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:

Systemic clinical tumor regressions and potentiation of PD1 blockade with in situ vaccination

Abstract

Indolent non-Hodgkin’s lymphomas (iNHLs) are incurable with standard therapy and are poorly responsive to checkpoint blockade. Although lymphoma cells are efficiently killed by primed T cells, in vivo priming of anti-lymphoma T cells has been elusive. Here, we demonstrate that lymphoma cells can directly prime T cells, but in vivo immunity still requires cross-presentation. To address this, we developed an in situ vaccine (ISV), combining Flt3L, radiotherapy, and a TLR3 agonist, which recruited, antigen-loaded and activated intratumoral, cross-presenting dendritic cells (DCs). ISV induced anti-tumor CD8+ T cell responses and systemic (abscopal) cancer remission in patients with advanced stage iNHL in an ongoing trial (NCT01976585). Non-responding patients developed a population of PD1+CD8+ T cells after ISV, and murine tumors became newly responsive to PD1 blockade, prompting a follow-up trial of the combined therapy. Our data substantiate that recruiting and activating intratumoral, cross-priming DCs is achievable and critical to anti-tumor T cell responses and PD1-blockade efficacy.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Cross-presentation is required to control lymphoma growth in vivo.
Fig. 2: In situ vaccination with FLt3L, XRT and pICLC induces systemic tumor immunity dependent on cross-presenting dendritic cells.
Fig. 3: In situ vaccination mediates tumor infiltration by CD8+ T cells and augments checkpoint blockade therapy.
Fig. 4: ISV induces TLR3+ DCs and PD1+ T cells in patients with indolent B cell lymphoma.
Fig. 5: In situ vaccination induces systemic tumor regression in patients with indolent B cell lymphoma.

Similar content being viewed by others

Data availability

The datasets supporting the findings presented in this study are available from the corresponding author upon reasonable request. All requests for data and materials will be promptly reviewed by the Icahn School of Medicine at Mount Sinai to verify whether the request is subject to any intellectual property or confidentiality obligations. Patient-related data were generated as part of a clinical trial and may be subject to patient confidentiality. Any data that can be shared will be released via a Material Transfer Agreement.

References

  1. Locke, F. L. et al. Phase 1 results of ZUMA-1: a multicenter study of KTE-C19 anti-CD19 CAR T cell therapy in refractory aggressive lymphoma. Mol. Ther. 25, 285–295 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Bollard, C. M. et al. Sustained complete responses in patients with lymphoma receiving autologous cytotoxic T lymphocytes targeting Epstein-Barr virus latent membrane proteins. J. Clin. Oncol. 32, 798–808 (2014).

    Article  CAS  PubMed  Google Scholar 

  3. Bargou, R. et al. Tumor regression in cancer patients by very low doses of a T cell-engaging antibody. Science 321, 974–977 (2008).

    Article  CAS  PubMed  Google Scholar 

  4. Levy, R. et al. Active idiotypic vaccination versus control immunotherapy for follicular lymphoma. J. Clin. Oncol. 32, 1797–1803 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Schuster, S. J. et al. Vaccination with patient-specific tumor-derived antigen in first remission improves disease-free survival in follicular lymphoma. J. Clin. Oncol. 29, 2787–2794 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Sharma, P., Hu-Lieskovan, S., Wargo, J. A. & Ribas, A. Primary, adaptive, and acquired resistance to cancer immunotherapy. Cell 168, 707–723 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Ding, W. et al. Pembrolizumab in patients with CLL and Richter transformation or with relapsed CLL. Blood 129, 3419–3427 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Lesokhin, A. M. et al. Nivolumab in patients with relapsed or refractory hematologic malignancy: preliminary results of a phase Ib study. J. Clin. Oncol. 34, 2698–2704 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Hildner, K. et al. Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity. Science 322, 1097–1100 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Sánchez-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).

    Article  PubMed  Google Scholar 

  13. Spranger, S. et al. Density of immunogenic antigens does not explain the presence or absence of the T-cell-inflamed tumor microenvironment in melanoma. Proc. Natl Acad. Sci. USA 113, E7759–E7768 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Liao, J. et al. Converting lymphoma cells into potent antigen-presenting cells for interferon-induced tumor regression. Cancer Immunol. Res. 5, 560–570 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. de Charette, M., Marabelle, A. & Houot, R. Turning tumour cells into antigen presenting cells: The next step to improve cancer immunotherapy? Eur. J. Cancer 68, 134–147 (2016).

    Article  PubMed  Google Scholar 

  16. Agudo, J. et al. Quiescent tissue stem cells evade immune surveillance. Immunity 48, 271–285 e275 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Gajewski, T. F., Schreiber, H. & Fu, Y. X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 14, 1014–1022 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Anandasabapathy, N. et al. Efficacy and safety of CDX-301, recombinant human Flt3L, at expanding dendritic cells and hematopoietic stem cells in healthy human volunteers. Bone Marrow Transplant 50, 924–930 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Maraskovsky, E. et al. Dramatic increase in the numbers of functionally mature dendritic cells in Flt3 ligand-treated mice: multiple dendritic cell subpopulations identified. J. Exp. Med. 184, 1953–1962 (1996).

    Article  CAS  PubMed  Google Scholar 

  20. Anandasabapathy, N. et al. Classical Flt3L-dependent dendritic cells control immunity to protein vaccine. J. Exp. Med. 211, 1875–1891 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Barry, K. C. et al. A natural killer-dendritic cell axis defines checkpoint therapy-responsive tumor microenvironments. Nat. Med. 24, 1178–1191 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Merad, M., Sugie, T., Engleman, E. G. & Fong, L. In vivo manipulation of dendritic cells to induce therapeutic immunity. Blood 99, 1676–1682 (2002).

    Article  CAS  PubMed  Google Scholar 

  23. Schiavoni, G. et al. ICSBP is essential for the development of mouse type I interferon-producing cells and for the generation and activation of CD8α+ dendritic cells. J. Exp. Med. 196, 1415–1425 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Suzuki, S. et al. Critical roles of interferon regulatory factor 4 in CD11bhighCD8alpha- dendritic cell development. Proc. Natl Acad. Sci. USA 101, 8981–8986 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Bachem, A. et al. Expression of XCR1 characterizes the Batf3-dependent lineage of dendritic cells capable of antigen cross-presentation. Front. Immunol. 3, 214 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Sancho, D. et al. Tumor therapy in mice via antigen targeting to a novel, DC-restricted C-type lectin. J. Clin. Invest 118, 2098–2110 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Apetoh, L. et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 13, 1050–1059 (2007).

    Article  CAS  PubMed  Google Scholar 

  28. Obeid, M. et al. Calreticulin exposure dictates the immunogenicity of cancer cell death. Nat. Med. 13, 54–61 (2007).

    Article  CAS  PubMed  Google Scholar 

  29. Sauter, B. et al. Consequences of cell death: exposure to necrotic tumor cells, but not primary tissue cells or apoptotic cells, induces the maturation of immunostimulatory dendritic cells. J. Exp. Med. 191, 423–434 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Tang, D., Kang, R., Zeh, H. J. & Lotze, M. T. High-mobility group box 1 and cancer. Biochim. Biophys. Acta 1799, 131–140 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Yanai, H. et al. HMGB proteins function as universal sentinels for nucleic-acid-mediated innate immune responses. Nature 462, 99–103 (2009).

    Article  CAS  PubMed  Google Scholar 

  32. Guilliams, M. et al. Dendritic cells, monocytes and macrophages: a unified nomenclature based on ontogeny. Nat. Rev. Immunol. 14, 571–578 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Cheson, B. D. et al. Recommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classification. J. Clin. Oncol. 32, 3059–3068 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  34. Chaperot, L. et al. Functional expression of CD80 and CD86 allows immunogenicity of malignant B cells from non-Hodgkin’s lymphomas. Exp. Hematol. 27, 479–488 (1999).

    Article  CAS  PubMed  Google Scholar 

  35. Aoi, T., Nakano, H., Tanaka, Y. & Kakiuchi, T. Enhancement of antigen-presenting ability of B lymphoma cells by partial inhibition of protein synthesis through inducing B7-1 expression. Immunology 91, 212–218 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Brody, J. D. et al. In situ vaccination with a TLR9 agonist induces systemic lymphoma regression: a phase I/II study. J. Clin. Oncol. 28, 4324–4332 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Kim, Y. H. et al. In situ vaccination against mycosis fungoides by intratumoral injection of a TLR9 agonist combined with radiation: a phase 1/2 study. Blood 119, 355–363 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Rook, A. H. et al. Topical resiquimod can induce disease regression and enhance T-cell effector functions in cutaneous T-cell lymphoma. Blood 126, 1452–1461 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. El Tawdy, A. M. et al. Toll-like receptor (TLR)7 expression in mycosis fungoides and psoriasis: a case-control study. Clin. Exp. Dermatol. 42, 172–177 (2017).

    Article  CAS  PubMed  Google Scholar 

  40. Jarrousse, V. et al. Toll-like receptors 2, 4 and 9 expression in cutaneous T-cell lymphoma (mycosis fungoides and Sézary syndrome). Eur. J. Dermatol. 16, 636–641 (2006).

    CAS  PubMed  Google Scholar 

  41. Freedman, R. S. et al. Pilot study of Flt3 ligand comparing intraperitoneal with subcutaneous routes on hematologic and immunologic responses in patients with peritoneal carcinomatosis and mesotheliomas. Clin. Cancer Res. 9, 5228–5237 (2003).

    CAS  PubMed  Google Scholar 

  42. Morse, M. A. et al. Preoperative mobilization of circulating dendritic cells by Flt3 ligand administration to patients with metastatic colon cancer. J. Clin. Oncol. 18, 3883–3893 (2000).

    Article  CAS  PubMed  Google Scholar 

  43. Gasparetto, C. et al. Mobilization of dendritic cells from patients with breast cancer into peripheral blood stem cell leukapheresis samples using Flt-3-Ligand and G-CSF or GM-CSF. Cytokine 18, 8–19 (2002).

    Article  CAS  PubMed  Google Scholar 

  44. Disis, M. L. et al. Flt3 ligand as a vaccine adjuvant in association with HER-2/neu peptide-based vaccines in patients with HER-2/neu-overexpressing cancers. Blood 99, 2845–2850 (2002).

    Article  CAS  PubMed  Google Scholar 

  45. Anders, K. et al. Oncogene-targeting T cells reject large tumors while oncogene inactivation selects escape variants in mouse models of cancer. Cancer Cell 20, 755–767 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wolkers, M. C., Stoetter, G., Vyth-Dreese, F. A. & Schumacher, T. N. Redundancy of direct priming and cross-priming in tumor-specific CD8+T cell responses. J. Immunol. 167, 3577–3584 (2001).

    Article  CAS  PubMed  Google Scholar 

  47. Ochsenbein, A. F. et al. Roles of tumour localization, second signals and cross priming in cytotoxic T-cell induction. Nature 411, 1058–1064 (2001).

    Article  CAS  PubMed  Google Scholar 

  48. Hargadon, K. M. et al. Incomplete differentiation of antigen-specific CD8 T cells in tumor-draining lymph nodes. J. Immunol. 177, 6081–6090 (2006).

    Article  CAS  PubMed  Google Scholar 

  49. Chevalier, N. et al. Analysis of dendritic cell subpopulations in follicular lymphoma with respect to the tumor immune microenvironment. Leuk. Lymphoma 57, 2150–2160 (2016).

    Article  CAS  PubMed  Google Scholar 

  50. Chang, K. C., Huang, G. C., Jones, D. & Lin, Y. H. Distribution patterns of dendritic cells and T cells in diffuse large B-cell lymphomas correlate with prognoses. Clin. Cancer Res. 13, 6666–6672 (2007).

    Article  CAS  PubMed  Google Scholar 

  51. 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 

  52. Mayordomo, J. I. et al. Bone marrow-derived dendritic cells pulsed with synthetic tumour peptides elicit protective and therapeutic antitumour immunity. Nat. Med. 1, 1297–1302 (1995).

    Article  CAS  PubMed  Google Scholar 

  53. Hawiger, D. et al. Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivo. J. Exp. Med. 194, 769–779 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Abuodeh, Y., Venkat, P. & Kim, S. Systematic review of case reports on the abscopal effect. Curr. Probl. Cancer 40, 25–37 (2016).

    Article  PubMed  Google Scholar 

  55. Rees, G. J. Abscopal regression in lymphoma: a mechanism in common with total body irradiation? Clin. Radiol. 32, 475–480 (1981).

    Article  CAS  PubMed  Google Scholar 

  56. Kurlander, R., Stein, R. S. & Roth, D. Hyperkalemia complicating splenic irradiation of chronic lymphocytic leukemia. Cancer 36, 926–930 (1975).

    Article  CAS  PubMed  Google Scholar 

  57. Sham, R. L. The abscopal effect and chronic lymphocytic leukemia. Am. J. Med. 98, 307–308 (1995).

    Article  CAS  PubMed  Google Scholar 

  58. Andtbacka, R. H. et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J. Clin. Oncol. 33, 2780–2788 (2015).

    Article  CAS  PubMed  Google Scholar 

  59. Postow, M. A. et al. Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N. Engl. J. Med. 372, 2006–2017 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Antonia, S. J. et al. Nivolumab alone and nivolumab plus ipilimumab in recurrent small-cell lung cancer (CheckMate 032): a multicentre, open-label, phase 1/2 trial. Lancet Oncol. 17, 883–895 (2016).

    Article  CAS  PubMed  Google Scholar 

  61. Sanjana, N. E., Shalem, O. & Zhang, F. Improved vectors and genome-wide libraries for CRISPR screening. Nat. Methods 11, 783–784 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Chan, W. F., Perez-Diez, A., Razavy, H. & Anderson, C. C. The ability of natural tolerance to be applied to allogeneic tissue: determinants and limits. Biol. Direct 2, 10 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  63. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank the ISMMS human immune monitoring core for help with mass cytometry experiments and Luminex assay; the ISMMS flow cytometry core; and the CCMS animal facility. We also thank N. Sadek and C. Kappauff for technical assistance and N. Bhardwaj for critical review of the manuscript. J.D.B. was supported by a Damon Runyon Cancer Research Foundation Clinical Investigator Award (no. 45367), by an Investigator Studies Program (MSIP) Award (no. 10677), and by a Cancer Research Institute - Clinic & Laboratory Integration Program (CLIP, no. 45367) Award. L.H. was supported by a research fellowship of the German research foundation (DFG, HA 7431/1-1). B.D.B. and M.M. were supported by NIH U19AI128949.

Author information

Authors and Affiliations

Authors

Contributions

L.H. designed the study, performed and analyzed experiments, interpreted the results and wrote the manuscript. T.U.M. was involved in human sample collection and analysis. R.U. performed and analyzed cell culture experiments. J.S.-A, M.D., S.H. and Y.Z. performed immunohistochemistry. D.O. was involved in human sample collection. M.Y. and H.M. provided CDX-301. A.M.S. provided pICLC. A.H.R. performed and analyzed mass cytometry experiments. B.D.B. and M.M. helped design experiments and wrote the manuscript. J.D.B. conceived the study, supervised the design and execution of the study, interpreted the results and wrote the manuscript.

Corresponding author

Correspondence to Joshua D. Brody.

Ethics declarations

Competing interests

Research support for these studies was provided by Merck & Co., Celldex Therapeutics, Oncovir, Inc. and Genentech. J.D.B. has received research funding from Acerta Pharma, Bristol Myers Squibb, Genentech, Gilead Sciences, Seattle Genetics, Pharmacyclics and Celgene. M.Y. and H.M. are employed by Celldex Therapeutics. A.M.S. is employed by Oncovir, Inc. The authors declare no other competing financial interests.

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 Cross-presentation is required to control lymphoma growth in vivo.

a, GFP- and mCherry-expressing tumor cell lines (ratio1:1) were cocultured with JEDI TCs pre-activated with anti-CD3/anti-CD8. Tumor cell killing was analyzed after 3 d. One representative out of three independent experiments is shown. b, A20-GFP lymphoma-bearing Balb/c- and C57Bl/6-Rag1–/– mice were injected with 5 × 103 GFP-specific JEDI CD8+ TCs, and tumors were analyzed for transferred TCs by flow cytometry 2 weeks after transfer. Representative dot plots from n = 9 (Balb/c) or n = 15 (B6) mice from two independent experiments. c, WT mice were inoculated with A20 tumors and tumor were stained for TCs (CD3) and DC (CD11c) after 3 weeks. Representative FACS plots (left), immunofluorescence (middle) and quantification of intratumoral TCs and DC numbers (right). n = 10 from two independent experiments. Data are presented as mean ± s.e.m .

Extended Data Fig. 2 Flt3L induces recruitment of CD103 precursors.

a,b, Tumor-bearing Balb/c mice were treated with Flt3L and analyzed for DC expansion in the tumor (a) and bone marrow (b). n = 8 mice pooled from two independent experiments. One-way ANOVA with Tukey’s post test. c, Representative dot plots of CD11b+ and CD103+ cells among all intratumoral dendritic cells (one out of four independent experiments) and of IRF8+ and IRF4+ cells among DN dendritic cells in the tumor (one out of two independent experiments). d,e, Intratumoral DC subsets were isolated from Flt3L-treated mice by flow sorting and cultured in the presence of bone marrow feeder cells with or without Flt3L (d) or cocultured with irradiated A20 cells (e). CD103 and XCR1 (d) or CD11b (e) expression was analyzed after 3 d of culture. One representative out of two independent experiments with n = 3 or 4 mice each is shown. f, Splenocytes from Flt3L-treated mice were irradiated with 9 Gy in vitro and analyzed for CD103 expression on DC subsets after 3 d. One representative out of four independent cell culture samples is shown. Data are presented as mean ± s.e.m.

Extended Data Fig. 3 local XRT of the tumor induces maturation and activation of TLR3+ dendritic cells.

a,b, Splenocytes isolated from Flt3L-treated mice were cultured in the presence of supernatant from irradiated A20 cells and either a HMGB1-blocking antibody or TLR agonists. Activation of dendritic cells was analyzed after 48 h. One-way ANOVA with Tukey’s post test. Representative experiment (n = 2 biologically independent cell culture samples) out of three independent shown. c, HMGB1 levels in the serum of irradiated mice (0 Gy: n = 5, 9 Gy: n = 6, 9 Gy no tumor: n = 3; left panel) or supernatants of irradiated A20 cells (24 h: n = 7, 48 h n = 5; right panel). Data pooled from two or three independent experiments, respectively. One-way ANOVA with Tukey’s post test. d,e, Flt3L-treated splenocytes were cultured with irradiated GFP+ A20 cells with or without pIC for 24 h and then cocultured with JEDI splenocytes for 4 d. Activation of GFP-specific JEDI CD8+ TCs was analyzed by proliferation. Data pooled from five independent experiments (n = 10 biologically independent cell culture samples). One-way ANOVA with Tukey’s post test. f, Tumor-bearing WT or Batf3–/– mice were treated with Flt3L and analyzed for intratumoral DC subsets 1 d after the last injection. Representative plots from n = 4 mice per group. Data are presented as mean ± s.e.m.

Extended Data Fig. 4 In situ vaccination induces upregulation of checkpoint molecules and augments checkpoint blockade therapy.

ac, A20-GFP tumor-bearing mice were treated with Flt3L, XRT and pICLC as indicated. 1 × 106 JEDI CD8+ TCs were adoptively transferred 1 d after XRT and intratumoral TCs were analyzed by flow cytometry 7 d after transfer. a, CD25 expression on GFP-specific vs. non-specific CD8+ TCs in the tumor and TdLN of ISV-treated mice. n = 3, unpaired two-tailed t test. b, TIM3 and LAG3 expression on bulk CD8+ TC in the tumor of untreated and ISV-treated mice. n = 3, unpaired two-tailed t test. c, PD1 expression on GFP-specific vs. non-specific CD8+ TCs in the TdLN of untreated and ISV-treated mice. n = 3, unpaired two-tailed t test. d, A20-lymphoma-bearing mice were treated with ISV, and i.t. DCs were analyzed for PD-L1 expression after three injections of pICLC. n = 6 (untreated) or n = 7 (F/X/IC) from two independent experiments, unpaired two-tailed t test. e, A20-lymphoma-bearing mice were treated with ISV and systemic anti-PD1 or isotype control antibody and followed for tumor growth and survival after indicated treatment. n = 18, except Flt3L+XRT+pIC n = 19 and XRT + pIC + anti-PD1 n = 21; two-way ANOVA with Bonferroni post test. f, IFNγ production and CD137 expression by splenic CD8+ TCs isolated from tumor-bearing mice treated as indicated 1 week after the last pICLC injection and cocultured with irradiated tumor cells for 48 h. n = 4 mice per group, one representative experiment out of two independent experiments shown, one-way ANOVA with Tukey’s post test. Data are presented as mean ± s.e.m.

Extended Data Fig. 5 In situ vaccination induces DC expansion and intratumoral TC infiltration in patients with indolent B cell lymphoma.

a,b,11 patients with indolent non-Hodgkin’s B cell lymphoma were treated with intratumoral Flt3L, local XRT of the tumor and intratumoral pICLC. PBMCs collected before the start of treatment and 1 d after the last Flt3L injection were analyzed by CyTOF. a, Relative numbers of DC cell subsets among all live cells. Paired two-tailed t test. b, Relative numbers of classical DC subsets among all cDC. Paired two-tailed t test. c, PBMCs collected before and after Flt3L treatment were stimulated with pIC, and cytokine production in the supernatant was analyzed by Luminex. n = 11, one-way ANOVA with Tukey’s post test. d,e, tumor biopsies from one patient with partial response collected before treatment and 6 months post vaccination were analyzed for presence of lymphoma (κ-LC+) B cells (d) and TCs (e) by flow cytometry. f, Pearson correlation of frequency of DC subsets pretreatment with abscopal BOR (n = 11). Data are presented as mean ± s.e.m.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hammerich, L., Marron, T.U., Upadhyay, R. et al. Systemic clinical tumor regressions and potentiation of PD1 blockade with in situ vaccination. Nat Med 25, 814–824 (2019). https://doi.org/10.1038/s41591-019-0410-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-019-0410-x

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