Abstract The intratumoral mycobiome plays a crucial role in the tumor microenvironment, but its impact on renal cell carcinoma (RCC) remains unclear. We collected and quantitatively profiled the intratumoral mycobiome data from 1044 patients with RCC across four international cohorts, of which 466 patients received immunotherapy. Patients were stratified into mycobiota ecology-depauperate and mycobiota ecology-flourishing (MEF) groups based on fungal abundance. The MEF group had worse prognosis, higher fungal diversity, down-regulated lipid catabolism, and exhausted CD8^+ T cells. We developed the intratumoral mycobiota signature and intratumoral mycobiota-related genes expression signature, which robustly predicted prognosis and immunotherapy outcomes in RCC and other cancers. Aspergillus tanneri was identified as a potential key fungal species influencing RCC prognosis. Our findings suggest that the intratumoral mycobiome suppresses lipid catabolism and induces T cell exhaustion in RCC. __________________________________________________________________ Intratumoral mycobiome modulates tumor microenvironment, affecting immunotherapy outcomes in renal cancer. INTRODUCTION Renal cell carcinoma (RCC) constitutes approximately 90% of all malignant kidney tumors, with clear cell renal cell carcinoma (ccRCC) being the most prevalent subtype, characterized by high metastatic potential and poor prognosis ([70]1–[71]4). The intratumoral microbiome, a crucial component of the tumor microenvironment, has been demonstrated to exist widely in various tumor tissues and is closely associated with cancer initiation, progression, metastasis, and treatment outcomes ([72]5–[73]11). Intratumoral microbiota can influence the host through multiple aspects, such as modulating immunogenicity and altering metabolic activities, which consequently affect the biological behavior of the tumor and patient prognosis ([74]12–[75]16). While previous studies have mainly focused on the bacterial microbiota, less attention has been given to the fungal microbiota (mycobiota), which is also an important element of the human microbiome. Existing evidence has confirmed that mycobiota may play a crucial role in the occurrence and progression of tumors ([76]17, [77]18). Therefore, further investigation into the role of intratumoral mycobiota in RCC is of great significance. Immune checkpoint inhibitors (ICIs) enhance the capacity of the immune system to attack cancer cells by blocking or inhibiting immune checkpoint molecules ([78]19, [79]20). However, only 10 to 20% of patients with RCC benefit from immunotherapy, and the complex mechanisms behind this are still being explored ([80]21, [81]22). Recent studies have suggested that the intratumoral microbiome may modulate the tumor immune microenvironment, subsequently influencing patients’ responses to immunotherapy ([82]14, [83]16, [84]23). In colorectal and gastrointestinal cancers, the intestinal mycobiota in patients has been demonstrated to be closely associated with prognosis, tumor progression, and response to immunotherapy ([85]24–[86]26). Studies of the intratumoral mycobiota have reported that it plays an important role in various types of tumors by modulating immune responses and regulating metabolic processes ([87]6, [88]27–[89]29). However, the impact of intratumoral mycobiota on patients with RCC and its role in immunotherapy remain to be elucidated. In this study, we integrated multiple clinical cohorts and multi-omics datasets to investigate the impact of the intratumoral mycobiome on the tumor microenvironment, prognosis, and immunotherapy outcomes in patients with RCC. RESULTS Prevalence of mycobiota in RCC and its association with prognosis To comprehensively investigate the ecology of intratumoral mycobiota in RCC, our study incorporated a total of 1084 patients with RCC from five distinct cohorts, encompassing 21 countries. The intratumoral mycobiome data of 522 patients with RCC were derived from the study by Narunsky-Haziza et al. ([90]18) [The Cancer Genome Atlas Kidney Renal Clear Cell Carcinoma (TCGA-KIRC)]. We collected data from 56 patients with primary RCC at West China Hospital of Sichuan University (WCH-RCC) ([91]30). Two large-scale, international, multicenter RCC immunotherapy clinical trials—IMmotion150 ([92]31) (n = 162) and IMmotion151 ([93]32) (n = 304)—were included. By processing raw RNA sequencing (RNA-seq) data through a microbial assignment pipeline, we obtained the intratumoral mycobiome profile of patients in the WCH-RCC, IMmotion150, and IMmotion151 cohorts (see Materials and Methods). Moreover, we retrospectively collected tumor tissues from a cohort of 40 patients with RCC at Shanghai Changhai Hospital (CHH-RCC). This cohort was used to corroborate the impact of intratumoral mycobiota on the host immune microenvironment. To further expand our research, we included a total of 2338 patients who received ICI therapy from 19 cohorts encompassing different tumor types, including non–small cell lung cancer (NSCLC), metastatic urothelial carcinoma (mUC), melanoma, and so on ([94]Fig. 1, A and B, and table S1) ([95]33–[96]50). Fig. 1. Overall study design. [97]Fig. 1. [98]Open in a new tab (A) Basic information of all cohorts included in this study. The world map showed the countries or regions of origin for the RCC cohorts included in the relevant studies. (B) Study flowchart. NSCLC, non–small cell lung cancer; mUC, metastatic urothelial cancer; CRC, colorectal cancer; RCC, renal cell carcinoma; PDAC, pancreatic ductal adenocarcinoma. Among the 35 cancer types included in TCGA, RCC exhibited a higher proportion of intratumoral fungal reads relative to total reads compared to other cancers, with marked heterogeneity across patients ([99]18). In our study, we included 522 patients with RCC from TCGA, all of which exhibited the presence of at least one fungal species. Ascomycota and Basidiomycota constituted the dominant fungal phyla in RCC (fig. S1). We characterized the intratumoral mycobiota landscape of TCGA-KIRC patients at the class level ([100]Fig. 2A). Notably, Dothideomycetes and Tremellomycetes were predominant in more than three-fifths (327 of 522) of the samples, whereas Sordariomycetes dominated approximately one-sixth (88 of 522). In nearly one-sixth (79 of 522) of the samples, Eurotiomycetes and Malasseziomycetes were more common. These findings suggested substantial heterogeneity in the intratumoral mycobiota composition among different patients with RCC. To validate the presence of mycobiota in RCC tissues, we performed fluorescence in situ hybridization (FISH) staining on tissue sections from 40 patients with RCC in the CHH-RCC cohort using fungal-specific 28S rRNA probes. All these samples exhibited positive 28S probe fluorescence of varying intensities, further confirming the widespread presence of mycobiota in the tumor tissues of patients with RCC ([101]Fig. 2B). Fig. 2. Intratumoral mycobiota landscape in RCC and group construction. [102]Fig. 2. [103]Open in a new tab (A) Composition of intratumoral fungi (class level) and clinical information of TCGA-KIRC patients. From inner to outer, the mapped information includes OS, gender, age, AJCC stage, T stage, N stage, M stage, and the relative abundance of intratumoral fungi at the class level in each sample. (B) Fluorescence in situ hybridization (FISH) showing the presence of fungi in tumor tissues of patients with RCC. Scale bar, 100 μm (10×) and 20 μm (40×). (C and D) Consensus clustering based on the intratumoral mycobiota abundance in TCGA-KIRC. (C) Consensus score matrix for all samples when k = 2. (D) Consensus matrix CDF curves for different k values. (E) Kaplan-Meier curves of OS for the two groups (named group A and group B) obtained based on consensus clustering [P = 2.1 × 10^−4, HR (95% CI) =1.78 (1.31 to 2.42)]. (F) Differences in various clinical characteristics between group A and group B, with significance tested using the chi-square test. We further explored the heterogeneity and clinical implications of the intratumoral mycobiome in patients with RCC. The optimal number of clusters (k = 2) was determined based on the consensus score matrix and cumulative distribution function (CDF) curve ([104]Fig. 2, C and D). Using consensus clustering based on the intratumoral mycobiota profiles, TCGA-KIRC patients were stratified into two clusters (group A and group B). Patients in group B displayed significantly poorer prognosis compared to group A [P = 2.1 × 10^−4, hazard ratio (HR) =1.78, 95% confidence interval (CI) =1.31 to 2.42, [105]Fig. 2E]. In addition, group B comprised a higher proportion of patients with advanced American Joint Committee on Cancer (AJCC) stages (P = 0.003) and T stages (P = 0.009), as well as a higher proportion of patients with metastasis (M stage: P = 0.033). However, there was no difference in age between two groups (P = 0.432; [106]Fig. 2F). Higher fungal diversity and complexity in the poorer prognosis group The multiple taxonomic levels of fungi exhibited heterogeneity between group A and group B ([107]Fig. 3, A and B, and fig. S2, A and B). The alpha diversity analysis revealed significantly higher intratumoral fungal richness and diversity in group B, compared to group A (P < 0.001, [108]Fig. 3, C to E, and fig. S2C). Beta diversity analysis revealed distinct fungal communities between group A and group B [permutational multivariate analysis of variance (PERMANOVA) P < 0.001, [109]Fig. 3F]. Linear discriminant analysis effect size (LEfSe) was used to further identify the significantly differed fungi between group A and group B [linear discriminant analysis (LDA) score > 2, P.adj < 0.05]. At the phylum level, Mucoromycota and Microsporidia were found to be significantly enriched in group B. Regarding the class level, 16 taxa, including Eurotiomycetes, Sordariomycetes, and Malasseziomycetes, exhibited higher abundance in group B, while Xylonomycetes were more enriched in group A. In terms of the order level, a total of 32 taxa, including Hypocreales and Eurotiales, revealed significantly higher abundance in group B. In contrast, only Xylonales, Eremomycetales, and Dothideales were enriched in group A ([110]Fig. 3G). As for the family and genus levels, 53 and 88 taxa, respectively, were significantly enriched in group B, while only 2 and 2 taxa were enriched in group A (table S2). At each taxonomic level, group B exhibited a greater number of enriched taxa. Furthermore, fungal co-occurrence network analysis revealed that the network complexity and the degree of fungal interactions were markedly higher in group B, with higher number of nodes, edges, and average degree ([111]Fig. 3, H and I, and table S3). The dominant fungal genera showed differences between the co-occurrence networks of group A and group B. In group A, the predominant taxa were Fusarium (27.2%) and Malassezia (16.3%), while in group B, the top taxa were Aspergillus (20.7%) and Fusarium (6.8%). Notably, Aspergillus and Fusarium occupied relatively critical positions in the fungal communities of both groups. Together, these results suggested that the intratumoral fungal community in group B manifested a more intricate and interconnected structure compared to that of group A. Fig. 3. Heterogeneity of fungal communities between groups. [112]Fig. 3. [113]Open in a new tab (A and B) Relative abundance of fungi at different taxonomic levels in group A and group B, showing only the top eight taxa. (A) Family level; (B) genus level. (C to E) Comparison of fungal alpha diversity between group A and group B using three indices: Shannon, Simpson, and Chao 1. (F) Comparison of fungal community differences between group A and group B based on beta diversity using principal coordinate analysis (PCoA) with Bray-Curtis distance, statistically tested using PERMANOVA. (G) Identification of fungi with the abundance differences between group A and group B using LEfSe analysis. The bar plot showed the results at the phylum, class, and order levels (LDA score > 2, FDR <0.05). (H and I) Fungal community co-occurrence networks and network topology parameters for group A and group B. The nodes implied individual fungi, and edges represented significant Spearman correlations between fungi (|ρ| > 0.80, FDR < 0.05). The nodes and edges of the networks were colored according to fungi (genus level), with the top eight most abundant fungi assigned different colors and the remaining fungi colored gray. The blue edges of the network represented the positive correlations between fungi. The thickness of the edges was the weight of the correlation. (H) Interaction network for group A (node: 21, edges: 46, modularity: 0.23, average degree: 4.38). (I) Interaction network for group B (node: 120, edges: 1396, modularity: 0.28, average degree: 23.27). (J) Group definition based on the features of fungal communities. LEfSe, linear discriminant analysis effect size; LDA, linear discriminant analysis; PERMANOVA, permutational multivariate analysis of variance. ***P < 0.001. Collectively, the fungal communities demonstrated marked distinctions between group A and group B. Multiple community feature analyses revealed that the intratumoral fungal community in group B had greater diversity and complexity than that of group A. To more precisely describe the distinct community attributes of the two groups, we designated group A as the mycobiota ecology-depauperate (MED) group, while group B was termed the mycobiota ecology-flourishing (MEF) group ([114]Fig. 3J). Down-regulation of lipid catabolism pathway in the MEF group To elucidate the mechanisms by which the intratumoral mycobiota influences the host microenvironment, clinical outcomes, and tumor characteristics, we investigated the differences in host biology between the MEF and MED groups. Differential expression analysis identified 110 up-regulated and 276 down-regulated genes in the MEF group compared to the MED group [|logFC| > 1, false discovery rate (FDR) < 0.05, fig. S3A]. These differentially expressed genes (DEGs) were primarily enriched in metabolic and immunological pathways, including lipid metabolism, amino acid metabolism, and interleukin-related signaling ([115]Fig. 4, A and B). In addition, we assessed the somatic mutation landscape in the MEF and MED groups, identifying 24 genes with significantly different mutation frequencies (P < 0.05, fig. S3B). The three genes exhibiting the most significant differences were GRIN2B, SPHKAP, and LAMA2, all of which have been previously implicated in tumor progression ([116]51–[117]53). Fig. 4. Unique metabolic characteristics and immune microenvironment between different groups. [118]Fig. 4. [119]Open in a new tab (A and B) Metabolic and immune-related pathways obtained by ORA based on DEGs between groups (different ribbons represented different sources of gene sets). Pink, GO database; light blue, KEGG database; lime green, Reactome database; orange, WikiPathways; purple, Hallmark database. (C) Lipid catabolism pathways significantly down-regulated in the MEF group compared to the MED group in GSEA. (D) Spearman correlation between lipid catabolism pathways and differentially abundant fungi (genus level) between MED and MEF groups. Lipid catabolism pathways were obtained from GSEA results (FDR < 0.05), and differentially abundant fungi were obtained from LEfSe results (LDA score > 2, FDR < 0.05). The size of the bubbles represented the level of correlation. The color of the bubbles indicated the FDR level: blue, negative correlation; red, positive correlation. (E) Proportions of major cells in the immune microenvironment of MED and MEF groups obtained using the xCell. (F) Proportions of CD8^+ T cells in MED and MEF groups based on CIBERSORT, EPIC, TIMER, and quanTIseq. (G) Proportions of CD8^+ T cell subtypes within MED and MEF groups obtained using gene sets provided by Zheng et al. ([120]59). Tn, naïve T cells; Tem, effector memory T cells; Temra, terminally differentiated effector memory or effector; KIR, killer cell immunoglobulin-like receptors; Trm, tissue-resident memory T cells; Tex, exhausted T cells; ISG, interferon-stimulated genes. (H) Scores of terminal Tex in MED and MEF groups. (I) Expression levels of exhausted CD8^+ T cell–related markers (CTLA-4, LAG-3, and PDCD1) in MED and MEF groups. (J) Expression levels of immunosuppressive genes IDO1, SLAMF7, and VEGFA in MED and MEF groups. ORA, overrepresentation analysis; DEGs, differentially expressed genes; GSEA, gene set enrichment analysis; NES: normalized enrichment score; TME: tumor microenvironment. *P < 0.05, **P < 0.01. Gene set enrichment analysis (GSEA) showed significant down-regulation of multiple lipid metabolism pathways in the MEF group compared to the MED group, including lipid catabolism, neutral lipid catabolism, and glycerolipid catabolism ([121]Fig. 4C and fig. S3C). Furthermore, the peroxisome proliferator-activated receptor (PPAR) pathway, closely linked to lipid metabolic activity ([122]54), was also down-regulated in the MEF group (fig. S3D). These findings denoted that the metabolic activity of patients in the MEF group underwent substantial changes, potentially further leading to the accumulation of lipid accumulation within the tumor microenvironment. The association between lipid accumulation in RCC and poor prognosis has been well documented in previous studies ([123]55, [124]56). To gain further insights, we evaluated the specific changes in lipid catabolism pathways among the different groups. The expression patterns of key metabolic genes were found to be in concordance with the observed pathway changes. Of particular note, CPT1A, members of the acyl-CoA dehydrogenase family, and PPARs exhibited lower expression levels in the MEF group (fig. S3E). To explore the potential relationship between intratumoral fungi and lipid metabolism pathways, we performed a correlation analysis between the differentially abundant fungal genera (LDA score > 2, FDR < 0.05) and lipid metabolism pathways. We observed that some fungal genera exhibited significant correlations with multiple lipid metabolism pathways (FDR < 0.05). Among these, Coccidioides and Trichoderma exhibited robust negative correlations with several lipid metabolism pathways. In contrast, Anthracocystis and Coniophora displayed positive correlations with multiple lipid metabolism pathways ([125]Fig. 4D). Elevated exhausted CD8^+ T cell infiltration in the MEF group Lipids play an important role in modulating immune responses to tumor and their accumulation within the microenvironment inducing dysfunction of CD8^+ tumor-infiltrating lymphocytes ([126]57, [127]58). The immune infiltration analysis using xCell showed a higher proportion of CD8^+ T cells in the MEF group compared to the MED group ([128]Fig. 4E). To verify the composition of CD8^+ T cells across different groups, we integrated four immune infiltration algorithms: Cell-type Identification By Estimating Relative Subsets Of RNA Transcripts (CIBERSORT), Estimating the Proportion of Immune and Cancer cells (EPIC), Tumor IMmune Estimation Resource (TIMER), and quantify the fractions of ten immune cell types from bulk RNA-sequencing data (quanTIseq). All algorithms confirmed a relatively higher proportion of CD8^+ T cells in the MEF group, with quanTIseq showing statistical significance (P < 0.05, [129]Fig. 4F). To further investigate the immune microenvironment characteristics, we calculated the composition of different CD8^+ T cell subtypes in the two groups ([130]59). The results exhibited significantly higher proportions of terminal and TCF7^+ exhausted CD8^+ T (Tex) cells in the MEF group ([131]Fig. 4G). We also observed higher proportions of GZMK^+ effector memory T (Tem) cells and ZNF683^+CXCR6^+ tissue-resident memory T (Trm) cells in the MEF group (P < 0.01). They participate in and dominate the two pathways from naive T cells to terminal Tex cells, playing a critical role in the development trajectory of T cell exhaustion ([132]59). We further evaluated the exhaustion scores and Tex subtypes of different groups ([133]60, [134]61). The results suggested a significantly higher exhaustion score in the MEF group, with high infiltration of all Tex subtypes ([135]Fig. 4H and fig. S3F). Although the overall regulatory T (Treg) cell levels did not present substantial differences between groups (fig. S3G), both TNFRSF9^+ Treg and ISG^+ Treg subtypes had higher proportions in the MEF group (fig. S3H). Among the Tex-related markers, the expression levels of cytotoxic T-lymphocyte antigen 4 (CTLA-4) and Lymphocyte-activation gene 3 (LAG-3) were significantly higher in the MEF group (P < 0.05, [136]Fig. 4I). Meanwhile, the PDCD1 was also relatively up-regulated, although not reaching statistical significance (P = 0.070, [137]Fig. 4I). Furthermore, we found significantly higher expression of indoleamine 2,3-dioxygenase 1 (IDO1), signaling lymphocytic activation molecule family member 7 (SLAMF7), and vascular endothelial growth factor A (VEGFA) in the MEF group (P < 0.05, [138]Fig. 4J). These genes have been reported to play a crucial role in inducing T cell exhaustion ([139]62–[140]64). Intratumoral mycobiota signature predicted prognosis and immunotherapy response in patients with RCC To evaluate the association between the fungal abundance characteristics and patient prognosis, we developed an integrated machine learning (ML) framework. This framework was used to develop an intratumoral mycobiota signature (IMS) model associated with overall survival (OS). In TCGA-KIRC patients, a total of 224 fungal species were detected. We eliminated low-abundance species (abundance of 0 in more than 80% of samples) to reduce excessive noise in the model. Subsequently, fungi significantly associated with OS were screened using a univariate Cox regression model, and the abundances of the 40 screened fungi were input into the ML framework ([141]Fig. 5A). We constructed 100 different ML models and calculated the mean C-index of each model across all datasets (fig. S4A). The model with the highest mean C-index was selected for further investigation. The results showed that the combination of random survival forest (RSF) and bidirectional stepwise Cox regression [Stepcox (both)] has the optimal performance (mean C-index = 0.613, [142]Fig. 5B). In this ML algorithm combination, the RSF model evaluated and selected important variables based on the principle of minimum tree depth, obtaining 23 important species. Further application of Stepcox regression constructed the IMS model, which included seven fungal species ([143]Fig. 5C). Fig. 5. Construction of IMS to predict prognosis and immunotherapy response in patients with RCC through integrated machine learning. [144]Fig. 5. [145]Open in a new tab (A) Screening process for selecting fungi as input for machine learning model training. (B) Machine learning based on 40 fungi, establishing 100 predictive models and calculating the C-index of each model in all datasets (only the first 10 models were shown here; the complete result can be found in fig. S4A). (C) Information on the seven fungi in IMS. The left side showed the model coefficients of the fungi; the right side showed the HR and 95% CI of each fungus in TCGA-KIRC. (D) KM curve of OS obtained by applying IMS scores in TCGA-KIRC [P = 1.6 × 10^−10; HR (95% CI) = 2.73 (1.98 to 3.76)]. (E) KM curve of OS obtained by applying IMS scores in WCH-RCC [P = 0.016; HR (95% CI) =4.25 (1.18 to 15.3)]. (F) ROC curve for OS prediction based on IMS scores in TCGA-KIRC (AUC = 0.682, 95% CI: 0.633 to 0.731). (G) ROC curve for OS prediction based on IMS scores in WCH-RCC (AUC = 0.671, 95% CI: 0.500 to 0.842). (H) PFS curve obtained by applying IMS scores in the IMmotion150 cohort of patients receiving immunotherapy [P = 0.0095, HR (95% CI) =1.96 (1.17 to 3.29)]. (I) PFS curve obtained by applying IMS scores in the IMmotion151 cohort of patients receiving immunotherapy [P = 0.018, HR (95% CI) =1.42 (1.06 to 1.90)]. (J) Clinical response rates to immunotherapy [complete response (CR)/partial response (PR) and stable disease (SD)/progressive disease (PD)] in the high and low IMS score groups of the IMmotion150 cohort, with significance tested using the chi-square test. (K) Clinical response rates to immunotherapy (CR/PR/SD/PD) in the high and low IMS score groups of the IMmotion151 cohort, with significance tested using the chi-square test. We divided patients with RCC into high-score (IMS-High) and low-score (IMS-Low) groups based on their IMS scores. Patients in the IMS-High group had significantly worse OS compared to the IMS-Low group in both the TCGA-KIRC cohort (training dataset) and the WCH-RCC cohort (validation dataset) [TCGA-KIRC: P = 1.6 × 10^−10; HR (95% CI) = 2.73 (1.98 to 3.76); WCH-RCC: P = 0.016; HR (95% CI) =4.25 (1.18 to 15.3); [146]Fig. 5, D and E]. The receiver operating characteristic (ROC) curves showed that the area under the curve (AUC) of IMS scores was 0.682 (95% CI: 0.633 to 0.731; [147]Fig. 5F) in TCGA-KIRC and 0.671 (95% CI: 0.500 to 0.842; [148]Fig. 5G) in WCH-RCC. Moreover, the multivariate Cox regression model indicated that IMS remained a significant prognostic factor after adjusting for clinical characteristics including age, AJCC stage, and TNM stage (P < 0.05, fig. S4, B and C). The above evidence substantiated the reliability and robustness of IMS in predicting the prognosis of patients with RCC. Previous studies have suggested that the intratumoral mycobiome is closely associated with the tumor immune microenvironment and may influence the efficacy of ICIs in treated patients ([149]28, [150]65). Accordingly, we further assessed the relationship between IMS and the prognosis of patients receiving immunotherapy in two international multicenter RCC cohorts (IMmotion150 and IMmotion151). The IMS-High group manifested significantly worse progression-free survival (PFS) compared to the IMS-Low group [IMmotion150: P = 0.0095, HR (95% CI) =1.96 (1.17 to 3.29); IMmotion151: P = 0.018, HR (95% CI) =1.42 (1.06 to 1.90), [151]Fig. 5, H and I]. The IMS-High group had a significantly lower response rate to immunotherapy (IMmotion150: P = 0.044; IMmotion151: P = 0.003, [152]Fig. 5, J and K). Aspergillus tanneri as the key fungus influencing RCC prognosis We carried out an in-depth analysis of the fungi in IMS. To identify the key species, we integrated the results from LEfSe, survival analysis, and ML through the framework of “rationality-congruence assessment” (RCA) (see Materials and Methods, fig. S5A). We performed a multidimensional interpretation of the fungi to select the key species with consistent performance through the framework. Among the seven fungi involved in the IMS, only Aspergillus tanneri achieved an RCA score of 6 (fig. S5B). The feature importance analysis also revealed that the importance of A. tanneri in the IMS model was higher than the other species ([153]Fig. 5C). Consequently, we considered A. tanneri as the key fungus and executed further analyses. In the TCGA-KIRC cohort, A. tanneri was detected in nearly 80% (415/522) of tumor tissues. It showed a significant positive correlation with the alpha diversity of the fungal community (Shannon, Simpson, ACE, and Chao1: P < 0.001, fig. S6A). Survival analysis showed that the high-abundance group of A. tanneri had a significantly worse OS [P = 2.2 × 10^−8, HR (95% CI) = 2.31 (1.71 to 3.13), [154]Fig. 6A]. In the high–A. tanneri abundance group, the tumor progression was more advanced as demonstrated by higher AJCC stage, T stage, and M stage (fig. S6B). Moreover, we noticed that the patients in the high–A. tanneri abundance group, the IMS-High group, and the MEF group had a high degree of overlap ([155]Fig. 6B), all showing poorer prognosis. We further confirmed the presence of A. tanneri in tumor tissues within the CHH-RCC cohort through quantitative polymerase chain reaction (qPCR) analysis (fig. S6C). The above results indicated that A. tanneri had a notable impact on the prognosis of patients with RCC. Fig. 6. A. tanneri as a key fungus influencing prognosis in patients with RCC. [156]Fig. 6. [157]Open in a new tab (A) KM curve of OS obtained by grouping TCGA-KIRC patients into high- and low-abundance groups based on A. tanneri abundance [P = 2.2 × 10^−8, HR (95% CI) = 2.31 (1.71 to 3.13)]. (B) Sankey diagram showed the distribution of samples in TCGA-KIRC from consensus clustering groups based on mycobiota abundance to high and low IMS score groups obtained from machine learning, and then to high and low A. tanneri abundance groups. MED and MEF groups obtained from consensus clustering; IMS: High and low score groups obtained based on IMS from machine learning; A. tanneri abundance: Groups obtained based on high and low A. tanneri abundance. (C) Spearman correlation between the seven fungi in IMS and lipid catabolism pathways. Lipid catabolism pathways were selected from GSEA results (FDR < 0.05). The size of the bubbles represented the level of correlation. The color of the bubbles indicated the FDR level: blue, negative correlation; red, positive correlation. (D) Proportions of CD8^+ T cells between high and low A. tanneri abundance groups obtained from five algorithms (TCGA-KIRC). (E) Expression levels of exhausted CD8^+ T cell–related markers (CTLA-4, LAG-3, and PDCD1) in high and low A. tanneri abundance groups. (F and G) Representative images of A. tanneri FISH staining and multiplex immunofluorescence in the CHH-RCC cohort. FISH: A. tanneri (red); DAPI (blue). Scale bar, 80 μm (10×) and 20 μm (40×). Multiplex immunofluorescence: positive cells detected, CD8 (red), PD-1 (green), LAG-3 (purple), CTLA-4 (orange), and DAPI (blue). Scale bar, 80 μm (10×) and 20 μm (40×). (H to J) Spearman correlation scatter plot between A. tanneri and PD-1^+ CD8^+ T cells (H), LAG-3^+ CD8^+ T cells (I), and CTLA-4^+ CD8^+ T cells (J) in the CHH-RCC cohort. *P < 0.05; **P < 0.01. The correlation analysis showed that A. tanneri was significantly negatively correlated with lipid catabolism pathways (FDR < 0.01, [158]Fig. 6C). In addition, A. tanneri had a negative correlation with the PPAR pathway (P = 0.002, ρ = −0.136, fig. S6D). Notably, among all the fungi found so far, the A. tanneri NIH1004 strain has been reported to have the strongest specialized metabolic capacity ([159]66). Together, there may be a complex and close cross-talk between A. tanneri and host metabolic activities. To further explore the potential impact mechanism of A. tanneri on the host, we compared the composition of the immune microenvironment between the high- and low-abundance group of A. tanneri. Similar to the MEF group, the proportion of CD8^+ T cells was higher in the high–A. tanneri abundance group with worse prognosis (P < 0.05, fig. S6E). The multiple immune infiltration algorithms provided consistent evidence ([160]Fig. 6D). Meanwhile, we found that Treg cells showed significantly higher infiltration in the high–A. tanneri abundance group (fig. S6E). In terms of the expression of Tex-related markers, CTLA-4, LAG-3, and PDCD1 were up-regulated in the high–A. tanneri abundance group (CTLA-4: P = 0.038, LAG-3: P = 0.004, PDCD1: P = 0.050; [161]Fig. 6E). In patients with CHH-RCC, FISH revealed the widespread presence of A. tanneri in tumor tissues ([162]Fig. 6, F and G). To confirm the association between A. tanneri and T cell exhaustion, we performed a correlation analysis on the results of FISH and multiplex immunofluorescence. We found that A. tanneri abundance exhibited a significant positive correlation with PD-1 and LAG-3 (PD-1: ρ = 0.423, P = 0.007; LAG-3: ρ = 0.351, P = 0.027; [163]Fig. 6, H and I). However, no notable correlation was observed between A. tanneri and CTLA-4 (P = 0.945, [164]Fig. 6J). These results suggested that A. tanneri may influence patient prognosis by modulating the immune microenvironment. Intratumoral mycobiota-related gene expression signature predicted prognosis and immunotherapy response In this study, we found a substantial association between the intratumoral mycobiota and patient prognosis in patients with RCC. Considering that the interaction between fungi and the host is generally reflected in gene expression patterns, we investigated the gene expression signatures related to intratumoral mycobiota. An ML prognostic model was constructed based on these signatures. Three distinct gene sets were defined to screen for gene expression features associated with intratumoral mycobiota. The first one included genes significantly correlated with at least one fungal species that had differential abundance between the MEF and MED groups (FDR < 0.05). The second gene set consisted of DEGs between the MEF and MED groups (|logFC| > 1, FDR < 0.05). The third set encompassed genes significantly associated with prognosis (univariate Cox regression model: P < 0.05). By taking the intersection of the three sets, we obtained a gene collection associated with the characteristics of the intratumoral mycobiome (n = 69, [165]Fig. 7A). On the basis of these 69 genes and through the integration of ML algorithms, we constructed the intratumoral mycobiota-related genes expression signature (IMRGES). Each model was fitted in the training set TCGA-KIRC and the validation set WCH-RCC, and the mean C-index of the models was calculated ([166]Fig. 7B and fig. S7A). Among all ML models, the combination of bidirectional stepwise Cox regression and RSF exhibited the best performance with the highest mean C-index (mean C-index = 0.925). Eventually, this combination was used to construct the IMRGES model, which included 23 genes ([167]Fig. 7C). Fig. 7. Construction of IMRGES through integrated machine learning based on intratumoral fungi-related gene sets. [168]Fig. 7. [169]Open in a new tab (A) Venn diagram showing the intersection of three conditional gene sets (n = 69). Fungi-related genes: genes correlated with at least one fungal species that had differential abundance between the MEF and MED groups (FDR < 0.05); prognostic genes: genes that can independently predict patient prognosis (Cox proportional hazards test P < 0.05); DEGs: differentially expressed genes between MED and MEF groups (|logFC| > 1, FDR < 0.05). (B) Machine learning based on 69 fungi-related genes, establishing 92 predictive models and calculating the C-index of each model in all datasets (only the first 15 models were shown here; the complete result can be found in fig. S7A). (C) Importance index of the 23 genes in the selected IMRGES. (D) KM curve of OS for IMRGES scores in the TCGA-KIRC cohort [P = 2.4 × 10^−120; HR (95% CI) =52 (31 to 86)]. (E) KM curve of OS for IMRGES scores in the WCH-RCC cohort [P = 2.8 × 10^−7; HR (95% CI) =16 (4 to 66)]. (F) KM curve of PFS for IMRGES scores in the IMmotion150 cohort of patients receiving immunotherapy [P = 0.039, HR (95% CI) =1.55 (1.02 to 2.34)]. (G) KM curve of PFS for IMRGES scores in the IMmotion151 cohort of patients receiving immunotherapy [P = 0.012, HR (95% CI) =1.52 (1.10 to 2.11)]. (H) Clinical response rates to immunotherapy (CR/PR/SD/PD) in the high and low IMRGES score groups of the IMmotion150 and IMmotion151 cohorts, with significance tested using the chi-square test. (I) Forest plot showing the results of a meta-analysis of the impact of IMRGES on OS in patients with different cancers at the pan-cancer level. The squares represented the study-specific HRs. The horizontal lines implied the 95% CI. The diamond indicated the summary HR and its corresponding 95% CI. The dashed vertical line represented the summary HR. According to the IMRGES, patients were divided into a high–IMRGES score group (IMRGES-High) and a low–IMRGES score group (IMRGES-Low). In the training set TCGA-KIRC and the validation set WCH-RCC, the IMRGES-High group demonstrated significantly worse OS [TGCA-KIRC: P = 2.4 × 10^−120; HR (95% CI) =52 (31 to 87); WCH-RCC: P = 2.8 × 10^−7; HR (95% CI) =16 (4 to 66); [170]Fig. 7, D and E]. The ROC curve showed that the AUC of the IMRGES score was 0.952 (95% CI = 0.935 to 0.970; fig. S7B) in TCGA-KIRC and 0.869 (95% CI = 0.744 to 0.994; fig. S7C) in WCH-RCC. In addition, multivariate Cox regression analysis revealed that IMRGES remained significant (P < 0.05, fig. S7, D and E) after adjusting for clinical characteristics such as age, AJCC stage, and TNM stage, indicating that IMRGES has an independent predictive effect on the OS of patients with RCC. In the RCC cohort receiving immunotherapy, IMRGES also demonstrated the capability to differentiate patients’ PFS [IMmotion150: P = 0.039, HR (95% CI) =1.55 (1.02 to 2.34), [171]Fig. 7F; IMmotion151: P = 0.012, HR (95% CI) =1.52 (1.10 to 2.11), [172]Fig. 7G]. Furthermore, it exhibited promising predictive performance for the response of patients with RCC to immunotherapy (IMmotion150: P = 0.026; IMmotion151: P = 0.201; [173]Fig. 7H). To further validate the predictive potential of IMRGES, we collected transcriptome data from 2338 patients across six cancer types for extensive validation. All patients underwent ICI therapy. We conducted a meta-analysis to examine the relationship between IMRGES and OS in these cohorts, using a random-effects model to account for intercohort heterogeneity (P < 0.01, I^2 = 59%, [174]Fig. 7I). Our findings indicated that, at the pan-cancer level, higher IMRGES scores were frequently associated with poor patient OS (HR = 1.506, 95% CI =1.195 to 1.897, P < 0.01, [175]Fig. 7I). DISCUSSION In this study, we systematically investigated the intratumoral mycobiota in patients with RCC, revealing the heterogeneity of fungal communities among different patients and their correlation with prognosis and immune response. The widespread presence of fungi was detected in RCC tumor tissues. On the basis of the intratumoral mycobiome abundance, patients could be divided into two groups, MED and MEF groups. In the MEF group, the prognosis of patients was significantly worse compared to the MED group. Prior researches have demonstrated that the diversity of intratumoral bacterial microbiota was frequently associated with patient prognosis ([176]7, [177]11, [178]67). These findings underscore the intimate relationship between the intratumoral microbiota and patients. Nevertheless, the connection between the intratumoral mycobiota and the prognosis and immunotherapy outcomes of patients with RCC remained unexplored. Our in-depth analysis in this study revealed that the MEF group harbored a significantly higher fungal community diversity and complexity, implying that the heterogeneity of the intratumoral mycobiota was also closely related with patient prognosis. The MEF group displayed significant down-regulation of lipid catabolism pathways and an immune microenvironment characterized by high infiltration of Tex cells. Moreover, we constructed the IMS based on the fungal community, which could robustly discriminate the prognosis and response to immunotherapy of patients with RCC. We also pinpointed A. tanneri as a potential key species influencing the prognosis of patients with RCC. Ultimately, by developing the IMRGES, we extended the predictive capability of fungi to the transcriptome level and further applied it to other cancer types, thereby expanding the application value of our research. Lipid metabolism reprogramming is a hallmark of cancer that frequently shapes a unique tumor microenvironment and cancer cell phenotype, playing a pivotal role in tumor development and progression ([179]56, [180]68). In the MEF group, several pathways related to lipid catabolism were significantly down-regulated, and the expression of certain key genes involved in lipid metabolism was markedly reduced. This change potentially leads to the continuous accumulation of lipids in the tumor microenvironment. Lipid accumulation is a prominent feature in RCC, especially in ccRCC, signifying malignant tumor progression and poor prognosis ([181]69, [182]70). Moreover, we found correlations between certain intratumoral fungal genus and lipid metabolism pathways. These results suggested that the intratumoral mycobiota may influence tumor progression by modulating host metabolic activities. Prior studies have shown that intratumoral microbes and their derived metabolites can affect the host’s metabolic patterns. They played a role in regulating the tumor microenvironment and affecting long-term prognosis and immunotherapy efficacy ([183]5, [184]15, [185]27, [186]71–[187]73). The intratumoral mycobiota stimulates cancer cells to secrete interleukin-33 (IL-33), which, in turn, recruits and activates T[H]2 and innate lymphoid cells 2 within the pancreatic ductal adenocarcinoma (PDAC) tumor microenvironment and further stimulates the tumor growth ([188]27). Liu et al. ([189]29) have found that Aspergillus sydowii could accelerate the lung tumor progression by proliferation and stimulation myeloid-derived suppressor cells through IL-1β. This process leads to the suppression of cytotoxic T lymphocyte cell activity and the accumulation of PD-1^+ CD8^+ T cells. The intratumoral microbiota could also mediate the function of immune effector cells in the tumor microenvironment, influencing antitumor immunity and tumor biological behavior by altering tumor immunogenicity ([190]9, [191]71, [192]74–[193]77). In our study, the MEF group had a highly exhausted state of CD8^+ T cells despite their high infiltration. Simultaneously, this group showed high expression of T cell inhibitory receptors in the tumor tissue, and certain Treg subtypes were significantly up-regulated. These Treg cells played a crucial role in tumor immune escape and could induce the generation of Tex cells ([194]59, [195]78–[196]80). The immune microenvironment in the MEF was in a highly suppressed state according to the above analyses. Evidence from various studies have pointed that lipid metabolism imbalance in tumor tissues can induce CD8^+ T cell dysfunction ([197]57, [198]58, [199]81). In RCC, patients with worse prognosis always have tumor immune microenvironments enriched with CD8^+ T cells exhibiting reduced cytotoxic activity ([200]41, [201]82). Therefore, in the MEF group, the significant down-regulation of lipid catabolism pathways may lead to lipid accumulation in the tumor microenvironment, thereby inhibiting the antitumor activity of CD8^+ T cells and causing them to exhibit an exhausted state. Compared to the MED group, we observed significantly higher expression levels of IDO1, SLAMF7, and VEGFA in the MEF group. These genes have been reported to play a role in promoting or sustaining T cell exhaustion within the tumor microenvironment ([202]62–[203]64). IDO1 has been shown to drive immunosuppressive effects through the metabolism of tryptophan into kynurenine. It would ultimately suppress effector T cell responses and foster an environment conducive to T cell dysfunction. Similarly, SLAMF7 activation can induce multiple inhibitory receptors on T cells and reprogram them toward a more exhausted phenotype. VEGFA, a potent mediator of angiogenesis, also exerts immunomodulatory functions by impairing T cell infiltration, enhancing the presence of immunosuppressive cells, and contributing to the up-regulation of T cell exhaustion markers. Therefore, beyond the notable up-regulation of immune exhaustion markers such as LAG3, the higher expression of genes capable of indirectly suppressing T cell function may also play a crucial role in molding the T cell exhaustion state. Notably, IDO1 has been identified as a crucial immunoregulatory enzyme connecting host metabolic pathways and microbial signals in previous studies ([204]83, [205]84). Zhang et al. ([206]85) found that the gut commensal bacterium Dubosiella newyorkensis produces l-lysine (Lys), which activates IDO1 and thereby induces the immunotolerogenic capacity of dendritic cells. This metabolic circuitry ultimately gives rise to an immunosuppressive microenvironment. While much attention has focused on bacterial contributions, fungal constituents may similarly alter or amplify IDO1-related pathways. In conclusion, evaluating IDO1 expression alongside fungal community changes may provide insights into the broader role of host-mycobiota metabolic cross-talk in shaping antitumor responses. Recent studies have highlighted the intimate association between intratumoral microbiome, particularly bacterial microbiota, and the prognosis or outcome of immunotherapy ([207]5, [208]14, [209]86). However, the relationship between intratumoral mycobiota and immunotherapy remains unclear. In the context of the gut microbiome, gut fungi have exhibited superior predictive capabilities compared to gut bacteria in terms of patient response to ICIs ([210]25, [211]87). In our study, we observed that intratumoral mycobiota in RCC potentially played an essential role in reshaping the immune microenvironment. By using integrated ML algorithms, we identified the most representative and influential fungi combinations from the fungal microecological community to construct the IMS. This signature could be used to predict patient survival and differentiate patients’ responses to immunotherapy. Furthermore, applying the framework of RCA, we identified A. tanneri as a potentially key fungal species influencing the RCC fungal community. Medema et al. ([212]66) have indicated that A. tanneri had the most robust metabolic capabilities among currently found fungal species. Our findings demonstrated that A. tanneri was intimately associated with the host immune microenvironment. Consequently, there existed a potential nexus between intratumoral fungi and immunotherapy efficacy. Further studies are warranted to elucidate the intricate interaction mechanisms between fungi and the tumor immune microenvironment ([213]18, [214]27, [215]88). Our research had certain limitations. Although we have demonstrated the critical role of A. tanneri in RCC through various analyses, the difficulty in obtaining and culturing this species posed technical challenges that currently prevented experimental verification of its effects in RCC. We have not investigated the potential impact of the metabolic products derived from the intratumoral mycobiota and A. tanneri on the tumor microenvironment. Moreover, our study only deeply explored A. tanneri, while other important components of the intratumoral mycobiota required further attention and investigation. In conclusion, by integrating data from multiple large-scale clinical cohorts and multi-omics datasets, this study systematically unveiled the heterogeneity of the RCC intratumoral mycobiota and its intricate associations with host metabolism and immune status. These findings offered different perspectives for future research on the role of intratumoral mycobiota in tumor development and immunotherapy. Further comprehensive exploration into the fungal communities within tumors may provide unique insights into the mechanisms underlying tumor occurrence and progression. In addition, such research could guide the development of precise immunotherapeutic strategies and advance the application of precision medicine approaches in the field of oncology. MATERIALS AND METHODS Study population and sample collection The intratumoral mycobiome data for TCGA-KIRC were derived from a comprehensive pan-cancer study investigating the fungal ecologies ([216]18). The RNA-seq data, mutation data, and clinical information for TCGA-KIRC were obtained using the TCGAbiolink R package ([217]89). We collected data from 56 patients with WCH-RCC ([218]30). In addition, we included patients who received ICI therapy from two large-scale, international, multicenter RCC clinical trials, IMmotion150 ([219]31) (n = 162) and IMmotion151 ([220]32) (n = 304). In both cohorts, all patient samples were collected before treatment initiation, and the patients had not received any prior systemic therapy for RCC before undergoing immunotherapy. In IMmotion150 cohort, patients received either atezolizumab monotherapy or atezolizumab plus bevacizumab. In IMmotion151 cohort, patients received atezolizumab plus bevacizumab. A summary table of baseline information for patients in different cohorts is shown in table S4. Moreover, we included transcriptome data from 2338 patients receiving immunotherapy across six types of cancers, including melanoma, NSCLC, mUC, colorectal cancer (CRC), RCC, and PDAC. For multiplex immunofluorescence and FISH, we retrospectively collected 40 patients who were clinically diagnosed with ccRCC and underwent renal cancer surgery at Changhai Hospital between 2019 and 2021 in our study (CHH-RCC). The baseline characteristics of patients were described in table S4. The inclusion criteria were as follows: (i) preoperative imaging and clinical evaluations indicated resectable renal tumors; (ii) postoperative pathological diagnosis confirmed ccRCC; and (iii) comprehensive initial medical records were available, including detailed medical history, imaging data, and preoperative laboratory test results. The exclusion criteria were as follows: (i) receipt of targeted therapy, immunotherapy, interventional therapy, or other antitumor treatments before surgery; (ii) antibiotic use within 1 month before surgery; (iii) postoperative pathological findings indicating non-ccRCC subtypes; and (iv) presence of congenital autoimmune diseases or other malignancies documented in the medical records. This study was approved by the Ethics Committee of Changhai Hospital (ethical approval no. CHEC2021-091). Following surgical resection, tumor tissues were fixed in 10% neutral buffered formalin and embedded in paraffin blocks for preservation. We sectioned these samples into 4- to 5-μm-thick slices for the study. For qPCR, tumor tissues from 16 patients who underwent radical nephrectomy for RCC were collected from Changhai Hospital. Patient recruitment and sample collection were approved by the Ethics Committee of Changhai Hospital (no. CHEC2021-091). Before the start of this study, written informed consent was obtained from each participant. All cases were pathologically confirmed as ccRCC post-surgery. To ensure the integrity and sterility of the samples, we performed strict aseptic procedures. Tumor tissues were collected from patients under sterile conditions in the operating room. Tissue samples were excised within 30 min of removal from the body using sterile surgical instruments. The tumor tissues were carefully dissected, rinsed with sterile saline, and immediately stored in sterile cryovials. Samples were transferred to liquid nitrogen tanks within 15 min of collection. To control for environmental contamination, four 1.5-ml sterile saline tubes were used as negative controls. During the sampling process, the cryovials containing sterile saline were left open and exposed to the operating room environment for 5 min. Negative controls were transported and stored under the same conditions as the tissue samples. These controls were subsequently used to identify and exclude potential contamination introduced during tissue collection and downstream processing. Microbial assignments A microbial assignments workflow was established based on the research by Narunsky-Haziza et al. ([221]18). (i) Host depletion: We constructed a host depletion pipeline using a combination of fastp ([222]90), Minimap2 ([223]91), and SAMtools ([224]92). Initially, quality control was conducted on the raw RNA-seq data using fastp, and low-quality reads (based on default fastp parameters) were eliminated. Quality-controlled reads were subsequently aligned to the human genome GRCh38.p14 and the Phi X 174 viral genome using Minimap2. Successfully aligned reads were removed using SAMtools. (ii) Microbial assignments: The host-depleted sequences were aligned to a standard microbial reference genome constructed based on the RefSeq release 220 (rep220) database using Bowtie2 ([225]93). Last, the alignment results were inputted into Woltka ([226]94) to obtain a microbial abundance matrix based on operational genomic units. All reference genomes were downloaded in September 2023. Transcriptomic quantification The raw reads were subjected to quality control using fastp (under the same conditions as previously mentioned) and then the expression of transcripts was quantified using Salmon v1.10.2 ([227]95) based on the human reference genome (GRCh38.p14). The gene-level expression matrix was obtained using the tximport package. With the exception of the TCGA-KIRC, WCH-RCC, IMmotion150, and IMmotion151 cohorts, the source of transcriptomic data for the remaining datasets is shown in table S1. Consensus clustering Consensus clustering was performed based on the intratumoral mycobiome abundance in RCC. The optimal number of clusters was identified using the consensus score matrix and CDF curve. This process was executed using the ConsensuClusterPlus ([228]96) R package. Microbial signature analysis The alpha diversity indices, including ACE, Chao1, Shannon, and Simpson, were calculated using the vegan package ([229]97). The Wilcoxon rank-sum test was used to compare the statistical differences in alpha diversity between two groups. Principal coordinate analysis (PCoA) based on Bray-Curtis distances was used to assess the beta diversity of the fungal communities. PERMANOVA was performed to compare the differences in fungal community structure between groups, and visualization was conducted using the microeco ([230]98) package. LEfSe was applied to identify differentially abundant fungal taxa from the phylum to genus level (LDA score > 2, FDR < 0.05). To compare the ecological complexity of the fungal communities in different groups, we calculated the Spearman correlation between species to construct a co-occurrence network. Species with a prevalence below 10% in the sample were excluded from the analysis. The P values for the correlations were corrected using the FDR method. The threshold for the existence of interactions between species was set as a Spearman correlation >0.8 and an FDR <0.05. Subsequently, we visualized the network structure and computed their topological characteristics, including the number of nodes and edges, average degree, and other indicators. Differential gene expression analysis and pathway enrichment analysis To explore the potential differences in biological characteristics between the MED and MEF groups, the edgeR ([231]99) package was used to identify DEGs between the groups. The thresholds for |fold change| and FDR were set to >2 and <0.05, respectively. Subsequently, we performed overrepresentation analysis (ORA) on the DEGs, comprehensively considering the Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Ontology (GO), Reactome, WikiPathways, and Hallmark databases for pathway annotation. ORA was conducted using the BioEnricher package. GSEA ([232]100) was performed using the clusterProfiler ([233]101) package to identify up-regulated and down-regulated pathways in the MED group. The gene sets were obtained from the MSigDB database. The enrichment P values were corrected for multiple testing using the FDR method. Visualization was performed using the GseaVis ([234]102) package. Single-sample GSEA (ssGSEA) was conducted using the “GSVA” ([235]103) package. Immune infiltration We performed immune infiltration analysis using the IOBR ([236]104) package. To ensure the robustness of the assessment, we used five immune infiltration algorithms, including CIBERSORT ([237]105), EPIC ([238]106), TIMER ([239]107), xCell ([240]108), and quanTIseq ([241]109). In addition, we further evaluated cell subtypes and calculated exhaustion scores based on ssGSEA. The composition of CD8^+ T cell subtypes and Treg subtypes was computed using gene sets provided by Zheng et al. ([242]59) and Li et al. ([243]61). The exhaustion scores were calculated based on the exhaustion-related gene sets provided by Sade-Feldman et al. ([244]60). Integrated ML framework To further investigate the impact of intratumoral fungi on patient prognosis and immunotherapy, we integrated 10 ML algorithms to predict OS, including CoxBoost, generalized boosted regression modeling, Lasso, Ridge, elastic net (Enet) ([245]110), RSF ([246]111), partial least squares regression for Cox ([247]112), survival support vector machine ([248]113), supervised principal components ([249]114), and stepwise Cox. A total of 100 algorithm combinations were obtained to identify the most representative and stable species or genes in the complex intratumoral fungal microecological environment. Some combinations were skipped due to their inability to effectively reduce the dimensionality of the feature space. We constructed the IMS and IMRGES from the mycobiome and transcriptome levels, respectively. For IMS, the sparsity and high discreteness of species abundance in the fungal community could introduce noise and affect the accuracy of IMS. Therefore, we first removed low-abundance species (fungi with zero abundance in more than 80% of the samples). Subsequently, fungi significantly associated with OS were identified through univariate Cox analysis and served as candidate inputs for the model. For IMRGES, to maximize the transfer of fungal characteristics to the genome and enhance its broad applicability, we defined three major gene sets to serve as “bridges.” Set 1: Fungal-related genes. We calculated the Spearman correlation between each gene and all fungal species that had differential abundance between the MEF and MED groups, applying FDR to correct the correlation P values. Genes significantly correlated with at least one fungus were retained (FDR < 0.05). Set 2: DEGs. We identified DEGs between the MED and MEF groups (|logFC| > 1, FDR < 0.05), which were closely related to the fungal community ecology. Set 3: Prognosis genes. We ensured that each gene could predict the OS of patients (Cox P < 0.05). The intersection of genes from the three gene sets was considered to effectively characterize the fungal features and served as input for our model. We calculated the C-index of each model in the training and validation sets. The highest mean C-index was used as the basis for model selection. Subsequently, on the basis of the lastly selected model, we calculated the IMS score or IMRGES score for each patient. We then determined the optimal cutoff value using the survminer package, dividing all patients into high-score and low-score groups. We conducted a meta-analysis using Onlinemeta to investigate the impact of IMRGES on OS in patients receiving immunotherapy across six cancer types. Because of significant heterogeneity observed among the cohorts (P < 0.65, I^2 = 59%), a random-effects model was used. Rationality-congruence assessment For the seven fungi in IMS, we constructed the framework of RCA by integrating the results of LEfSe, survival analysis, pathway analysis, and ML (fig. S5A). This framework allowed for the comprehensive screening of key species using a cumulative scoring method based on the consistency of multiple evidence. We combined the four analysis results in pairs, yielding a total of six combinations for testing. To evaluate the performance of fungi in the paired test combinations, we proposed the principle of “directionality” consistency. The directionality referred to the inference of the potential direct or indirect impact of fungi on prognosis based on the findings of previous studies and their performance in the four analyses (tests). In any test, if a fungus showed an unfavorable impact on survival, its directionality in that test is considered unfavorable and scored as −1. Conversely, if it has a favorable impact on prognosis, it is scored as +1. Specifically, in LEfSe, patients in group B had significantly worse prognosis compared to those in group A. If a fungus was enriched in group B, it was considered to have a potential unfavorable impact on prognosis, and the score for this test was recorded as −1. Conversely, fungi enriched in group A were scored as +1. If not enriched in either group, it was assigned a score of 0. For survival analysis, we directly referred to the HR of the fungi, where HR > 1 could be defined as a risk factor and scored as −1. In pathway analysis, we mainly considered the impact of lipid catabolism pathways on survival. In RCC, if any lipid catabolism pathways are significantly down-regulated, it may lead to lipid metabolism imbalance and lipid accumulation, which often indicates poor prognosis ([250]55). Therefore, if a fungus was negatively correlated with any lipid catabolism pathway, we inferred that the higher its abundance, the more likely lipid accumulation occurred in the tumor microenvironment, and the more unfavorable it is for patient survival, scoring −1. Conversely, if a fungus was positively correlated with lipid catabolism pathways, it was scored +1, and if it had no significant correlation, it was scored 0. In ML, patients in the high–IMS score group showed significantly worse prognosis compared to the low–IMS score group. We made judgments based on the coefficient of the fungus in the model. If the coefficient > 0, it indicated a positive contribution to the model and may be associated with worse prognosis, scoring −1. The score of a single paired test combination was the product of the scores of the two tests (+1, 0, or −1). A score of +1 indicates that the two tests have consistent directionality, meaning the paired test is passed. Species that passed all paired test combinations, accumulating a score of +6, were considered to have passed the RCA. DNA extraction and qPCR quantification Approximately 200 mg of tissue was collected from each ccRCC tumor mass for DNA extraction. Total DNA was extracted using the QIAamp PowerFecal (Pro) DNA Kit according to the manufacturer’s instructions. Briefly, tumor tissues were thoroughly ground in liquid nitrogen using a sterile mortar and pestle. Subsequently, C1 buffer was added, and the samples were vortexed at maximum speed for 10 min. The samples were then centrifuged at 15,000g at 4°C for 10 min, after which the supernatant was discarded. DNA was further purified according to the manufacturer’s instructions. The quality of the extracted DNA was assessed using a NanoDrop spectrophotometer, and all samples had the ratio of the absorbance values of 260 nm versus 280 nm (A[260]/A[280] ratios) between 1.8 and 2.0. The study was designed with three experimental groups: ccRCC tumor blocks obtained under sterile postsurgical conditions, environmental controls, and nontemplate controls (detection of contamination of the PCR reagents). All procedures were performed under strict sterile conditions. For qPCR, we used the Takara Premix Ex Taq (Takara-#RR390A) kit to detect the presence of A. tanneri within ccRCC samples. The qPCR reaction program was as follows: initial denaturation at 94°C for 10 min, followed by 40 cycles of denaturation at 94°C for 15 s and annealing/extension at 60°C for 60 s. For each sample, three replicate wells were prepared with 15 ng of DNA per well. The mean CT value of the triplicates was used as the CT value for A. tanneri in that sample. Multiplex immunofluorescence Tissue sections were treated with EDTA antigen retrieval buffer. After circling with an immunohistochemistry pen, the sections were immersed in a 3% hydrogen peroxide solution for 25 min at room temperature in the dark. Subsequently, bovine serum albumin was added and incubated for 30 min, followed by the addition of the primary antibody and overnight incubation at 4°C. After washing with PBS, the corresponding fluorescent-labeled secondary antibody was incubated for 50 min. This process was repeated for the other three antibodies. 4′,6-Diamidino-2-phenylindole (DAPI) staining was added at room temperature and incubated for 10 min. Analysis was performed using ImageJ (v 1.54g). A nuclear segmentation algorithm was applied on the DAPI channel to identify and count cells. The number of positive and negative cells for each marker was quantified. Fluorescence in situ hybridization Formalin-fixed paraffin-embedded sections were immersed in diethylpryrocarbonate water and hydrolyzed with lysozyme (10 mg/ml) for 30 min to disrupt the fungal cell walls. Subsequently, probes targeting the fungal 28S rRNA gene (AGGTGCCCCGAAGGGCGCTA) and A. tanneri–specific probe (TAGCGCCCTTCGGGGCACCT) were labeled with Cy3 fluorophore (excitation wavelength: 555 nm, emission wavelength: 590 nm) ([251]28). The intensity of the fluorescent signal from A. tanneri was quantified using ImageJ (v 1.54g). Statistical analysis Comparisons of continuous variables between two groups were performed using the Wilcoxon rank-sum test. The chi-square test was used to compare categorical variables. The correlation between two continuous variables was evaluated using the Spearman correlation test. Cox regression and Kaplan-Meier analyses were conducted using the survival package. The ROC curves for the ML models were obtained using the pROC package. The ggplot2 package was used for graphical visualization. All statistical analyses and plotting were performed in R (version 4.2.3). All statistical tests were two-sided, and a P value less than 0.05 was considered statistically significant. Acknowledgments