Dendritic Cells: The Long and Evolving Road towards Successful Targetability in Cancer
Abstract
:1. Introduction
2. Distinct Origin and Development of DCs
Monocytes
3. DCs: The Most Potent APC of the Body
4. Identification of New DC Subsets
DC Subpopulation | Localization | ID Markers | Maturation Molecules Activation/Inhibition | Ontogeny and Function | References |
---|---|---|---|---|---|
Classical Type I DC (cDC1) or DC1 | <0.01% of CD45+ cells in the blood of healthy donors. <0.1% of CD45+ cells in tissues. Blood Lymph node paracortex Other tissues (e.g., tonsil, spleen, skin, lung, intestine, ileum, payers’ patch, liver, and lymph nodes). | CD141+ Low/moderate CD11c, CD11b and SIRPα (Signal Regulatory Protein Alpha, CD172) CD103+ (αE integrin) CD1c− in circulation CD1c+/− in tissues (e.g., skin and lungs) CD59+ SLAMF8+ CD26+ CD8α+ Negative for moDCs markers: CD14, CD16, CD209 and SIRP CD45RA− CD123− | DEC-205+ (dendritic and epithelial cells-205) XCR1+ (the chemokine receptor X-C Motif Chemokine Receptor 1) maturation marker CLEC9A+ (C type lectin receptor Clec9A, CD370) Axl+ receptor tyrosine kinase CADM1+ (Cell adhesion molecule 1) CD135+ (Flt3L receptor) High expression of MHCII IDO1/IDO2 (Indoleamine 2,3 Dioxygenase 1 and 2 TLR-3 and TLR-9 BTLA (B and T Lymphocyte Attenuator, CD272) Tim3 and PD-L1 inhibitory receptors Higher CD40 and lower CD86 expression than cDC2s | Relatively homogenous population in comparison to cDC2. Polarize activated CD4+ T cells toward Th1 and away from Th2 phenotype. Major source of IL-12 in vivo. Immune responses against cancer and pathogens infection. CLEC9+ CD141+ cells are producers of IFNγ and CD8+ T cell activation by cross presentation in response to TLR-3 ligation. | [41,64,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96] |
Classical Type 2 DC (cDC2) | Lymph nodes in subcapsular sinus. Two distinct subsets based upon CD5 expression CD5high and CD5low in blood and skin. | CD11b+ CD11c+ CD33+ CD13+ CD172α+ Discriminatory marker CD1c (BDCA1) CD5high DCs express high levels of cDC2-specific genes, while CD5low DCs preferentially express monocytes-related genes. A population with analogous phenotype CD1c+CD14+CD5low cDCs (VCAM, FCN1, S100A8, S100A9) was named DC3. Their ontogeny and functions remain unknown. | Mgl2 or CLEC12A, CD2 FCεR1 SIRPα CCR2 CCR6 TLR2, TLR4–6, TLR8 and TLR9 Dependent on IRF4, IRF2, TRAF6, KLF2, RelB, RBP-J transcription factors and Notch signaling. The CD5high cells have strong IRF4, CCR7, CD207, TLR3 expression. Additional markers: CLEC10A VEGFA, FC R2A. | Secretion of inflammatory cytokines: TNF-α, IL-12p70, IL-23, TNF-α, IL-1, IL-6, IL-8, IL-12, IL-18 and chemokines: CCL3, CCL4 and CXCL8. Uptake of exogenous antigens, specifically tumor antigen. T immune responses against extracellular bacterial and fungal pathogens. Polarization of Th17, Th2, Th1, Th22, Treg and cytotoxic responses. By the induction of Th2 responses are involved in humoral responses via IL-6 and IL-1β secretion. Higher potential to induce CD4+ T cell proliferation than cDC1. CD5high cells induce IL-10 producing Treg cells, whereas CD5low cells induce IFNγ producing T cells. | [31,33,41,64,88,90,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112] |
DC2 | DC2Bs have been detected in the spleen apparently are absent in circulation. | CD1c+ Compared to DC3 have slightly higher expression of: MHCII CD1c CD11c CD5 Unique expression of CD32b CD301 (macrophage galactose-type C-type lectin, CLEC10A) FC R1A (the alpha chain of the high affinity receptor for IgE). Two subpopulations by CLEC10A expression: DC2A: CD1low CLEC10A− CLECL4high DC2B: CD1c+ CLEC10A+ CLEC4Alow | TFs, gene associated with lipid antigen presentation and metabolism (CD1E, NPC2, PSAP) have found to be enriched in DC2Bs, whereas CD3E in DC2A subset. | DC2 cells show greater secretory ability of CCL19, IL-10, IL-12B and IL-18 than DC3 cells. DC2B exhibited pro-inflammatory potential with an increased expression of IL-1B. DC2A have anti-inflammatory phenotype characterized by higher levels of transcript for amphiregulin (AREG), IDO1, the immunomodulatory receptor CD300a and IL-22 binding protein. | [41,78,98,113,114] |
DC3 | Expanded DC3 populations have been observed in blood of patients with systemic lupus erythematosus (LE) and in skin of patients with psoriasis | CD1c+ CD14 low/high CD5low Gene signature (CD14, S100A9, S100A8). Unique expression of CD163 (a scavenger receptor and PPR for bacterial) and CD36. | CD88− BTLA− Additional key markers VCAM, LYZ and ANXA1 Expresses unique markers enriched for antigen processing, MHCII and leukocytes activation. Express CCR7, co-stimulatory molecules (CD80, CD86, CD70, CD40), T cell attracting chemokines (CCL5, CCL19, CCL17, CCL22, CXCL9, CXCL10, CXCL11 and CXCL13). | DC3 cells are characterized by acute and chronic inflammatory gene signature. DC3 function in vivo and its contribution to pathological conditions are still uncertain. Produce IL1B and IL23A during pathogenesis. Produce high amount of IL-12p70, IL-13, IL-17 and IL-10, TNF-α and IL-1β, CCL2, CCL1, CCL3, CXCL1, CXCL3 and CXCL5. DC3s induce TRM differentiation from naïve CD8+T cells. | [41,73,75,78,100,112,115] |
CD16+ DC or DC4 | Lin− HLA-DR+ CD11c+ CD14low/high CD16+ CD141− CD1c− High expression of PPP1R14A and DAB2 | Slan+ cells? CD85d (ILT-4) and CD85h (ILT-1) CD115 CD31 SLC7A7 CD98 Siglec-10 | Gene of sets related to type I interferon signaling and virus response. IFN-α/β signaling? Further studies are needed to clarify the phenotype and function of this newly described subset. | [41,98,116,117,118] | |
AXL+ SIGLEC6+ DC (AS DC) or DC5 | 2–3% of all DC compartment Two subsets identified: CD123+ CD11c− AS DCs (0.7%, of the CD45+ HLADR+ fraction) CD123−/low CD11c+ AS DCs (1.7% of the CD45+ HLADR+ fraction) Found in secondary lymphoid organs, like tonsils, co-localizing with T cells in situ. | AXL+ SIGLEC6+ SIGLEC1+ SIGLEC2+ The commonly markers used to identify pDCs (CD123, CD303 and CD304, CD123, CD11c) have also been found. The identity of AS DCs has not been clarified given their heterogeneity, specifically with a spectral expression for CD123, CD5, or CD11c. | CD221 and CD169, CD39 and IRAP AXL+ SIGLEC6+ DCscan differentiate into cDC2 showing a progenitor potential for cDCs. | As DCs do not secrete IFNα, induce T cell activation (like cDCs), Treg formation or B cell activation. Both CD123+ CD11c− AS DCs and CD123−/low CD11c+ AS DCs subsets are potent stimulators of allogenic CD4+ and CD8+ T cell proliferation. | [41,71,77,114,117,119] |
pDC or DC6 | 0.2–0.4% of total CD45+ circulating cells. | Lin− CD11c− CD11b− CD13− CD123/IL3R+ CD303/BDCA2+ CD304/BDCA4+ Used as a discriminative lineage marker to identify pDCs: ILT7 (immunoglobulin-like transcript 7) CD4 BCL11A TCF4 The co-staining of CD123 with HLA-DR is essential to exclude HLA-DR-granulocytic populations expressing CD123. CD68 is a marker useful for distinguishing them from DC1 and DC2. Named by Villani et al. as DC6, pDC differentiation is modulated by IRF8, IRF7, E2.2, Runx2, SpiB, IRF4, BCL11A and ZEB2 expression. Dual myeloid-lymphoid origin | At steady state, pDCs exhibits low levels of MHC class I and II, and low to undetectable level of costimulatory molecules CD40, CD80 and CD86. PD-L1 is negligible on their surface and very low expression of Tim3 is present at basal and stimulatory conditions. After stimulation, pDCs express IFN-α/β and differentiate into mature MHC class I/IIhigh, CD80+, CD40+ CD86+. TLR7 and TLR9 CCR2, CCR5, CXCR3 and CXCR4 chemokine receptors has been also detected | Natural IFN-α producing cells in response to virus, IL-3, and bacterial components. Moderate amounts of TNF-α and IL-6 after viral stimulation. Signaling pathway for CLEC4C, NRP1 and IL3RA. Co-stimulatory and activation of NK cell, cDCs, T cells and B cells. Tolerogenic functions by inducing T cell depletion, CD4 T cell anergy and T reg differentiation. Cross-priming post-antigen presentation by driving antigen transfer to bystander DCs through pDCs-derived exosomes. | [41,64,88,114,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134] |
Monocyte-derived DC | HLA-DR+ CD11+ CD1c+ CD1a+ CD1b+ FcεRI+ Negative for: CD16, CD163, CD206, CD209, CD14 and CD11b. Recently described LSP-1 as a marker distinguishing from monocytes, macrophages, cDCs or DC3 | Mainly express CCR2 and CXC3CR1. | Inflammatory DCs or Tip (TNFα and inducible nitric oxide synthase producing). Produce IL-6, IL-23, IL-1β and TNFα upon activation. Bacterial phagocytosis, iNOS-dependent bacterial killing and tissue toxicity. Depending on the inflammatory stimulus, they induce T helper (Th1), Th17 or Th2 responses. Induce CD4+ T proliferation and cross-present to CD8+ T cells. | [31,38,88,135,136,137,138,139,140,141,142,143,144,145,146,147,148]. | |
mregDC | This population has been found in inflammatory environments, including solid tumors and Crohn’s disease lesions. | This population has been defined as CCR7+ DCs, or “activated DCs” or “mature DCs enriched in immunoregulatory molecules” (mregDCs) with the expression of LAMP3. Regulatory markers CD274, CD200, FAS and ALDH1A2 | Low levels of TLR signaling genes and increased levels of migratory genes the expression of maturation markers (CCR7, CD40, RELB and CD83) | Modulating Th2 response due to the expression of IL4R, IL4I1, CCL17, CCL22 and BCL2L1 genes. In human and mouse non- small cell lung cancers, the mregDC program is expressed by canonical DC1s and DC2s upon uptake of tumor antigens. The role of this new subset is still under exploration both in homeostatic and pathological conditions. | [41,98,122,140,149,150] |
5. DC Crosstalk within the Tumor Microenvironment
5.1. Tumor-Infiltrating DCs Subsets (TIDCs)
5.1.1. Tumor Infiltrating cDC1
5.1.2. cDC2
5.1.3. pDCs
5.1.4. mregDCs
5.1.5. moDCs
5.2. DC Subset Crosstalk: The Driving Force of the Anti-Tumor Response
5.3. Peripheral Blood DC Subsets (PBDCs)
6. Targetability of DCs in Cancer
6.1. Immunogenic Cell Death and DC Activation Induced by Radiotherapy and Chemotherapy
6.2. Inhibitory Molecules Regulating DCs
6.2.1. STAT3 Inhibitors
6.2.2. IDO Inhibitors
6.2.3. Immune Checkpoint Blockade
PD-1/PD-L1 Axis
CTLA-4 Inhibition
6.3. Molecules for DC Activation
6.3.1. TLR Agonists
6.3.2. STING
6.3.3. CD40 Agonist
7. Moving towards a DC—Based Personalized Cancer Therapy
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Flamand, V.; Sornasse, T.; Thielemans, K.; Demanet, C.; Bakkus, M.; Bazin, H.; Tielemans, F.; Leo, O.; Urbain, J.; Moser, M. Murine dendritic cells pulsed in vitro with tumor antigen induce tumor resistance in vivo. Eur. J. Immunol. 1994, 24, 605–610. [Google Scholar] [CrossRef] [PubMed]
- Coulie, P.G.; Van den Eynde, B.J.; van der Bruggen, P.; Boon, T. Tumour antigens recognized by T lymphocytes: At the core of cancer immunotherapy. Nat. Rev. Cancer 2014, 14, 135–146. [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] [PubMed]
- Guilliams, M.; Ginhoux, F.; Jakubzick, C.; Naik, S.H.; Onai, N.; Schraml, B.U.; Segura, E.; Tussiwand, R.; Yona, S. Dendritic cells, monocytes and macrophages: A unified nomenclature based on ontogeny. Nat. Rev. Immunol. 2014, 14, 571–578. [Google Scholar] [CrossRef]
- Ginhoux, F.; Guilliams, M. Tissue-Resident Macrophage Ontogeny and Homeostasis. Immunity 2016, 44, 439–449. [Google Scholar] [CrossRef] [PubMed]
- Naik, S.H.; Metcalf, D.; van Nieuwenhuijze, A.; Wicks, I.; Wu, L.; O’Keeffe, M.; Shortman, K. Intrasplenic steady-state dendritic cell precursors that are distinct from monocytes. Nat. Immunol. 2006, 7, 663–671. [Google Scholar] [CrossRef] [PubMed]
- Varol, C.; Landsman, L.; Fogg, D.K.; Greenshtein, L.; Gildor, B.; Margalit, R.; Kalchenko, V.; Geissmann, F.; Jung, S. Monocytes give rise to mucosal, but not splenic, conventional dendritic cells. J. Exp. Med. 2007, 204, 171–180. [Google Scholar] [CrossRef] [PubMed]
- Onai, N.; Obata-Onai, A.; Schmid, M.A.; Ohteki, T.; Jarrossay, D.; Manz, M.G. Identification of clonogenic common Flt3+M-CSFR+ plasmacytoid and conventional dendritic cell progenitors in mouse bone marrow. Nat. Immunol. 2007, 8, 1207–1216. [Google Scholar] [CrossRef]
- Velten, L.; Haas, S.F.; Raffel, S.; Blaszkiewicz, S.; Islam, S.; Hennig, B.P.; Hirche, C.; Lutz, C.; Buss, E.C.; Nowak, D.; et al. Human haematopoietic stem cell lineage commitment is a continuous process. Nat. Cell Biol. 2017, 19, 271–281. [Google Scholar] [CrossRef]
- Notta, F.; Zandi, S.; Takayama, N.; Dobson, S.; Gan, O.I.; Wilson, G.; Kaufmann, K.B.; McLeod, J.; Laurenti, E.; Dunant, C.F.; et al. Distinct routes of lineage development reshape the human blood hierarchy across ontogeny. Science 2016, 351, aab2116. [Google Scholar] [CrossRef] [Green Version]
- Karamitros, D.; Stoilova, B.; Aboukhalil, Z.; Hamey, F.; Reinisch, A.; Samitsch, M.; Quek, L.; Otto, G.; Repapi, E.; Doondeea, J.; et al. Single-cell analysis reveals the continuum of human lympho-myeloid progenitor cells. Nat. Immunol. 2018, 19, 85–97. [Google Scholar] [CrossRef] [PubMed]
- Li, G.; Hao, W.; Hu, W. Transcription factor PU.1 and immune cell differentiation (Review). Int. J. Mol. Med. 2020, 46, 1943–1950. [Google Scholar] [CrossRef] [PubMed]
- Nutt, S.L.; Metcalf, D.; D’Amico, A.; Polli, M.; Wu, L. Dynamic regulation of PU.1 expression in multipotent hematopoietic progenitors. J. Exp. Med. 2005, 201, 221–231. [Google Scholar] [CrossRef] [PubMed]
- Kanada, S.; Nishiyama, C.; Nakano, N.; Suzuki, R.; Maeda, K.; Hara, M.; Kitamura, N.; Ogawa, H.; Okumura, K. Critical role of transcription factor PU.1 in the expression of CD80 and CD86 on dendritic cells. Blood 2011, 117, 2211–2222. [Google Scholar] [CrossRef]
- Yashiro, T.; Hara, M.; Ogawa, H.; Okumura, K.; Nishiyama, C. Critical Role of Transcription Factor PU.1 in the Function of the OX40L/TNFSF4 Promoter in Dendritic Cells. Sci. Rep. 2016, 6, 34825. [Google Scholar] [CrossRef]
- Zhu, X.J.; Yang, Z.F.; Chen, Y.; Wang, J.; Rosmarin, A.G. PU.1 is essential for CD11c expression in CD8(+)/CD8(-) lymphoid and monocyte-derived dendritic cells during GM-CSF or FLT3L-induced differentiation. PLoS ONE 2012, 7, e52141. [Google Scholar] [CrossRef]
- Zhang, D.E.; Hohaus, S.; Voso, M.T.; Chen, H.M.; Smith, L.T.; Hetherington, C.J.; Tenen, D.G. Function of PU.1 (Spi-1), C/EBP, and AML1 in early myelopoiesis: Regulation of multiple myeloid CSF receptor promoters. Curr. Top. Microbiol. Immunol. 1996, 211, 137–147. [Google Scholar] [CrossRef]
- Swirski, F.K.; Nahrendorf, M.; Etzrodt, M.; Wildgruber, M.; Cortez-Retamozo, V.; Panizzi, P.; Figueiredo, J.L.; Kohler, R.H.; Chudnovskiy, A.; Waterman, P.; et al. Identification of splenic reservoir monocytes and their deployment to inflammatory sites. Science 2009, 325, 612–616. [Google Scholar] [CrossRef]
- Doulatov, S.; Notta, F.; Eppert, K.; Nguyen, L.T.; Ohashi, P.S.; Dick, J.E. Revised map of the human progenitor hierarchy shows the origin of macrophages and dendritic cells in early lymphoid development. Nat. Immunol. 2010, 11, 585–593. [Google Scholar] [CrossRef]
- Kawamura, S.; Onai, N.; Miya, F.; Sato, T.; Tsunoda, T.; Kurabayashi, K.; Yotsumoto, S.; Kuroda, S.; Takenaka, K.; Akashi, K.; et al. Identification of a Human Clonogenic Progenitor with Strict Monocyte Differentiation Potential: A Counterpart of Mouse cMoPs. Immunity 2017, 46, 835–848.e4. [Google Scholar] [CrossRef]
- Ma, O.; Hong, S.; Guo, H.; Ghiaur, G.; Friedman, A.D. Granulopoiesis requires increased C/EBPalpha compared to monopoiesis, correlated with elevated Cebpa in immature G-CSF receptor versus M-CSF receptor expressing cells. PLoS ONE 2014, 9, e95784. [Google Scholar] [CrossRef]
- Kurotaki, D.; Yamamoto, M.; Nishiyama, A.; Uno, K.; Ban, T.; Ichino, M.; Sasaki, H.; Matsunaga, S.; Yoshinari, M.; Ryo, A.; et al. IRF8 inhibits C/EBPalpha activity to restrain mononuclear phagocyte progenitors from differentiating into neutrophils. Nat. Commun. 2014, 5, 4978. [Google Scholar] [CrossRef] [PubMed]
- Kurotaki, D.; Osato, N.; Nishiyama, A.; Yamamoto, M.; Ban, T.; Sato, H.; Nakabayashi, J.; Umehara, M.; Miyake, N.; Matsumoto, N.; et al. Essential role of the IRF8-KLF4 transcription factor cascade in murine monocyte differentiation. Blood 2013, 121, 1839–1849. [Google Scholar] [CrossRef] [PubMed]
- Platzer, B.; Jorgl, A.; Taschner, S.; Hocher, B.; Strobl, H. RelB regulates human dendritic cell subset development by promoting monocyte intermediates. Blood 2004, 104, 3655–3663. [Google Scholar] [CrossRef]
- Chandra, J.; Kuo, P.T.; Hahn, A.M.; Belz, G.T.; Frazer, I.H. Batf3 selectively determines acquisition of CD8(+) dendritic cell phenotype and function. Immunol. Cell. Biol. 2017, 95, 215–223. [Google Scholar] [CrossRef]
- Aliberti, J.; Schulz, O.; Pennington, D.J.; Tsujimura, H.; Reis e Sousa, C.; Ozato, K.; Sher, A. Essential role for ICSBP in the in vivo development of murine CD8alpha + dendritic cells. Blood 2003, 101, 305–310. [Google Scholar] [CrossRef]
- Jackson, J.T.; Hu, Y.; Liu, R.; Masson, F.; D’Amico, A.; Carotta, S.; Xin, A.; Camilleri, M.J.; Mount, A.M.; Kallies, A.; et al. Id2 expression delineates differential checkpoints in the genetic program of CD8α+ and CD103+ dendritic cell lineages. EMBO J. 2011, 30, 2690–2704. [Google Scholar] [CrossRef]
- Meredith, M.M.; Liu, K.; Darrasse-Jeze, G.; Kamphorst, A.O.; Schreiber, H.A.; Guermonprez, P.; Idoyaga, J.; Cheong, C.; Yao, K.H.; Niec, R.E.; et al. Expression of the zinc finger transcription factor zDC (Zbtb46, Btbd4) defines the classical dendritic cell lineage. J. Exp. Med. 2012, 209, 1153–1165. [Google Scholar] [CrossRef]
- McKenna, H.J.; Stocking, K.L.; Miller, R.E.; Brasel, K.; De Smedt, T.; Maraskovsky, E.; Maliszewski, C.R.; Lynch, D.H.; Smith, J.; Pulendran, B.; et al. Mice lacking flt3 ligand have deficient hematopoiesis affecting hematopoietic progenitor cells, dendritic cells, and natural killer cells. Blood 2000, 95, 3489–3497. [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] [Green Version]
- Collin, M.; Bigley, V. Human dendritic cell subsets: An update. Immunology 2018, 154, 3–20. [Google Scholar] [CrossRef] [PubMed]
- Carotta, S.; Dakic, A.; D’Amico, A.; Pang, S.H.M.; Greig, K.T.; Nutt, S.L.; Wu, L. The Transcription Factor PU.1 Controls Dendritic Cell Development and Flt3 Cytokine Receptor Expression in a Dose-Dependent Manner. Immunity 2010, 32, 628–641. [Google Scholar] [CrossRef] [PubMed]
- Chopin, M.; Lun, A.T.; Zhan, Y.; Schreuder, J.; Coughlan, H.; D’Amico, A.; Mielke, L.A.; Almeida, F.F.; Kueh, A.J.; Dickins, R.A.; et al. Transcription Factor PU.1 Promotes Conventional Dendritic Cell Identity and Function via Induction of Transcriptional Regulator DC-SCRIPT. Immunity 2019, 50, 77–90.e75. [Google Scholar] [CrossRef] [PubMed]
- Passlick, B.; Flieger, D.; Ziegler-Heitbrock, H.W. Identification and characterization of a novel monocyte subpopulation in human peripheral blood. Blood 1989, 74, 2527–2534. [Google Scholar] [CrossRef] [PubMed]
- Ziegler-Heitbrock, L.; Ancuta, P.; Crowe, S.; Dalod, M.; Grau, V.; Hart, D.N.; Leenen, P.J.; Liu, Y.J.; MacPherson, G.; Randolph, G.J.; et al. Nomenclature of monocytes and dendritic cells in blood. Blood 2010, 116, e74–e80. [Google Scholar] [CrossRef]
- Ziegler-Heitbrock, L.; Hofer, T.P. Toward a refined definition of monocyte subsets. Front. Immunol. 2013, 4, 23. [Google Scholar] [CrossRef]
- Ziegler-Heitbrock, L. Blood Monocytes and Their Subsets: Established Features and Open Questions. Front. Immunol. 2015, 6, 423. [Google Scholar] [CrossRef]
- Patel, A.A.; Zhang, Y.; Fullerton, J.N.; Boelen, L.; Rongvaux, A.; Maini, A.A.; Bigley, V.; Flavell, R.A.; Gilroy, D.W.; Asquith, B.; et al. The fate and lifespan of human monocyte subsets in steady state and systemic inflammation. J. Exp. Med. 2017, 214, 1913–1923. [Google Scholar] [CrossRef]
- Korkosz, M.; Bukowska-Strakova, K.; Sadis, S.; Grodzicki, T.; Siedlar, M. Monoclonal antibodies against macrophage colony-stimulating factor diminish the number of circulating intermediate and nonclassical (CD14(++)CD16(+)/CD14(+)CD16(++)) monocytes in rheumatoid arthritis patient. Blood 2012, 119, 5329–5330. [Google Scholar] [CrossRef]
- Venneri, M.A.; De Palma, M.; Ponzoni, M.; Pucci, F.; Scielzo, C.; Zonari, E.; Mazzieri, R.; Doglioni, C.; Naldini, L. Identification of proangiogenic TIE2-expressing monocytes (TEMs) in human peripheral blood and cancer. Blood 2007, 109, 5276–5285. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Wong, K.L.; Tai, J.J.; Wong, W.C.; Han, H.; Sem, X.; Yeap, W.H.; Kourilsky, P.; Wong, S.C. Gene expression profiling reveals the defining features of the classical, intermediate, and nonclassical human monocyte subsets. Blood 2011, 118, e16–e31. [Google Scholar] [CrossRef] [PubMed]
- Ozanska, A.; Szymczak, D.; Rybka, J. Pattern of human monocyte subpopulations in health and disease. Scand. J. Immunol. 2020, 92, e12883. [Google Scholar] [CrossRef]
- Zawada, A.M.; Rogacev, K.S.; Rotter, B.; Winter, P.; Marell, R.R.; Fliser, D.; Heine, G.H. SuperSAGE evidence for CD14++CD16+ monocytes as a third monocyte subset. Blood 2011, 118, e50–e61. [Google Scholar] [CrossRef] [PubMed]
- Lund, H.; Boysen, P.; Akesson, C.P.; Lewandowska-Sabat, A.M.; Storset, A.K. Transient Migration of Large Numbers of CD14(++) CD16(+) Monocytes to the Draining Lymph Node after Onset of Inflammation. Front. Immunol. 2016, 7, 322. [Google Scholar] [CrossRef]
- Hasegawa, H.; Matsumoto, T. Mechanisms of Tolerance Induction by Dendritic Cells In Vivo. Front. Immunol. 2018, 9, 350. [Google Scholar] [CrossRef]
- Wakkach, A.; Fournier, N.; Brun, V.; Breittmayer, J.-P.; Cottrez, F.; Groux, H. Characterization of Dendritic Cells that Induce Tolerance and T Regulatory 1 Cell Differentiation In Vivo. Immunity 2003, 18, 605–617. [Google Scholar] [CrossRef]
- Austyn, J.M.; Kupiec-Weglinski, J.W.; Hankins, D.F.; Morris, P.J. Migration patterns of dendritic cells in the mouse. Homing to T cell-dependent areas of spleen, and binding within marginal zone. J. Exp. Med. 1988, 167, 646–651. [Google Scholar] [CrossRef]
- Sozzani, S. Dendritic cell trafficking: More than just chemokines. Cytokine Growth Factor Rev. 2005, 16, 581–592. [Google Scholar] [CrossRef]
- de Winde, C.M.; Munday, C.; Acton, S.E. Molecular mechanisms of dendritic cell migration in immunity and cancer. Med. Microbiol. and Immunol. 2020, 209, 515–529. [Google Scholar] [CrossRef]
- Alvarez, D.; Vollmann, E.H.; von Andrian, U.H. Mechanisms and consequences of dendritic cell migration. Immunity 2008, 29, 325–342. [Google Scholar] [CrossRef]
- Andreae, S.; Piras, F.; Burdin, N.; Triebel, F. Maturation and Activation of Dendritic Cells Induced by Lymphocyte Activation Gene-3 (CD223). J. Immunol. 2002, 168, 3874–3880. [Google Scholar] [CrossRef]
- Vargas, P.; Maiuri, P.; Bretou, M.; Sáez, P.J.; Pierobon, P.; Maurin, M.; Chabaud, M.; Lankar, D.; Obino, D.; Terriac, E.; et al. Innate control of actin nucleation determines two distinct migration behaviours in dendritic cells. Nat. Cell Biol. 2016, 18, 43–53. [Google Scholar] [CrossRef] [PubMed]
- Worbs, T.; Hammerschmidt, S.I.; Förster, R. Dendritic cell migration in health and disease. Nat. Rev. Immunol. 2017, 17, 30–48. [Google Scholar] [CrossRef]
- Lukacs-Kornek, V.; Engel, D.; Tacke, F.; Kurts, C. The role of chemokines and their receptors in dendritic cell biology. Front. Biosci. 2008, 13, 2238–2252. [Google Scholar] [CrossRef] [PubMed]
- Del Prete, A.; Shao, W.H.; Mitola, S.; Santoro, G.; Sozzani, S.; Haribabu, B. Regulation of dendritic cell migration and adaptive immune response by leukotriene B4 receptors: A role for LTB4 in up-regulation of CCR7 expression and function. Blood 2007, 109, 626–631. [Google Scholar] [CrossRef] [PubMed]
- Sadik, C.D.; Luster, A.D. Lipid-cytokine-chemokine cascades orchestrate leukocyte recruitment in inflammation. J. Leukoc. Biol. 2012, 91, 207–215. [Google Scholar] [CrossRef]
- Majumdar, R.; Sixt, M.; Parent, C.A. New paradigms in the establishment and maintenance of gradients during directed cell migration. Curr. Opin. Cell Biol. 2014, 30, 33–40. [Google Scholar] [CrossRef]
- Tiberio, L.; Del Prete, A.; Schioppa, T.; Sozio, F.; Bosisio, D.; Sozzani, S. Chemokine and chemotactic signals in dendritic cell migration. Cell Mol. Immunol 2018, 15, 346–352. [Google Scholar] [CrossRef] [PubMed]
- Eksioglu, E.A.; Eisen, S.; Reddy, V. Dendritic cells as therapeutic agents against cancer. Front. Biosci. (Landmark Ed.) 2010, 15, 321–347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kotsias, F.; Cebrian, I.; Alloatti, A. Chapter Two—Antigen processing and presentation. In International Review of Cell and Molecular Biology; Lhuillier, C., Galluzzi, L., Eds.; Academic Press: Cambridge, MA, USA, 2019; Volume 348, pp. 69–121. [Google Scholar]
- Lin, M.L.; Zhan, Y.; Villadangos, J.A.; Lew, A.M. The cell biology of cross-presentation and the role of dendritic cell subsets. Immunol. Cell Biol. 2008, 86, 353–362. [Google Scholar] [CrossRef] [PubMed]
- Rosalia, R.A.; Quakkelaar, E.D.; Redeker, A.; Khan, S.; Camps, M.; Drijfhout, J.W.; Silva, A.L.; Jiskoot, W.; van Hall, T.; van Veelen, P.A.; et al. Dendritic cells process synthetic long peptides better than whole protein, improving antigen presentation and T-cell activation. Eur. J. Immunol. 2013, 43, 2554–2565. [Google Scholar] [CrossRef] [PubMed]
- Carenza, C.; Calcaterra, F.; Oriolo, F.; Di Vito, C.; Ubezio, M.; Della Porta, M.G.; Mavilio, D.; Della Bella, S. Costimulatory Molecules and Immune Checkpoints Are Differentially Expressed on Different Subsets of Dendritic Cells. Front. Immunol. 2019, 10, 1325. [Google Scholar] [CrossRef]
- Fujii, T.; Naing, A.; Rolfo, C.; Hajjar, J. Biomarkers of response to immune checkpoint blockade in cancer treatment. Crit. Rev. Oncol. Hematol. 2018, 130, 108–120. [Google Scholar] [CrossRef] [PubMed]
- Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef]
- Waisman, A.; Lukas, D.; Clausen, B.E.; Yogev, N. Dendritic cells as gatekeepers of tolerance. Semin. Immunopathol. 2017, 39, 153–163. [Google Scholar] [CrossRef]
- Helft, J.; Anjos-Afonso, F.; van der Veen, A.G.; Chakravarty, P.; Bonnet, D.; Reis e Sousa, C. Dendritic Cell Lineage Potential in Human Early Hematopoietic Progenitors. Cell Rep. 2017, 20, 529–537. [Google Scholar] [CrossRef]
- Palucka, K.; Banchereau, J. Dendritic-cell-based therapeutic cancer vaccines. Immunity 2013, 39, 38–48. [Google Scholar] [CrossRef]
- Nutt, S.L.; Chopin, M. Transcriptional Networks Driving Dendritic Cell Differentiation and Function. Immunity 2020, 52, 942–956. [Google Scholar] [CrossRef]
- Alcántara-Hernández, M.; Leylek, R.; Wagar, L.E.; Engleman, E.G.; Keler, T.; Marinkovich, M.P.; Davis, M.M.; Nolan, G.P.; Idoyaga, J. High-Dimensional Phenotypic Mapping of Human Dendritic Cells Reveals Interindividual Variation and Tissue Specialization. Immunity 2017, 47, 1037–1050.e6. [Google Scholar] [CrossRef] [Green Version]
- Guilliams, M.; Dutertre, C.A.; Scott, C.L.; McGovern, N.; Sichien, D.; Chakarov, S.; Van Gassen, S.; Chen, J.; Poidinger, M.; De Prijck, S.; et al. Unsupervised High-Dimensional Analysis Aligns Dendritic Cells across Tissues and Species. Immunity 2016, 45, 669–684. [Google Scholar] [CrossRef]
- Worah, K.; Mathan, T.S.M.; Vu Manh, T.P.; Keerthikumar, S.; Schreibelt, G.; Tel, J.; Duiveman-de Boer, T.; Sköld, A.E.; van Spriel, A.B.; de Vries, I.J.M.; et al. Proteomics of Human Dendritic Cell Subsets Reveals Subset-Specific Surface Markers and Differential Inflammasome Function. Cell Rep. 2016, 16, 2953–2966. [Google Scholar] [CrossRef]
- Heidkamp, G.F.; Sander, J.; Lehmann, C.H.K.; Heger, L.; Eissing, N.; Baranska, A.; Lühr, J.J.; Hoffmann, A.; Reimer, K.C.; Lux, A.; et al. Human lymphoid organ dendritic cell identity is predominantly dictated by ontogeny, not tissue microenvironment. Sci. Immunol. 2016, 1, eaai7677. [Google Scholar] [CrossRef] [PubMed]
- Dutertre, C.A.; Becht, E.; Irac, S.E.; Khalilnezhad, A.; Narang, V.; Khalilnezhad, S.; Ng, P.Y.; van den Hoogen, L.L.; Leong, J.Y.; Lee, B.; et al. Single-Cell Analysis of Human Mononuclear Phagocytes Reveals Subset-Defining Markers and Identifies Circulating Inflammatory Dendritic Cells. Immunity 2019, 51, 573–589.e8. [Google Scholar] [CrossRef] [PubMed]
- Calzetti, F.; Tamassia, N.; Micheletti, A.; Finotti, G.; Bianchetto-Aguilera, F.; Cassatella, M.A. Human dendritic cell subset 4 (DC4) correlates to a subset of CD14(dim/-)CD16(++) monocytes. J. Allergy Clin. Immunol. 2018, 141, 2276–2279.e2273. [Google Scholar] [CrossRef] [PubMed]
- See, P.; Dutertre, C.A.; Chen, J.; Günther, P.; McGovern, N.; Irac, S.E.; Gunawan, M.; Beyer, M.; Händler, K.; Duan, K.; et al. Mapping the human DC lineage through the integration of high-dimensional techniques. Science 2017, 356, eaag3009. [Google Scholar] [CrossRef] [PubMed]
- Balan, S.; Arnold-Schrauf, C.; Abbas, A.; Couespel, N.; Savoret, J.; Imperatore, F.; Villani, A.C.; Vu Manh, T.P.; Bhardwaj, N.; Dalod, M. Large-Scale Human Dendritic Cell Differentiation Revealing Notch-Dependent Lineage Bifurcation and Heterogeneity. Cell Rep. 2018, 24, 1902–1915.e6. [Google Scholar] [CrossRef] [PubMed]
- Chu, C.C.; Ali, N.; Karagiannis, P.; Di Meglio, P.; Skowera, A.; Napolitano, L.; Barinaga, G.; Grys, K.; Sharif-Paghaleh, E.; Karagiannis, S.N.; et al. Resident CD141 (BDCA3)+ dendritic cells in human skin produce IL-10 and induce regulatory T cells that suppress skin inflammation. J. Exp. Med. 2012, 209, 935–945. [Google Scholar] [CrossRef]
- Haniffa, M.; Shin, A.; Bigley, V.; McGovern, N.; Teo, P.; See, P.; Wasan, P.S.; Wang, X.N.; Malinarich, F.; Malleret, B.; et al. Human tissues contain CD141hi cross-presenting dendritic cells with functional homology to mouse CD103+ nonlymphoid dendritic cells. Immunity 2012, 37, 60–73. [Google Scholar] [CrossRef]
- Granot, T.; Senda, T.; Carpenter, D.J.; Matsuoka, N.; Weiner, J.; Gordon, C.L.; Miron, M.; Kumar, B.V.; Griesemer, A.; Ho, S.H.; et al. Dendritic Cells Display Subset and Tissue-Specific Maturation Dynamics over Human Life. Immunity 2017, 46, 504–515. [Google Scholar] [CrossRef] [Green Version]
- Gerner, M.Y.; Casey, K.A.; Kastenmuller, W.; Germain, R.N. Dendritic cell and antigen dispersal landscapes regulate T cell immunity. J. Exp. Med. 2017, 214, 3105–3122. [Google Scholar] [CrossRef] [PubMed]
- Mair, F.; Liechti, T. Comprehensive Phenotyping of Human Dendritic Cells and Monocytes. Cytom. Part. A 2021, 99, 231–242. [Google Scholar] [CrossRef] [PubMed]
- Cao, L.; Chang, H.; Shi, X.; Peng, C.; He, Y. Keratin mediates the recognition of apoptotic and necrotic cells through dendritic cell receptor DEC205/CD205. Proc. Natl. Acad. Sci. USA 2016, 113, 13438–13443. [Google Scholar] [CrossRef]
- Hossain, M.K.; Wall, K.A. Use of Dendritic Cell Receptors as Targets for Enhancing Anti-Cancer Immune Responses. Cancers 2019, 11, 418. [Google Scholar] [CrossRef] [PubMed]
- Bachem, A.; Güttler, 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]
- Crozat, K.; Guiton, R.; Guilliams, M.; Henri, S.; Baranek, T.; Schwartz-Cornil, I.; Malissen, B.; Dalod, M. Comparative genomics as a tool to reveal functional equivalences between human and mouse dendritic cell subsets. Immunol. Rev. 2010, 234, 177–198. [Google Scholar] [CrossRef] [PubMed]
- Matsuo, K.; Yoshie, O.; Kitahata, K.; Kamei, M.; Hara, Y.; Nakayama, T. Recent Progress in Dendritic Cell-Based Cancer Immunotherapy. Cancers 2021, 13, 2495. [Google Scholar] [CrossRef] [PubMed]
- Chevalier, M.F.; Bohner, P.; Pieraerts, C.; Lhermitte, B.; Gourmaud, J.; Nobile, A.; Rotman, S.; Cesson, V.; Martin, V.; Legris, A.S.; et al. Immunoregulation of Dendritic Cell Subsets by Inhibitory Receptors in Urothelial Cancer. Eur. Urol. 2017, 71, 854–857. [Google Scholar] [CrossRef] [PubMed]
- Murphy, T.L.; Grajales-Reyes, G.E.; Wu, X.; Tussiwand, R.; Briseño, C.G.; Iwata, A.; Kretzer, N.M.; Durai, V.; Murphy, K.M. Transcriptional Control of Dendritic Cell Development. Annu. Rev. Immunol. 2016, 34, 93–119. [Google Scholar] [CrossRef]
- Yu, C.I.; Becker, C.; Metang, P.; Marches, F.; Wang, Y.; Toshiyuki, H.; Banchereau, J.; Merad, M.; Palucka, A.K. Human CD141+ dendritic cells induce CD4+ T cells to produce type 2 cytokines. J. Immunol. 2014, 193, 4335–4343. [Google Scholar] [CrossRef] [Green Version]
- Böttcher, 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]
- Gutiérrez-Martínez, E.; Planès, R.; Anselmi, G.; Reynolds, M.; Menezes, S.; Adiko, A.C.; Saveanu, L.; Guermonprez, P. Cross-Presentation of Cell-Associated Antigens by MHC Class I in Dendritic Cell Subsets. Front. Immunol. 2015, 6, 363. [Google Scholar] [CrossRef] [PubMed]
- Martínez-López, M.; Iborra, S.; Conde-Garrosa, R.; Sancho, D. Batf3-dependent CD103+ dendritic cells are major producers of IL-12 that drive local Th1 immunity against Leishmania major infection in mice. Eur. J. Immunol. 2015, 45, 119–129. [Google Scholar] [CrossRef] [PubMed]
- Schreibelt, G.; Klinkenberg, L.J.J.; Cruz, L.J.; Tacken, P.J.; Tel, J.; Kreutz, M.; Adema, G.J.; Brown, G.D.; Figdor, C.G.; de Vries, I.J.M. The C-type lectin receptor CLEC9A mediates antigen uptake and (cross-)presentation by human blood BDCA3+ myeloid dendritic cells. Blood 2012, 119, 2284–2292. [Google Scholar] [CrossRef] [PubMed]
- Theisen, D.; Murphy, K. The role of cDC1s in vivo: CD8 T cell priming through cross-presentation. F1000Research 2017, 6, 98. [Google Scholar] [CrossRef] [PubMed]
- Durai, V.; Murphy, K.M. Functions of Murine Dendritic Cells. Immunity 2016, 45, 719–736. [Google Scholar] [CrossRef] [PubMed]
- Brown, C.C.; Gudjonson, H.; Pritykin, Y.; Deep, D.; Lavallée, 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]
- Schlitzer, A.; Sivakamasundari, V.; Chen, J.; Sumatoh, H.R.; Schreuder, J.; Lum, J.; Malleret, B.; Zhang, S.; Larbi, A.; Zolezzi, F.; et al. Identification of cDC1- and cDC2-committed DC progenitors reveals early lineage priming at the common DC progenitor stage in the bone marrow. Nat. Immunol. 2015, 16, 718–728. [Google Scholar] [CrossRef]
- Bourdely, P.; Anselmi, G.; Vaivode, K.; Ramos, R.N.; Missolo-Koussou, Y.; Hidalgo, S.; Tosselo, J.; Nuñez, N.; Richer, W.; Vincent-Salomon, A.; et al. Transcriptional and Functional Analysis of CD1c(+) Human Dendritic Cells Identifies a CD163(+) Subset Priming CD8(+)CD103(+) T Cells. Immunity 2020, 53, 335–352.e8. [Google Scholar] [CrossRef]
- 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. 2020, 11, 606805. [Google Scholar] [CrossRef]
- Binnewies, M.; Mujal, A.M.; Pollack, J.L.; Combes, A.J.; Hardison, E.A.; Barry, K.C.; Tsui, J.; Ruhland, M.K.; Kersten, K.; Abushawish, M.A.; et al. Unleashing Type-2 Dendritic Cells to Drive Protective Antitumor CD4(+) T Cell Immunity. Cell 2019, 177, 556–571.e16. [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]
- Gerner, M.Y.; Torabi-Parizi, P.; Germain, R.N. Strategically localized dendritic cells promote rapid T cell responses to lymph-borne particulate antigens. Immunity 2015, 42, 172–185. [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]
- Nizzoli, G.; Krietsch, J.; Weick, A.; Steinfelder, S.; Facciotti, F.; Gruarin, P.; Bianco, A.; Steckel, B.; Moro, M.; Crosti, M.; et al. Human CD1c+ dendritic cells secrete high levels of IL-12 and potently prime cytotoxic T-cell responses. Blood 2013, 122, 932–942. [Google Scholar] [CrossRef]
- Durai, V.; Bagadia, P.; Granja, J.M.; Satpathy, A.T.; Kulkarni, D.H.; Davidson, J.T.; Wu, R.; Patel, S.J.; Iwata, A.; Liu, T.-T.; et al. Cryptic activation of an Irf8 enhancer governs cDC1 fate specification. Nature Immunology 2019, 20, 1161–1173. [Google Scholar] [CrossRef] [PubMed]
- Persson, E.K.; Uronen-Hansson, H.; Semmrich, M.; Rivollier, A.; Hägerbrand, K.; Marsal, J.; Gudjonsson, S.; Håkansson, 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]
- Vander Lugt, B.; Khan, A.A.; Hackney, J.A.; Agrawal, S.; Lesch, J.; Zhou, M.; Lee, W.P.; Park, S.; Xu, M.; DeVoss, J.; et al. Transcriptional programming of dendritic cells for enhanced MHC class II antigen presentation. Nat. Immunol. 2014, 15, 161–167. [Google Scholar] [CrossRef]
- Yin, X.; Yu, H.; Jin, X.; Li, J.; Guo, H.; Shi, Q.; Yin, Z.; Xu, Y.; Wang, X.; Liu, R.; et al. Human Blood CD1c+ Dendritic Cells Encompass CD5high and CD5low Subsets That Differ Significantly in Phenotype, Gene Expression, and Functions. J. Immunol. 2017, 198, 1553–1564. [Google Scholar] [CrossRef]
- Bakdash, G.; Buschow, S.I.; Gorris, M.A.; Halilovic, A.; Hato, S.V.; Sköld, A.E.; Schreibelt, G.; Sittig, S.P.; Torensma, R.; Duiveman-de Boer, T.; et al. Expansion of a BDCA1+CD14+ Myeloid Cell Population in Melanoma Patients May Attenuate the Efficacy of Dendritic Cell Vaccines. Cancer Res. 2016, 76, 4332–4346. [Google Scholar] [CrossRef] [Green Version]
- Cytlak, U.; Resteu, A.; Pagan, S.; Green, K.; Milne, P.; Maisuria, S.; McDonald, D.; Hulme, G.; Filby, A.; Carpenter, B.; et al. Differential IRF8 Transcription Factor Requirement Defines Two Pathways of Dendritic Cell Development in Humans. Immunity 2020, 53, 353–370.e8. [Google Scholar] [CrossRef]
- Heger, L.; Balk, S.; Lühr, J.J.; Heidkamp, G.F.; Lehmann, C.H.K.; Hatscher, L.; Purbojo, A.; Hartmann, A.; Garcia-Martin, F.; Nishimura, S.-I.; et al. CLEC10A Is a Specific Marker for Human CD1c+ Dendritic Cells and Enhances Their Toll-Like Receptor 7/8-Induced Cytokine Secretion. Front. Immunol. 2018, 9, 744. [Google Scholar] [CrossRef] [PubMed]
- Villar, J.; Segura, E. Decoding the Heterogeneity of Human Dendritic Cell Subsets. Trends Immunol. 2020, 41, 1062–1071. [Google Scholar] [CrossRef]
- Nakamizo, S.; Dutertre, C.-A.; Khalilnezhad, A.; Zhang, X.M.; Lim, S.; Lum, J.; Koh, G.; Foong, C.; Yong, P.J.A.; Tan, K.J.; et al. Single-cell analysis of human skin identifies CD14+ type 3 dendritic cells co-producing IL1B and IL23A in psoriasis. J. Exp. Med. 2021, 218, e20202345. [Google Scholar] [CrossRef]
- MacDonald, K.P.A.; Munster, D.J.; Clark, G.J.; Dzionek, A.; Schmitz, J.; Hart, D.N.J. Characterization of human blood dendritic cell subsets. Blood 2002, 100, 4512–4520. [Google Scholar] [CrossRef] [PubMed]
- Clark, G.J.; Silveira, P.A.; Hogarth, P.M.; Hart, D.N.J. The cell surface phenotype of human dendritic cells. Semin. Cell Dev. Biol. 2019, 86, 3–14. [Google Scholar] [CrossRef]
- van Leeuwen-Kerkhoff, N.; Lundberg, K.; Westers, T.M.; Kordasti, S.; Bontkes, H.J.; de Gruijl, T.D.; Lindstedt, M.; van de Loosdrecht, A.A. Transcriptional profiling reveals functional dichotomy between human slan(+) non-classical monocytes and myeloid dendritic cells. J. Leukoc. Biol. 2017, 102, 1055–1068. [Google Scholar] [CrossRef] [PubMed]
- Zhang, H.; Gregorio, J.D.; Iwahori, T.; Zhang, X.; Choi, O.; Tolentino, L.L.; Prestwood, T.; Carmi, Y.; Engleman, E.G. A distinct subset of plasmacytoid dendritic cells induces activation and differentiation of B and T lymphocytes. Proc. Natl. Acad. Sci. USA 2017, 114, 1988–1993. [Google Scholar] [CrossRef]
- Li, G.; Cheng, L.; Su, L. Phenotypic and Functional Study of Human Plasmacytoid Dendritic Cells. Curr. Protoc. 2021, 1, e50. [Google Scholar] [CrossRef]
- Reizis, B. Plasmacytoid Dendritic Cells: Development, Regulation, and Function. Immunity 2019, 50, 37–50. [Google Scholar] [CrossRef] [Green Version]
- Rhodes, J.W.; Tong, O.; Harman, A.N.; Turville, S.G. Human Dendritic Cell Subsets, Ontogeny, and Impact on HIV Infection. Front. Immunol. 2019, 10, 1088. [Google Scholar] [CrossRef]
- Dzionek, A.; Fuchs, A.; Schmidt, P.; Cremer, S.; Zysk, M.; Miltenyi, S.; Buck, D.W.; Schmitz, J. BDCA-2, BDCA-3, and BDCA-4: Three markers for distinct subsets of dendritic cells in human peripheral blood. J. Immunol. 2000, 165, 6037–6046. [Google Scholar] [CrossRef] [PubMed]
- Summers, K.L.; Hock, B.D.; McKenzie, J.L.; Hart, D.N. Phenotypic characterization of five dendritic cell subsets in human tonsils. Am. J. Pathol. 2001, 159, 285–295. [Google Scholar] [CrossRef]
- Strobl, H.; Scheinecker, C.; Riedl, E.; Csmarits, B.; Bello-Fernandez, C.; Pickl, W.F.; Majdic, O.; Knapp, W. Identification of CD68+lin- peripheral blood cells with dendritic precursor characteristics. J. Immunol. 1998, 161, 740–748. [Google Scholar] [PubMed]
- Liu, Y.J. IPC: Professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursors. Annu. Rev. Immunol. 2005, 23, 275–306. [Google Scholar] [CrossRef] [PubMed]
- Perussia, B.; Fanning, V.; Trinchieri, G. A leukocyte subset bearing HLA-DR antigens is responsible for in vitro alpha interferon production in response to viruses. Nat. Immun. Cell Growth Regul. 1985, 4, 120–137. [Google Scholar]
- Siegal, F.P.; Kadowaki, N.; Shodell, M.; Fitzgerald-Bocarsly, P.A.; Shah, K.; Ho, S.; Antonenko, S.; Liu, Y.J. The nature of the principal type 1 interferon-producing cells in human blood. Science 1999, 284, 1835–1837. [Google Scholar] [CrossRef]
- Asselin-Paturel, C.; Trinchieri, G. Production of type I interferons: Plasmacytoid dendritic cells and beyond. J. Exp. Med. 2005, 202, 461–465. [Google Scholar] [CrossRef]
- Grouard, G.; Rissoan, M.C.; Filgueira, L.; Durand, I.; Banchereau, J.; Liu, Y.J. The enigmatic plasmacytoid T cells develop into dendritic cells with interleukin (IL)-3 and CD40-ligand. J. Exp. Med. 1997, 185, 1101–1111. [Google Scholar] [CrossRef]
- Tel, J.; Schreibelt, G.; Sittig, S.P.; Mathan, T.S.; Buschow, S.I.; Cruz, L.J.; Lambeck, A.J.; Figdor, C.G.; de Vries, I.J. Human plasmacytoid dendritic cells efficiently cross-present exogenous Ags to CD8+ T cells despite lower Ag uptake than myeloid dendritic cell subsets. Blood 2013, 121, 459–467. [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]
- Guéry, L.; Hugues, S. Tolerogenic and activatory plasmacytoid dendritic cells in autoimmunity. Front. Immunol. 2013, 4, 59. [Google Scholar] [CrossRef] [PubMed]
- Fu, C.; Peng, P.; Loschko, J.; Feng, L.; Pham, P.; Cui, W.; Lee, K.P.; Krug, A.B.; Jiang, A. Plasmacytoid dendritic cells cross-prime naive CD8 T cells by transferring antigen to conventional dendritic cells through exosomes. Proc. Natl. Acad. Sci. USA 2020, 117, 23730–23741. [Google Scholar] [CrossRef]
- 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]
- Chometon, T.Q.; Siqueira, M.D.S.; Sant Anna, J.C.; Almeida, M.R.; Gandini, M.; Martins de Almeida Nogueira, A.C.; Antas, P.R.Z. A protocol for rapid monocyte isolation and generation of singular human monocyte-derived dendritic cells. PLoS ONE 2020, 15, e0231132. [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]
- 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]
- Tang-Huau, T.L.; Segura, E. Human in vivo-differentiated monocyte-derived dendritic cells. Semin. Cell Dev. Biol. 2019, 86, 44–49. [Google Scholar] [CrossRef]
- Tang-Huau, T.-L.; Gueguen, P.; Goudot, C.; Durand, M.; Bohec, M.; Baulande, S.; Pasquier, B.; Amigorena, S.; Segura, E. Human in vivo-generated monocyte-derived dendritic cells and macrophages cross-present antigens through a vacuolar pathway. Nat. Commun. 2018, 9, 2570. [Google Scholar] [CrossRef]
- Zilionis, R.; Engblom, C.; Pfirschke, C.; Savova, V.; Zemmour, D.; Saatcioglu, H.D.; Krishnan, I.; Maroni, G.; Meyerovitz, C.V.; Kerwin, C.M.; et al. Single-Cell Transcriptomics of Human and Mouse Lung Cancers Reveals Conserved Myeloid Populations across Individuals and Species. Immunity 2019, 50, 1317–1334.e10. [Google Scholar] [CrossRef]
- Moutel, S.; Beugnet, A.; Schneider, A.; Lombard, B.; Loew, D.; Amigorena, S.; Perez, F.; Segura, E. Surface LSP-1 Is a Phenotypic Marker Distinguishing Human Classical versus Monocyte-Derived Dendritic Cells. iScience 2020, 23, 100987. [Google Scholar] [CrossRef] [PubMed]
- Rydström, A.; Wick, M.J. Monocyte recruitment, activation, and function in the gut-associated lymphoid tissue during oral Salmonella infection. J. Immunol. 2007, 178, 5789–5801. [Google Scholar] [CrossRef] [PubMed]
- Guilliams, M.; Movahedi, K.; Bosschaerts, T.; VandenDriessche, T.; Chuah, M.K.; Hérin, M.; Acosta-Sanchez, A.; Ma, L.; Moser, M.; Van Ginderachter, J.A.; et al. IL-10 Dampens TNF/Inducible Nitric Oxide Synthase-Producing Dendritic Cell-Mediated Pathogenicity during Parasitic Infection. J. Immunol. 2009, 182, 1107–1118. [Google Scholar] [CrossRef] [PubMed]
- Gu, F.F.; Wu, J.J.; Liu, Y.Y.; Hu, Y.; Liang, J.Y.; Zhang, K.; Li, M.; Wang, Y.; Zhang, Y.A.; Liu, L. Human inflammatory dendritic cells in malignant pleural effusions induce Th1 cell differentiation. Cancer Immunol. Immunother. 2020, 69, 779–788. [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] [PubMed]
- Hohl, T.M.; Rivera, A.; Lipuma, L.; Gallegos, A.; Shi, C.; Mack, M.; Pamer, E.G. Inflammatory monocytes facilitate adaptive CD4 T cell responses during respiratory fungal infection. Cell Host Microbe. 2009, 6, 470–481. [Google Scholar] [CrossRef]
- Le Borgne, M.; Etchart, N.; Goubier, A.; Lira, S.A.; Sirard, J.C.; van Rooijen, N.; Caux, C.; Aït-Yahia, S.; Vicari, A.; Kaiserlian, D.; et al. Dendritic cells rapidly recruited into epithelial tissues via CCR6/CCL20 are responsible for CD8+ T cell crosspriming in vivo. Immunity 2006, 24, 191–201. [Google Scholar] [CrossRef]
- Maier, B.; Leader, A.M.; Chen, S.T.; Tung, N.; Chang, C.; LeBerichel, J.; Chudnovskiy, A.; Maskey, S.; Walker, L.; Finnigan, J.P.; et al. A conserved dendritic-cell regulatory program limits antitumour immunity. Nature 2020, 580, 257–262. [Google Scholar] [CrossRef]
- Martin, J.C.; Chang, C.; Boschetti, G.; Ungaro, R.; Giri, M.; Grout, J.A.; Gettler, K.; Chuang, L.S.; Nayar, S.; Greenstein, A.J.; et al. Single-Cell Analysis of Crohn’s Disease Lesions Identifies a Pathogenic Cellular Module Associated with Resistance to Anti-TNF Therapy. Cell 2019, 178, 1493–1508.e20. [Google Scholar] [CrossRef]
- Labani-Motlagh, A.; Ashja-Mahdavi, M.; Loskog, A. The Tumor Microenvironment: A Milieu Hindering and Obstructing Antitumor Immune Responses. Front. Immunol. 2020, 11, 940. [Google Scholar] [CrossRef]
- 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]
- Hubert, M.; Gobbini, E.; Bendriss-Vermare, N.; Caux, C.; Valladeau-Guilemond, J. Human Tumor-Infiltrating Dendritic Cells: From in Situ Visualization to High-Dimensional Analyses. Cancers 2019, 11, 1082. [Google Scholar] [CrossRef]
- Karthaus, N.; Torensma, R.; Tel, J. Deciphering the message broadcast by tumor-infiltrating dendritic cells. Am. J. Pathol. 2012, 181, 733–742. [Google Scholar] [CrossRef] [PubMed]
- Conejo-Garcia, J.R.; Rutkowski, M.R.; Cubillos-Ruiz, J.R. State-of-the-art of regulatory dendritic cells in cancer. Pharmacol. Ther. 2016, 164, 97–104. [Google Scholar] [CrossRef]
- Veglia, F.; Gabrilovich, D.I. Dendritic cells in cancer: The role revisited. Curr. Opin. Immunol. 2017, 45, 43–51. [Google Scholar] [CrossRef]
- Torres-Aguilar, H.; Aguilar-Ruiz, S.R.; González-Pérez, G.; Munguía, R.; Bajaña, S.; Meraz-Ríos, M.A.; Sánchez-Torres, C. Tolerogenic dendritic cells generated with different immunosuppressive cytokines induce antigen-specific anergy and regulatory properties in memory CD4+ T cells. J. Immunol. 2010, 184, 1765–1775. [Google Scholar] [CrossRef]
- Ghiringhelli, F.; Puig, P.E.; Roux, S.; Parcellier, A.; Schmitt, E.; Solary, E.; Kroemer, G.; Martin, F.; Chauffert, B.; Zitvogel, L. Tumor cells convert immature myeloid dendritic cells into TGF-beta-secreting cells inducing CD4+CD25+ regulatory T cell proliferation. J. Exp. Med. 2005, 202, 919–929. [Google Scholar] [CrossRef]
- Lutz, M.B.; Schuler, G. Immature, semi-mature and fully mature dendritic cells: Which signals induce tolerance or immunity? Trends Immunol. 2002, 23, 445–449. [Google Scholar] [CrossRef]
- Munn, D.H.; Sharma, M.D.; Lee, J.R.; Jhaver, K.G.; Johnson, T.S.; Keskin, D.B.; Marshall, B.; Chandler, P.; Antonia, S.J.; Burgess, R.; et al. Potential regulatory function of human dendritic cells expressing indoleamine 2,3-dioxygenase. Science 2002, 297, 1867–1870. [Google Scholar] [CrossRef]
- Zhu, S.; Yang, N.; Wu, J.; Wang, X.; Wang, W.; Liu, Y.-J.; Chen, J. Tumor microenvironment-related dendritic cell deficiency: A target to enhance tumor immunotherapy. Pharmacol. Res. 2020, 159, 104980. [Google Scholar] [CrossRef]
- Jochems, C.; Schlom, J. Tumor-infiltrating immune cells and prognosis: The potential link between conventional cancer therapy and immunity. Exp. Biol. Med. (Maywood) 2011, 236, 567–579. [Google Scholar] [CrossRef] [Green Version]
- Chang, W.J.; Du, Y.; Zhao, X.; Ma, L.Y.; Cao, G.W. Inflammation-related factors predicting prognosis of gastric cancer. World J. Gastroenterol. 2014, 20, 4586–4596. [Google Scholar] [CrossRef] [PubMed]
- Ma, Y.; Shurin, G.V.; Peiyuan, Z.; Shurin, M.R. Dendritic cells in the cancer microenvironment. J. Cancer 2013, 4, 36–44. [Google Scholar] [CrossRef] [PubMed]
- Michea, P.; Noël, F.; Zakine, E.; Czerwinska, U.; Sirven, P.; Abouzid, O.; Goudot, C.; Scholer-Dahirel, A.; Vincent-Salomon, A.; Reyal, F.; et al. Adjustment of dendritic cells to the breast-cancer microenvironment is subset specific. Nat. Immunol. 2018, 19, 885–897. [Google Scholar] [CrossRef] [PubMed]
- Tran Janco, J.M.; Lamichhane, P.; Karyampudi, L.; Knutson, K.L. Tumor-infiltrating dendritic cells in cancer pathogenesis. J. Immunol. 2015, 194, 2985–2991. [Google Scholar] [CrossRef]
- Mastelic-Gavillet, B.; Sarivalasis, A.; Lozano, L.E.; Wyss, T.; Inoges, S.; de Vries, I.J.M.; Dartiguenave, F.; Jichlinski, P.; Derrè, L.; Coukos, G.; et al. Quantitative and qualitative impairments in dendritic cell subsets of patients with ovarian or prostate cancer. Eur. J. Cancer 2020, 135, 173–182. [Google Scholar] [CrossRef] [PubMed]
- Santegoets, S.J.; Stam, A.G.; Lougheed, S.M.; Gall, H.; Jooss, K.; Sacks, N.; Hege, K.; Lowy, I.; Scheper, R.J.; Gerritsen, W.R.; et al. Myeloid derived suppressor and dendritic cell subsets are related to clinical outcome in prostate cancer patients treated with prostate GVAX and ipilimumab. J. Immunother. Cancer 2014, 2, 31. [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]
- Hammerich, L.; Marron, T.U.; Upadhyay, R.; Svensson-Arvelund, J.; Dhainaut, M.; Hussein, S.; Zhan, Y.; Ostrowski, D.; Yellin, M.; Marsh, H.; et al. Systemic clinical tumor regressions and potentiation of PD1 blockade with in situ vaccination. Nat. Med. 2019, 25, 814–824. [Google Scholar] [CrossRef]
- Salmon, H.; Idoyaga, J.; Rahman, A.; Leboeuf, M.; Remark, R.; Jordan, S.; Casanova-Acebes, M.; Khudoynazarova, M.; Agudo, J.; Tung, N.; et al. Expansion and Activation of CD103(+) Dendritic Cell Progenitors at the Tumor Site Enhances Tumor Responses to Therapeutic PD-L1 and BRAF Inhibition. Immunity 2016, 44, 924–938. [Google Scholar] [CrossRef]
- Sandel, M.H.; Dadabayev, A.R.; Menon, A.G.; Morreau, H.; Melief, C.J.; Offringa, R.; van der Burg, S.H.; Janssen-van Rhijn, C.M.; Ensink, N.G.; Tollenaar, R.A.; et al. Prognostic value of tumor-infiltrating dendritic cells in colorectal cancer: Role of maturation status and intratumoral localization. Clin. Cancer Res. 2005, 11, 2576–2582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bell, D.; Chomarat, P.; Broyles, D.; Netto, G.; Harb, G.M.; Lebecque, S.; Valladeau, J.; Davoust, J.; Palucka, K.A.; Banchereau, J. In breast carcinoma tissue, immature dendritic cells reside within the tumor, whereas mature dendritic cells are located in peritumoral areas. J Exp Med 1999, 190, 1417–1426. [Google Scholar] [CrossRef] [PubMed]
- Feijoó, E.; Alfaro, C.; Mazzolini, G.; Serra, P.; Peñuelas, I.; Arina, A.; Huarte, E.; Tirapu, I.; Palencia, B.; Murillo, O.; et al. Dendritic cells delivered inside human carcinomas are sequestered by interleukin-8. Int. J. Cancer 2005, 116, 275–281. [Google Scholar] [CrossRef]
- Yuan, A.; Steigen, S.E.; Goll, R.; Vonen, B.; Husbekk, A.; Cui, G.; Florholmen, J. Dendritic cell infiltration pattern along the colorectal adenoma-carcinoma sequence. Apmis 2008, 116, 445–456. [Google Scholar] [CrossRef]
- Spranger, S.; Bao, R.; Gajewski, T.F. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature 2015, 523, 231–235. [Google Scholar] [CrossRef] [PubMed]
- Roberts, E.W.; Broz, M.L.; Binnewies, M.; Headley, M.B.; Nelson, A.E.; Wolf, D.M.; Kaisho, T.; Bogunovic, D.; Bhardwaj, N.; Krummel, M.F. Critical Role for CD103(+)/CD141(+) Dendritic Cells Bearing CCR7 for Tumor Antigen Trafficking and Priming of T Cell Immunity in Melanoma. Cancer Cell 2016, 30, 324–336. [Google Scholar] [CrossRef]
- Hirao, M.; Onai, N.; Hiroishi, K.; Watkins, S.C.; Matsushima, K.; Robbins, P.D.; Lotze, M.T.; Tahara, H. CC Chemokine Receptor-7 on Dendritic Cells Is Induced after Interaction with Apoptotic Tumor Cells: Critical Role in Migration from the Tumor Site to Draining Lymph Nodes. Cancer Res. 2000, 60, 2209–2217. [Google Scholar]
- Burgoyne, P.; Hayes, A.J.; Cooper, R.S.; Le Brocq, M.L.; Hansell, C.A.H.; Campbell, J.D.; Graham, G.J. Sorting for CCR7-positivity enriches for a dendritic cell population with enhanced antigen-presenting capacity and anti-tumour potency. bioRxiv 2019, 755488. [Google Scholar] [CrossRef]
- Gabrilovich, D.I.; Ostrand-Rosenberg, S.; Bronte, V. Coordinated regulation of myeloid cells by tumours. Nat. Rev. Immunol. 2012, 12, 253–268. [Google Scholar] [CrossRef]
- Almand, B.; Resser, J.R.; Lindman, B.; Nadaf, S.; Clark, J.I.; Kwon, E.D.; Carbone, D.P.; Gabrilovich, D.I. Clinical Significance of Defective Dendritic Cell Differentiation in Cancer. Clin. Cancer Res. 2000, 6, 1755–1766. [Google Scholar]
- Gabrilovich, D.I.; Chen, H.L.; Girgis, K.R.; Cunningham, H.T.; Meny, G.M.; Nadaf, S.; Kavanaugh, D.; Carbone, D.P. Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat. Med. 1996, 2, 1096–1103. [Google Scholar] [CrossRef]
- Bharadwaj, U.; Li, M.; Zhang, R.; Chen, C.; Yao, Q. Elevated interleukin-6 and G-CSF in human pancreatic cancer cell conditioned medium suppress dendritic cell differentiation and activation. Cancer Res. 2007, 67, 5479–5488. [Google Scholar] [CrossRef]
- Stock, A.; Booth, S.; Cerundolo, V. Prostaglandin E2 suppresses the differentiation of retinoic acid-producing dendritic cells in mice and humans. J. Exp. Med. 2011, 208, 761–773. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hargadon, K. Tumor-Altered Dendritic Cell Function: Implications for Anti-Tumor Immunity. Front. Immunol. 2013, 4, 192. [Google Scholar] [CrossRef]
- Menetrier-Caux, C.; Montmain, G.; Dieu, M.C.; Bain, C.; Favrot, M.C.; Caux, C.; Blay, J.Y. Inhibition of the differentiation of dendritic cells from CD34(+) progenitors by tumor cells: Role of interleukin-6 and macrophage colony-stimulating factor. Blood 1998, 92, 4778–4791. [Google Scholar] [CrossRef]
- Lavin, Y.; Kobayashi, S.; Leader, A.; Amir, E.-a.D.; Elefant, N.; Bigenwald, C.; Remark, R.; Sweeney, R.; Becker, C.D.; Levine, J.H.; et al. Innate Immune Landscape in Early Lung Adenocarcinoma by Paired Single-Cell Analyses. Cell 2017, 169, 750–765.e17. [Google Scholar] [CrossRef]
- Zong, J.; Keskinov, A.A.; Shurin, G.V.; Shurin, M.R. Tumor-derived factors modulating dendritic cell function. Cancer Immunol. Immunother. 2016, 65, 821–833. [Google Scholar] [CrossRef] [PubMed]
- Shurin, M.R.; Yurkovetsky, Z.R.; Tourkova, I.L.; Balkir, L.; Shurin, G.V. Inhibition of CD40 expression and CD40-mediated dendritic cell function by tumor-derived IL-10. Int. J. Cancer 2002, 101, 61–68. [Google Scholar] [CrossRef]
- Murugaiyan, G.; Martin, S.; Saha, B. Levels of CD40 expression on dendritic cells dictate tumour growth or regression. Clin. Exp. Immunol. 2007, 149, 194–202. [Google Scholar] [CrossRef]
- Oh, S.A.; Wu, D.-C.; Cheung, J.; Navarro, A.; Xiong, H.; Cubas, R.; Totpal, K.; Chiu, H.; Wu, Y.; Comps-Agrar, L.; et al. PD-L1 expression by dendritic cells is a key regulator of T-cell immunity in cancer. Nature Cancer 2020, 1, 681–691. [Google Scholar] [CrossRef] [PubMed]
- Herbst, R.S.; Soria, J.C.; Kowanetz, M.; Fine, G.D.; Hamid, O.; Gordon, M.S.; Sosman, J.A.; McDermott, D.F.; Powderly, J.D.; Gettinger, S.N.; et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014, 515, 563–567. [Google Scholar] [CrossRef]
- Mayoux, M.; Roller, A.; Pulko, V.; Sammicheli, S.; Chen, S.; Sum, E.; Jost, C.; Fransen, M.F.; Buser, R.B.; Kowanetz, M.; et al. Dendritic cells dictate responses to PD-L1 blockade cancer immunotherapy. Sci. Transl. Med. 2020, 12, eaav7431. [Google Scholar] [CrossRef]
- Miller, T.J.; Anyaegbu, C.C.; Lee-Pullen, T.F.; Spalding, L.J.; Platell, C.F.; McCoy, M.J. PD-L1+ dendritic cells in the tumor microenvironment correlate with good prognosis and CD8+ T cell infiltration in colon cancer. Cancer Sci. 2021, 112, 1173–1183. [Google Scholar] [CrossRef] [PubMed]
- Lin, H.; Wei, S.; Hurt, E.M.; Green, M.D.; Zhao, L.; Vatan, L.; Szeliga, W.; Herbst, R.; Harms, P.W.; Fecher, L.A.; et al. Host expression of PD-L1 determines efficacy of PD-L1 pathway blockade-mediated tumor regression. J. Clin. Investig. 2018, 128, 805–815. [Google Scholar] [CrossRef] [PubMed]
- Leone, P.; Shin, E.-C.; Perosa, F.; Vacca, A.; Dammacco, F.; Racanelli, V. MHC Class I Antigen Processing and Presenting Machinery: Organization, Function, and Defects in Tumor Cells. JNCI J. Natl. Cancer Inst. 2013, 105, 1172–1187. [Google Scholar] [CrossRef]
- Whiteside, T.L.; Stanson, J.; Shurin, M.R.; Ferrone, S. Antigen-Processing Machinery in Human Dendritic Cells: Up-Regulation by Maturation and Down-Regulation by Tumor Cells. J. Immunol. 2004, 173, 1526–1534. [Google Scholar] [CrossRef]
- Tourkova, I.L.; Shurin, G.V.; Chatta, G.S.; Perez, L.; Finke, J.; Whiteside, T.L.; Ferrone, S.; Shurin, M.R. Restoration by IL-15 of MHC Class I Antigen-Processing Machinery in Human Dendritic Cells Inhibited by Tumor-Derived Gangliosides. J. Immunol. 2005, 175, 3045–3052. [Google Scholar] [CrossRef] [PubMed]
- Tourkova, I.L.; Shurin, G.V.; Ferrone, S.; Shurin, M.R. Interferon regulatory factor 8 mediates tumor-induced inhibition of antigen processing and presentation by dendritic cells. Cancer Immunol. Immunother. 2009, 58, 567–574. [Google Scholar] [CrossRef]
- Herber, D.L.; Cao, W.; Nefedova, Y.; Novitskiy, S.V.; Nagaraj, S.; Tyurin, V.A.; Corzo, A.; Cho, H.I.; Celis, E.; Lennox, B.; et al. Lipid accumulation and dendritic cell dysfunction in cancer. Nat. Med. 2010, 16, 880–886. [Google Scholar] [CrossRef] [PubMed]
- Qian, J.; Olbrecht, S.; Boeckx, B.; Vos, H.; Laoui, D.; Etlioglu, E.; Wauters, E.; Pomella, V.; Verbandt, S.; Busschaert, P.; et al. A pan-cancer blueprint of the heterogeneous tumor microenvironment revealed by single-cell profiling. Cell Res. 2020, 30, 745–762. [Google Scholar] [CrossRef]
- Kim, N.; Kim, H.K.; Lee, K.; Hong, Y.; Cho, J.H.; Choi, J.W.; Lee, J.-I.; Suh, Y.-L.; Ku, B.M.; Eum, H.H.; et al. Single-cell RNA sequencing demonstrates the molecular and cellular reprogramming of metastatic lung adenocarcinoma. Nature Commun. 2020, 11, 2285. [Google Scholar] [CrossRef] [PubMed]
- Cillo, A.R.; Kürten, C.H.L.; Tabib, T.; Qi, Z.; Onkar, S.; Wang, T.; Liu, A.; Duvvuri, U.; Kim, S.; Soose, R.J.; et al. Immune Landscape of Viral- and Carcinogen-Driven Head and Neck Cancer. Immunity 2020, 52, 183–199.e9. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.; He, Y.; Luo, N.; Patel, S.J.; Han, Y.; Gao, R.; Modak, M.; Carotta, S.; Haslinger, C.; Kind, D.; et al. Landscape and Dynamics of Single Immune Cells in Hepatocellular Carcinoma. Cell 2019, 179, 829–845.e20. [Google Scholar] [CrossRef] [PubMed]
- Nirschl, C.J.; Suárez-Fariñas, M.; Izar, B.; Prakadan, S.; Dannenfelser, R.; Tirosh, I.; Liu, Y.; Zhu, Q.; Devi, K.S.P.; Carroll, S.L.; et al. IFNγ-Dependent Tissue-Immune Homeostasis Is Co-opted in the Tumor Microenvironment. Cell 2017, 170, 127–141.e15. [Google Scholar] [CrossRef]
- Ji, A.L.; Rubin, A.J.; Thrane, K.; Jiang, S.; Reynolds, D.L.; Meyers, R.M.; Guo, M.G.; George, B.M.; Mollbrink, A.; Bergenstråhle, J.; et al. Multimodal Analysis of Composition and Spatial Architecture in Human Squamous Cell Carcinoma. Cell 2020, 182, 497–514.e422. [Google Scholar] [CrossRef]
- Zhang, L.; Li, Z.; Skrzypczynska, K.M.; Fang, Q.; Zhang, W.; O’Brien, S.A.; He, Y.; Wang, L.; Zhang, Q.; Kim, A.; et al. Single-Cell Analyses Inform Mechanisms of Myeloid-Targeted Therapies in Colon Cancer. Cell 2020, 181, 442–459.e29. [Google Scholar] [CrossRef]
- Zhang, Y.; Narayanan, S.P.; Mannan, R.; Raskind, G.; Wang, X.; Vats, P.; Su, F.; Hosseini, N.; Cao, X.; Kumar-Sinha, C.; et al. Single-cell analyses of renal cell cancers reveal insights into tumor microenvironment, cell of origin, and therapy response. Proc. Natl. Acad. Sci. USA 2021, 118, e2103240118. [Google Scholar] [CrossRef]
- Shi, W.; Li, X.; Porter, J.L.; Ostrodi, D.H.; Yang, B.; Li, J.; Wang, Y.; Zhang, J.; Bai, L.; Jiao, S. Level of plasmacytoid dendritic cells is increased in non-small cell lung carcinoma. Tumour. Biol. 2014, 35, 2247–2252. [Google Scholar] [CrossRef]
- Li, S.; Wu, J.; Zhu, S.; Liu, Y.J.; Chen, J. Disease-Associated Plasmacytoid Dendritic Cells. Front. Immunol. 2017, 8, 1268. [Google Scholar] [CrossRef]
- O’Donnell, R.K.; Mick, R.; Feldman, M.; Hino, S.; Wang, Y.; Brose, M.S.; Muschel, R.J. Distribution of dendritic cell subtypes in primary oral squamous cell carcinoma is inconsistent with a functional response. Cancer Lett. 2007, 255, 145–152. [Google Scholar] [CrossRef]
- Hartmann, E.; Wollenberg, B.; Rothenfusser, S.; Wagner, M.; Wellisch, D.; Mack, B.; Giese, T.; Gires, O.; Endres, S.; Hartmann, G. Identification and Functional Analysis of Tumor-Infiltrating Plasmacytoid Dendritic Cells in Head and Neck Cancer. Cancer Res. 2003, 63, 6478–6487. [Google Scholar]
- Gerhard, G.M.; Bill, R.; Messemaker, M.; Klein, A.M.; Pittet, M.J. Tumor-infiltrating dendritic cell states are conserved across solid human cancers. J. Exp. Med. 2020, 218, e20200264. [Google Scholar] [CrossRef] [PubMed]
- Sosa Cuevas, E.; Ouaguia, L.; Mouret, S.; Charles, J.; De Fraipont, F.; Manches, O.; Valladeau-Guilemond, J.; Bendriss-Vermare, N.; Chaperot, L.; Aspord, C. BDCA1+ cDC2s, BDCA2+ pDCs and BDCA3+ cDC1s reveal distinct pathophysiologic features and impact on clinical outcomes in melanoma patients. Clin. Transl. Immunol. 2020, 9, e1190. [Google Scholar] [CrossRef]
- Broz, M.L.; Binnewies, M.; Boldajipour, B.; Nelson, A.E.; Pollack, J.L.; Erle, D.J.; Barczak, A.; Rosenblum, M.D.; Daud, A.; Barber, D.L.; et al. Dissecting the tumor myeloid compartment reveals rare activating antigen-presenting cells critical for T cell immunity. Cancer Cell 2014, 26, 638–652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barry, K.C.; Hsu, J.; Broz, M.L.; Cueto, F.J.; Binnewies, M.; Combes, A.J.; Nelson, A.E.; Loo, K.; Kumar, R.; Rosenblum, M.D.; et al. A natural killer–dendritic cell axis defines checkpoint therapy–responsive tumor microenvironments. Nature Med. 2018, 24, 1178–1191. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.S.; Radford, K.J. Chapter Three—The role of dendritic cells in cancer. In International Review of Cell and Molecular Biology; Lhuillier, C., Galluzzi, L., Eds.; Academic Press: Cambridge, MA, USA, 2019; Volume 348, pp. 123–178. [Google Scholar]
- Hildner, K.; Edelson, B.T.; Purtha, W.E.; Diamond, M.; Matsushita, H.; Kohyama, M.; Calderon, B.; Schraml, B.U.; Unanue, E.R.; Diamond, M.S.; et al. Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity. Science 2008, 322, 1097–1100. [Google Scholar] [CrossRef]
- Theisen, D.J.; Davidson, J.T.; Briseño, C.G.; Gargaro, M.; Lauron, E.J.; Wang, Q.; Desai, P.; Durai, V.; Bagadia, P.; Brickner, J.R.; et al. WDFY4 is required for cross-presentation in response to viral and tumor antigens. Science 2018, 362, 694–699. [Google Scholar] [CrossRef]
- Caminschi, I.; Proietto, A.I.; Ahmet, F.; Kitsoulis, S.; Shin Teh, J.; Lo, J.C.Y.; Rizzitelli, A.; Wu, L.; Vremec, D.; van Dommelen, S.L.H.; 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] [PubMed]
- Böttcher, 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]
- Meyer, M.A.; Baer, J.M.; Knolhoff, B.L.; Nywening, T.M.; Panni, R.Z.; Su, X.; Weilbaecher, K.N.; Hawkins, W.G.; Ma, C.; Fields, R.C.; et al. Breast and pancreatic cancer interrupt IRF8-dependent dendritic cell development to overcome immune surveillance. Nat. Commun. 2018, 9, 1250. [Google Scholar] [CrossRef]
- Luke, J.J.; Bao, R.; Sweis, R.F.; Spranger, S.; Gajewski, T.F. WNT/β-catenin Pathway Activation Correlates with Immune Exclusion across Human Cancers. Clin. Cancer Res. 2019, 25, 3074–3083. [Google Scholar] [CrossRef]
- Harlin, H.; Meng, Y.; Peterson, A.C.; Zha, Y.; Tretiakova, M.; Slingluff, C.; McKee, M.; Gajewski, T.F. Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer Res. 2009, 69, 3077–3085. [Google Scholar] [CrossRef]
- Ruffell, B.; Chang-Strachan, D.; Chan, V.; Rosenbusch, A.; Ho, C.M.; Pryer, N.; Daniel, D.; Hwang, E.S.; Rugo, H.S.; Coussens, L.M. Macrophage IL-10 blocks CD8+ T cell-dependent responses to chemotherapy by suppressing IL-12 expression in intratumoral dendritic cells. Cancer Cell 2014, 26, 623–637. [Google Scholar] [CrossRef] [PubMed]
- Mattiuz, R.; Brousse, C.; Ambrosini, M.; Cancel, J.-C.; Bessou, G.; Mussard, J.; Sanlaville, A.; Caux, C.; Bendriss-Vermare, N.; Valladeau-Guilemond, J.; et al. Type 1 conventional dendritic cells and interferons are required for spontaneous CD4+ and CD8+ T-cell protective responses to breast cancer. Clin. Transl. Immunol. 2021, 10, e1305. [Google Scholar] [CrossRef] [PubMed]
- Diamond, M.S.; Kinder, M.; Matsushita, H.; Mashayekhi, M.; Dunn, G.P.; Archambault, J.M.; Lee, H.; Arthur, C.D.; White, J.M.; Kalinke, U.; et al. Type I interferon is selectively required by dendritic cells for immune rejection of tumors. J. Exp. Med. 2011, 208, 1989–2003. [Google Scholar] [CrossRef] [PubMed]
- Hubert, M.; Gobbini, E.; Couillault, C.; Manh, T.-P.V.; Doffin, A.-C.; Berthet, J.; Rodriguez, C.; Ollion, V.; Kielbassa, J.; Sajous, C.; et al. IFN-III is selectively produced by cDC1 and predicts good clinical outcome in breast cancer. Sci. Immunol. 2020, 5, eaav3942. [Google Scholar] [CrossRef] [PubMed]
- Chiba, S.; Baghdadi, M.; Akiba, H.; Yoshiyama, H.; Kinoshita, I.; Dosaka-Akita, H.; Fujioka, Y.; Ohba, Y.; Gorman, J.V.; Colgan, J.D.; et al. Tumor-infiltrating DCs suppress nucleic acid-mediated innate immune responses through interactions between the receptor TIM-3 and the alarmin HMGB1. Nat. Immunol. 2012, 13, 832–842. [Google Scholar] [CrossRef]
- de Mingo Pulido, Á.; Gardner, A.; Hiebler, S.; Soliman, H.; Rugo, H.S.; Krummel, M.F.; Coussens, L.M.; Ruffell, B. TIM-3 Regulates CD103(+) Dendritic Cell Function and Response to Chemotherapy in Breast Cancer. Cancer Cell 2018, 33, 60–74.e66. [Google Scholar] [CrossRef]
- Anderson, A.C.; Joller, N.; Kuchroo, V.K. Lag-3, Tim-3, and TIGIT: Co-inhibitory Receptors with Specialized Functions in Immune Regulation. Immunity 2016, 44, 989–1004. [Google Scholar] [CrossRef]
- Bol, K.F.; Tel, J.; de Vries, I.J.; Figdor, C.G. Naturally circulating dendritic cells to vaccinate cancer patients. Oncoimmunology 2013, 2, e23431. [Google Scholar] [CrossRef]
- Di Blasio, S.; van Wigcheren, G.F.; Becker, A.; van Duffelen, A.; Gorris, M.; Verrijp, K.; Stefanini, I.; Bakker, G.J.; Bloemendal, M.; Halilovic, A.; et al. The tumour microenvironment shapes dendritic cell plasticity in a human organotypic melanoma culture. Nat. Commun. 2020, 11, 2749. [Google Scholar] [CrossRef] [PubMed]
- Santegoets, S.J.; Duurland, C.L.; Jordanova, E.J.; van Ham, V.J.; Ehsan, I.; Loof, N.M.; Narang, V.; Dutertre, C.A.; Ginhoux, F.; van Egmond, S.L.; et al. CD163+ cytokine-producing cDC2 stimulate intratumoral type 1 T cell responses in HPV16-induced oropharyngeal cancer. J. Immunother. Cancer 2020, 8, e001053. [Google Scholar] [CrossRef] [PubMed]
- Zitvogel, L.; Galluzzi, L.; Kepp, O.; Smyth, M.J.; Kroemer, G. Type I interferons in anticancer immunity. Nat. Rev. Immunol. 2015, 15, 405–414. [Google Scholar] [CrossRef] [PubMed]
- Kießler, M.; Plesca, I.; Sommer, U.; Wehner, R.; Wilczkowski, F.; Müller, L.; Tunger, A.; Lai, X.; Rentsch, A.; Peuker, K.; et al. Tumor-infiltrating plasmacytoid dendritic cells are associated with survival in human colon cancer. J. Immunother Cancer 2021, 9, e001813. [Google Scholar] [CrossRef]
- Wang, J.-B.; Huang, X.; Li, F.-R. Impaired dendritic cell functions in lung cancer: A review of recent advances and future perspectives. Cancer Commun. 2019, 39, 43. [Google Scholar] [CrossRef]
- Tel, J.; Aarntzen, E.H.; Baba, T.; Schreibelt, G.; Schulte, B.M.; Benitez-Ribas, D.; Boerman, O.C.; Croockewit, S.; Oyen, W.J.; van Rossum, M.; et al. Natural human plasmacytoid dendritic cells induce antigen-specific T-cell responses in melanoma patients. Cancer Res. 2013, 73, 1063–1075. [Google Scholar] [CrossRef]
- Labidi-Galy, S.I.; Sisirak, V.; Meeus, P.; Gobert, M.; Treilleux, I.; Bajard, A.; Combes, J.D.; Faget, J.; Mithieux, F.; Cassignol, A.; et al. Quantitative and functional alterations of plasmacytoid dendritic cells contribute to immune tolerance in ovarian cancer. Cancer Res. 2011, 71, 5423–5434. [Google Scholar] [CrossRef]
- Han, N.; Zhang, Z.; Liu, S.; Ow, A.; Ruan, M.; Yang, W.; Zhang, C. Increased tumor-infiltrating plasmacytoid dendritic cells predicts poor prognosis in oral squamous cell carcinoma. Arch. Oral Biol. 2017, 78, 129–134. [Google Scholar] [CrossRef]
- Treilleux, I.; Blay, J.Y.; Bendriss-Vermare, N.; Ray-Coquard, I.; Bachelot, T.; Guastalla, J.P.; Bremond, A.; Goddard, S.; Pin, J.J.; Barthelemy-Dubois, C.; et al. Dendritic cell infiltration and prognosis of early stage breast cancer. Clin. Cancer Res. 2004, 10, 7466–7474. [Google Scholar] [CrossRef]
- Labidi-Galy, S.I.; Treilleux, I.; Goddard-Leon, S.; Combes, J.D.; Blay, J.Y.; Ray-Coquard, I.; Caux, C.; Bendriss-Vermare, N. Plasmacytoid dendritic cells infiltrating ovarian cancer are associated with poor prognosis. Oncoimmunology 2012, 1, 380–382. [Google Scholar] [CrossRef]
- Gadalla, R.; Hassan, H.; Ibrahim, S.A.; Abdullah, M.S.; Gaballah, A.; Greve, B.; El-Deeb, S.; El-Shinawi, M.; Mohamed, M.M. Tumor microenvironmental plasmacytoid dendritic cells contribute to breast cancer lymph node metastasis via CXCR4/SDF-1 axis. Breast Cancer Res. Treat. 2019, 174, 679–691. [Google Scholar] [CrossRef] [PubMed]
- van der Touw, W.; Chen, H.M.; Pan, P.Y.; Chen, S.H. LILRB receptor-mediated regulation of myeloid cell maturation and function. Cancer Immunol. Immunother. 2017, 66, 1079–1087. [Google Scholar] [CrossRef] [PubMed]
- Jahrsdörfer, B.; Vollmer, A.; Blackwell, S.E.; Maier, J.; Sontheimer, K.; Beyer, T.; Mandel, B.; Lunov, O.; Tron, K.; Nienhaus, G.U.; et al. Granzyme B produced by human plasmacytoid dendritic cells suppresses T-cell expansion. Blood 2010, 115, 1156–1165. [Google Scholar] [CrossRef] [PubMed]
- Perrot, I.; Blanchard, D.; Freymond, N.; Isaac, S.; Guibert, B.; Pachéco, Y.; Lebecque, S. Dendritic cells infiltrating human non-small cell lung cancer are blocked at immature stage. J. Immunol. 2007, 178, 2763–2769. [Google Scholar] [CrossRef] [Green Version]
- Yang, L.L.; Mao, L.; Wu, H.; Chen, L.; Deng, W.W.; Xiao, Y.; Li, H.; Zhang, L.; Sun, Z.J. pDC depletion induced by CD317 blockade drives the antitumor immune response in head and neck squamous cell carcinoma. Oral Oncol. 2019, 96, 131–139. [Google Scholar] [CrossRef]
- Thiel, A.; Pries, R.; Jeske, S.; Trenkle, T.; Wollenberg, B. Effect of head and neck cancer supernatant and CpG-oligonucleotides on migration and IFN-alpha production of plasmacytoid dendritic cells. Anticancer Res. 2009, 29, 3019–3025. [Google Scholar]
- Sisirak, V.; Faget, J.; Gobert, M.; Goutagny, N.; Vey, N.; Treilleux, I.; Renaudineau, S.; Poyet, G.; Labidi-Galy, S.I.; Goddard-Leon, S.; et al. Impaired IFN-α production by plasmacytoid dendritic cells favors regulatory T-cell expansion that may contribute to breast cancer progression. Cancer Res. 2012, 72, 5188–5197. [Google Scholar] [CrossRef]
- Camisaschi, C.; De Filippo, A.; Beretta, V.; Vergani, B.; Villa, A.; Vergani, E.; Santinami, M.; Cabras, A.D.; Arienti, F.; Triebel, F.; et al. Alternative activation of human plasmacytoid DCs in vitro and in melanoma lesions: Involvement of LAG-3. J. Investig. Dermatol 2014, 134, 1893–1902. [Google Scholar] [CrossRef]
- Combes, A.; Camosseto, V.; N’Guessan, P.; Argüello, R.J.; Mussard, J.; Caux, C.; Bendriss-Vermare, N.; Pierre, P.; Gatti, E. BAD-LAMP controls TLR9 trafficking and signalling in human plasmacytoid dendritic cells. Nat. Commun. 2017, 8, 913. [Google Scholar] [CrossRef]
- Koucký, V.; Bouček, J.; Fialová, A. Immunology of Plasmacytoid Dendritic Cells in Solid Tumors: A Brief Review. Cancers 2019, 11, 470. [Google Scholar] [CrossRef]
- 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]
- Pedroza-Gonzalez, A.; Zhou, G.; Vargas-Mendez, E.; Boor, P.P.; Mancham, S.; Verhoef, C.; Polak, W.G.; Grünhagen, D.; Pan, Q.; Janssen, H.; et al. Tumor-infiltrating plasmacytoid dendritic cells promote immunosuppression by Tr1 cells in human liver tumors. Oncoimmunology 2015, 4, e1008355. [Google Scholar] [CrossRef] [PubMed]
- Aspord, C.; Leccia, M.T.; Charles, J.; Plumas, J. Plasmacytoid dendritic cells support melanoma progression by promoting Th2 and regulatory immunity through OX40L and ICOSL. Cancer Immunol. Res. 2013, 1, 402–415. [Google Scholar] [CrossRef] [PubMed]
- Faget, J.; Sisirak, V.; Blay, J.Y.; Caux, C.; Bendriss-Vermare, N.; Ménétrier-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]
- Poropatich, K.; Dominguez, D.; Chan, W.-C.; Andrade, J.; Zha, Y.; Wray, B.; Miska, J.; Qin, L.; Cole, L.; Coates, S.; et al. OX40+ plasmacytoid dendritic cells in the tumor microenvironment promote antitumor immunity. J. Clin. Investig. 2020, 130, 3528–3542. [Google Scholar] [CrossRef]
- von Bergwelt-Baildon, M.S.; Popov, A.; Saric, T.; Chemnitz, J.; Classen, S.; Stoffel, M.S.; Fiore, F.; Roth, U.; Beyer, M.; Debey, S.; et al. CD25 and indoleamine 2,3-dioxygenase are up-regulated by prostaglandin E2 and expressed by tumor-associated dendritic cells in vivo: Additional mechanisms of T-cell inhibition. Blood 2006, 108, 228–237. [Google Scholar] [CrossRef]
- Speeckaert, R.; Vermaelen, K.; van Geel, N.; Autier, P.; Lambert, J.; Haspeslagh, M.; van Gele, M.; Thielemans, K.; Neyns, B.; Roche, N.; et al. Indoleamine 2,3-dioxygenase, a new prognostic marker in sentinel lymph nodes of melanoma patients. Eur. J. Cancer 2012, 48, 2004–2011. [Google Scholar] [CrossRef]
- Schütz, C.; Inselmann, S.; Saussele, S.; Dietz, C.T.; Müller, M.C.; Eigendorff, E.; Brendel, C.A.; Metzelder, S.K.; Brümmendorf, T.H.; Waller, C.; et al. Expression of the CTLA-4 ligand CD86 on plasmacytoid dendritic cells (pDC) predicts risk of disease recurrence after treatment discontinuation in CML. Leukemia 2018, 32, 1054. [Google Scholar] [CrossRef]
- Ray, A.; Das, D.S.; Song, Y.; Richardson, P.; Munshi, N.C.; Chauhan, D.; Anderson, K.C. Targeting PD1-PDL1 immune checkpoint in plasmacytoid dendritic cell interactions with T cells, natural killer cells and multiple myeloma cells. Leukemia 2015, 29, 1441–1444. [Google Scholar] [CrossRef]
- Yan, Y.; Zhang, G.-X.; Gran, B.; Fallarino, F.; Yu, S.; Li, H.; Cullimore, M.L.; Rostami, A.; Xu, H. IDO Upregulates Regulatory T Cells via Tryptophan Catabolite and Suppresses Encephalitogenic T Cell Responses in Experimental Autoimmune Encephalomyelitis. J. Immunol. 2010, 185, 5953–5961. [Google Scholar] [CrossRef]
- Garris, C.S.; Arlauckas, S.P.; Kohler, R.H.; Trefny, M.P.; Garren, S.; Piot, C.; Engblom, C.; Pfirschke, C.; Siwicki, M.; Gungabeesoon, J.; et al. Successful Anti-PD-1 Cancer Immunotherapy Requires T Cell-Dendritic Cell Crosstalk Involving the Cytokines IFN-γ and IL-12. Immunity 2018, 49, 1148–1161.e7. [Google Scholar] [CrossRef] [PubMed]
- 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. 2017, 5, 1098–1108. [Google Scholar] [CrossRef] [PubMed]
- Kuhn, S.; Yang, J.; Ronchese, F. Monocyte-Derived Dendritic Cells Are Essential for CD8(+) T Cell Activation and Antitumor Responses After Local Immunotherapy. Front. Immunol. 2015, 6, 584. [Google Scholar] [CrossRef] [PubMed]
- Schetters, S.T.T.; Rodriguez, E.; Kruijssen, L.J.W.; Crommentuijn, M.H.W.; Boon, L.; Van den Bossche, J.; Den Haan, J.M.M.; Van Kooyk, Y. Monocyte-derived APCs are central to the response of PD1 checkpoint blockade and provide a therapeutic target for combination therapy. J. Immunother. Cancer 2020, 8, e000588. [Google Scholar] [CrossRef]
- ITO, S.; NAGATA, Y.; SUSUMU, S.; YONEDA, A.; MATSUO, M.; YUI, K.; UDONO, H.; EGUCHI, S.; KANEMATSU, T. Phenotypic Analysis of Monocyte-derived Dendritic Cells Loaded with Tumor Antigen with Heat-shock Cognate Protein-70. Anticancer Res. 2012, 32, 4897–4904. [Google Scholar]
- Wang, M.; Shi, J.; Wan, Y.; Li, J.; Yuan, Y. A subset of myeloid dendritic cells derived from peripheral blood monocytes represented a predominant subset characterized by their potential tumor-inhibiting activity. Vitro Cell Dev. Biol. Anim. 2009, 45, 398–404. [Google Scholar] [CrossRef]
- Bellik, L.; Gerlini, G.; Parenti, A.; Ledda, F.; Pimpinelli, N.; Neri, B.; Pantalone, D. Role of conventional treatments on circulating and monocyte-derived dendritic cells in colorectal cancer. Clin. Immunol 2006, 121, 74–80. [Google Scholar] [CrossRef]
- Orsini, G.; Legitimo, A.; Failli, A.; Ferrari, P.; Nicolini, A.; Spisni, R.; Miccoli, P.; Consolini, R. Defective generation and maturation of dendritic cells from monocytes in colorectal cancer patients during the course of disease. Int. J. Mol. Sci. 2013, 14, 22022–22041. [Google Scholar] [CrossRef]
- Kiertscher, S.M.; Luo, J.; Dubinett, S.M.; Roth, M.D. Tumors Promote Altered Maturation and Early Apoptosis of Monocyte-Derived Dendritic Cells. J. Immunol. 2000, 164, 1269–1276. [Google Scholar] [CrossRef]
- Devalaraja, S.; To, T.K.J.; Folkert, I.W.; Natesan, R.; Alam, M.Z.; Li, M.; Tada, Y.; Budagyan, K.; Dang, M.T.; Zhai, L.; et al. Tumor-Derived Retinoic Acid Regulates Intratumoral Monocyte Differentiation to Promote Immune Suppression. Cell 2020, 180, 1098–1114.e1016. [Google Scholar] [CrossRef]
- Santana-Magal, N.; Farhat-Younis, L.; Gutwillig, A.; Gleiberman, A.; Rasoulouniriana, D.; Tal, L.; Netanely, D.; Shamir, R.; Blau, R.; Feinmesser, M.; et al. Melanoma-Secreted Lysosomes Trigger Monocyte-Derived Dendritic Cell Apoptosis and Limit Cancer Immunotherapy. Cancer Res. 2020, 80, 1942–1956. [Google Scholar] [CrossRef] [PubMed]
- Shinde, P.; Fernandes, S.; Melinkeri, S.; Kale, V.; Limaye, L. Compromised functionality of monocyte-derived dendritic cells in multiple myeloma patients may limit their use in cancer immunotherapy. Sci. Res. 2018, 8, 5705. [Google Scholar] [CrossRef] [PubMed]
- Noubade, R.; Majri-Morrison, S.; Tarbell, K.V. Beyond cDC1: Emerging Roles of DC Crosstalk in Cancer Immunity. Front. Immunol. 2019, 10, 1014. [Google Scholar] [CrossRef] [PubMed]
- Hor, J.L.; Whitney, P.G.; Zaid, A.; Brooks, A.G.; Heath, W.R.; Mueller, S.N. Spatiotemporally Distinct Interactions with Dendritic Cell Subsets Facilitates CD4+ and CD8+ T Cell Activation to Localized Viral Infection. Immunity 2015, 43, 554–565. [Google Scholar] [CrossRef]
- Brewitz, A.; Eickhoff, S.; Dähling, S.; Quast, T.; Bedoui, S.; Kroczek, R.A.; Kurts, C.; Garbi, N.; Barchet, W.; Iannacone, M.; et al. CD8(+) T Cells Orchestrate pDC-XCR1(+) Dendritic Cell Spatial and Functional Cooperativity to Optimize Priming. Immunity 2017, 46, 205–219. [Google Scholar] [CrossRef]
- Allan, R.S.; Waithman, J.; Bedoui, S.; Jones, C.M.; Villadangos, J.A.; Zhan, Y.; Lew, A.M.; Shortman, K.; Heath, W.R.; Carbone, F.R. Migratory dendritic cells transfer antigen to a lymph node-resident dendritic cell population for efficient CTL priming. Immunity 2006, 25, 153–162. [Google Scholar] [CrossRef] [Green Version]
- Quezada, S.A.; Simpson, T.R.; Peggs, K.S.; Merghoub, T.; Vider, J.; Fan, X.; Blasberg, R.; Yagita, H.; Muranski, P.; Antony, P.A.; et al. Tumor-reactive CD4(+) T cells develop cytotoxic activity and eradicate large established melanoma after transfer into lymphopenic hosts. J. Exp. Med. 2010, 207, 637–650. [Google Scholar] [CrossRef]
- Kennedy, R.; Celis, E. Multiple roles for CD4+ T cells in anti-tumor immune responses. Immunol. Rev. 2008, 222, 129–144. [Google Scholar] [CrossRef]
- Greyer, M.; Whitney, P.G.; Stock, A.T.; Davey, G.M.; Tebartz, C.; Bachem, A.; Mintern, J.D.; Strugnell, R.A.; Turner, S.J.; Gebhardt, T.; et al. T Cell Help Amplifies Innate Signals in CD8(+) DCs for Optimal CD8(+) T Cell Priming. Cell Rep. 2016, 14, 586–597. [Google Scholar] [CrossRef]
- van Beek, J.J.P.; Flórez-Grau, G.; Gorris, M.A.J.; Mathan, T.S.M.; Schreibelt, G.; Bol, K.F.; Textor, J.; de Vries, I.J.M. Human pDCs Are Superior to cDC2s in Attracting Cytolytic Lymphocytes in Melanoma Patients Receiving DC Vaccination. Cell Rep. 2020, 30, 1027–1038.e1024. [Google Scholar] [CrossRef]
- Yang, H.; Zhang, Q.; Xu, M.; Wang, L.; Chen, X.; Feng, Y.; Li, Y.; Zhang, X.; Cui, W.; Jia, X. CCL2-CCR2 axis recruits tumor associated macrophages to induce immune evasion through PD-1 signaling in esophageal carcinogenesis. Mol. Cancer 2020, 19, 41. [Google Scholar] [CrossRef]
- Abraham, D.; Zins, K.; Sioud, M.; Lucas, T.; Schäfer, R.; Stanley, E.R.; Aharinejad, S. Stromal cell-derived CSF-1 blockade prolongs xenograft survival of CSF-1-negative neuroblastoma. Int. J. Cancer 2010, 126, 1339–1352. [Google Scholar] [CrossRef] [PubMed]
- Yamamoto, T.; Yanagimoto, H.; Satoi, S.; Toyokawa, H.; Yamao, J.; Kim, S.; Terakawa, N.; Takahashi, K.; Kwon, A.H. Circulating myeloid dendritic cells as prognostic factors in patients with pancreatic cancer who have undergone surgical resection. J. Surg. Res. 2012, 173, 299–308. [Google Scholar] [CrossRef] [PubMed]
- Carenza, C.; Franzese, S.; Calcaterra, F.; Mavilio, D.; Della Bella, S. Comprehensive Phenotyping of Dendritic Cells in Cancer Patients by Flow Cytometry. Cytometry A 2021, 99, 218–230. [Google Scholar] [CrossRef] [PubMed]
- Kini Bailur, J.; Gueckel, B.; Pawelec, G. Prognostic impact of high levels of circulating plasmacytoid dendritic cells in breast cancer. J. Transl. Med. 2016, 14, 151. [Google Scholar] [CrossRef] [PubMed]
- Yanagimoto, H.; Takai, S.; Satoi, S.; Toyokawa, H.; Takahashi, K.; Terakawa, N.; Kwon, A.H.; Kamiyama, Y. Impaired function of circulating dendritic cells in patients with pancreatic cancer. Clin. Immunol. 2005, 114, 52–60. [Google Scholar] [CrossRef]
- Wojas, K.; Tabarkiewicz, J.; Jankiewicz, M.; Roliński, J. Dendritic cells in peripheral blood of patients with breast and lung cancer--a pilot study. Folia Histochem. Cytobiol 2004, 42, 45–48. [Google Scholar]
- Takahashi, K.; Toyokawa, H.; Takai, S.; Satoi, S.; Yanagimoto, H.; Terakawa, N.; Araki, H.; Kwon, A.H.; Kamiyama, Y. Surgical influence of pancreatectomy on the function and count of circulating dendritic cells in patients with pancreatic cancer. Cancer Immunol. Immunother. 2006, 55, 775–784. [Google Scholar] [CrossRef]
- Tjomsland, V.; Sandström, P.; Spångeus, A.; Messmer, D.; Emilsson, J.; Falkmer, U.; Falkmer, S.; Magnusson, K.E.; Borch, K.; Larsson, M. Pancreatic adenocarcinoma exerts systemic effects on the peripheral blood myeloid and plasmacytoid dendritic cells: An indicator of disease severity? BMC Cancer 2010, 10, 87. [Google Scholar] [CrossRef]
- Ferrari, S.; Malugani, F.; Rovati, B.; Porta, C.; Riccardi, A.; Danova, M. Flow cytometric analysis of circulating dendritic cell subsets and intracellular cytokine production in advanced breast cancer patients. Oncol. Rep. 2005, 14, 113–120. [Google Scholar] [CrossRef]
- Bella, S.D.; Gennaro, M.; Vaccari, M.; Ferraris, C.; Nicola, S.; Riva, A.; Clerici, M.; Greco, M.; Villa, M.L. Altered maturation of peripheral blood dendritic cells in patients with breast cancer. Br. J. Cancer 2003, 89, 1463–1472. [Google Scholar] [CrossRef] [PubMed]
- Lee, Y.S.; O’Brien, L.J.; Walpole, C.M.; Pearson, F.E.; Leal-Rojas, I.M.; Masterman, K.-A.; Atkinson, V.; Barbour, A.; Radford, K.J. Human CD141+ dendritic cells (cDC1) are impaired in patients with advanced melanoma but can be targeted to enhance anti-PD-1 in a humanized mouse model. J. Immunother. Cancer 2021, 9, e001963. [Google Scholar] [CrossRef] [PubMed]
- Almeida, J.; Bueno, C.; Alguero, M.C.; Sanchez, M.L.; Cañizo, M.C.; Fernandez, M.E.; Vaquero, J.M.; Laso, F.J.; Escribano, L.; San Miguel, J.F.; et al. Extensive characterization of the immunophenotype and pattern of cytokine production by distinct subpopulations of normal human peripheral blood MHC II+/lineage- cells. Clin. Exp. Immunol. 1999, 118, 392–401. [Google Scholar] [CrossRef] [PubMed]
- Adhikaree, J.; Franks, H.A.; Televantos, C.; Vaghela, P.; Kaur, A.P.; Walker, D.; Schmitz, M.; Jackson, A.M.; Patel, P.M. Impaired circulating myeloid CD1c+ dendritic cell function in human glioblastoma is restored by p38 inhibition—Implications for the next generation of DC vaccines. Oncoimmunology 2019, 8, 1593803. [Google Scholar] [CrossRef]
- Sakakura, K.; Chikamatsu, K.; Takahashi, K.; Whiteside, T.L.; Furuya, N. Maturation of circulating dendritic cells and imbalance of T-cell subsets in patients with squamous cell carcinoma of the head and neck. Cancer Immunol. Immunother. 2006, 55, 151–159. [Google Scholar] [CrossRef]
- Liu, W.; Zhao, J.; Li, Q.; Wang, Q.; Zhou, Y.; Tong, Z. Gastric cancer patients have elevated plasmacytoid and CD1c+ dendritic cells in the peripheral blood. Oncol. Lett. 2018, 15, 5087–5092. [Google Scholar] [CrossRef]
- Wertel, I.; Polak, G.; Bednarek, W.; Barczyński, B.; Roliński, J.; Kotarski, J. Dendritic cell subsets in the peritoneal fluid and peripheral blood of women suffering from ovarian cancer. Cytom. B Clin. Cytom. 2008, 74, 251–258. [Google Scholar] [CrossRef]
- Curiel, T.J.; Cheng, P.; Mottram, P.; Alvarez, X.; Moons, L.; Evdemon-Hogan, M.; Wei, S.; Zou, L.; Kryczek, I.; Hoyle, G.; et al. Dendritic cell subsets differentially regulate angiogenesis in human ovarian cancer. Cancer Res. 2004, 64, 5535–5538. [Google Scholar] [CrossRef]
- Mohty, M.; Isnardon, D.; Vey, N.; Brière, F.; Blaise, D.; Olive, D.; Gaugler, B. Low blood dendritic cells in chronic myeloid leukaemia patients correlates with loss of CD34+/CD38- primitive haematopoietic progenitors. Br. J. Haematol 2002, 119, 115–118. [Google Scholar] [CrossRef]
- Ratta, M.; Fagnoni, F.; Curti, A.; Vescovini, R.; Sansoni, P.; Oliviero, B.; Fogli, M.; Ferri, E.; Della Cuna, G.R.; Tura, S.; et al. Dendritic cells are functionally defective in multiple myeloma: The role of interleukin-6. Blood 2002, 100, 230–237. [Google Scholar] [CrossRef]
- Boissel, N.; Rousselot, P.; Raffoux, E.; Cayuela, J.M.; Maarek, O.; Charron, D.; Degos, L.; Dombret, H.; Toubert, A.; Rea, D. Defective blood dendritic cells in chronic myeloid leukemia correlate with high plasmatic VEGF and are not normalized by imatinib mesylate. Leukemia 2004, 18, 1656–1661. [Google Scholar] [CrossRef] [PubMed]
- Paek, S.H.; Kim, H.G.; Lee, J.W.; Woo, J.; Kwon, H.; Kim, J.B.; Lim, W.; Kim, J.R.; Moon, B.I.; Paik, N.S. Circulating Plasmacytoid and Myeloid Dendritic Cells in Breast Cancer Patients: A Pilot Study. J. Breast Cancer 2019, 22, 29–37. [Google Scholar] [CrossRef] [PubMed]
- Azizi, E.; Carr, A.J.; Plitas, G.; Cornish, A.E.; Konopacki, C.; Prabhakaran, S.; Nainys, J.; Wu, K.; Kiseliovas, V.; Setty, M.; et al. Single-Cell Map of Diverse Immune Phenotypes in the Breast Tumor Microenvironment. Cell 2018, 174, 1293–1308.e36. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Yang, J.; Jiang, J.; Zhuang, Y.; He, W. Function and subsets of dendritic cells and natural killer cells were decreased in gastric cancer. Int. J. Clin. Exp. Pathol. 2014, 7, 8304–8311. [Google Scholar] [PubMed]
- Tabarkiewicz, J.; Rybojad, P.; Jablonka, A.; Rolinski, J. CD1c+ and CD303+ dendritic cells in peripheral blood, lymph nodes and tumor tissue of patients with non-small cell lung cancer. Oncol. Rep. 2008, 19, 237–243. [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]
- Carvalho, H.A.; Villar, R.C. Radiotherapy and immune response: The systemic effects of a local treatment. Clinics (Sao Paulo) 2018, 73, e557s. [Google Scholar] [CrossRef]
- Obeid, M.; Panaretakis, T.; Joza, N.; Tufi, R.; Tesniere, A.; van Endert, P.; Zitvogel, L.; Kroemer, G. Calreticulin exposure is required for the immunogenicity of gamma-irradiation and UVC light-induced apoptosis. Cell Death Differ. 2007, 14, 1848–1850. [Google Scholar] [CrossRef]
- Perregaux, D.G.; McNiff, P.; Laliberte, R.; Conklyn, M.; Gabel, C.A. ATP acts as an agonist to promote stimulus-induced secretion of IL-1 beta and IL-18 in human blood. J. Immunol. 2000, 165, 4615–4623. [Google Scholar] [CrossRef]
- Ghiringhelli, F.; Apetoh, L.; Tesniere, A.; Aymeric, L.; Ma, Y.; Ortiz, C.; Vermaelen, K.; Panaretakis, T.; Mignot, G.; Ullrich, E.; et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1beta-dependent adaptive immunity against tumors. Nat. Med. 2009, 15, 1170–1178. [Google Scholar] [CrossRef]
- Aymeric, L.; Apetoh, L.; Ghiringhelli, F.; Tesniere, A.; Martins, I.; Kroemer, G.; Smyth, M.J.; Zitvogel, L. Tumor cell death and ATP release prime dendritic cells and efficient anticancer immunity. Cancer Res. 2010, 70, 855–858. [Google Scholar] [CrossRef] [PubMed]
- Gameiro, S.R.; Jammeh, M.L.; Wattenberg, M.M.; Tsang, K.Y.; Ferrone, S.; Hodge, J.W. Radiation-induced immunogenic modulation of tumor enhances antigen processing and calreticulin exposure, resulting in enhanced T-cell killing. Oncotarget 2014, 5, 403–416. [Google Scholar] [CrossRef] [PubMed]
- Lim, J.Y.; Gerber, S.A.; Murphy, S.P.; Lord, E.M. Type I interferons induced by radiation therapy mediate recruitment and effector function of CD8(+) T cells. Cancer Immunol. Immunother. 2014, 63, 259–271. [Google Scholar] [CrossRef] [PubMed]
- Korbelik, M.; Zhang, W.; Merchant, S. Involvement of damage-associated molecular patterns in tumor response to photodynamic therapy: Surface expression of calreticulin and high-mobility group box-1 release. Cancer Immunol. Immunother. 2011, 60, 1431–1437. [Google Scholar] [CrossRef]
- Sistigu, A.; Yamazaki, T.; Vacchelli, E.; Chaba, K.; Enot, D.P.; Adam, J.; Vitale, I.; Goubar, A.; Baracco, E.E.; Remedios, C.; et al. Cancer cell-autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat.Med. 2014, 20, 1301–1309. [Google Scholar] [CrossRef]
- Bernard, J.J.; Cowing-Zitron, C.; Nakatsuji, T.; Muehleisen, B.; Muto, J.; Borkowski, A.W.; Martinez, L.; Greidinger, E.L.; Yu, B.D.; Gallo, R.L. Ultraviolet radiation damages self noncoding RNA and is detected by TLR3. Nat. Med. 2012, 18, 1286–1290. [Google Scholar] [CrossRef]
- Apetoh, L.; Ghiringhelli, F.; Tesniere, A.; Obeid, M.; Ortiz, C.; Criollo, A.; Mignot, G.; Maiuri, M.C.; Ullrich, E.; Saulnier, P.; et al. Toll-like receptor 4-dependent contribution of the immune system to anticancer chemotherapy and radiotherapy. Nat. Med. 2007, 13, 1050–1059. [Google Scholar] [CrossRef]
- Vanpouille-Box, C.; Alard, A.; Aryankalayil, M.J.; Sarfraz, Y.; Diamond, J.M.; Schneider, R.J.; Inghirami, G.; Coleman, C.N.; Formenti, S.C.; Demaria, S. DNA exonuclease Trex1 regulates radiotherapy-induced tumour immunogenicity. Nat. Commun. 2017, 8, 15618. [Google Scholar] [CrossRef]
- Deng, L.; Liang, H.; Xu, M.; Yang, X.; Burnette, B.; Arina, A.; Li, X.D.; Mauceri, H.; Beckett, M.; Darga, T.; et al. STING-Dependent Cytosolic DNA Sensing Promotes Radiation-Induced Type I Interferon-Dependent Antitumor Immunity in Immunogenic Tumors. Immunity 2014, 41, 843–852. [Google Scholar] [CrossRef]
- Hou, Y.; Liang, H.; Rao, E.; Zheng, W.; Huang, X.; Deng, L.; Zhang, Y.; Yu, X.; Xu, M.; Mauceri, H.; et al. Non-canonical NF-kappaB Antagonizes STING Sensor-Mediated DNA Sensing in Radiotherapy. Immunity 2018, 49, 490–503.e494. [Google Scholar] [CrossRef]
- Surace, L.; Lysenko, V.; Fontana, A.O.; Cecconi, V.; Janssen, H.; Bicvic, A.; Okoniewski, M.; Pruschy, M.; Dummer, R.; Neefjes, J.; et al. Complement is a central mediator of radiotherapy-induced tumor-specific immunity and clinical response. Immunity 2015, 42, 767–777. [Google Scholar] [CrossRef] [PubMed]
- Gorin, J.B.; Menager, J.; Gouard, S.; Maurel, C.; Guilloux, Y.; Faivre-Chauvet, A.; Morgenstern, A.; Bruchertseifer, F.; Cherel, M.; Davodeau, F.; et al. Antitumor immunity induced after alpha irradiation. Neoplasia 2014, 16, 319–328. [Google Scholar] [CrossRef] [PubMed]
- Zou, S.; Tong, Q.; Liu, B.; Huang, W.; Tian, Y.; Fu, X. Targeting STAT3 in Cancer Immunotherapy. Mol. Cancer 2020, 19, 145. [Google Scholar] [CrossRef] [PubMed]
- Yu, H.; Lee, H.; Herrmann, A.; Buettner, R.; Jove, R. Revisiting STAT3 signalling in cancer: New and unexpected biological functions. Nat. Rev. Cancer 2014, 14, 736–746. [Google Scholar] [CrossRef] [PubMed]
- Siersbaek, R.; Scabia, V.; Nagarajan, S.; Chernukhin, I.; Papachristou, E.K.; Broome, R.; Johnston, S.J.; Joosten, S.E.P.; Green, A.R.; Kumar, S.; et al. IL6/STAT3 Signaling Hijacks Estrogen Receptor alpha Enhancers to Drive Breast Cancer Metastasis. Cancer Cell 2020, 38, 412–423.e419. [Google Scholar] [CrossRef] [PubMed]
- Khatib, A.; Solaimuthu, B.; Ben Yosef, M.; Abu Rmaileh, A.; Tanna, M.; Oren, G.; Schlesinger Frisch, M.; Axelrod, J.H.; Lichtenstein, M.; Shaul, Y.D. The glutathione peroxidase 8 (GPX8)/IL-6/STAT3 axis is essential in maintaining an aggressive breast cancer phenotype. Proc. Natl. Acad. Sci. USA 2020, 117, 21420–21431. [Google Scholar] [CrossRef] [PubMed]
- Deng, J.; Liu, Y.; Lee, H.; Herrmann, A.; Zhang, W.; Zhang, C.; Shen, S.; Priceman, S.J.; Kujawski, M.; Pal, S.K.; et al. S1PR1-STAT3 signaling is crucial for myeloid cell colonization at future metastatic sites. Cancer Cell 2012, 21, 642–654. [Google Scholar] [CrossRef] [Green Version]
- Nefedova, Y.; Huang, M.; Kusmartsev, S.; Bhattacharya, R.; Cheng, P.; Salup, R.; Jove, R.; Gabrilovich, D. Hyperactivation of STAT3 is involved in abnormal differentiation of dendritic cells in cancer. J. Immunol. 2004, 172, 464–474. [Google Scholar] [CrossRef]
- Nefedova, Y.; Cheng, P.; Gilkes, D.; Blaskovich, M.; Beg, A.A.; Sebti, S.M.; Gabrilovich, D.I. Activation of dendritic cells via inhibition of Jak2/STAT3 signaling. J. Immunol. 2005, 175, 4338–4346. [Google Scholar] [CrossRef]
- Oosterhoff, D.; Lougheed, S.; van de Ven, R.; Lindenberg, J.; van Cruijsen, H.; Hiddingh, L.; Kroon, J.; van den Eertwegh, A.J.; Hangalapura, B.; Scheper, R.J.; et al. Tumor-mediated inhibition of human dendritic cell differentiation and function is consistently counteracted by combined p38 MAPK and STAT3 inhibition. Oncoimmunology 2012, 1, 649–658. [Google Scholar] [CrossRef]
- Uhlen, M.; Fagerberg, L.; Hallstrom, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, A.; Kampf, C.; Sjostedt, E.; Asplund, A.; et al. Proteomics. Tissue-based map of the human proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef] [PubMed]
- van Baren, N.; Van den Eynde, B.J. Tryptophan-degrading enzymes in tumoral immune resistance. Front. Immunol. 2015, 6, 34. [Google Scholar] [CrossRef] [PubMed]
- Thuring, M.; Knuchel, R.; Picchetta, L.; Keller, D.; Schmidli, T.S.; Provenzano, M. The Prognostic Value of Indoleamine-2,3-Dioxygenase Gene Expression in Urine of Prostate Cancer Patients Undergoing Radical Prostatectomy as First Treatment of Choice. Front. Immunol. 2020, 11, 1244. [Google Scholar] [CrossRef] [PubMed]
- Pan, K.; Wang, H.; Chen, M.S.; Zhang, H.K.; Weng, D.S.; Zhou, J.; Huang, W.; Li, J.J.; Song, H.F.; Xia, J.C. Expression and prognosis role of indoleamine 2,3-dioxygenase in hepatocellular carcinoma. J. Cancer Res. Clin. Oncol. 2008, 134, 1247–1253. [Google Scholar] [CrossRef] [PubMed]
- Riesenberg, R.; Weiler, C.; Spring, O.; Eder, M.; Buchner, A.; Popp, T.; Castro, M.; Kammerer, R.; Takikawa, O.; Hatz, R.A.; et al. Expression of indoleamine 2,3-dioxygenase in tumor endothelial cells correlates with long-term survival of patients with renal cell carcinoma. Clin. Cancer Res. 2007, 13, 6993–7002. [Google Scholar] [CrossRef]
- Smith, C.; Chang, M.Y.; Parker, K.H.; Beury, D.W.; DuHadaway, J.B.; Flick, H.E.; Boulden, J.; Sutanto-Ward, E.; Soler, A.P.; Laury-Kleintop, L.D.; et al. IDO is a nodal pathogenic driver of lung cancer and metastasis development. Cancer Discov. 2012, 2, 722–735. [Google Scholar] [CrossRef]
- Brandacher, G.; Perathoner, A.; Ladurner, R.; Schneeberger, S.; Obrist, P.; Winkler, C.; Werner, E.R.; Werner-Felmayer, G.; Weiss, H.G.; Gobel, G.; et al. Prognostic value of indoleamine 2,3-dioxygenase expression in colorectal cancer: Effect on tumor-infiltrating T cells. Clin. Cancer Res. 2006, 12, 1144–1151. [Google Scholar] [CrossRef] [Green Version]
- Ino, K.; Yamamoto, E.; Shibata, K.; Kajiyama, H.; Yoshida, N.; Terauchi, M.; Nawa, A.; Nagasaka, T.; Takikawa, O.; Kikkawa, F. Inverse correlation between tumoral indoleamine 2,3-dioxygenase expression and tumor-infiltrating lymphocytes in endometrial cancer: Its association with disease progression and survival. Clin. Cancer Res. 2008, 14, 2310–2317. [Google Scholar] [CrossRef]
- Feng, G.S.; Dai, W.; Gupta, S.L.; Werner-Felmayer, G.; Wachter, H.; Takikawa, O.; Taylor, M.W. Analysis of interferon-gamma resistant mutants that are possibly defective in their signaling mechanism. Mol. Gen. Genet. 1991, 230, 91–96. [Google Scholar] [CrossRef]
- Litzenburger, U.M.; Opitz, C.A.; Sahm, F.; Rauschenbach, K.J.; Trump, S.; Winter, M.; Ott, M.; Ochs, K.; Lutz, C.; Liu, X.; et al. Constitutive IDO expression in human cancer is sustained by an autocrine signaling loop involving IL-6, STAT3 and the AHR. Oncotarget 2014, 5, 1038–1051. [Google Scholar] [CrossRef]
- Fujigaki, H.; Saito, K.; Fujigaki, S.; Takemura, M.; Sudo, K.; Ishiguro, H.; Seishima, M. The signal transducer and activator of transcription 1alpha and interferon regulatory factor 1 are not essential for the induction of indoleamine 2,3-dioxygenase by lipopolysaccharide: Involvement of p38 mitogen-activated protein kinase and nuclear factor-kappaB pathways, and synergistic effect of several proinflammatory cytokines. J. Biochem 2006, 139, 655–662. [Google Scholar] [CrossRef] [PubMed]
- Grohmann, U.; Orabona, C.; Fallarino, F.; Vacca, C.; Calcinaro, F.; Falorni, A.; Candeloro, P.; Belladonna, M.L.; Bianchi, R.; Fioretti, M.C.; et al. CTLA-4-Ig regulates tryptophan catabolism in vivo. Nat. Immunol. 2002, 3, 1097–1101. [Google Scholar] [CrossRef] [PubMed]
- Mellor, A.L.; Baban, B.; Chandler, P.; Marshall, B.; Jhaver, K.; Hansen, A.; Koni, P.A.; Iwashima, M.; Munn, D.H. Cutting edge: Induced indoleamine 2,3 dioxygenase expression in dendritic cell subsets suppresses T cell clonal expansion. J. Immunol. 2003, 171, 1652–1655. [Google Scholar] [CrossRef] [PubMed]
- Munn, D.H.; Sharma, M.D.; Hou, D.; Baban, B.; Lee, J.R.; Antonia, S.J.; Messina, J.L.; Chandler, P.; Koni, P.A.; Mellor, A.L. Expression of indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes. J. Clin. Investig. 2004, 114, 280–290. [Google Scholar] [CrossRef]
- Wang, Y.; Yang, B.H.; Li, H.; Cao, S.; Ren, X.B.; Yu, J.P. IDO(+) DCs and signalling pathways. Curr Cancer Drug Targets 2013, 13, 278–288. [Google Scholar] [CrossRef]
- Lanzinger, M.; Jurgens, B.; Hainz, U.; Dillinger, B.; Raberger, J.; Fuchs, D.; Heitger, A. Ambivalent effects of dendritic cells displaying prostaglandin E2-induced indoleamine 2,3-dioxygenase. Eur. J. Immunol. 2012, 42, 1117–1128. [Google Scholar] [CrossRef]
- Curti, A.; Pandolfi, S.; Valzasina, B.; Aluigi, M.; Isidori, A.; Ferri, E.; Salvestrini, V.; Bonanno, G.; Rutella, S.; Durelli, I.; et al. Modulation of tryptophan catabolism by human leukemic cells results in the conversion of CD25- into CD25+ T regulatory cells. Blood 2007, 109, 2871–2877. [Google Scholar] [CrossRef]
- Sioud, M.; Saeboe-Larssen, S.; Hetland, T.E.; Kaern, J.; Mobergslien, A.; Kvalheim, G. Silencing of indoleamine 2,3-dioxygenase enhances dendritic cell immunogenicity and antitumour immunity in cancer patients. Int. J. Oncol. 2013, 43, 280–288. [Google Scholar] [CrossRef]
- Liu, M.; Li, Z.; Yao, W.; Zeng, X.; Wang, L.; Cheng, J.; Ma, B.; Zhang, R.; Min, W.; Wang, H. IDO inhibitor synergized with radiotherapy to delay tumor growth by reversing T cell exhaustion. Mol. Med. Rep. 2020, 21, 445–453. [Google Scholar] [CrossRef]
- Brown, Z.J.; Yu, S.J.; Heinrich, B.; Ma, C.; Fu, Q.; Sandhu, M.; Agdashian, D.; Zhang, Q.; Korangy, F.; Greten, T.F. Indoleamine 2,3-dioxygenase provides adaptive resistance to immune checkpoint inhibitors in hepatocellular carcinoma. Cancer Immunol. Immunother. 2018, 67, 1305–1315. [Google Scholar] [CrossRef]
- Gomes, B.; Driessens, G.; Bartlett, D.; Cai, D.; Cauwenberghs, S.; Crosignani, S.; Dalvie, D.; Denies, S.; Dillon, C.P.; Fantin, V.R.; et al. Characterization of the Selective Indoleamine 2,3-Dioxygenase-1 (IDO1) Catalytic Inhibitor EOS200271/PF-06840003 Supports IDO1 as a Critical Resistance Mechanism to PD-(L)1 Blockade Therapy. Mol. Cancer Ther. 2018, 17, 2530–2542. [Google Scholar] [CrossRef] [PubMed]
- Le Naour, J.; Galluzzi, L.; Zitvogel, L.; Kroemer, G.; Vacchelli, E. Trial watch: IDO inhibitors in cancer therapy. Oncoimmunology 2020, 9, 1777625. [Google Scholar] [CrossRef] [PubMed]
- Keir, M.E.; Butte, M.J.; Freeman, G.J.; Sharpe, A.H. PD-1 and its ligands in tolerance and immunity. Annu. Rev. Immunol. 2008, 26, 677–704. [Google Scholar] [CrossRef] [PubMed]
- Abiko, K.; Matsumura, N.; Hamanishi, J.; Horikawa, N.; Murakami, R.; Yamaguchi, K.; Yoshioka, Y.; Baba, T.; Konishi, I.; Mandai, M. IFN-gamma from lymphocytes induces PD-L1 expression and promotes progression of ovarian cancer. Br. J. Cancer 2015, 112, 1501–1509. [Google Scholar] [CrossRef]
- Morimoto, Y.; Kishida, T.; Kotani, S.I.; Takayama, K.; Mazda, O. Interferon-beta signal may up-regulate PD-L1 expression through IRF9-dependent and independent pathways in lung cancer cells. Biochem. Biophys. Res. Commun. 2018, 507, 330–336. [Google Scholar] [CrossRef]
- Patel, S.A.; Minn, A.J. Combination Cancer Therapy with Immune Checkpoint Blockade: Mechanisms and Strategies. Immunity 2018, 48, 417–433. [Google Scholar] [CrossRef]
- Chaudhri, A.; Xiao, Y.; Klee, A.N.; Wang, X.; Zhu, B.; Freeman, G.J. PD-L1 Binds to B7-1 Only In Cis on the Same Cell Surface. Cancer Immunol. Res. 2018, 6, 921–929. [Google Scholar] [CrossRef] [Green Version]
- Wu, Y.; Chen, W.; Xu, Z.P.; Gu, W. PD-L1 Distribution and Perspective for Cancer Immunotherapy-Blockade, Knockdown, or Inhibition. Front. Immunol. 2019, 10, 2022. [Google Scholar] [CrossRef]
- Bar, N.; Costa, F.; Das, R.; Duffy, A.; Samur, M.; McCachren, S.; Gettinger, S.N.; Neparidze, N.; Parker, T.L.; Bailur, J.K.; et al. Differential effects of PD-L1 versus PD-1 blockade on myeloid inflammation in human cancer. JCI Insight 2020, 5, e129353. [Google Scholar] [CrossRef]
- Sanchez-Paulete, A.R.; Cueto, F.J.; Martinez-Lopez, M.; Labiano, S.; Morales-Kastresana, A.; Rodriguez-Ruiz, M.E.; Jure-Kunkel, M.; Azpilikueta, A.; Aznar, M.A.; Quetglas, J.I.; et al. Cancer Immunotherapy with Immunomodulatory Anti-CD137 and Anti-PD-1 Monoclonal Antibodies Requires BATF3-Dependent Dendritic Cells. Cancer Discov. 2016, 6, 71–79. [Google Scholar] [CrossRef]
- 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] [PubMed]
- Haymaker, C.L.; Kim, D.; Uemura, M.; Vence, L.M.; Phillip, A.; McQuail, N.; Brown, P.D.; Fernandez, I.; Hudgens, C.W.; Creasy, C.; et al. Metastatic Melanoma Patient Had a Complete Response with Clonal Expansion after Whole Brain Radiation and PD-1 Blockade. Cancer Immunol. Res. 2017, 5, 100–105. [Google Scholar] [CrossRef] [PubMed]
- Gaudreau, P.O.; Negrao, M.V.; Mitchell, K.G.; Reuben, A.; Corsini, E.M.; Li, J.; Karpinets, T.V.; Wang, Q.; Diao, L.; Wang, J.; et al. Neoadjuvant Chemotherapy Increases Cytotoxic T Cell, Tissue Resident Memory T Cell, and B Cell Infiltration in Resectable NSCLC. J. Thorac. Oncol. 2021, 16, 127–139. [Google Scholar] [CrossRef] [PubMed]
- Diab, A.; Tannir, N.M.; Bentebibel, S.E.; Hwu, P.; Papadimitrakopoulou, V.; Haymaker, C.; Kluger, H.M.; Gettinger, S.N.; Sznol, M.; Tykodi, S.S.; et al. Bempegaldesleukin (NKTR-214) plus Nivolumab in Patients with Advanced Solid Tumors: Phase I Dose-Escalation Study of Safety, Efficacy, and Immune Activation (PIVOT-02). Cancer Discov. 2020, 10, 1158–1173. [Google Scholar] [CrossRef] [PubMed]
- Ferris, R.L.; Blumenschein, G., Jr.; Fayette, J.; Guigay, J.; Colevas, A.D.; Licitra, L.; Harrington, K.; Kasper, S.; Vokes, E.E.; Even, C.; et al. Nivolumab for Recurrent Squamous-Cell Carcinoma of the Head and Neck. N. Engl. J. Med. 2016, 375, 1856–1867. [Google Scholar] [CrossRef]
- Ocadlikova, D.; Lecciso, M.; Broto, J.M.; Scotlandi, K.; Cavo, M.; Curti, A.; Palmerini, E. Sunitinib Exerts In Vitro Immunomodulatory Activity on Sarcomas via Dendritic Cells and Synergizes With PD-1 Blockade. Front. Immunol. 2021, 12, 577766. [Google Scholar] [CrossRef]
- Chu, T.H.; Vo, M.C.; Park, H.S.; Lakshmi, T.J.; Jung, S.H.; Kim, H.J.; Lee, J.J. Potent anti-myeloma efficacy of dendritic cell therapy in combination with pomalidomide and programmed death-ligand 1 blockade in a preclinical model of multiple myeloma. Cancer Immunol. Immunother. 2021, 70, 31–45. [Google Scholar] [CrossRef]
- Chen, Q.; Yin, H.; Pu, N.; Zhang, J.; Zhao, G.; Lou, W.; Wu, W. Chemokine C-C motif ligand 21 synergized with programmed death-ligand 1 blockade restrains tumor growth. Cancer Sci. 2021, 112, 4457–4469. [Google Scholar] [CrossRef]
- Walker, L.S.; Sansom, D.M. The emerging role of CTLA4 as a cell-extrinsic regulator of T cell responses. Nat. Rev. Immunol. 2011, 11, 852–863. [Google Scholar] [CrossRef]
- Selby, M.J.; Engelhardt, J.J.; Quigley, M.; Henning, K.A.; Chen, T.; Srinivasan, M.; Korman, A.J. Anti-CTLA-4 antibodies of IgG2a isotype enhance antitumor activity through reduction of intratumoral regulatory T cells. Cancer Immunol. Res. 2013, 1, 32–42. [Google Scholar] [CrossRef]
- Kirkwood, J.M.; Lorigan, P.; Hersey, P.; Hauschild, A.; Robert, C.; McDermott, D.; Marshall, M.A.; Gomez-Navarro, J.; Liang, J.Q.; Bulanhagui, C.A. Phase II trial of tremelimumab (CP-675,206) in patients with advanced refractory or relapsed melanoma. Clin. Cancer Res. 2010, 16, 1042–1048. [Google Scholar] [CrossRef] [PubMed]
- Ribas, A. Overcoming immunologic tolerance to melanoma: Targeting CTLA-4 with tremelimumab (CP-675,206). Oncologist 2008, 13 (Suppl. 4), 10–15. [Google Scholar] [CrossRef] [PubMed]
- Chiarion-Sileni, V.; Pigozzo, J.; Ascierto, P.A.; Simeone, E.; Maio, M.; Calabro, L.; Marchetti, P.; De Galitiis, F.; Testori, A.; Ferrucci, P.F.; et al. Ipilimumab retreatment in patients with pretreated advanced melanoma: The expanded access programme in Italy. Br. J. Cancer 2014, 110, 1721–1726. [Google Scholar] [CrossRef] [PubMed]
- Camacho, L.H.; Antonia, S.; Sosman, J.; Kirkwood, J.M.; Gajewski, T.F.; Redman, B.; Pavlov, D.; Bulanhagui, C.; Bozon, V.A.; Gomez-Navarro, J.; et al. Phase I/II trial of tremelimumab in patients with metastatic melanoma. J. Clin. Oncol. 2009, 27, 1075–1081. [Google Scholar] [CrossRef] [PubMed]
- Saenger, Y.M.; Wolchok, J.D. The heterogeneity of the kinetics of response to ipilimumab in metastatic melanoma: Patient cases. Cancer Immun. 2008, 8, 1. [Google Scholar]
- Du, X.; Tang, F.; Liu, M.; Su, J.; Zhang, Y.; Wu, W.; Devenport, M.; Lazarski, C.A.; Zhang, P.; Wang, X.; et al. A reappraisal of CTLA-4 checkpoint blockade in cancer immunotherapy. Cell Res. 2018, 28, 416–432. [Google Scholar] [CrossRef]
- Chen, X.; Shao, Q.; Hao, S.; Zhao, Z.; Wang, Y.; Guo, X.; He, Y.; Gao, W.; Mao, H. CTLA-4 positive breast cancer cells suppress dendritic cells maturation and function. Oncotarget 2017, 8, 13703–13715. [Google Scholar] [CrossRef]
- Esmaily, M.; Masjedi, A.; Hallaj, S.; Nabi Afjadi, M.; Malakotikhah, F.; Ghani, S.; Ahmadi, A.; Sojoodi, M.; Hassannia, H.; Atyabi, F.; et al. Blockade of CTLA-4 increases anti-tumor response inducing potential of dendritic cell vaccine. J. Control. Release 2020, 326, 63–74. [Google Scholar] [CrossRef]
- Son, C.H.; Bae, J.H.; Shin, D.Y.; Lee, H.R.; Choi, Y.J.; Jo, W.S.; Ho Jung, M.; Kang, C.D.; Yang, K.; Park, Y.S. CTLA-4 blockade enhances antitumor immunity of intratumoral injection of immature dendritic cells into irradiated tumor in a mouse colon cancer model. J. Immunother. 2014, 37, 1–7. [Google Scholar] [CrossRef]
- Urban-Wojciuk, Z.; Khan, M.M.; Oyler, B.L.; Fahraeus, R.; Marek-Trzonkowska, N.; Nita-Lazar, A.; Hupp, T.R.; Goodlett, D.R. The Role of TLRs in Anti-cancer Immunity and Tumor Rejection. Front. Immunol. 2019, 10, 2388. [Google Scholar] [CrossRef]
- Slattery, M.L.; Herrick, J.S.; Bondurant, K.L.; Wolff, R.K. Toll-like receptor genes and their association with colon and rectal cancer development and prognosis. Int. J. Cancer 2012, 130, 2974–2980. [Google Scholar] [CrossRef] [PubMed]
- Li, G.; Zheng, Z. Toll-like receptor 3 genetic variants and susceptibility to hepatocellular carcinoma and HBV-related hepatocellular carcinoma. Tumour Biol. 2013, 34, 1589–1594. [Google Scholar] [CrossRef] [PubMed]
- Yeyeodu, S.T.; Kidd, L.R.; Oprea-Ilies, G.M.; Burns, B.G.; Vancleave, T.T.; Shim, J.Y.; Kimbro, K.S. IRAK4 and TLR3 Sequence Variants may Alter Breast Cancer Risk among African-American Women. Front. Immunol. 2013, 4, 338. [Google Scholar] [CrossRef] [PubMed]
- Hasimu, A.; Ge, L.; Li, Q.Z.; Zhang, R.P.; Guo, X. Expressions of Toll-like receptors 3, 4, 7, and 9 in cervical lesions and their correlation with HPV16 infection in Uighur women. Chin. J. Cancer 2011, 30, 344–350. [Google Scholar] [CrossRef] [PubMed]
- Zeljic, K.; Supic, G.; Jovic, N.; Kozomara, R.; Brankovic-Magic, M.; Obrenovic, M.; Magic, Z. Association of TLR2, TLR3, TLR4 and CD14 genes polymorphisms with oral cancer risk and survival. Oral Dis. 2014, 20, 416–424. [Google Scholar] [CrossRef] [PubMed]
- Mitchell, D.; Yong, M.; Schroder, W.; Black, M.; Tirrell, M.; Olive, C. Dual stimulation of MyD88-dependent Toll-like receptors induces synergistically enhanced production of inflammatory cytokines in murine bone marrow-derived dendritic cells. J. Infect. Dis. 2010, 202, 318–329. [Google Scholar] [CrossRef]
- 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. Nat. Immunol. 2005, 6, 769–776. [Google Scholar] [CrossRef]
- Warger, T.; Osterloh, P.; Rechtsteiner, G.; Fassbender, M.; Heib, V.; Schmid, B.; Schmitt, E.; Schild, H.; Radsak, M.P. Synergistic activation of dendritic cells by combined Toll-like receptor ligation induces superior CTL responses in vivo. Blood 2006, 108, 544–550. [Google Scholar] [CrossRef] [Green Version]
- Guinn, Z.P.; Petro, T.M. IFN-gamma synergism with poly I:C reduces growth of murine and human cancer cells with simultaneous changes in cell cycle and immune checkpoint proteins. Cancer Lett. 2018, 438, 1–9. [Google Scholar] [CrossRef]
- Di, S.; Zhou, M.; Pan, Z.; Sun, R.; Chen, M.; Jiang, H.; Shi, B.; Luo, H.; Li, Z. Combined Adjuvant of Poly I:C Improves Antitumor Effects of CAR-T Cells. Front. Oncol. 2019, 9, 241. [Google Scholar] [CrossRef]
- Karapetyan, L.; Luke, J.J.; Davar, D. Toll-Like Receptor 9 Agonists in Cancer. Onco. Targets. Ther. 2020, 13, 10039–10060. [Google Scholar] [CrossRef] [PubMed]
- Sato-Kaneko, F.; Yao, S.; Ahmadi, A.; Zhang, S.S.; Hosoya, T.; Kaneda, M.M.; Varner, J.A.; Pu, M.; Messer, K.S.; Guiducci, C.; et al. Combination immunotherapy with TLR agonists and checkpoint inhibitors suppresses head and neck cancer. JCI Insight 2017, 2, e93397. [Google Scholar] [CrossRef] [PubMed]
- Murad, Y.M.; Clay, T.M.; Lyerly, H.K.; Morse, M.A. CPG-7909 (PF-3512676, ProMune): Toll-like receptor-9 agonist in cancer therapy. Expert. Opin. Biol. Ther. 2007, 7, 1257–1266. [Google Scholar] [CrossRef] [PubMed]
- Mullins, S.R.; Vasilakos, J.P.; Deschler, K.; Grigsby, I.; Gillis, P.; John, J.; Elder, M.J.; Swales, J.; Timosenko, E.; Cooper, Z.; et al. Intratumoral immunotherapy with TLR7/8 agonist MEDI9197 modulates the tumor microenvironment leading to enhanced activity when combined with other immunotherapies. J. Immunother. Cancer 2019, 7, 244. [Google Scholar] [CrossRef] [PubMed]
- Okamoto, M.; Furuichi, S.; Nishioka, Y.; Oshikawa, T.; Tano, T.; Ahmed, S.U.; Takeda, K.; Akira, S.; Ryoma, Y.; Moriya, Y.; et al. Expression of toll-like receptor 4 on dendritic cells is significant for anticancer effect of dendritic cell-based immunotherapy in combination with an active component of OK-432, a streptococcal preparation. Cancer Res. 2004, 64, 5461–5470. [Google Scholar] [CrossRef]
- Shetab Boushehri, M.A.; Lamprecht, A. TLR4-Based Immunotherapeutics in Cancer: A Review of the Achievements and Shortcomings. Mol. Pharm. 2018, 15, 4777–4800. [Google Scholar] [CrossRef]
- Sun, L.; Wu, J.; Du, F.; Chen, X.; Chen, Z.J. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 2013, 339, 786–791. [Google Scholar] [CrossRef]
- Ishikawa, H.; Ma, Z.; Barber, G.N. STING regulates intracellular DNA-mediated, type I interferon-dependent innate immunity. Nature 2009, 461, 788–792. [Google Scholar] [CrossRef] [Green Version]
- Ishikawa, H.; Barber, G.N. STING is an endoplasmic reticulum adaptor that facilitates innate immune signalling. Nature 2008, 455, 674–678. [Google Scholar] [CrossRef]
- Garcia-Diaz, A.; Shin, D.S.; Moreno, B.H.; Saco, J.; Escuin-Ordinas, H.; Rodriguez, G.A.; Zaretsky, J.M.; Sun, L.; Hugo, W.; Wang, X.; et al. Interferon Receptor Signaling Pathways Regulating PD-L1 and PD-L2 Expression. Cell Rep. 2017, 19, 1189–1201. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Cheng, H.; Yuan, H.; Xu, Q.; Shu, C.; Zhang, Y.; Xu, P.; Tan, J.; Rui, Y.; Li, P.; et al. Antitumor Activity of cGAMP via Stimulation of cGAS-cGAMP-STING-IRF3 Mediated Innate Immune Response. Sci. Rep. 2016, 6, 19049. [Google Scholar] [CrossRef] [PubMed]
- Marcus, A.; Mao, A.J.; Lensink-Vasan, M.; Wang, L.; Vance, R.E.; Raulet, D.H. Tumor-Derived cGAMP Triggers a STING-Mediated Interferon Response in Non-tumor Cells to Activate the NK Cell Response. Immunity 2018, 49, 754–763.e754. [Google Scholar] [CrossRef]
- Tang, C.H.; Zundell, J.A.; Ranatunga, S.; Lin, C.; Nefedova, Y.; Del Valle, J.R.; Hu, C.C. Agonist-Mediated Activation of STING Induces Apoptosis in Malignant B Cells. Cancer Res. 2016, 76, 2137–2152. [Google Scholar] [CrossRef] [PubMed]
- Zhang, C.X.; Ye, S.B.; Ni, J.J.; Cai, T.T.; Liu, Y.N.; Huang, D.J.; Mai, H.Q.; Chen, Q.Y.; He, J.; Zhang, X.S.; et al. STING signaling remodels the tumor microenvironment by antagonizing myeloid-derived suppressor cell expansion. Cell Death Differ. 2019, 26, 2314–2328. [Google Scholar] [CrossRef]
- Woo, S.R.; Fuertes, M.B.; Corrales, L.; Spranger, S.; Furdyna, M.J.; Leung, M.Y.; Duggan, R.; Wang, Y.; Barber, G.N.; Fitzgerald, K.A.; et al. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity 2014, 41, 830–842. [Google Scholar] [CrossRef] [PubMed]
- Demaria, O.; De Gassart, A.; Coso, S.; Gestermann, N.; Di Domizio, J.; Flatz, L.; Gaide, O.; Michielin, O.; Hwu, P.; Petrova, T.V.; et al. STING activation of tumor endothelial cells initiates spontaneous and therapeutic antitumor immunity. Proc. Natl. Acad. Sci. USA 2015, 112, 15408–15413. [Google Scholar] [CrossRef]
- Fu, J.; Kanne, D.B.; Leong, M.; Glickman, L.H.; McWhirter, S.M.; Lemmens, E.; Mechette, K.; Leong, J.J.; Lauer, P.; Liu, W.; et al. STING agonist formulated cancer vaccines can cure established tumors resistant to PD-1 blockade. Sci. Transl. Med. 2015, 7, 283ra252. [Google Scholar] [CrossRef]
- Sen, T.; Rodriguez, B.L.; Chen, L.; Corte, C.M.D.; Morikawa, N.; Fujimoto, J.; Cristea, S.; Nguyen, T.; Diao, L.; Li, L.; et al. Targeting DNA Damage Response Promotes Antitumor Immunity through STING-Mediated T-cell Activation in Small Cell Lung Cancer. Cancer Discov. 2019, 9, 646–661. [Google Scholar] [CrossRef] [Green Version]
- Wang, H.; Hu, S.; Chen, X.; Shi, H.; Chen, C.; Sun, L.; Chen, Z.J. cGAS is essential for the antitumor effect of immune checkpoint blockade. Proc. Natl. Acad. Sci. USA 2017, 114, 1637–1642. [Google Scholar] [CrossRef]
- Amouzegar, A.; Chelvanambi, M.; Filderman, J.N.; Storkus, W.J.; Luke, J.J. STING Agonists as Cancer Therapeutics. Cancers 2021, 13, 2695. [Google Scholar] [CrossRef]
- Mackey, M.F.; Gunn, J.R.; Maliszewsky, C.; Kikutani, H.; Noelle, R.J.; Barth, R.J., Jr. Dendritic cells require maturation via CD40 to generate protective antitumor immunity. J. Immunol. 1998, 161, 2094–2098. [Google Scholar] [PubMed]
- Kalantari, T.; Ciric, B.; Kamali-Sarvestani, E.; Rostami, A. Bone marrow dendritic cells deficient for CD40 and IL-23p19 are tolerogenic in vitro. Iran. J. Basic. Med. Sci. 2020, 23, 287–292. [Google Scholar] [CrossRef]
- Djureinovic, D.; Wang, M.; Kluger, H.M. Agonistic CD40 Antibodies in Cancer Treatment. Cancers 2021, 13, 1302. [Google Scholar] [CrossRef] [PubMed]
- van Mierlo, G.J.; den Boer, A.T.; Medema, J.P.; van der Voort, E.I.; Fransen, M.F.; Offringa, R.; Melief, C.J.; Toes, R.E. CD40 stimulation leads to effective therapy of CD40(-) tumors through induction of strong systemic cytotoxic T lymphocyte immunity. Proc. Natl. Acad. Sci. USA 2002, 99, 5561–5566. [Google Scholar] [CrossRef]
- Mangsbo, S.M.; Broos, S.; Fletcher, E.; Veitonmaki, N.; Furebring, C.; Dahlen, E.; Norlen, P.; Lindstedt, M.; Totterman, T.H.; Ellmark, P. The human agonistic CD40 antibody ADC-1013 eradicates bladder tumors and generates T-cell-dependent tumor immunity. Clin. Cancer Res. 2015, 21, 1115–1126. [Google Scholar] [CrossRef] [PubMed]
- Byrne, K.T.; Betts, C.B.; Mick, R.; Sivagnanam, S.; Bajor, D.L.; Laheru, D.A.; Chiorean, E.G.; O’Hara, M.H.; Liudahl, S.M.; Newcomb, C.; et al. Neoadjuvant Selicrelumab, an Agonist CD40 Antibody, Induces Changes in the Tumor Microenvironment in Patients with Resectable Pancreatic Cancer. Clin. Cancer Res. 2021, 27, 4574–4586. [Google Scholar] [CrossRef]
- Rech, A.J.; Dada, H.; Kotzin, J.J.; Henao-Mejia, J.; Minn, A.J.; Twyman-Saint Victor, C.; Vonderheide, R.H. Radiotherapy and CD40 Activation Separately Augment Immunity to Checkpoint Blockade in Cancer. Cancer Res. 2018, 78, 4282–4291. [Google Scholar] [CrossRef] [PubMed]
- Ngiow, S.F.; Young, A.; Blake, S.J.; Hill, G.R.; Yagita, H.; Teng, M.W.; Korman, A.J.; Smyth, M.J. Agonistic CD40 mAb-Driven IL12 Reverses Resistance to Anti-PD1 in a T-cell-Rich Tumor. Cancer Res. 2016, 76, 6266–6277. [Google Scholar] [CrossRef] [Green Version]
- Zhang, J.; Li, Y.; Yang, S.; Zhang, L.; Wang, W. Anti-CD40 mAb enhanced efficacy of anti-PD1 against osteosarcoma. J. Bone Oncol. 2019, 17, 100245. [Google Scholar] [CrossRef]
- Bajor, D.L.; Xu, X.; Torigian, D.A.; Mick, R.; Garcia, L.R.; Richman, L.P.; Desmarais, C.; Nathanson, K.L.; Schuchter, L.M.; Kalos, M.; et al. Immune activation and a 9-year ongoing complete remission following CD40 antibody therapy and metastasectomy in a patient with metastatic melanoma. Cancer Immunol. Res. 2014, 2, 1051–1058. [Google Scholar] [CrossRef]
- Johnson, P.; Challis, R.; Chowdhury, F.; Gao, Y.; Harvey, M.; Geldart, T.; Kerr, P.; Chan, C.; Smith, A.; Steven, N.; et al. Clinical and biological effects of an agonist anti-CD40 antibody: A Cancer Research UK phase I study. Clin. Cancer Res. 2015, 21, 1321–1328. [Google Scholar] [CrossRef] [PubMed]
- Sanborn Rachel, G.N.; O’Hara, M. 34th Annual Meeting & Pre-Conference Programs of the Society for Immunotherapy of Cancer (SITC 2019): Part 2: National Harbor, MD, USA. 10 November 2019. P827. Phase 1 study of the CD40 agonist monoclonal antibody (mAb); CDX-1140 alone and in combination with CDX-301 (rhFLT3L) in patients with advanced cancers. J. Immunother. Cancer 2019, 7, 283. [Google Scholar] [CrossRef]
- Browaeys, R.; Saelens, W.; Saeys, Y. NicheNet: Modeling intercellular communication by linking ligands to target genes. Nat. Methods 2020, 17, 159–162. [Google Scholar] [CrossRef] [PubMed]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Marmonti, E.; Oliva-Ramirez, J.; Haymaker, C. Dendritic Cells: The Long and Evolving Road towards Successful Targetability in Cancer. Cells 2022, 11, 3028. https://doi.org/10.3390/cells11193028
Marmonti E, Oliva-Ramirez J, Haymaker C. Dendritic Cells: The Long and Evolving Road towards Successful Targetability in Cancer. Cells. 2022; 11(19):3028. https://doi.org/10.3390/cells11193028
Chicago/Turabian StyleMarmonti, Enrica, Jacqueline Oliva-Ramirez, and Cara Haymaker. 2022. "Dendritic Cells: The Long and Evolving Road towards Successful Targetability in Cancer" Cells 11, no. 19: 3028. https://doi.org/10.3390/cells11193028
APA StyleMarmonti, E., Oliva-Ramirez, J., & Haymaker, C. (2022). Dendritic Cells: The Long and Evolving Road towards Successful Targetability in Cancer. Cells, 11(19), 3028. https://doi.org/10.3390/cells11193028