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:

Tumor immunoevasion by the conversion of effector NK cells into type 1 innate lymphoid cells

An Author Correction to this article was published on 21 March 2024

This article has been updated

Abstract

Avoiding destruction by immune cells is a hallmark of cancer, yet how tumors ultimately evade control by natural killer (NK) cells remains incompletely defined. Using global transcriptomic and flow-cytometry analyses and genetically engineered mouse models, we identified the cytokine-TGF-β-signaling-dependent conversion of NK cells (CD49aCD49b+Eomes+) into intermediate type 1 innate lymphoid cell (intILC1) (CD49a+CD49b+Eomes+) populations and ILC1 (CD49a+CD49bEomesint) populations in the tumor microenvironment. Strikingly, intILC1s and ILC1s were unable to control local tumor growth and metastasis, whereas NK cells favored tumor immunosurveillance. Experiments with an antibody that neutralizes the cytokine TNF suggested that escape from the innate immune system was partially mediated by TNF-producing ILC1s. Our findings provide new insight into the plasticity of group 1 ILCs in the tumor microenvironment and suggest that the TGF-β-driven conversion of NK cells into ILC1s is a previously unknown mechanism by which tumors escape surveillance by the innate immune system.

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

Figure 1: Three distinct populations of group 1 ILCs infiltrate MCA1956 tumors.
Figure 2: The conversion of NK cells into ILC1-like cells is dependent on TGF-β signaling.
Figure 3: intILC1s have the greatest proliferative capacity among group 1 ILC subsets in vivo.
Figure 4: TGF-β induces the conversion of NK cells into intILC1s in the tumor microenvironment.
Figure 5: TGF-β signaling limits tumor immunosurveillance by converting tumor NK cells into intILC1s and ILC1s.
Figure 6: TGF-β signaling abolishes control of metastasis by the innate immune system.
Figure 7: Functional differences among tumor group 1 ILC subsets.
Figure 8: IFN-γ and TNF control tumor growth and metastasis differentially.

Similar content being viewed by others

Change history

References

  1. Guillerey, C., Huntington, N.D. & Smyth, M.J. Targeting natural killer cells in cancer immunotherapy. Nat. Immunol. 17, 1025–1036 (2016).

    Google Scholar 

  2. Yang, L., Pang, Y. & Moses, H.L. TGF-beta and immune cells: an important regulatory axis in the tumor microenvironment and progression. Trends Immunol. 31, 220–227 (2010).

    Google Scholar 

  3. Wrzesinski, S.H., Wan, Y.Y. & Flavell, R.A. Transforming growth factor-β and the immune response: implications for anticancer therapy. Clin. Cancer Res. 13, 5262–5270 (2007).

    Google Scholar 

  4. Smyth, M.J., Strobl, S.L., Young, H.A., Ortaldo, J.R. & Ochoa, A.C. Regulation of lymphokine-activated killer activity and pore-forming protein gene expression in human peripheral blood CD8+ T lymphocytes. Inhibition by transforming growth factor-beta. J. Immunol. 146, 3289–3297 (1991).

    Google Scholar 

  5. Donatelli, S.S. et al. TGF-β-inducible microRNA-183 silences tumor-associated natural killer cells. Proc. Natl. Acad. Sci. USA 111, 4203–4208 (2014).

    Google Scholar 

  6. Wilson, E.B. et al. Human tumour immune evasion via TGF-β blocks NK cell activation but not survival allowing therapeutic restoration of anti-tumour activity. PLoS One 6, e22842 (2011).

    Google Scholar 

  7. Smyth, M.J. et al. CD4+CD25+ T regulatory cells suppress NK cell-mediated immunotherapy of cancer. J. Immunol. 176, 1582–1587 (2006).

    Google Scholar 

  8. Viel, S. et al. TGF-β inhibits the activation and functions of NK cells by repressing the mTOR pathway. Sci. Signal. 9, ra19 (2016).

    Google Scholar 

  9. Sojka, D.K. et al. Tissue-resident natural killer (NK) cells are cell lineages distinct from thymic and conventional splenic NK cells. eLife 3, e01659 (2014).

    Google Scholar 

  10. Constantinides, M.G., McDonald, B.D., Verhoef, P.A. & Bendelac, A. A committed precursor to innate lymphoid cells. Nature 508, 397–401 (2014).

    Google Scholar 

  11. Fuchs, A. ILC1s in tissue inflammation and infection. Front. Immunol. 7, 104 (2016).

    Google Scholar 

  12. Robinette, M.L. et al. Transcriptional programs define molecular characteristics of innate lymphoid cell classes and subsets. Nat. Immunol. 16, 306–317 (2015).

    Google Scholar 

  13. Seillet, C. et al. Differential requirement for Nfil3 during NK cell development. J. Immunol. 192, 2667–2676 (2014).

    Google Scholar 

  14. Gasteiger, G., Fan, X., Dikiy, S., Lee, S.Y. & Rudensky, A.Y. Tissue residency of innate lymphoid cells in lymphoid and nonlymphoid organs. Science 350, 981–985 (2015).

    Google Scholar 

  15. Klose, C.S. et al. Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages. Cell 157, 340–356 (2014).

    Google Scholar 

  16. Spits, H., Bernink, J.H. & Lanier, L. NK cells and type 1 innate lymphoid cells: partners in host defense. Nat. Immunol. 17, 758–764 (2016).

    Google Scholar 

  17. Seillet, C. & Belz, G.T. Differentiation and diversity of subsets in group 1 innate lymphoid cells. Int. Immunol. 28, 3–11 (2016).

    Google Scholar 

  18. Vallentin, B. et al. Innate Lymphoid Cells in Cancer. Cancer Immunol. Res. 3, 1109–1114 (2015).

    Google Scholar 

  19. Morvan, M.G. & Lanier, L.L. NK cells and cancer: you can teach innate cells new tricks. Nat. Rev. Cancer 16, 7–19 (2016).

    Google Scholar 

  20. Cortez, V.S. et al. Transforming growth factor-β signaling guides the differentiation of innate lymphoid cells in salivary glands. Immunity 44, 1127–1139 (2016).

    Google Scholar 

  21. Keskin, D.B. et al. TGFbeta promotes conversion of CD16+ peripheral blood NK cells into CD16 NK cells with similarities to decidual NK cells. Proc. Natl. Acad. Sci. USA 104, 3378–3383 (2007).

    Google Scholar 

  22. Narni-Mancinelli, E. et al. Fate mapping analysis of lymphoid cells expressing the NKp46 cell surface receptor. Proc. Natl. Acad. Sci. USA 108, 18324–18329 (2011).

    Google Scholar 

  23. Ruijtenberg, S. & van den Heuvel, S. Coordinating cell proliferation and differentiation: antagonism between cell cycle regulators and cell type-specific gene expression. Cell Cycle 15, 196–212 (2016).

    Google Scholar 

  24. Liberzon, A. et al. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 1, 417–425 (2015).

    Google Scholar 

  25. Smyth, M.J. et al. Differential tumor surveillance by natural killer (NK) and NKT cells. J. Exp. Med. 191, 661–668 (2000).

    Google Scholar 

  26. Smyth, M.J., Crowe, N.Y. & Godfrey, D.I. NK cells and NKT cells collaborate in host protection from methylcholanthrene-induced fibrosarcoma. Int. Immunol. 13, 459–463 (2001).

    Google Scholar 

  27. Sathe, P. et al. Innate immunodeficiency following genetic ablation of Mcl1 in natural killer cells. Nat. Commun. 5, 4539 (2014).

    Google Scholar 

  28. Knight, D.A. et al. Host immunity contributes to the anti-melanoma activity of BRAF inhibitors. J. Clin. Invest. 123, 1371–1381 (2013).

    Google Scholar 

  29. Krasnova, Y., Putz, E.M., Smyth, M.J. & Souza-Fonseca-Guimaraes, F. Bench to bedside: NK cells and control of metastasis. Clin. Immunol. 177, 50–59 (2017).

    Google Scholar 

  30. Chaput, N. et al. Phase I clinical trial combining imatinib mesylate and IL-2: HLA-DR+ NK cell levels correlate with disease outcome. OncoImmunology 2, e23080 (2013).

    Google Scholar 

  31. Ménard, C. et al. Natural killer cell IFN-gamma levels predict long-term survival with imatinib mesylate therapy in gastrointestinal stromal tumor-bearing patients. Cancer Res. 69, 3563–3569 (2009).

    Google Scholar 

  32. Fuchs, A. et al. Intraepithelial type 1 innate lymphoid cells are a unique subset of IL-12- and IL-15-responsive IFN-γ-producing cells. Immunity 38, 769–781 (2013).

    Google Scholar 

  33. Crome, S.Q. et al. A distinct innate lymphoid cell population regulates tumor-associated T cells. Nat. Med. 23, 368–375 (2017).

    Google Scholar 

  34. Dadi, S. et al. Cancer immunosurveillance by tissue-resident innate lymphoid cells and innate-like T cells. Cell 164, 365–377 (2016).

    Google Scholar 

  35. Pikovskaya, O. et al. Cutting edge: eomesodermin is sufficient to direct type 1 innate lymphocyte development into the conventional NK lineage. J. Immunol. 196, 1449–1454 (2016).

    Google Scholar 

  36. Cortez, V.S. et al. SMAD4 impedes the conversion of NK cells into ILC1-like cells by curtailing non-canonical TGF-β signaling. Nat. Immunol. doi:10.1038/ni.3809 (2016).

  37. Hayakawa, Y. et al. IFN-gamma-mediated inhibition of tumor angiogenesis by natural killer T-cell ligand, α-galactosylceramide. Blood 100, 1728–1733 (2002).

    Google Scholar 

  38. Ikeda, H., Old, L.J. & Schreiber, R.D. The roles of IFNγ in protection against tumor development and cancer immunoediting. Cytokine Growth Factor Rev. 13, 95–109 (2002).

    Google Scholar 

  39. Balkwill, F. Tumour necrosis factor and cancer. Nat. Rev. Cancer 9, 361–371 (2009).

    Google Scholar 

  40. Baluk, P. et al. TNF-α drives remodeling of blood vessels and lymphatics in sustained airway inflammation in mice. J. Clin. Invest. 119, 2954–2964 (2009).

    Google Scholar 

  41. Sainson, R.C. et al. TNF primes endothelial cells for angiogenic sprouting by inducing a tip cell phenotype. Blood 111, 4997–5007 (2008).

    Google Scholar 

  42. Gill, S. et al. Rapid development of exhaustion and down-regulation of eomesodermin limit the antitumor activity of adoptively transferred murine natural killer cells. Blood 119, 5758–5768 (2012).

    Google Scholar 

  43. Doisne, J.M. et al. Composition, development, and function of uterine innate lymphoid cells. J. Immunol. 195, 3937–3945 (2015).

    Google Scholar 

  44. Levi, I. et al. Characterization of tumor infiltrating natural killer cell subset. Oncotarget 6, 13835–13843 (2015).

    Google Scholar 

  45. Bruno, A. et al. The proangiogenic phenotype of natural killer cells in patients with non-small cell lung cancer. Neoplasia 15, 133–142 (2013).

    Google Scholar 

  46. Lima, P.D., Zhang, J., Dunk, C., Lye, S.J. & Croy, B.A. Leukocyte driven-decidual angiogenesis in early pregnancy. Cell. Mol. Immunol. 11, 522–537 (2014).

    Google Scholar 

  47. Vacca, P. et al. Crosstalk between decidual NK and CD14+ myelomonocytic cells results in induction of Tregs and immunosuppression. Proc. Natl. Acad. Sci. USA 107, 11918–11923 (2010).

    Google Scholar 

  48. Arteaga, C.L. et al. Anti-transforming growth factor (TGF)-β antibodies inhibit breast cancer cell tumorigenicity and increase mouse spleen natural killer cell activity. Implications for a possible role of tumor cell/host TGF-β interactions in human breast cancer progression. J. Clin. Invest. 92, 2569–2576 (1993).

    Google Scholar 

  49. Terabe, M. et al. Synergistic enhancement of CD8+ T cell-mediated tumor vaccine efficacy by an anti-transforming growth factor-β monoclonal antibody. Clin. Cancer Res. 15, 6560–6569 (2009).

    Google Scholar 

  50. Morris, J.C. et al. Phase I study of GC1008 (fresolimumab): a human anti-transforming growth factor-β (TGFβ) monoclonal antibody in patients with advanced malignant melanoma or renal cell carcinoma. PLoS One 9, e90353 (2014).

    Google Scholar 

  51. Akhurst, R.J. & Hata, A. Targeting the TGFβ signalling pathway in disease. Nat. Rev. Drug Discov. 11, 790–811 (2012).

    Google Scholar 

  52. Viant, C. et al. Transforming growth factor-β and Notch ligands act as opposing environmental cues in regulating the plasticity of type 3 innate lymphoid cells. Sci. Signal. 9, ra46 (2016).

    Google Scholar 

  53. Kara, E.E. et al. CCR2 defines in vivo development and homing of IL-23-driven GM-CSF-producing Th17 cells. Nat. Commun. 6, 8644 (2015).

    Google Scholar 

  54. Johnstone, C.N. et al. Functional and molecular characterisation of EO771.LMB tumours, a new C57BL/6-mouse-derived model of spontaneously metastatic mammary cancer. Dis. Model. Mech. 8, 237–251 (2015).

    Google Scholar 

  55. Blake, S.J. et al. Suppression of metastases using a new lymphocyte checkpoint target for cancer immunotherapy. Cancer Discov. 6, 446–459 (2016).

    Google Scholar 

  56. Souza-Fonseca-Guimaraes, F. et al. NK cells require IL-28R for optimal in vivo activity. Proc. Natl. Acad. Sci. USA 112, E2376–E2384 (2015).

    Google Scholar 

  57. Delconte, R.B. et al. CIS is a potent checkpoint in NK cell-mediated tumor immunity. Nat. Immunol. 17, 816–824 (2016).

    Google Scholar 

  58. Rautela, J. et al. Loss of Host Type-I IFN signaling accelerates metastasis and impairs NK-cell antitumor function in multiple models of breast cancer. Cancer Immunol. Res. 3, 1207–1217 (2015).

    Google Scholar 

  59. Ngiow, S.F. et al. A threshold level of intratumor CD8+ T-cell PD1 expression dictates therapeutic response to anti-PD1. Cancer Res. 75, 3800–3811 (2015).

    Google Scholar 

  60. Liao, Y., Smyth, G.K. & Shi, W. The Subread aligner: fast, accurate and scalable read mapping by seed-and-vote. Nucleic Acids Res. 41, e108 (2013).

    Google Scholar 

  61. Law, C.W., Chen, Y., Shi, W. & Smyth, G.K. voom: Precision weights unlock linear model analysis tools for RNA-seq read counts. Genome Biol. 15, R29 (2014).

    Google Scholar 

  62. Ritchie, M.E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Google Scholar 

  63. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550 (2005).

    Google Scholar 

  64. Lee, E., Chuang, H.Y., Kim, J.W., Ideker, T. & Lee, D. Inferring pathway activity toward precise disease classification. PLOS Comput. Biol. 4, e1000217 (2008).

    Google Scholar 

  65. Bald, T. et al. Ultraviolet-radiation-induced inflammation promotes angiotropism and metastasis in melanoma. Nature 507, 109–113 (2014).

    Google Scholar 

Download references

Acknowledgements

We thank R. Schreiber (Washington University School of Medicine) for MCA1956 fibrosarcoma cells and anti-IFN-γ and anti-TNF hybridomas; the animal house and flow cytometry facilities at QIMR Berghofer Medical Research Institute and Walter and Eliza Hall Institute of Medical Research; E. Loza, K. Elder, L. Town, L. Spencer, T. Camilleri and T. Kratina, for mouse breeding, maintenance and genotyping; and K. MacDonald, D. Smith, A. Kallies, L. Beattie, R. Allan, G. Hill and S. Nutt for discussion, comments and advice on this project. Supported by the National Health and Medical Research Council of Australia (Senior Principal Research Fellowship 1078671 to M.J.S.; Peter Doherty Early Career Fellowship 1088703 to F.S.-F.-G. and 1124690 to T.B.; project grant 1027472 to G.T.B.; Elizabeth Blackburn NHMRC Fellowship to G.T.B.; Independent Research Institute Infrastructure Support scheme grant to G.T.B.; project grants 1066770 & 1057852 N.D.H.; and RD Wright Career development Fellowship 1112113 to N.W.), the Cancer Research Institute Clinical and Laboratory Integration Programs (M.J.S. and N.D.H.), Queensland Institute of Medical Research Berghofer International PhD Scholarship (Y.G. and J.Y.), University of Queensland International Scholarship (Y.G. and J.Y.), the National Breast Cancer Foundation (PF-15-008 to F.S.-F.-G.), Cure Cancer Australia (Priority-Driven Young Investigator Project Grant 1082709 and 1120725 to F.S.-F.-G.), European Molecular Biology Organization (long-term fellowship ALTF 945-2015 to T.B.), the Naito Foundation (K.N.), Cancer Council Queensland (PhD fellowship to A.Y.), Griffith University (PhD scholarships to S.S.N.), Inserm-Avenir-Grant (L.B.), Ligue Nationale Contre le Cancer (L.B.), Fondation ARC Pour la Recherche sur le Cancer (L.B.), the Victorian State Government Operational Infrastructure Scheme (G.T.B.), the Harry J Lloyd Charitable Trust (Melanoma Research Grant to N.D.H.) and the DFG Excellence Cluster Immunosensation (EXC 1023 to M.H.).

Author information

Authors and Affiliations

Authors

Contributions

Y.G., F.S.-F.-G., T.B., N.D.H., K.N. and M.J.S. designed research, supervised work and wrote the paper; Y.G., F.S.-F.-G., T.B., A.Y., S.F.N., J.R., S.J.B., J.Y., J.S.L., M.M., L.Z., N.D.H., K.N. and M.J.S. performed research; Y.G., F.S.-F.-G., T.B., S.S.N., A.Y., S.F.N., J.R., J.S., N.W., S.J.B., J.Y., M.M., L.Z., M.W.L.T., G.T.B., C.R.E., N.D.H., K.N., M.H. and M.J.S. analyzed data; and L.B., E.V., K.T. and G.T.B. provided experimental materials.

Competing Interests StatementM.J.S. has research agreements with Bristol-Myers Squibb, Corvus Pharmaceuticals and Aduro Biotech; E.V. is a cofounder and shareholder in Innate Pharma; and N.D.H. and J.R. are cofounders and shareholders in oNKo-Innate.

Corresponding author

Correspondence to Mark J Smyth.

Ethics declarations

Competing interests

M.J.S. has research agreements with Bristol-Myers Squibb, Corvus Pharmaceuticals and Aduro Biotech; E.V. is a cofounder and shareholder in Innate Pharma; and N.D.H. and J.R. are cofounders and shareholders in oNKo-Innate.

Integrated supplementary information

Supplementary Figure 1 Similarities among group 1 ILC subsets from tumor, liver and spleen.

(a) Representative flow cytometric plots illustrating the gating strategy for sorting of group 1 ILC subsets from MCA1956 tumors for transcriptomic analyses. (b) Representative flow cytometric plots illustrating post-sorting purity of tumor NK cells, intILC1s and ILC1s. (c) Heatmap visualizing gene expression profiles from NK cells and ILC1s isolated from liver/spleen (GSE52047) clustered by tumor NK cell and ILC1 gene signatures. (d) Quantification of gene signature expression by combined z-scores. Horizontal bars, median; boxes, 25th to 75th quartile; ‘whiskers’, 10th and 90th quartile. Statistical analysis by pairwise two-sided t-tests with Benjamini & Hochberg (FDR) correction for multiple testing. **P < 0.01, ****P < 0.0001; ns, non-significant. Sp: spleen; Liv: liver.

Supplementary Figure 2 TGF-b promotes NK cell conversion both in vitro and in vivo.

(a) Flow cytometric characterization of group 1 ILC composition based on CD49a and CD49b expression in livers and spleens of indicated transgenic mice. (b) Liver group 1 ILC composition as determined by flow cytometry based on CD49a and Eomes expression in indicated transgenic mice. (c) Corresponding quantification of liver group 1 ILC subsets amongst indicated transgenic mice. Data shown as mean ± s.e.m.; Mann-Whitney U-test; **P < 0.01. (a-c) Data represent n = 5 of one experiment. (d,e) Percentage and phenotype of NK-derived ILC1s (CD49a+Eomes) in NK cell cultured for 5 days with 25 ng/mL rIL-15/IL-15Rα complex and TGF-β1 at indicated concentration. Statistical comparisons of in vitro NK cell-derived ILC1 percentage (d) and TRAILhiDNAM-1hi cell percentage in in vitro NK cell-derived ILC1s (e) at different rTGF-β1 concentrations. Data shown as mean ± s.e.m. of 5 replicates per group of one experiment. *P < 0.05, ***P < 0.001, ****P < 0.0001 determined by one-way ANOVA and Tukey’s multiple comparison test. (f) Purity of intILC1s and NK cells sorted from pooled spleens of indicated transgenic mice. (g) CD49a and CD49b expression profile of cells as sorted in (f) after cultured in serum-free medium in presence of 50 ng/mL rIL-15 for 4 days (n = 3 of one experiment). (h) Expression of CD49a and CD49b on splenic NK cells cultured in serum-free medium supplemented as indicated over time (n = 3 of one experiment). (i) Corresponding expression of CD49a and CD49b on liver ILC1s (n = 3 of one experiment).

Supplementary Figure 3 Analysis of proliferation-associated gene sets from liver and splenic NK cells and ILC1s.

Heatmap visualizing E2F gene set expression (a) and G2M checkpoint gene set expression (b) of liver/spleen NK cells and ILC1s (GSE52047) clustered by tumor NK cell and ILC1 gene signatures (left of each panel). Quantifications of gene set expression (right of each panel) by combined z-scores were compared by pairwise two-sided t-tests with Benjamini & Hochberg (FDR) correction for multiple testing. Horizontal lines in whisker boxplots represent quartiles. *P < 0.05; ns, non-significant. Sp: spleen; Liv: liver. Horizontal bars, median; boxes, 25th to 75th quartile; ‘whiskers’, 10th and 90th quartile.

Supplementary Figure 4 Mcl1FL mice are deficient in group 1 ILCs, while anti-asGM1 ‘preferentially’ depletes the tumor microenvironment of NK cells and intILC1s.

(a) Representative flow cytometric plots showing NK1.1+NKp46+ cells (left) and CD49a+CD49b ILC1s and CD49aCD49b+ NK cells (right) in the liver (top panel) and spleen (bottom panel) of Mcl1WT and Mcl1FL mice. (b) Corresponding quantifications of cell population in the liver (top panel) and spleen (bottom panel) as indicated. Rag2−/−γc−/− mice were used as negative control (mean ± s.e.m.; n = 5 for liver, n = 4 for spleen and n = 2 for Rag2−/−γc−/− mice from two independent experiments; unpaired two-sided t-tests; *P < 0.05, **P < 0.01). (c) Experimental setup for the treatment of MCA1956 tumor-bearing WT mice with two doses of 50 μg anti-asGM1 antibody or control IgG i.p. for two consecutive days. (d,e) Representative flow cytometric plots showing percentage of group 1 ILCs (in live CD45+Lin population) (d) and group 1 ILC composition (e) in different tissues from mice treated as indicated (n = 5 for ctrl IgG treated group and n = 6 for anti-asGM1 antibody treated group of two independent experiments). (f-h) Corresponding quantifications of group 1 ILC subset number in the liver (f), spleen (g) and MCA1956 tumor (h) of mice treated as indicated (mean ± s.e.m.; n = 5 for ctrl IgG treated group and n = 6 for anti-asGM1 antibody treated group of two independent experiments; Mann-Whitney U-test; *P < 0.05, **P < 0.01).

Supplementary Figure 5 TGF-b signaling fosters tumor growth and NK cell conversion in SM1WT1 tumors.

(a) Representative flow cytometric plots showing group 1 ILC composition in SM1WT1 melanomas harvested from indicated transgenic mice at day 24 after tumor injection. ND, not determined. (b) Corresponding quantification of tumor group 1 ILC subsets (mean ± s.e.m.; n = 8 for Ncr1cre/wt mice, n = 7 for RIIFL mice and RICA-FL mice of two independent experiments; one-way ANOVA and Tukey’s multiple comparison test; ***P < 0.001, ****P < 0.0001). (c) Tumor growth of SM1WT1 melanomas in indicated transgenic mice (mean ± s.e.m.; n = 10 for Ncr1cre/wt, RICA-FL and RICA-WT mice, n = 5 for Mcl1WT and Mcl1FL mice of two independent experiments; one-way ANOVA and Tukey’s multiple comparison test; **P < 0.01). (d) Tumor growth of SM1WT1 melanomas in RIIFL and RIIWT mice treated with 50 μg control IgG or anti-asGM1 antibody i.p. on day -1, 0, 7, 14 and 21 before or after tumor cell injection at day 0 (mean ± s.e.m.; n = 10 for RIIFL and RIIWT mice of two independent experiments; one-way ANOVA and Tukey’s multiple comparison test; ***P < 0.001, ****P < 0.0001).

Supplementary Figure 6 TGF-β signaling in NKp46+ cells controls lung metastasis.

Lung metastasis in WT or indicated transgenic mice following i.v. injection of 2 × 105 RM-1 prostate carcinoma cells (a) and 3.5 × 105 EO771-LMB mCherry+ breast cancer cells (b-d). Total fluorescence radiant efficiency (b), relative mCherry mRNA expression (c) and representative fluorescence imaging (d) of lung metastases were shown. Results shown as mean ± s.e.m. and represent 3 mice per group (RICA-WT), 6 mice per group (RIIWT, RIIFL and RICA-FL), and 15 mice per group (WT and Ncr1cre/wt) for (a) and 6 mice per group (Ncr1cre/wt and RICA-FL), 5 mice per group (RIIFL and Mcl1FL) for (b-d) of one experiment. *P < 0.05, **P < 0.01, ****P < 0.0001 determined by one-way ANOVA and Tukey’s multiple comparison test.

Supplementary Figure 7 Expression of inhibitory immune cell receptors on tumor group 1 ILC subsets.

(a) Comparisons of inhibitory immune cell receptor expression among tumor group 1 ILC subsets isolated from s.c. transplanted MCA1956 tumors (mean ± s.e.m.; n = 5 of two experiments; one-way ANOVA and Tukey’s multiple comparison test; *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001; N.D., not detected). (b) Representative flow cytometric plots showing the expression of indicated receptors in tumor group 1 ILC subsets isolated from primary MCA-induced fibrosarcomas (n = 5 of one experiment). (c) Representative histogram showing NKG2A expression in tumor group 1 ILC subsets isolated from s.c. MCA1956 tumors (n = 5 of two independent experiments).

Supplementary Figure 8 Antibody-mediated neutralization of TNF impairs the growth of SM1WT1 melanomas.

(a) IFN-γ (left) and TNF (middle) production by tumor group 1 ILC subsets from SM1WT1 melanomas after 4 h stimulation by PMA/ionomycin. Percentages of cytokine producing cells were determined by flow cytometry and ratios of IFN-γ/TNF-producing cells (right) were calculated (mean ± s.e.m.; n = 7 tumors from one experiment; one-way ANOVA and Tukey’s multiple comparison; *P < 0.05, **P < 0.01). (b) Experimental setup for antibody-mediated cytokine neutralization in SM1WT1 melanoma-bearing mice. (c) Tumor growth in RIIWT (left) and RIIFL (right) mice treated as indicated (mean ± s.e.m.; n = 5 mice per group of two independent experiments; one-way ANOVA and Tukey’s multiple comparison test; ***P < 0.001, ****P < 0.0001). (d) Representative flow cytometric plots showing the gating strategy for the analyses of CXCR6 expression on CD3CD56dim and CD3CD56bright human NK cells from PBMC of healthy volunteers (HV) and GIST patients as well as tumor infiltrating lymphocytes (TILs) from GIST patients. (e) Representative flow cytometric plots showing the gating strategy for the analyses of CXCR6 expression on NK1.1+NKp46+ tumor group 1 ILCs in MCA1956 tumors (n = 5 per group of two independent experiments).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 and Supplementary Tables 1 and 2 (PDF 1237 kb)

Life Sciences Reporting Summary (PDF 71 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gao, Y., Souza-Fonseca-Guimaraes, F., Bald, T. et al. Tumor immunoevasion by the conversion of effector NK cells into type 1 innate lymphoid cells. Nat Immunol 18, 1004–1015 (2017). https://doi.org/10.1038/ni.3800

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3800

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