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.

  • Review Article
  • Published:

Dendritic-cell immunotherapy: from ex vivo loading to in vivo targeting

Key Points

  • Dendritic cells (DCs) are key regulators of innate and adaptive immune responses. They are used in clinical trials to induce immune responses directed against specific antigens.

  • Clinical trials exploring DC-based vaccines mostly involve autologous DCs that are cultured and loaded with antigens ex vivo, which is a costly procedure. Targeting antigens to DC surface receptors in vivo bypasses the necessity for ex vivo DC culturing, allowing large-scale application of DC-based vaccination therapy.

  • Preclinical mouse studies for targeted delivery of antigens to DCs in vivo show successful induction of humoral and cellular responses. Targeting tumour antigens to DC surface receptors has been shown to protect mice from growing tumours and even cure them of existing tumours.

  • Strategies aimed at inducing immunity require means to mature the targeted DCs Without a proper maturation stimulus, targeted delivery of antigen might result in the induction of tolerance.

  • The specific receptor that is targeted may affect the quality of the immune response owing to differences in intracellular receptor routing, signalling pathways and expression patterns. For instance, antigens delivered to receptors preferentially expressed by the mouse CD8+ DC subset predominantly induce cellular responses, whereas antigens delivered to CD8 DC subset mainly induce humoral responses.

  • Advancing knowledge on the intracellular routing of antigen allows for the design of more potent vaccines. The use of powerful DC maturation stimuli, agents that facilitate antigen to escape from endosomes or means to prevent rapid antigen degradation have been shown to enhance humoral responses, cellular responses, or both.

Abstract

The realization that dendritic cells (DCs) orchestrate innate and adaptive immune responses has stimulated research on harnessing DCs to create more effective vaccines. Early clinical trials exploring autologous DCs that were loaded with antigens ex vivo to induce T-cell responses have provided proof of principle. Here, we discuss how direct targeting of antigens to DC surface receptors in vivo might replace laborious and expensive ex vivo culturing, and facilitate large-scale application of DC-based vaccination therapies.

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

Access options

Buy this article

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

Figure 1: Intracellular fate of antigens targeted to DC surface receptors.
Figure 2: Vaccines delivering MHC class I antigens.

Similar content being viewed by others

References

  1. Banchereau, J. & Steinman, R. M. Dendritic cells and the control of immunity. Nature 392, 245–252 (1998).

    Article  CAS  PubMed  Google Scholar 

  2. Celis, E. & Chang, T. W. Antibodies to hepatitis B surface antigen potentiate the response of human T lymphocyte clones to the same antigen. Science 224, 297–299 (1984).

    CAS  PubMed  Google Scholar 

  3. Chang, T. W. Regulation of immune response by antibodies: the importance of antibody and monocyte Fc receptor interaction in T-cell activation. Immunol. Today 6, 245–249 (1985).

    CAS  PubMed  Google Scholar 

  4. Snider, D. P. & Segal, D. M. Targeted antigen presentation using crosslinked antibody heteroaggregates. J. Immunol. 139, 1609–1616 (1987).

    CAS  PubMed  Google Scholar 

  5. Carayanniotis, G. & Barber, B. H. Adjuvant-free IgG responses induced with antigen coupled to antibodies against class II MHC. Nature 327, 59–61 (1987).

    CAS  PubMed  Google Scholar 

  6. Snider, D. P., Kaubisch, A. & Segal, D. M. Enhanced antigen immunogenicity induced by bispecific antibodies. J. Exp. Med. 171, 1957–1963 (1990).

    CAS  PubMed  Google Scholar 

  7. Figdor, C. G., van Kooyk, Y. & Adema, G. J. C-type lectin receptors on dendritic cells and Langerhans cells. Nature Rev. Immunol. 2, 77–84 (2002).

    CAS  Google Scholar 

  8. Keler, T., Ramakrishna, V. & Fanger, M. W. Mannose receptor-targeted vaccines. Expert Opin. Biol. Ther. 4, 1953–1962 (2004).

    CAS  PubMed  Google Scholar 

  9. Karanikas, V. et al. Antibody and T cell responses of patients with adenocarcinoma immunized with mannan-MUC1 fusion protein. J. Clin. Invest. 100, 2783–2792 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Apostolopoulos, V. et al. Pilot phase III immunotherapy study in early-stage breast cancer patients using oxidized mannan-MUC1. Breast Cancer Res. 8, R27 (2006).

    PubMed  PubMed Central  Google Scholar 

  11. He, L. Z. et al. A novel human cancer vaccine elicits cellular responses to the tumor-associated antigen, human chorionic gonadotropin β. Clin. Cancer Res. 10, 1920–1927 (2004).

    CAS  PubMed  Google Scholar 

  12. Ramakrishna, V. et al. Mannose receptor targeting of tumor antigen pmel17 to human dendritic cells directs anti-melanoma T cell responses via multiple HLA molecules. J. Immunol. 172, 2845–2852 (2004).

    CAS  PubMed  Google Scholar 

  13. He, L. Z. et al. Antigenic targeting of the human mannose receptor induces tumor immunity. J. Immunol. 178, 6259–6267 (2007).

    CAS  PubMed  Google Scholar 

  14. Tarlinton, D. & Lew, A. Antigen to the node: B cells go native. Immunity 26, 388–390 (2007).

    CAS  PubMed  Google Scholar 

  15. Mahnke, K. et al. The dendritic cell receptor for endocytosis, DEC-205, can recycle and enhance antigen presentation via major histocompatibility complex class II-positive lysosomal compartments. J. Cell Biol. 151, 673–684 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Inaba, K. et al. Tissue distribution of the DEC-205 protein that is detected by the monoclonal antibody NLDC-145. I. Expression on dendritic cells and other subsets of mouse leukocytes. Cell. Immunol. 163, 148–156 (1995).

    CAS  PubMed  Google Scholar 

  17. Bonifaz, L. et al. Efficient targeting of protein antigen to the dendritic cell receptor DEC-205 in the steady state leads to antigen presentation on major histocompatibility complex class I products and peripheral CD8+ T cell tolerance. J. Exp. Med. 196, 1627–1638 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Hawiger, D. et al. Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivo. J. Exp. Med. 194, 769–779 (2001). References 17 and 18 show that targeting antigen to DCs without providing a maturation stimulus induces tolerance.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Mahnke, K., Qian, Y. J., Knop, J. & Enk, A. H. Induction of CD4+/CD25+ regulatory T cells by targeting of antigens to immature dendritic cells. Blood 101, 4862–4869 (2003).

    CAS  PubMed  Google Scholar 

  20. Bonifaz, L. C. et al. In vivo targeting of antigens to maturing dendritic cells via the DEC-205 receptor improves T cell vaccination. J. Exp. Med. 199, 815–824 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. van Broekhoven, C. L., Parish, C. R., Demangel, C., Britton, W. J. & Altin, J. G. Targeting dendritic cells with antigen-containing liposomes: a highly effective procedure for induction of antitumor immunity and for tumor immunotherapy. Cancer Res. 64, 4357–4365 (2004).

    CAS  PubMed  Google Scholar 

  22. Boscardin, S. B. et al. Antigen targeting to dendritic cells elicits long-lived T cell help for antibody responses. J. Exp. Med. 203, 599–606 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Mahnke, K. et al. Targeting of antigens to activated dendritic cells in vivo cures metastatic melanoma in mice. Cancer Res. 65, 7007–7012 (2005).

    CAS  PubMed  Google Scholar 

  24. Bozzacco, L. et al. DEC-205 receptor on dendritic cells mediates presentation of HIV gag protein to CD8+ T cells in a spectrum of human MHC I haplotypes. Proc. Natl Acad. Sci. USA 104, 1289–1294 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Kato, M. et al. Expression of human DEC-205 (CD205) multilectin receptor on leukocytes. Int. Immunol. 18, 857–869 (2006).

    CAS  PubMed  Google Scholar 

  26. Geijtenbeek, T. B. H. et al. Identification of DC-SIGN, a novel dendritic cell-specific ICAM-3 receptor that supports primary immune responses. Cell 100, 575–585 (2000).

    CAS  PubMed  Google Scholar 

  27. Soilleux, E. J. et al. Constitutive and induced expression of DC-SIGN on dendritic cell and macrophage subpopulations in situ and in vitro. J. Leukoc. Biol. 71, 445–457 (2002).

    CAS  PubMed  Google Scholar 

  28. Gramberg, T., Caminschi, I., Wegele, A., Hofmann, H. & Pohlmann, S. Evidence that multiple defects in murine DC-SIGN inhibit a functional interaction with pathogens. Virology 345, 482–491 (2005).

    PubMed  Google Scholar 

  29. Caminschi, I. et al. Functional comparison of mouse CIRE/mouse DC-SIGN and human DC-SIGN. Int. Immunol. 18, 741–753 (2006).

    CAS  PubMed  Google Scholar 

  30. Tacken, P. J. et al. Effective induction of naive and recall T cell responses by targeting antigen to human dendritic cells via a humanized anti-DC-SIGN antibody. Blood 106, 1278–1285 (2005).

    CAS  PubMed  Google Scholar 

  31. Granelli-Piperno, A. et al. Dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin/CD209 is abundant on macrophages in the normal human lymph node and is not required for dendritic cell stimulation of the mixed leukocyte reaction. J. Immunol. 175, 4265–4273 (2005).

    CAS  PubMed  Google Scholar 

  32. Shortman, K. & Liu, Y. J. Mouse and human dendritic cell subtypes. Nature Rev. Immunol. 2, 151–161 (2002).

    CAS  Google Scholar 

  33. den Haan, J. M. M., Lehar, S. M. & Bevan, M. J. CD8+ but not CD8 dendritic cells cross-prime cytotoxic T cells in vivo. J. Exp. Med. 192, 1685–1696 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Dudziak, D. et al. Differential antigen processing by dendritic cell subsets in vivo. Science 315, 107–111 (2007).

    CAS  PubMed  Google Scholar 

  35. Caminschi, I. et al. Molecular cloning of F4/80-like-receptor, a seven-span membrane protein expressed differentially by dendritic cell and monocyte-macrophage subpopulations. J. Immunol. 167, 3570–3576 (2001).

    CAS  PubMed  Google Scholar 

  36. Caminschi, I. et al. Molecular cloning of a C-type lectin superfamily protein differentially expressed by CD8α splenic dendritic cells. Mol. Immunol. 38, 365–373 (2001).

    CAS  PubMed  Google Scholar 

  37. Vremec, D. & Shortman, K. Dendritic cell subtypes in mouse lymphoid organs: cross-correlation of surface markers, changes with incubation, and differences among thymus, spleen, and lymph nodes. J. Immunol. 159, 565–573 (1997).

    CAS  PubMed  Google Scholar 

  38. McLellan, A. D. et al. Anatomic location and T-cell stimulatory functions of mouse dendritic cell subsets defined by CD4 and CD8 expression. Blood 99, 2084–2093 (2002).

    CAS  PubMed  Google Scholar 

  39. Corbett, A. J. et al. Antigen delivery via two molecules on the CD8 dendritic cell subset induces humoral immunity in the absence of conventional 'danger'. Eur. J Immunol. 35, 2815–2825 (2005).

    CAS  PubMed  Google Scholar 

  40. Carter, R. W., Thompson, C., Reid, D. M., Wong, S. Y. C. & Tough, D. F. Preferential induction of CD4+ T cell responses through in vivo targeting of antigen to dendritic cell-associated C-type lectin-1. J. Immunol. 177, 2276–2284 (2006). Together with references 34 and 39 , this study demonstrates that the targeting of receptors preferentially expressed on two different DC subsets can result in disparate immune responses.

    CAS  PubMed  Google Scholar 

  41. Dakappagari, N. et al. Internalizing antibodies to the C-type lectins, L-SIGN and DC-SIGN, inhibit viral glycoprotein binding and deliver antigen to human dendritic cells for the induction of T cell responses. J. Immunol. 176, 426–440 (2006).

    CAS  PubMed  Google Scholar 

  42. Pasquier, B. et al. Identification of FcαRI as an inhibitory receptor that controls inflammation: dual role of FcRγ ITAM. Immunity 22, 31–42 (2005).

    CAS  PubMed  Google Scholar 

  43. Herre, J. et al. Dectin-1 uses novel mechanisms for yeast phagocytosis in macrophages. Blood 104, 4038–4045 (2004).

    CAS  PubMed  Google Scholar 

  44. Woof, J. M. & Burton, D. R. Human antibody-Fc receptor interactions illuminated by crystal structures. Nature Rev. Immunol. 4, 89–99 (2004).

    CAS  Google Scholar 

  45. Nimmerjahn, F. & Ravetch, J. V. Fcγ receptors: old friends and new family members. Immunity 24, 19–28 (2006).

    CAS  PubMed  Google Scholar 

  46. Kanazawa, N. Dendritic cell immunoreceptors: C-type lectin receptors for pattern-recognition and signaling on antigen-presenting cells. J. Dermatol. Sci. 45, 77–86 (2007).

    CAS  PubMed  Google Scholar 

  47. Underhill, D. M., Rossnagle, E., Lowell, C. A. & Simmons, R. M. Dectin-1 activates Syk tyrosine kinase in a dynamic subset of macrophages for reactive oxygen production. Blood 106, 2543–2550 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Rogers, N. C. et al. Syk-dependent cytokine induction by Dectin-1 reveals a novel pattern recognition pathway for C type lectins. Immunity 22, 507–517 (2005).

    CAS  PubMed  Google Scholar 

  49. Gantner, B. N., Simmons, R. M., Canavera, S. J., Akira, S. & Underhill, D. M. Collaborative induction of inflammatory responses by Dectin-1 and Toll-like receptor 2. J. Exp. Med. 197, 1107–1117 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Geijtenbeek, T. B. H. et al. Mycobacteria target DC-SIGN to suppress dendritic cell function. J. Exp. Med. 197, 7–17 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Dzionek, A. et al. BDCA-2, a novel plasmacytoid dendritic cell-specific type II C-type lectin, mediates antigen capture and is a potent inhibitor of interferon α/β induction. J. Exp. Med. 194, 1823–1834 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. de Vries, I. J. et al. Maturation of dendritic cells is a prerequisite for inducing immune responses in advanced melanoma patients. Clin. Cancer Res. 9, 5091–5100 (2003).

    CAS  PubMed  Google Scholar 

  53. Dhodapkar, M. V., Steinman, R. M., Krasovsky, J., Munz, C. & Bhardwaj, N. Antigen-specific inhibition of effector T cell function in humans after injection of immature dendritic cells. J. Exp. Med. 193, 233–238 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Fujii, S. i., Shimizu, K., Smith, C., Bonifaz, L. & Steinman, R. M. Activation of natural killer T cells by α-Galactosylceramide rapidly induces the full maturation of dendritic cells in vivo and thereby acts as an adjuvant for combined CD4 and CD8 T cell immunity to a coadministered protein. J. Exp. Med. 198, 267–279 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. van Duin, D., Medzhitov, R. & Shaw, A. C. Triggering TLR signaling in vaccination. Trends Immunol. 27, 49–55 (2006).

    CAS  PubMed  Google Scholar 

  56. Datta, S. K. et al. A subset of Toll-like receptor ligands induces cross-presentation by bone marrow-derived dendritic cells. J. Immunol. 170, 4102–4110 (2003).

    CAS  PubMed  Google Scholar 

  57. Wilson, N. S. et al. Systemic activation of dendritic cells by Toll-like receptor ligands or malaria infection impairs cross-presentation and antiviral immunity. Nature Immunol. 7, 165–172 (2006). This paper shows that systemic exposure to TLR ligands prior to vaccination downmodulates cross-presentation.

    CAS  Google Scholar 

  58. Butler, M. et al. Altered expression and endocytic function of CD205 in human dendritic cells, and detection of a CD205–DCL-1 fusion protein upon dendritic cell maturation. Immunology 120, 362–371 (2006).

    PubMed  Google Scholar 

  59. Jackson, D. C. et al. A totally synthetic vaccine of generic structure that targets Toll-like receptor 2 on dendritic cells and promotes antibody or cytotoxic T cell responses. Proc. Natl Acad. Sci. USA 101, 15440–15445 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Zhang, L. et al. An adenoviral vector cancer vaccine that delivers a tumor-associated antigen/CD40-ligand fusion protein to dendritic cells. Proc. Natl Acad. Sci. USA 100, 15101–15106 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Delneste, Y. et al. Involvement of LOX-1 in dendritic cell-mediated antigen cross-presentation. Immunity 17, 353–362 (2002).

    CAS  PubMed  Google Scholar 

  62. Wille-Reece, U., Wu, C. y., Flynn, B. J., Kedl, R. M. & Seder, R. A. Immunization with HIV-1 Gag protein conjugated to a TLR7/8 agonist results in the generation of HIV-1 Gag-specific Th1 and CD8+ T cell responses. J. Immunol. 174, 7676–7683 (2005).

    CAS  PubMed  Google Scholar 

  63. Cho, H. J. et al. Immunostimulatory DNA-based vaccines induce cytotoxic lymphocyte activity by a T-helper cell-independent mechanism. Nature Biotech. 18, 509–514 (2000).

    CAS  Google Scholar 

  64. Yarovinsky, F., Kanzler, H., Hieny, S., Coffman, R. L. & Sher, A. Toll-like receptor recognition regulates immunodominance in an antimicrobial CD4+ T cell response. Immunity 25, 655–664 (2006).

    CAS  PubMed  Google Scholar 

  65. Blander, J. M. & Medzhitov, R. Toll-dependent selection of microbial antigens for presentation by dendritic cells. Nature 440, 808–812 (2006). An elegant study demonstrating that TLR ligands need to be physically linked to the antigen to be efficiently presented by MHC class II.

    CAS  PubMed  Google Scholar 

  66. Napolitani, G., Rinaldi, A., Bertoni, F., Sallusto, F. & Lanzavecchia, A. Selected Toll-like receptor agonist combinations synergistically trigger a T helper type 1-polarizing program in dendritic cells. Nature Immunol. 6, 769–776 (2005).

    CAS  Google Scholar 

  67. Ramakrishna, V. et al. Toll-like receptor activation enhances cell-mediated immunity induced by an antibody vaccine targeting human dendritic cells. J. Transl. Med. 5, 5 (2007).

    PubMed  PubMed Central  Google Scholar 

  68. Burgdorf, S., Kautz, A., Bohnert, V., Knolle, P. A. & Kurts, C. Distinct pathways of antigen uptake and intracellular routing in CD4 and CD8 T cell activation. Science 316, 612–616 (2007).

    CAS  PubMed  Google Scholar 

  69. Carbone, F. R. & Bevan, M. J. Class I-restricted processing and presentation of exogenous cell-associated antigen in vivo. J. Exp. Med. 171, 377–387 (1990).

    CAS  PubMed  Google Scholar 

  70. Reis e Sousa, C. & Germain, R. N. Major histocompatibility complex class I presentation of peptides derived from soluble exogenous antigen by a subset of cells engaged in phagocytosis. J. Exp. Med. 182, 841–851 (1995).

    CAS  PubMed  Google Scholar 

  71. Brossart, P. & Bevan, M. J. Presentation of exogenous protein antigens on major histocompatability complex class I molecules by dendritic cells: pathway of presentation and regulation by cytokines. Blood 90, 1594–1599 (1997).

    CAS  PubMed  Google Scholar 

  72. Guermonprez, P. et al. ER-phagosome fusion defines an MHC class I cross-presentation compartment in dendritic cells. Nature 425, 397–402 (2003).

    CAS  PubMed  Google Scholar 

  73. Ackerman, A. L., Kyritsis, C., Tampe, R. & Cresswell, P. Access of soluble antigens to the endoplasmic reticulum can explain cross-presentation by dendritic cells. Nature Immunol. 6, 107–113 (2005).

    CAS  Google Scholar 

  74. Yewdell, J. W., Bennink, J. R. & Hosaka, Y. Cells process exogenous proteins for recognition by cytotoxic T lymphocytes. Science 239, 637–640 (1988).

    CAS  PubMed  Google Scholar 

  75. Moore, M. W., Carbone, F. R. & Bevan, M. J. Introduction of soluble protein into the class I pathway of antigen processing and presentation. Cell 54, 777–785 (1988).

    CAS  PubMed  Google Scholar 

  76. Melikov, K. & Chernomordik, L. V. Arginine-rich cell penetrating peptides: from endosomal uptake to nuclear delivery. Cell. Mol. Life Sci. 62, 2739–2749 (2005).

    CAS  PubMed  Google Scholar 

  77. Batchu, R. B. et al. Protein transduction of dendritic cells for NY-ESO-1-based immunotherapy of myeloma. Cancer Res. 65, 10041–10049 (2005).

    CAS  PubMed  Google Scholar 

  78. Kim, D. T. et al. Introduction of soluble proteins into the MHC class I pathway by conjugation to an HIV tat peptide. J. Immunol. 159, 1666–1668 (1997).

    CAS  PubMed  Google Scholar 

  79. Shibagaki, N. & Udey, M. C. Dendritic cells transduced with protein antigens induce cytotoxic lymphocytes and elicit antitumor immunity. J. Immunol. 168, 2393–2401 (2002).

    CAS  PubMed  Google Scholar 

  80. Kielian, M. & Rey, F. A. Virus membrane-fusion proteins: more than one way to make a hairpin. Nature Rev. Microbiol. 4, 67–76 (2006).

    CAS  Google Scholar 

  81. Mastrobattista, E. et al. Functional characterization of an endosome-disruptive peptide and its application in cytosolic delivery of immunoliposome-entrapped proteins. J. Biol. Chem. 277, 27135–27143 (2002).

    CAS  PubMed  Google Scholar 

  82. Plank, C., Oberhauser, B., Mechtler, K., Koch, C. & Wagner, E. The influence of endosome-disruptive peptides on gene transfer using synthetic virus-like gene transfer systems. J. Biol. Chem. 269, 12918–12924 (1994).

    CAS  PubMed  Google Scholar 

  83. Wadia, J. S., Stan, R. V. & Dowdy, S. F. Transducible TAT-HA fusogenic peptide enhances escape of TAT-fusion proteins after lipid raft macropinocytosis. Nature Med. 10, 310–315 (2004).

    CAS  PubMed  Google Scholar 

  84. Laus, R., Graddis, T. J., Hakim, I. & Vidovic, D. Enhanced major histocompatibility complex class I-dependent presentation of antigens modified with cationic and fusogenic peptides. Nature Biotechnol. 18, 1269–1272 (2000).

    CAS  Google Scholar 

  85. Schwarze, S. R., Ho, A., Vocero-Akbani, A. & Dowdy, S. F. In vivo protein transduction: delivery of a biologically active protein into the mouse. Science 285, 1569–1572 (1999).

    CAS  PubMed  Google Scholar 

  86. Lisziewicz, J. et al. DermaVir: a novel topical vaccine for HIV/AIDS. J. Invest. Dermatol. 124, 160–169 (2005).

    CAS  PubMed  Google Scholar 

  87. Lisziewicz, J. et al. Control of viral rebound through therapeutic immunization with DermaVir. AIDS 19, 35–43 (2005).

    PubMed  Google Scholar 

  88. O'Connor, T. P. & Crystal, R. G. Genetic medicines: treatment strategies for hereditary disorders. Nature Rev. Genet. 7, 261–276 (2006).

    CAS  PubMed  Google Scholar 

  89. Walsh, G. Biopharmaceutical benchmarks 2006. Nature Biotechnol. 24, 769–776 (2006).

    CAS  Google Scholar 

  90. Tillman, B. W. et al. Maturation of dendritic cells accompanies high-efficiency gene transfer by a CD40-targeted adenoviral vector. J. Immunol. 162, 6378–6383 (1999).

    CAS  PubMed  Google Scholar 

  91. Korokhov, N. et al. High efficiency transduction of dendritic cells by adenoviral vectors targeted to DC-SIGN. Cancer Biol. Ther. 4, 289–294 (2005).

    CAS  PubMed  Google Scholar 

  92. Hedley, S. J. et al. An adenovirus vector with a chimeric fiber incorporating stabilized single chain antibody achieves targeted gene delivery. Gene Ther. 13, 88–94 (2005).

    Google Scholar 

  93. Lennon-Dumenil, A. M. et al. Analysis of protease activity in live antigen-presenting cells shows regulation of the phagosomal proteolytic contents during dendritic cell activation. J. Exp. Med. 196, 529–540 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Delamarre, L., Pack, M., Chang, H., Mellman, I. & Trombetta, E. S. Differential lysosomal proteolysis in antigen-presenting cells determines antigen fate. Science 307, 1630–1634 (2005).

    CAS  PubMed  Google Scholar 

  95. Delamarre, L., Couture, R., Mellman, I. & Trombetta, E. S. Enhancing immunogenicity by limiting susceptibility to lysosomal proteolysis. J. Exp. Med. 203, 2049–2055 (2006). This report indicates that the immunogenicity of antigens can be enhanced by preventing rapid degradation.

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Waeckerle-Men, Y. & Groettrup, M. PLGA microspheres for improved antigen delivery to dendritic cells as cellular vaccines. Adv. Drug Deliv. Rev. 57, 475–482 (2005).

    CAS  PubMed  Google Scholar 

  97. Savina, A. et al. NOX2 controls phagosomal pH to regulate antigen processing during crosspresentation by dendritic cells. Cell 126, 205–218 (2006).

    CAS  PubMed  Google Scholar 

  98. Accapezzato, D. et al. Chloroquine enhances human CD8+ T cell responses against soluble antigens in vivo. J. Exp. Med. 202, 817–828 (2005). This study shows that the cross-presentation of antigens is enhanced by a relatively simple trick: preventing endosomal acidification by co-administering the antimalarial drug chloroquine.

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Kloetzel, P. M. Generation of major histocompatibility complex class I antigens: functional interplay between proteasomes and TPPII. Nature Immunol. 5, 661–669 (2004).

    CAS  Google Scholar 

  100. Toes, R. E. M. et al. Discrete cleavage motifs of constitutive and immunoproteasomes revealed by quantitative analysis of cleavage products. J. Exp. Med. 194, 1–12 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Gaczynska, M., Rock, K. L. & Goldberg, A. L. γ-Interferon and expression of MHC genes regulate peptide hydrolysis by proteasomes. Nature 365, 264–267 (1993).

    CAS  PubMed  Google Scholar 

  102. Van Kaer, L. et al. Altered peptidase and viral-specific T cell response in LMP2 mutant mice. Immunity 1, 533–541 (1994).

    CAS  PubMed  Google Scholar 

  103. Basler, M., Moebius, J., Elenich, L., Groettrup, M. & Monaco, J. J. An altered T cell repertoire in MECL-1-deficient mice. J. Immunol. 176, 6665–6672 (2006).

    CAS  PubMed  Google Scholar 

  104. Chapatte, L. et al. Processing of tumor-associated antigen by the proteasomes of dendritic cells controls in vivo T-cell responses. Cancer Res. 66, 5461–5468 (2006). An elegant example of how T cells recognizing peptide epitopes that are not generated by the immunoproteasome can be primed by DCs expressing the peptide, but not the protein, antigen.

    CAS  PubMed  Google Scholar 

  105. Curiel, T. J. Tregs and rethinking cancer immunotherapy. J. Clin. Invest. 117, 1167–1174 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  106. Ratzinger, G. et al. Mature human Langerhans cells derived from CD34+ hematopoietic progenitors stimulate greater cytolytic T lymphocyte activity in the absence of bioactive IL-12p70, by either single peptide presentation or cross-priming, than do dermal-interstitial or monocyte-derived dendritic cells. J. Immunol. 173, 2780–2791 (2004).

    CAS  PubMed  Google Scholar 

  107. Benitez-Ribas, D. et al. Plasmacytoid dendritic cells of melanoma patients present exogenous proteins to CD4+ T cells after FcγRII-mediated uptake. J. Exp. Med. 203, 1629–1635 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Zhang, J. et al. Characterization of Siglec-H as a novel endocytic receptor expressed on murine plasmacytoid dendritic cell precursors. Blood 107, 3600–3608 (2006).

    CAS  PubMed  Google Scholar 

  109. Hsu, F. J. et al. Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells. Nature Med. 2, 52–58 (1996).

    CAS  PubMed  Google Scholar 

  110. Adema, G. J., de Vries, I. J., Punt, C. J. & Figdor, C. G. Migration of dendritic cell based cancer vaccines: in vivo veritas? Curr. Opin. Immunol. 17, 170–174 (2005).

    CAS  PubMed  Google Scholar 

  111. Mignot, G., Roux, S., Thery, C., Segura, E. & Zitvogel, L. Prospects for exosomes in immunotherapy of cancer. J. Cell. Mol. Med. 10, 376–388 (2006).

    CAS  PubMed  Google Scholar 

  112. Engering, A. et al. The dendritic cell-specific adhesion receptor DC-SIGN internalizes antigen for presentation to T cells. J. Immunol. 168, 2118–2126 (2002).

    CAS  PubMed  Google Scholar 

  113. Heijnen, I. A. et al. Antigen targeting to myeloid-specific human FcγRI/CD64 triggers enhanced antibody responses in transgenic mice. J. Clin. Invest. 97, 331–338 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Liu, Y. et al. Regulated expression of FcγR in human dendritic cells controls cross-presentation of antigen-antibody complexes. J. Immunol. 177, 8440–8447 (2006).

    CAS  PubMed  Google Scholar 

  115. Mende, I. et al. Highly efficient antigen targeting to M-DC8+ dendritic cells via FcγRIII/CD16-specific antibody conjugates. Int. Immunol. 17, 539–547 (2005).

    CAS  PubMed  Google Scholar 

  116. Otten, M. A., Groenveld, I., van De Winkel, J. G. & van Egmond, M. Inefficient antigen presentation via the IgA Fc receptor (FcαRI) on dendritic cells. Immunobiol. 211, 503–510 (2006).

    CAS  Google Scholar 

  117. Fayolle, C., Sebo, P., Ladant, D., Ullmann, A. & Leclerc, C. In vivo induction of CTL responses by recombinant adenylate cyclase of Bordetella pertussis carrying viral CD8+ T cell epitopes. J. Immunol. 156, 4697–4706 (1996).

    CAS  PubMed  Google Scholar 

  118. Fayolle, C. et al. Delivery of multiple epitopes by recombinant detoxified adenylate cyclase of Bordetella pertussis induces protective antiviral immunity. J. Virol. 75, 7330–7338 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  119. Saron, M. F. et al. Anti-viral protection conferred by recombinant adenylate cyclase toxins from Bordetella pertussis carrying a CD8+ T cell epitope from lymphocytic choriomeningitis virus. Proc. Natl Acad. Sci. USA 94, 3314–3319 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  120. Sebo, P. et al. In vivo induction of CTL responses by recombinant adenylate cyclase of Bordetella pertussis carrying multiple copies of a viral CD8+ T-cell epitope. FEMS Immunol. Med. Microbiol. 26, 167–173 (1999).

    CAS  PubMed  Google Scholar 

  121. Romani, N. et al. Proliferating dendritic cell progenitors in human blood. J. Exp. Med. 180, 83–93 (1994).

    CAS  PubMed  Google Scholar 

  122. Nestle, F. O. et al. Vaccination of melanoma patients with peptide- or tumorlysate-pulsed dendritic cells. Nature Med. 4, 328–332 (1998).

    CAS  PubMed  Google Scholar 

  123. Holtl, L. et al. Cellular and humoral immune responses in patients with metastatic renal cell carcinoma after vaccination with antigen pulsed dendritic cells. J. Urol. 161, 777–782 (1999).

    CAS  PubMed  Google Scholar 

  124. Holtl, L. et al. Immunotherapy of metastatic renal cell carcinoma with tumor lysate-pulsed autologous dendritic cells. Clin. Cancer Res. 8, 3369–3376 (2002).

    CAS  PubMed  Google Scholar 

  125. Thurner, B. et al. Vaccination with Mage-3A1 peptide-pulsed mature, monocyte-derived dendritic cells expands specific cytotoxic T cells and induces regression of some metastases in advanced stage IV melanoma. J. Exp. Med. 190, 1669–1678 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Dhodapkar, M. V. et al. Rapid generation of broad T-cell immunity in humans after a single injection of mature dendritic cells. J. Clin. Invest. 104, 173–180 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  127. Mackensen, A. et al. Phase I study in melanoma patients of a vaccine with peptide-pulsed dendritic cells generated in vitro from CD34+ hematopoietic progenitor cells. Int. J Cancer 86, 385–392 (2000).

    CAS  PubMed  Google Scholar 

  128. Banchereau, J. et al. Immune and clinical responses in patients with metastatic melanoma to CD34+ progenitor-derived dendritic cell vaccine. Cancer Res. 61, 6451–6458 (2001).

    CAS  PubMed  Google Scholar 

  129. Fong, L. et al. Altered peptide ligand vaccination with Flt3 ligand expanded dendritic cells for tumor immunotherapy. Proc. Natl Acad. Sci. USA 98, 8809–8814 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  130. 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 

  131. Mullins, D. W. et al. Route of immunization with peptide-pulsed dendritic cells controls the distribution of memory and effector T cells in lymphoid tissues and determines the pattern of regional tumor control. J. Exp. Med. 198, 1023–1034 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  132. Dhodapkar, M. V. & Steinman, R. M. Antigen-bearing immature dendritic cells induce peptide-specific CD8+ regulatory T cells in vivo in humans. Blood 100, 174–177 (2002).

    CAS  PubMed  Google Scholar 

  133. Jonuleit, H. et al. A comparison of two types of dendritic cell as adjuvants for the induction of melanoma-specific T-cell responses in humans following intranodal injection. Int. J. Cancer 93, 243–251 (2001).

    CAS  PubMed  Google Scholar 

  134. Lau, R. et al. Phase I trial of intravenous peptide-pulsed dendritic cells in patients with metastatic melanoma. J. Immunother. 24, 66–78 (2001).

    CAS  PubMed  Google Scholar 

  135. Fong, L. et al. Dendritic cell-based xenoantigen vaccination for prostate cancer immunotherapy. J. Immunol. 167, 7150–7156 (2001).

    CAS  PubMed  Google Scholar 

  136. Kikuchi, T. et al. Results of a phase I clinical trial of vaccination of glioma patients with fusions of dendritic and glioma cells. Cancer Immunol. Immunother. 50, 337–344 (2001).

    CAS  PubMed  Google Scholar 

  137. Schuler-Thurner, B. et al. Rapid induction of tumor-specific type 1 T helper cells in metastatic melanoma patients by vaccination with mature, cryopreserved, peptide-loaded monocyte-derived dendritic cells. J. Exp. Med. 195, 1279–1288 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. 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 

  139. Nair, S. K. et al. Induction of tumor-specific cytotoxic T lymphocytes in cancer patients by autologous tumor RNA-transfected dendritic cells. Ann. Surg. 235, 540–549 (2002).

    PubMed  PubMed Central  Google Scholar 

  140. Nair, S. K. et al. Induction of carcinoembryonic antigen (CEA)-specific cytotoxic T-lymphocyte responses in vitro using autologous dendritic cells loaded with CEA peptide or CEA RNA in patients with metastatic malignancies expressing CEA. Int. J. Cancer 82, 121–124 (1999).

    CAS  PubMed  Google Scholar 

  141. Heiser, A. et al. Autologous dendritic cells transfected with prostate-specific antigen RNA stimulate CTL responses against metastatic prostate tumors. J. Clin. Invest. 109, 409–417 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  142. Pecher, G., Haring, A., Kaiser, L. & Thiel, E. Mucin gene (MUC1) transfected dendritic cells as vaccine: results of a phase I/II clinical trial. Cancer Immunol. Immunother. 51, 669–673 (2002).

    CAS  PubMed  Google Scholar 

  143. Banchereau, J. et al. Immune and clinical outcomes in patients with stage IV melanoma vaccinated with peptide-pulsed dendritic cells derived from CD34+ progenitors and activated with type I interferon. J. Immunother. 28, 505–516 (2005).

    CAS  PubMed  Google Scholar 

  144. Trakatelli, M. et al. A new dendritic cell vaccine generated with interleukin-3 and interferon-β induces CD8+ T cell responses against NA17-A2 tumor peptide in melanoma patients. Cancer Immunol. Immunother. 55, 469–474 (2006).

    CAS  PubMed  Google Scholar 

  145. Escudier, B. et al. Vaccination of metastatic melanoma patients with autologous dendritic cell (DC) derived-exosomes: results of the first phase I clinical trial. J. Transl. Med. 3, 10 (2005).

    PubMed  PubMed Central  Google Scholar 

  146. Dannull, J. et al. Enhancement of vaccine-mediated antitumor immunity in cancer patients after depletion of regulatory T cells. J. Clin. Invest. 115, 3623–3633 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Holtl, L. et al. Allogeneic dendritic cell vaccination against metastatic renal cell carcinoma with or without cyclophosphamide. Cancer Immunol. Immunother. 54, 663–670 (2005).

    PubMed  Google Scholar 

  148. Spisek, R. et al. Bortezomib enhances dendritic cell (DC)-mediated induction of immunity to human myeloma via exposure of cell surface heat shock protein 90 on dying tumor cells: therapeutic implications. Blood 109, 4839–4845 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Hildenbrand, B. et al. Immunotherapy of patients with hormone-refractory prostate carcinoma pre-treated with interferon-γ and vaccinated with autologous PSA-peptide loaded dendritic cells — a pilot study. Prostate 67, 500–508 (2007).

    CAS  PubMed  Google Scholar 

  150. Marten, A. et al. Allogeneic dendritic cells fused with tumor cells: preclinical results and outcome of a clinical phase I/II trial in patients with metastatic renal cell carcinoma. Hum. Gene Ther. 14, 483–494 (2003).

    PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Carl G. Figdor.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

FURTHER INFORMATION

Carl G. Figdor's homepage

Glossary

Adjuvant

An agent that does not have any specific antigenic effect in itself, but stimulates the immune system to increase the response to antigens.

Cross-presentation

The mechanism by which certain antigen-presenting cells take up, process and present extracellular antigens on MHC class I molecules to stimulate CD8+ T cells.

Immunoproteasome

The standard proteasome is composed of 14 α and β subunits, of which three, β1, β2 and β5, are involved in peptide-bond cleavage. Interferon-γ induces the expression of the immunosubunits β1i, β2i and β5i that can replace the catalytic subunits of the standard proteasome to generate the immunoproteasome, which has distinct cleavage-site preferences.

C-type lectin receptor

A receptor belonging to the family of Ca+-dependent lectins that share primary structural homology in their carbohydrate-recognition domains.

Regulatory T cell

(TReg cell). A specialized subpopulation of CD4+ T cells that suppresses immune responses to maintain tolerance to (self) antigens.

B16 melanoma model

A well-characterized model to study tumour growth in C57BL/6 mice. There are many sublines of the B16 mouse melanoma cell line, each with its own characteristics.

Hypervariable domains

Three regions within the immunoglobulin variable region that are highly divergent. Together they form a surface that is complementary to the antigen.

Framework regions

Regions adjoining the hypervariable domains, located at the N terminus of the immunoglobulin.

Humanized antibody

Genetically engineered antibody in which the hypervariable domains of a non-human antibody have been transplanted onto a human antibody.

ITAM

(Immunoreceptor tyrosine-based activation motif). A structural motif containing a tyrosine residue that is found in the cytoplasmic tails of several signalling molecules. The consensus sequence consists of Tyr-Xaa-Xaa-Leu/Ile, and the tyrosine is a target for phosphorylation by Src tyrosine kinases and subsequent binding of proteins containing SRC homologue 2 domains.

ITIM

(Immunoreceptor tyrosine-based inhibitory motif). A structural motif found in the cytoplasmic domains of many receptors that negatively regulates intracellular signalling complexes. The consensus sequence consists of Ile/Val-Xaa-Tyr-Xaa-Xaa-Leu/Val.

Toll-like receptors

(TLRs). A family of membrane-spanning proteins that recognize structurally conserved molecules that are shared by various microorganisms. Signalling through TLRs generally results in immune activation.

Single chain antibody

An antibody consisting of only one heavy and one light chain.

Poly (D,L-lactide-co-glycolide) microspheres

Biodegradable microparticles suitable for drug or antigen delivery, consisting of a polymeric ester of lactic and glycolic acid that is approved for application in humans.

Lysosomotropic

Having affinity for, and thus accumulating in, lysosomes. Lysosomotropic weak bases that are capable of crossing biological membranes selectively accumulate in acidic compartments by protonation, thereby affecting organelle pH and function.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Tacken, P., de Vries, I., Torensma, R. et al. Dendritic-cell immunotherapy: from ex vivo loading to in vivo targeting. Nat Rev Immunol 7, 790–802 (2007). https://doi.org/10.1038/nri2173

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1038/nri2173

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