Integrated multi-omics analysis identifies CD73 as a prognostic biomarker and immunotherapy response predictor in head and neck squamous cell carcinoma

Background Advances in tumor immunotherapy have been developed for patients with advanced recurrent or metastatic (R/M) HNSCC. However, the response of most HNSCC patients to immune checkpoint inhibitors (ICI) remains unsatisfactory. CD73 is a promising target for tumor immunotherapy, but its role in HNSCC remains insufficient. In this study, we aim to explore the function of CD73 in HNSCC. Methods Transcriptomic and clinical data of TCGA-HNSC were downloaded from UCSC Xena for analysis of CD73 mRNA expression and prognosis. Immunohistochemical assay were performed to validate the expression of CD73 in tumor tissues and its relationship with CD8+ T cells. GSEA analysis was performed with the “clusterProfiler” R package. Immune infiltration analysis was calculated with ESTIMATE, CIBERSORT and MCP-counter algorithms. Single-cell transcriptomic data was originated from GSE103322. Cell clustering, annotation and CD73 expression were from the TISCH database. Correlation data between CD73 and tumor signatures were obtained from the CancerSEA database. Somatic mutation data were obtained from TCGA-HNSC and analyzed by “maftools” R package. Immune efficacy prediction was performed using TIDE algorithm and validated with the IMvigor210 cohort. Results Compared with normal tissues, both mRNA and protein expressions of CD73 were elevated in tumor tissues (P = 9.7×10-10, P = 7.6×10-5, respectively). Kaplan-Meier analysis revealed that patients with high expression of CD73 had worse overall survival (log-rank P = 0.0094), and CD73 could be used as a diagnostic factor for HNSCC (AUC = 0.778). Both bulk RNA-seq and single-cell RNA-seq analysis showed that high CD73 expression can promote EMT and metastasis, samples with high CD73 expression had reduced CD8+ T cells. Furthermore, it was found that CD73-high group was more prone to have mutations in TP53, HRAS and CDKN2A, and were negatively correlated with TMB (P = 0.0055) and MSI (P = 0.00034). Mutational signature analysis found that CD73 was associated with APOBEC signature. Immunotherapy efficacy analysis showed that CD73-high group was less sensitive to immune efficacy. Conclusions Our results demonstrate that CD73 has an inhibitory effect on the tumor microenvironment, and is more likely to be unresponsive to ICI therapy. Collectively, targeting CD73 may provide new insights for tumor targeted therapy and/or immunotherapy.

[1]  Ruixia Wang,et al.  CD73 facilitates invadopodia formation and boosts malignancy of head and neck squamous cell carcinoma via the MAPK signaling pathway , 2022, Cancer science.

[2]  Ming Yan,et al.  CD73 in small extracellular vesicles derived from HNSCC defines tumour‐associated immunosuppression mediated by macrophages in the microenvironment , 2022, Journal of extracellular vesicles.

[3]  Qi Zhang,et al.  Metabolic modulation of immune checkpoints and novel therapeutic strategies in cancer. , 2022, Seminars in cancer biology.

[4]  A. Jemal,et al.  Cancer statistics, 2022 , 2022, CA: a cancer journal for clinicians.

[5]  G. Jin,et al.  Elimination of acquired resistance to PD-1 blockade via the concurrent depletion of tumour cells and immunosuppressive cells , 2021, Nature Biomedical Engineering.

[6]  Xiaochen Bo,et al.  clusterProfiler 4.0: A universal enrichment tool for interpreting omics data , 2021, Innovation.

[7]  C. Supuran,et al.  Small-molecule CD73 inhibitors for the immunotherapy of cancer: a patent and literature review (2017–present) , 2021, Expert opinion on therapeutic patents.

[8]  Zhiyu Qian,et al.  Immunotherapy Strategy Targeting Programmed Cell Death Ligand 1 and CD73 with Macrophage-Derived Mimetic Nanovesicles to Treat Bladder Cancer. , 2021, Molecular pharmaceutics.

[9]  C. R. Leemans,et al.  Head and neck squamous cell carcinoma , 2020, Nature Reviews Disease Primers.

[10]  Fan Zhang,et al.  TISCH: a comprehensive web resource enabling interactive single-cell transcriptome visualization of tumor microenvironment , 2020, bioRxiv.

[11]  Bin Zhang,et al.  Targeting CD73 to augment cancer immunotherapy. , 2020, Current opinion in pharmacology.

[12]  L. Buisseret,et al.  The adenosine pathway in immuno-oncology , 2020, Nature Reviews Clinical Oncology.

[13]  G. Dubyak,et al.  CD8+CD73+ T cells in the tumor microenvironment of head and neck cancer patients are linked to diminished T cell infiltration and activation in tumor tissue , 2020, European journal of immunology.

[14]  Huidong Shi,et al.  CD73 on cancer-associated fibroblasts enhanced by the A2B-mediated feedforward circuit enforces an immune checkpoint , 2020, Nature Communications.

[15]  Hung-Ming Wang,et al.  Pembrolizumab alone or with chemotherapy versus cetuximab with chemotherapy for recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-048): a randomised, open-label, phase 3 study , 2019, The Lancet.

[16]  S. Karam,et al.  Radioimmunotherapy for the treatment of head and neck cancer. , 2019, The Lancet. Oncology.

[17]  Yong Wan,et al.  CD73: an emerging checkpoint for cancer immunotherapy. , 2019, Immunotherapy.

[18]  M. Stratton,et al.  Characterizing Mutational Signatures in Human Cancer Cell Lines Reveals Episodic APOBEC Mutagenesis , 2019, Cell.

[19]  Wei Liu,et al.  CancerSEA: a cancer single-cell state atlas , 2018, Nucleic Acids Res..

[20]  Yassen Assenov,et al.  Maftools: efficient and comprehensive analysis of somatic variants in cancer , 2018, Genome research.

[21]  G. Freeman,et al.  Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response , 2018, Nature Medicine.

[22]  A. Kulkarni,et al.  Specific blockade CD73 alters the “exhausted” phenotype of T cells in head and neck squamous cell carcinoma , 2018, International journal of cancer.

[23]  Anh Nhat Tran,et al.  HPAanalyze: an R package that facilitates the retrieval and analysis of the Human Protein Atlas data , 2018, BMC Bioinformatics.

[24]  L. Emens,et al.  Targeting adenosine for cancer immunotherapy , 2018, Journal of Immunotherapy for Cancer.

[25]  M. Smyth,et al.  Targeting immunosuppressive adenosine in cancer , 2017, Nature Reviews Cancer.

[26]  S. Robson,et al.  The ectonucleotidases CD39 and CD73: Novel checkpoint inhibitor targets , 2017, Immunological reviews.

[27]  R. Bourgon,et al.  Atezolizumab as first-line treatment in cisplatin-ineligible patients with locally advanced and metastatic urothelial carcinoma: a single-arm, multicentre, phase 2 trial , 2017, The Lancet.

[28]  P. Laurent-Puig,et al.  Estimating the population abundance of tissue-infiltrating immune and stromal cell populations using gene expression , 2016, Genome Biology.

[29]  Bin Zhu,et al.  Edinburgh Research Explorer Association of germline variants in the APOBEC3 region with cancer risk and enrichment with APOBEC-signature mutations in tumors , 2022 .

[30]  J. Mesirov,et al.  The Molecular Signatures Database Hallmark Gene Set Collection , 2015 .

[31]  C. Swanton,et al.  APOBEC Enzymes: Mutagenic Fuel for Cancer Evolution and Heterogeneity. , 2015, Cancer discovery.

[32]  Ash A. Alizadeh,et al.  Robust enumeration of cell subsets from tissue expression profiles , 2015, Nature Methods.

[33]  G. von Heijne,et al.  Tissue-based map of the human proteome , 2015, Science.

[34]  Matthew E. Ritchie,et al.  limma powers differential expression analyses for RNA-sequencing and microarray studies , 2015, Nucleic acids research.

[35]  G. Getz,et al.  Inferring tumour purity and stromal and immune cell admixture from expression data , 2013, Nature Communications.

[36]  Guangchuang Yu,et al.  clusterProfiler: an R package for comparing biological themes among gene clusters. , 2012, Omics : a journal of integrative biology.

[37]  C. Cole,et al.  COSMIC: the catalogue of somatic mutations in cancer , 2011, Genome Biology.

[38]  A. McKenna,et al.  The Mutational Landscape of Head and Neck Squamous Cell Carcinoma , 2011, Science.

[39]  Shicai Wang,et al.  COSMIC: the Catalogue Of Somatic Mutations In Cancer , 2018, Nucleic Acids Res..

[40]  Xavier Robin,et al.  pROC: an open-source package for R and S+ to analyze and compare ROC curves , 2011, BMC Bioinformatics.

[41]  Renaud Gaujoux,et al.  A flexible R package for nonnegative matrix factorization , 2010, BMC Bioinformatics.

[42]  Erik K. Malm,et al.  A Human Protein Atlas for Normal and Cancer Tissues Based on Antibody Proteomics* , 2005, Molecular & Cellular Proteomics.