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:

Recruitment and activation of type 3 innate lymphoid cells promote antitumor immune responses

Abstract

Tumors poorly infiltrated by T cells are more resistant to immunogenic chemotherapies and checkpoint inhibition than highly infiltrated tumors. Using murine models, we found that CCR6+ type 3 innate lymphoid cells (ILC3s) can trigger an increase in the number of T cells infiltrating a tumor. Shortly after administration of cisplatin chemotherapy, production of the chemokine CCL20 and proinflammatory cytokine IL-1β at the tumor site led to the recruitment and activation of ILC3s. Within the tumor, ILC3 production of the chemokine CXCL10 was responsible for the recruitment of CD4+ and CD8+ T lymphocytes to the tumor. ILC3-dependent infiltration of T cells was essential for antitumor immune responses and increased the efficacy of checkpoint inhibition. Thus, we reveal an essential role of CCL20 and IL-1β, which promote ILC3-dependent antitumor immunity and enhance tumor sensitivity to immunotherapy.

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: ILC3s are increased early in the tumor bed after platin-based chemotherapy.
Fig. 2: ILC3s are essential for the antitumor efficacy of cisplatin.
Fig. 3: ILC3s are crucial actors in the antitumor efficacy of immunotherapies targeting immune checkpoint inhibitors.
Fig. 4: ILC3 production of CXCL10 recruits CD4+ and CD8+ T lymphocytes to the tumor.
Fig. 5: CCL20 is involved in the recruitment of ILC3s to the tumor.
Fig. 6: IL-1β activates ILC3s in the tumor.
Fig. 7: CCL20 and IL-1β can turn a cold tumor hot.

Similar content being viewed by others

Data availability

The RNA-seq data generated in this paper have been deposited in the Gene Expression Omnibus database under accession number GSE171656. All other data needed to evaluate the conclusions in the paper are reported in the article or source data files. Source data are provided with this paper.

References

  1. Galon, J. et al. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science 313, 1960–1964 (2006).

    Article  CAS  PubMed  Google Scholar 

  2. Larkin, J. et al. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N. Engl. J. Med. 381, 1535–1546 (2019).

    Article  CAS  PubMed  Google Scholar 

  3. Tumeh, P. C. et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515, 568–571 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Matsutani, S. et al. Significance of tumor-infiltrating lymphocytes before and after neoadjuvant therapy for rectal cancer. Cancer Sci. 109, 966–979 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Irshad, S. et al. RORγt+ innate lymphoid cells promote lymph node metastasis of breast cancers.Cancer Res. 77, 1083–1096 (2017).

    Article  CAS  PubMed  Google Scholar 

  6. Kirchberger, S. et al. Innate lymphoid cells sustain colon cancer through production of interleukin-22 in a mouse model. J. Exp. Med. 210, 917–931 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Eisenring, M., vom Berg, J., Kristiansen, G., Saller, E. & Becher, B. IL-12 initiates tumor rejection via lymphoid tissue-inducer cells bearing the natural cytotoxicity receptor NKp46. Nat. Immunol. 11, 1030–1038 (2010).

    Article  CAS  PubMed  Google Scholar 

  8. Nussbaum, K. et al. Tissue microenvironment dictates the fate and tumor-suppressive function of type 3 ILCs. J. Exp. Med. 214, 2331–2347 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Dieu-Nosjean, M.-C. et al. Long-term survival for patients with non-small-cell lung cancer with intratumoral lymphoid structures. J. Clin. Oncol. 26, 4410–4417 (2008).

    Article  CAS  PubMed  Google Scholar 

  10. Ben-Sasson, S. Z. et al. IL-1 enhances expansion, effector function, tissue localization, and memory response of antigen-specific CD8 T cells. J. Exp. Med. 210, 491–502 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Voronov, E. & Apte, R. N. IL-1 in colon inflammation, colon carcinogenesis and invasiveness of colon cancer. Cancer Microenviron. 8, 187–200 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Mortha, A. et al. Microbiota-dependent crosstalk between macrophages and ILC3 promotes intestinal homeostasis. Science 343, 1249288 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Zhou, L. et al. Innate lymphoid cells support regulatory T cells in the intestine through interleukin-2. Nature 568, 405–409 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Hirota, K. et al. Preferential recruitment of CCR6-expressing Th17 cells to inflamed joints via CCL20 in rheumatoid arthritis and its animal model. J. Exp. Med. 204, 2803–2812 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Kleinewietfeld, M. et al. CCR6 expression defines regulatory effector/memory-like cells within the CD25+CD4+ T-cell subset. Blood 105, 2877–2886 (2005).

    Article  CAS  PubMed  Google Scholar 

  16. Ito, T., Carson, W. F., Cavassani, K. A., Connett, J. M. & Kunkel, S. L. CCR6 as a mediator of immunity in the lung and gut. Exp. Cell. Res. 317, 613–619 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Li, Z. et al. Epidermal Notch1 recruits RORγ+ group 3 innate lymphoid cells to orchestrate normal skin repair. Nat. Commun. 7, 11394 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Gandhi, L. et al. Pembrolizumab plus chemotherapy in metastatic non-small-cell lung cancer. N. Engl. J. Med. 378, 2078–2092 (2018).

    Article  CAS  PubMed  Google Scholar 

  19. Withers, D. R. et al. Transient inhibition of ROR-γt therapeutically limits intestinal inflammation by reducing TH17 cells and preserving group 3 innate lymphoid cells. Nat. Med. 22, 319–323 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Huh, J. R. et al. Digoxin and its derivatives suppress TH17 cell differentiation by antagonizing RORγt activity. Nature 472, 486–490 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Xiao, S. et al. Small-molecule RORγt antagonists inhibit T helper 17 cell transcriptional network by divergent mechanisms. Immunity 40, 477–489 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Mebius, R. E., Rennert, P. & Weissman, I. L. Developing lymph nodes collect CD4+CD3- LTbeta+ cells that can differentiate to APC, NK cells, and follicular cells but not T or B cells. Immunity 7, 493–504 (1997).

    Article  CAS  PubMed  Google Scholar 

  23. Carrega, P. et al. NCR+ ILC3 concentrate in human lung cancer and associate with intratumoral lymphoid structures. Nat. Commun. 6, 8280 (2015).

    Article  CAS  PubMed  Google Scholar 

  24. Maurice, N. J., McElrath, M. J., Andersen-Nissen, E., Frahm, N. & Prlic, M. CXCR3 enables recruitment and site-specific bystander activation of memory CD8+ T cells. Nat. Commun. 10, 4987 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Takatori, H. et al. Lymphoid tissue inducer-like cells are an innate source of IL-17 and IL-22. J. Exp. Med. 206, 35–41 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Godec, J. et al. Compendium of immune signatures identifies conserved and species-specific biology in response to inflammation. Immunity 44, 194–206 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Paget, C. et al. CD3bright signals on γδ T cells identify IL-17A-producing Vγ6Vδ1+ T cells. Immunol. Cell Biol. 93, 198–212 (2015).

    Article  CAS  PubMed  Google Scholar 

  28. Koh, J. et al. IL23-producing human lung cancer cells promote tumor growth via conversion of innate lymphoid cell 1 (ILC1) into ILC3. Clin. Cancer Res. 25, 4026–4037 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Munneke, J. M. et al. Activated innate lymphoid cells are associated with a reduced susceptibility to graft-versus-host disease. Blood 124, 812–821 (2014).

    Article  CAS  PubMed  Google Scholar 

  30. Trabanelli, S. et al. CD127+ innate lymphoid cells are dysregulated in treatment naïve acute myeloid leukemia patients at diagnosis. Haematologica 100, e257–e260 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Kuo, P. T. et al. The role of CXCR3 and its chemokine ligands in skin disease and cancer. Front. Med. 5, 271 (2018).

    Article  Google Scholar 

  32. Sato, Y. et al. CXCL10 expression status is prognostic in patients with advanced thoracic esophageal squamous cell carcinoma. Ann. Surg. Oncol. 23, 936–942 (2016).

    Article  PubMed  Google Scholar 

  33. Sonnenberg, G. F., Monticelli, L. A., Elloso, M. M., Fouser, L. A. & Artis, D. CD4+ lymphoid tissue-inducer cells promote innate immunity in the gut. Immunity 34, 122–134 (2011).

    Article  CAS  PubMed  Google Scholar 

  34. Ranasinghe, R. & Eri, R. Modulation of the CCR6-CCL20 axis: a potential therapeutic target in inflammation and cancer. Medicina (Kaunas) 54, 88 (2018).

    Article  Google Scholar 

  35. Wang, D. et al. Colorectal cancer cell-derived CCL20 recruits regulatory T cells to promote chemoresistance via FOXO1/CEBPB/NF-κB signaling. J. Immunother. Cancer 7, 215 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  36. Spranger, S., Dai, D., Horton, B. & Gajewski, T. F. Tumor-residing Batf3 dendritic cells are required for effector T cell trafficking and adoptive T cell therapy. Cancer Cell 31, 711–723 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Rébé, C. & Ghiringhelli, F. Interleukin-1β and cancer. Cancers 12, 1791 (2020).

    Article  PubMed Central  Google Scholar 

  38. Ridker, P. M. et al. Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: exploratory results from a randomised, double-blind, placebo-controlled trial. Lancet 390, 1833–1842 (2017).

    Article  CAS  PubMed  Google Scholar 

  39. Bruchard, M. et al. Chemotherapy-triggered cathepsin B release in myeloid-derived suppressor cells activates the Nlrp3 inflammasome and promotes tumor growth. Nat. Med. 19, 57–64 (2013).

    Article  CAS  PubMed  Google Scholar 

  40. Ghiringhelli, F. et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1β–dependent adaptive immunity against tumors. Nat. Med. 15, 1170–1178 (2009).

    Article  CAS  PubMed  Google Scholar 

  41. Martínez-Reza, I. et al. Calcitriol inhibits the proliferation of triple-negative breast cancer cells through a mechanism involving the proinflammatory cytokines IL-1 β and TNF- α. J. Immunol. Res. 2019, 6384278 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Mitsunaga, S. et al. Serum levels of IL-6 and IL-1β can predict the efficacy of gemcitabine in patients with advanced pancreatic cancer. Br. J. Cancer 108, 2063–2069 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Kim, J.-W. et al. Clinical implications of VEGF, TGF-β1, and IL-1β in patients with advanced non-small cell lung cancer. Cancer Res. Treat. 45, 325–333 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank T.C. Wu (Johns Hopkins School of Medicine, Baltimore, MD) for providing TC-1 cells. This work was supported by the Association pour la Recherche sur le Cancer (M.B. and F.G.), an ISITE-BFC senior fellowship (F.G.), LabEx LipSTIC (Lipoprotéines et Santé) grant ANR-11-LABX0021 (F.G.), Ligue Contre Le Cancer CCIR EST (F.V.,M.B. and F.G.) and the Fondation de France (M.B.). F.G.’s team is supported by Equipe labélisée Ligue Nationale Contre le Cancer.

Author information

Authors and Affiliations

Authors

Contributions

M.B. contributed to conceptualization, investigation, visualization, writing and funding acquisition. M.G. contributed to investigation and validation. R.B., A.P., C.H., E. Barsac, C. Racoeur and F.C., C. Paul, C. Rébé contributed to investigation. E. Ballot and C.T. contributed to formal analysis. B.R., T.B. and C. Paget contributed resources. F.V. supervised the study and acquired funding. F.G. supervised the study and contributed to funding acquisition, conceptualization and writing.

Corresponding author

Correspondence to Mélanie Bruchard.

Ethics declarations

Competing interests

F.G. received honoraria for oral communication from Lilly, Sanofi, BMS, Astra Zeneca and Amgen; received funding for clinical trials by Astra Zeneca; received a travel grant from Roche France, Amgen and Servier; and is an advisory board member for Merck Serano, Amgen, Roche France and Sanofi. All other authors have no competing interests.

Peer review

Peer review information

Nature Immunology thanks Kingston Mills, Shigeo Koyasu and Cyril Seillet for their contribution to the peer review of this work. Zoltan Fehervari was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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 Cisplatin induced immune recruitment to the poorly immune infiltrated cancer TC-1 model.

Therapeutic response of WT mice bearing a a, TC-1 n = 7, b, MC38, n = 5 and c, 4T1 n = 10 tumors treated with ICB on days 10-12-14, reflected by tumor growth kinetics. d, Therapeutic response of WT mice bearing urethane induced lung tumors treated with α-PD-L1, as reflected by the number of tumor foci. n = 9, results are shown as mean ± SEM. Flow cytometry analysis of e, CD45+ cells and f, CD4+ cells and CD8+ cells from TC-1, 4T1, urethane carcinogen induced model and MC38 used as positive control untreated tumors. Results show a number of cells for 300 000 total viable cells from each tumor, n = 5. g–n Flow cytometry analysis of immune cells infiltrating TC-1 tumors of WT mice treated with CDDP. Tumors were harvested and analyzed one, three or seven days after CDDP. Results show the numbers of cells from 300 000 total viable cells from each tumor. n = 5. Numbers of g, CD45+ cells. h, CD8+ cells. i, CD4+ cells. j, B cells. k, CD11c+ cells. l, CD11b+ cells. m, Myeloid-derived suppressor cells (MDSCs). n, TCRγδ+ cells. o–p two-dimensional tSNE representation of ILCs separated in 5 clusters from TC-1 tumors of WT mice treated with CDDP. Tumors were harvested and analyzed one, three or seven days after CDDP, n = 5. o, tSNE from all sequenced cells or p, separated by time points. Results are shown as mean ± SD and are representative of three independent experiments. Statistical significance was determined using two tailed multiple unpaired T Test (ac), two tailed unpaired T test (d), one-way anova (e) and two-way anova and Tukey’s multiple comparison test (fn).

Source data

Extended Data Fig. 2 Gating strategies.

Gating strategies of flow cytometry data used throughout the study of a, ILCs, b, lymphocytes and c, DCs, myeloid cells and eosinophils.

Extended Data Fig. 3 Effects of oxaliplatin and carboplatin on the immune infiltrate of the TC-1 tumor.

Therapeutic responses of WT and nude mice bearing a TC-1 tumor treated with a, Cisplatin (CDDP) n = 8, b, Carboplatin (Cb) n = 6 or c, Oxaliplatin (Ox) n = 6, reflected by tumor growth kinetics. Results are shown as mean ± SEM. d–l Flow cytometry analysis of immune cells infiltrating the tumor one and seven days after an Ox or Cb treatment. Numbers are shown for 300 000 total viable cells from each tumor. n = 5. Numbers of d, ILC1s, e, ILC2s, f, CD45+ cells, g, CD8+ cells, h, CD4+ cells, i, B cells, j, CD11c+ cells, k, CD11b+ cells, l, MDSCs. Results are shown as mean ± SD and are representative of three independent experiments. Statistical significance was determined using two-way anova and Tukey’s multiple comparison test (al).

Source data

Extended Data Fig. 4 Effects of paclitaxel, doxorubicin and 5-fluorouracil on the immune infiltrate of the TC-1 tumor.

a–g, Flow cytometry analysis of immune cells infiltrating the tumor seven days after a Paclitaxel (Paclitxl), a Doxorubicine (Doxo) or a 5FU treatment. Numbers are shown for 300 000 total viable cells from each TC-1 tumor. n = 5. Numbers of a, ILC3s one and seven days after treatment, b, CD45+ cells, c, CD4+ and CD8+ cells, d, B cells, e, CD11c+ cells, f, CD11b+ cells, g, MDSCs. h, Therapeutic responses of WT mice bearing a 4T1 tumor treated with CDDP at day 10, reflected by tumor growth kinetics. n = 7. Tumor growth results are shown as mean ± SEM. i–o Flow cytometry analysis of immune cells infiltrating the tumor one and seven days after CDDP. Numbers are shown for 300 000 total viable cells from each 4T1 tumors. n = 5. Numbers of i, CD45+ cells, j, CCR6+ ILC3s, k, CD4+ and CD8+ cells, l, B cells, m, CD11c+ cells, n, CD11b+ cells, o, MDSCs. Results are shown as mean ± SD and are representative of three independent experiments. Statistical significance was determined using two-way anova and Tukey’s multiple comparison test (a, c, g, io), one-way anova (b, df) or using two tailed multiple unpaired T Test (h).

Source data

Extended Data Fig. 5 Importance of RORγt+ cells in the anti-tumor immune response.

Flow cytometry analysis of a, RORγt+IL-17+ Th17 and b, CCR6+ and NKP46+ IL-17+ ILC3s cells from WT mice intestine treated with PBS, GSK805 or Digoxin n = 5 and of c, RORγt+ Th17 and d, CCR6+ ILC3 from TC-1 tumor seven days after CDDP treated with Digoxin or GSK805, n = 7. Numbers for 300 000 CD45+ cells. e-p Flow cytometry analysis of immune cells infiltrating TC-1 tumors one, three and seven days after a CDDP and or Digoxin treatment. Numbers are shown for 300 000 total viable cells from each TC-1 tumor. n = 7. Numbers of e, ILC3s, f, ILC2s, g, ILC1s, h, NKs, i, CD8 + cells, j, CD4 + cells, k, B cells, l, DCs, m, macrophages, n, M-MDSCs, o, PMN-MDSCs and p, eosinophils. Results are shown as mean ± SD and are representative of three independent experiments. Statistical significance was determined using one-way anova (ad) and two-way anova and Tukey’s multiple comparison test (eo).

Source data

Extended Data Fig. 6 Importance of ILC3s in the antitumor immune response.

a, Therapeutic response of WT mice bearing urethane induced lung carcinoma to CDDP, Digoxin and GSK805 fourteen days after the CDDP treatment, reflected by the number of tumor foci, n = 10. b, Therapeutic response of BALB/c WT mice bearing 4T1 tumors to CDDP alone or in combination with Digoxin as reflected by tumor growth kinetics. n = 5. c, Flow cytometry analysis of the numbers of NK and ILCs from TC-1 tumors. Numbers are shown for 300 000 total viable cells from each TC-1 tumor, n = 5. d–g, Flow cytometry analysis of immune populations in the spleen of Rorcfl/flCd4cre and littermate control mice, n = 5. Percentages of d, CCR6+ ILC3s in ILCs and of e, Th17 in CD4+ cells from the spleen. f, flow cytometry analysis of the numbers of CCR6+ ILC3 in TC-1 tumors of Rorcfl/flCd4cre mice or littermate control mice. n = 5 g, Percentage of Th17 cells found in the spleen of Rorcfl/flCd4cre mice seven days after an adoptive transfer of Th17, identified by their cytotracker expression. n = 5. h, Therapeutic response of littermate controls Rorcfl/flCd4+/+ mice bearing TC-1 tumors to CDDP alone or in combination with Digoxin as in Fig. 2d, as reflected by tumor growth kinetics. n = 6. Flow cytometry expression of checkpoint inhibitors on i, CD4+ cells and j, CD8+ cells from TC-1 tumors on WT mice seven days after a CDDP treatment. n = 5. k, Therapeutic response of littermate controls Rorcfl/flCd4+/+ mice bearing TC-1 tumors to CDDP in combination with anti-CTLA-4 and Digoxin as in Fig.3b, reflected by tumor growth. n = 5. l, Therapeutic response of WT mice bearing a TC-1 tumor treated with anti-CTLA-4 as in Fig. 3a, reflected by tumor growth. n = 7. m, Therapeutic response of littermate controls Rorcfl/flCd4+/+ mice bearing MC38 tumors to ICB in combination with Digoxin as in Fig. 3g, reflected by tumor growth. n = 5. All tumor growth results are shown as mean ± SEM and experiments followed the experimental setting depicted above each data or referred to the main figures; other data are shown as mean ± SD and are representative of three independent experiments. Statistical significance was determined using two-way anova and Tukey’s multiple comparison test (b, c, f, hm), two tailed unpaired T-test (d, e), and one-way anova (a).

Source data

Extended Data Fig. 7 Impact of CDDP on CXCL10 production by intratumor immune cells.

Violin plot showing expression level of a, Lta and b, Ltb genes in cells belonging to the ILC3 CCR6 cluster at each time point, measured by sc RNA-seq from TC-1 tumors, as described in Fig. 1. c–m, Flow cytometry analysis of CXCL10 expressing cells from TC-1 tumors treated with PBS or CDDP, one, three and seven days before. Percentages of CXCL10+ in c, ILC1s, d, ILC2s, e, NKs, f, CD4+ cells, g, CD8+ cells, h, DCs, i, B cells, j, macrophages, k, M-MDSCs, l, PMN-MDSCs, m, eosinophils, n = 6. n, numbers of eosinophils from tumors treated as in c-m n = 6. Numbers are shown for 300 000 total viable cells from each TC-1 tumor. o, representative flow cytometry plot of CXCL10 expression. Results are shown as mean ± SD and are representative of three independent experiments. Statistical significance was determined using two-way anova and Tukey’s multiple comparison test (cn).

Source data

Extended Data Fig. 8 Impact of CXCR3 on CDDP driven intratumor immune recruitment.

a–g Flow cytometry analysis of immune populations in TC-1 tumors treated with anti-CXCR3 and CDDP and analyzed seven days after CDDP. Numbers are shown for 300 000 total viable cells from each TC-1 tumor. n = 5. Numbers of a, CCR6+ ILC3s, b, CD45+ cells, c, CD8+ and CD4+ cells, d, B cells, e, CD11c+ cells, f, CD11b+ cells and g, MDSCs. h, representative flow cytometry plot of KI67, IL-2, Granzyme B, IFNγ and TNFα expression. Results are shown as mean ± SD and are representative of three independent experiments. Statistical significance was determined using one-way anova (a, b, df) and two-ways anova plus Tukey’s multiple comparison test (c, g).

Source data

Extended Data Fig. 9 CCL20 is an important chemokine in CDDP triggered intratumor immune response.

KI67 expression on CCR6+ ILC3s from TC-1 tumors one or seven days after CDDP by a, flow cytometry. n = 5. and by b, sc RNA-seq. c, Fold increase of cytokine expressions relative to PBS in TC-1 tumors treated with CDDP using 14 cytokine bioplex. n = 5. d, Flow cytometry analysis of the percentage of cells from TC-1 tumors expressing CCL20 two days after a CDDP treatment. n = 5. e CCL20 production in vitro by TC-1 cells treated with various chemotherapies at their IC50 for two days. n = 3. f–j Flow cytometry analysis seven days after CDDP of immune cells infiltrating TC-1 tumors treated with α-CCL20 antibody. Numbers are shown for 300 000 total viable cells from each TC-1 tumor. n = 5. Numbers of f, CD45+ cells. g, B cells. h, CD11c+ cells. i, CD11b+ cells and j, MDSCs. k, Flow cytometry analysis of CCL20 expression by TC-1 cells transfected with a control Sh RNA (Sh C-) or directed against CCL20 (Sh CCL20) and treated in vitro with PBS or CDDP. n = 6. l, Therapeutic response of TC-1 Sh C- and TC-1 Sh CCL20 to CDDP as reflected by tumor growth kinetics. n = 17. m, representative flow cytometry plot of CCL20 expression in TC-1, MC38 and DCs, n, Flow cytometry analysis of the percentage of cells from MC38 tumors expressing CCL20 two days after an ICB treatment. EC: endothelial cells, MSC: mesenchymal stromal cells MC: myeloid cells DC: dendritic cells TC: tumor cells n = 5. All tumor growth results are shown as mean ± SEM and experiments followed the experimental setting depicted above each data or referred to the main figures and are representative of three independent experiments; other data are shown as mean ± SD and are representative of three independent experiments. Statistical significance was determined using one-way anova (ei), two-way anova and Tukey’s multiple comparison test (ad, jn).

Source data

Extended Data Fig. 10 IL-1β is a key factor for antitumor immune responses.

a, Flow cytometry analysis of Caspase-1 cleavage in myeloid cells MSC-2 treated in vitro with LPS and CDDP or LPS and Nigericin as a positive control for 24 h. n = 6. b, IL-1β production by myeloid cells MSC-2 treated in vitro with LPS and CDDP or LPS and Nigericin as a positive control for 24 h. n = 4. c-g, In vivo Flow cytometry analysis of Pro-IL-1β expression in immune cells from TC-1 tumors seven day after a CDDP or a PBS treatment Results show the numbers of cells from 300 000 total viable cells from each tumor. n = 5. Numbers of c, eosinophils, d, macrophages, e, M-MDSCs, f, PMN-MDSCs, g, DCs. h, Representative flow cytometry plot of CD69 expression in ILC3s. i, Therapeutic response of WT and TCRδ-/- mice bearing TC-1 tumors treated with CDDP or PBS reflected by tumor growth following the experimental setting depicted above. Flow cytometry analysis of the numbers of j, CD4+ and CD8+ cells of MC38 tumors from WT mice treated with ICB and IL-1RA as in Fig. 6l. n = 5. Numbers are shown for 300 000 total viable cells from each MC38 tumor. Therapeutic response of k, littermate controls Rorcfl/flCd4+/+ mice, n = 5, and l, Rorc–/– mice, n = 5, bearing MC38 tumors to ICB in combination with IL-1RA reflected by tumor growth following the experimental setting depicted above each data. m, Therapeutic response of WT mice bearing a MC38 tumor treated with rIL-1β and rCCL20 alone or in combination with ICB, reflected by tumor growth following the experimental setting depicted above, n = 5. All tumor growth results are shown as mean ± SEM and experiments followed the experimental setting depicted above each data or referred to the main figures and are representative of three independent experiments.; other data are shown as mean ± SD and are representative of three independent experiments. Statistical significance was determined using one-way anova (a, b), two-way anova and Tukey’s multiple comparison test (cg, im).

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 Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

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.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 10

Statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bruchard, M., Geindreau, M., Perrichet, A. et al. Recruitment and activation of type 3 innate lymphoid cells promote antitumor immune responses. Nat Immunol 23, 262–274 (2022). https://doi.org/10.1038/s41590-021-01120-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-01120-y

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