Abstract
Cancer remains a major global health challenge, with incidence and mortality rates continuing to rise. Metabolic reprogramming, a hallmark of cancer, not only sustains rapid proliferation but also shapes an immunosuppressive tumor microenvironment. Among metabolic enzymes, sphingosine kinase 1 (SPHK1) plays a key role in sphingolipid signaling by regulating the balance between sphingosine-1-phosphate and sphingosine. This regulation influences both cell fate and immune responses. However, the role of SPHK1 as a potential “metabolic immune checkpoint” across various cancers, as well as its implications for prognosis and immunotherapy, remains insufficiently explored. In this pan-cancer study, we analyzed SPHK1 expression using RNA-seq data from The Cancer Genome Atlas, which includes 33 cancer types. We also examined its clinical association. We then validated SPHK1 expression at the mRNA and protein levels in clinical samples of head and neck squamous cell carcinoma (HNSC), stomach adenocarcinoma (STAD), and liver hepatocellular carcinoma (LIHC) using RT‑qPCR and immunohistochemistry, and assessed its effect on cancer cell viability using the CCK‑8 assay. Furthermore, we conducted integrated analyses to evaluate the relationship between SPHK1 expression and key immunological features, including immune cell infiltration, tumor mutation burden (TMB), microsatellite instability (MSI), and immune checkpoint gene expression. These analyses aimed to delineate SPHK1’s role in immune modulation. We observed significant upregulation of SPHK1 in multiple cancers, especially in HNSC, STAD, and LIHC. We also confirmed its ability to enhance cancer cell viability. High SPHK1 expression is consistently associated with poor patient survival, supporting its prognostic value. Importantly, comprehensive immunological analyses revealed that SPHK1 expression is closely linked to immunosuppressive features across cancers, including altered immune cell infiltration and elevated expression of established immune checkpoint molecules, positioning SPHK1 as a key regulator linking metabolic dysregulation to immune evasion. Our findings suggest that SPHK1 acts as an oncogene and prognostic biomarker. Additionally, it functions as a novel “metabolic immune checkpoint” across multiple cancer types. SPHK1 may bridge sphingolipid metabolism with tumor immune suppression and represents a potential promising integrated target for metabolically informed immunotherapy strategies.
Similar content being viewed by others
Data availability
The data used for bioinformatic analyses during the current study are original from public databases, including The Cancer Genome Atlas (TCGA) (available at https://portal.gdc.cancer.gov/), the Human Protein Atlas (HPA) (accessible at https://www.proteinatlas.org/), The cBioPortal platform (available at https://www.cbioportal.org/), The UALCAN database (http://ualcan.path.uab.edu/), BioGRID (https://thebiogrid.org/), STRING(https://cn.string-db.org/), GDSC (https://www.cancerrxgene.org/), CTRP (https://portals.broadinstitute.org/ctrp/) and CellMiner (https://discover.nci.nih.gov/cellminer/home.do).The data used for the validation experiment are available from the corresponding author upon reasonable request. All scripts and processed data matrices for this study were uploaded to a public GitHub repository accessible via the following link: https://github.com/xhbuestc/SPHK1-pan-cancer-analysis/tree/main/Code; https://github.com/xhbuestc/SPHK1-pan-cancer-analysis/tree/main/Processed%20matrics.
Abbreviations
- ACC :
-
Adrenocortical carcinoma
- BLCA:
-
Bladder urothelial carcinoma
- BRCA:
-
Breast invasive carcinoma
- CESC:
-
Cervical squamous cell carcinoma and endocervical adenocarcinoma
- CHOL:
-
Cholangiocarcinoma
- COAD:
-
Colon adenocarcinoma
- DLBC:
-
Lymphoid neoplasm diffuse large B-cell lymphoma
- ESCA:
-
Esophageal carcinoma
- GBM:
-
Glioblastoma multiforme
- HNSC:
-
Head and neck squamous cell carcinoma
- KICH:
-
Kidney chromophobe
- KIRC:
-
Kidney renal clear cell carcinoma
- KIRP:
-
Kidney renal papillary cell carcinoma
- LAML:
-
Acute myeloid leukemia
- LGG:
-
Brain lower grade glioma
- LIHC:
-
Liver hepatocellular carcinoma
- LUAD:
-
Lung adenocarcinoma
- LUSC:
-
Lung squamous cell carcinoma
- MESO:
-
Mesothelioma
- OV:
-
Ovarian serous cystadenocarcinoma
- PAAD:
-
Pancreatic adenocarcinoma
- PCPG:
-
Pheochromocytoma and paraganglioma
- PRAD:
-
Prostate adenocarcinoma
- READ:
-
Rectum adenocarcinoma
- SARC:
-
Sarcoma
- SKCM:
-
Skin cutaneous melanoma
- STAD:
-
Stomach adenocarcinoma
- TGCT:
-
Testicular germ cell tumors
- THCA:
-
Thyroid carcinoma
- THYM:
-
Thymoma
- UCEC:
-
Uterine corpus endometrial carcinoma
- UCS:
-
Uterine carcinosarcoma
- UVM:
-
Uveal melanoma
- OS:
-
Overall survival
- DSS:
-
Disease specific survival
- PFI:
-
Progress free interval
- GO:
-
Gene ontology
- KEGG:
-
Kyoto encyclopedia of genes and genomes
- IHC :
-
Immunohistochemistry
- ROC:
-
Receiver operating characteristic
- GDSC:
-
Genomics of drug sensitivity in cancer
- CTRP:
-
The cancer therapeutics response portal
- Cor:
-
Correlation coefficient
- TCGA:
-
The cancer Genome atlas
References
Qi, J. et al. National and subnational trends in cancer burden in China, 2005–20: an analysis of national mortality surveillance data. Lancet Public Health 8, e943–e955. https://doi.org/10.1016/s2468-2667(23)00211-6 (2023).
Bedard, P. L., Hyman, D. M., Davids, M. S. & Siu, L. L. Small molecules, big impact: 20 years of targeted therapy in oncology. Lancet 395, 1078–1088. https://doi.org/10.1016/s0140-6736(20)30164-1 (2020).
Ikeda, H. et al. Immune evasion through mitochondrial transfer in the tumour microenvironment. Nature 638, 225–236. https://doi.org/10.1038/s41586-024-08439-0 (2025).
Chen, Y. et al. Long non-coding RNAs-sphingolipid metabolism nexus: Potential targets for cancer treatment. Pharmacol. Res. 210, 107539. https://doi.org/10.1016/j.phrs.2024.107539 (2024).
Xie, Y. et al. Dysregulation of sphingolipid metabolism contributes to the pathogenesis of chronic myeloid leukemia. Cell Death Dis. 16, 282. https://doi.org/10.1038/s41419-025-07594-0 (2025).
Chen, Y. et al. SPK1/S1P axis confers gastrointestinal stromal tumors (GISTs) resistance of imatinib. Gastric Cancer 26, 26–43. https://doi.org/10.1007/s10120-022-01332-7 (2023).
Gupta, P. et al. Tumor derived extracellular vesicles drive T cell exhaustion in tumor microenvironment through sphingosine mediated signaling and impacting immunotherapy outcomes in ovarian cancer. Adv. Sci. (Weinh) 9, e2104452. https://doi.org/10.1002/advs.202104452 (2022).
Feng, Y. et al. Aberrant lipid metabolism reshapes the immune landscape in bone metastasis of nasopharyngeal carcinoma. J. Immunother. Cancer https://doi.org/10.1136/jitc-2025-012134 (2025).
Chen, D. et al. SPHK1 potentiates colorectal cancer progression and metastasis via regulating autophagy mediated by TRAF6-induced ULK1 ubiquitination. Cancer Gene Ther. 31, 410–419. https://doi.org/10.1038/s41417-023-00711-1 (2024).
Kao, W. H. et al. SPHK1 promotes bladder cancer metastasis via PD-L2/c-Src/FAK signaling cascade. Cell Death Dis 15, 678. https://doi.org/10.1038/s41419-024-07044-3 (2024).
Colaprico, A. et al. TCGAbiolinks: An R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res 44, e71. https://doi.org/10.1093/nar/gkv1507 (2016).
Johnson, W. E., Li, C. & Rabinovic, A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 8, 118–127. https://doi.org/10.1093/biostatistics/kxj037 (2007).
Liao, C. & Wang, X. TCGAplot: an R package for integrative pan-cancer analysis and visualization of TCGA multi-omics data. BMC Bioinformatics 24, 483. https://doi.org/10.1186/s12859-023-05615-3 (2023).
Kanehisa, M., Sato, Y., Kawashima, M., Furumichi, M. & Tanabe, M. KEGG as a reference resource for gene and protein annotation. Nucleic Acids Res. 44, D457-462. https://doi.org/10.1093/nar/gkv1070 (2016).
Kanehisa, M. & Goto, S. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 28, 27–30. https://doi.org/10.1093/nar/28.1.27 (2000).
Kanehisa, M., Furumichi, M., Sato, Y., Matsuura, Y. & Ishiguro-Watanabe, M. KEGG: Biological systems database as a model of the real world. Nucleic Acids Res. 53, D672–D677. https://doi.org/10.1093/nar/gkae909 (2025).
Siegel, R. L., Giaquinto, A. N. & Jemal, A. Cancer statistics, 2024. CA Cancer J. Clin. 74, 12–49. https://doi.org/10.3322/caac.21820 (2024).
Meric-Bernstam, F. et al. Efficacy and safety of Trastuzumab Deruxtecan in patients with HER2-expressing solid tumors: primary results from the DESTINY-PanTumor02 phase II trial. J. Clin. Oncol. 42, 47–58. https://doi.org/10.1200/JCO.23.02005 (2024).
Bae, G. E. et al. Increased sphingosine kinase 1 expression predicts distant metastasis and poor outcome in patients with colorectal cancer. Anticancer Res. 39, 663–670. https://doi.org/10.21873/anticanres.13161 (2019).
Bao, J.-M. et al. Overexpression of SPHK1 associated with targeted therapy resistance in predicting poor prognosis in renal cell carcinoma. Trans. Cancer Res. 12, 572–584. https://doi.org/10.21037/tcr-22-417 (2023).
Zhao, J. et al. ChREBP-mediated choline deprivation and chemokine secretion shape Tumor-associated macrophages to promote immune evasion. Cancer Res. 85, 4701–4717. https://doi.org/10.1158/0008-5472.CAN-25-0235 (2025).
Luca, A. C. et al. Loss of SPHK1 fuels inflammation to drive KRAS-mutated lung adenocarcinoma. Cancer Lett. 623, 217733. https://doi.org/10.1016/j.canlet.2025.217733 (2025).
Zhan, Y. et al. Targeting SPHK1 in macrophages remodels the tumor microenvironment and enhances anti-PD-1 immunotherapy efficacy in colorectal cancer liver metastasis. Cancer Commun. (Lond) 45, 1203–1228. https://doi.org/10.1002/cac2.70047 (2025).
Fang, Q. et al. SPHK1 promotes HNSCC immune evasion by regulating the MMP1-PD-L1 axis. Theranostics 14, 7199–7218. https://doi.org/10.7150/thno.102390 (2024).
Lan, B. et al. Triggering of endoplasmic reticulum stress via ATF4-SPHK1 signaling promotes glioblastoma invasion and chemoresistance. Cell Death Dis. 15, 552. https://doi.org/10.1038/s41419-024-06936-8 (2024).
Qin, Z. et al. SPHK1 contributes to cisplatin resistance in bladder cancer cells via the NONO/STAT3 axis. Int. J. Mol. Med. https://doi.org/10.3892/ijmm.2021.5037 (2021).
Dixit, D. et al. S1PR1 inhibition induces proapoptotic signaling in T cells and limits humoral responses within lymph nodes. J. Clin. Invest. https://doi.org/10.1172/JCI174984 (2024).
Wang, G. et al. Sphingosine 1-phosphate receptor 2 promotes the onset and progression of non-alcoholic fatty liver disease-related hepatocellular carcinoma through the PI3K/AKT/mTOR pathway. Discov. Oncol. 14, 4. https://doi.org/10.1007/s12672-023-00611-8 (2023).
Xu, G., Yang, Z., Sun, Y., Dong, H. & Ma, J. Interaction of microRNAs with sphingosine kinases, sphingosine-1 phosphate, and sphingosine-1 phosphate receptors in cancer. Discov. Oncol. 12, 33. https://doi.org/10.1007/s12672-021-00430-9 (2021).
Grbcic, P. et al. Dual sphingosine kinase inhibitor SKI-II enhances sensitivity to 5-fluorouracil in hepatocellular carcinoma cells via suppression of osteopontin and FAK/IGF-1R signalling. Biochem. Biophys. Res. Commun. 487, 782–788. https://doi.org/10.1016/j.bbrc.2017.04.100 (2017).
Acknowledgements
We thank The Cancer Genome Atlas (TCGA) (available at https://portal.gdc.cancer.gov/), the Human Protein Atlas (HPA) (accessible at https://www.proteinatlas.org/), the cBioPortal platform (available at https://www.cbioportal.org/), the UALCAN database (http://ualcan.path.uab.edu/), BioGRID (https://thebiogrid.org/), STRING (https://cn.string-db.org/), KEGG (www.kegg.jp/kegg/kegg1.html), GDSC (https://www.cancerrxgene.org/), CTRP (https://portals.broadinstitute.org/ctrp/) and CellMiner (https://discover.nci.nih.gov/cellminer/home.do) for providing open datasets for the analyses.
Funding
This study was supported by Sichuan Science and Technology Program (grant number: 2025ZNSFSC0577); Technology Innovation Research and Development Project of Chengdu Science and Technology Bureau (grant number: 2021-YF05-02064-SN); the healthy department of Sichuan Province (grant number: 20PJ116); Fujian Provincial Natural Science Foundation of China (Grant number: 2024J01644); The major Science and Technology Planning Project of Jingmen City, Hubei Province, China (Grant Number: 2023YFZD033).
Author information
Authors and Affiliations
Contributions
Qiuju Wang: Conceptualization, Methodology, Validation, Formal analysis, Investigation, Data curation, Writing Original Draft, Writing—review & editing, Funding acquisition; Lei Wang: Methodology, Validation, Formal analysis, Investigation, Data curation, Writing—review & editing, Visualization, Funding acquisition; Guodong Zhong: Methodology, Validation, Formal analysis, Investigation, Software, Data curation, Writing—review & editing. Hao Luo and Qiao He: Validation, Formal analysis, Investigation, Resources, Data curation, Visualization. Yan Chen: Conceptualization, Methodology, Validation, Formal analysis, Investigation, Resources, Data curation, Writing—review & editing, Visualization, Project administration; Wei Li: Methodology, Validation, Formal analysis, Writing—review & editing, Funding acquisition.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
Below is the link to the electronic supplementary material.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.
About this article
Cite this article
Wang, L., Zhong, G., Luo, H. et al. The emerging role of SPHK1 at the immune-metabolic interface: a pan-cancer integrative analysis. Sci Rep (2026). https://doi.org/10.1038/s41598-026-35350-7
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41598-026-35350-7


