAGE and transferred onto PVDF membranes. Membranes were blocked with 5% non-fat milk in TBST for 1 hour at room temperature. Membranes were then incubated overnight at 4°C with primary antibodies diluted in blocking buffer. The following primary antibodies were used: IRF4 (11247-2-AP, Proteintech), CXCL13 (10927-1-AP, Proteintech), BCL6 (21187-1-AP, Proteintech), PD-L1 (28076-1-AP, Proteintech), and GAPDH (60004-1-Ig, Proteintech). After washing with TBST, membranes were incubated with horseradish peroxidase (HRP)-conjugated secondary antibodies for 1 hour at room temperature. The following secondary antibodies were used: Goat anti-Rabbit IgG (H+L) (SA00001-2, Proteintech) and Goat anti-Mouse IgG (H+L) (SA00001-1, Proteintech). Bands were visualized using an enhanced chemiluminescence (ECL) detection kit (36222ES, Yeason) and imaged using ChemiDoc Imaging System (Bio-Rad). Cellular Functional Experiments In Vitro The proliferation capability of cell lines was detected by colony formation assay and CCK-8 assay, respectively. For colony formation assay, we seeded 786-O or 769-P into 6-well plates with 1000 cells per well. After 14 days of culture in complete medium, the plates were gently washed twice with PBS, fixed with 4% paraformaldehyde for 20 minutes at room temperature, and then stained with 0.1% Crystal Violet solution for 20 minutes at room temperature. Following staining, plates were carefully washed with water to remove excess stain and allowed to air dry before being photographed on a luminous board. The colonies were quantified if they contained more than 50 cells. CCK-8 assay was conducted in 96-well plates, with each well containing 2000 cells in 100 µl of complete culture medium. Measurements were taken on day 1, 2, 3, 4, and 5 after incubation, respectively, by CCK-8 assay kit (Beyotime). After adding 10 µl CCK-8 reagent to each well, cell culture proceeded for an additional 2 hours shielded from light. Subsequently, absorbance was measured at the wavelength of 450 nm using Microplate Spectrophotometer (BioTek Instruments Inc.). Transwell assay was performed to assess cell invasion. Transwell chambers (Corning) coated with Matrigel (Corning) were loaded with 200 µl FBS-free culture medium containing 20,000 cells. Then, they were placed in 24-well plates, which contained 800 µl complete culture medium per well. After culturing for 24 hours, non-invaded cells remaining on the upper surface of the Transwell membrane were carefully removed using a cotton swab. The chambers were then washed with PBS, and the invaded cells on the lower surface were fixed with 4% paraformaldehyde for 20 minutes at room temperature. After fixation, the membranes were stained with 0.1% Crystal Violet solution for 20 minutes. Excess stain was removed by washing with PBS, and the membranes were allowed to air dry before being carefully excised, mounted on slides, and photographed. Wound healing assay was used to evaluate cell migration. Cells were seeded into 6-well plates and allowed to grow until confluent, and a scratch was then made to create a wound. After 24 hours of incubation in FBS-free culture medium, the wound gap area was photographed and measured. Flow Cytometry Assay Flow cytometry was performed to examine the apoptosis rate of cells. Cells were labeled with Annexin V-PE and 7-AAD (Multi Sciences) following the manufacturer's instructions. Then, detection was carried out using the LSRFortessa flow cytometer (BD Biosciences), and results were analyzed with CytExpert software. Single-Cell RNA Sequencing and Data Preprocessing In this study, we collected scRNA-seq data from 19 ccRCC samples sourced from [134]GSE207493. The single-cell data from the 10x platform were preprocessed following the standard protocol outlined in the R package Seurat [135]^75. Doublets were eliminated using DoubletFinder[136]^76, and low-quality cells were filtered out based on specific criteria: cells with fewer than 200 or more than 6000 detected genes, or those with a mitochondrial gene ratio exceeding 20%. Additionally, low-expression genes, defined as those expressed in fewer than 5 cells, were also excluded. Cell cycle status was assessed using the CellCycleScoring() function, and potential interference from cell cycle effects was mitigated through the regression algorithm implemented in ScaleData(). Following data normalization and standardization, Harmony was employed to correct for batch effects, and the top 2000 highly variable genes were selected. Dimensionality reduction was executed using Principal Component Analysis (PCA), with the first 15 principal components chosen for t-SNE visualization. Cell clustering was performed using the FindClusters() function, with a resolution parameter set to 0.5. Finally, cell subpopulations were manually annotated based on classical marker genes and references from