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:

Intratumoral follicular regulatory T cells curtail anti-PD-1 treatment efficacy

Abstract

Immune-checkpoint blockade (ICB) has shown remarkable clinical success in boosting antitumor immunity. However, the breadth of its cellular targets and specific mode of action remain elusive. We find that tumor-infiltrating follicular regulatory T (TFR) cells are prevalent in tumor tissues of several cancer types. They are primarily located within tertiary lymphoid structures and exhibit superior suppressive capacity and in vivo persistence as compared with regulatory T cells, with which they share a clonal and developmental relationship. In syngeneic tumor models, anti-PD-1 treatment increases the number of tumor-infiltrating TFR cells. Both TFR cell deficiency and the depletion of TFR cells with anti-CTLA-4 before anti-PD-1 treatment improve tumor control in mice. Notably, in a cohort of 271 patients with melanoma, treatment with anti-CTLA-4 followed by anti-PD-1 at progression was associated with better a survival outcome than monotherapy with anti-PD-1 or anti-CTLA-4, anti-PD-1 followed by anti-CTLA-4 at progression or concomitant combination therapy.

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: Tumor-infiltrating TFR cells are highly prevalent in human cancers.
Fig. 2: Tumor-infiltrating TFR cells are primarily located in the TLS.
Fig. 3: Comparison of human tumor-infiltrating TREG and TFR cells.
Fig. 4: Frequency and functional responsiveness of TFR cells in murine tumor models.
Fig. 5: Intratumoral TFR cells gradually increase over time.
Fig. 6: TFR cells are highly responsive to ICB.
Fig. 7: Clinical benefit of sequential ICB.

Similar content being viewed by others

Data availability

Expression data have been deposited in the Gene Expression Omnibus database under the Super Series accession number GSE132297. This Super Series includes data from human and mouse samples. Source data are provided with this paper.

Code availability

Scripts used for this study are available in our repository on GitHub (https://github.com/vijaybioinfo/TFR_2021). An explanation of each is included, as well as version changes.

References

  1. De Simone, M. et al. Transcriptional landscape of human tissue lymphocytes unveils uniqueness of tumor-infiltrating T regulatory cells. Immunity 45, 1135–1147 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Linterman, M. A. et al. Foxp3+ follicular regulatory T cells control the germinal center response. Nat. Med. 17, 975–982 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Sage, P. T., Paterson, A. M., Lovitch, S. B. & Sharpe, A. H. The coinhibitory receptor CTLA-4 controls B cell responses by modulating T follicular helper, T follicular regulatory, and T regulatory cells. Immunity 41, 1026–1039 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Sage, P. T., Alvarez, D., Godec, J., Von Andrian, U. H. & Sharpe, A. H. Circulating T follicular regulatory and helper cells have memory-like properties. J. Clin. Invest. 124, 5191–5204 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Sage, P. T., Francisco, L. M., Carman, C. V. & Sharpe, A. H. The receptor PD-1 controls follicular regulatory T cells in the lymph nodes and blood. Nat. Immunol. 14, 152–161 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Vanderleyden, I. et al. Follicular regulatory T cells can access the germinal center independently of CXCR5. Cell Rep. 30, 611–619 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Brenna, E. et al. CD4+ T follicular helper cells in human tonsils and blood are clonally convergent but divergent from non-Tfh CD4+ cells. Cell Rep. 30, 137–152 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Romão, V. C. et al. Human blood Tfr cells are indicators of ongoing humoral activity not fully licensed with suppressive function. Sci. Immunol. 2, eaan1487 (2017).

    Article  PubMed  Google Scholar 

  9. Ritvo, P.-G. G. et al. Tfr cells lack IL-2Rα but express decoy IL-1R2 and IL-1Ra and suppress the IL-1–dependent activation of Tfh cells. Sci. Immunol. 2, eaan0368 (2017).

    Article  PubMed  Google Scholar 

  10. Botta, D. et al. Dynamic regulation of T follicular regulatory cell responses by interleukin 2 during influenza infection. Nat. Immunol. 18, 1249–1260 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Helmink, B. A. et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 577, 549–555 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  13. Petitprez, F. et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature 577, 556–560 (2020).

    Article  CAS  PubMed  Google Scholar 

  14. Hollern, D. P. et al. B cells and T follicular helper cells mediate response to checkpoint inhibitors in high mutation burden mouse models of breast cancer. Cell 179, 1191–1206 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Chung, Y. et al. Follicular regulatory T cells expressing Foxp3 and Bcl-6 suppress germinal center reactions. Nat. Med. 17, 983–988.

  16. Chen, X. et al. Cutting edge: expression of TNFR2 defines a maximally suppressive subset of mouse CD4+CD25+FoxP3+ T regulatory cells: applicability to tumor-infiltrating T regulatory cells. J. Immunol. 180, 6467–6471 (2008).

    Article  CAS  PubMed  Google Scholar 

  17. Huang, C.-T. et al. Role of LAG-3 in regulatory T cells. Immunity 21, 503–513 (2004).

    Article  CAS  PubMed  Google Scholar 

  18. Joller, N. et al. Treg cells expressing the coinhibitory molecule TIGIT selectively inhibit proinflammatory Th1 and Th17 cell responses. Immunity 40, 569–581 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Hayatsu, N. et al. Analyses of a mutant Foxp3 allele reveal BATF as a critical transcription factor in the differentiation and accumulation of tissue regulatory T cells. Immunity 47, 268–283 (2017).

    Article  CAS  PubMed  Google Scholar 

  20. Guo, X. et al. Global characterization of T cells in non-small-cell lung cancer by single-cell sequencing. Nat. Med. 24, 978–985 (2018).

    Article  CAS  PubMed  Google Scholar 

  21. Plitas, G. et al. Regulatory T cells exhibit distinct features in human breast cancer. Immunity 45, 1122–1134 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Miller, B. C. et al. Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade. Nat. Immunol. 20, 326–336 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. 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 

  24. Utzschneider, D. T. et al. T cell factor 1-expressing memory-like CD8+ T cells sustain the immune response to chronic viral infections. Immunity 45, 415–427 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Siddiqui, I. et al. Intratumoral Tcf1+PD-1+CD8+ T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity 50, 195–211 (2019).

    Article  CAS  PubMed  Google Scholar 

  26. Sade-Feldman, M. et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ahmadzadeh, M. et al. Tumor-infiltrating human CD4+ regulatory T cells display a distinct TCR repertoire and exhibit tumor and neoantigen reactivity. Sci. Immunol. 4, eaao4310 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Xu, L. et al. The kinase mTORC1 promotes the generation and suppressive function of follicular regulatory T cells. Immunity 47, 538–551 (2017).

    Article  CAS  PubMed  Google Scholar 

  29. Kniemeyer, O., Brakhage, A. A., Ferreira, F., Wallner, M. & Sawitzki, B. Regulatory T cell specificity directs tolerance versus allergy against aeroantigens in humans. Cell 167, 1067–1078 (2016).

    Article  PubMed  CAS  Google Scholar 

  30. Szabo, P. A., Miron, M. & Farber, D. L. Location, location, location: tissue resident memory T cells in mice and humans. Sci. Immunol. 4, eaas9673 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Page, N. et al. Expression of the DNA-binding factor TOX promotes the encephalitogenic potential of microbe-induced autoreactive CD8+ T cells. Immunity 48, 937–950 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Scott, A. C. et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 571, 270–274 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Tam, E. M. et al. Antibody-mediated targeting of TNFR2 activates CD8+ T cells in mice and promotes antitumor immunity. Sci. Transl. Med. 11, eaax0720 (2019).

    Article  PubMed  Google Scholar 

  34. Fu, W. et al. Deficiency in T follicular regulatory cells promotes autoimmunity. J. Exp. Med. 215, 815–825 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Wu, H. et al. Follicular regulatory T cells repress cytokine production by follicular helper T cells and optimize IgG responses in mice. Eur. J. Immunol. 46, 1152–1161 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kleffel, S. et al. Melanoma cell-intrinsic PD-1 receptor functions promote tumor growth. Cell 162, 1242–1256 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Zhang, R. et al. An obligate cell-intrinsic function for CD28 in Tregs. J. Clin. Invest. 123, 580–593 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Franckaert, D. et al. Promiscuous Foxp3-cre activity reveals a differential requirement for CD28 in Foxp3+ and Foxp3T cells. Immunol. Cell Biol. 93, 417–423 (2015).

    Article  CAS  PubMed  Google Scholar 

  39. Simpson, T. R. et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J. Exp. Med. 210, 1695–1710 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Wolchok, J. D. et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 377, 1345–1356 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Sage, P. T. et al. Suppression by TFR cells leads to durable and selective inhibition of B cell effector function. Nat. Immunol. 17, 1436–1446 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Champiat, S. et al. Hyperprogressive disease is a new pattern of progression in cancer patients treated by anti-PD-1/PD-L1. Clin. Cancer Res. 23, 1920–1928 (2017).

    Article  CAS  PubMed  Google Scholar 

  43. Knorr, D. A. & Ravetch, J. V. Immunotherapy and hyperprogression: unwanted outcomes, unclear mechanism. Clin. Cancer Res. 25, 904–906 (2019).

    Article  CAS  PubMed  Google Scholar 

  44. Locci, M. et al. Human circulating PD-1+CXCR3CXCR5+ memory Tfh cells are highly functional and correlate with broadly neutralizing HIV antibody responses. Immunity 39, 758–769 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Juneja, V. R. et al. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J. Exp. Med. 214, 895–904 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Webster, K. E. et al. In vivo expansion of Treg cells with IL-2-mAb complexes: induction of resistance to EAE and long-term acceptance of islet allografts without immunosuppression. J. Exp. Med. 206, 751–760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Ganesan, A. P. et al. Tissue-resident memory features are linked to the magnitude of cytotoxic T cell responses in human lung cancer. Nat. Immunol. 18, 940–950 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Singh, D. et al. CD4+ follicular helper-like T cells are key players in anti-tumor immunity. Preprint at bioRxiv https://doi.org/10.1101/2020.01.08.898346 (2020).

  49. Engel, I. et al. Innate-like functions of natural killer T cell subsets result from highly divergent gene programs. Nat. Immunol. 17, 728–739 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    Article  CAS  PubMed  Google Scholar 

  51. Trapnell, C., Pachter, L. & Salzberg, S. L. TopHat: discovering splice junctions with RNA-Seq. Bioinformatics 25, 1105–1111 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows–Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  55. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Patil, V. S. et al. Precursors of human CD4+ cytotoxic T lymphocytes identified by single-cell transcriptome analysis. Sci. Immunol. 3, 8664 (2018).

    Article  Google Scholar 

  58. Langfelder, P. & Horvath, S. WGCNA: an R package for weighted correlation network analysis. BMC Bioinformatics 9, 559 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Mellone, M. et al. Induction of fibroblast senescence generates a non-fibrogenic myofibroblast phenotype that differentially impacts on cancer prognosis. Aging 9, 114–132 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Ottensmeier, C. H. et al. Upregulated glucose metabolism correlates inversely with CD8+ T-cell infiltration and survival in squamous cell carcinoma. Cancer Res. 76, 4136–4148 (2016).

    Article  CAS  PubMed  Google Scholar 

  61. 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 

  62. Zheng, C. et al. Landscape of infiltrating T cells in liver cancer revealed by single-cell sequencing. Cell 169, 1342–1356 (2017).

    Article  CAS  PubMed  Google Scholar 

  63. Savas, P. et al. Single-cell profiling of breast cancer T cells reveals a tissue-resident memory subset associated with improved prognosis. Nat. Med. 24, 986–993 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  65. Puram, S. V. et al. Single-cell transcriptomic analysis of primary and metastatic tumor ecosystems in head and neck cancer. Cell 171, 1611–1624 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Jerby-Arnon, L. et al. A cancer cell program promotes T cell exclusion and resistance to checkpoint blockade. Cell 175, 984–997.e24 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. 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 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Finak, G. et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. 16, 278 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Kharchenko, P. V., Silberstein, L. & Scadden, D. T. Bayesian approach to single-cell differential expression analysis. Nat. Methods 11, 740–742 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Meckiff, B. J. et al. Imbalance of regulatory and cytotoxic SARS-CoV-2-reactive CD4+ T cells in COVID-19. Cell 183, 1340–1353 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank L. Chudley, K. McCann, O. Wood, M. Chamberlain, K. Amer, D. Jeffrey, M. Lane, C. Fixmer, M. Lopez, N. Graham, M. Machado, T. Mellows and B. Johnson for their assistance with the recruitment of study subjects and processing of samples. We thank M. Wheater for providing access to the clinical data from the joint practice with I.K. and C.H.O. We thank A. Upadhye for contributions to the experimental work. We thank J. B. Lilley for his help with the data collection and analysis of the survival cohort. We thank C. Kim, D. Hinz and C. Dillingham for their assistance with cell sorting (FACSAria Fusion Cell Sorter; grant no. S10 RR027366); S. Liang, A. Wang and H. Simon for assistance with library preparation, next-generation sequencing using an Illumina HiSeq 2500 (NIH grant no. S10OD016262) and NovaSeq6000 (grant no. S10OD025052-01). We thank the members of the Vijayanand laboratory for their assistance with editing the figures and manuscript. We thank J. Linden and S. Fuchs for providing B16F10-OVA and MC38-OVA tumor cell lines, respectively. This work was supported by the Wessex Clinical Research Network and the National Institute of Health Research, UK (sample collection), the William K. Bowes Jr Foundation (P.V.), Whitaker Foundation (T.S.-E. and C.H.O.), a Cancer Research UK Centres Network Accelerator Award Grant (grant no. A21998; T.S.-E. and C.H.O.), the Faculty of Medicine of the University of Southampton (T.S.-E. and C.H.O.) and Cancer Research UK (J.C. and C.H.O.). The funders have no role in the study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

S.E., J.C., P.S.F., T.S.-E., F.A., C.H.O. and P.V., conceived the work. S.E. and J.C. performed experiments. S.E., J.C., B.P., C.R.-S. and A.M. analyzed data under the supervision of F.A., C.H.O. and P.V. C.J.H. performed the immunohistochemical analyses under the supervision of G.J.T. A.A., E.W., S.J.C., I.K. and S.E. assisted with patient recruitment, obtaining consent and sample collection. S.E. wrote the first draft of the manuscript that was revised and edited by P.S.F., F.A., C.H.O. and P.V.

Corresponding author

Correspondence to Pandurangan Vijayanand.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available. L. A. Dempsey was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Selection criteria for the integrated single-cell analysis and gating strategies.

a, Violin plots depicting single-cell expression levels for BCL6, CXCR5 and FOXP3 transcripts (left panel) in tumor-infiltrating CD4+ T cells of an exemplary dataset61; dotted lines indicate threshold used for defining positive cells. The scatter plot (right panel) shows expression levels of BCL6 and CXCR5 transcripts in FOXP3-expressing CD4+ T cells b, Gating strategy (surface panel) to sort tumor-infiltrating TREG (LINCD45+CD3+CD4+CXCR5CD127CD25+) and TFR (LINCD45+CD3+CD4+CXCR5+GITR+) cells is shown in the representative FACS plots. c, Gating strategy (intracellular panel) to identify tumor-infiltrating TREG (LINCD45+CD3+CD4+CXCR5FOXP3+BCL-6-) and TFR (LINCD45+CD3+CD4+BCL-6+FOXP3+) cells is shown in the representative FACS plots. d, Representative immunohistochemistry staining for one of the ten NSCLC patients in (Fig. 1d–i) is shown, PanCK (white), CD4 (light blue), CXCR5 (yellow), CD20 (magenta) FOXP3 (green) and BCL-6 (red), scale bars are 25 μm.

Extended Data Fig. 2 Transcriptome analysis of murine TFR cells and characterization of TFR cells in murine tumors.

a, Schematic of immunization model in which mice were immunized intraperitoneally (i.p.) with Ovalbumin in complete Freund’s adjuvant, Ovalbumin in Monophosphoryl Lipid A or mock PBS. b, tSNE plot of TEFF (CD19CD45+CD3+CD4+CXCR5GITRCD25CD62LCD44+), TREG (CD19CD45+CD3+CD4+CXCR5GITR+CD25+), TFH (CD19CD45+CD3+CD4+CXCR5+GITR) and TFR (CD19CD45+CD3+CD4+CXCR5+GITR+). Each symbol represents data from an individual mouse sample (n = 9 for TEFF, n = 11 for TREG, n = 11 for TFH, n = 11 for TFR) that passed quality controls. c, Euler diagrams show the overlap of differentially expressed genes (left, upregulated in TFR, right, downregulated in TFR) in TFR cells compared to the indicated cell types. d, Heatmap comparing gene signatures of TEFF, TREG, TFH and TFR cells. Depicted are transcripts that change in expression more than 2-fold with a DEseq2 adjusted-P value of ≤ 0.05. e, Log-transformed RNA-seq expression values for each of the indicated differentially expressed genes. Each symbol represents an individual sample, data are mean + /− s.e.m. f, Representative histogram plot showing MFI of the surface expression of indicated markers in human tumor-infiltrating TFR cells (n = 4).

Source data

Extended Data Fig. 3 Transcriptome analysis of human tumor-infiltrating TFR cells.

a, Weighted gene co-expression network analysis (WGCNA) depicted as a Topological Overlap Matrix (TOM) heatmap. It included all genes used in the WGCNA analysis and each row and column correspond to a single gene. Red color indicates the degree of topological overlap. The signed network was generated with bulk RNA-seq data of sorted cells enriched for tumor-infiltrating TREG (LINCD45+CD3+CD4+CXCR5CD127CD25+) and TFR (LINCD45+CD3+CD4+CXCR5+GITR+) populations respectively from 10 treatment naïve NSCLC patients (as described in Fig. 2a–d). b, Spearman correlation analysis of the modules identified in (a), depicting module correlation with TFR phenotype. Genes in the pink module are visualized in Gephi, BCL6 and FOXP3 are highlighted. c, Ingenuity pathway analysis of genes in pink module (b). Shown are the top 5 canonical pathways ordered by P value. d, flow cytometric analysis of the frequency (upper panel, P = 0.002 for indicated comparison) and MFI (lower panel, P = 0.002 for indicated comparison) of Ki67-expressing cells, representative histogram plots (right panel) for tumor-infiltrating CD8+ T cells, TREG and TFR cells from n = 10 NSCLC patient samples (described in Fig. 1e,f). e, Heatmap comparing gene expression signatures of enriched population of tumor-infiltrating TREG cells (green) and TFR cells (yellow). Depicted are transcripts that change in expression more than 2-fold with an adjusted-P value of ≤ 0.05. f, Weighted gene co-expression network analysis visualized in Gephi, the nodes are colored and sized according to the number of edges (connections), and the edge thickness is proportional to the edge weight (strength of correlation). The top 10 most differentially expressed genes between TREG and TFR cells are highlighted. g, flow cytometric analysis of the frequency of tumor-infiltrating TCF-1+ TREG and TFR cells from n = 5 NSCLC patient samples, P = 0.0159). Data are mean + /− s.e.m. Significance for comparisons were computed using two-tailed Wilcoxon matched-pairs signed-rank test between TREG and TFR cells (d) or two-tailed Mann–Whitney test (g).

Source data

Extended Data Fig. 4 Cell-trajectory analysis of human TREG and TFR cells from primary tumor tissue and metastasized tumor-infiltrated lymph nodes.

a, Single-cell pseudotime trajectory of cells in cluster 1 (TREG cells) and cluster 6 (TFR cells) (left) or cells from primary tumor tissue or metastatic tumor-infiltrated lymph nodes (right) constructed using the Monocle3 algorithm. b, Normalized gene expression of IL1R2, CCR8, TNFRSF9, TNFRSF18 and PDCD1 on pseudotime path as in (a).

Extended Data Fig. 5 TCR-seq analysis of tumor-infiltrating TREG and TFR cells.

a, the pie chart illustrates the mean percentage of TFR clonotypes that were shared with TREG cells (light blue) and non-TREG cells (gray) respectively, from 4 patients with the highest numbers of clonally expanded FOXP3-expressing cells from a published single-cell RNA-seq dataset20. The lower panel plot displays the percentage of TFR clonotypes that overlap with 4-1BB or 4-1BB+ tumor-infiltrating TREG cells. b, Euler diagram depicting the degree of clonal overlap between TREG, TFH and TFR cells. c, Representative TraCer plot of patient 101020 depicting all clonally expanded cells, color indicates the type of tumor-infiltrating CD4+ T cells: non-TREG (gray, FOXP3), 4-1BB TREG (green), 4-1BB+ TREG (red) and TFR (yellow) cells. d, Single-cell pseudotime trajectory of 4-1BB, 4-1BB+ TREG, clonally expanded, TCR-sharing TREG and TFR cells (indicated with colored circles) constructed using the Monocle3 algorithm. e, Correlation of Monocle component 1 (x-axis) with the genes commonly unregulated in 4-1BB+ TREG, clonally expanded, TCR-sharing TREG and TFR cells compared to 4-1BB TREG cells (y-axis). The solid line represents LOESS fitting between the shared signature and Monocle component 1. f, flow cytometric analysis of the frequency (left panel, P = 0.002 for indicated comparison), MFI (middle panel, P = 0.002 for indicated comparison) for 4-1BB expression in tumor-infiltrating CD8+ T cells, TREG and TFR cells (n = 10 treatment naïve NSCLC patients as in Fig. 2a–d). Data are mean + /− s.e.m. Significance for comparisons were computed using two-tailed Wilcoxon matched-pairs signed-rank test between TREG and TFR cells.

Source data

Extended Data Fig. 6 Characterization of murine TFR cells in an immunization and cancer setting.

a, Gating strategy to identify tumor-infiltrating TREG (CD19CD45+CD3+CD4+BCL-6FOXP3+) and TFR (CD19CD45+CD3+CD4+BCL-6+FOXP3+) cells in B16F10-OVA inoculated mice at d21 (upper panel), shown are representative FACS plots. The FACS plots in the lower panel illustrate intracellular expression of BCL-6 in the indicated cell types (left panel), expression of GITR (middle upper panel), KI-67 (right upper panel), PD-1 (middle lower panel), and CTLA-4 (right lower panel) versus FOXP3 in CD4+ T cells. b, Contour plots depicting the expression levels of FOXP3 in the indicated cell populations from (Fig. 4d). c, Luminex analysis of supernatants from an in vitro proliferation assay (repeat of in vitro suppression assay experiment in Fig. 4g,h), depicted is the concentration of secreted IFN-γ, IL-2 and TNF. d, Flow-cytometric analysis of the frequency of tumor-infiltrating TREG and TFR cells (P = 0.0025 in MC38-OVA, n = 5 mice for day 14 and n = 7 mice for day 21; P = 0.0017 in B16F10-OVA, n = 10 mice for day 14 and n = 6 mice for day 21) in indicated tumor models at indicated time points. Data are mean + /− s.e.m., Significance for comparisons were computed using two-tailed Mann–Whitney test (d). Data in b-d are representative of two independent experiments.

Source data

Extended Data Fig. 7 Human TFR cells are responsive to anti-PD-1 therapy.

a, Heatmap comparing gene signatures of human tumor-infiltrating TFR cells pre- (n = 21 patients) and post- (n = 26 patients) anti-PD-1 therapy20. TFR cells from 5 patients (P2, P3, P12, P15, P20) receiving anti-PD-1 monotherapy were combined. IPA analysis of transcripts (n = 98) more highly expressed post anti-PD-1 treatment (right upper panel) and transcripts that overlap with CD28 signaling, ICOS-ICOSL signaling and T cell receptor signaling are highlighted (right lower panel and heatmap). b-i, Mice were s.c. inoculated with B16F10-OVA cells and treated with tamoxifen (days 5-8 and days 11-14) and anti-PD-1 Abs (day 9). Tumor volume (b,f), TFR cell frequencies (c, P = n.s., g, P = 0.035), eGFP cell frequencies (d, P = 0.0025, h, P = 0.0012) and FOXP3 frequencies (e,i) for n = 6 Foxp3eGFP-cre-ERT2 mice, n = 7 Foxp3eGFP-cre-ERT2/wt x Bcl6fl/fl mice, n = 7 Foxp3eGFP-cre-ERT2 mice and n = 5 Foxp3eGFP-cre-ERT2/wt mice. Data are mean + /− s.e.m., Significance for comparisons were computed using two-tailed Mann–Whitney test (b-i). Data in b-i are representative of two independent experiments.

Source data

Extended Data Fig. 8 Murine TFR cells are depleted by anti-CTLA-4 thereapy.

a,b, Foxp3YFPcre/YFPcre Bcl6fl/fl (TFR knockout) mice or Foxp3YFPcre/YFPcre Bcl6+/+ control mice were s.c. inoculated with B16F10-OVA cells and treated with isotype control or anti-PD-1 Abs at indicated time points, frequency and Ki-67 expression of CD8+ T cells and CD4+ T cells in tumor-draining lymph nodes of mice treated as indicated in, n = 7 mice for ctrl+isotype ctrl, n = 6 mice for ctrl+anti-PD-1, n = 9 mice for the two TFR ko groups. c, Mice were s.c. inoculated with B16F10-OVA or MC38-OVA cells and treated with anti-CTLA-4 Abs at day 10 and day 13. Flow-cytometric analysis of the frequency of tumor-infiltrating TREG and TFR cells, as well as fold depletion of both cell types following anti-CTLA-4 therapy in the B16F10-OVA model (left panel, n = 9 mice, P = 0.0435) and MC38-OVA model (right panel, n = 5 mice, P = 0.0079). d, Survival curves of an independent cohort of melanoma patients (n = 29) stratified into TFRhi (>5.075% of CD4+ cells co-expressing FOXP3 and BCL-6) and TFRlo (<5.075% of cells co-expressing FOXP3 and BCL-6) e, IHC analysis of the frequency of FOXP3+BCL6+ TFR cells with a cutoff (orange line) set to upper limit of normal of 5.075% pertaining to (Extended Data Fig. 8d), P = 0.0654. f, Survival curves of melanoma patients stratified into CXCR5hi (frequency of CXCR5 + cells >8.336%) and CXCR5lo (frequency of CXCR5 + cells <8.336%). g, IHC analysis of the frequency of CXCR5 + cells with a cutoff (orange line) set to upper limit of normal of 8.375% pertaining to (Extended Data Fig. 8f), P = 0.0002. Data are mean + /− s.e.m., Significance for comparisons were computed using two-tailed Mann–Whitney test (c,e,g) or Mantel–Cox test (d,f). Data in (a–c) are representative of two independent experiments.

Source data

Supplementary information

Source data

Source Data Fig. 1

Source data integrated analysis.

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 Fig. 7

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. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Eschweiler, S., Clarke, J., Ramírez-Suástegui, C. et al. Intratumoral follicular regulatory T cells curtail anti-PD-1 treatment efficacy. Nat Immunol 22, 1052–1063 (2021). https://doi.org/10.1038/s41590-021-00958-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-00958-6

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