Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Single-cell and spatial transcriptomics reveal mechanisms of radioresistance and immune escape in recurrent nasopharyngeal carcinoma

Abstract

Radiotherapy resistance and immune evasion are prominent features of recurrent nasopharyngeal carcinoma (rNPC). However, their mechanisms remain incompletely understood. Here, we conducted single-cell and spatial transcriptomics analysis of 39 tumors from 24 patients to reveal the microenvironmental differences between primary and rNPC. Specific MCAM+ cancer-associated fibroblasts are significantly enriched in rNPC, where they promote tumor radioresistance through the collagen IV–ITGA2–FAK–AKT axis. Furthermore, we found that collagen IV suppresses the infiltration of T cells, and we identified mechanisms of immune escape in rNPC. We uncovered the presence and function of CD8 ZNF683 cells in rNPC with lower cytotoxicity. The abundance of B cells and tertiary lymphoid structures significantly diminishes in rNPC. Finally, we confirmed that CD47–SIRPα commonly existed between myeloid and malignant cells in rNPC. This study provides an in-depth understanding of the mechanism of radioresistance and immune evasion in rNPC as well as highlighting critical preliminary targets for curing rNPC.

This is a preview of subscription content, access via your institution

Access options

Buy this article

USD 39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: ScRNA-seq and spatial profiling of the pNPC and rNPC TME.
Fig. 2: Characteristics of malignant cells in PT and RT samples and their effects on the tumor ecosystem.
Fig. 3: MCAM-positive fibroblasts were identified in rNPC and were found to promote the radioresistance of NPC tumor cells.
Fig. 4: MCAM+ mCAFs promoted the radioresistance of NPC tumor cells through the COL4–ITGA2–FAK–AKT pathway.
Fig. 5: Intratumoral collagen IV levels were correlated with T cell suppression and immune escape in patients with NPC.
Fig. 6: CD8+ T cells maintain a memory phenotype with weak cytotoxicity in rNPC.
Fig. 7: TLSs were depleted in rNPC.
Fig. 8: SIRPα+ myeloid cells reside near CD47+ malignant cells and may inhibit antigen presentation in rNPC.

Similar content being viewed by others

Data availability

Raw scRNA-seq, spatial transcriptomics and WES data of pNPC and rNPC samples have been deposited in the Genome Sequence Archive of the Beijing Institute of Genomics (BIG) Data Center at the Beijing Institute of Genomics, Chinese Academy of Science, under accession number HRA010466 (accessible at http://bigd.big.ac.cn/gsa-human). Bulk RNA-seq data of human-NPC cell lines after indicated treatment were downloaded from the GSA-Human database under accession number HRA010688 (accessible at http://bigd.big.ac.cn/gsa-human). The WES data for 43 RT and 32 PT previously reported by our team are under accession numbers HRA000052 and HRA000053, which can be accessed at http://bigd.big.ac.cn/gsa-human. The bulk RNA-seq data from 88 PT samples reported in a previous publication26 are under accession number GSE102349. Source data are provided with this paper.

Code availability

The custom code used in this study is available at https://github.com/zhanglabtools/NPC_scRNA_Spatial_code. This repository has been archived at Zenodo (https://doi.org/10.5281/zenodo.15104454)78.

References

  1. Wang, Y. Q. et al. Prognostic significance of tumor-infiltrating lymphocytes in nondisseminated nasopharyngeal carcinoma: a large-scale cohort study. Int. J. Cancer 142, 2558–2566 (2018).

    Article  PubMed  CAS  Google Scholar 

  2. Wagner, J. et al. A single-cell atlas of the tumor and immune ecosystem of human breast cancer. Cell 177, 1330–1345.e18 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Xue, R. et al. Liver tumour immune microenvironment subtypes and neutrophil heterogeneity. Nature 612, 141–147 (2022).

    Article  PubMed  CAS  Google Scholar 

  4. Chen, X. & Song, E. The theory of tumor ecosystem. Cancer Commun. (Lond.) 42, 587–608 (2022).

    Article  PubMed  Google Scholar 

  5. Chua, M. L. K., Wee, J. T. S., Hui, E. P. & Chan, A. T. C. Nasopharyngeal carcinoma. Lancet 387, 1012–1024 (2016).

    Article  PubMed  Google Scholar 

  6. Ng, W. T. et al. Clinical outcomes and patterns of failure after intensity-modulated radiotherapy for nasopharyngeal carcinoma. Int. J. Radiat. Oncol. Biol. Phys. 79, 420–428 (2011).

    Article  PubMed  Google Scholar 

  7. Zhao, C. et al. Locoregional control and mild late toxicity after reducing target volumes and radiation doses in patients with locoregionally advanced nasopharyngeal carcinoma treated with induction chemotherapy (IC) followed by concurrent chemoradiotherapy: 10-year results of a phase 2 study. Int. J. Radiat. Oncol. Biol. Phys. 104, 836–844 (2019).

    Article  PubMed  Google Scholar 

  8. You, R. et al. Hyperfractionation compared with standard fractionation in intensity-modulated radiotherapy for patients with locally advanced recurrent nasopharyngeal carcinoma: a multicentre, randomised, open-label, phase 3 trial. Lancet 401, 917–927 (2023).

    Article  PubMed  Google Scholar 

  9. Hsu, C. et al. Safety and antitumor activity of pembrolizumab in patients with programmed death-ligand 1-positive nasopharyngeal carcinoma: results of the KEYNOTE-028 study. J. Clin. Oncol. 35, 4050–4056 (2017).

    Article  PubMed  CAS  Google Scholar 

  10. Ma, B. B. Y. et al. Antitumor activity of nivolumab in recurrent and metastatic nasopharyngeal carcinoma: an international, multicenter study of the Mayo Clinic Phase 2 Consortium (NCI-9742). J. Clin. Oncol. 36, 1412–1418 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Fang, W. et al. Camrelizumab (SHR-1210) alone or in combination with gemcitabine plus cisplatin for nasopharyngeal carcinoma: results from two single-arm, phase 1 trials. Lancet Oncol. 19, 1338–1350 (2018).

    Article  PubMed  CAS  Google Scholar 

  12. Wang, F. H. et al. Efficacy, safety, and correlative biomarkers of toripalimab in previously treated recurrent or metastatic nasopharyngeal carcinoma: a phase II clinical trial (POLARIS-02). J. Clin. Oncol. 39, 704–712 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. McGranahan, N. et al. Allele-specific HLA loss and immune escape in lung cancer evolution. Cell 171, 1259–1271.e11 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. House, I. G. et al. Macrophage-derived CXCL9 and CXCL10 are required for antitumor immune responses following immune checkpoint blockade. Clin. Cancer Res. 26, 487–504 (2020).

    Article  PubMed  CAS  Google Scholar 

  15. Rosenthal, R. et al. Neoantigen-directed immune escape in lung cancer evolution. Nature 567, 479–485 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Sun, Y. et al. Single-cell landscape of the ecosystem in early-relapse hepatocellular carcinoma. Cell 184, 404–421.e16 (2021).

    Article  PubMed  CAS  Google Scholar 

  17. Azizi, E. et al. Single-cell map of diverse immune phenotypes in the breast tumor microenvironment. Cell 174, 1293–1308.e36 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Lambrechts, D. et al. Phenotype molding of stromal cells in the lung tumor microenvironment. Nat. Med. 24, 1277–1289 (2018).

    Article  PubMed  CAS  Google Scholar 

  19. Marx, V. Method of the year: spatially resolved transcriptomics. Nat. Methods 18, 9–14 (2021).

    Article  PubMed  CAS  Google Scholar 

  20. Chen, Y. P. et al. Single-cell transcriptomics reveals regulators underlying immune cell diversity and immune subtypes associated with prognosis in nasopharyngeal carcinoma. Cell Res. 30, 1024–1042 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Jin, S. et al. Single-cell transcriptomic analysis defines the interplay between tumor cells, viral infection, and the microenvironment in nasopharyngeal carcinoma. Cell Res. 30, 950–965 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Liu, Y. et al. Tumour heterogeneity and intercellular networks of nasopharyngeal carcinoma at single cell resolution. Nat. Commun. 12, 741 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Gong, L. et al. Comprehensive single-cell sequencing reveals the stromal dynamics and tumor-specific characteristics in the microenvironment of nasopharyngeal carcinoma. Nat. Commun. 12, 1540 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. You, R. et al. Clonal mutations activate the NF-κB pathway to promote recurrence of nasopharyngeal carcinoma. Cancer Res. 79, 5930–5943 (2019).

    Article  PubMed  CAS  Google Scholar 

  25. Ding, X. et al. Camrelizumab plus apatinib in patients with recurrent or metastatic nasopharyngeal carcinoma: an open-label, single-arm, phase II study. J. Clin. Oncol. 41, 2571–2583 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Zhang, L. et al. Genomic analysis of nasopharyngeal carcinoma reveals TME-based subtypes. Mol. Cancer Res. 15, 1722–1732 (2017).

    Article  PubMed  CAS  Google Scholar 

  27. Palla, G. et al. Squidpy: a scalable framework for spatial omics analysis. Nat. Methods 19, 171–178 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Chen, Y. et al. Oncogenic collagen I homotrimers from cancer cells bind to α3β1 integrin and impact tumor microbiome and immunity to promote pancreatic cancer. Cancer Cell 40, 818–834.e9 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Kobayashi, H. et al. The origin and contribution of cancer-associated fibroblasts in colorectal carcinogenesis. Gastroenterology 162, 890–906 (2022).

    Article  PubMed  CAS  Google Scholar 

  30. Egeblad, M., Rasch, M. G. & Weaver, V. M. Dynamic interplay between the collagen scaffold and tumor evolution. Curr. Opin. Cell Biol. 22, 697–706 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Shin, H. J., Gil, M. & Lee, I. S. Association of elevated expression levels of COL4A1 in stromal cells with an immunosuppressive tumor microenvironment in low-grade glioma, pancreatic adenocarcinoma, skin cutaneous melanoma, and stomach adenocarcinoma. J. Pers. Med. 12, 534 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Cao, J. et al. The single-cell transcriptional landscape of mammalian organogenesis. Nature 566, 496–502 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Cabrita, R. et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 577, 561–565 (2020).

    Article  PubMed  CAS  Google Scholar 

  34. Ladanyi, A. et al. Prognostic impact of B-cell density in cutaneous melanoma. Cancer Immunol. Immunother. 60, 1729–1738 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Messina, J. L. et al. 12-Chemokine gene signature identifies lymph node-like structures in melanoma: Potential for patient selection for immunotherapy? Sci. Rep. 2, 765 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Cipponi, A. et al. Neogenesis of lymphoid structures and antibody responses occur in human melanoma metastases. Cancer Res. 72, 3997–4007 (2012).

    Article  PubMed  CAS  Google Scholar 

  37. Sautes-Fridman, C., Petitprez, F., Calderaro, J. & Fridman, W. H. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat. Rev. Cancer 19, 307–325 (2019).

    Article  PubMed  CAS  Google Scholar 

  38. Yu, Y. et al. Cancer-associated fibroblasts induce epithelial–mesenchymal transition of breast cancer cells through paracrine TGF-β signalling. Br. J. Cancer 110, 724–732 (2014).

    Article  PubMed  CAS  Google Scholar 

  39. Straussman, R. et al. Tumour micro-environment elicits innate resistance to RAF inhibitors through HGF secretion. Nature 487, 500–504 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Yang, H. et al. Multi-scale integrative analyses identify THBS2+ cancer-associated fibroblasts as a key orchestrator promoting aggressiveness in early-stage lung adenocarcinoma. Theranostics 12, 3104–3130 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Ma, Z. et al. Interferon-dependent SLC14A1+ cancer-associated fibroblasts promote cancer stemness via WNT5A in bladder cancer. Cancer Cell 40, 1550–1565.e7 (2022).

    Article  PubMed  CAS  Google Scholar 

  42. Tang, L. L. et al. The Chinese Society of Clinical Oncology (CSCO) clinical guidelines for the diagnosis and treatment of nasopharyngeal carcinoma. Cancer Commun. (Lond.) 41, 1195–1227 (2021).

    Article  PubMed  Google Scholar 

  43. Melnikova, V. O. & Bar-Eli, M. Bioimmunotherapy for melanoma using fully human antibodies targeting MCAM/MUC18 and IL-8. Pigment Cell Res. 19, 395–405 (2006).

    Article  PubMed  CAS  Google Scholar 

  44. Giannelli, G. et al. Biomarkers and overall survival in patients with advanced hepatocellular carcinoma treated with TGF-βRI inhibitor galunisertib. PLoS ONE 15, e0222259 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Wick, A. et al. Phase 1b/2a study of galunisertib, a small molecule inhibitor of transforming growth factor-beta receptor I, in combination with standard temozolomide-based radiochemotherapy in patients with newly diagnosed malignant glioma. Invest. New Drugs 38, 1570–1579 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Melisi, D. et al. TGFβ receptor inhibitor galunisertib is linked to inflammation- and remodeling-related proteins in patients with pancreatic cancer. Cancer Chemother. Pharmacol. 83, 975–991 (2019).

    Article  PubMed  CAS  Google Scholar 

  47. Yamazaki, T. et al. Galunisertib plus neoadjuvant chemoradiotherapy in patients with locally advanced rectal cancer: a single-arm, phase 2 trial. Lancet Oncol. 23, 1189–1200 (2022).

    Article  PubMed  CAS  Google Scholar 

  48. De, P. et al. An integrin-targeted, pan-isoform, phosphoinositide-3 kinase inhibitor, SF1126, has activity against multiple myeloma in vivo. Cancer Chemother. Pharmacol. 71, 867–881 (2013).

    Article  PubMed  CAS  Google Scholar 

  49. Chien, A. J. et al. MK-2206 and standard neoadjuvant chemotherapy improves response in patients with human epidermal growth factor receptor 2-positive and/or hormone receptor-negative breast cancers in the I-SPY 2 trial. J. Clin. Oncol. 38, 1059–1069 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Badalamente, M. A., Hurst, L. C. & Hentz, V. R. Collagen as a clinical target: nonoperative treatment of Dupuytren’s disease. J. Hand. Surg. Am. 27, 788–798 (2002).

    Article  PubMed  Google Scholar 

  51. Islam, M. S. et al. Extracellular matrix and Hippo signaling as therapeutic targets of antifibrotic compounds for uterine fibroids. Clin. Transl. Med. 11, e475 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Xu, J. Y., Wei, X. L., Wang, Y. Q. & Wang, F. H. Current status and advances of immunotherapy in nasopharyngeal carcinoma. Ther. Adv. Med. Oncol. 14, 17588359221096214 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Zemek, R. M. et al. Sensitization to immune checkpoint blockade through activation of a STAT1/NK axis in the tumor microenvironment. Sci. Transl. Med. 11, eaav7816 (2019).

    Article  PubMed  Google Scholar 

  54. Parry, E. M. et al. ZNF683 marks a CD8+ T cell population associated with anti-tumor immunity following anti-PD-1 therapy for Richter syndrome. Cancer Cell 41, 1803–1816.e8 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Willingham, S. B. et al. The CD47-signal regulatory protein alpha (SIRPa) interaction is a therapeutic target for human solid tumors. Proc. Natl Acad. Sci. USA 109, 6662–6667 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Lakhani, N. J. et al. Evorpacept alone and in combination with pembrolizumab or trastuzumab in patients with advanced solid tumours (ASPEN-01): a first-in-human, open-label, multicentre, phase 1 dose-escalation and dose-expansion study. Lancet Oncol. 22, 1740–1751 (2021).

    Article  PubMed  CAS  Google Scholar 

  57. Advani, R. et al. CD47 blockade by Hu5F9-G4 and rituximab in non-Hodgkin’s lymphoma. N. Engl. J. Med. 379, 1711–1721 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Peluso, M. O. et al. The fully human anti-CD47 antibody SRF231 exerts dual-mechanism antitumor activity via engagement of the activating receptor CD32a. J. Immunother. Cancer 8, e000413 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Ansell, S. M. et al. Phase I study of the CD47 blocker TTI-621 in patients with relapsed or refractory hematologic malignancies. Clin. Cancer Res. 27, 2190–2199 (2021).

    Article  PubMed  CAS  Google Scholar 

  60. Dodagatta-Marri, E. et al. Integrin αvβ8 on T cells suppresses anti-tumor immunity in multiple models and is a promising target for tumor immunotherapy. Cell Rep. 36, 109309 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Wolock, S. L., Lopez, R. & Klein, A. M. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell Syst. 8, 281–291.e9 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Traag, V. A., Waltman, L. & van Eck, N. J. From Louvain to Leiden: guaranteeing well-connected communities. Sci. Rep. 9, 5233 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Becht, E. et al. Dimensionality reduction for visualizing single-cell data using UMAP. Nat. Biotechnol. 37, 38–44 (2019).

    Article  CAS  Google Scholar 

  65. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Tran, H. T. N. et al. A benchmark of batch-effect correction methods for single-cell RNA sequencing data. Genome Biol. 21, 12 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Peng, J. et al. Single-cell RNA-seq highlights intra-tumoral heterogeneity and malignant progression in pancreatic ductal adenocarcinoma. Cell Res. 29, 725–738 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Gulati, G. S. et al. Single-cell transcriptional diversity is a hallmark of developmental potential. Science 367, 405–411 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Yu, G., Wang, L. G., Han, Y. & He, Q. Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 16, 284–287 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Szklarczyk, D. et al. The STRING database in 2021: customizable protein–protein networks, and functional characterization of user-uploaded gene/measurement sets. Nucleic Acids Res. 49, D605–D612 (2021).

    Article  PubMed  CAS  Google Scholar 

  72. Shannon, P. et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 13, 2498–2504 (2003).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Dimitrov, D. et al. Comparison of methods and resources for cell–cell communication inference from single-cell RNA-seq data. Nat. Commun. 13, 3224 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Kolde, R., Laur, S., Adler, P. & Vilo, J. Robust rank aggregation for gene list integration and meta-analysis. Bioinformatics 28, 573–580 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  75. Schubert, M. et al. Perturbation-response genes reveal signaling footprints in cancer gene expression. Nat. Commun. 9, 20 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Badia, I. M. P. et al. decoupleR: ensemble of computational methods to infer biological activities from omics data. Bioinform Adv 2, vbac016 (2022).

    Article  Google Scholar 

  77. Ru, B., Huang, J., Zhang, Y., Aldape, K. & Jiang, P. Estimation of cell lineages in tumors from spatial transcriptomics data. Nat. Commun. 14, 568 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. You, R. et al. Custom code for study: single-cell and spatial transcriptomics reveal mechanisms of radioresistance and immune escape in recurrent nasopharyngeal carcinoma. Zenodo https://doi.org/10.5281/zenodo.15104454 (2025).

Download references

Acknowledgements

Funding was provided by the National Natural Science Foundation of China (no. 82230034 to M.Y.C., 82373407 to R.Y., 82471134 to Y.P.L. and 82203231 to T.L.X.), the Guangzhou Science and Technology Plan Project (no. 202103010001 to M.Y.C.), the National Key Research and Development Program of China (no. 2021YFA1302500 to S.H.Z.), Guangzhou Science and Technology Plan Projects (no. 2023A04J1760 to T.L.X.) and Jiangxi Provincial Natural Science Foundation of China (no. 20212BDH80023 to M.Y.C.).

Author information

Authors and Affiliations

Authors

Contributions

M.Y.C., S.H.Z., T.L.X., Y.P.L. and R.Y. conceived and designed the project and the strategy. R.Y., X.Z., P.Y.H., Y.J.H., Y.P.L. and M.Y.C. collected and created the samples. S.H.Z., Q.L.S., K.N.D., H.S.X., X.Y.S. and R.Q.X. designed and performed the computation and statistical analysis. C.L., X.L., C.L.H., Y.L.X., J.H.W., T.Y., H.F.L., Z.N.W., K.M.C., T.L.X. and R.Y. designed and performed the experiments. W.M.H. conducted the pathological evaluation. M.Y.C., S.H.Z., T.L.X., Y.P.L. and R.Y. supervised the study. All authors wrote, reviewed and edited the manuscript.

Corresponding authors

Correspondence to Rui You, Youping Liu, Tianliang Xia, Shihua Zhang or Mingyuan Chen.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Genetics thanks Wei Dai and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 ScRNA-seq and spatial profiling of the TME of pNPC and rNPC, related to Fig. 1.

(a) Histogram indicating the total number of detected genes (left panel) and the number of read counts (right panel). (b) Violin plot showing the expression levels of 8 known housekeeping genes in all cells. (c) UMAP plot for cells before integration, cells are colored according to the origin of patients (left panel), disease states (middle panel), and cell types (right panel). (d) Boxplot showing the acceptance rate calculated by kBET in uncorrected single cells and harmony-corrected ones. (e) Heatmap of the copy number variants (CNV) analysis, inferred from the scRNA-seq data. Each row indicates a single cell, NK and B cells were used as references. (f) Violin plot showing the CNV scores of inferred malignant cells and normal epithelial cells. Two-sided Wilcoxon rank-sum test. (g) UMAP plot showing the expression levels of marker genes, defined for major cell types. (h) Histogram indicating the ratio of cells in PT (blue) and RT (red) tissue samples. (i) Boxplot showing the number of detected genes of the indicated patients per spot across spatial data. (j-k) Count of programs in PT (blue) and RT (red) samples in Fig. 1f.

Extended Data Fig. 2 Characteristics of malignant cells in PT and RT samples, related to Fig. 2.

(a) Volcano plot shows differentially expressed genes (DEGs) between PT (blue) and RT (red) malignant cells, genes with top10 fold-changes are shown for each group. (b) Gene Set Enrichment Analysis (GSEA) and Gene Ontology of the DEGs between malignant cells from PT (blue) and RT (red) samples, based on the subontology of biological processes. (c) The proportion of significant interactions from fibroblasts to malignant cells compared with significant interactions from other immune cells to malignant cells inferred by LIANA. For fibroblasts, the proportion was calculated using the interaction number between fibroblasts (source) and tumor cells (target) as the numerator and the total interactions sent from fibroblasts as the denominator. For other immune cells, the proportion was calculated using the interaction number between other immune cells (source) and tumor cells (target) as the numerator and the total interactions sent from other immune cells as the denominator. Two-sided proportions z-test. (d) Schematic diagram illustrating the construction of radioresistant cell lines. (e) Representative images of the colony-forming assay of radiation-resistant (R, red) and wild type (W, blue) C666 and TW03 cell lines after being irradiated at doses of 0, 2, 4, 6, and 8 Gy. The right panel indicates the survival fractions of each cell line. Two-sided paired t-test. Scale bars, 5 mm. (f) The Spearman correlation between mutation load and the signature scores of four cancer cell-intrinsic transcriptional programs within PT and RT. Two-sided Spearman’s correlation test. (g) The Spearman correlation between mutation load and the ratios of various cell populations in TME within PT and RT. Two-sided Spearman’s correlation test. (h) Characterization of malignant cells and fibroblasts on spatial transcriptomics data using cell-type deconvolution. Scale bars, 500 µm. (i) Bar chart showing the number of significant L-R interactions from immune cells to malignant cells in PT (blue) and RT (red) samples.

Source data

Extended Data Fig. 3 MCAM+ fibroblasts were identified in rNPC and promoted the radioresistance of NPC tumor cells, related to Fig. 3.

(a) Dotplot showing the normalized expression of marker genes belonging to the clusters defined in Fig. 3a. (b) Track plot showing the normalized expression of mCAF markers (ACTA2, POSTN) and the iCAF marker (CFD) in 4 CAF subclusters. Gene expression is represented by height. (c) Heatmap showing the normalized expression of MCAM and ACTA2 for each cell type. (d) Enrichment analysis of the top 50 DEGs based on the subtypes of CAFs, calculated using Metascape. (e) The primary fibroblasts were cultured from PT (left) and RT (right) samples. The images were taken by a microscope. Scale bars, 400 µm. (f) CAFs from PT and RT tissues were isolated and detected by flow cytometry. (g) MTT assay of NPC cell lines co-cultured with MCAM+ or MCAM CAFs, with PBS as control. Two-sided paired t-test. (h) Representative immunofluorescent staining images show the expression of DAPI, Ki67, and MCAM in MCAM+ mCAFs and MCAM CAFs. Scale bars, 50 µm. Two-sided t-test. (i) TW03 and C666 cells were co-cultured with MCAM+ mCAFs or MCAM CAFs, after irradiation at the doses of 10 Gy, 15 Gy, and 20 Gy. The distribution of γ-H2AX foci was visualized by immunofluorescence staining. The cell nuclei were counter-stained with DAPI (blue). Scale bars, 20 µm. (j) Representative images of the colony-forming assay of NPC cell lines after being co-cultured with MCAM+ mCAFs or MCAM CAFs, followed by different irradiation dose gradients (10 Gy, 15 Gy, and 20 Gy). Scale bars, 5 mm. (k) C666 cells (1 × 106) were injected alone or combined with MCAM+ or MCAM CAFs (3:1) subcutaneously into the haunch of each nude mouse (n = 6 per group). After 6-8 days, all NCG mice were treated with local radiotherapy at a dose of 2 Gy for 5 consecutive days. (l) Representative IHC images immunofluorescent staining images show the expression of PANCK, DAPI, ACTA2, and MCAM in MCAM+ mCAFs and MCAM CAFs. Scale bars, 50 µm.

Source data

Extended Data Fig. 4 MCAM+ mCAFs promoted the radioresistance of NPC cells via the COL4-ITGA2-PI3K-AKT axis, related to Fig. 4.

(a) The expression of representative markers between MCAM+ and MCAM mCAFs cells in the single cell (log (CPM + 1)) and bulk RNA seq cohort (log (FPKM + 1)). CPM, Counts Per Million; FPKM, Fragments Per Kilobase Million. (b-c) The percentage of apoptotic cells in the C666 cell line (b) and the TW03 cell line (c) was quantified by flow cytometry, treated with recombinant PDGFA or IGFBP7 protein, and irradiation at a dose of 10 Gy (n = 3 per group). Quantification represented by a bar plot (mean ± SD), right. Two-sided t-test. (d) Western blotting for the expression of COL4A1 in MCAM+ mCAFs and COL4A1KD MCAM+ mCAFs. (e) ELISA for type IV collagen in the supernatant of MCAM+ CAFs and COL4A1KD MCAM+ mCAFs, ng/ml (n = 3 per group). Two-sided t-test. (f) The correlation between the ratio of malignant cells and the expression of ITGA2 and ITGA3 based on RNA-seq data from Validation cohort 4. Two-sided Spearman’s correlation test. (g) Western blotting for the expression of COL4A1, COL4A2, vimentin, and α-SMA in MCAM+ mCAFs with or without knockdown COL4A1. (h) Representative images of the colony-forming assay of NPC cell lines after being treated with different concentration gradients of Collagen IV followed by irradiation (10 Gy) (n = 3 per group). Quantification represented by a bar plot (mean ± SD), right. Two-sided t-test. Scale bars, 5 mm. (i) Representative images of PANCK, ITGA2, ITGA3, and collagen IV in situ IHC staining in NPC samples. Scale bars, 50 µm. (j) Boxplot showing the expression of ITGA2 (left) and ITGA3 (right) in wide type (W, blue) and radioresistant type (R, Red) cell lines (n = 3 per group). One-sided paired t-test. (k) HEK293T cells were transfected with Flag-COL4A1, HA-ITGA2, or the related vector control and then subjected to immunoprecipitation and immunoblot with the designated antibodies.

Source data

Extended Data Fig. 5 The formation of MCAM+ mCAFs was induced via TGF-β signaling, related to Fig. 4.

(a-b) Western blotting for key proteins of the FAK-AKT pathway in the indicated NPC cell lines, with or without collagen IV pretreatment (a), and with or without ITGA2 knockdown following collagen IV pretreatment (b). (c) TW03 and C666 cells were treated with or without the AKT inhibitor MK2206 in the presence of type IV collagen. After irradiation at a dose of 10 Gy, the distribution of γ-H2AX foci was visualized by immunofluorescence staining (n = 3 per group). The cell nuclei were counter-stained with DAPI (blue). Scale bars, 20 µm. (d) Volcano plot shows DEGs between C666 bulk samples (left) and RT sh-ITGA2 C666 bulk sample (right). The names of the reported irradiation-resistance related TFs are indicated in the plot1-6. (e) Correlation between the expression of TFs which highly expressed in C666 bulk samples and the cell score for FAK-PI3K_AKT in single malignant cells. Two-sided Spearman’s correlation test. (f) Enrichment of the TFs that are highly expressed in C666 bulk samples (versus sh-ITGA2 C666). (g) C666 cells combined with MCAM+ mCAFs (3:1) were injected subcutaneously into NCG mice (n = 6 per group). The mice were randomly subjected to gavage with MK2206 or DMSO, and treated with radiotherapy at a dose of 2 Gy for 5 consecutive days. (h) The deconvolution score of malignant and the score of the TGF-β pathway in malignant spots based on the ST data. Scale bars, 500 µm. (i) Violin plots showing the TGF-β score in malignant cells from PT (blue, n = 3,241) and RT (red, n = 11,105) samples based on the scRNA-seq data. Two-sided Wilcoxon rank-sum test. (j) Boxplot showing the TGF-β scores of NPC cells from wild type (W) and radioresistant tumor cells (R) based on the bulk RNA seq data (n = 3 per group). One-sided paired t-test. (k) Western blotting for key proteins in MCAM CAFs after being treated with recombinant TGF-β in different concentration gradients. (l) MTT assay was performed after MCAM mCAFs cells were treated with increasing doses of TGF-β. Two-sided ANOVA test.

Source data

Extended Data Fig. 6 The construction and validation of HuHSC-NCG mice, related to Fig. 5.

(a) Schematic illustration of the construction process of the humanized mouse model. (b) After establishing the humanized mouse model, the percentage of mCD45 cells, hCD45 cells, hCD19 cells, and hCD3 cells among PBMC was detected in 18 of the mouse models by flow cytometry. (c) Western blotting for the expression of Col4a1 in MCAM+ CAFs and Col4a1 knockout MCAM+ CAFs. (d) Scatter plot showing the correlations between the expression level of COL4A1 and the ratio of CD8+ T, and CD4+ T cells in spatial data. Two-sided Spearman’s correlation test. (e) C666 cells (1×106) were combined with MCAM+ mCAFs with or without COL4A1 KO and injected subcutaneously into HuHSC-NCG mice followed by irradiation (n = 5 per group). Box plot showing the ratio of CD8+T cells and subgroups of CD8+ T cells in the tumor mice using flow cytometry. Two-sided t-test.

Source data

Extended Data Fig. 7 CD8+ T cells maintain a memory phenotype with weak cytotoxicity in rNPC, related to Fig. 6.

(a) Heatmap showing the expression of marker genes for each cluster defined in Fig. 6a. (b) Heatmap indicating the expression of selected gene sets in T/NK subtypes, including naive, resident, inhibitory, cytokines, co-stimulatory, transcriptional factors (TF), and cell type. (c) Boxplots illustrating the ratios of NK subtypes in PT (blue) and RT (red) tumors, respectively. Two-sided Wilcoxon rank-sum test. (d) UMAP plot showing the gene sets for CD8+ T cells, including naive, cytotoxic, and exhausted. (e) Tree map showing the correspondence between the subtypes of CD8+ T cells and other functional CD8+ T cell types of including naive, cytotoxic, and exhausted. (f) Pseudotime-ordered analysis of CD8+ T cells from pNPC and rNPC samples. UMAP plots are labeled by pseudotime (left panel), as well as the naive, cytotoxic, and exhausted CD8+ cell types (right panel). (g) Negative gates of ZNF683 and granzyme B in FACS assays.

Extended Data Fig. 8 Tertiary lymphoid structures are depleted in rNPC, related to Fig. 7.

(a-b) Violin plots showing the percentages of various immune-cell types in TLS (green) and other tissues (orange) in two representative patients. Two-sided Wilcoxon rank-sum test. (c-d) Violin plots showing the percentages of B, CD4+ T, and CD8+ T cell subclusters in TLS (green) and other tissues (orange) in two representative patients. Two-sided Wilcoxon rank-sum test. (e-f) Kaplan-Meier analysis showing the PFS of patients from validation cohort 4, stratified by low (green) or high (blue) expression of MS4A1 (n) and BANK1 (o). Two-sided log-rank test. (g) Kaplan-Meier analysis showing the PFS of patients from validation cohort 4, stratified by low (green) or high (blue) ratio of B cells. Two-sided log-rank test.

Extended Data Fig. 9 SIRPα+ myeloid cells reside near CD47+ malignant cells to inhibit antigen-presentation in rNPC, related to Fig. 8.

(a) UMAP plot showing myeloid cells, color-coded according to PT (blue) and RT (red). (b) t-SNE plot showing the expression of marker genes for the identification of the indicated cell subtypes in Fig. 8a. (c) Scatterplots showing M1 and M2 scores (left panel), as well as pro- and anti-inflammatory scores (right panel) for each color-coded macrophage subtypes. (d) Colocalization scores of TME cells to malignant cells in PT (left) and RT (right). (e) Differences in colocalization scores of TME cells to malignant cells between PT and RT samples. (f) Bar chart showing the number of significant LR interactions from malignant to myeloid subtypes in PT (blue) and RT (red) samples. (g) Scatterplot showing the association between the expression of SIRPA in myeloid subtypes and the distance of these subclusters to CD47+ malignant cells in PT samples, inferred from spatial transcriptomics (ST). Two-sided Spearman’s correlation test.

Extended Data Fig. 10

Schematic diagram of the mechanism of radioresistance and immune escape in rNPC and potential targeted treatment strategies.

Supplementary information

Supplementary Information

Supplementary Notes 1–13, Supplementary Figs. 1–4 and References.

Reporting Summary

Peer Review File

Supplementary Tables 1–7

1. Clinical information for cohort. a, Clinical information of patients in discovery cohort 1. b, Clinical information of patients in validation cohort 1. c, Clinical information of patients in validation cohort 2. 2. All the relevant gene sets used in the current study. 3. Multivariate analysis of the proportions of MCAM+ mCAFs and other clinical characteristics in LRRFS and overall survival. 4. Communication P value and neighborhood enrichment score on spatial transcriptomics data. 5. The proportions and count of leukocytes in the humanized mouse model. 6. The comparison of the percentages of cell types and subtypes in TLS versus other tissues. 7. Relevant sequence used in this article. a, The RT–qPCR sequence for Notch3, Sox4, Atf3, Col4a1, Sox9, and Mcam. b, The siRNA sequence for Notch3, Sox9, Atf2, Atf3, Notch3, Sox4. c, The shRNA sequence for Itga2, Col4a1.

Source data

Source Data Fig. 1

Unprocessed western blots.

Source Data Fig. 3

Source Data from biological experiments.

Source Data Fig. 4

Source Data from biological experiments.

Source Data Fig. 5

Source Data from biological experiments.

Source Data Fig. 6

Source Data from biological experiments.

Source Data Fig. 7

Source Data from biological experiments.

Source Data Fig. 8

Source Data from biological experiments.

Source Data Extended Data Fig. 2

Source Data from biological experiments.

Source Data Extended Data Fig. 3

Source Data from biological experiments.

Source Data Extended Data Fig. 4

Source Data from biological experiments.

Source Data Extended Data Fig. 5

Source Data from biological experiments.

Source Data Extended Data Fig. 6

Source Data from biological experiments.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

You, R., Shen, Q., Lin, C. et al. Single-cell and spatial transcriptomics reveal mechanisms of radioresistance and immune escape in recurrent nasopharyngeal carcinoma. Nat Genet 57, 1950–1965 (2025). https://doi.org/10.1038/s41588-025-02253-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Version of record:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41588-025-02253-8

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer