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.

  • Analysis
  • Published:

Single-cell meta-analyses reveal responses of tumor-reactive CXCL13+ T cells to immune-checkpoint blockade

Abstract

Immune-checkpoint blockade (ICB) therapies represent a paradigm shift in the treatment of human cancers; however, it remains incompletely understood how tumor-reactive T cells respond to ICB across tumor types. Here, we demonstrate that measuring CXCL13 expression could effectively identify both precursor and terminally differentiated tumor-reactive CD8+ T cells within tumors. Applying this approach, we performed meta-analyses of published single-cell data for CXCL13+CD8+ T cells in 225 samples from 102 patients treated with ICB across five cancer types. We found that CXCL13+CD8+ T cells were correlated with favorable responses to ICB, and the treatment further increased such cells in responsive tumors. In addition, CXCL13+ tumor-reactive subsets exhibited variable responses to ICB in distinct contexts, likely due to different degrees of exhaustion-related immunosuppression. Our integrated analyses provide insights into mechanisms underlying ICB and suggest that bolstering precursor tumor-reactive CD8+ T cells might provide an effective therapeutic approach to improve cancer treatment.

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

Access options

Buy this article

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

Fig. 1: CXCL13 as a key marker of tumor-specific CD8+ T cells.
Fig. 2: CXCL13+ tumor-reactive T cells correlate with response to ICB.
Fig. 3: CXCL13+ tumor-reactive T cell subsets.
Fig. 4: The subset distribution of CXCL13+ tumor-reactive T cells.
Fig. 5: Clonal revival and peripheral T cell dynamics.
Fig. 6: Dynamics of tumor-infiltrating CD4+ T cell subsets following treatment.

Similar content being viewed by others

Data availability

Newly generated bulk TCR-seq data that support the findings of this study have been deposited in the Gene Expression Omnibus (GEO) under accession code GSE17994. Previously published scRNA-seq, scTCR-seq and bulk TCR-seq data reanalyzed here are available under accession codes GSE179994 (NSCLC data by Liu et al.6), GSE173321 (NSCLC data by Caushi et al.13), GSE123814 (BCC and SCC data4), GSE169246 (breast cancer data by Zhang et al.7), GSE180268 (HNSCC data by Eberhardt et al.14) and SRZ190804 (RCC data by Krishna et al.20). The breast cancer data by Bassez et al.5 were downloaded from http://biokey.lambrechtslab.org. The RCC data by Bi et al.19 were download from https://singlecell.broadinstitute.org/single_cell/study/SCP1288. An interactive web server for analyzing and visualizing the scRNA-seq data is available at http://metaICB.cancer-pku.cn/. All other relevant data are available from the corresponding authors upon reasonable request. Source data are provided with this paper.

Code availability

All custom code used to generate the results in this study has been deposited in a GitHub repository at https://github.com/PaulingLiu/metaICB.

References

  1. McLane, L. M., Abdel-Hakeem, M. S. & Wherry, E. J. CD8 T cell exhaustion during chronic viral infection and cancer. Annu. Rev. Immunol. 37, 457–495 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Sharma, P. & Allison, J. P. The future of immune checkpoint therapy. Science 348, 56–61 (2015).

    Article  PubMed  Google Scholar 

  3. Hellmann, M. D. et al. Nivolumab plus ipilimumab as first-line treatment for advanced non-small-cell lung cancer (CheckMate 012): results of an open-label, phase 1, multicohort study. Lancet Oncol. 18, 31–41 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Yost, K. E. et al. Clonal replacement of tumor-specific T cells following PD-1 blockade. Nat. Med. 25, 1251–1259 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Bassez, A. et al. A single-cell map of intratumoral changes during anti-PD1 treatment of patients with breast cancer. Nat. Med. 27, 820–832 (2021).

    Article  CAS  PubMed  Google Scholar 

  6. Liu, B. et al. Temporal single-cell tracing reveals clonal revival and expansion of precursor exhausted T cells during anti-PD-1 therapy in lung cancer. Nat. Cancer 3, 108–121 (2022).

    Article  CAS  PubMed  Google Scholar 

  7. Zhang, Y. et al. Single-cell analyses reveal key immune cell subsets associated with response to PD-L1 blockade in triple-negative breast cancer. Cancer Cell 39, 1578–1593 (2021).

  8. Simoni, Y. et al. Bystander CD8+ T cells are abundant and phenotypically distinct in human tumour infiltrates. Nature 557, 575–579 (2018).

    Article  CAS  PubMed  Google Scholar 

  9. Ahmadzadeh, M. et al. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 114, 1537–1544 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Matsuzaki, J. et al. Tumor-infiltrating NY-ESO-1–specific CD8+ T cells are negatively regulated by LAG-3 and PD-1 in human ovarian cancer. Proc. Natl Acad. Sci. USA 107, 7875–7880 (2010).

    Article  PubMed Central  Google Scholar 

  11. Thommen, D. S. et al. A transcriptionally and functionally distinct PD-1+ CD8+ T cell pool with predictive potential in non-small-cell lung cancer treated with PD-1 blockade. Nat. Med. 24, 994–1004 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Duhen, T. et al. Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors. Nat. Commun. 9, 2724 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Caushi, J. X. et al. Transcriptional programs of neoantigen-specific TIL in anti-PD-1-treated lung cancers. Nature 596, 126–132 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Eberhardt, C. S. et al. Functional HPV-specific PD-1+ stem-like CD8 T cells in head and neck cancer. Nature 597, 279–284 (2021).

    Article  CAS  PubMed  Google Scholar 

  15. Oliveira, G. et al. Phenotype, specificity and avidity of antitumour CD8+ T cells in melanoma. Nature 596, 119–125 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Han, A., Glanville, J., Hansmann, L. & Davis, M. M. Linking T-cell receptor sequence to functional phenotype at the single-cell level. Nat. Biotechnol. 32, 684–692 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. van der Leun, A. M., Thommen, D. S. & Schumacher, T. N. CD8+ T cell states in human cancer: insights from single-cell analysis. Nat. Rev. Cancer 20, 218–232 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Workel, H. H. et al. A transcriptionally distinct CXCL13+CD103+CD8+ T-cell population is associated with B-cell recruitment and neoantigen load in human cancer. Cancer Immunol. Res. 7, 784–796 (2019).

    Article  CAS  PubMed  Google Scholar 

  19. Bi, K. et al. Tumor and immune reprogramming during immunotherapy in advanced renal cell carcinoma. Cancer Cell 39, 649–661 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Krishna, C. et al. Single-cell sequencing links multiregional immune landscapes and tissue-resident T cells in ccRCC to tumor topology and therapy efficacy. Cancer Cell 39, 662–677 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Yarchoan, M., Hopkins, A. & Jaffee, E. M. Tumor mutational burden and response rate to PD-1 inhibition. N. Engl. J. Med. 377, 2500–2501 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Zheng, L. et al. Pan-cancer single-cell landscape of tumor-infiltrating T cells. Science 374, abe6474 (2021).

    Article  PubMed  Google Scholar 

  23. Yarchoan, M. et al. PD-L1 expression and tumor mutational burden are independent biomarkers in most cancers. JCI Insight 4, e126908 (2019).

  24. Beltra, J.-C. et al. Developmental relationships of four exhausted CD8+ T cell subsets reveals underlying transcriptional and epigenetic landscape control mechanisms. Immunity 52, 825–841 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Wei, S. C. et al. Distinct cellular mechanisms underlie anti-CTLA-4 and anti-PD-1 checkpoint blockade. Cell 170, 1120–1133 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hudson, W. H. et al. Proliferating transitory T cells with an effector-like transcriptional signature emerge from PD-1+ stem-like CD8+ T cells during chronic Infection. Immunity 51, 1043–1058 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Wykes, M. N. & Lewin, S. R. Immune checkpoint blockade in infectious diseases. Nat. Rev. Immunol. 18, 91–104 (2018).

    Article  CAS  PubMed  Google Scholar 

  28. Wang, J. et al. Fibrinogen-like protein 1 is a major immune inhibitory ligand of LAG-3. Cell 176, 334–347 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Huang, Y.-H. et al. CEACAM1 regulates TIM-3-mediated tolerance and exhaustion. Nature 517, 386–390 (2015).

    Article  CAS  PubMed  Google Scholar 

  30. Dammeijer, F. et al. The PD-1/PD-L1-checkpoint restrains T cell immunity in tumor-draining lymph nodes. Cancer Cell 38, 685–700 (2020).

    Article  CAS  PubMed  Google Scholar 

  31. Chen, D. S. & Mellman, I. Oncology meets immunology: the cancer-immunity cycle. Immunity 39, 1–10 (2013).

    Article  PubMed  Google Scholar 

  32. Muthuswamy, R. et al. NF-κB hyperactivation in tumor tissues allows tumor-selective reprogramming of the chemokine microenvironment to enhance the recruitment of cytolytic T effector cells. Cancer Res. 72, 3735–3743 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Valpione, S. et al. Immune-awakening revealed by peripheral T cell dynamics after one cycle of immunotherapy. Nat. Cancer 1, 210–221 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Zhang, L. et al. Lineage tracking reveals dynamic relationships of T cells in colorectal cancer. Nature 564, 268–272 (2018).

    Article  CAS  PubMed  Google Scholar 

  35. Lowery, F. J. et al. Molecular signatures of antitumor neoantigen-reactive T cells from metastatic human cancers. Science 375, 877–884 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Cohen, M. et al. The interaction of CD4+ helper T cells with dendritic cells shapes the tumor microenvironment and immune checkpoint blockade response. Nat. Cancer 3, 303–317 (2022).

    Article  CAS  PubMed  Google Scholar 

  37. Zheng, C. et al. Transcriptomic profiles of neoantigen-reactive T cells in human gastrointestinal cancers. Cancer Cell 40, 410–423 (2022).

    Article  PubMed  Google Scholar 

  38. Baitsch, L. et al. Exhaustion of tumor-specific CD8+ T cells in metastases from melanoma patients. J. Clin. Invest. 121, 2350–2360 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Li, H. et al. Dysfunctional CD8 T cells form a proliferative, dynamically regulated compartment within human melanoma. Cell 176, 775–789 (2019).

    Article  CAS  PubMed  Google Scholar 

  40. Braun, D. A. et al. Interplay of somatic alterations and immune infiltration modulates response to PD-1 blockade in advanced clear cell renal cell carcinoma. Nat. Med. 26, 909–918 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hahn, W. C. et al. An expanded universe of cancer targets. Cell 184, 1142–1155 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Palmer, A. C., Izar, B., Hwangbo, H. & Sorger, P. K. Predictable clinical benefits without evidence of synergy in trials of combination therapies with immune-checkpoint inhibitors. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 28, 368–377 (2022).

    Article  Google Scholar 

  43. Connolly, K. A. et al. A reservoir of stem-like CD8+ T cells in the tumor-draining lymph node preserves the ongoing antitumor immune response. Sci. Immunol. 6, eabg7836 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Im, S. J. et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Jerby-Arnon, L. et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 175, 984–997 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Shen, M. et al. Pharmacological disruption of the MTDH–SND1 complex enhances tumor antigen presentation and synergizes with anti-PD-1 therapy in metastatic breast cancer. Nat. Cancer 3, 60–74 (2022).

    Article  CAS  PubMed  Google Scholar 

  47. Eisenhauer, E. A. et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur. J. Cancer 45, 228–247 (2009).

    Article  CAS  PubMed  Google Scholar 

  48. Liu, B. et al. An entropy-based metric for assessing the purity of single cell populations. Nat. Commun. 11, 3155 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Polański, K. et al. BBKNN: fast batch alignment of single cell transcriptomes. Bioinformatics 36, 964–965 (2020).

    PubMed  Google Scholar 

  51. 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 

  52. Li, C. et al. SciBet as a portable and fast single cell type identifier. Nat. Commun. 11, 1818 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  54. Bergen, V., Lange, M., Peidli, S., Wolf, F. A. & Theis, F. J. Generalizing RNA velocity to transient cell states through dynamical modeling. Nat. Biotechnol. 38, 1408–1414 (2020).

    Article  CAS  PubMed  Google Scholar 

  55. Harding, F. A., McArthur, J. G., Gross, J. A., Raulet, D. H. & Allison, J. P. CD28-mediated signalling co-stimulates murine T cells and prevents induction of anergy in T-cell clones. Nature 356, 607–609 (1992).

    Article  CAS  PubMed  Google Scholar 

  56. Alquicira-Hernandez, J. & Powell, J. E. Nebulosa recovers single cell gene expression signals by kernel density estimation. Bioinformatics 37, 2485–2487 (2021).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Z.Z. is supported by the National Key R&D Program of China (2020YFE0202200), the National Natural Science Foundation of China (81988101, 91959000, 91942307, 31991171), the Beijing Municipal Science and Technology Commission (Z201100005320014 and Z211100003321005), the Changping Laboratory and the Beijing Advanced Innovation Center for Genomics. Part of the analysis was performed on the High Performance Computing and National Center for Protein Sciences at Peking University.

Author information

Authors and Affiliations

Authors

Contributions

Z.Z. conceived and designed this study. B.L. conceived and performed bioinformatic data analysis and interpreted the results. Y.Z., D.W. and X. H. assisted with the data analysis and provided valuable discussion. B.L. and Z.Z. wrote the manuscript with input from all the authors.

Corresponding author

Correspondence to Zemin Zhang.

Ethics declarations

Competing interests

Z.Z. is a founder of Analytical Biosciences and an advisor for InnoCare Pharma and ArsenalBio. All financial interests are unrelated to this study. The other authors declare no competing interests.

Peer review

Peer review information

Nature Cancer thanks Benjamin Izar and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Characterization of tumor-specific CD8 T cells.

a, Volcano plot showing differentially expressed genes between bystander CD8 T cells and tumor-reactive CD8 T cells from post-ICB non-responsive tumors; N = 6 tumors. Each red dot denotes an individual gene with adjusted P value < 0.01 (two-sided t-test) and fold change ≥ 0.2. b, Volcano plot showing differentially expressed genes between bystander CD8 T cells and tumor-reactive CD8 T cells from N = 13 treatment-naïve tumors. Each red dot denotes an individual gene with adjusted P value < 0.01 (two-sided t-test) and fold change ≥ 0.2. c, Gene expression heatmap for tumor-specific and bystander CD8 T cells. The tumor-reactive CD8 T cells shown here were from N = 13 treatment-naive tumors. d, Average CXCL13 expression in bystander CD8 T cell clones and qualified (clonal size > 3; left) and all (right) tumor-specific CD8 T cell clones from treatment-naïve and post-ICB non-responsive (NR) tumors; N = 19 tumors. e, Performance summary of the CXCL13-selection-based strategy in identifying tumor-reactive CD8 T cells in terms of the clone and cell numbers, corresponding to d. f-h, UMAP plots showing the expression levels of certain signature genes in the three CD8 T cell groups; N = 7 tumors. i, Gene expression heatmap for tumor-specific and bystander CD8 T cells from pre- and post-treatment responsive lung tumors (Liu et al. NSCLC data); N = 7 tumors for both left and right panels.

Source data

Extended Data Fig. 2 Identification of tumor-specific CD8 T cell clones.

a, Determination and classification of n = 31 tumor-specific and n = 57 bystander CD8 T cell clones by using CXCL13 expression and exhaustion score. Data were summarized from pre-treatment responsive BCC and SCC tumors; N = 15 patients. b, ROC curves for the performance of individual exhaustion-related genes and CXCL13 in discriminating tumor-specific from bystander CD8 T cell clones from post-treatment responsive tumors. The antigen specificities of post-treatment CD8 T cell clones were determined based on matched pre-treatment clones in a. The sample size is the same as in a. c, Determination and classification of n = 44 tumor-specific and n = 29 bystander CD8 T cell clones by using CXCL13 expression and exhaustion score. Data were summarized from pre-treatment responsive breast tumors; N = 4 patients. d, ROC curves for the performance of individual exhaustion-related genes and CXCL13 in discriminating tumor-specific from bystander CD8 T cell clones from post-treatment responsive tumors. The antigen specificities of post-treatment CD8 T cell clones were determined based on matched pre-treatment clones in c. The sample size is the same as in c.

Source data

Extended Data Fig. 3 Isolation of CXCL13 + CD8 T cell clones and T cell expansion.

a, Density plots showing the distribution of CXCL13 and CD8A expression across all T cell clones. For each dataset, the threshold for identifying CXCL13- or CD8A-positive clones was determined as the inflection point of the density curve (the vertical line). Data were summarized from n = 20 (the Liu et al. data), 15 (the Caushi et al. data), 22 (BCC), 7 (SCC), 53 (breast cancer cohort 1), 22 (breast cancer cohort 2), 12 (breast cancer cohort 3), 7 (RCC; Krishna et al.) and 4 (RCC; Bi et al.) tumor samples. b, Comparison of the number of expanded CXCL13+ clonotypes (clonal size ≥ 2 and ≥ 20, respectively) between treatment responses. Samples analyzed here were the same as in a, presented as mean ± s.e.m. Two-sided Wilcoxon test.

Source data

Extended Data Fig. 4 Clustering of CXCL13 + tumor-reactive T cells.

a, UMAP plot of 27,155 CXCL13+ CD8 T cells from N = 15 post-ICB lung tumor samples from the Caushi et al. data, color-coded by clusters. b, UMAP plots showing the expression levels of certain signature genes in different cell clusters from a. c, TOX gene expression in the tumor-specific CD8 T cell subsets from N = 20 tumor samples from 10 patients from the Liu et al. NSCLC dataset. d, UMAP plots of CXCL13+ CD8 T cells from pre- and post-ICB tumors for other immunotherapy datasets. N = 20, 22, 7, 53, 22, 7, and 4 tumor samples for the 7 datasets, respectively (from left to right and top to bottom). e, Comparison of the stem-like T cell signature scores and terminally differentiated (TD) signature scores in different cell clusters from a and d, color-coded by clusters. Precursor-like T cells were not identified in the two RCC datasets, and we therefore included the reference data as comparisons when analyzing TD cells. The reference data were from the Eberhardt et al. study and included experimentally validated stem-like and TD cells. Each dot represents one cell while the center line indicates the median value. Data were summarized from n = 13,538, 27,155, 1,944, 4,360, 3,325, 542, 7,290, 3,305 cells from the 8 datasets, respectively. The tumor sample size for each dataset is the same as in a and d. The lower and upper hinges represent the 25th and 75th percentiles, respectively, and whiskers denote 1.5× interquartile range. ***P < 2.2×10−16, two-sided t-test.

Source data

Extended Data Fig. 5 Epigenetic features of CXCL13 + tumor-reactive T cell subsets.

a, ATAC-seq tracks showing the chromatin accessibility in the CD8A, CCR7 and CXCR4 loci for the three CXCL13+ CD8 T cell clusters from N = 4 patients. b, Correlation between the residual tumor size and the proportion of precursor-like cells in all CXCL13+ tumor-reactive CD8 T cells after treatment. Data were summarized from n = 15 patients. P value was determined by two-tailed linear regression t-test.

Source data

Extended Data Fig. 6 Clonal replacement and CXCL13 + tumor-reactive T cell clones in blood.

a, Relationship of clonal replacement with baseline T cell infiltration (left) and the gap of the time between two matched samplings before and after treatment (right). Data were from n = 7 patients from the Liu et al. NSCLC study. P value was determined by a two-tailed linear regression t-test. b, c, Scatter plots of the clone size of each detected CXCL13+ CD8 T cell clone in tumor (y axis) and blood (x axis) for patients with NSCLC (b) and BCC (c); N = 14, 7 and 63 clones for samples NSCLC (P010), NSCLC (P013) and BCC (su001), respectively. Each dot represents one clone. d, Comparison of the number of CXCL13+ tumor-reactive CD8 T cell clonotypes detected in pre- and post-treatment blood for the 3 responsive patients from the Liu et al. NSCLC study. P value was determined by a two-sided paired t-test.

Source data

Extended Data Fig. 7 Dynamics of CD4 T cell subsets following treatment.

a-c, Comparison of the frequency of Tregs (a), other Th (b) and naïve (c) CD4 T cells in all CD4 T cells from pre- and post-treatment responsive and non-responsive tumors. Data were summarized from n = 20, 15, 22, 7, 53, 22, 12, 4 and 7 tumor samples from datasets 1-9, respectively. R, responsive; NR, non-responsive; MPR, major pathologic response. Each dot represents one sample while the center line indicates the median value. The lower and upper hinges represent the 25th and 75th percentiles, respectively, and whiskers denote 1.5× interquartile range.

Source data

Supplementary information

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical Source Data

Rights and permissions

Springer Nature or its licensor 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

Liu, B., Zhang, Y., Wang, D. et al. Single-cell meta-analyses reveal responses of tumor-reactive CXCL13+ T cells to immune-checkpoint blockade. Nat Cancer 3, 1123–1136 (2022). https://doi.org/10.1038/s43018-022-00433-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s43018-022-00433-7

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