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:

Adoptive cellular therapy with T cells expressing the dendritic cell growth factor Flt3L drives epitope spreading and antitumor immunity

Abstract

Adoptive cell therapies using genetically engineered T cell receptor or chimeric antigen receptor T cells are emerging forms of immunotherapy that redirect T cells to specifically target cancer. However, tumor antigen heterogeneity remains a key challenge limiting their efficacy against solid cancers. Here, we engineered T cells to secrete the dendritic cell (DC) growth factor Fms-like tyrosine kinase 3 ligand (Flt3L). Flt3L-secreting T cells expanded intratumoral conventional type 1 DCs and substantially increased host DC and T cell activation when combined with immune agonists poly (I:C) and anti-4-1BB. Importantly, combination therapy led to enhanced inhibition of tumor growth and the induction of epitope spreading towards antigens beyond those recognized by adoptively transferred T cells in solid tumor models of T cell receptor and chimeric antigen receptor T cell therapy. Our data suggest that augmenting endogenous DCs is a promising strategy to overcome the clinical problem of antigen-negative tumor escape following adoptive cell 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: Flt3L drives DC proliferation and T cell–dependent rejection of solid tumors.
Fig. 2: Flt3L-generated CD103+ DCs induce superior T cell expansion in vitro.
Fig. 3: Adoptive transfer of Flt3L-secreting T cells promotes intratumoral CD103+ cDC1 proliferation and infiltration of endogenous CD8+ T cells.
Fig. 4: Flt3L-secreting T cells elicit enhanced inhibition of tumor growth.
Fig. 5: Flt3L-secreting T cells and immune adjuvants induce oligoclonal expansion of host T cells.
Fig. 6: Flt3L-secreting CAR T cells and immune adjuvants enhance antitumor immunity in a host cDC1- and T cell–dependent manner.
Fig. 7: Combination therapy with Flt3L-secreting CAR T cells induces epitope spreading.

Similar content being viewed by others

Data availability

The data generated or analyzed to support the findings of this study are available in this article and from the corresponding author upon request without restrictions. Source data for Figs. 1–7 and Extended Data Figs. 1–9 are presented with the paper.

References

  1. Mardiana, S., Solomon, B. J., Darcy, P. K. & Beavis, P. A. Supercharging adoptive T cell therapy to overcome solid tumor-induced immunosuppression. Sci. Transl. Med. 11, eaaw2293 (2019).

    PubMed  CAS  Google Scholar 

  2. Mardiana, S., Lai, J., House, I. G., Beavis, P. A. & Darcy, P. K. Switching on the green light for chimeric antigen receptor T-cell therapy. Clin. Transl. Immunology 8, e1046 (2019).

    PubMed  PubMed Central  Google Scholar 

  3. June, C. H., O’Connor, R. S., Kawalekar, O. U., Ghassemi, S. & Milone, M. C. CAR T cell immunotherapy for human cancer. Science 359, 1361–1365 (2018).

    CAS  PubMed  Google Scholar 

  4. Majzner, R. G. & Mackall, C. L. Tumor antigen escape from CAR T-cell therapy. Cancer Discov. 8, 1219–1226 (2018).

    PubMed  CAS  Google Scholar 

  5. Jackson, H. J. & Brentjens, R. J. Overcoming antigen escape with CAR T-cell therapy. Cancer Discov. 5, 1238–1240 (2015).

    PubMed  PubMed Central  CAS  Google Scholar 

  6. Salmon, H. et al. Expansion and activation of CD103+ dendritic cell progenitors at the tumor site enhances tumor responses to therapeutic PD-L1 and BRAF inhibition. Immunity 44, 924–938 (2016).

    PubMed  PubMed Central  CAS  Google Scholar 

  7. Roberts, E. W. et al. Critical role for CD103+/CD141+ dendritic cells bearing CCR7 for tumor antigen trafficking and priming of T cell immunity in melanoma. Cancer Cell 30, 324–336 (2016).

    PubMed  PubMed Central  CAS  Google Scholar 

  8. Broz, M. L. et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell 26, 638–652 (2014).

    PubMed  PubMed Central  CAS  Google Scholar 

  9. Xu, Y. et al. Closely related T-memory stem cells correlate with in vivo expansion of CAR.CD19-T cells and are preserved by IL-7 and IL-15. Blood 123, 3750–3759 (2014).

    PubMed  PubMed Central  CAS  Google Scholar 

  10. Alizadeh, D. et al. IL15 enhances CAR-T cell antitumor activity by reducing mTORC1 activity and preserving their stem cell memory phenotype. Cancer Immunol. Res. 7, 759–772 (2019).

    PubMed  PubMed Central  CAS  Google Scholar 

  11. Gattinoni, L., Klebanoff, C. A. & Restifo, N. P. Paths to stemness: building the ultimate antitumour T cell. Nat. Rev. Cancer 12, 671–684 (2012).

    PubMed  PubMed Central  CAS  Google Scholar 

  12. Brentjens, R. J. et al. Safety and persistence of adoptively transferred autologous CD19-targeted T cells in patients with relapsed or chemotherapy refractory B-cell leukemias. Blood 118, 4817–4828 (2011).

    PubMed  PubMed Central  CAS  Google Scholar 

  13. Wang, L. X. et al. Tumor ablation by gene-modified T cells in the absence of autoimmunity. Cancer Res. 70, 9591–9598 (2010).

    PubMed  CAS  Google Scholar 

  14. Schlitzer, A. et al. Identification of cDC1- and cDC2-committed DC progenitors reveals early lineage priming at the common DC progenitor stage in the bone marrow. Nat. Immunol. 16, 718–728 (2015).

    CAS  PubMed  Google Scholar 

  15. Grajales-Reyes, G. E. et al. Batf3 maintains autoactivation of Irf8 for commitment of a CD8ɑ+ conventional DC clonogenic progenitor. Nat. Immunol. 16, 708–717 (2015).

    PubMed  PubMed Central  CAS  Google Scholar 

  16. Sichien, D. et al. IRF8 transcription factor controls survival and function of terminally differentiated conventional and plasmacytoid dendritic cells, respectively. Immunity 45, 626–640 (2016).

    PubMed  CAS  Google Scholar 

  17. Guilliams, M. et al. Unsupervised high-dimensional analysis aligns dendritic cells across tissues and species. Immunity 45, 669–684 (2016).

    PubMed  PubMed Central  CAS  Google Scholar 

  18. Durai, V. et al. Cryptic activation of an Irf8 enhancer governs cDC1 fate specification. Nat. Immunol. 20, 1161–1173 (2019).

    PubMed  PubMed Central  CAS  Google Scholar 

  19. Binnewies, M. et al. Unleashing type-2 dendritic cells to drive protective antitumor CD4+ T cell immunity. Cell 177, 556–571.e16 (2019).

    PubMed  PubMed Central  CAS  Google Scholar 

  20. Bottcher, J. P. et al. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell 172, 1022–1037.e14 (2018).

    PubMed  PubMed Central  CAS  Google Scholar 

  21. Ruffell, B. et al. Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell 26, 623–637 (2014).

    PubMed  PubMed Central  CAS  Google Scholar 

  22. Marigo, I. et al. T cell cancer therapy requires CD40-CD40L activation of tumor necrosis factor and inducible nitric-oxide-synthase-producing dendritic cells. Cancer Cell 30, 651 (2016).

    PubMed  CAS  Google Scholar 

  23. Sade-Feldman, M. et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013.e20 (2018).

    PubMed  PubMed Central  CAS  Google Scholar 

  24. 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.e10 (2019).

    PubMed  CAS  Google Scholar 

  25. Fuertes, M. B. et al. Host type I IFN signals are required for antitumor CD8+ T cell responses through CD8ɑ+ dendritic cells. J. Exp. Med. 208, 2005–2016 (2011).

    PubMed  PubMed Central  CAS  Google Scholar 

  26. Hammerich, L. et al. Systemic clinical tumor regressions and potentiation of PD1 blockade with in situ vaccination. Nat. Med. 25, 814–824 (2019).

    PubMed  CAS  Google Scholar 

  27. Segal, N. H. et al. Phase I study of single-agent utomilumab (PF-05082566), a 4-1BB/CD137 agonist, in patients with advanced cancer. Clin. Cancer Res. 24, 1816–1823 (2018).

    PubMed  CAS  Google Scholar 

  28. Mardiana, S. et al. A multifunctional role for adjuvant anti-4-1BB therapy in augmenting antitumor response by chimeric antigen receptor T cells. Cancer Res. 77, 1296–1309 (2017).

    PubMed  CAS  Google Scholar 

  29. Sanchez-Paulete, A. R. et al. Cancer immunotherapy with immunomodulatory anti-CD137 and anti-PD-1 monoclonal antibodies requires BATF3-dependent dendritic cells. Cancer Discov. 6, 71–79 (2016).

    PubMed  CAS  Google Scholar 

  30. Canale, F. P. et al. CD39 expression defines cell exhaustion in tumor-infiltrating CD8+ T cells—response. Cancer Res. 78, 5175 (2018).

    PubMed  CAS  Google Scholar 

  31. John, L. B. et al. Anti-PD-1 antibody therapy potently enhances the eradication of established tumors by gene-modified T cells. Clin. Cancer Res. 19, 5636–5646 (2013).

    PubMed  CAS  Google Scholar 

  32. Beavis, P. A. et al. Targeting the adenosine 2A receptor enhances chimeric antigen receptor T cell efficacy. J. Clin. Invest. 127, 929–941 (2017).

    PubMed  PubMed Central  Google Scholar 

  33. Michie, J. et al. Antagonism of IAPs enhances CAR T-cell efficacy. Cancer Immunol. Res. 7, 183–192 (2019).

    PubMed  CAS  Google Scholar 

  34. Rafiq, S. et al. Targeted delivery of a PD-1-blocking scFv by CAR-T cells enhances anti-tumor efficacy in vivo. Nat. Biotechnol. 36, 847–856 (2018).

    PubMed  PubMed Central  CAS  Google Scholar 

  35. Koneru, M., Purdon, T. J., Spriggs, D., Koneru, S. & Brentjens, R. J. IL-12 secreting tumor-targeted chimeric antigen receptor T cells eradicate ovarian tumors. Oncoimmunology 4, e994446 (2015).

    PubMed  PubMed Central  Google Scholar 

  36. Newick, K. et al. Augmentation of CAR T-cell trafficking and antitumor efficacy by blocking protein kinase A localization. Cancer Immunol. Res. 4, 541–551 (2016).

    PubMed  PubMed Central  CAS  Google Scholar 

  37. Chang, Z. L. et al. Rewiring T-cell responses to soluble factors with chimeric antigen receptors. Nat. Chem. Biol. 14, 317–324 (2018).

    PubMed  PubMed Central  CAS  Google Scholar 

  38. Moon, E. K. et al. Expression of a functional CCR2 receptor enhances tumor localization and tumor eradication by retargeted human T cells expressing a mesothelin-specific chimeric antibody receptor. Clin. Cancer Res. 17, 4719–4730 (2011).

    PubMed  PubMed Central  CAS  Google Scholar 

  39. Chmielewski, M., Kopecky, C., Hombach, A. A. & Abken, H. IL-12 release by engineered T cells expressing chimeric antigen receptors can effectively muster an antigen-independent macrophage response on tumor cells that have shut down tumor antigen expression. Cancer Res. 71, 5697–5706 (2011).

    PubMed  CAS  Google Scholar 

  40. Avanzi, M. P. et al. Engineered tumor-targeted T cells mediate enhanced anti-tumor efficacy both directly and through activation of the endogenous immune system. Cell Rep. 23, 2130–2141 (2018).

    PubMed  PubMed Central  CAS  Google Scholar 

  41. Chmielewski, M. & Abken, H. CAR T cells releasing IL-18 convert to T-Bethigh FoxO1low effectors that exhibit augmented activity against advanced solid tumors. Cell Rep. 21, 3205–3219 (2017).

    PubMed  CAS  Google Scholar 

  42. Kuhn, N. F. et al. CD40 ligand-modified chimeric antigen receptor T cells enhance antitumor function by eliciting an endogenous antitumor response. Cancer Cell 35, 473–488.e6 (2019).

    PubMed  PubMed Central  CAS  Google Scholar 

  43. Adachi, K. et al. IL-7 and CCL19 expression in CAR-T cells improves immune cell infiltration and CAR-T cell survival in the tumor. Nat. Biotechnol. 36, 346–351 (2018).

    PubMed  CAS  Google Scholar 

  44. de Mingo Pulido, A. et al. TIM-3 regulates CD103+ dendritic cell function and response to chemotherapy in breast cancer. Cancer Cell 33, 60–74.e6 (2018).

    PubMed  PubMed Central  Google Scholar 

  45. Beavis, P. A. et al. Dual PD-1 and CTLA-4 checkpoint blockade promotes antitumor immune responses through CD4+Foxp3 cell-mediated modulation of CD103+ dendritic cells. Cancer Immunol. Res. 6, 1069–1081 (2018).

    PubMed  CAS  Google Scholar 

  46. 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.e4 (2017).

    PubMed  PubMed Central  CAS  Google Scholar 

  47. Durai, V. et al. Altered compensatory cytokine signaling underlies the discrepancy between Flt3 −/− and Flt3l −/− mice. J. Exp. Med. 215, 1417–1435 (2018).

    PubMed  PubMed Central  CAS  Google Scholar 

  48. Brown, C. E. et al. Regression of glioblastoma after chimeric antigen receptor T-cell therapy. N. Engl. J. Med. 375, 2561–2569 (2016).

    PubMed  PubMed Central  CAS  Google Scholar 

  49. Adusumilli, P. S. et al. Regional delivery of mesothelin-targeted CAR T cell therapy generates potent and long-lasting CD4-dependent tumor immunity. Sci. Transl. Med. 6, 261ra151 (2014).

    PubMed  PubMed Central  Google Scholar 

  50. Tchou, J. et al. Safety and efficacy of intratumoral injections of chimeric antigen receptor (CAR) T cells in metastatic breast cancer. Cancer Immunol. Res. 5, 1152–1161 (2017).

    PubMed  PubMed Central  CAS  Google Scholar 

  51. Piechocki, M. P., Ho, Y. S., Pilon, S. & Wei, W. Z. Human ErbB-2 (Her-2) transgenic mice: a model system for testing Her-2 based vaccines. J. Immunol. 171, 5787–5794 (2003).

    PubMed  CAS  Google Scholar 

  52. Darcy, P. K. et al. Redirected perforin-dependent lysis of colon carcinoma by ex vivo genetically engineered CTL. J. Immunol. 164, 3705–3712 (2000).

    PubMed  CAS  Google Scholar 

  53. Haynes, N. M. et al. Redirecting mouse CTL against colon carcinoma: superior signaling efficacy of single-chain variable domain chimeras containing TCR-ζ vs Fc ε RI-γ. J. Immunol. 166, 182–187 (2001).

    PubMed  CAS  Google Scholar 

  54. Haynes, N. et al. Single-chain antigen recognition receptors that costimulate potent rejection of established experimental tumors. Blood 100, 3155–3163 (2002).

    PubMed  CAS  Google Scholar 

  55. Duong, C. P. et al. Engineering T cell function using chimeric antigen receptors identified using a DNA library approach. PLoS ONE 8, e63037 (2013).

    PubMed  PubMed Central  CAS  Google Scholar 

  56. Naik, S. H. et al. Development of plasmacytoid and conventional dendritic cell subtypes from single precursor cells derived in vitro and in vivo. Nat. Immunol. 8, 1217–1226 (2007).

    PubMed  CAS  Google Scholar 

  57. Mayer, C. T. et al. Selective and efficient generation of functional Batf3-dependent CD103+ dendritic cells from mouse bone marrow. Blood 124, 3081–3091 (2014).

    PubMed  PubMed Central  CAS  Google Scholar 

  58. Helft, J. et al. GM-CSF mouse bone marrow cultures comprise a heterogeneous population of CD11c+MHCII+ macrophages and dendritic cells. Immunity 42, 1197–1211 (2015).

    PubMed  CAS  Google Scholar 

  59. Chen, E. Y. et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14, 128 (2013).

    PubMed  PubMed Central  Google Scholar 

  60. Kuleshov, M. V. et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).

    PubMed  PubMed Central  CAS  Google Scholar 

  61. Betel, I. The occurrence of cytochromes in nuclei isolated from rat thymus. Biochim. Biophys. Acta 143, 62–69 (1967).

    PubMed  CAS  Google Scholar 

  62. Zotos, D. et al. IL-21 regulates germinal center B cell differentiation and proliferation through a B cell–intrinsic mechanism. J. Exp. Med. 207, 365–378 (2010).

    PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We acknowledge assistance from the staff of the Animal Facility, the Centre for Advanced Histology and Microscopy, the Flow Cytometry Facility, the Molecular Genomics Core and the Pathology departments at the Peter MacCallum Cancer Centre. We also acknowledge the contribution of consumer advocates K. Gill, M. Rear and G. Sissing for this study. This work was funded by the National Health and Medical Research Council (NHMRC; grant nos. 1062580, 1143976 and 1150425), the Cancer Council Victoria (grant no. 1156382), the National Breast Cancer Foundation (grant no. IIRS-19-016 19-22), the CLEARbridge Foundation and the Tour de Cure Foundation. P.A.B. is supported by a National Breast Cancer Foundation Fellowship (ID no. ECF-17-005). P.K.D., A.M.L. and K.K. are supported by NHMRC Research Fellowships (grant nos. APP1041828, APP1080321 and APP1102792, respectively).

Author information

Authors and Affiliations

Authors

Contributions

J.L., S.M., P.A.B. and P.K.D. designed the experiments, developed the methodology, analyzed and interpreted data and wrote the manuscript. J.L., S.M., P.A.B., P.K.D., I.G.H., K.S., M.A.H., L.G., A.X.Y.C., K.L.T., E.V.P. and J.D.C. performed experiments and acquired data. E.M.C., A.M.L., B.J.S., J.A.T., K.K., M.E., S.J.V. and J.W. provided technical support and advice on data analysis and interpretation. P.A.B. and P.K.D. supervised the study and were responsible for coordination and strategy.

Corresponding authors

Correspondence to Phillip K. Darcy or Paul A. Beavis.

Ethics declarations

Competing interests

P.A.B. and P.K.D. are inventors on a patent filed for the following study (filing no. PCT/AU2019/050660).

Additional information

Peer review information 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.

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

Extended data

Extended Data Fig. 1 Tumor-derived Flt3L mediates tumor regression. Related to Fig. 1.

a, Flt3L levels from 24JK-Her2 and MC38 cell lines transduced to express Cherry control (Cherry) or Flt3L Cherry (Cherry FL). Data represent technical replicates from one of two independent experiments with similar results. b, Tumor and serum Flt3L levels from hHer2 mice 6 days post inoculation with 24JK-Her2 Cherry or Cherry FL cells (3 mice/group), from one of two independent experiments. c, Tumor measurement (2-way ANOVA) and survival (tumor area > 100mm2) of C57BL/6 mice subcutaneously injected with 5 × 104 MC38 expressing Flt3L cherry (Cherry FL, n = 7) or cherry control (Cherry, n = 8) (two-tailed Mantel-Cox, two pooled independent experiments). d, Tumor myeloid cell gating strategy. Lineage markers include Thy1.2, CD19, B220/CD45R, NK1.1 and Ly6G. e, Frequency of tumor infiltrating CD4+ T cells, Treg, CD11b+ cDC2, macrophages and neutrophils 9 days post tumor inoculation with 24JK-Her2 Cherry FL or control. Bars represent mean ± SEM, b, e Unpaired Student’s t-test, symbols represent individual mice.

Source data

Extended Data Fig. 2 Characterization of in vitro differentiated BM DCs. Related to Fig. 2.

a, In vitro differentiated BM DC gating strategy. b, Contour plots of BM cells differentiated with (FL-DCs) or without (GM-DCs) exogenous Flt3L (20 ng/mL). c, IFN-γ and TNF levels in supernatants of 4 × 104 naive OT-I T cells co-cultured for 5 days with OVA257-264 peptide (0.1 μg/mL) pulsed GM- or FL-DCs at a 2:1 ratio. Where indicated, DCs were activated with poly (I:C) (25 μg/mL) for 24 hours prior to co-culture. Bars represent mean ± SEM of technical triplicates from one of three independent experiments with similar results. d, Schematic illustration of OT-I T cell transduction and preconditioning with cytokines IL-7 and IL-15 (IL-7/15).

Source data

Extended Data Fig. 3 Preconditioning regimens for lymphodepletion and cDC1 expansion following OT-I FL therapy.

a, Analysis of splenic immune cell subsets 2 days after total body irradiation (TBI), representative of one of two independent experiments with similar results. b, Fold change of immune cell subsets normalized to control non-irradiated mice. c, Comparison of splenic immune cell subsets after preconditioning with titrated doses of TBI or cyclophosphamide (CP). d, Fold change of immune cell subsets after 0.5 Gy TBI or 50 mg/kg CP normalized to control mice. e, hHer2 mice were irradiated at indicated doses and subcutaneously inoculated with 5 × 105 Flt3L expressing 24JK-Her2 cells (n = 6 mice/group), from of one of two independent experiments (2-way ANOVA). f–g, Absolute numbers of BM pre-cDC, pre-cDC1 and pre-cDC2 per femur, and splenic cDC1 and cDC2 populations 7 days post adoptive transfer, after preconditioning with 0.5 Gy TBI or 50 mg/kg CP. Bars represent mean ± SEM, a, f, g 1-way ANOVA, symbols represent individual mice.

Source data

Extended Data Fig. 4 Flt3L-secreting T cells promote DC proliferation and tumor infiltration of endogenous CD8+ T cells. Related to Fig. 3.

a, BM pre-cDC gating strategy. Lineage markers include Thy1.2, CD19, and NK1.1. b, Frequency of pre-cDC1 and pre-cDC2 as a percentage of total BM pre-cDC (NT n = 4 mice, OT-I and FL n = 6 mice/group). c, Absolute numbers of macrophages and neutrophils per mg of tumor, and d, Frequency of tumor cDC1, cDC2 and cDC1 precursors as a percentage of total tumor DCs post adoptive transfer (NT n = 4, OT-1 n = 6, FL n = 5). e, Adoptive transfer of E0771-OVA tumor-bearing mice with 2 × 107 antigen-specific OT-I T cells or antigen non-specific pmel-1 T cells transduced to secrete Flt3L (OT-I FL and pmel-1 FL, respectively). (Left) Expression of surface NGFR marker in OT-I FL and pmel-1 FL cells. (Mid) Flt3L from tumor supernatants (day 7). (Right) Absolute numbers of tumor cDC1 and cDC1 precursors on day 7 post adoptive transfer with OT-I FL or pmel-1 FL. f, Representative contour plots of tumor cDC1 and cDC2 respectively secreting IL-12 and TNF. g, Absolute numbers of migratory and resident dLN DCs (day 5 NT n = 4 mice, OT-I n = 6, FL n = 5; day 7 NT and FL n = 8/group, OT-I n = 11) and h, splenic cDC1, cDC2, macrophages and neutrophils (NT n = 3 – 4/group, OT-I n = 6/group, FL = 5 – 6/group). Bars represent mean ± SEM, b–e, g, h 1-way ANOVA, a–g Data representative of one of two independent experiments with similar results, d,e Symbols represent individual mice.

Source data

Extended Data Fig. 5 Treg and CD62L+TCF1+ CD8+ T cells in tumors.

a, Absolute numbers of Treg per mg tumor on day 7 post adoptive transfer (1-way ANOVA), from one of two independent experiments. b, Frequency of CD62L+TCF-1+ OT-I or endogenous CD8+ T cells in the tumor on day 7 post adoptive transfer from two pooled independent experiments (unpaired Student’s t-test). Symbols represent individual mice.

Source data

Extended Data Fig. 6 Characterization of the effects of adjuvants in vitro and in vivo.

a, Activated BM DCs were pulsed with OVA257-264 before co-culture with naive OT-I T cells (1:2). Frequencies of CD69+ and 4-1BB+ CD8+ OT-I T cells day 5 post co-culture, data represent technical triplicates from one of three independent experiments with similar results. b, Survival (tumor area > 100mm2) from one of two independent experiments (two-tailed Mantel-Cox) of mice subcutaneously inoculated with 5 × 105 MC38 Cherry FL cells or control. Ten days later, mice were injected with poly (I:C:) (200 μg) and/or anti-4-1BB (100 μg). Subsequent poly (I:C) were administered on days 4 and 8 post initial treatment. c, E0771-OVA tumor bearing C57BL/6 WT mice were treated accordingly with OT-I FL, poly (I:C), anti-4-1BB (top; NT and FL + anti-4-1BB n = 5 mice/group, rest n = 4/group; 2-way ANOVA) or adjuvants with no T cells (bottom; n = 5 mice/group). Arrows (top) indicate adjuvants (days 5 and 8). Data points from NT, OT-I FL and OT-I FL + poly (I:C) + anti-4-1BB were included in Fig. 4c pooled data. d, Significantly upregulated genes based on GO term ‘Response to type I Interferon’ (GO: 0034340) (day 6). e, Representative CD40+CD86+ activated DCs (day 7). f–g, Absolute numbers of PMA and ionomycin stimulated, IFN-γ-producing CD8+ T cells on day 7 post adoptive transfer from 2 – 4 pooled independent experiments, symbols represent individual mice, 1-way ANOVA. h, hHer2 mice were subcutaneously injected with 1 × 106 MC38-Her2, preconditioned and treated with 1 × 107 CAR T or CAR T FL cells on days 5 and 6 post tumor inoculation (NT and FL n = 5 mice/group, CAR T n = 6, CAR T + adjuvants n = 12, FL + adjuvants n = 13, 2-way ANOVA, Sidak’s multiple comparison). Arrow indicate adjuvants on day 5 post adoptive transfer. a, c, f–h Bars represent mean ± SEM.

Source data

Extended Data Fig. 7 In vitro and in vivo characterization of CD4+ CAR T cells.

a, Frequency of mCherry+ CD4+ and CD8+ CAR T cells from bulk transduced CAR T cell population prior to adoptive transfer, from two pooled independent experiments with n = 2 technical replicates per experiment. b, Frequency of CD4+ and CD8+ mCherry+ CAR T cells in the dLN of E0771-OVA-Her2 tumor-bearing mice on 7 days post adoptive transfer (CAR n = 8 mice, FL n = 9, CAR T + adjuvants n = 12, FL + adjuvants n = 12). Bars represent mean ± SEM.

Source data

Extended Data Fig. 8 Safety assessment of Flt3L-secreting T cells and adjuvant therapy.

hHer2 transgenic mice were inoculated with 4 × 105 E0771-OVA-Her2 tumor cells and treated with transduced CAR T cells as per Fig. 6f. Mice were monitored for changes in weight. Alternatively, sera and organs were assessed at 48 hours post adjuvant therapy for toxicity studies. a, Liver function and/or damage was assessed by serum alanine transaminase (ALT), aspartate aminotransferase (AST), total albumin, bilirubin, and gamma glutamyl transferase (GGT) levels. b, Kidney function and/or damage was assessed by serum urea and creatinine levels. c, Sera were assessed for levels of IFN-γ, IL-1β, IL-6, GM-CSF, G-CSF, MCP-1 and TNF cytokines. d, Percentage weight change relative to day 0 measurements. Dotted line (90%) indicates the maximum ethical limit allowed for weight reduction. Arrows indicate adjuvant administration (NT n = 5 mice, CAR T and FL + Isotype n = 6 mice/group, CAR T and FL + adjuvants n = 8/group). e, Representative hematoxylin and eosin histology staining of cerebellum, liver, kidney, lung and spleen of control non-treated (NT), CAR T or CAR T FL adjuvant treated mice (3 mice/group). f, Sera were assessed for antinuclear antibodies as an indication of autoimmunity (n = 5 mice/group). Sera from NZB/W (with proteinuria; n = 3) and C57BL/6 mice (n = 3) immunized with an irrelevant antigen (NP-KLH) were respectively used as positive and negative controls. Bars represent mean ± SEM, a–c Symbols represent individual mice, 1-way ANOVA.

Source data

Extended Data Fig. 9 Assessment of BM hematopoietic stem cells and progenitors.

a, Gating strategy for BM populations. b, Absolute numbers of hematopoietic stem cells (HSC), common lymphoid progenitors (CLP), common dendritic progenitors (CDP) and myeloid and dendritic progenitors (MDP) in the BM from E0771-OVA tumor-bearing mice 7 or 14 days post adoptive transfer with Flt3L-secreting OT-I T cells. Data shown as mean ± SEM of n = 5 mice/group, 1-way ANOVA.

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1 and 2.

Reporting Summary

Supplementary Table

Information on antibodies, reagents, software and service platforms.

Source data

Source Data Fig. 1

Statistical Source Data for Fig. 1.

Source Data Fig. 2

Statistical Source Data for Fig. 2.

Source Data Fig. 3

Statistical Source Data for Fig. 3.

Source Data Fig. 4

Statistical Source Data for Fig. 4.

Source Data Fig. 5

Statistical Source Data for Fig. 5.

Source Data Fig. 6

Statistical Source Data for Fig. 6.

Source Data Fig. 7

Statistical Source Data for Fig. 7.

Source Data Extended Data Fig. 1

Statistical Source Data for Extended Data Fig. 1.

Source Data Extended Data Fig. 2

Statistical Source Data for Extended Data Fig. 2.

Source Data Extended Data Fig. 3

Statistical Source Data for Extended Data Fig. 3.

Source Data Extended Data Fig. 4

Statistical Source Data for Extended Data Fig. 4.

Source Data Extended Data Fig. 5

Statistical Source Data for Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Statistical Source Data for Extended Data Fig. 6.

Source Data Extended Data Fig. 7

Statistical Source Data for Extended Data Fig. 7.

Source Data Extended Data Fig. 8

Statistical Source Data for Extended Data Fig. 8.

Source Data Extended Data Fig. 9

Statistical Source Data for Extended Data Fig. 9.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lai, J., Mardiana, S., House, I.G. et al. Adoptive cellular therapy with T cells expressing the dendritic cell growth factor Flt3L drives epitope spreading and antitumor immunity. Nat Immunol 21, 914–926 (2020). https://doi.org/10.1038/s41590-020-0676-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-0676-7

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing