Recent Progress in Dendritic Cell-Based Cancer Immunotherapy
Abstract
Simple Summary
Abstract
1. Introduction
2. DC Subsets and Their Functions
2.1. cDC1
2.2. cDC2
2.3. pDC
2.4. moDC
2.5. LCs
3. Use of DEC205 and CLEC9A in cDC1-Targeting Vaccines
4. Differential Expression of Chemokine Receptors by DC Subsets
4.1. Use of Chemokines to Target DCs
4.2. The Role of the XCL1-XCR1 Axis in cDC1-CD8+ T-Cell Interactions
4.3. Use of Fusion Antigens Targeting XCR1 as CTL-inducing Vaccines
4.4. Generation of highly Active XCL1 and its Use as an Adjuvant for Antigen Delivery to cDC1s
4.5. Induction of Memory CD8+ CTLs by the Highly Active XCL1 Adjuvant
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Ganesh, K.; Massague, J. Targeting metastatic cancer. Nat. Med. 2021, 27, 34–44. [Google Scholar] [CrossRef] [PubMed]
- Dobosz, P.; Dzieciatkowski, T. The Intriguing History of Cancer Immunotherapy. Front. Immunol. 2019, 10, 2965. [Google Scholar] [CrossRef] [PubMed]
- Carlson, R.D.; Flickinger, J.C., Jr.; Snook, A.E. Talkin’ Toxins: From Coley’s to Modern Cancer Immunotherapy. Toxins (Basel) 2020, 12, 241. [Google Scholar] [CrossRef] [PubMed]
- Burnet, F.M. The concept of immunological surveillance. Prog. Exp. Tumor Res. 1970, 13, 1–27. [Google Scholar]
- Thomas, L. On immunosurveillance in human cancer. Yale J. Biol. Med. 1982, 55, 329–333. [Google Scholar]
- Esfahani, K.; Roudaia, L.; Buhlaiga, N.; Del Rincon, S.V.; Papneja, N.; Miller, W.H., Jr. A review of cancer immunotherapy: From the past, to the present, to the future. Curr. Oncol. 2020, 27, S87–S97. [Google Scholar] [CrossRef]
- Dunn, G.P.; Bruce, A.T.; Ikeda, H.; Old, L.J.; Schreiber, R.D. Cancer immunoediting: From immunosurveillance to tumor escape. Nat. Immunol. 2002, 3, 991–998. [Google Scholar] [CrossRef]
- O’Donnell, J.S.; Teng, M.W.L.; Smyth, M.J. Cancer immunoediting and resistance to T cell-based immunotherapy. Nat. Rev. Clin. Oncol. 2019, 16, 151–167. [Google Scholar] [CrossRef]
- Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef]
- He, X.; Xu, C. Immune checkpoint signaling and cancer immunotherapy. Cell Res. 2020, 30, 660–669. [Google Scholar] [CrossRef]
- Bonaventura, P.; Shekarian, T.; Alcazer, V.; Valladeau-Guilemond, J.; Valsesia-Wittmann, S.; Amigorena, S.; Caux, C.; Depil, S. Cold Tumors: A Therapeutic Challenge for Immunotherapy. Front. Immunol. 2019, 10, 168. [Google Scholar] [CrossRef]
- Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010, 363, 711–723. [Google Scholar] [CrossRef] [PubMed]
- Topalian, S.L.; Hodi, F.S.; Brahmer, J.R.; Gettinger, S.N.; Smith, D.C.; McDermott, D.F.; Powderly, J.D.; Carvajal, R.D.; Sosman, J.A.; Atkins, M.B.; et al. Safety, activity, and immune correlates of anti-PD-1 antibody in cancer. N. Engl. J. Med. 2012, 366, 2443–2454. [Google Scholar] [CrossRef]
- Brahmer, J.R.; Tykodi, S.S.; Chow, L.Q.; Hwu, W.J.; Topalian, S.L.; Hwu, P.; Drake, C.G.; Camacho, L.H.; Kauh, J.; Odunsi, K.; et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N. Engl. J. Med. 2012, 366, 2455–2465. [Google Scholar] [CrossRef]
- van der Bruggen, P.; Traversari, C.; Chomez, P.; Lurquin, C.; De Plaen, E.; Van den Eynde, B.; Knuth, A.; Boon, T. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science 1991, 254, 1643–1647. [Google Scholar] [CrossRef]
- Saxena, M.; van der Burg, S.H.; Melief, C.J.M.; Bhardwaj, N. Therapeutic cancer vaccines. Nat. Rev. Cancer 2021, 1–19. [Google Scholar] [CrossRef]
- Colbert, J.D.; Cruz, F.M.; Rock, K.L. Cross-presentation of exogenous antigens on MHC I molecules. Curr. Opin. Immunol. 2020, 64, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Wculek, S.K.; Cueto, F.J.; Mujal, A.M.; Melero, I.; Krummel, M.F.; Sancho, D. Dendritic cells in cancer immunology and immunotherapy. Nat. Rev. Immunol. 2020, 20, 7–24. [Google Scholar] [CrossRef] [PubMed]
- Hilligan, K.L.; Ronchese, F. Antigen presentation by dendritic cells and their instruction of CD4+ T helper cell responses. Cell. Mol. Immunol. 2020, 17, 587–599. [Google Scholar] [CrossRef]
- Eisenbarth, S.C. Dendritic cell subsets in T cell programming: Location dictates function. Nat. Rev. Immunol. 2019, 19, 89–103. [Google Scholar] [CrossRef]
- Anderson, D.A., 3rd; Dutertre, C.A.; Ginhoux, F.; Murphy, K.M. Genetic models of human and mouse dendritic cell development and function. Nat. Rev. Immunol. 2021, 21, 101–115. [Google Scholar] [CrossRef] [PubMed]
- Bottcher, J.P.; Reis e Sousa, C. The Role of Type 1 Conventional Dendritic Cells in Cancer Immunity. Trends Cancer 2018, 4, 784–792. [Google Scholar] [CrossRef] [PubMed]
- den Haan, J.M.; Lehar, S.M.; Bevan, M.J. CD8(+) but not CD8(-) dendritic cells cross-prime cytotoxic T cells in vivo. J. Exp. Med. 2000, 192, 1685–1696. [Google Scholar] [CrossRef] [PubMed]
- Pooley, J.L.; Heath, W.R.; Shortman, K. Cutting edge: Intravenous soluble antigen is presented to CD4 T cells by CD8- dendritic cells, but cross-presented to CD8 T cells by CD8+ dendritic cells. J. Immunol. 2001, 166, 5327–5330. [Google Scholar] [CrossRef] [PubMed]
- Steinman, R.M.; Cohn, Z.A. Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution. J. Exp. Med. 1973, 137, 1142–1162. [Google Scholar] [CrossRef]
- Steinman, R.M.; Cohn, Z.A. Identification of a novel cell type in peripheral lymphoid organs of mice. II. Functional properties in vitro. J. Exp. Med. 1974, 139, 380–397. [Google Scholar]
- Guermonprez, P.; Valladeau, J.; Zitvogel, L.; Thery, C.; Amigorena, S. Antigen presentation and T cell stimulation by dendritic cells. Annu. Rev. Immunol. 2002, 20, 621–667. [Google Scholar] [CrossRef]
- Patente, T.A.; Pinho, M.P.; Oliveira, A.A.; Evangelista, G.C.M.; Bergami-Santos, P.C.; Barbuto, J.A.M. Human Dendritic Cells: Their Heterogeneity and Clinical Application Potential in Cancer Immunotherapy. Front. Immunol. 2019, 9, 3176. [Google Scholar] [CrossRef]
- Merad, M.; Sathe, P.; Helft, J.; Miller, J.; Mortha, A. The dendritic cell lineage: Ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu. Rev. Immunol. 2013, 31, 563–604. [Google Scholar] [CrossRef]
- Spranger, S.; Dai, D.; Horton, B.; Gajewski, T.F. Tumor-Residing Batf3 Dendritic Cells Are Required for Effector T Cell Trafficking and Adoptive T Cell Therapy. Cancer Cell 2017, 31, 711–723.e4. [Google Scholar] [CrossRef]
- Bachem, A.; Hartung, E.; Guttler, S.; Mora, A.; Zhou, X.; Hegemann, A.; Plantinga, M.; Mazzini, E.; Stoitzner, P.; Gurka, S.; et al. Expression of XCR1 Characterizes the Batf3-Dependent Lineage of Dendritic Cells Capable of Antigen Cross-Presentation. Front. Immunol. 2012, 3, 214. [Google Scholar] [CrossRef]
- Calabro, S.; Liu, D.; Gallman, A.; Nascimento, M.S.; Yu, Z.; Zhang, T.T.; Chen, P.; Zhang, B.; Xu, L.; Gowthaman, U.; et al. Differential Intrasplenic Migration of Dendritic Cell Subsets Tailors Adaptive Immunity. Cell Rep. 2016, 16, 2472–2485. [Google Scholar] [CrossRef] [PubMed]
- Segura, E.; Durand, M.; Amigorena, S. Similar antigen cross-presentation capacity and phagocytic functions in all freshly isolated human lymphoid organ-resident dendritic cells. J. Exp. Med. 2013, 210, 1035–1047. [Google Scholar] [CrossRef]
- Audsley, K.M.; McDonnell, A.M.; Waithman, J. Cross-Presenting XCR1(+) Dendritic Cells as Targets for Cancer Immunotherapy. Cells 2020, 9, 565. [Google Scholar] [CrossRef]
- Dorner, B.G.; Dorner, M.B.; Zhou, X.; Opitz, C.; Mora, A.; Guttler, S.; Hutloff, A.; Mages, H.W.; Ranke, K.; Schaefer, M.; et al. Selective expression of the chemokine receptor XCR1 on cross-presenting dendritic cells determines cooperation with CD8+ T cells. Immunity 2009, 31, 823–833. [Google Scholar] [CrossRef] [PubMed]
- Henri, S.; Poulin, L.F.; Tamoutounour, S.; Ardouin, L.; Guilliams, M.; de Bovis, B.; Devilard, E.; Viret, C.; Azukizawa, H.; Kissenpfennig, A.; et al. CD207+ CD103+ dermal dendritic cells cross-present keratinocyte-derived antigens irrespective of the presence of Langerhans cells. J. Exp. Med. 2010, 207, 189–206. [Google Scholar] [CrossRef]
- Contreras, V.; Urien, C.; Guiton, R.; Alexandre, Y.; Vu Manh, T.P.; Andrieu, T.; Crozat, K.; Jouneau, L.; Bertho, N.; Epardaud, M.; et al. Existence of CD8alpha-like dendritic cells with a conserved functional specialization and a common molecular signature in distant mammalian species. J. Immunol. 2010, 185, 3313–3325. [Google Scholar] [CrossRef] [PubMed]
- Crozat, K.; Guiton, R.; Contreras, V.; Feuillet, V.; Dutertre, C.A.; Ventre, E.; Vu Manh, T.P.; Baranek, T.; Storset, A.K.; Marvel, J.; et al. The XC chemokine receptor 1 is a conserved selective marker of mammalian cells homologous to mouse CD8alpha+ dendritic cells. J. Exp. Med. 2010, 207, 1283–1292. [Google Scholar] [CrossRef] [PubMed]
- Bachem, A.; Guttler, S.; Hartung, E.; Ebstein, F.; Schaefer, M.; Tannert, A.; Salama, A.; Movassaghi, K.; Opitz, C.; Mages, H.W.; et al. Superior antigen cross-presentation and XCR1 expression define human CD11c+CD141+ cells as homologues of mouse CD8+ dendritic cells. J. Exp. Med. 2010, 207, 1273–1281. [Google Scholar] [CrossRef] [PubMed]
- Huysamen, C.; Willment, J.A.; Dennehy, K.M.; Brown, G.D. CLEC9A is a novel activation C-type lectin-like receptor expressed on BDCA3+ dendritic cells and a subset of monocytes. J. Biol. Chem. 2008, 283, 16693–16701. [Google Scholar] [CrossRef]
- Collin, M.; Bigley, V. Human dendritic cell subsets: An update. Immunology 2018, 154, 3–20. [Google Scholar] [CrossRef] [PubMed]
- Lafaille, F.G.; Pessach, I.M.; Zhang, S.Y.; Ciancanelli, M.J.; Herman, M.; Abhyankar, A.; Ying, S.W.; Keros, S.; Goldstein, P.A.; Mostoslavsky, G.; et al. Impaired intrinsic immunity to HSV-1 in human iPSC-derived TLR3-deficient CNS cells. Nature 2012, 491, 769–773. [Google Scholar] [CrossRef]
- Villani, A.C.; Satija, R.; Reynolds, G.; Sarkizova, S.; Shekhar, K.; Fletcher, J.; Griesbeck, M.; Butler, A.; Zheng, S.; Lazo, S.; et al. Single-cell RNA-seq reveals new types of human blood dendritic cells, monocytes, and progenitors. Science 2017, 356, eaah4573. [Google Scholar] [CrossRef] [PubMed]
- Hochrein, H.; Shortman, K.; Vremec, D.; Scott, B.; Hertzog, P.; O’Keeffe, M. Differential production of IL-12, IFN-alpha, and IFN-gamma by mouse dendritic cell subsets. J. Immunol. 2001, 166, 5448–5455. [Google Scholar] [CrossRef]
- Fukao, T.; Frucht, D.M.; Yap, G.; Gadina, M.; O’Shea, J.J.; Koyasu, S. Inducible expression of Stat4 in dendritic cells and macrophages and its critical role in innate and adaptive immune responses. J. Immunol. 2001, 166, 4446–4455. [Google Scholar] [CrossRef] [PubMed]
- Liu, S.; Cai, X.; Wu, J.; Cong, Q.; Chen, X.; Li, T.; Du, F.; Ren, J.; Wu, Y.T.; Grishin, N.V.; et al. Phosphorylation of innate immune adaptor proteins MAVS, STING, and TRIF induces IRF3 activation. Science 2015, 347, aaa2630. [Google Scholar] [CrossRef] [PubMed]
- Molenkamp, B.G.; van Leeuwen, P.A.; Meijer, S.; Sluijter, B.J.; Wijnands, P.G.; Baars, A.; van den Eertwegh, A.J.; Scheper, R.J.; de Gruijl, T.D. Intradermal CpG-B activates both plasmacytoid and myeloid dendritic cells in the sentinel lymph node of melanoma patients. Clin. Cancer Res. 2007, 13, 2961–2969. [Google Scholar] [CrossRef] [PubMed]
- Tussiwand, R.; Everts, B.; Grajales-Reyes, G.E.; Kretzer, N.M.; Iwata, A.; Bagaitkar, J.; Wu, X.; Wong, R.; Anderson, D.A.; Murphy, T.L.; et al. Klf4 expression in conventional dendritic cells is required for T helper 2 cell responses. Immunity 2015, 42, 916–928. [Google Scholar] [CrossRef] [PubMed]
- Persson, E.K.; Uronen-Hansson, H.; Semmrich, M.; Rivollier, A.; Hagerbrand, K.; Marsal, J.; Gudjonsson, S.; Hakansson, U.; Reizis, B.; Kotarsky, K.; et al. IRF4 transcription-factor-dependent CD103(+)CD11b(+) dendritic cells drive mucosal T helper 17 cell differentiation. Immunity 2013, 38, 958–969. [Google Scholar] [CrossRef] [PubMed]
- Shin, C.; Han, J.A.; Choi, B.; Cho, Y.K.; Do, Y.; Ryu, S. Intrinsic features of the CD8alpha(-) dendritic cell subset in inducing functional T follicular helper cells. Immunol. Lett. 2016, 172, 21–28. [Google Scholar] [CrossRef]
- Sittig, S.P.; Bakdash, G.; Weiden, J.; Skold, A.E.; Tel, J.; Figdor, C.G.; de Vries, I.J.; Schreibelt, G. A Comparative Study of the T Cell Stimulatory and Polarizing Capacity of Human Primary Blood Dendritic Cell Subsets. Mediators Inflamm. 2016, 2016, 3605643. [Google Scholar] [CrossRef] [PubMed]
- Nizzoli, G.; Larghi, P.; Paroni, M.; Crosti, M.C.; Moro, M.; Neddermann, P.; Caprioli, F.; Pagani, M.; De Francesco, R.; Abrignani, S.; et al. IL-10 promotes homeostatic proliferation of human CD8(+) memory T cells and, when produced by CD1c(+) DCs, shapes naive CD8(+) T-cell priming. Eur. J. Immunol. 2016, 46, 1622–1632. [Google Scholar] [CrossRef] [PubMed]
- Laoui, D.; Keirsse, J.; Morias, Y.; Van Overmeire, E.; Geeraerts, X.; Elkrim, Y.; Kiss, M.; Bolli, E.; Lahmar, Q.; Sichien, D.; et al. The tumour microenvironment harbours ontogenically distinct dendritic cell populations with opposing effects on tumour immunity. Nat. Commun. 2016, 7, 13720. [Google Scholar] [CrossRef] [PubMed]
- Durai, V.; Murphy, K.M. Functions of Murine Dendritic Cells. Immunity 2016, 45, 719–736. [Google Scholar] [CrossRef]
- Hasegawa, H.; Matsumoto, T. Mechanisms of Tolerance Induction by Dendritic Cells In Vivo. Front. Immunol. 2018, 9, 350. [Google Scholar] [CrossRef]
- Piccioli, D.; Tavarini, S.; Borgogni, E.; Steri, V.; Nuti, S.; Sammicheli, C.; Bardelli, M.; Montagna, D.; Locatelli, F.; Wack, A. Functional specialization of human circulating CD16 and CD1c myeloid dendritic-cell subsets. Blood 2007, 109, 5371–5379. [Google Scholar] [CrossRef]
- Brown, C.C.; Gudjonson, H.; Pritykin, Y.; Deep, D.; Lavallee, V.P.; Mendoza, A.; Fromme, R.; Mazutis, L.; Ariyan, C.; Leslie, C.; et al. Transcriptional Basis of Mouse and Human Dendritic Cell Heterogeneity. Cell 2019, 179, 846–863.e24. [Google Scholar] [CrossRef]
- Gilliet, M.; Cao, W.; Liu, Y.J. Plasmacytoid dendritic cells: Sensing nucleic acids in viral infection and autoimmune diseases. Nat. Rev. Immunol. 2008, 8, 594–606. [Google Scholar] [CrossRef]
- Sathe, P.; Vremec, D.; Wu, L.; Corcoran, L.; Shortman, K. Convergent differentiation: Myeloid and lymphoid pathways to murine plasmacytoid dendritic cells. Blood 2013, 121, 11–19. [Google Scholar] [CrossRef]
- Shigematsu, H.; Reizis, B.; Iwasaki, H.; Mizuno, S.; Hu, D.; Traver, D.; Leder, P.; Sakaguchi, N.; Akashi, K. Plasmacytoid dendritic cells activate lymphoid-specific genetic programs irrespective of their cellular origin. Immunity 2004, 21, 43–53. [Google Scholar] [CrossRef]
- Swiecki, M.; Colonna, M. The multifaceted biology of plasmacytoid dendritic cells. Nat. Rev. Immunol. 2015, 15, 471–485. [Google Scholar] [CrossRef] [PubMed]
- Bao, M.; Liu, Y.J. Regulation of TLR7/9 signaling in plasmacytoid dendritic cells. Protein Cell 2013, 4, 40–52. [Google Scholar] [CrossRef] [PubMed]
- Blanco, P.; Palucka, A.K.; Gill, M.; Pascual, V.; Banchereau, J. Induction of dendritic cell differentiation by IFN-alpha in systemic lupus erythematosus. Science 2001, 294, 1540–1543. [Google Scholar] [CrossRef]
- Nestle, F.O.; Conrad, C.; Tun-Kyi, A.; Homey, B.; Gombert, M.; Boyman, O.; Burg, G.; Liu, Y.J.; Gilliet, M. Plasmacytoid predendritic cells initiate psoriasis through interferon-alpha production. J. Exp. Med. 2005, 202, 135–143. [Google Scholar] [CrossRef] [PubMed]
- Faget, J.; Sisirak, V.; Blay, J.Y.; Caux, C.; Bendriss-Vermare, N.; Menetrier-Caux, C. ICOS is associated with poor prognosis in breast cancer as it promotes the amplification of immunosuppressive CD4(+) T cells by plasmacytoid dendritic cells. Oncoimmunology 2013, 2, e23185. [Google Scholar] [CrossRef] [PubMed]
- Conrad, C.; Gregorio, J.; Wang, Y.H.; Ito, T.; Meller, S.; Hanabuchi, S.; Anderson, S.; Atkinson, N.; Ramirez, P.T.; Liu, Y.J.; et al. Plasmacytoid dendritic cells promote immunosuppression in ovarian cancer via ICOS costimulation of Foxp3(+) T-regulatory cells. Cancer Res. 2012, 72, 5240–5249. [Google Scholar] [CrossRef]
- Le Mercier, I.; Poujol, D.; Sanlaville, A.; Sisirak, V.; Gobert, M.; Durand, I.; Dubois, B.; Treilleux, I.; Marvel, J.; Vlach, J.; et al. Tumor promotion by intratumoral plasmacytoid dendritic cells is reversed by TLR7 ligand treatment. Cancer Res. 2013, 73, 4629–4640. [Google Scholar] [CrossRef]
- Stary, G.; Bangert, C.; Tauber, M.; Strohal, R.; Kopp, T.; Stingl, G. Tumoricidal activity of TLR7/8-activated inflammatory dendritic cells. J. Exp. Med. 2007, 204, 1441–1451. [Google Scholar] [CrossRef]
- Kalb, M.L.; Glaser, A.; Stary, G.; Koszik, F.; Stingl, G. TRAIL(+) human plasmacytoid dendritic cells kill tumor cells in vitro: mechanisms of imiquimod- and IFN-alpha-mediated antitumor reactivity. J. Immunol. 2012, 188, 1583–1591. [Google Scholar] [CrossRef]
- Segura, E.; Amigorena, S. Inflammatory dendritic cells in mice and humans. Trends Immunol. 2013, 34, 440–445. [Google Scholar] [CrossRef]
- Perez, C.R.; De Palma, M. Engineering dendritic cell vaccines to improve cancer immunotherapy. Nat. Commun. 2019, 10, 5408. [Google Scholar] [CrossRef] [PubMed]
- Schlitzer, A.; McGovern, N.; Ginhoux, F. Dendritic cells and monocyte-derived cells: Two complementary and integrated functional systems. Semin. Cell Dev. Biol. 2015, 41, 9–22. [Google Scholar] [CrossRef] [PubMed]
- Serbina, N.V.; Salazar-Mather, T.P.; Biron, C.A.; Kuziel, W.A.; Pamer, E.G. TNF/iNOS-producing dendritic cells mediate innate immune defense against bacterial infection. Immunity 2003, 19, 59–70. [Google Scholar] [CrossRef]
- Leon, B.; Lopez-Bravo, M.; Ardavin, C. Monocyte-derived dendritic cells formed at the infection site control the induction of protective T helper 1 responses against Leishmania. Immunity 2007, 26, 519–531. [Google Scholar] [CrossRef]
- Wollenberg, A.; Kraft, S.; Hanau, D.; Bieber, T. Immunomorphological and ultrastructural characterization of Langerhans cells and a novel, inflammatory dendritic epidermal cell (IDEC) population in lesional skin of atopic eczema. J. Investig. Dermatol. 1996, 106, 446–453. [Google Scholar] [CrossRef]
- Wollenberg, A.; Mommaas, M.; Oppel, T.; Schottdorf, E.M.; Gunther, S.; Moderer, M. Expression and function of the mannose receptor CD206 on epidermal dendritic cells in inflammatory skin diseases. J. Investig. Dermatol. 2002, 118, 327–334. [Google Scholar] [CrossRef]
- Eguiluz-Gracia, I.; Bosco, A.; Dollner, R.; Melum, G.R.; Lexberg, M.H.; Jones, A.C.; Dheyauldeen, S.A.; Holt, P.G.; Baekkevold, E.S.; Jahnsen, F.L. Rapid recruitment of CD14(+) monocytes in experimentally induced allergic rhinitis in human subjects. J. Allergy Clin. Immunol. 2016, 137, 1872–1881.e12. [Google Scholar] [CrossRef]
- Grimm, M.C.; Pullman, W.E.; Bennett, G.M.; Sullivan, P.J.; Pavli, P.; Doe, W.F. Direct evidence of monocyte recruitment to inflammatory bowel disease mucosa. J. Gastroenterol. Hepatol. 1995, 10, 387–395. [Google Scholar] [CrossRef]
- Sallusto, F.; Lanzavecchia, A. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. J. Exp. Med. 1994, 179, 1109–1118. [Google Scholar] [CrossRef]
- Nakano, H.; Lin, K.L.; Yanagita, M.; Charbonneau, C.; Cook, D.N.; Kakiuchi, T.; Gunn, M.D. Blood-derived inflammatory dendritic cells in lymph nodes stimulate acute T helper type 1 immune responses. Nat. Immunol. 2009, 10, 394–402. [Google Scholar] [CrossRef]
- Plantinga, M.; Guilliams, M.; Vanheerswynghels, M.; Deswarte, K.; Branco-Madeira, F.; Toussaint, W.; Vanhoutte, L.; Neyt, K.; Killeen, N.; Malissen, B.; et al. Conventional and monocyte-derived CD11b(+) dendritic cells initiate and maintain T helper 2 cell-mediated immunity to house dust mite allergen. Immunity 2013, 38, 322–335. [Google Scholar] [CrossRef] [PubMed]
- Segura, E.; Touzot, M.; Bohineust, A.; Cappuccio, A.; Chiocchia, G.; Hosmalin, A.; Dalod, M.; Soumelis, V.; Amigorena, S. Human inflammatory dendritic cells induce Th17 cell differentiation. Immunity 2013, 38, 336–348. [Google Scholar] [CrossRef] [PubMed]
- Larson, S.R.; Atif, S.M.; Gibbings, S.L.; Thomas, S.M.; Prabagar, M.G.; Danhorn, T.; Leach, S.M.; Henson, P.M.; Jakubzick, C.V. Ly6C(+) monocyte efferocytosis and cross-presentation of cell-associated antigens. Cell Death Differ. 2016, 23, 997–1003. [Google Scholar] [CrossRef] [PubMed]
- Shin, K.S.; Jeon, I.; Kim, B.S.; Kim, I.K.; Park, Y.J.; Koh, C.H.; Song, B.; Lee, J.M.; Lim, J.; Bae, E.A.; et al. Monocyte-Derived Dendritic Cells Dictate the Memory Differentiation of CD8(+) T Cells During Acute Infection. Front. Immunol. 2019, 10, 1887. [Google Scholar] [CrossRef]
- Costa, E.; Corda, M.G.; Guidotti, A. On a brain polypeptide functioning as a putative effector for the recognition sites of benzodiazepine and beta-carboline derivatives. Neuropharmacology 1983, 22, 1481–1492. [Google Scholar] [CrossRef]
- Mody, N.; Dubey, S.; Sharma, R.; Agrawal, U.; Vyas, S.P. Dendritic cell-based vaccine research against cancer. Expert Rev. Clin. Immunol. 2015, 11, 213–232. [Google Scholar] [CrossRef]
- 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 2002, 100, 2858–2866. [Google Scholar] [CrossRef] [PubMed]
- Morse, M.A.; Coleman, R.E.; Akabani, G.; Niehaus, N.; Coleman, D.; Lyerly, H.K. Migration of human dendritic cells after injection in patients with metastatic malignancies. Cancer Res. 1999, 59, 56–58. [Google Scholar]
- Chorro, L.; Sarde, A.; Li, M.; Woollard, K.J.; Chambon, P.; Malissen, B.; Kissenpfennig, A.; Barbaroux, J.B.; Groves, R.; Geissmann, F. Langerhans cell (LC) proliferation mediates neonatal development, homeostasis, and inflammation-associated expansion of the epidermal LC network. J. Exp. Med. 2009, 206, 3089–3100. [Google Scholar] [CrossRef]
- Hoeffel, G.; Wang, Y.; Greter, M.; See, P.; Teo, P.; Malleret, B.; Leboeuf, M.; Low, D.; Oller, G.; Almeida, F.; et al. Adult Langerhans cells derive predominantly from embryonic fetal liver monocytes with a minor contribution of yolk sac-derived macrophages. J. Exp. Med. 2012, 209, 1167–1181. [Google Scholar] [CrossRef]
- Sere, K.; Baek, J.H.; Ober-Blobaum, J.; Muller-Newen, G.; Tacke, F.; Yokota, Y.; Zenke, M.; Hieronymus, T. Two distinct types of Langerhans cells populate the skin during steady state and inflammation. Immunity 2012, 37, 905–916. [Google Scholar] [CrossRef] [PubMed]
- Doebel, T.; Voisin, B.; Nagao, K. Langerhans Cells - The Macrophage in Dendritic Cell Clothing. Trends Immunol. 2017, 38, 817–828. [Google Scholar] [CrossRef] [PubMed]
- Deckers, J.; Hammad, H.; Hoste, E. Langerhans Cells: Sensing the Environment in Health and Disease. Front. Immunol. 2018, 9, 93. [Google Scholar] [CrossRef] [PubMed]
- Romani, N.; Clausen, B.E.; Stoitzner, P. Langerhans cells and more: Langerin-expressing dendritic cell subsets in the skin. Immunol. Rev. 2010, 234, 120–141. [Google Scholar] [CrossRef] [PubMed]
- Nagao, K.; Ginhoux, F.; Leitner, W.W.; Motegi, S.; Bennett, C.L.; Clausen, B.E.; Merad, M.; Udey, M.C. Murine epidermal Langerhans cells and langerin-expressing dermal dendritic cells are unrelated and exhibit distinct functions. Proc. Natl. Acad. Sci. U S A 2009, 106, 3312–3317. [Google Scholar] [CrossRef]
- Matejuk, A. Skin Immunity. Arch. Immunol. Ther. Exp. (Warsz) 2018, 66, 45–54. [Google Scholar] [CrossRef]
- Nguyen, A.V.; Soulika, A.M. The Dynamics of the Skin’s Immune System. Int. J. Mol. Sci. 2019, 20, 1811. [Google Scholar] [CrossRef]
- Kubo, A.; Nagao, K.; Yokouchi, M.; Sasaki, H.; Amagai, M. External antigen uptake by Langerhans cells with reorganization of epidermal tight junction barriers. J. Exp. Med. 2009, 206, 2937–2946. [Google Scholar] [CrossRef]
- Igyarto, B.Z.; Haley, K.; Ortner, D.; Bobr, A.; Gerami-Nejad, M.; Edelson, B.T.; Zurawski, S.M.; Malissen, B.; Zurawski, G.; Berman, J.; et al. Skin-resident murine dendritic cell subsets promote distinct and opposing antigen-specific T helper cell responses. Immunity 2011, 35, 260–272. [Google Scholar] [CrossRef]
- Mathers, A.R.; Janelsins, B.M.; Rubin, J.P.; Tkacheva, O.A.; Shufesky, W.J.; Watkins, S.C.; Morelli, A.E.; Larregina, A.T. Differential capability of human cutaneous dendritic cell subsets to initiate Th17 responses. J. Immunol. 2009, 182, 921–933. [Google Scholar] [CrossRef]
- Levin, C.; Bonduelle, O.; Nuttens, C.; Primard, C.; Verrier, B.; Boissonnas, A.; Combadiere, B. Critical Role for Skin-Derived Migratory DCs and Langerhans Cells in TFH and GC Responses after Intradermal Immunization. J. Invest. Dermatol. 2017, 137, 1905–1913. [Google Scholar] [CrossRef] [PubMed]
- Ouchi, T.; Kubo, A.; Yokouchi, M.; Adachi, T.; Kobayashi, T.; Kitashima, D.Y.; Fujii, H.; Clausen, B.E.; Koyasu, S.; Amagai, M.; et al. Langerhans cell antigen capture through tight junctions confers preemptive immunity in experimental staphylococcal scalded skin syndrome. J. Exp. Med. 2011, 208, 2607–2613. [Google Scholar] [CrossRef] [PubMed]
- Nakajima, S.; Igyarto, B.Z.; Honda, T.; Egawa, G.; Otsuka, A.; Hara-Chikuma, M.; Watanabe, N.; Ziegler, S.F.; Tomura, M.; Inaba, K.; et al. Langerhans cells are critical in epicutaneous sensitization with protein antigen via thymic stromal lymphopoietin receptor signaling. J. Allergy Clin. Immunol. 2012, 129, 1048–1055.e6. [Google Scholar] [CrossRef] [PubMed]
- Yoshiki, R.; Kabashima, K.; Sugita, K.; Atarashi, K.; Shimauchi, T.; Tokura, Y. IL-10-producing Langerhans cells and regulatory T cells are responsible for depressed contact hypersensitivity in grafted skin. J. Invest. Dermatol. 2009, 129, 705–713. [Google Scholar] [CrossRef] [PubMed]
- Gomez de Aguero, M.; Vocanson, M.; Hacini-Rachinel, F.; Taillardet, M.; Sparwasser, T.; Kissenpfennig, A.; Malissen, B.; Kaiserlian, D.; Dubois, B. Langerhans cells protect from allergic contact dermatitis in mice by tolerizing CD8(+) T cells and activating Foxp3(+) regulatory T cells. J. Clin. Investig. 2012, 122, 1700–1711. [Google Scholar] [CrossRef]
- Dudziak, D.; Kamphorst, A.O.; Heidkamp, G.F.; Buchholz, V.R.; Trumpfheller, C.; Yamazaki, S.; Cheong, C.; Liu, K.; Lee, H.W.; Park, C.G.; et al. Differential antigen processing by dendritic cell subsets in vivo. Science 2007, 315, 107–111. [Google Scholar] [CrossRef]
- Wang, B.; Zaidi, N.; He, L.Z.; Zhang, L.; Kuroiwa, J.M.; Keler, T.; Steinman, R.M. Targeting of the non-mutated tumor antigen HER2/neu to mature dendritic cells induces an integrated immune response that protects against breast cancer in mice. Breast Cancer Res. 2012, 14, R39. [Google Scholar] [CrossRef]
- Idoyaga, J.; Lubkin, A.; Fiorese, C.; Lahoud, M.H.; Caminschi, I.; Huang, Y.; Rodriguez, A.; Clausen, B.E.; Park, C.G.; Trumpfheller, C.; et al. Comparable T helper 1 (Th1) and CD8 T-cell immunity by targeting HIV gag p24 to CD8 dendritic cells within antibodies to Langerin, DEC205, and Clec9A. Proc. Natl. Acad. Sci. USA 2011, 108, 2384–2389. [Google Scholar] [CrossRef]
- Charalambous, A.; Oks, M.; Nchinda, G.; Yamazaki, S.; Steinman, R.M. Dendritic cell targeting of survivin protein in a xenogeneic form elicits strong CD4+ T cell immunity to mouse survivin. J. Immunol. 2006, 177, 8410–8421. [Google Scholar] [CrossRef]
- Hawiger, D.; Inaba, K.; Dorsett, Y.; Guo, M.; Mahnke, K.; Rivera, M.; Ravetch, J.V.; Steinman, R.M.; Nussenzweig, M.C. Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivo. J. Exp. Med. 2001, 194, 769–779. [Google Scholar] [CrossRef]
- Sancho, D.; Mourao-Sa, D.; Joffre, O.P.; Schulz, O.; Rogers, N.C.; Pennington, D.J.; Carlyle, J.R.; Reis e Sousa, C. Tumor therapy in mice via antigen targeting to a novel, DC-restricted C-type lectin. J. Clin. Invest. 2008, 118, 2098–2110. [Google Scholar] [CrossRef] [PubMed]
- Tullett, K.M.; Leal Rojas, I.M.; Minoda, Y.; Tan, P.S.; Zhang, J.G.; Smith, C.; Khanna, R.; Shortman, K.; Caminschi, I.; Lahoud, M.H.; et al. Targeting CLEC9A delivers antigen to human CD141(+) DC for CD4(+) and CD8(+)T cell recognition. JCI Insight 2016, 1, e87102. [Google Scholar] [CrossRef] [PubMed]
- Masterman, K.A.; Haigh, O.L.; Tullett, K.M.; Leal-Rojas, I.M.; Walpole, C.; Pearson, F.E.; Cebon, J.; Schmidt, C.; O’Brien, L.; Rosendahl, N.; et al. Human CLEC9A antibodies deliver NY-ESO-1 antigen to CD141(+) dendritic cells to activate naive and memory NY-ESO-1-specific CD8(+) T cells. J. Immunother. Cancer 2020, 8, e000691. [Google Scholar] [CrossRef] [PubMed]
- Pearson, F.E.; Tullett, K.M.; Leal-Rojas, I.M.; Haigh, O.L.; Masterman, K.A.; Walpole, C.; Bridgeman, J.S.; McLaren, J.E.; Ladell, K.; Miners, K.; et al. Human CLEC9A antibodies deliver Wilms’ tumor 1 (WT1) antigen to CD141(+) dendritic cells to activate naive and memory WT1-specific CD8(+) T cells. Clin. Transl. Immunol. 2020, 9, e1141. [Google Scholar] [CrossRef]
- Joffre, O.P.; Sancho, D.; Zelenay, S.; Keller, A.M.; Reis e Sousa, C. Efficient and versatile manipulation of the peripheral CD4+ T-cell compartment by antigen targeting to DNGR-1/CLEC9A. Eur. J. Immunol. 2010, 40, 1255–1265. [Google Scholar] [CrossRef]
- Li, J.; Ahmet, F.; Sullivan, L.C.; Brooks, A.G.; Kent, S.J.; De Rose, R.; Salazar, A.M.; Reis e Sousa, C.; Shortman, K.; Lahoud, M.H.; et al. Antibodies targeting Clec9A promote strong humoral immunity without adjuvant in mice and non-human primates. Eur. J. Immunol. 2015, 45, 854–864. [Google Scholar] [CrossRef]
- Caminschi, I.; Proietto, A.I.; Ahmet, F.; Kitsoulis, S.; Shin Teh, J.; Lo, J.C.; Rizzitelli, A.; Wu, L.; Vremec, D.; van Dommelen, S.L.; et al. The dendritic cell subtype-restricted C-type lectin Clec9A is a target for vaccine enhancement. Blood 2008, 112, 3264–3273. [Google Scholar] [CrossRef]
- Mittal, D.; Vijayan, D.; Putz, E.M.; Aguilera, A.R.; Markey, K.A.; Straube, J.; Kazakoff, S.; Nutt, S.L.; Takeda, K.; Hill, G.R.; et al. Interleukin-12 from CD103(+) Batf3-Dependent Dendritic Cells Required for NK-Cell Suppression of Metastasis. Cancer Immunol. Res. 2018, 5, 1098–1108. [Google Scholar] [CrossRef]
- Alexandre, Y.O.; Ghilas, S.; Sanchez, C.; Le Bon, A.; Crozat, K.; Dalod, M. XCR1+ dendritic cells promote memory CD8+ T cell recall upon secondary infections with Listeria monocytogenes or certain viruses. J. Exp. Med. 2016, 213, 75–92. [Google Scholar] [CrossRef]
- Wong, J.L.; Berk, E.; Edwards, R.P.; Kalinski, P. IL-18-primed helper NK cells collaborate with dendritic cells to promote recruitment of effector CD8+ T cells to the tumor microenvironment. Cancer Res. 2013, 73, 4653–4662. [Google Scholar] [CrossRef]
- Vu Manh, T.P.; Elhmouzi-Younes, J.; Urien, C.; Ruscanu, S.; Jouneau, L.; Bourge, M.; Moroldo, M.; Foucras, G.; Salmon, H.; Marty, H.; et al. Defining Mononuclear Phagocyte Subset Homology Across Several Distant Warm-Blooded Vertebrates Through Comparative Transcriptomics. Front. Immunol. 2015, 6, 299. [Google Scholar] [CrossRef] [PubMed]
- Hartung, E.; Becker, M.; Bachem, A.; Reeg, N.; Jakel, A.; Hutloff, A.; Weber, H.; Weise, C.; Giesecke, C.; Henn, V.; et al. Induction of potent CD8 T cell cytotoxicity by specific targeting of antigen to cross-presenting dendritic cells in vivo via murine or human XCR1. J. Immunol. 2015, 194, 1069–1079. [Google Scholar] [CrossRef] [PubMed]
- Mizumoto, Y.; Hemmi, H.; Katsuda, M.; Miyazawa, M.; Kitahata, Y.; Miyamoto, A.; Nakamori, M.; Ojima, T.; Matsuda, K.; Nakamura, M.; et al. Anticancer effects of chemokine-directed antigen delivery to a cross-presenting dendritic cell subset with immune checkpoint blockade. Br. J. Cancer 2020, 122, 1185–1193. [Google Scholar] [CrossRef] [PubMed]
- Hossain, M.K.; Wall, K.A. Use of Dendritic Cell Receptors as Targets for Enhancing Anti-Cancer Immune Responses. Cancers (Basel) 2019, 11, 418. [Google Scholar] [CrossRef] [PubMed]
- Jiang, W.; Swiggard, W.J.; Heufler, C.; Peng, M.; Mirza, A.; Steinman, R.M.; Nussenzweig, M.C. The receptor DEC-205 expressed by dendritic cells and thymic epithelial cells is involved in antigen processing. Nature 1995, 375, 151–155. [Google Scholar] [CrossRef]
- Shrimpton, R.E.; Butler, M.; Morel, A.S.; Eren, E.; Hue, S.S.; Ritter, M.A. CD205 (DEC-205): A recognition receptor for apoptotic and necrotic self. Mol. Immunol. 2009, 46, 1229–1239. [Google Scholar] [CrossRef]
- Guo, M.; Gong, S.; Maric, S.; Misulovin, Z.; Pack, M.; Mahnke, K.; Nussenzweig, M.C.; Steinman, R.M. A monoclonal antibody to the DEC-205 endocytosis receptor on human dendritic cells. Hum. Immunol. 2000, 61, 729–738. [Google Scholar] [CrossRef]
- Kato, M.; Neil, T.K.; Fearnley, D.B.; McLellan, A.D.; Vuckovic, S.; Hart, D.N. Expression of multilectin receptors and comparative FITC-dextran uptake by human dendritic cells. Int. Immunol. 2000, 12, 1511–1519. [Google Scholar] [CrossRef]
- Inaba, K.; Swiggard, W.J.; Inaba, M.; Meltzer, J.; Mirza, A.; Sasagawa, T.; Nussenzweig, M.C.; Steinman, R.M. 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. 1995, 163, 148–156. [Google Scholar] [CrossRef] [PubMed]
- Sancho, D.; Joffre, O.P.; Keller, A.M.; Rogers, N.C.; Martinez, D.; Hernanz-Falcon, P.; Rosewell, I.; Reis e Sousa, C. Identification of a dendritic cell receptor that couples sensing of necrosis to immunity. Nature 2009, 458, 899–903. [Google Scholar] [CrossRef]
- Ahrens, S.; Zelenay, S.; Sancho, D.; Hanc, P.; Kjaer, S.; Feest, C.; Fletcher, G.; Durkin, C.; Postigo, A.; Skehel, M.; et al. F-actin is an evolutionarily conserved damage-associated molecular pattern recognized by DNGR-1, a receptor for dead cells. Immunity 2012, 36, 635–645. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.G.; Czabotar, P.E.; Policheni, A.N.; Caminschi, I.; Wan, S.S.; Kitsoulis, S.; Tullett, K.M.; Robin, A.Y.; Brammananth, R.; van Delft, M.F.; et al. The dendritic cell receptor Clec9A binds damaged cells via exposed actin filaments. Immunity 2012, 36, 646–657. [Google Scholar] [CrossRef] [PubMed]
- Tullett, K.M.; Lahoud, M.H.; Radford, K.J. Harnessing Human Cross-Presenting CLEC9A(+)XCR1(+) Dendritic Cells for Immunotherapy. Front. Immunol. 2014, 5, 239. [Google Scholar] [CrossRef] [PubMed]
- Iborra, S.; Izquierdo, H.M.; Martinez-Lopez, M.; Blanco-Menendez, N.; Reis e Sousa, C.; Sancho, D. The DC receptor DNGR-1 mediates cross-priming of CTLs during vaccinia virus infection in mice. J. Clin. Investig. 2012, 122, 1628–1643. [Google Scholar] [CrossRef]
- Murray, D.P.; Watson, R.D.; Zezulka, A.V.; Murray, R.G.; Littler, W.A. Plasma catecholamine levels in acute myocardial infarction: Influence of beta-adrenergic blockade and relation to central hemodynamics. Am. Heart J. 1988, 115, 38–44. [Google Scholar] [CrossRef]
- Zlotnik, A.; Yoshie, O. The chemokine superfamily revisited. Immunity 2012, 36, 705–716. [Google Scholar] [CrossRef]
- Bachelerie, F.; Ben-Baruch, A.; Burkhardt, A.M.; Combadiere, C.; Farber, J.M.; Graham, G.J.; Horuk, R.; Sparre-Ulrich, A.H.; Locati, M.; Luster, A.D.; et al. International Union of Basic and Clinical Pharmacology. LXXXIX. Update on the extended family of chemokine receptors and introducing a new nomenclature for atypical chemokine receptors. Pharmacol. Rev. 2013, 66, 1–79. [Google Scholar]
- Yoshie, O.; Imai, T.; Nomiyama, H. Chemokines in immunity. Adv. Immunol. 2001, 78, 57–110. [Google Scholar]
- Sallusto, F.; Palermo, B.; Lenig, D.; Miettinen, M.; Matikainen, S.; Julkunen, I.; Forster, R.; Burgstahler, R.; Lipp, M.; Lanzavecchia, A. Distinct patterns and kinetics of chemokine production regulate dendritic cell function. Eur. J. Immunol. 1999, 29, 1617–1625. [Google Scholar] [CrossRef]
- Dieu, M.C.; Vanbervliet, B.; Vicari, A.; Bridon, J.M.; Oldham, E.; Ait-Yahia, S.; Briere, F.; Zlotnik, A.; Lebecque, S.; Caux, C. Selective recruitment of immature and mature dendritic cells by distinct chemokines expressed in different anatomic sites. J. Exp. Med. 1998, 188, 373–386. [Google Scholar] [CrossRef]
- Campbell, M.J.; Esserman, L.; Byars, N.E.; Allison, A.C.; Levy, R. Idiotype vaccination against murine B cell lymphoma. Humoral and cellular requirements for the full expression of antitumor immunity. J. Immunol. 1990, 145, 1029–1036. [Google Scholar] [PubMed]
- Bosteels, C.; Fierens, K.; De Prijck, S.; Van Moorleghem, J.; Vanheerswynghels, M.; De Wolf, C.; Chalon, A.; Collignon, C.; Hammad, H.; Didierlaurent, A.M.; et al. CCR2- and Flt3-Dependent Inflammatory Conventional Type 2 Dendritic Cells Are Necessary for the Induction of Adaptive Immunity by the Human Vaccine Adjuvant System AS01. Front. Immunol. 2021, 11, 606805. [Google Scholar] [CrossRef] [PubMed]
- Penna, G.; Vulcano, M.; Roncari, A.; Facchetti, F.; Sozzani, S.; Adorini, L. Cutting edge: Differential chemokine production by myeloid and plasmacytoid dendritic cells. J. Immunol. 2002, 169, 6673–6676. [Google Scholar] [CrossRef] [PubMed]
- Krug, A.; Uppaluri, R.; Facchetti, F.; Dorner, B.G.; Sheehan, K.C.; Schreiber, R.D.; Cella, M.; Colonna, M. IFN-producing cells respond to CXCR3 ligands in the presence of CXCL12 and secrete inflammatory chemokines upon activation. J. Immunol. 2002, 169, 6079–6083. [Google Scholar] [CrossRef] [PubMed]
- Megjugorac, N.J.; Young, H.A.; Amrute, S.B.; Olshalsky, S.L.; Fitzgerald-Bocarsly, P. Virally stimulated plasmacytoid dendritic cells produce chemokines and induce migration of T and NK cells. J. Leukoc. Biol. 2004, 75, 504–514. [Google Scholar] [CrossRef] [PubMed]
- Chow, K.V.; Lew, A.M.; Sutherland, R.M.; Zhan, Y. Monocyte-Derived Dendritic Cells Promote Th Polarization, whereas Conventional Dendritic Cells Promote Th Proliferation. J. Immunol. 2016, 196, 624–636. [Google Scholar] [CrossRef]
- Sutti, S.; Bruzzi, S.; Heymann, F.; Liepelt, A.; Krenkel, O.; Toscani, A.; Ramavath, N.N.; Cotella, D.; Albano, E.; Tacke, F. CX3CR1 Mediates the Development of Monocyte-Derived Dendritic Cells during Hepatic Inflammation. Cells 2019, 8, 1099. [Google Scholar] [CrossRef]
- Biragyn, A.; Tani, K.; Grimm, M.C.; Weeks, S.; Kwak, L.W. Genetic fusion of chemokines to a self tumor antigen induces protective, T-cell dependent antitumor immunity. Nat. Biotechnol. 1999, 17, 253–258. [Google Scholar] [CrossRef]
- Biragyn, A.; Surenhu, M.; Yang, D.; Ruffini, P.A.; Haines, B.A.; Klyushnenkova, E.; Oppenheim, J.J.; Kwak, L.W. Mediators of innate immunity that target immature, but not mature, dendritic cells induce antitumor immunity when genetically fused with nonimmunogenic tumor antigens. J. Immunol. 2001, 167, 6644–6653. [Google Scholar] [CrossRef]
- Toumbis, M.; Zervas, J.; Anagnostopoulou, O.; Konstantopoulos, K.; Krimbeni, G.; Kotsovoulou, V.; Fessas, F. HLA antigens and bronchogenic carcinoma in the Greek population. Acta Oncol. 1991, 30, 575–578. [Google Scholar] [CrossRef]
- Kelner, G.S.; Kennedy, J.; Bacon, K.B.; Kleyensteuber, S.; Largaespada, D.A.; Jenkins, N.A.; Copeland, N.G.; Bazan, J.F.; Moore, K.W.; Schall, T.J.; et al. Lymphotactin: A cytokine that represents a new class of chemokine. Science 1994, 266, 1395–1399. [Google Scholar] [CrossRef]
- Yoshida, T.; Imai, T.; Kakizaki, M.; Nishimura, M.; Yoshie, O. Molecular cloning of a novel C or gamma type chemokine, SCM-1. FEBS Lett. 1995, 360, 155–159. [Google Scholar] [CrossRef]
- Hedrick, J.A.; Saylor, V.; Figueroa, D.; Mizoue, L.; Xu, Y.; Menon, S.; Abrams, J.; Handel, T.; Zlotnik, A. Lymphotactin is produced by NK cells and attracts both NK cells and T cells in vivo. J. Immunol. 1997, 158, 1533–1540. [Google Scholar]
- Kennedy, J.; Kelner, G.S.; Kleyensteuber, S.; Schall, T.J.; Weiss, M.C.; Yssel, H.; Schneider, P.V.; Cocks, B.G.; Bacon, K.B.; Zlotnik, A. Molecular cloning and functional characterization of human lymphotactin. J. Immunol. 1995, 155, 203–209. [Google Scholar] [PubMed]
- Yoshida, T.; Imai, T.; Kakizaki, M.; Nishimura, M.; Takagi, S.; Yoshie, O. Identification of single C motif-1/lymphotactin receptor XCR1. J. Biol. Chem. 1998, 273, 16551–16554. [Google Scholar] [CrossRef] [PubMed]
- Yoshida, T.; Izawa, D.; Nakayama, T.; Nakahara, K.; Kakizaki, M.; Imai, T.; Suzuki, R.; Miyasaka, M.; Yoshie, O. Molecular cloning of mXCR1, the murine SCM-1/lymphotactin receptor. FEBS Lett. 1999, 458, 37–40. [Google Scholar] [CrossRef]
- Crozat, K.; Tamoutounour, S.; Vu Manh, T.P.; Fossum, E.; Luche, H.; Ardouin, L.; Guilliams, M.; Azukizawa, H.; Bogen, B.; Malissen, B.; et al. Cutting edge: Expression of XCR1 defines mouse lymphoid-tissue resident and migratory dendritic cells of the CD8alpha+ type. J. Immunol. 2011, 187, 4411–4415. [Google Scholar] [CrossRef]
- Bottcher, J.P.; Bonavita, E.; Chakravarty, P.; Blees, H.; Cabeza-Cabrerizo, M.; Sammicheli, S.; Rogers, N.C.; Sahai, E.; Zelenay, S.; Reis e Sousa, C. NK Cells Stimulate Recruitment of cDC1 into the Tumor Microenvironment Promoting Cancer Immune Control. Cell 2018, 172, 1022–1037.e14. [Google Scholar] [CrossRef]
- Bodder, J.; Zahan, T.; van Slooten, R.; Schreibelt, G.; de Vries, I.J.M.; Florez-Grau, G. Harnessing the cDC1-NK Cross-Talk in the Tumor Microenvironment to Battle Cancer. Front. Immunol. 2021, 11, 631713. [Google Scholar] [CrossRef] [PubMed]
- Fossum, E.; Grodeland, G.; Terhorst, D.; Tveita, A.A.; Vikse, E.; Mjaaland, S.; Henri, S.; Malissen, B.; Bogen, B. Vaccine molecules targeting Xcr1 on cross-presenting DCs induce protective CD8+ T-cell responses against influenza virus. Eur. J. Immunol. 2015, 45, 624–635. [Google Scholar] [CrossRef]
- Terhorst, D.; Fossum, E.; Baranska, A.; Tamoutounour, S.; Malosse, C.; Garbani, M.; Braun, R.; Lechat, E.; Crameri, R.; Bogen, B.; et al. Laser-assisted intradermal delivery of adjuvant-free vaccines targeting XCR1+ dendritic cells induces potent antitumoral responses. J. Immunol. 2015, 194, 5895–5902. [Google Scholar] [CrossRef]
- Lysen, A.; Braathen, R.; Gudjonsson, A.; Tesfaye, D.Y.; Bogen, B.; Fossum, E. Dendritic cell targeted Ccl3- and Xcl1-fusion DNA vaccines differ in induced immune responses and optimal delivery site. Sci. Rep. 2019, 9, 1820. [Google Scholar] [CrossRef]
- Matsuo, K.; Kitahata, K.; Kawabata, F.; Kamei, M.; Hara, Y.; Takamura, S.; Oiso, N.; Kawada, A.; Yoshie, O.; Nakayama, T. A Highly Active Form of XCL1/Lymphotactin Functions as an Effective Adjuvant to Recruit Cross-Presenting Dendritic Cells for Induction of Effector and Memory CD8(+) T Cells. Front. Immunol. 2018, 9, 2775. [Google Scholar] [CrossRef]
- Tuinstra, R.L.; Peterson, F.C.; Kutlesa, S.; Elgin, E.S.; Kron, M.A.; Volkman, B.F. Interconversion between two unrelated protein folds in the lymphotactin native state. Proc. Natl. Acad. Sci. USA 2008, 105, 5057–5062. [Google Scholar] [CrossRef]
- Fox, J.C.; Nakayama, T.; Tyler, R.C.; Sander, T.L.; Yoshie, O.; Volkman, B.F. Structural and agonist properties of XCL2, the other member of the C-chemokine subfamily. Cytokine 2015, 71, 302–311. [Google Scholar] [CrossRef]
- Fox, J.C.; Thomas, M.A.; Dishman, A.F.; Larsen, O.; Nakayama, T.; Yoshie, O.; Rosenkilde, M.M.; Volkman, B.F. Structure-function guided modeling of chemokine-GPCR specificity for the chemokine XCL1 and its receptor XCR1. Sci. Signal. 2019, 12, eaat4128. [Google Scholar] [CrossRef]
- Kamei, M.; Matsuo, K.; Imanishi, H.; Hara, Y.; Quen, Y.S.; Kamiyama, F.; Oiso, N.; Kawada, A.; Okada, N.; Nakayama, T. Transcutaneous immunization with a highly active form of XCL1 as a vaccine adjuvant using a hydrophilic gel patch elicits long-term CD8(+) T cell responses. J. Pharmacol. Sci. 2020, 143, 182–187. [Google Scholar] [CrossRef] [PubMed]
- Nizard, M.; Roussel, H.; Diniz, M.O.; Karaki, S.; Tran, T.; Voron, T.; Dransart, E.; Sandoval, F.; Riquet, M.; Rance, B.; et al. Induction of resident memory T cells enhances the efficacy of cancer vaccine. Nat. Commun. 2017, 8, 15221. [Google Scholar] [CrossRef]
- Klebanoff, C.A.; Gattinoni, L.; Torabi-Parizi, P.; Kerstann, K.; Cardones, A.R.; Finkelstein, S.E.; Palmer, D.C.; Antony, P.A.; Hwang, S.T.; Rosenberg, S.A.; et al. Central memory self/tumor-reactive CD8+ T cells confer superior antitumor immunity compared with effector memory T cells. Proc. Natl. Acad. Sci. USA 2005, 102, 9571–9576. [Google Scholar] [CrossRef]
- Manjarrez-Orduno, N.; Menard, L.C.; Kansal, S.; Fischer, P.; Kakrecha, B.; Jiang, C.; Cunningham, M.; Greenawalt, D.; Patel, V.; Yang, M.; et al. Circulating T Cell Subpopulations Correlate With Immune Responses at the Tumor Site and Clinical Response to PD1 Inhibition in Non-Small Cell Lung Cancer. Front. Immunol. 2018, 9, 1613. [Google Scholar] [CrossRef]
- Hu, Z.; Ott, P.A.; Wu, C.J. Towards personalized, tumour-specific, therapeutic vaccines for cancer. Nat. Rev. Immunol. 2018, 18, 168–182. [Google Scholar] [CrossRef]
- Galluzzi, L.; Buque, A.; Kepp, O.; Zitvogel, L.; Kroemer, G. Immunogenic cell death in cancer and infectious disease. Nat. Rev. Immunol. 2017, 17, 97–111. [Google Scholar] [CrossRef]
- Joshi, N.S.; Cui, W.; Chandele, A.; Lee, H.K.; Urso, D.R.; Hagman, J.; Gapin, L.; Kaech, S.M. Inflammation directs memory precursor and short-lived effector CD8(+) T cell fates via the graded expression of T-bet transcription factor. Immunity 2007, 27, 281–295. [Google Scholar] [CrossRef]
- Pipkin, M.E.; Sacks, J.A.; Cruz-Guilloty, F.; Lichtenheld, M.G.; Bevan, M.J.; Rao, A. Interleukin-2 and inflammation induce distinct transcriptional programs that promote the differentiation of effector cytolytic T cells. Immunity 2010, 32, 79–90. [Google Scholar] [CrossRef] [PubMed]
- Williams, M.A.; Tyznik, A.J.; Bevan, M.J. Interleukin-2 signals during priming are required for secondary expansion of CD8+ memory T cells. Nature 2006, 441, 890–893. [Google Scholar] [CrossRef]
- Hermans, I.F.; Ritchie, D.S.; Yang, J.; Roberts, J.M.; Ronchese, F. CD8+ T cell-dependent elimination of dendritic cells in vivo limits the induction of antitumor immunity. J. Immunol. 2000, 164, 3095–3101. [Google Scholar] [CrossRef]
- Yang, J.; Huck, S.P.; McHugh, R.S.; Hermans, I.F.; Ronchese, F. Perforin-dependent elimination of dendritic cells regulates the expansion of antigen-specific CD8+ T cells in vivo. Proc. Natl. Acad. Sci. USA 2006, 103, 147–152. [Google Scholar] [CrossRef]
- Glenn, G.M.; O’Hagan, D.T. Adjuvants: Progress, regress and pandemic preparedness. Expert Rev. Vaccines 2007, 6, 651–652. [Google Scholar] [CrossRef][Green Version]
- Ishii, K.J.; Akira, S. Toll or toll-free adjuvant path toward the optimal vaccine development. J. Clin. Immunol. 2007, 27, 363–371. [Google Scholar] [CrossRef]
- Steinhagen, F.; Kinjo, T.; Bode, C.; Klinman, D.M. TLR-based immune adjuvants. Vaccine 2011, 29, 3341–3355. [Google Scholar] [CrossRef]
- Vijay, K. Toll-like receptors in immunity and inflammatory diseases: Past, present, and future. Int. Immunopharmacol. 2018, 59, 391–412. [Google Scholar] [CrossRef] [PubMed]
- Mastelic-Gavillet, B.; Balint, K.; Boudousquie, C.; Gannon, P.O.; Kandalaft, L.E. Personalized Dendritic Cell Vaccines-Recent Breakthroughs and Encouraging Clinical Results. Front. Immunol. 2019, 10, 766. [Google Scholar] [CrossRef] [PubMed]
- Baldin, A.V.; Savvateeva, L.V.; Bazhin, A.V.; Zamyatnin, A.A., Jr. Dendritic Cells in Anticancer Vaccination: Rationale for Ex Vivo Loading or In Vivo Targeting. Cancers (Basel) 2020, 12, 590. [Google Scholar] [CrossRef] [PubMed]
- Nagarsheth, N.; Wicha, M.S.; Zou, W. Chemokines in the cancer microenvironment and their relevance in cancer immunotherapy. Nat. Rev. Immunol. 2017, 17, 559–572. [Google Scholar] [CrossRef]
- Bobanga, I.D.; Petrosiute, A.; Huang, A.Y. Chemokines as Cancer Vaccine Adjuvants. Vaccines (Basel) 2013, 1, 444–462. [Google Scholar] [CrossRef]
- Darvin, P.; Toor, S.M.; Sasidharan Nair, V.; Elkord, E. Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp. Mol. Med. 2018, 50, 1–11. [Google Scholar] [CrossRef]
- Murciano-Goroff, Y.R.; Warner, A.B.; Wolchok, J.D. The future of cancer immunotherapy: Microenvironment-targeting combinations. Cell Res. 2020, 30, 507–519. [Google Scholar] [CrossRef]
- Rice, A.E.; Latchman, Y.E.; Balint, J.P.; Lee, J.H.; Gabitzsch, E.S.; Jones, F.R. An HPV-E6/E7 immunotherapy plus PD-1 checkpoint inhibition results in tumor regression and reduction in PD-L1 expression. Cancer Gene Ther. 2015, 22, 454–462. [Google Scholar] [CrossRef]
- Antonios, J.P.; Soto, H.; Everson, R.G.; Orpilla, J.; Moughon, D.; Shin, N.; Sedighim, S.; Yong, W.H.; Li, G.; Cloughesy, T.F.; et al. PD-1 blockade enhances the vaccination-induced immune response in glioma. JCI Insight 2016, 1, e87059. [Google Scholar] [CrossRef]
- Curran, M.A.; Glisson, B.S. New Hope for Therapeutic Cancer Vaccines in the Era of Immune Checkpoint Modulation. Annu. Rev. Med. 2019, 70, 409–424. [Google Scholar] [CrossRef]
- Le, D.T.; Lutz, E.; Uram, J.N.; Sugar, E.A.; Onners, B.; Solt, S.; Zheng, L.; Diaz, L.A., Jr.; Donehower, R.C.; Jaffee, E.M.; et al. Evaluation of ipilimumab in combination with allogeneic pancreatic tumor cells transfected with a GM-CSF gene in previously treated pancreatic cancer. J. Immunother. 2013, 36, 382–389. [Google Scholar] [CrossRef] [PubMed]
- Weber, J.S.; Kudchadkar, R.R.; Yu, B.; Gallenstein, D.; Horak, C.E.; Inzunza, H.D.; Zhao, X.; Martinez, A.J.; Wang, W.; Gibney, G.; et al. Safety, efficacy, and biomarkers of nivolumab with vaccine in ipilimumab-refractory or -naive melanoma. J. Clin. Oncol. 2013, 31, 4311–4318. [Google Scholar] [CrossRef] [PubMed]
- Gibney, G.T.; Kudchadkar, R.R.; DeConti, R.C.; Thebeau, M.S.; Czupryn, M.P.; Tetteh, L.; Eysmans, C.; Richards, A.; Schell, M.J.; Fisher, K.J.; et al. Safety, correlative markers, and clinical results of adjuvant nivolumab in combination with vaccine in resected high-risk metastatic melanoma. Clin. Cancer Res. 2015, 21, 712–720. [Google Scholar] [CrossRef]
- Mougel, A.; Terme, M.; Tanchot, C. Therapeutic Cancer Vaccine and Combinations With Antiangiogenic Therapies and Immune Checkpoint Blockade. Front. Immunol. 2019, 10, 467. [Google Scholar] [CrossRef]
Target Molecule | Type | Expressing Cell | Function | Application (Ref) |
---|---|---|---|---|
DEC-205 | C-type lectin receptor | cDC1s, cDC2s, B cells, T cells, NK cells | Antigen recognition (apoptotic and necrotic cells) | [106,107,108,109,110] |
CLEC9A | C-type lectin receptor | cDC1s, some pDCs, monocytes | Antigen recognition/endocytosis (cross-presentation) | [111,112,113,114,115,116,117] |
XCR1 | Chemokine receptor | cDC1s | Cell migration | [118,119,120,121,122,123] |
Chemokine | Chemokine Receptor | Immune Response | Ref. |
---|---|---|---|
CCL7 | CCR1, CCR2, CCR3 | Antigen-specific IgG response Antitumor effect | [149,150] |
CCL20 | CCR6 | Antigen-specific IgG response Antitumor effect | [149,150] |
CCL21 | CCR7 | Antigen-specific IgG response | [150] |
XCL1 | XCR1 | CTL response, Antitumor effect Antiviral effect | [118,119,120,121,122,123] |
Type | Immunization Method | Immune Response | Ref |
---|---|---|---|
Targeting cDC1 | |||
Antigen-XCL1 fusion protein | Injection | CTL responses combined with LPS | [118] |
Antigen-conjugated anti-XCR1 antibody | Injection | CTL responses combined with LPS | [118] |
Antigen peptide-XCL1 fusion protein | Injection | CTL responses, antitumor immunity, enhance CTL responses with anti-PD1 | [119] |
Antigen-XCL1 dimeric fusion protein | DNA vaccine | CTL responses, Influenza virus protection | [120,122] |
Laser-assisted intradermal delivery | CD4+/CD8+ T-cell responses, antitumor immunity | [121] | |
Recruiting cDC1 | |||
Mixture of antigen and a highly active form of XCL1 | Injection | Effector and memory CTL responses, antitumor immunity | [123] |
Transcutaneous device delivery | Enhance memory CTL responses, antitumor immunity | [163] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Matsuo, K.; Yoshie, O.; Kitahata, K.; Kamei, M.; Hara, Y.; Nakayama, T. Recent Progress in Dendritic Cell-Based Cancer Immunotherapy. Cancers 2021, 13, 2495. https://doi.org/10.3390/cancers13102495
Matsuo K, Yoshie O, Kitahata K, Kamei M, Hara Y, Nakayama T. Recent Progress in Dendritic Cell-Based Cancer Immunotherapy. Cancers. 2021; 13(10):2495. https://doi.org/10.3390/cancers13102495
Chicago/Turabian StyleMatsuo, Kazuhiko, Osamu Yoshie, Kosuke Kitahata, Momo Kamei, Yuta Hara, and Takashi Nakayama. 2021. "Recent Progress in Dendritic Cell-Based Cancer Immunotherapy" Cancers 13, no. 10: 2495. https://doi.org/10.3390/cancers13102495
APA StyleMatsuo, K., Yoshie, O., Kitahata, K., Kamei, M., Hara, Y., & Nakayama, T. (2021). Recent Progress in Dendritic Cell-Based Cancer Immunotherapy. Cancers, 13(10), 2495. https://doi.org/10.3390/cancers13102495