Opinion statement
The primary objective of this study is to evaluate the effectiveness of cancer vaccines containing genetically modified dendritic cells (DCs) in inducing transformational immune responses. This paper sheds considerable light on DCs’ function in advancing treatment techniques. This objective is achieved by thoroughly analyzing the many facets of DCs and their strategic integration into cancer treatment. Due to their role as immune response regulators, DCs can potentially enhance cancer treatment strategies. DCs have the potential to revolutionize immunotherapy, as shown by a comprehensive analysis of their numerous characteristics. The review deftly transitions from examining the fundamentals of preclinical research to delving into the complexities of clinical implementation while acknowledging the inherent challenges in translating DC vaccine concepts into tangible progress. The analysis also emphasizes the potential synergistic outcomes that can be achieved by combining DC vaccines with established pharmaceuticals, thereby emphasizing the importance of employing a holistic approach to enhance treatment efficacy. Despite the existence of transformative opportunities, advancement is hindered by several obstacles. The exhaustive analysis of technical complexities, regulatory dynamics, and upcoming challenges provides valuable insights for overcoming obstacles requiring strategic navigation to incorporate DC vaccines successfully. This document provides a comprehensive analysis of the developments in DC-based immunotherapy, concentrating on its potential to transform cancer therapy radically.
Similar content being viewed by others
References and Recommended Reading
Papers of particular interest, published recently, have been highlighted as: • Of importance
Mehralizadeh H, et al. Cytokine sustained delivery for cancer therapy; special focus on stem cell-and biomaterial-based delivery methods. Pathol-Res Pract. 2023;247:154528.
Shabani S, Moghadam MF, Gargari SL. Isolation and characterization of a novel GRP78-specific single-chain variable fragment (scFv) using ribosome display method. Med Oncol. 2021;38(9):115.
Liu J, et al. Cancer vaccines as promising immuno-therapeutics: platforms and current progress. J Hematol Oncol. 2022;15(1):28.
Kaboli PJ, et al. Shedding light on triple-negative breast cancer with Trop2-targeted antibody-drug conjugates. Am J Cancer Res. 2022;12(4):1671.
Carlson RD, Flickinger JC Jr, Snook AE. Talkin’toxins: from Coley’s to modern cancerimmunotherapy. Toxins. 2020;12(4):241.
• Sabado RL, Balan S, Bhardwaj N. Dendritic cell-based immunotherapy. Cell Res. 2017;27(1):74–95. This review discussed how current approaches apply DC vaccines to improve anti-tumor immunity in clinic.
Okamoto M, et al. Dendritic cell-based vaccine for pancreatic cancer in Japan. World J Gastrointest Pharmacol Ther. 2016;7(1):133.
Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature. 2007;449(7161):419–26.
Steinman RM. Decisions about dendritic cells: past, present, and future. Annu Rev Immunol. 2012;30:1–22.
• Plumas J. Harnessing dendritic cells for innovative therapeutic cancer vaccines. Curr Opin Oncol. 2022;34(2):161–8. This article reviewed recent research using allogeneic DCs as alternatives to autologous DCs to develop innovative therapeutic cancer vaccines.
Abbasi S, et al. Chimeric antigen receptor T (CAR-T) cells: novel cell therapy for hematological malignancies. Cancer Med. 2023;12(7):7844–58.
Kozani PS, Shabani S. Adverse events and side effects of chimeric antigen receptor (CAR) t cell therapy in patients with hematologic malignancies. Trends Med Sci. 2021;1(1):620374.
Saxena M, et al. Therapeutic cancer vaccines. Nat Rev Cancer. 2021;21(6):360–78.
Andtbacka RH, et al. Talimogene laherparepvec improves durable response rate in patients with advanced melanoma. J Clin Oncol. 2015;33(25):2780–8.
Ribas A, et al. Oncolytic virotherapy promotes intratumoral T cell infiltration and improves anti-PD-1 immunotherapy. Cell. 2018;174(4):1031–2.
Harari A, et al. Antitumour dendritic cell vaccination in a priming and boosting approach. Nat Rev Drug Discov. 2020;19(9):635–52.
Tolouei AE, et al. Gingival mesenchymal stem cell therapy, immune cells, and immunoinflammatory application. Mol Biol Rep. 2023:1–9.
Fu C, et al. DC-based vaccines for cancer immunotherapy Vaccines. 2020;8(4):706.
Eisenbarth SC. Dendritic cell subsets in T cell programming: location dictates function. Nat Rev Immunol. 2019;19(2):89–103.
Guermonprez P, et al. Antigen presentation and T cell stimulation by dendritic cells. Annu Rev Immunol. 2002;20(1):621–67.
Patente TA, et al. Human dendritic cells: their heterogeneity and clinical application potential in cancer immunotherapy. Front Immunol. 2019;9:3176.
Hilligan KL, Ronchese F. Antigen presentation by dendritic cells and their instruction of CD4+ T helper cell responses. Cell Mol Immunol. 2020;17(6):587–99.
Wculek SK, et al. Dendritic cells in cancer immunology and immunotherapy. Nat Rev Immunol. 2020;20(1):7–24. This article reviewed different DC subsets immunity and tolerance in cancer settings and immunotherapy strategies.
Anderson DA III, et al. Genetic models of human and mouse dendritic cell development and function. Nat Rev Immunol. 2021;21(2):101–15.
Villani AC, et al. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science. 2017;356(6335):eaah4573.
Collin M, Bigley V. Human dendritic cell subsets: an update. Immunology. 2018;154(1):3–20.
Haniffa M, Collin M, Ginhoux F. Ontogeny and functional specialization of dendritic cells in human and mouse. Adv Immunol. 2013;120:1–49.
Haniffa M, Collin M, Ginhoux F. Identification of human tissue cross-presenting dendritic cells: a new target for cancer vaccines. Oncoimmunol. 2013;2(3): e23140.
Jongbloed SL, Kassianos AJ, McDonald KJ, et al. Human CD141+ (BDCA-3)+ DCs represent a unique myeloid DC subset that cross-presents necrotic cell antigens. J Exp Med. 2010;207(6):1247–60.
Brown CC, et al. Transcriptional basis of mouse and human dendritic cell heterogeneity. Cell. 2019;179(4):846–63.
Yamazaki C, et al. Critical roles of a dendritic cell subset expressing a chemokine receptor, XCR1. J Immunol. 2013;190(12):6071–82.
Dhodapkar MV, et al. Antigen-specific inhibition of effector T cell function in humans after injection of immature dendritic cells. J Exp Med. 2001;193(2):233–8.
Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nat Rev Cancer. 2012;12(4):265–77.
Bonaccorsi I, et al. Novel perspectives on dendritic cell-based immunotherapy of cancer. Immunol Lett. 2013;155(1–2):6–10.
Palucka K, Banchereau J, Mellman I. Designing vaccines based on biology of human dendritic cell subsets. Immunity. 2010;33(4):464–78.
Banchereau J, et al. Immune and clinical responses in patients with metastatic melanoma to CD34+ progenitor-derived dendritic cell vaccine. Cancer Res. 2001;61(17):6451–8.
Mody N, et al. Dendritic cell-based vaccine research against cancer. Expert Rev Clin Immunol. 2015;11(2):213–32.
Trefzer U, et al. Hybrid cell vaccination for cancer immune therapy: first clinical trial with metastatic melanoma. Int J Cancer. 2000;85(5):618–26.
Trefzer U, et al. Tumour-dendritic hybrid cell vaccination for the treatment of patients with malignant melanoma: immunological effects and clinical results. Vaccine. 2005;23(17–18):2367–73.
Krause SW, et al. The treatment of patients with disseminated malignant melanoma by vaccination with autologous cell hybrids of tumor cells and dendritic cells. J Immunother. 2002;25(5):421–8.
Maeng HM, et al. Phase I clinical trial of an autologous dendritic cell vaccine against HER2 shows safety and preliminary clinical efficacy. Front Oncol. 2021;11: 789078.
Aspord C, et al. HLA-A* 0201+ plasmacytoid dendritic cells provide a cell-based immunotherapy for melanoma patients. J Invest Dermatol. 2012;132(10):2395–406.
Fabre JW. The allogeneic response and tumor immunity. Nat Med. 2001;7(6):649–52.
Bol KF, et al. The clinical application of cancer immunotherapy based on naturally circulating dendritic cells. J Immunother Cancer. 2019;7(1):1–3. This article discussed recent clinical developments and future prospects of natural DC-based immunotherapy.
Kikuchi T, et al. Vaccination of glioma patients with fusions of dendritic and glioma cells and recombinant human interleukin 12. J Immunother. 2004;27(6):452–9.
Homma S, et al. Cancer immunotherapy using dendritic/tumour-fusion vaccine induces elevation of serum anti-nuclear antibody with better clinical responses. Clin Exp Immunol. 2006;144(1):41–7.
Akasaki Y, et al. Antitumor effect of immunizations with fusions of dendritic and glioma cells in a mouse brain tumor model. J Immunother. 2001;24(2):106–13.
Haenssle HA, et al. Hybrid cell vaccination in metastatic melanoma: clinical and immunologic results of a phase I/II study. J Immunother. 2004;27(2):147–55.
Avigan DE, et al. Phase I/II study of vaccination with electrofused allogeneic dendritic cells/autologous tumor-derived cells in patients with stage IV renal cell carcinoma. J Immunother. 2007;30(7):749–61.
Vo MC, et al. Lenalidomide enhances the function of dendritic cells generated from patients with multiple myeloma. Exp Hematol. 2017;46:48–55.
Sakamaki I, et al. Lenalidomide enhances the protective effect of a therapeutic vaccine and reverses immune suppression in mice bearing established lymphomas. Leukemia. 2014;28(2):329–37.
Nahas MR, et al. Hypomethylating agent alters the immune microenvironment in acute myeloid leukaemia (AML) and enhances the immunogenicity of a dendritic cell/AML vaccine. Br J Haematol. 2019;185(4):679–90.
Rosenblatt J, et al. PD-1 blockade by CT-011, anti PD-1 antibody, enhances ex-vivo T cell responses to autologous dendritic/myeloma fusion vaccine. J Immunother. 2011;34(5):409.
Isazadeh A, et al. Resistance mechanisms to immune checkpoints blockade by monoclonal antibody drugs in cancer immunotherapy: focus on myeloma. J Cell Physiol. 2021;236(2):791–805.
Ghahremani Dehbokri S, et al. CTLA-4: as an immunosuppressive immune checkpoint in breast cancer. Cur Mol Med. 2023;23(6):521–6.
Nagle SJ, Garfall AL, Stadtmauer EA. The promise of chimeric antigen receptor engineered T cells in the treatment of hematologic malignancies. Cancer J. 2016;22(1):27.
Gu YZ, Zhao X, Song XR. Ex vivo pulsed dendritic cell vaccination against cancer. Acta Pharmacol Sin. 2020;41(7):959–69.
Anguille S, et al. Dendritic cells as pharmacological tools for cancer immunotherapy. Pharmacol Rev. 2015;67(4):731–53.
Tesfatsion DA. Dendritic cell vaccine against leukemia: advances and perspectives. Immunotherapy. 2014;6(4):485–96.
Rodríguez-Ruiz ME, et al. Combined immunotherapy encompassing intratumoral poly-ICLC, dendritic-cell vaccination and radiotherapy in advanced cancer patients. Ann Oncol. 2018;29(5):1312–9.
Amberger DC, Schmetzer HM. Dendritic cells of leukemic origin: specialized antigen-presenting cells as potential treatment tools for patients with myeloid leukemia. Transfus Med Hemother. 2020;47(6):432–43.
Fong L, et al. Dendritic cells injected via different routes induce immunity in cancer patients. J Immunol. 2001;166(6):4254–9.
Morse MA, 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. 1999;5(6):1331–8.
Barratt-Boyes SM, Watkins SC, Finn OJ. Migration of cultured chimpanzee dendritic cells following intravenous and subcutaneous injection. Adv Exp Med Biol. 1997;417:71–5.
Ando K, et al. Intravenous dendritic cell administration enhances suppression of lung metastasis induced by carbon-ion irradiation. J Radiat Res. 2017;58(4):446–55.
Gargett T, et al. Phase I trial of Lipovaxin-MM, a novel dendritic cell-targeted liposomal vaccine for malignant melanoma. Cancer Immunol Immunother. 2018;67(9):1461–72.
Schmitt A, Hus I, Schmitt M. Dendritic cell vaccines for leukemia patients. Expert Rev Anticancer Ther. 2007;7(3):275–83.
Gilliet M, et al. Intranodal injection of semimature monocyte-derived dendritic cells induces T helper type 1 responses to protein neoantigen. Blood. 2003;102(1):36–42.
Willekens B, et al. Tolerogenic dendritic cell-based treatment for multiple sclerosis (MS): a harmonised study protocol for two phase I clinical trials comparing intradermal and intranodal cell administration. BMJ Open. 2019;9(9): e030309.
Morisaki T, et al. Intranodal administration of neoantigen peptide-loaded dendritic cell vaccine elicits epitope-specific T cell responses and clinical effects in a patient with chemorefractory ovarian cancer with malignant ascites. Immunol Invest. 2021;50(5):562–79.
Castro JE, et al. Gene immunotherapy of chronic lymphocytic leukemia: a phase I study of intranodally injected adenovirus expressing a chimeric CD154 molecule. Cancer Res. 2012;72(12):2937–48.
Karthaus N, Torensma R, Tel J. Deciphering the message broadcast by tumor-infiltrating dendritic cells. Am J Pathol. 2012;181(3):733–42.
Isazadeh H, et al. Advances in siRNA delivery approaches in cancer therapy: challenges and opportunities. Mol Biol Rep. 2023:1–5.
Zhang Y, et al. Noncoding RNAs link metabolic reprogramming to immune microenvironment in cancers. J Hematol Oncol. 2021;14(1):1–26.
Liu Y, Zhou X, Wang X. Targeting the tumor microenvironment in B-cell lymphoma: challenges and opportunities. J Hematol Oncol. 2021;14(1):1–7.
Cranmer LD, Trevor KT, Hersh EM. Clinical applications of dendritic cell vaccination in the treatment of cancer. Cancer Immunol Immunother. 2004;53:275–306.
Cui Y, et al. Immune response, clinical outcome and safety of dendritic cell vaccine in combination with cytokine-induced killer cell therapy in cancer patients. Oncol Lett. 2013;6(2):537–41.
Amos SM, et al. Autoimmunity associated with immunotherapy of cancer. Blood, The Journal of the American Society of Hematology. 2011;118(3):499–509.
Leonhartsberger N, et al. Quality of life during dendritic cell vaccination against metastatic renal cell carcinoma. Cancer Immunol Immunother. 2012;61:1407–13.
Madan RA, et al. Therapeutic cancer vaccines in prostate cancer: the paradox of improved survival without changes in time to progression. Oncologist. 2010;15(9):969–75.
Aarntzen EH, et al. Targeting CD4+ T-helper cells improves the induction of antitumor responses in dendritic cell–based vaccination. Cancer Res. 2013;73(1):19–29.
Tel J, et al. Natural human plasmacytoid dendritic cells induce antigen-specific T-cell responses in melanoma patients. Cancer Res. 2013;73(3):1063–75.
Roddie H, et al. Phase I/II study of vaccination with dendritic-like leukaemia cells for the immunotherapy of acute myeloid leukaemia. Br J Haematol. 2006;133(2):152–7.
Ji YS, Park SK, Ryu S. Whole leukemia cell vaccines: past progress and future directions. Vaccine. 2020;38(22):3811–20.
Zhao W, Zhao G, Wang B. Revisiting GM-CSF as an adjuvant for therapeutic vaccines. Cell Mol Immunol. 2018;15(2):187–9.
Bondanza A, et al. Inhibition of phosphatidylserine recognition heightens the immunogenicity of irradiated lymphoma cells in vivo. J Exp Med. 2004;200(9):1157–65.
O’Brien LJ, Guillerey C, Radford KJ. Can dendritic cell vaccination prevent leukemia relapse? Cancers. 2019;11(6):875.
Garg AD, Coulie PG, Van den Eynde BJ, Agostinis P. Integrating next-generation dendritic cell vaccines into the current cancer immunotherapy landscape. Trends Immunol. 2017;38(8):577–93.
van Beek JJ, et al. Human blood myeloid and plasmacytoid dendritic cells cross activate each other and synergize in inducing NK cell cytotoxicity. Oncoimmunology. 2016;5(10): e1227902.
Van Acker HH, et al. Desirable cytolytic immune effector cell recruitment by interleukin-15 dendritic cells. Oncotarget. 2017;8(8):13652.
Van Ee TJ, et al. BDCA1+ CD14+ immunosuppressive cells in cancer, a potential target? Vaccines. 2018;6(3):65.
Curti A, et al. Indoleamine 2, 3-dioxygenase-expressing leukemic dendritic cells impair a leukemia-specific immune response by inducing potent T regulatory cells. Haematologica. 2010;95(12):2022.
Draube A, et al. Dendritic cell based tumor vaccination in prostate and renal cell cancer: a systematic review and meta-analysis. PLoS ONE. 2011;6(4): e18801.
Carreno BM, et al. IL-12p70–producing patient DC vaccine elicits Tc1-polarized immunity. J Clin Invest. 2013;123(8):3383–94.
Breton G, et al. Circulating precursors of human CD1c+ and CD141+ dendritic cells. J Exp Med. 2015;212(3):401–13.
Bennaceur K, et al. Dendritic cells dysfunction in tumour environment. Cancer Lett. 2008;272(2):186–96.
Bronte V, Mocellin S. Suppressive influences in the immune response to cancer. J Immunother. 2009;32(1):1–1.
Author information
Authors and Affiliations
Contributions
S.T. conceived and directed the project, contributed to the content discussion, and reviewed and edited the article. P.L. drafted the article, and L.J. and X.B. reviewed and edited the article and contributed to the content discussion.
Corresponding author
Ethics declarations
Conflict of Interest
The authors declare no competing interests.
Human and Animal Rights and Informed Consent
This article does not contain any studies with human or animal subjects performed by any of the authors.
Additional information
Publisher's Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Li, P., Jia, L., Bian, X. et al. Application of Engineered Dendritic Cell Vaccines in Cancer Immunotherapy: Challenges and Opportunities. Curr. Treat. Options in Oncol. 24, 1703–1719 (2023). https://doi.org/10.1007/s11864-023-01143-7
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1007/s11864-023-01143-7