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.

  • Perspective
  • Published:

Antitumour dendritic cell vaccination in a priming and boosting approach

Abstract

Mobilizing antitumour immunity through vaccination potentially constitutes a powerful anticancer strategy but has not yet provided robust clinical benefits in large patient populations. Although major hurdles still exist, we believe that currently available strategies for vaccines that target dendritic cells or use them to present antitumour antigens could be integrated into existing clinical practice using prime–boost approaches. In the priming phase, these approaches capitalize on either standard treatment modalities to trigger in situ vaccination and release tumour antigens or vaccination with dendritic cells loaded with tumour lysates or patient-specific neoantigens. In a second boost phase, personalized synthetic vaccines specifically boost T cells that were triggered during the priming phase. This immunotherapy approach has been enabled by the substantial recent improvements in dendritic cell vaccines. In this Perspective, we discuss these improvements, highlight how the prime–boost approach can be translated into clinical practice and provide solutions for various anticipated hurdles.

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: Mechanisms involved in tumour immunogenicity induced by standard treatment modalities.
Fig. 2: Key aspects involved in the design of whole tumour-based anticancer therapeutic vaccines.
Fig. 3: Potential sources of tumour neoantigens.
Fig. 4: A dendritic cell-based synthetic vaccine in a priming and boost approach.

Similar content being viewed by others

References

  1. Zhang, L. et al. Intratumoral T cells, recurrence, and survival in epithelial ovarian cancer. N. Engl. J. Med. 348, 203–213 (2003).

    CAS  PubMed  Google Scholar 

  2. Fu, Q. et al. Prognostic value of tumor-infiltrating lymphocytes in melanoma: a systematic review and meta-analysis. Oncoimmunology 8, 1593806 (2019).

    PubMed  PubMed Central  Google Scholar 

  3. Ingold Heppner, B., Loibl, S. & Denkert, C. Tumor-infiltrating lymphocytes: a promising biomarker in breast cancer. Breast Care 11, 96–100 (2016).

    PubMed  PubMed Central  Google Scholar 

  4. Beatty, G. L. & Gladney, W. L. Immune escape mechanisms as a guide for cancer immunotherapy. Clin. Cancer Res. 21, 687–692 (2015).

    CAS  PubMed  Google Scholar 

  5. O’Keeffe, M., Mok, W. H. & Radford, K. J. Human dendritic cell subsets and function in health and disease. Cell Mol. Life Sci. 72, 4309–4325 (2015).

    PubMed  Google Scholar 

  6. Banchereau, J. et al. Immunobiology of dendritic cells. Annu. Rev. Immunol. 18, 767–811 (2000).

    CAS  PubMed  Google Scholar 

  7. Disis, M. L. Mechanism of action of immunotherapy. Semin. Oncol. 41 (Suppl. 5), 3–13 (2014).

    Google Scholar 

  8. Martin Lluesma, S., Wolfer, A., Harari, A. & Kandalaft, L. E. Cancer vaccines in ovarian cancer: how can we improve? Biomedicines 4, 10 (2016).

    PubMed Central  Google Scholar 

  9. Small, E. J. et al. Placebo-controlled phase III trial of immunologic therapy with sipuleucel-T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer. J. Clin. Oncol. 24, 3089–3094 (2006).

    CAS  PubMed  Google Scholar 

  10. Garg, A. D., Coulie, P. G., Van den Eynde, B. J. & Agostinis, P. Integrating next-generation dendritic cell vaccines into the current cancer immunotherapy landscape. Trends Immunol. 38, 577–593 (2017).

    CAS  PubMed  Google Scholar 

  11. Odunsi, K. et al. Vaccination with an NY-ESO-1 peptide of HLA class I/II specificities induces integrated humoral and T cell responses in ovarian cancer. Proc. Natl Acad. Sci. USA 104, 12837–12842 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Zhao, L., Zhang, M. & Cong, H. Advances in the study of HLA-restricted epitope vaccines. Hum. Vaccin. Immunother. 9, 2566–2577 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Bezu, L. et al. Trial watch: peptide-based vaccines in anticancer therapy. Oncoimmunology 7, e1511506 (2018).

    PubMed  PubMed Central  Google Scholar 

  14. Chiang, C. L., Coukos, G. & Kandalaft, L. E. Whole tumor antigen vaccines: where are we? Vaccines 3, 344–372 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Alspach, E. et al. MHC-II neoantigens shape tumour immunity and response to immunotherapy. Nature 574, 696–701 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Melief, C. J. & van der Burg, S. H. Immunotherapy of established (pre)malignant disease by synthetic long peptide vaccines. Nat. Rev. Cancer 8, 351–360 (2008).

    CAS  PubMed  Google Scholar 

  17. Dikov, M. M. et al. Differential roles of vascular endothelial growth factor receptors 1 and 2 in dendritic cell differentiation. J. Immunol. 174, 215–222 (2005).

    CAS  PubMed  Google Scholar 

  18. Rabinowich, H. et al. Lymphocyte apoptosis induced by Fas ligand-expressing ovarian carcinoma cells. Implications for altered expression of T cell receptor in tumor-associated lymphocytes. J. Clin. Invest. 101, 2579–2588 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Groh, V., Wu, J., Yee, C. & Spies, T. Tumour-derived soluble MIC ligands impair expression of NKG2D and T-cell activation. Nature 419, 734 (2002).

    CAS  PubMed  Google Scholar 

  20. Loercher, A. E., Nash, M. A., Kavanagh, J. J., Platsoucas, C. D. & Freedman, R. S. Identification of an IL-10-producing HLA-DR-negative monocyte subset in the malignant ascites of patients with ovarian carcinoma that inhibits cytokine protein expression and proliferation of autologous T cells. J. Immunol. 163, 6251–6260 (1999).

    CAS  PubMed  Google Scholar 

  21. Li, M. O., Wan, Y. Y., Sanjabi, S., Robertson, A.-K. L. & Flavell, R. A. Transforming growth factor-β regulation of immune responses. Annu. Rev. Immunol. 24, 99–146 (2006).

    CAS  PubMed  Google Scholar 

  22. Vandenberk, L., Belmans, J., Van Woensel, M., Riva, M. & Van Gool, S. W. Exploiting the immunogenic potential of cancer cells for improved dendritic cell vaccines. Front. Immunol. 6, 663 (2015).

    PubMed  Google Scholar 

  23. Mookerjee, A., Graciotti, M., Kandalaft, L. E. & Kandalaft, L. A cancer vaccine with dendritic cells differentiated with GM-CSF and IFNalpha and pulsed with a squaric acid treated cell lysate improves T cell priming and tumor growth control in a mouse model. Bioimpacts 8, 211–221 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Tanyi, J. L. et al. Personalized cancer vaccine effectively mobilizes antitumor T cell immunity in ovarian cancer. Sci. Transl Med. 10, eaao5931 (2018).

    PubMed  Google Scholar 

  25. Kroemer, G., Galluzzi, L., Kepp, O. & Zitvogel, L. Immunogenic cell death in cancer therapy. Annu. Rev. Immunol. 31, 51–72 (2013).

    CAS  PubMed  Google Scholar 

  26. Kawasaki, T. & Kawai, T. Toll-like receptor signaling pathways. Front. Immunol. 5, 461 (2014).

    PubMed  PubMed Central  Google Scholar 

  27. Chiang, C. L. & Kandalaft, L. E. In vivo cancer vaccination: which dendritic cells to target and how? Cancer Treat. Rev. 71, 88–101 (2018).

    CAS  PubMed  Google Scholar 

  28. Su, Z. et al. Immunological and clinical responses in metastatic renal cancer patients vaccinated with tumor RNA-transfected dendritic cells. Cancer Res. 63, 2127–2133 (2003).

    CAS  PubMed  Google Scholar 

  29. Kyte, J. A. et al. Phase I/II trial of melanoma therapy with dendritic cells transfected with autologous tumor-mRNA. Cancer Gene Ther. 13, 905–918 (2006).

    CAS  PubMed  Google Scholar 

  30. Vik-Mo, E. O. et al. Therapeutic vaccination against autologous cancer stem cells with mRNA-transfected dendritic cells in patients with glioblastoma. Cancer Immunol. Immunother. 62, 1499–1509 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Markov, O., Oshchepkova, A. & Mironova, N. Immunotherapy based on dendritic cell-targeted/-derived extracellular vesicles — a novel strategy for enhancement of the anti-tumor immune response. Front. Pharmacol. 10, 1152 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Patel, G. K. et al. Comparative analysis of exosome isolation methods using culture supernatant for optimum yield, purity and downstream applications. Sci. Rep. 9, 5335 (2019).

    PubMed  PubMed Central  Google Scholar 

  33. Whiteside, T. L. Tumor-derived exosomes and their role in cancer progression. Adv. Clin. Chem. 74, 103–141 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Palucka, A. K. et al. Dendritic cells loaded with killed allogeneic melanoma cells can induce objective clinical responses and MART-1 specific CD8+ T-cell immunity. J. Immunother. 29, 545–557 (2006).

    CAS  PubMed  Google Scholar 

  35. Di Nicola, M. et al. Vaccination with autologous tumor-loaded dendritic cells induces clinical and immunologic responses in indolent B-cell lymphoma patients with relapsed and measurable disease: a pilot study. Blood 113, 18–27 (2009).

    PubMed  Google Scholar 

  36. Ernstoff, M. S. et al. Developing a rational tumor vaccine therapy for renal cell carcinoma: immune yin and yang. Clin. Cancer Res. 13, 733s–740s (2007).

    CAS  PubMed  Google Scholar 

  37. Neller, M. A., Lopez, J. A. & Schmidt, C. W. Antigens for cancer immunotherapy. Semin. Immunol. 20, 286–295 (2008).

    CAS  PubMed  Google Scholar 

  38. Bol, K. F., Schreibelt, G., Gerritsen, W. R., de Vries, I. J. & Figdor, C. G. Dendritic cell-based immunotherapy: state of the art and beyond. Clin. Cancer Res. 22, 1897–1906 (2016).

    CAS  PubMed  Google Scholar 

  39. Galluzzi, L., Senovilla, L., Zitvogel, L. & Kroemer, G. The secretally: immunostimulation by anticancer drugs. Nat. Rev. Drug Discov. 11, 215–233 (2012).

    CAS  PubMed  Google Scholar 

  40. Mikyskova, R. et al. Dendritic cells pulsed with tumor cells killed by high hydrostatic pressure induce strong immune responses and display therapeutic effects both in murine TC-1 and TRAMP-C2 tumors when combined with docetaxel chemotherapy. Int. J. Oncol. 48, 953–964 (2016).

    CAS  PubMed  Google Scholar 

  41. Casares, N. et al. Caspase-dependent immunogenicity of doxorubicin-induced tumor cell death. J. Exp. Med. 202, 1691–1701 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  43. Martin Lluesma, S., Graciotti, M., Chiang, C. L. & Kandalaft, L. E. Does the immunocompetent status of cancer patients have an impact on therapeutic DC vaccination strategies? Vaccines 6, 79 (2018).

    PubMed Central  Google Scholar 

  44. Demaria, S., Coleman, C. N. & Formenti, S. C. Radiotherapy: changing the game in immunotherapy. Trends Cancer 2, 286–294 (2016).

    PubMed  PubMed Central  Google Scholar 

  45. Formenti, S. C. & Demaria, S. Radiation therapy to convert the tumor into an in situ vaccine. Int. J. Radiat. Oncol. Biol. Phys. 84, 879–880 (2012).

    PubMed  Google Scholar 

  46. Golden, E. B., Demaria, S., Schiff, P. B., Chachoua, A. & Formenti, S. C. An abscopal response to radiation and ipilimumab in a patient with metastatic non-small cell lung cancer. Cancer Immunol. Res. 1, 365–372 (2013).

    PubMed  PubMed Central  Google Scholar 

  47. Herrera, F. G., Fyles, A. & Milosevic, M. In regard to Ishikawa et al.: Cyclooxygenase-2 impairs treatment effect of radiotherapy for cervical cancer by inhibition of radiation-induced apoptosis (Int J Radiat Oncol Biol Phys 2006;66:1347–1355). Int. J. Radiat. Oncol. Biol. Phys. 68, 959–960; author reply 960 (2007).

    PubMed  Google Scholar 

  48. Hiniker, S. M. et al. A systemic complete response of metastatic melanoma to local radiation and immunotherapy. Transl Oncol. 5, 404–407 (2012).

    PubMed  PubMed Central  Google Scholar 

  49. Postow, M. A. et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N. Engl. J. Med. 366, 925–931 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Tabi, Z. et al. Resistance of CD45RA- T cells to apoptosis and functional impairment, and activation of tumor-antigen specific T cells during radiation therapy of prostate cancer. J. Immunol. 185, 1330–1339 (2010).

    CAS  PubMed  Google Scholar 

  51. Herrera, F. G. et al. 50-Gy stereotactic body radiation therapy to the dominant intraprostatic nodule: results from a phase 1a/b trial. Int. J. Radiat. Oncol. Biol. Phys. 103, 320–334 (2019).

    PubMed  Google Scholar 

  52. Kato, T. et al. Characterization of the cryoablation-induced immune response in kidney cancer patients. Oncoimmunology 6, e1326441 (2017).

    PubMed  PubMed Central  Google Scholar 

  53. Yakkala, C., Chiang, C. L., Kandalaft, L., Denys, A. & Duran, R. Cryoablation and immunotherapy: an enthralling synergy to confront the tumors. Front. Immunol. 10, 2283 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Machlenkin, A. et al. Combined dendritic cell cryotherapy of tumor induces systemic antimetastatic immunity. Clin. Cancer Res. 11, 4955–4961 (2005).

    CAS  PubMed  Google Scholar 

  55. Udagawa, M. et al. Enhancement of immunologic tumor regression by intratumoral administration of dendritic cells in combination with cryoablative tumor pretreatment and Bacillus Calmette-Guerin cell wall skeleton stimulation. Clin. Cancer Res. 12, 7465–7475 (2006).

    CAS  PubMed  Google Scholar 

  56. Collin, M. & Bigley, V. Human dendritic cell subsets: an update. Immunology 154, 3–20 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Wculek, S. K. et al. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 20, 7–24 (2020).

    CAS  PubMed  Google Scholar 

  58. Osugi, Y., Vuckovic, S. & Hart, D. N. Myeloid blood CD11c+ dendritic cells and monocyte-derived dendritic cells differ in their ability to stimulate T lymphocytes. Blood 100, 2858–2866 (2002).

    CAS  PubMed  Google Scholar 

  59. Prue, R. L. et al. A phase I clinical trial of CD1c (BDCA-1)+ dendritic cells pulsed with HLA-A*0201 peptides for immunotherapy of metastatic hormone refractory prostate cancer. J. Immunother. 38, 71–76 (2015).

    CAS  PubMed  Google Scholar 

  60. Davis, I. D. et al. Blood dendritic cells generated with Flt3 ligand and CD40 ligand prime CD8+ T cells efficiently in cancer patients. J. Immunother. 29, 499–511 (2006).

    PubMed  Google Scholar 

  61. Westdorp, H. et al. Blood-derived dendritic cell vaccinations induce immune responses that correlate with clinical outcome in patients with chemo-naive castration-resistant prostate cancer. J. Immunother. Cancer 7, 302 (2019).

    PubMed  PubMed Central  Google Scholar 

  62. Hsu, J. L. et al. A blood dendritic cell vaccine for acute myeloid leukemia expands anti-tumor T cell responses at remission. Oncoimmunology 7, e1419114 (2018).

    PubMed  PubMed Central  Google Scholar 

  63. Koucky, V., Boucek, J. & Fialova, A. Immunology of plasmacytoid dendritic cells in solid tumors: a brief review. Cancers 11, 470 (2019).

    CAS  PubMed Central  Google Scholar 

  64. Perez, C. R. & De Palma, M. Engineering dendritic cell vaccines to improve cancer immunotherapy. Nat. Commun. 10, 5408 (2019).

    PubMed  PubMed Central  Google Scholar 

  65. Bol, K. F. et al. The clinical application of cancer immunotherapy based on naturally circulating dendritic cells. J. Immunother. Cancer 7, 109 (2019).

    PubMed  PubMed Central  Google Scholar 

  66. Guo, C. et al. Therapeutic cancer vaccines: past, present, and future. Adv. Cancer Res. 119, 421–475 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Marciani, D. J. Vaccine adjuvants: role and mechanisms of action in vaccine immunogenicity. Drug Discov. Today 8, 934–943 (2003).

    CAS  PubMed  Google Scholar 

  68. Obeid, J., Hu, Y. & Slingluff, C. L. Jr. Vaccines, adjuvants, and dendritic cell activators–current status and future challenges. Semin. Oncol. 42, 549–561 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Saxena, M. & Bhardwaj, N. Turbocharging vaccines: emerging adjuvants for dendritic cell based therapeutic cancer vaccines. Curr. Opin. Immunol. 47, 35–43 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Kong, B. Y. et al. On the other side: manipulating the immune checkpoint landscape of dendritic cells to enhance cancer immunotherapy. Front. Oncol. 9, 50–50 (2019).

    PubMed  PubMed Central  Google Scholar 

  71. Brown, J. A. et al. Blockade of programmed death-1 ligands on dendritic cells enhances T cell activation and cytokine production. J. Immunol. 170, 1257–1266 (2003).

    CAS  PubMed  Google Scholar 

  72. Rosenblatt, J. et al. PD-1 blockade by CT-011, anti-PD-1 antibody, enhances ex vivo T-cell responses to autologous dendritic cell/myeloma fusion vaccine. J. Immunother. 34, 409–418 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Son, C. H. et al. CTLA-4 blockade enhances antitumor immunity of intratumoral injection of immature dendritic cells into irradiated tumor in a mouse colon cancer model. J. Immunother. 37, 1–7 (2014).

    CAS  PubMed  Google Scholar 

  74. Ribas, A. et al. Dendritic cell vaccination combined with CTLA4 blockade in patients with metastatic melanoma. Clin. Cancer Res. 15, 6267–6276 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Shimabukuro-Vornhagen, A. et al. The immunosuppressive factors IL-10, TGF-beta, and VEGF do not affect the antigen-presenting function of CD40-activated B cells. J. Exp. Clin. Cancer Res. 31, 47 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Kondo, E. et al. CD40-activated B cells can be generated in high number and purity in cancer patients: analysis of immunogenicity and homing potential. Clin. Exp. Immunol. 155, 249–256 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Wennhold, K., Shimabukuro-Vornhagen, A. & von Bergwelt-Baildon, M. B cell-based cancer immunotherapy. Transfus. Med. Hemother. 46, 36–46 (2019).

    PubMed  PubMed Central  Google Scholar 

  78. Andreesen, R., Hennemann, B. & Krause, S. W. Adoptive immunotherapy of cancer using monocyte-derived macrophages: rationale, current status, and perspectives. J. Leukoc. Biol. 64, 419–426 (1998).

    CAS  PubMed  Google Scholar 

  79. Lee, S., Kivimae, S., Dolor, A. & Szoka, F. C. Macrophage-based cell therapies: the long and winding road. J. Control. Rel. 240, 527–540 (2016).

    CAS  Google Scholar 

  80. Dohnal, A. M. et al. Phase I study of tumor Ag-loaded IL-12 secreting semi-mature DC for the treatment of pediatric cancer. Cytotherapy 9, 755–770 (2007).

    CAS  PubMed  Google Scholar 

  81. Fong, L., Brockstedt, D., Benike, C., Wu, L. & Engleman, E. G. Dendritic cells injected via different routes induce immunity in cancer patients. J. Immunol. 166, 4254–4259 (2001).

    CAS  PubMed  Google Scholar 

  82. West, E. et al. Clinical grade OK432-activated dendritic cells: in vitro characterization and tracking during intralymphatic delivery. J. Immunother. 32, 66–78 (2009).

    CAS  PubMed  Google Scholar 

  83. Salgaller, M. L. et al. Report of immune monitoring of prostate cancer patients undergoing T-cell therapy using dendritic cells pulsed with HLA-A2-specific peptides from prostate-specific membrane antigen (PSMA). Prostate 35, 144–151 (1998).

    CAS  PubMed  Google Scholar 

  84. Morse, M. A. et al. A phase I study of active immunotherapy with carcinoembryonic antigen peptide (CAP-1)-pulsed, autologous human cultured dendritic cells in patients with metastatic malignancies expressing carcinoembryonic antigen. Clin. Cancer Res. 5, 1331–1338 (1999).

    CAS  PubMed  Google Scholar 

  85. Barratt-Boyes, S. M., Watkins, S. C. & Finn, O. J. Migration of cultured chimpanzee dendritic cells following intravenous and subcutaneous injection. Adv. Exp. Med. Biol. 417, 71–75 (1997).

    CAS  PubMed  Google Scholar 

  86. Nestle, F. O. et al. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat. Med. 4, 328–332 (1998).

    CAS  PubMed  Google Scholar 

  87. Koski, G. K., Cohen, P. A., Roses, R. E., Xu, S. & Czerniecki, B. J. Reengineering dendritic cell-based anti-cancer vaccines. Immunol. Rev. 222, 256–276 (2008).

    CAS  PubMed  Google Scholar 

  88. Lambert, L. A. et al. Intranodal immunization with tumor lysate-pulsed dendritic cells enhances protective antitumor immunity. Cancer Res. 61, 641–646 (2001).

    CAS  PubMed  Google Scholar 

  89. Seyfizadeh, N., Muthuswamy, R., Mitchell, D. A., Nierkens, S. & Seyfizadeh, N. Migration of dendritic cells to the lymph nodes and its enhancement to drive anti-tumor responses. Crit. Rev. Oncol. Hematol. 107, 100–110 (2016).

    PubMed  Google Scholar 

  90. Aarntzen, E. H. et al. Targeting of 111In-labeled dendritic cell human vaccines improved by reducing number of cells. Clin. Cancer Res. 19, 1525–1533 (2013).

    CAS  PubMed  Google Scholar 

  91. Aarntzen, E. H. et al. Reducing cell number improves the homing of dendritic cells to lymph nodes upon intradermal vaccination. Oncoimmunology 2, e24661 (2013).

    PubMed  PubMed Central  Google Scholar 

  92. Weiden, J., Tel, J. & Figdor, C. G. Synthetic immune niches for cancer immunotherapy. Nat. Rev. Immunol. 18, 212–219 (2018).

    CAS  PubMed  Google Scholar 

  93. Hori, Y., Winans, A. M., Huang, C. C., Horrigan, E. M. & Irvine, D. J. Injectable dendritic cell-carrying alginate gels for immunization and immunotherapy. Biomaterials 29, 3671–3682 (2008).

    CAS  PubMed  Google Scholar 

  94. Verma, V. et al. Activated dendritic cells delivered in tissue compatible biomatrices induce in-situ anti-tumor CTL responses leading to tumor regression. Oncotarget 7, 39894–39906 (2016).

    PubMed  PubMed Central  Google Scholar 

  95. Aurisicchio, L. & Ciliberto, G. Genetic cancer vaccines: current status and perspectives. Expert. Opin. Biol. Ther. 12, 1043–1058 (2012).

    CAS  PubMed  Google Scholar 

  96. Graciotti, M., Berti, C., Klok, H. A. & Kandalaft, L. The era of bioengineering: how will this affect the next generation of cancer immunotherapy? J. Transl Med. 15, 142 (2017).

    PubMed  PubMed Central  Google Scholar 

  97. Calmeiro, J. et al. Biomaterial-based platforms for in situ dendritic cell programming and their use in antitumor immunotherapy. J. Immunother. Cancer 7, 238 (2019).

    PubMed  PubMed Central  Google Scholar 

  98. Manolova, V. et al. Nanoparticles target distinct dendritic cell populations according to their size. Eur. J. Immunol. 38, 1404–1413 (2008).

    CAS  PubMed  Google Scholar 

  99. Cruz, L. J. et al. Targeting nanoparticles to CD40, DEC-205 or CD11c molecules on dendritic cells for efficient CD8+ T cell response: a comparative study. J. Control. Rel. 192, 209–218 (2014).

    CAS  Google Scholar 

  100. Kranz, L. M. et al. Systemic RNA delivery to dendritic cells exploits antiviral defence for cancer immunotherapy. Nature 534, 396–401 (2016).

    PubMed  Google Scholar 

  101. Ali, O. A., Emerich, D., Dranoff, G. & Mooney, D. J. In situ regulation of DC subsets and T cells mediates tumor regression in mice. Sci. Transl Med. 1, 8ra19 (2009).

    PubMed  PubMed Central  Google Scholar 

  102. Ali, O. A., Huebsch, N., Cao, L., Dranoff, G. & Mooney, D. J. Infection-mimicking materials to program dendritic cells in situ. Nat. Mater. 8, 151–158 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Tran Janco, J. M., Lamichhane, P., Karyampudi, L. & Knutson, K. L. Tumor-infiltrating dendritic cells in cancer pathogenesis. J. Immunol. 194, 2985–2991 (2015).

    PubMed  Google Scholar 

  104. Karthaus, N., Torensma, R. & Tel, J. Deciphering the message broadcast by tumor-infiltrating dendritic cells. Am. J. Pathol. 181, 733–742 (2012).

    CAS  PubMed  Google Scholar 

  105. Herr, H. W. et al. Intravesical bacillus Calmette-Guerin therapy prevents tumor progression and death from superficial bladder cancer: ten-year follow-up of a prospective randomized trial. J. Clin. Oncol. 13, 1404–1408 (1995).

    CAS  PubMed  Google Scholar 

  106. Carpentier, A. et al. Phase 1 trial of a CpG oligodeoxynucleotide for patients with recurrent glioblastoma. Neuro Oncol. 8, 60–66 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Fricke, I. & Gabrilovich, D. I. Dendritic cells and tumor microenvironment: a dangerous liaison. Immunol. Invest. 35, 459–483 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Laoui, D. et al. The tumour microenvironment harbours ontogenically distinct dendritic cell populations with opposing effects on tumour immunity. Nat. Commun. 7, 13720 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Vigneron, N., Stroobant, V., Van den Eynde, B. J. & van der Bruggen, P. Database of T cell-defined human tumor antigens: the 2013 update. Cancer Immun. 13, 15 (2013).

    PubMed  PubMed Central  Google Scholar 

  110. Cobbold, M. et al. MHC class I-associated phosphopeptides are the targets of memory-like immunity in leukemia. Sci. Transl Med. 5, 203ra125 (2013).

    PubMed  PubMed Central  Google Scholar 

  111. Lin, M. H. et al. Immunological evaluation of a novel HLA-A2 restricted phosphopeptide of tumor associated antigen, TRAP1, on cancer therapy. Vaccine X 1, 100017 (2019).

    PubMed  PubMed Central  Google Scholar 

  112. Rizvi, N. A. et al. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348, 124–128 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Snyder, A. et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 371, 2189–2199 (2014).

    PubMed  PubMed Central  Google Scholar 

  114. McGranahan, N. & Swanton, C. Clonal heterogeneity and tumor evolution: past, present, and the future. Cell 168, 613–628 (2017).

    CAS  PubMed  Google Scholar 

  115. Wolf, Y. et al. UVB-induced tumor heterogeneity diminishes immune response in melanoma. Cell 179, 219–235 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Gejman, R. S. et al. Rejection of immunogenic tumor clones is limited by clonal fraction. eLife 7, e41090 (2018).

    PubMed  PubMed Central  Google Scholar 

  117. Jamal-Hanjani, M. et al. Tracking the evolution of non-small-cell lung cancer. N. Engl. J. Med. 376, 2109–2121 (2017).

    CAS  PubMed  Google Scholar 

  118. Robbins, P. F. et al. A pilot trial using lymphocytes genetically engineered with an NY-ESO-1-reactive T-cell receptor: long-term follow-up and correlates with response. Clin. Cancer Res. 21, 1019–1027 (2015).

    CAS  PubMed  Google Scholar 

  119. Porter, D. L. et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci. Transl Med. 7, 303ra139 (2015).

    PubMed  PubMed Central  Google Scholar 

  120. Yamamoto, T. N., Kishton, R. J. & Restifo, N. P. Developing neoantigen-targeted T cell–based treatments for solid tumors. Nat. Med. 25, 1488–1499 (2019).

    CAS  PubMed  Google Scholar 

  121. McGranahan, N. et al. Clonal status of actionable driver events and the timing of mutational processes in cancer evolution. Sci. Transl Med. 7, 283ra254 (2015).

    Google Scholar 

  122. Schumacher, T. N. & Schreiber, R. D. Neoantigens in cancer immunotherapy. Science 348, 69–74 (2015).

    CAS  PubMed  Google Scholar 

  123. Laumont, C. M. et al. Global proteogenomic analysis of human MHC class I-associated peptides derived from non-canonical reading frames. Nat. Commun. 7, 10238 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Sebestyen, E. et al. Large-scale analysis of genome and transcriptome alterations in multiple tumors unveils novel cancer-relevant splicing networks. Genome Res. 26, 732–744 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Uenaka, A. et al. Identification of a unique antigen peptide pRL1 on BALB/c RL male 1 leukemia recognized by cytotoxic T lymphocytes and its relation to the Akt oncogene. J. Exp. Med. 180, 1599–1607 (1994).

    CAS  PubMed  Google Scholar 

  126. Coulie, P. G. et al. A mutated intron sequence codes for an antigenic peptide recognized by cytolytic T lymphocytes on a human melanoma. Proc. Natl Acad. Sci. USA 92, 7976–7980 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Robbins, P. F. et al. The intronic region of an incompletely spliced gp100 gene transcript encodes an epitope recognized by melanoma-reactive tumor-infiltrating lymphocytes. J. Immunol. 159, 303–308 (1997).

    CAS  PubMed  Google Scholar 

  128. Lupetti, R. et al. Translation of a retained intron in tyrosinase-related protein (TRP) 2 mRNA generates a new cytotoxic T lymphocyte (CTL)-defined and shared human melanoma antigen not expressed in normal cells of the melanocytic lineage. J. Exp. Med. 188, 1005–1016 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Wang, R. F., Parkhurst, M. R., Kawakami, Y., Robbins, P. F. & Rosenberg, S. A. Utilization of an alternative open reading frame of a normal gene in generating a novel human cancer antigen. J. Exp. Med. 183, 1131–1140 (1996).

    CAS  PubMed  Google Scholar 

  130. Wang, R. F. et al. A breast and melanoma-shared tumor antigen: T cell responses to antigenic peptides translated from different open reading frames. J. Immunol. 161, 3598–3606 (1998).

    CAS  PubMed  Google Scholar 

  131. Rosenberg, S. A. et al. Identification of BING-4 cancer antigen translated from an alternative open reading frame of a gene in the extended MHC class II region using lymphocytes from a patient with a durable complete regression following immunotherapy. J. Immunol. 168, 2402–2407 (2002).

    CAS  PubMed  Google Scholar 

  132. Ronsin, C. et al. A non-AUG-defined alternative open reading frame of the intestinal carboxyl esterase mRNA generates an epitope recognized by renal cell carcinoma-reactive tumor-infiltrating lymphocytes in situ. J. Immunol. 163, 483–490 (1999).

    CAS  PubMed  Google Scholar 

  133. Mayrand, S. M., Schwarz, D. A. & Green, W. R. An alternative translational reading frame encodes an immunodominant retroviral CTL determinant expressed by an immunodeficiency-causing retrovirus. J. Immunol. 160, 39–50 (1998).

    CAS  PubMed  Google Scholar 

  134. Van Den Eynde, B. J. et al. A new antigen recognized by cytolytic T lymphocytes on a human kidney tumor results from reverse strand transcription. J. Exp. Med. 190, 1793–1800 (1999).

    PubMed Central  Google Scholar 

  135. Attig, J. et al. LTR retroelement expansion of the human cancer transcriptome and immunopeptidome revealed by de novo transcript assembly. Genome Res. 29, 1578–1590 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Chong, C. et al. Integrated proteogenomic deep sequencing and analytics accurately identify non-canonical peptides in tumor immunopeptidomes. Nat. Commun. 11, 1293 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Laumont, C. M. et al. Noncoding regions are the main source of targetable tumor-specific antigens. Sci. Transl Med. 10, eaau5516 (2018).

    CAS  PubMed  Google Scholar 

  138. Smart, A. C. et al. Intron retention is a source of neoepitopes in cancer. Nat. Biotechnol. 36, 1056–1058 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  139. Zhao, Q. et al. Proteogenomics uncovers a vast repertoire of shared tumor-specific antigens in ovarian cancer. Cancer Immunol. Res. https://doi.org/10.1158/2326-6066.CIR-19-0541 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  140. Abelin, J. G. et al. Defining HLA-II ligand processing and binding rules with mass spectrometry enhances cancer epitope prediction. Immunity 51, 766–779 (2019).

    CAS  PubMed  Google Scholar 

  141. Abelin, J. G. et al. Mass spectrometry profiling of HLA-associated peptidomes in mono-allelic cells enables more accurate epitope prediction. Immunity 46, 315–326 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Bassani-Sternberg, M. et al. Deciphering HLA-I motifs across HLA peptidomes improves neo-antigen predictions and identifies allostery regulating HLA specificity. PLoS Comput. Biol. 13, e1005725 (2017).

    PubMed  PubMed Central  Google Scholar 

  143. Bassani-Sternberg, M. & Gfeller, D. Unsupervised HLA peptidome deconvolution improves ligand prediction accuracy and predicts cooperative effects in peptide-HLA interactions. J. Immunol. 197, 2492–2499 (2016).

    CAS  PubMed  Google Scholar 

  144. Guillaume, P. et al. The C-terminal extension landscape of naturally presented HLA-I ligands. Proc. Natl Acad. Sci. USA 115, 5083–5088 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Jurtz, V. et al. NetMHCpan-4.0: improved peptide-MHC class I interaction predictions integrating eluted ligand and peptide binding affinity data. J. Immunol. https://doi.org/10.4049/jimmunol.1700893 (2017).

  146. Mei, S. et al. A comprehensive review and performance evaluation of bioinformatics tools for HLA class I peptide-binding prediction. Brief Bioinform. https://doi.org/10.1093/bib/bbz051 (2019).

    Article  Google Scholar 

  147. Muller, M., Gfeller, D., Coukos, G. & Bassani-Sternberg, M. ‘Hotspots’ of antigen presentation revealed by human leukocyte antigen ligandomics for neoantigen prioritization. Front. Immunol. 8, 1367 (2017).

    PubMed  PubMed Central  Google Scholar 

  148. Bassani-Sternberg, M. et al. Direct identification of clinically relevant neoepitopes presented on native human melanoma tissue by mass spectrometry. Nat. Commun. 7, 13404 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Ebrahimi-Nik, H. et al. Mass spectrometry driven exploration reveals nuances of neoepitope-driven tumor rejection. JCI Insight 5, e129152 (2019).

    Google Scholar 

  150. Schmidt, J. et al. In silico and cell-based analyses reveal strong divergence between prediction and observation of T-cell–recognized tumor antigen T-cell epitopes. J. Biol. Chem. 292, 11840–11849 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Calis, J. J. et al. Properties of MHC class I presented peptides that enhance immunogenicity. PLoS Comput. Biol. 9, e1003266 (2013).

    PubMed  PubMed Central  Google Scholar 

  152. Bjerregaard, A. M. et al. An analysis of natural T cell responses to predicted tumor neoepitopes. Front. Immunol. 8, 1566 (2017).

    PubMed  PubMed Central  Google Scholar 

  153. Duan, F. et al. Genomic and bioinformatic profiling of mutational neoepitopes reveals new rules to predict anticancer immunogenicity. J. Exp. Med. 211, 2231–2248 (2014).

    PubMed  PubMed Central  Google Scholar 

  154. Jorgensen, K. W., Rasmussen, M., Buus, S. & Nielsen, M. NetMHCstab - predicting stability of peptide-MHC-I complexes; impacts for cytotoxic T lymphocyte epitope discovery. Immunology 141, 18–26 (2014).

    CAS  PubMed  Google Scholar 

  155. Garcia-Garijo, A., Fajardo, C. A. & Gros, A. Determinants for neoantigen identification. Front. Immunol. 10, 1392 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  156. Finn, O. J. Human tumor antigens yesterday, today, and tomorrow. Cancer Immunol. Res. 5, 347–354 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Hundal, J. et al. pVACtools: a computational toolkit to identify and visualize cancer neoantigens. Cancer Immunol. Res. 8, 409–420 (2020).

    PubMed  PubMed Central  Google Scholar 

  158. Hundal, J. et al. pVAC-Seq: a genome-guided in silico approach to identifying tumor neoantigens. Genome Med. 8, 11 (2016).

    PubMed  PubMed Central  Google Scholar 

  159. Bjerregaard, A. M., Nielsen, M., Hadrup, S. R., Szallasi, Z. & Eklund, A. C. MuPeXI: prediction of neo-epitopes from tumor sequencing data. Cancer Immunol. Immunother. 66, 1123–1130 (2017).

    CAS  PubMed  Google Scholar 

  160. Ott, P. A. et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 547, 217–221 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Sahin, U. et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 547, 222–226 (2017).

    CAS  PubMed  Google Scholar 

  162. Carreno, B. M. et al. Cancer immunotherapy. A dendritic cell vaccine increases the breadth and diversity of melanoma neoantigen-specific T cells. Science 348, 803–808 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Hilf, N. et al. Actively personalized vaccination trial for newly diagnosed glioblastoma. Nature 565, 240–245 (2019).

    CAS  PubMed  Google Scholar 

  164. Keskin, D. B. et al. Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial. Nature 565, 234–239 (2019).

    CAS  PubMed  Google Scholar 

  165. DeVault, V. L. et al. Vaccine neoantigens empirically identified through the ex vivo ATLAS platform promote potent therapeutic responses to cancer in mice. SITC https://sitc.planion.com/Web.User/AbstractDet?ACCOUNT=SITC&ABSID=12169&CONF=SITC19&ssoOverride=OFF&CKEY=) (2019).

  166. Gfeller, D. & Bassani-Sternberg, M. Predicting antigen presentation-what could we learn from a million peptides? Front. Immunol. 9, 1716 (2018).

    PubMed  PubMed Central  Google Scholar 

  167. Chen, B. et al. Predicting HLA class II antigen presentation through integrated deep learning. Nat. Biotechnol. 37, 1332–1343 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Racle, J. et al. Robust prediction of HLA class II epitopes by deep motif deconvolution of immunopeptidomes. Nat. Biotechnol. 37, 1283–1286 (2019).

    CAS  PubMed  Google Scholar 

  169. Sarivalasis, A. et al. A phase I/II trial comparing autologous dendritic cell vaccine pulsed either with personalized peptides (PEP-DC) or with tumor lysate (OC-DC) in patients with advanced high-grade ovarian serous carcinoma. J. Transl Med. 17, 391 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  170. Anguille, S., Smits, E. L., Lion, E., van Tendeloo, V. F. & Berneman, Z. N. Clinical use of dendritic cells for cancer therapy. Lancet Oncol. 15, e257–e267 (2014).

    CAS  PubMed  Google Scholar 

  171. Figdor, C. G., de Vries, I. J. M., Lesterhuis, W. J. & Melief, C. J. M. Dendritic cell immunotherapy: mapping the way. Nat. Med. 10, 475–480 (2004).

    CAS  PubMed  Google Scholar 

  172. Bol, K. F. et al. Favorable overall survival in stage III melanoma patients after adjuvant dendritic cell vaccination. Oncoimmunology 5, e1057673 (2016).

    PubMed  Google Scholar 

  173. Nakai, N., Hartmann, G., Kishimoto, S. & Katoh, N. Dendritic cell vaccination in human melanoma: relationships between clinical effects and vaccine parameters. Pigment. Cell Melanoma Res. 23, 607–619 (2010).

    CAS  PubMed  Google Scholar 

  174. Failli, A., Legitimo, A., Orsini, G., Romanini, A. & Consolini, R. Numerical defect of circulating dendritic cell subsets and defective dendritic cell generation from monocytes of patients with advanced melanoma. Cancer Lett. 337, 184–192 (2013).

    CAS  PubMed  Google Scholar 

  175. Legitimo, A., Consolini, R., Failli, A., Orsini, G. & Spisni, R. Dendritic cell defects in the colorectal cancer. Hum. Vaccin. Immunother. 10, 3224–3235 (2014).

    PubMed  PubMed Central  Google Scholar 

  176. Ormandy, L. A. et al. Direct ex vivo analysis of dendritic cells in patients with hepatocellular carcinoma. World J. Gastroenterol. 12, 3275–3282 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  177. Della Bella, S. et al. Altered maturation of peripheral blood dendritic cells in patients with breast cancer. Br. J. Cancer 89, 1463–1472 (2003).

    PubMed  PubMed Central  Google Scholar 

  178. Yanagimoto, H. et al. Impaired function of circulating dendritic cells in patients with pancreatic cancer. Clin. Immunol. 114, 52–60 (2005).

    CAS  PubMed  Google Scholar 

  179. Tucci, M. et al. The immune escape in melanoma: role of the impaired dendritic cell function. Expert. Rev. Clin. Immunol. 10, 1395–1404 (2014).

    CAS  PubMed  Google Scholar 

  180. Orsini, G. et al. Defective generation and maturation of dendritic cells from monocytes in colorectal cancer patients during the course of disease. Int. J. Mol. Sci. 14, 22022–22041 (2013).

    PubMed  PubMed Central  Google Scholar 

  181. van Willigen, W. W. et al. Dendritic cell cancer therapy: vaccinating the right patient at the right time. Front. Immunol. 9, 2265 (2018).

    PubMed  PubMed Central  Google Scholar 

  182. Chan, T. A. et al. Development of tumor mutation burden as an immunotherapy biomarker: utility for the oncology clinic. Ann. Oncol. 30, 44–56 (2019).

    CAS  PubMed  Google Scholar 

  183. Yarchoan, M., Hopkins, A. & Jaffee, E. M. Tumor mutational burden and response rate to PD-1 inhibition. N. Engl. J. Med. 377, 2500–2501 (2017).

    PubMed  PubMed Central  Google Scholar 

  184. Rizvi, H. et al. Molecular determinants of response to anti-programmed cell death (PD)-1 and anti-programmed death-ligand 1 (PD-L1) blockade in patients with non-small-cell lung cancer profiled with targeted next-generation sequencing. J. Clin. Oncol. 36, 633–641 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Goodman, A. M. et al. Tumor mutational burden as an independent predictor of response to immunotherapy in diverse cancers. Mol. Cancer Ther. 16, 2598–2608 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  186. Zarogoulidis, P. et al. Nivolumab as first-line treatment in non-small cell lung cancer patients-key factors: tumor mutation burden and PD-L1 >/=50. Transl Lung Cancer Res. 7, S28–S30 (2018).

    PubMed  PubMed Central  Google Scholar 

  187. Park, S. E. et al. Clinical implication of tumor mutational burden in patients with HER2-positive refractory metastatic breast cancer. Oncoimmunology 7, e1466768 (2018).

    PubMed  PubMed Central  Google Scholar 

  188. Lauss, M. et al. Mutational and putative neoantigen load predict clinical benefit of adoptive T cell therapy in melanoma. Nat. Commun. 8, 1738 (2017).

    PubMed  PubMed Central  Google Scholar 

  189. Chen, D. S. & Mellman, I. Oncology meets immunology: the cancer-immunity cycle. Immunity 39, 1–10 (2013).

    PubMed  Google Scholar 

  190. Truxova, I. et al. Rationale for the combination of dendritic cell-based vaccination approaches with chemotherapy agents. Int. Rev. Cell Mol. Biol. 330, 115–156 (2017).

    CAS  PubMed  Google Scholar 

  191. Galluzzi, L., Buque, A., Kepp, O., Zitvogel, L. & Kroemer, G. Immunogenic cell death in cancer and infectious disease. Nat. Rev. Immunol. 17, 97–111 (2017).

    CAS  PubMed  Google Scholar 

  192. Bassani-Sternberg, M. et al. A phase Ib study of the combination of personalized autologous dendritic cell vaccine, aspirin, and standard of care adjuvant chemotherapy followed by nivolumab for resected pancreatic adenocarcinoma-a proof of antigen discovery feasibility in three patients. Front. Immunol. 10, 1832 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  193. Garg, A. D. et al. Dendritic cell vaccines based on immunogenic cell death elicit danger signals and T cell-driven rejection of high-grade glioma. Sci. Transl Med. 8, 328ra327 (2016).

    Google Scholar 

  194. Chen, F. et al. Neoantigen identification strategies enable personalized immunotherapy in refractory solid tumors. J. Clin. Invest. 129, 2056–2070 (2019).

    PubMed  PubMed Central  Google Scholar 

  195. Yang, W. et al. Immunogenic neoantigens derived from gene fusions stimulate T cell responses. Nat. Med. 25, 767–775 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  196. Sharma, G., Rive, C. M. & Holt, R. A. Rapid selection and identification of functional CD8(+) T cell epitopes from large peptide-coding libraries. Nat. Commun. 10, 4553 (2019).

    PubMed  PubMed Central  Google Scholar 

  197. Beard, R. E. et al. Multiple chimeric antigen receptors successfully target chondroitin sulfate proteoglycan 4 in several different cancer histologies and cancer stem cells. J. Immunother. Cancer 2, 25 (2014).

    PubMed  PubMed Central  Google Scholar 

  198. Zacharakis, N. et al. Immune recognition of somatic mutations leading to complete durable regression in metastatic breast cancer. Nat. Med. 24, 724–730 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  199. Bentzen, A. K. et al. Large-scale detection of antigen-specific T cells using peptide-MHC-I multimers labeled with DNA barcodes. Nat. Biotechnol. 34, 1037–1045 (2016).

    CAS  PubMed  Google Scholar 

  200. Newell, E. W. et al. Combinatorial tetramer staining and mass cytometry analysis facilitate T-cell epitope mapping and characterization. Nat. Biotechnol. 31, 623–629 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  201. Peng, S. et al. Sensitive detection and analysis of neoantigen-specific T cell populations from tumors and blood. Cell Rep. 28, 2728–2738 e2727 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  202. Zhang, S. Q. et al. High-throughput determination of the antigen specificities of T cell receptors in single cells. Nat. Biotechnol. https://doi.org/10.1038/nbt.4282 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  203. Bobisse, S. et al. Sensitive and frequent identification of high avidity neo-epitope specific CD8+ T cells in immunotherapy-naive ovarian cancer. Nat. Commun. 9, 1092 (2018).

    PubMed  PubMed Central  Google Scholar 

  204. Yossef, R. et al. Enhanced detection of neoantigen-reactive T cells targeting unique and shared oncogenes for personalized cancer immunotherapy. JCI Insight 3, e122467 (2018).

    PubMed Central  Google Scholar 

  205. Seliktar-Ofir, S. et al. Selection of shared and neoantigen-reactive T cells for adoptive cell therapy based on CD137 separation. Front. Immunol. 8, 1211 (2017).

    PubMed  PubMed Central  Google Scholar 

  206. Duhen, T. et al. Co-expression of CD39 and CD103 identifies tumor-reactive CD8 T cells in human solid tumors. Nat. Commun. 9, 2724 (2018).

    PubMed  PubMed Central  Google Scholar 

  207. Pasetto, A. et al. Tumor- and neoantigen-reactive T-cell receptors can be identified based on their frequency in fresh tumor. Cancer Immunol. Res. 4, 734–743 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  208. Scheper, W. et al. Low and variable tumor reactivity of the intratumoral TCR repertoire in human cancers. Nat. Med. 25, 89–94 (2019).

    CAS  PubMed  Google Scholar 

  209. Segaliny, A. I. et al. Functional TCR T cell screening using single-cell droplet microfluidics. Lab Chip 18, 3733–3749 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  210. Arnaud, M. et al. Biotechnologies to tackle the challenge of neoantigen identification. Curr. Opin. Biotechnol. 65, 52–59 (2020).

    CAS  PubMed  Google Scholar 

  211. Glass, D. G., McAlinden, N., Millington, O. R. & Wright, A. J. A minimally invasive optical trapping system to understand cellular interactions at onset of an immune response. PLoS ONE 12, e0188581 (2017).

    PubMed  PubMed Central  Google Scholar 

  212. Puech, P. H. et al. Force measurements of TCR/pMHC recognition at T cell surface. PLoS ONE 6, e22344 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  213. Huang, J., Edwards, L. J., Evavold, B. D. & Zhu, C. Kinetics of MHC-CD8 interaction at the T cell membrane. J. Immunol. 179, 7653–7662 (2007).

    CAS  PubMed  Google Scholar 

  214. Dura, B. et al. Profiling lymphocyte interactions at the single-cell level by microfluidic cell pairing. Nat. Commun. 6, 5940 (2015).

    CAS  PubMed  Google Scholar 

  215. Hoover, H. C. Jr., Surdyke, M., Dangel, R. B., Peters, L. C. & Hanna, M. G. Jr. Delayed cutaneous hypersensitivity to autologous tumor cells in colorectal cancer patients immunized with an autologous tumor cell: bacillus Calmette-Guerin vaccine. Cancer Res. 44, 1671–1676 (1984).

    PubMed  Google Scholar 

  216. Wallack, M. K. et al. Positive relationship of clinical and serologic responses to vaccinia melanoma oncolysate. Arch. Surg. 122, 1460–1463 (1987).

    CAS  PubMed  Google Scholar 

  217. Mitchell, M. S. et al. Effectiveness and tolerability of low-dose cyclophosphamide and low-dose intravenous interleukin-2 in disseminated melanoma [corrected]. J. Clin. Oncol. 6, 409–424 (1988).

    CAS  PubMed  Google Scholar 

  218. Elliott, G. T., McLeod, R. A., Perez, J. & Von Eschen, K. B. Interim results of a phase II multicenter clinical trial evaluating the activity of a therapeutic allogeneic melanoma vaccine (theraccine) in the treatment of disseminated malignant melanoma. Semin. Surg. Oncol. 9, 264–272 (1993).

    CAS  PubMed  Google Scholar 

  219. Wallack, M. K. et al. Surgical adjuvant active specific immunotherapy for patients with stage III melanoma: the final analysis of data from a phase III, randomized, double-blind, multicenter vaccinia melanoma oncolysate trial. J. Am. Coll. Surg. 187, 69–79 (1998).

    CAS  PubMed  Google Scholar 

  220. Chang, A. E. et al. A phase I trial of tumor lysate-pulsed dendritic cells in the treatment of advanced cancer. Clin. Cancer Res. 8, 1021–1032 (2002).

    CAS  PubMed  Google Scholar 

  221. Marten, A. et al. Therapeutic vaccination against metastatic renal cell carcinoma by autologous dendritic cells: preclinical results and outcome of a first clinical phase I/II trial. Cancer Immunol. Immunother. 51, 637–644 (2002).

    PubMed  Google Scholar 

  222. Nagayama, H. et al. Results of a phase I clinical study using autologous tumour lysate-pulsed monocyte-derived mature dendritic cell vaccinations for stage IV malignant melanoma patients combined with low dose interleukin-2. Melanoma Res. 13, 521–530 (2003).

    CAS  PubMed  Google Scholar 

  223. Jocham, D. et al. Adjuvant autologous renal tumour cell vaccine and risk of tumour progression in patients with renal-cell carcinoma after radical nephrectomy: phase III, randomised controlled trial. Lancet 363, 594–599 (2004).

    CAS  PubMed  Google Scholar 

  224. Sondak, V. K. et al. Adjuvant immunotherapy of resected, intermediate-thickness, node-negative melanoma with an allogeneic tumor vaccine: overall results of a randomized trial of the Southwest Oncology Group. J. Clin. Oncol. 20, 2058–2066 (2002).

    CAS  PubMed  Google Scholar 

  225. Yamanaka, R. et al. Vaccination of recurrent glioma patients with tumour lysate-pulsed dendritic cells elicits immune responses: results of a clinical phase I/II trial. Br. J. Cancer 89, 1172–1179 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  226. Powell, A., Creaney, J., Broomfield, S., Van Bruggen, I. & Robinson, B. Recombinant GM-CSF plus autologous tumor cells as a vaccine for patients with mesothelioma. Lung Cancer 52, 189–197 (2006).

    PubMed  Google Scholar 

  227. Kim, J. H. et al. Phase I/II study of immunotherapy using autologous tumor lysate-pulsed dendritic cells in patients with metastatic renal cell carcinoma. Clin. Immunol. 125, 257–267 (2007).

    CAS  PubMed  Google Scholar 

  228. Kuwabara, K. et al. Results of a phase I clinical study using dendritic cell vaccinations for thyroid cancer. Thyroid 17, 53–58 (2007).

    CAS  PubMed  Google Scholar 

  229. Ovali, E. et al. Active immunotherapy for cancer patients using tumor lysate pulsed dendritic cell vaccine: a safety study. J. Exp. Clin. Cancer Res. 26, 209–214 (2007).

    CAS  PubMed  Google Scholar 

  230. Dudek, A. Z. et al. Autologous large multivalent immunogen vaccine in patients with metastatic melanoma and renal cell carcinoma. Am. J. Clin. Oncol. 31, 173–181 (2008).

    PubMed  Google Scholar 

  231. Redman, B. G. et al. Phase Ib trial assessing autologous, tumor-pulsed dendritic cells as a vaccine administered with or without IL-2 in patients with metastatic melanoma. J. Immunother. 31, 591–598 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  232. Berntsen, A. et al. Therapeutic dendritic cell vaccination of patients with metastatic renal cell carcinoma: a clinical phase 1/2 trial. J. Immunother. 31, 771–780 (2008).

    CAS  PubMed  Google Scholar 

  233. Burgdorf, S. K. et al. Clinical responses in patients with advanced colorectal cancer to a dendritic cell based vaccine. Oncol. Rep. 20, 1305–1311 (2008).

    PubMed  Google Scholar 

  234. Schwaab, T. et al. Clinical and immunologic effects of intranodal autologous tumor lysate-dendritic cell vaccine with Aldesleukin (Interleukin 2) and IFN-{alpha}2a therapy in metastatic renal cell carcinoma patients. Clin. Cancer Res. 15, 4986–4992 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  235. Ardon, H. et al. Integration of autologous dendritic cell-based immunotherapy in the primary treatment for patients with newly diagnosed glioblastoma multiforme: a pilot study. J. Neurooncol. 99, 261–272 (2010).

    CAS  PubMed  Google Scholar 

  236. Um, S. J. et al. Phase I study of autologous dendritic cell tumor vaccine in patients with non-small cell lung cancer. Lung Cancer 70, 188–194 (2010).

    PubMed  Google Scholar 

  237. Trepiakas, R. et al. Vaccination with autologous dendritic cells pulsed with multiple tumor antigens for treatment of patients with malignant melanoma: results from a phase I/II trial. Cytotherapy 12, 721–734 (2010).

    CAS  PubMed  Google Scholar 

  238. Baek, S. et al. Combination therapy of renal cell carcinoma or breast cancer patients with dendritic cell vaccine and IL-2: results from a phase I/II trial. J. Transl Med. 9, 178 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  239. Cho, D. Y. et al. Adjuvant immunotherapy with whole-cell lysate dendritic cells vaccine for glioblastoma multiforme: a phase II clinical trial. World Neurosurg. 77, 736–744 (2012).

    PubMed  Google Scholar 

  240. Ellebaek, E. et al. Metastatic melanoma patients treated with dendritic cell vaccination, Interleukin-2 and metronomic cyclophosphamide: results from a phase II trial. Cancer Immunol. Immunother. 61, 1791–1804 (2012).

    CAS  PubMed  Google Scholar 

  241. Himoudi, N. et al. Lack of T-cell responses following autologous tumour lysate pulsed dendritic cell vaccination, in patients with relapsed osteosarcoma. Clin. Transl Oncol. 14, 271–279 (2012).

    CAS  PubMed  Google Scholar 

  242. Prins, R. M. et al. Comparison of glioma-associated antigen peptide-loaded versus autologous tumor lysate-loaded dendritic cell vaccination in malignant glioma patients. J. Immunother. 36, 152–157 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  243. Kamigaki, T. et al. Immunotherapy of autologous tumor lysate-loaded dendritic cell vaccines by a closed-flow electroporation system for solid tumors. Anticancer Res. 33, 2971–2976 (2013).

    CAS  PubMed  Google Scholar 

  244. Bapsy, P. P. et al. Open-label, multi-center, non-randomized, single-arm study to evaluate the safety and efficacy of dendritic cell immunotherapy in patients with refractory solid malignancies, on supportive care. Cytotherapy 16, 234–244 (2014).

    CAS  PubMed  Google Scholar 

  245. Baek, S. et al. Therapeutic DC vaccination with IL-2 as a consolidation therapy for ovarian cancer patients: a phase I/II trial. Cell Mol. Immunol. 12, 87–95 (2015).

    CAS  PubMed  Google Scholar 

  246. Caballero-Banos, M. et al. Phase II randomised trial of autologous tumour lysate dendritic cell plus best supportive care compared with best supportive care in pre-treated advanced colorectal cancer patients. Eur. J. Cancer 64, 167–174 (2016).

    PubMed  Google Scholar 

  247. Miwa, S. et al. Phase 1/2 study of immunotherapy with dendritic cells pulsed with autologous tumor lysate in patients with refractory bone and soft tissue sarcoma. Cancer 123, 1576–1584 (2017).

    CAS  PubMed  Google Scholar 

  248. Inoges, S. et al. A phase II trial of autologous dendritic cell vaccination and radiochemotherapy following fluorescence-guided surgery in newly diagnosed glioblastoma patients. J. Transl Med. 15, 104 (2017).

    PubMed  PubMed Central  Google Scholar 

  249. Liau, L. M. et al. First results on survival from a large phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma. J. Transl Med. 16, 142 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  250. Herbert, G. S. et al. Initial phase I/IIa trial results of an autologous tumor lysate, particle-loaded, dendritic cell (TLPLDC) vaccine in patients with solid tumors. Vaccine 36, 3247–3253 (2018).

    CAS  PubMed  Google Scholar 

  251. Buchroithner, J. et al. Audencel immunotherapy based on dendritic cells has no effect on overall and progression-free survival in newly diagnosed glioblastoma: a phase II randomized trial. Cancers 10, 372 (2018).

    CAS  PubMed Central  Google Scholar 

  252. Benitez-Ribas, D. et al. Immune response generated with the administration of autologous dendritic cells pulsed with an allogenic tumoral cell-lines lysate in patients with newly diagnosed diffuse intrinsic pontine glioma. Front. Oncol. 8, 127 (2018).

    PubMed  PubMed Central  Google Scholar 

  253. Rodriguez-Ruiz, M. E. et al. Combined immunotherapy encompassing intratumoral poly-ICLC, dendritic-cell vaccination and radiotherapy in advanced cancer patients. Ann. Oncol. 29, 1312–1319 (2018).

    CAS  PubMed  Google Scholar 

  254. Medzhitov, R. Recognition of microorganisms and activation of the immune response. Nature 449, 819–826 (2007).

    CAS  PubMed  Google Scholar 

  255. Chu, K. F. & Dupuy, D. E. Thermal ablation of tumours: biological mechanisms and advances in therapy. Nat. Rev. Cancer 14, 199–208 (2014).

    CAS  PubMed  Google Scholar 

  256. Saxena, M., Balan, S., Roudko, V. & Bhardwaj, N. Towards superior dendritic-cell vaccines for cancer therapy. Nat. Biomed. Eng. 2, 341–346 (2018).

    PubMed  PubMed Central  Google Scholar 

  257. Patente, T. A. et al. Human dendritic cells: their heterogeneity and clinical application potential in cancer immunotherapy. Front. Immunol. 9, 3176 (2018).

    CAS  PubMed  Google Scholar 

  258. Sheen, M. R. & Fiering, S. In situ vaccination: harvesting low hanging fruit on the cancer immunotherapy tree. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 11, e1524 (2019).

    PubMed  Google Scholar 

  259. Wang, Y.-J., Fletcher, R., Yu, J. & Zhang, L. Immunogenic effects of chemotherapy-induced tumor cell death. Genes. Dis. 5, 194–203 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  260. Pol, J. et al. Trial watch: immunogenic cell death inducers for anticancer chemotherapy. Oncoimmunology 4, e1008866 (2015).

    PubMed  PubMed Central  Google Scholar 

  261. Wu, J. & Waxman, D. J. Immunogenic chemotherapy: dose and schedule dependence and combination with immunotherapy. Cancer Lett. 419, 210–221 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  262. Ghiringhelli, F. et al. Metronomic cyclophosphamide regimen selectively depletes CD4+CD25+ regulatory T cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol. Immunother. 56, 641–648 (2007).

    CAS  PubMed  Google Scholar 

  263. Liu, Y., Cao, C. S., Yu, Y. & Si, Y. M. Thermal ablation in cancer. Oncol. Lett. 12, 2293–2295 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  264. Takaki, H. et al. Thermal ablation and immunomodulation: from preclinical experiments to clinical trials. Diagn. Interv. Imaging 98, 651–659 (2017).

    CAS  PubMed  Google Scholar 

  265. Nakagawa, H. et al. In vivo immunological antitumor effect of OK-432-stimulated dendritic cell transfer after radiofrequency ablation. Cancer Immunol. Immunother. 63, 347–356 (2014).

    CAS  PubMed  Google Scholar 

  266. Xu, H. et al. Synergism between cryoablation and GM-CSF: enhanced immune function of splenic dendritic cells in mice with glioma. Neuroreport 26, 346–353 (2015).

    CAS  PubMed  Google Scholar 

  267. den Brok, M. H. et al. Synergy between in situ cryoablation and TLR9 stimulation results in a highly effective in vivo dendritic cell vaccine. Cancer Res. 66, 7285–7292 (2006).

    Google Scholar 

  268. Levy, M. Y. et al. Cyclophosphamide unmasks an antimetastatic effect of local tumor cryoablation. J. Pharmacol. Exp. Ther. 330, 596–601 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  269. den Brok, M. H. et al. In situ tumor ablation creates an antigen source for the generation of antitumor immunity. Cancer Res. 64, 4024–4029 (2004).

    Google Scholar 

  270. Shi, L. et al. PD-1 blockade boosts radiofrequency ablation–elicited adaptive immune responses against tumor. Clin. Cancer Res. 22, 1173–1184 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  271. Thakur, A. et al. Induction of specific cellular and humoral responses against renal cell carcinoma after combination therapy with cryoablation and granulocyte-macrophage colony stimulating factor: a pilot study. J. Immunother. 34, 457–467 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  272. Niu, L. et al. Combination treatment with comprehensive cryoablation and immunotherapy in metastatic pancreatic cancer. Pancreas 42, 1143–1149 (2013).

    CAS  PubMed  Google Scholar 

  273. McArthur, H. L. et al. A pilot study of preoperative single-dose ipilimumab and/or cryoablation in women with early-stage breast cancer with comprehensive immune profiling. Clin. Cancer Res. 22, 5729–5737 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  274. Hlavata, Z. et al. The abscopal effect in the era of cancer immunotherapy: a spontaneous synergism boosting anti-tumor immunity? Target. Oncol. 13, 113–123 (2018).

    PubMed  Google Scholar 

  275. Golden, E. B. & Apetoh, L. Radiotherapy and immunogenic cell death. Semin. Radiat. Oncol. 25, 11–17 (2015).

    PubMed  Google Scholar 

  276. Matsumura, S. & Demaria, S. Up-regulation of the pro-inflammatory chemokine CXCL16 is a common response of tumor cells to ionizing radiation. Radiat. Res. 173, 418–425 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  277. Reits, E. A. et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J. Exp. Med. 203, 1259–1271 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  278. Bockel, S., Durand, B. & Deutsch, E. Combining radiation therapy and cancer immune therapies: from preclinical findings to clinical applications. Cancer Radiother. 22, 567–580 (2018).

    CAS  PubMed  Google Scholar 

  279. Janeway, C. A. Jr. & Medzhitov, R. Innate immune recognition. Annu. Rev. Immunol. 20, 197–216 (2002).

    CAS  PubMed  Google Scholar 

  280. Vacchelli, E. et al. Trial watch: FDA-approved Toll-like receptor agonists for cancer therapy. Oncoimmunology 1, 894–907 (2012).

    PubMed  PubMed Central  Google Scholar 

  281. Akira, S., Uematsu, S. & Takeuchi, O. Pathogen recognition and innate immunity. Cell 124, 783–801 (2006).

    CAS  PubMed  Google Scholar 

  282. Vacchelli, E. et al. Trial watch: toll-like receptor agonists for cancer therapy. Oncoimmunology 2, e25238 (2013).

    PubMed  PubMed Central  Google Scholar 

  283. Kaczanowska, S., Joseph, A. M. & Davila, E. TLR agonists: our best frenemy in cancer immunotherapy. J. Leukoc. Biol. 93, 847–863 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  284. Tomasicchio, M. et al. An autologous dendritic cell vaccine polarizes a Th-1 response which is tumoricidal to patient-derived breast cancer cells. Cancer Immunol. Immunother. 68, 71–83 (2019).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank K. Balint for assisting in collecting information in Table 1, P. Romero for critically reading the manuscript and G. Coukos for useful discussion.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lana E. Kandalaft.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Harari, A., Graciotti, M., Bassani-Sternberg, M. et al. Antitumour dendritic cell vaccination in a priming and boosting approach. Nat Rev Drug Discov 19, 635–652 (2020). https://doi.org/10.1038/s41573-020-0074-8

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41573-020-0074-8

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