Abstract The gene Sphingomyelin phosphodiesterase 2 (SMPD2), a member of the SMPD family, plays crucial roles in cell cycle progression and cell proliferation. However, the pathogenic implications of SMPD2 across various cancers remain poorly understood. Its potential involvement in lipid metabolism and immune-related processes within the tumor microenvironment has not been systematically characterized. To address these gaps, we conducted a comprehensive pan-cancer analysis of SMPD2. Using a range of computational tools, we investigated its role in tumor immune infiltration, immune evasion, tumor progression, therapy response, and prognosis across various cancer types. Our findings suggest that SMPD2 is widely expressed across cancers in The Cancer Genome Atlas (TCGA) and its expression levels are associated with tumor stages and clinical outcomes. Additionally, SMPD2 was found to be involved in tumor immune evasion across different cancer types. The methylation status of SMPD2 was inversely correlated with its mRNA expression levels, which were associated with dysfunctional T cell phenotypes and worse prognoses in diverse cancer cohorts. Furthermore, SMPD2 expression was linked to heterogeneous therapeutic outcomes across multiple cancer types, including variable responses to immune checkpoint blockade. Interestingly, SMPD2 demonstrated superior predictive capacity for treatment response and overall survival in immune checkpoint blockade sub-cohorts compared to three of the seven established biomarkers. While functional experiments are warranted, our results provide a data-driven, pan-cancer landscape of SMPD2 expression and its potential relevance to immune modulation and clinical outcomes Overall, SMPD2 may serve as a candidate biomarker for cancer prognosis and therapeutic response, and a potential target for future mechanistic studies. Supplementary Information The online version contains supplementary material available at 10.1007/s12672-025-03553-5. Keywords: SMPD2, Biomarker, Immune cell infiltration, Pan-cancer, Prognosis Introduction The rising incidence and mortality rates of cancer have garnered significant attention [[38]1]. In response, extensive research has been conducted to understand the mechanisms underlying cancer occurrence, development, and metastasis. Despite global advances in cancer therapy, overall survival rates remain unsatisfactory, highlighting the urgent need for reliable prognostic biomarkers. Ceramides, a class of bioactive sphingolipids, are key regulators of fundamental cellular processes such as cell death, proliferation, autophagy, and drug resistance [[39]2, [40]3]. They are also critical for cellular homeostasis, affecting migration, differentiation, stress adaptation, survival, and senescence [[41]4]. Neutral sphingomyelinases (nSMases) catalyze the hydrolysis of sphingomyelin to generate ceramide and have been implicated in diverse biological responses to environmental stress and inflammation [[42]5–[43]7]. Emerging evidence suggests that sphingolipid metabolism is intricately linked to antitumor immunity, particularly through its influence on immune cell function and response to immunotherapy. Among the nSMase family, SMPD2 (also known as nSMase1) stands out for its subcellular localization and functional roles. It is predominantly found in the endoplasmic reticulum (ER) and Golgi apparatus [[44]5, [45]8], and is critical for ceramide production in response to ER stress [[46]9, [47]10]. Notably, SMPD2 has been shown to regulate T cell receptor-induced apoptosis and modulate T cell signaling [[48]11], distinguishing it functionally from SMPD1 (lysosomal) and SMPD3 (exosomal/plasma membrane-associated). SMPD2 also contributes to tumor suppression in hepatocellular carcinoma by modulating the ceramide-to-sphingomyelin ratio [[49]12], while SMPD2-deficient mice exhibit enhanced liver tumor formation. Despite these findings, the role of SMPD2 in cancer immunology and its relevance across tumor types have not been systematically studied. These unique features make SMPD2 a biologically plausible candidate for pan-cancer exploration, particularly in the context of immune regulation and prognosis. The tumor microenvironment (TME) is a highly complex and heterogeneous ecosystem comprising malignant cells, fibroblasts, vasculature, immune, and immunosuppressive cells. This complexity fosters interactions that often lead to immune dysfunction, such as T-cell anergy, and impaired anti-tumor immune responses [[50]13]. Components like M2 macrophages [[51]14], cancer-associated fibroblasts (CAFs) [[52]15], regulatory T cells (Tregs) [[53]16], and myeloid-derived suppressor cells (MDSCs) [[54]17] play key roles in suppressing cytotoxic T cell function, thereby promoting tumor progression, metastasis, and resistance to immunotherapy. Sphingolipids, as structural components of cell membranes, regulate immune cell surface dynamics and modulate lymphocyte trafficking, inflammatory responses, and chemotaxis [[55]18–[56]20]. Certain chemotherapeutic agents also induce cancer cell death via ceramide accumulation [[57]21–[58]23]. In melanoma, nSMase2 has been shown to influence immune response and modulate anti-PD-1 therapy efficacy [[59]24]. However, the specific contributions of SMPD2 in shaping the immune landscape of tumors remain unclear. An integrated analysis of SMPD2 expression in cancer and its associations with immune contexture, prognosis, and therapy response may uncover novel insights into its clinical and biological significance. Systematically profiling diverse immune and stromal cell types across cancers remains challenging using experimental methods alone. In this context, bioinformatics approaches offer efficient and scalable alternatives by integrating multi-omics and clinical data to extract immune-related features. Such computational frameworks have become indispensable for cancer biomarker discovery, prognosis modeling, and therapeutic response prediction. In this study, we conducted a comprehensive pan-cancer analysis of SMPD2 using publicly available datasets. We investigated its expression patterns, epigenetic regulation, associations with immune cell infiltration and immune checkpoint expression, prognostic implications, and therapeutic relevance across multiple cancer types. Our goal was to delineate the potential role of SMPD2 in the tumor immune microenvironment and evaluate its utility as a biomarker and research target. Materials and methods Data acquisition We downloaded the uniformly processed and batch-corrected pan-cancer dataset TCGA TARGET GTEx (PANCAN; N = 19,131 samples; G = 60,499 genes) from the UCSC Xena platform ([60]https://xenabrowser.net/). We extracted the expression data of SMPD2 (ENSG00000135587) across all available samples. Expression values were log2(x + 0.001) transformed, and samples with zero expression were excluded prior to downstream analyses. Depending on the analysis, we retained tumor-derived samples (e.g., Primary Solid Tumor, Primary Tumor, Bone Marrow, Peripheral Blood, Metastatic) and, when appropriate, included normal tissue samples from TCGA or GTEx cohorts for comparison. Differential SMPD2 expression analysis To assess SMPD2 expression across different tumor stages, we employed the expression DIY module in GEPIA2 to generate pathological stage plots (I, II, III, IV) for TCGA cancer types. Additionally, we used the TNMplot module from the Kaplan-Meier (KM) plotter for differential gene expression analysis across tumor, normal, and metastatic tissues. The prognostic differences of SMPD2 expression across 38 tumor types were analyzed using the dichotomy method. Differential expression and survival analysis Differential expression of SMPD2 between tumor and normal tissues was analyzed across 34 cancer types using the Wilcoxon rank-sum test in R (version 3.6.4). Tumor types with fewer than three samples per group were excluded. For survival analysis, patients were stratified into high- and low-expression groups based on median SMPD2 expression. Kaplan–Meier (KM) survival curves, log-rank tests, and Cox proportional hazards regression models were used to evaluate overall survival (OS), disease-specific survival (DSS), and progression-free survival (PFS). Hazard ratios (HR) and 95% confidence intervals (CI) were calculated using the R packages survival and survminer. Immune cell infiltration analysis To estimate immune cell infiltration, we used multiple complementary methods: TIMER2.0 was applied to quantify the infiltration of six major immune cell types (B cells, CD4 + T cells, CD8 + T cells, neutrophils, macrophages, dendritic cells) across 38 tumor types. ESTIMATE scores, including ImmuneScore, StromalScore, and ESTIMATE Score, were computed using the R package estimate (v1.0.13), which infers tumor purity based on gene expression. xCell-based immune deconvolution was performed using the IOBR R package (v0.99.9), yielding enrichment scores for 67 immune and stromal cell types, including immunosuppressive populations such as Tregs, M2 macrophages, and fibroblasts. For all immune-related scores, Pearson correlation coefficients between SMPD2 expression and infiltration scores were calculated using the psych (version 2.1.6) corr.test function in R. Immune checkpoint co-expression analysis We curated a List of 60 immune checkpoint–related genes (24 inhibitory and 36 stimulatory) based on the study The Immune Landscape of Cancer [[61]25]. Pearson correlation was used to evaluate co-expression between SMPD2 and each checkpoint gene across tumor types. The strength of correlation was categorized as weak (r = 0.1–0.3), moderate (0.3–0.6), or strong (> 0.6). Tumor mutation burden, neoantigens, and functional enrichment We analyzed the correlation between SMPD2 expression and tumor mutation burden (TMB) using TCGA mutation data. Associations were evaluated using Spearman’s rank correlation. Differential expression and functional enrichment analysis Gene expression and corresponding clinical data of the target cancer type were retrieved using the TCGAbiolinks R package. Project identifiers and data types were specified to batch download high-throughput data via the GDC API. Standardized preprocessing was conducted to remove batch effects. Patients were stratified into high and low expression groups based on SMPD2 transcript levels. Differential gene expression analysis between the two groups was performed using the DESeq2 or edgeR package in R. Genes were considered significantly differentially expressed if they met both the adjusted p-value threshold (p < 0.05) and an absolute log₂ fold change of ≥ 1. Functional enrichment analysis of the identified differentially expressed genes (DEGs) was then conducted. Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis were performed using the clusterProfiler and org.Hs.eg.db packages in R. Enrichment results for biological processes, molecular functions, cellular components, and signaling pathways were visualized using bar plots. Epigenetic methylation analysis We used the TCGA methylation module of the UALCAN interactive web resource to analyze differential methylation levels of SMPD2 between tumor and paired normal tissues across TCGA cancer types. Promoter methylation levels were represented by beta (β) values ranging from 0 (unmethylated) to 1 (fully methylated). Different β value cutoff points were considered to indicate hypomethylation (β: 0.25–0.3) and hypermethylation (β: 0.5–0.7). We also used the TIDE server to assess the effects of methylation on dysfunctional T cell phenotypes and prognoses. Patient samples Noncancerous adjacent tissue and cancerous tissue of HCC were collected along with informed consent from patients under a protocol approved by the Medical Ethics Committee of Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China (No. NEFC-2021-394). All samples were obtained from the Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China. All experiments were performed in accordance with relevant guidelines and regulations. Immunohistochemistry analysis Formalin-fixed paraffin-embedded HCC specimens were treated with xylene and later rehydrated in alcohol at decreasing concentrations. Sections were boiled in citrate antigenic retrieval buffer (pH = 6.0) for 3 min, cooled to room temperature (RT), and endogenous peroxidase activity was quenched with 3% hydrogen peroxide for 10 min. Sections were blocked with 10% goat serum at RT for 30 min, then incubated with human SMPD2, human PD-L1, and human LAG-3 antibodies at 4 °C overnight. After washing, sections were incubated with a biotinylated secondary antibody at RT for 30 min, followed by a final wash with 1x PBS. Sections were then incubated with horseradish-conjugated streptavidin at RT for 20 min, developed with 3,3-diaminobenzidine tetrahydrochloride, and counterstained with hematoxylin. An IHC staining without primary antibody served as a negative control. Statistical analysis and multiple testing correction All statistical analyses were conducted using R version 3.6.4. For group comparisons, Wilcoxon Rank Sum tests were used. For correlation analyses, Pearson or Spearman methods were applied depending on data distribution. To account for multiple comparisons across expression, immune correlation, and survival analyses, we performed false discovery rate (FDR) correction using the Benjamini–Hochberg method. Unless otherwise specified, an FDR-adjusted p-value < 0.05 was considered statistically significant. In visualizations, significance thresholds were denoted as p < 0.05 (*), < 0.01 (**), and < 0.001 (***). Results Expression levels of SMPD2 Analysis of the GTEx dataset revealed that SMPD2 is expressed at low levels in muscle tissues but is most highly expressed in the testis (Fig. [62]1A). In the CCLE database, SMPD2 expression was lowest in kidney cell lines and highest in intestinal cell lines (Fig. [63]1B). We observed significant upregulation in 18 types of tumors, including GBM glioblastoma multiforme (p = 4.2 × 10^−37), GBMLGG glioblastoma multiforme and lower-grade glioma (p = 7.4 × 10^−51), LGG brain lower-grade glioma (p = 1.5 × 10^−31), UCEC uterine corpus endometrial carcinoma (p = 6.6 × 10^−8), BRCA breast invasive carcinoma (p = 1.2 × 10^−40), ESCA esophageal carcinoma (p = 1.3 × 10^−3), STES stomach and esophageal carcinoma (p = 7.9 × 10^−13), KIRP kidney renal papillary cell carcinoma (p = 2.1 × 10^−14), COAD colon adenocarcinoma (p = 3.7 × 10^−51), COADREAD colorectal adenocarcinoma (p = 7.4 × 10^−60), PRAD prostate adenocarcinoma (p = 1.7 × 10^−8), STAD stomach adenocarcinoma (p = 3.3 × 10^−16), LIHC liver hepatocellular carcinoma (p = 2.0 × 10^−30), BLCA bladder carcinoma (p = 4.6 × 10^−4), PAAD pancreatic adenocarcinoma (p = 7.5 × 10^−22), ALL acute lymphoblastic leukemia (p = 1.6 × 10^−20), LAML acute myeloid leukemia (p = 4.3 × 10^−47), CHOL cholangiocarcinoma (p = 4.6 × 10^−6). We also observed significant downregulation in 9 types of tumors, including LUAD lung adenocarcinoma (p = 6.0 × 10^−5), HNSC head and neck squamous cell carcinoma (p = 3.6 × 10^−8), KIRC kidney renal clear cell carcinoma (p = 7.8 × 10^−3), WT Wilms tumor (p = 6.8 × 10^−21), SKCM skin cutaneous melanoma (p = 6.0 × 10^−6), THCA thyroid carcinoma (p = 5.1 × 10^−60), TGCT testicular germ cell tumor (p = 1.1 × 10^−49), ACC adrenocortical carcinoma (p = 4.0 × 10^−8), and KICH kidney chromophobe (p = 5.6 × 10^−13) (Fig. [64]1C). We further analyzed the relationship between SMPD2 expression and tumor staging, and found that in ESCA esophageal carcinoma, SMPD2 is highly expressed in both early (Stage 1) and late (Stage 4) stages of the tumor. In KIRC kidney renal clear cell carcinoma and LIHC liver hepatocellular carcinoma, SMPD2 expression is slightly increased in the mid and late stages of the tumor (Fig. [65]1D). These findings suggest that SMPD2 expression may play a role in promoting tumor metastasis and enhancing the malignant phenotype of tumors. Fig. 1. [66]Fig. 1 [67]Open in a new tab SMPD2 expression in different tissue types; a SMPD2 gene expression levels analyzed based on the GTEx dataset; b SMPD2 gene expression levels analyzed based on the CCLE dataset; c Tumor vs. normal SMPD2 gene expression comparison; d the relationship between SMPD2 expression and tumor staging Prognostic relevance of SMPD2 expression in tumor To assess the correlation between SMPD2 expression and patient prognosis across the 38 tumor types, with results compiled as forest plots (p < 0.05). For Disease-specific survival (DSS), we observed that high expression of SMPD2 was associated with poor prognosis in five tumor types: TCGA-GBMLGG glioblastoma multiforme and lower-grade glioma (n = 619, p = 4.2 × 10^−12, HR = 2.34 [1.84, 2.97]), TCGA-LIHC (n = 341, p = 1.6 × 10^−4, HR = 1.62 [1.26, 2.08]), TCGA-SKCM skin cutaneous melanoma (n = 444, p = 0.01, HR = 1.22 [1.05, 1.42]), TCGA-SKCM metastatic skin cutaneous melanoma (n = 347, p = 0.02, HR = 1.21 [1.03, 1.44]), and TCGA-LAML acute myeloid leukemia (n = 209, p = 6.0 × 10^−3, HR = 1.47 [1.12, 1.93]). In contrast, low expression was associated with poor prognosis in one tumor type: TCGA-PAAD (n = 172, p = 2.4 × 10^−3, HR = 0.56 [0.38, 0.81]), as shown in Fig. [68]2A. Fig. 2. [69]Fig. 2 [70]Open in a new tab Prognostic relevance of SMPD2 expression in 38 tumor types. a, b A forest plot of hazard ratios of SMPD2 in 38 types of tumors Overall survival (OS) analysis revealed that high expression was associated with poor prognosis in five tumor types: TCGA-GBMLGG glioblastoma multiforme and lower-grade glioma (n = 619, p = 4.2 × 10^−12, HR = 2.34 [1.84, 2.97]), TCGA-LIHC liver hepatocellular carcinoma (n = 341, p = 1.6 × 10^−4, HR = 1.62 [1.26, 2.08]), TCGA-SKCM skin cutaneous melanoma (n = 444, p = 0.01, HR = 1.22 [1.05, 1.42]), TCGA-SKCM-M metastatic skin cutaneous melanoma (n = 347, p = 0.02, HR = 1.21 [1.03, 1.44]), and TCGA-LAML acute myeloid leukemia (n = 209, p = 6.0 × 10^−3, HR = 1.47 [1.12, 1.93]). In contrast, low expression was associated with poor prognosis in one tumor type: TCGA-PAAD pancreatic adenocarcinoma (n = 172, p = 2.4 × 10^−3, HR = 0.56 [0.38, 0.81]), as shown in Fig. [71]2B. To further illustrate the impact of SMPD2 expression levels on survival in different tumors, survival curves was plotted for high and low SMPD2 expression in the aforementioned cancers (Fig [72]S1). The data reveal that high SMPD2 expression is significantly associated with reduced survival rates in LIHC liver hepatocellular carcinoma (p < 0.01), SKCM skin cutaneous melanoma (p < 0.01), while low SMPD2 expression is related to reduced survival rates in PAAD pancreatic adenocarcinoma (p < 0.05). These results suggest that high SMPD2 expression may be a risk factor for adverse prognosis in ACC adrenocortical carcinoma, KIRC kidney renal clear cell carcinoma, LGG brain lower-grade glioma, LIHC liver hepatocellular carcinoma, and SKCM skin cutaneous melanoma, whereas low SMPD2 expression may indicate worse prognosis in UCEC uterine corpus endometrial carcinoma and PAAD pancreatic adenocarcinoma. Relationship between SMPD2 expression and immune cell infiltration To investigate the immunological relevance of SMPD2 in the tumor microenvironment, we comprehensively analyzed its association with immune cell infiltration, immune checkpoint gene expression, and tumor mutational characteristics across cancer types. SMPD2 expression exhibited diverse correlations with immune cell infiltration levels across cancers. Using the TIMER2.0 algorithm, we observed both positive and negative correlations between SMPD2 and the infiltration of B cells, CD4⁺ and CD8⁺ T cells, neutrophils, macrophages, and dendritic cells, indicating a heterogeneous immune regulatory landscape associated with SMPD2 (Fig. [73]3A). We further examined the relationship between SMPD2 and immune and stromal content using the ESTIMATE algorithm. SMPD2 expression showed significant correlations with ImmuneScore and StromalScore in 27 out of 44 cancer types, with the strongest associations found in cervical squamous cell carcinoma (CESC), thyroid carcinoma (THCA), and stomach adenocarcinoma (STAD), suggesting a link between SMPD2 and the tumor immune microenvironment (Fig. [74]3B). To assess the potential role of SMPD2 in immune escape, we analyzed its co-expression with immune checkpoint genes across cancers. SMPD2 was positively correlated with multiple checkpoint molecules, especially in cancers with unfavorable prognosis such as adrenocortical carcinoma (ACC), kidney renal clear cell carcinoma (KIRC), lower-grade glioma (LGG), liver hepatocellular carcinoma (LIHC), and skin cutaneous melanoma (SKCM) (Fig. [75]3C). In addition, we found that SMPD2 expression was significantly associated with tumor mutation burden (TMB) in several cancer types, including STAD, THCA, THYM, ESCA, LGG, LUAD, and PRAD, indicating that SMPD2 may reflect underlying genomic instability or immunogenicity (Fig. [76]3D). To further characterize its immunoregulatory profile, we examined the correlation between SMPD2 and 60 immune checkpoint–related genes (24 inhibitory, 36 stimulatory). The analysis revealed a predominant positive correlation pattern, particularly with inhibitory checkpoint genes such as PDCD1, CTLA4, and LAG3, implying a possible role of SMPD2 in promoting immune suppression (Fig. [77]3E). Finally, using xCell-based deconvolution, we observed that SMPD2 expression was positively correlated with regulatory T cells (Tregs), M2 macrophages, and fibroblasts in multiple cancer types, especially LIHC, LGG, KIRC, and SKCM. Conversely, it was negatively correlated with antitumor immune populations such as CD8⁺ T cells and Th1 cells in several other cancers (Fig. [78]3F). Together, these findings suggest that SMPD2 is closely associated with an immunosuppressive tumor microenvironment and may serve as a marker of immune dysfunction and evasion across multiple cancer types. Fig. 3. [79]Fig. 3 [80]Open in a new tab The SMPD2 expression correlated with immune infiltration. a The SMPD2 expression significantly correlated with the infiltration levels of various immune cells in the TIMER database. b Correlation analyses of the SMPD2 expression with ESTIMATE score in CESC, THCA, and STAD. c Correlation analyses of the SMPD2 expression with immune checkpoint genes in pan-cancer. d The relationship between the SMPD2 gene expression and TMB in diverse tumors e Co-expression analysis between SMPD2 and 60 immune checkpoint-related genes (24 inhibitory, 36 stimulatory) across pan-cancer, using Pearson correlation based on TCGA-TARGET datasets. f Correlation between SMPD2 expression and immune infiltration scores of 67 immune/stromal cell types calculated via xCell deconvolution (IOBR package), across 44 cancer types Transcriptomic and functional pathway alterations associated with SMPD2 overexpression Comparison of gene expression profiles between SMPD2-high and -low tumors revealed marked transcriptional differences. A volcano plot illustrates the global DEG distribution (Fig. [81]3A), and a heatmap of the top 50 genes highlights consistent differential expression patterns (Fig. [82]S2A). Functional enrichment analysis of the upregulated genes revealed strong associations with key metabolic and immune-related pathways. KEGG analysis identified significant enrichment in complement and coagulation cascades, cholesterol metabolism, steroid hormone biosynthesis, bile secretion, and drug metabolism via cytochrome P450 (Fig. [83]3B), indicating potential involvement of SMPD2 in both lipid metabolic reprogramming and immune modulation. In line with this, GO biological process analysis further highlighted enrichment in organic acid biosynthetic process, steroid metabolic process, and small molecule catabolic process, supporting a role for SMPD2 in shaping the metabolic landscape of tumors (Fig. [84]S2B). Collectively, these findings suggest that SMPD2 may contribute to tumor progression by orchestrating transcriptomic programs linked to metabolic adaptation and immunosuppressive microenvironmental remodeling. Fig. 4. [85]Fig. 4 [86]Open in a new tab Transcriptomic and pathway alterations associated with high SMPD2 expression. A Volcano plot showing differentially expressed genes (DEGs) between SMPD2-high and -low expression groups. Significantly upregulated and downregulated genes are highlighted in red and blue, respectively (|log₂FC| ≥ 1, adjusted p < 0.05). B KEGG pathway enrichment analysis of DEGs in the SMPD2-high group. The bar length represents gene count per pathway, and the color indicates the adjusted p-value Epigenetic modifications of SMPD2 is associated with dysfunctional T cell phenotypes and poor prognoses in cancer cohorts Promoter methylation status was analysed and it was found that SMPD2 is hypermethylated in PRAD, while hypomethylated in cancers such as BRCA, BLCA, LUAD, LUSC, THCA, UCEC, and READ (Fig. [87]5A). These findings indicate an inverse relationship between SMPD2 methylation and mRNA expression levels. Hypomethylation of SMPD2 was associated with dysfunctional T cell phenotypes (Fig. [88]5B) and shorter survival durations in brain, melanoma, metastatic melanoma, breast, kidney, and endometrium (Fig. [89]5C). Overall, these results suggest that SMPD2 methylation affects T cell function and prognosis through different mechanisms in various cancer types. Fig. 5. [90]Fig. 5 [91]Open in a new tab Methylation-mediated T cell phenotype dysregulation is associated with poor prognosis. A Boxplots showing differential SMPD2 methylation levels (beta values) between tumor and adjacent normal tissues across TCGA database. B Heatmap showing the roles of SMPD2 methylation in cytotoxic T-cell levels (CTLs), dysfunctional T-cell phenotypes, and risk factors of TCGA cancer cohorts. C Kaplan–Meier curves of overall survival differences between TCGA cancer cohorts with high methylation levels and those with low methylation levels of SMPD2 Association of SMPD2 expression with therapeutic responses The impact of SMPD2 expression on chemotherapeutic responses was investigated and it was found that SMPD2 expression did not significantly affect chemotherapy responses in ovarian cancer and colorectal cancer patients. However, higher SMPD2 expression was associated with favorable trends in treatment response in breast cancer and glioblastoma patients (Fig. [92]6A). Additionally, elevated SMPD2 expression was associated with clinical benefits from PD-L1 immune checkpoint blockade (ICB) in ovarian cancer and colorectal cancer patients (Fig. [93]6B). Fig. 6. [94]Fig. 6 [95]Open in a new tab SMPD2 expression associated with therapeutic responses. A The receiver operating characteristic (ROC) curve plot of the association between SMPD2 expression and responses to chemotherapy in breast, brain, colorectal, and ovarian cancer cohorts. B Kaplan–Meier curves (upper panel) of survival ratios as a measure of the immunotherapeutic response (immune checkpoint blockade) between cancer cohorts with high and those with low expression levels of SMPD2 Verification of SMPD2 expression in vitro and in vivo To further illustrate the expression pattern of SMPD2 in HCC, we investigated SMPD2 expression in HCC tissues collected from 24 cases in our center as stated above. SMPD2 expression in liver tissues from HCC patients was also visualized through immunostaining. We found that the protein level of SMPD2 was overexpressed in HCC tissues in majority of the enrolled patients, thus it was significantly overexpressed in HCC tissues than in general (Figs. [96]7). Moreover, our data also showed that the expression levels of SMPD2 was positively correlated with the expression levels of LAG-3, CTLA-4 and PDL-1 in HCC (Fig. [97]7). These findings provide preliminary in situ evidence suggesting a potential association between SMPD2 expression and an immunosuppressive tumor microenvironment in HCC. Further functional studies are needed to validate these observations. Fig. 7. [98]Fig. 7 [99]Open in a new tab Verification of SMPD2 expression in vitro and in vivo. a, b The expression of SMPD2, LAG3,CTLA-4, and PD-L1 protein was higher in liver cancer than normal liver tissue Conclusion In summary, our pan-cancer analysis revealed that SMPD2 is differentially expressed across multiple cancer types and is significantly associated with patient prognosis, immune features, and therapeutic responses. High SMPD2 expression predicted poorer survival outcomes in cancers such as ACC, KIRC, LGG, LIHC, and SKCM, while lower expression was unfavorable in UCEC and PAAD. SMPD2 expression was positively correlated with immune checkpoint molecules, immune cell infiltration, and tumor mutational burden, suggesting a potential role in immune evasion. Additionally, SMPD2 methylation status was linked to dysfunctional T cell phenotypes and clinical outcomes, further supporting its involvement in immune regulation. Importantly, transcriptomic and enrichment analyses revealed that SMPD2 may contribute to tumor progression by modulating key metabolic and immunosuppressive pathways, including lipid metabolism, steroid biosynthesis, and complement activation. These mechanisms may underlie its impact on therapeutic responses, especially in the context of chemotherapy and immune checkpoint blockade. Overall, SMPD2 emerges as a promising biomarker and potential therapeutic target in cancer. Future studies should aim to experimentally validate its mechanistic roles and assess its clinical utility in precision oncology. Discussion Our pan-cancer integrative analysis revealed that SMPD2 is aberrantly expressed across multiple cancer types and is closely associated with patient prognosis, tumor heterogeneity, and the tumor microenvironment. In several cancers, SMPD2 may act as a tumor-promoting factor, potentially through its roles in immune modulation and genomic instability. Expression patterns of SMPD2 Our analysis of the GTEx and CCLE databases demonstrated that SMPD2 is predominantly expressed in the testis and shows variable expression in different cancer cell lines. Notably, high SMPD2 expression was observed in several cancers, including BLCA, BRCA, CHOL, GBM, KIRP, LIHC, LUSC, PRAD, READ, THCA, and UCEC. This suggests that SMPD2 may play diverse roles depending on the tissue context. The observed low expression in certain cancers, such as HNSC and KICH, further highlight the tissue-specific regulation of SMPD2. Prognostic significance Our survival analysis indicates that SMPD2 expression significantly impacts prognosis in various cancers. High SMPD2 expression was associated with poor survival in ACC, KIRC, LGG, LIHC, and SKCM, suggesting that SMPD2 may act as an oncogene in these cases. Conversely, low SMPD2 expression was linked to poorer outcomes in UCEC and PAAD. These differential effects highlight the complex role of SMPD2 in cancer biology and its potential utility as a prognostic biomarker. Role in immune microenvironment We observed significant positive correlations between SMPD2 expression and key immune checkpoint molecules, including PD-1, PD-L1, and CTLA4, across multiple tumor types. This suggests that SMPD2 may contribute to immune evasion mechanisms and could represent a potential target for immunotherapy [[100]26]. Moreover, distinct immune checkpoint expression patterns were found between tumors with high and low SMPD2 expression, further supporting its potential role in shaping immune phenotypes [[101]27, [102]28]. The association between SMPD2 expression and immune subtypes also indicates its possible utility in stratifying patients for immune-based therapies. Transcriptomic and pathway mechanisms Transcriptome-wide comparisons revealed that high SMPD2 expression is associated with the upregulation of genes involved in lipid metabolism, steroid biosynthesis, and xenobiotic detoxification. These findings suggest that SMPD2 may promote tumor adaptation by enhancing metabolic plasticity, a hallmark of cancer cells that supports survival under stress and therapeutic pressure [[103]29, [104]30]. In parallel, the enrichment of complement and coagulation cascades implies a link to immunosuppressive microenvironment remodeling. Together, these results point to a potential role of SMPD2 in coordinating metabolic and immune-related programs that facilitate tumor progression. Epigenetic modifications Abnormal DNA methylation is an important epigenetic regulator involving tumorigenesis [[105]31], And lead to atypical T cell generation. Our analysis of SMPD2 promoter methylation demonstrated that hypermethylation and hypomethylation are associated with differential expression patterns and prognostic outcomes [[106]25, [107]32]. Specifically, hypomethylation of SMPD2 was linked to dysfunctional T cell phenotypes and poor survival in several cancers, while hypermethylation in breast cancer correlated with longer survival. These findings suggest that epigenetic regulation of SMPD2 may contribute to its diverse functional effects and highlight the need for further investigation into the underlying mechanisms. Therapeutic implications The impact of SMPD2 on therapeutic responses was also explored. While SMPD2 expression did not significantly affect chemotherapeutic responses in glioblastoma, ovarian, and colorectal cancers, it was associated with improved outcomes in breast cancer patients undergoing chemotherapy. Furthermore, higher SMPD2 expression correlated with better responses to PD-L1 immune checkpoint blockade in melanoma and GBM patients. PD-1 (PD) pathway blockade is a highly promising therapy [[108]33, [109]34], These results suggest that SMPD2 could be a valuable biomarker for predicting response to specific therapies, particularly immunotherapy. Validation and prospects In vitro validation of SMPD2 expression in various cancer tissues confirmed its differential expression, with overexpression observed in several tumor types. Notably, SMPD2 was highly expressed in HCC tissues, and its levels positively correlated with PD-L1 and LAG-3, indicating its potential role in creating an immunosuppressive microenvironment. Overall, our study highlights the multifaceted role of SMPD2 in cancer, from influencing tumor progression and immune interactions to affecting therapeutic responses. Future research should focus on elucidating the precise mechanisms by which SMPD2 regulates these processes and explore its potential as a therapeutic target or biomarker in cancer treatment. While our findings provide novel insights into the potential roles of SMPD2 in cancer, several limitations should be considered. First, our analyses were primarily based on large-scale transcriptomic and clinical data from public databases, which may introduce bias due to data heterogeneity. Second, although we identified significant correlations between SMPD2 and immune checkpoint molecules, functional validation through in vitro or in vivo experiments was not feasible in the current study due to experimental constraints. Further experimental studies are warranted to validate these associations and clarify the mechanistic roles of SMPD2 in tumor immunity. Supplementary Information Below is the link to the electronic supplementary material. [110]Supplementary Material 1.^ (179.6KB, jpg) [111]Supplementary Material 2.^ (96.4KB, jpg) Acknowledgements