1,204
Views
26
CrossRef citations to date
0
Altmetric
Commentary

New generation of dendritic cell vaccines

&
Pages 259-264 | Received 28 Sep 2012, Accepted 08 Oct 2012, Published online: 04 Jan 2013

Abstract

Dendritic cells (DC) play a pivotal role in the induction and regulation of immune responses, including the induction of cytotoxic T lymphocytes (CTL) responses. These are essential for the eradication of cancers and pathogens including HIV and malaria, for which there are currently no effective vaccines. New developments in our understanding of DC biology have identified the key DC subset responsible for CTL induction, which is now an attractive candidate to target for vaccination. These DC are characterized by expression of novel markers Clec9A and XCR1, and a specialized capacity to cross-present antigen (Ag) from tumors and pathogens that do not directly infect DC. New generation DC vaccines that specifically target the cross-presenting DC in vivo have already demonstrated potential in preclinical animal models but the challenge remains to translate these findings into clinically efficacous vaccines in man. This has been greatly facilitated by the recent identification of the equivalent Clec9A+XCR1+ cross-presenting DC in human lymphoid tissues and peripheral tissues that are key sites for vaccination administration. These findings combined with further studies on DC subset biology have important implications for the design of new CTL-mediated vaccines.

DC Vaccines for the induction of CTL against pathogens and cancers

The success of currently available vaccines is reliant on their ability to induce serum neutralizing antibodies. However, for the development of prophylactic and therapeutic vaccines against cancer and pathogens including HIV, malaria and tuberculosis there is now a large body of evidence to suggest that the induction of cytotoxic T cell (CTL) responses are important to provide protection and control established disease. Despite intensive efforts to develop vaccines designed to induce CTL responses, there are currently no effective vaccines for these diseases. Dendritic cells (DC) are the key antigen-presenting cells responsible for the initiation of CTL-mediated immune responses against cancers, intracellular pathogens and viruses. The existence of multiple DC subsets with specialized functions is now apparent in mice but translating this to humans has been a major challenge. Several recent studies have provided new insights into the DC network in human tissues. These findings have significant implications for the design of CTL-mediated vaccines.

The Complex Network of DC: Multiple subsets with specialized functions

The DC network is comprised of multiple subsets that differ in their ontology, location, phenotype and specialized function. The first division, evident in both mouse and man, occurs between plasmacytoid DC (pDC) and myeloid DC, the latter also referred to as conventional DC (cDC). PDC produce large amounts of type I IFNCitation1,Citation2 and act as a first line of defense against viral pathogens, though their role in the priming T cell responses remains controversial.Citation3 By contrast, cDC are considered the “professional” antigen (Ag) presenting cells critical for the activation of naïve T cells.Citation4,Citation5 The cDC are further divided into “lymphoid-resident” DC and “migratory DC.” The lymphoid-resident DC arrive in lymphoid organs as blood-borne precursors that develop into immature DC where they monitor the blood, lymphatics or other DC for pathogens.Citation5-Citation7 In the mouse, lymphoid-resident DC are further segregated into CD8+ DC and CD8- DC based on their expression of the CD8α chain.Citation5 The migratory DC do not develop in the lymphoid organs, but in the peripheral sites that they then monitor and sample for Ag. In the steady-state, and at an increased rate upon activation in response to pathogens or host intrinsic signals of damage, migratory DC travel to lymphoid tissues.Citation8 During this process they upregulate their co-stimulatory molecules and proceed to directly present their Ag to T cellsCitation9 or share the captured Ag with lymphoid-resident DC.Citation6 There are multiple subsets of migratory DC depending on the location they survey.Citation10,Citation11 Significant functional specializations are seen between the CD103-CD11b+ (referred to as CD11b+ DC) and CD103+CD11bloDC (referred to as CD103+DC) and the Langerhans’ cells (CD207+CD11b+CD103).Citation4 Lastly, a separate DC population, termed “inflammatory DC,” originates from monocytes and develops rapidly in response to inflammation or infection. These DC probably most closely resemble the monocyte-derived DC generated in vitro in the presence of GM-CSF/IL-4.Citation12-Citation14

Defining Cross-Presenting DC and Their Role in CTL-Mediated Immunity

Although by definition all cDC are capable of processing and presenting Ag and priming naïve T cell responses, only a small subset of migratory and lymphoid-resident cDC specialize in “cross-presentation,” that is the ability to present exogenous Ag in the context of MHC class I. Typically only endogenous Ag is presented in the context of MHC class I but cross-presenting DC sample Ag from other cells, circumventing the need to be directly infected by pathogens to acquire their Ag to prime CTL. In the mouse, the lymphoid-resident CD8+ DC and migratory CD103+ DC are the main cross-presenting DC and are crucial for the induction of CTL responses against cancers, viruses and other pathogenic infections.Citation15-Citation18 Indeed, there is strong evidence that these two DC subsets are closely related. Both CD8+ DC and CD103+ DC have a similar transcriptional signature,Citation19 require BatF3,Citation15 Id2Citation20 and IRF8Citation18,Citation20 for development and arise from a common precursor.Citation20 Initially, CD8+ DC and CD103+ DC were thought to be entirely dependent on Batf3 for development as exemplified by their absence in Batf3 deficient mice, which retained all other DC subsets.Citation15,Citation18,Citation21 More recent data suggests that Batf and Batf2 can compensate for Batf3.Citation22 Despite this, CD8+ DC and CD103+ DC are often referred to as Batf3-dependent DC. Delivering Ag and adjuvant directly to these DC is an attractive strategy for the induction of CTL and is, hence, being pursued in preclinical models.Citation10 Since human DC do not express CD8α, and CD103 is broadly expressed, translating the biology of mouse DC to human DC has been problematic. The discovery of several novel molecules exclusively expressed by these DC has permitted more refined phenotyping and functional insights and, importantly as discussed below, the identification of the human equivalents.

Bridging the Gap between Mouse and Human DC: Identification of conserved markers

In human, cDC have been classically defined as blood-lineage-marker negative, MHC class II+ and CD11c+ and are subdivided into CD1c (BDCA-1)+ and CD141 (BDCA-3)+ DC. There is now convincing evidence from a number of groups using genomics, phenotypic and functional approaches that CD141+ DC in blood and lymphoid tissues are the human equivalents of the mouse lymphoid-resident CD8+ DC.Citation23-Citation26 Like their mouse counterpart, CD141+ DC are efficient at cross-presentation, express TLR3 and respond to TLR3 ligation by producing IFN-λ.Citation27 However, CD141 is not an ideal defining marker since it is widely expressed on human cells and entirely absent from all mouse DC. Thus, the requirement for conserved markers between species has continued and was only fulfilled with the identification of the C-type-lectin, Clec9A, the chemokine receptor, XCR1 and the nectin-like protein, CADM1 (Necl2). Clec9A is a receptor for dead cells and a regulator of cross-priming,Citation28-Citation30 while XCR1 and CADM1 play a role in CD8+ T cell stimulation.Citation31,Citation32 All three molecules are expressed on mouse CD8+ DC and the human equivalent CD141+ DC. Clec9A and XCR1 are particularly important as they provide the means to identify the human equivalent of the murine migratory CD103+ DC. In the mouse, only the CD8+ DC and CD103+ DC express XCR1Citation19,Citation33 and both DC subsets express Clec9A.Citation34-Citation37 Importantly, human CD141+ DC expressing Clec9A and/or XCR1 have now been identified in lymphoid and non-lymphoid tissues, including skin, lung and gut.Citation25,Citation26,Citation36,Citation38,Citation39 This suggests that the Clec9A+XCR1+CD141+DC in peripheral tissues are the human equivalent of the mouse Clec9A+XCR1+CD103+ DC, which survey the periphery and traffic to the lymphoid organs. Given the high degree of conservation in tissue localization and genomic, phenotypic and functional similarities, a unified identity for the cross-presenting DC across multiple tissue subtypes and species is now achievable. Since Clec9A and XCR1, in conjunction, are the most specific defining markers across tissues and species, we hereafter refer to this population of cross-presenting DC as Clec9A+XCR1+ DC.

Targeting Cross-Presenting DC for Immunotherapy

CTL are stimulated by activated DC that have processed and are presenting Ag in the context of MHC class I in the lymph nodes. Therapeutic vaccines utilizing peptides, recombinant proteins, viral vectors, tumor cells or lysates are “non-targeted” and rely on these agents being captured by local DC and transported to the draining lymph node for presentation to T cells. In an effort to enhance the amount of tumor Ag presented by DC, an alternative approach involved differentiating DC from monocytes in vitro, loading these with Ag and adjuvants and injecting these into patients as therapeutic vaccines. Unfortunately, these therapeutic DC-based cancer vaccines are expensive, labor-intensive, require customization for each patient and ultimately have been of limited clinical benefit.Citation40,Citation41 A more efficient vaccine strategy is to deliver the Ag directly to DC in vivo. This has been achieved by immunizing with monoclonal antibodies (mAb) that recognize cell surface receptors expressed on DC and carry antigenic cargo. Targeting Ag to the DC subsets that are ideally equipped for cross-presentation and priming of CTL responses would inherently seem advantageous. In the mouse, it is the Clec9A+XCR1+ DC that play a key role in the induction of CTL and since their counterpart is conserved in humans, delivering Ag to this DC subset via mAb that recognize Clec9A or XCR1 is an attractive vaccine strategy. This is now a viable option, first due to the development of anti-Clec9A and anti-XCR1 mAb that can deliver Ag specifically to CLEC9A+XCR1+ DC in vivo,Citation33,Citation42 mediating cross-presentation and CTL induction.Citation34,Citation37,Citation43,Citation44 Second, it is now clear that these cells are located in tissues such as skinCitation38 and lungCitation36 where vaccine administration (i.e., intradermal or intranasal) is not only practical but has been clinically demonstrated to be more effective with lower doses of Ag compared with the standard injection routes (i.e., intramuscular, subcutaneous).Citation45,Citation46

Targeting Cross-Presenting DC in the Mouse: What have we learnt?

In the mouse, extensive work has been published on targeting Ag to DEC-205, a multi-lectin receptor expressed at high levels of CD8+ DC (reviewed elsewhereCitation10). This body of work has made two clear observations. First, effective priming of CTL requires the delivery of Ag in the presence of DC activation/maturation signalsCitation47,Citation48 and second, targeting the subset of DC that cross-present results in superior CD8 T cell responses.Citation49,Citation50 In the mouse, many other receptors have been exploited for the delivery of Ag and these studies have made a number of other salient points.Citation51 For example, it is logical to assume that the best receptor for Ag-delivery should only be expressed on DC; indeed promiscuous expression by other APC may prove detrimental. However, targeting Ag to DEC-205,Citation48 CD36,Citation52 CD11cCitation53 and Clec12A,Citation44,Citation54 all of which are expressed on multiple cell types, induced strong CD8 T cell responses. Importantly though, while the broad expression patterns of these receptors did not prevent the induction of CTL, it was the DC and not the other cells that were responsible for the priming of T cell immunity.Citation54-Citation56 Our own data also warns that not all receptors expressed by CD8+ DC will automatically be good vaccine targets. In this vein, though DEC-205, Clec9A and Langerin were comparable at promoting CD8 T cell responses,Citation57 delivering Ag to Clec12A, which is also expressed on CD8+ DC (as well as other DC subsets and non-DC) was significantly less effective at promoting cross-presentation.Citation44 The capacity of the individual receptor to promote cross-priming is critical when considering it a vaccine target. We and others have already confirmed that targeting Ag to Clec9A is extremely effective at promoting the priming of CTLCitation37,Citation44 and generating protective anti-tumor responses.Citation37 The question remaining to be answered is whether XCR1, the other receptor exclusively expressed on the cross-priming DC can be used to induce CTL. Since an anti-XCR1 mAb has recently been generated,Citation33,Citation42 it will be possible to compare Ag delivery to these receptors and determine which is most effective at promoting the induction of CTL. The last point that needs consideration is whether the mAb itself may affect immune outcome. In the case of Clec9A, one mAb elicits humoral responses in the absence of adjuvants, while another requires adjuvant.Citation34,Citation43,Citation44,Citation51 Since neither mAb appears to directly activate DC, it is difficult to reconcile these differences and this is the subject of a current collaborative study. However, in terms of inducing CTL responses, both of these mAb to Clec9A require co-administered adjuvants for efficacy,Citation37,Citation44 clearly indicating this will be the optimal targeting protocol for future clinical trials.

What is the Role of Other DC-Subsets in Anti-Tumor and Anti-Viral Immunity?

In the mouse, the crucial role for Clec9A+XCR1+ DC in the induction of anti-tumoral and anti-viral immune responses has been established,Citation15,Citation18 though the individual contributions of migratory vs. lymphoid-resident Clec9A+XCR1+ DC remains unknown. Even less is known about the contribution of lymphoid-resident CD8- DC and CD11b+ migratory DC, to anti-tumor and anti-viral immunity. There is some evidence that at least some subtypes of these DC are specialized at inducing CD4+ T cell responses.Citation9,Citation58-Citation60 This may be an important consideration in vaccine design for maximizing CD4 T helper and humoral immune responses. Another important function of migratory CD11b+ cDC may be to transfer Ag to lymphoid-resident CLEC9A+XCR1+ DCCitation6 but the significance of this process in CTL-mediated immunity is yet to be elucidated. The absence of a unique transcription factor defining the CD11b+ cDC subsets makes it difficult to discern their function in vivo. There are currently no definitive markers that clearly align the CD11b+ cDC subsets across different tissues and species.

In the mouse, under inflammatory conditions, monocytes can also acquire DC-like featuresCitation12 and effectively cross-present targeted Ag.Citation61 The equivalent of these in vivo-induced monocyte-derived DC remain to be identified in humans. Whether these monocyte-derived DC can be exploited for Ag-delivery and induction of CTL remains to be determined. Ultimately, it is the identification of definitive new markers that will facilitate the translation of mouse DC-biology to human DC-biology and identify the role these DC subsets play in cross-priming.

Which Adjuvant Will Be Most Effective?

One of the most crucial lessons of DC clinical trials has been the requirement for DC activation in order to generate CTL responses.Citation40,Citation41 This is also a pre-requisite for the induction of CTL in mice when Ag is delivered via mAb in vivo.Citation10 Indeed, delivering Ag and adjuvant simultaneously to DC enhances immunogenicity.Citation62-Citation66 In terms of DC immunotherapy this infers that the adjuvant must be delivered to the same DC subset being targeted with Ag. TLR ligands such as poly I:C (TLR3), MPL (TLR4) and CpG (TLR9) have all demonstrated efficacy as potent vaccine adjuvants in mice and non-human primates and are now being trialed in humans. Poly I:C is particularly effective as an adjuvant for T cell immunization that is well-tolerated in humans and elicits a Type I IFN signature that mimics live virus infection.Citation67 The study of mouse and human DC have alerted to one important interspecies difference, namely while all mouse Clec9A+XCR1+ DC express and TLR3, 4, 9Citation68 and respond to these TLR-ligands, human Clec9A+XCR1+ DC only express TLR3.Citation25,,Citation69 In light of this information, poly I:C or its stabilized analog (LC:IC) is currently one of the most attractive adjuvants to incorporate into vaccines aimed at targeting human Clec9A+XCR1+ DC.

Concluding Remarks and Future Directions

The discovery of the Clec9A+XCR1+ cross-presenting DC in mouse and man has provided new insights into the induction of CTL responses against viruses and tumors and paves the way for rational vaccine designs. Key questions that remain to be answered are: which molecule is the best receptor for Ag delivery, which adjuvant is most effective and ultimately, will targeting this DC subset alone be sufficient to generate protective immunity? In this regard further characterization of less well defined DC subsets and their role in viral and tumor immunity is essential. Though Clec9A+XCR1+ DC have been analyzed in their steady-state, quantitative and qualitative changes in response to infection and cancer may affect the ability to therapeutically target these DC in vivo and will need to be carefully evaluated in human disease settings. For cancer, in vivo targeting of DC will likely be most effective in combination with other agents that overcome the immunosuppressive environment such as targeting CTLA4 or PD1, or enhancing “immunogenic” tumor cell death with chemotherapeutic agents.Citation70 For priming viral immune responses, DC targeting may be more effective when used in combination with other vaccines as a prime-boost strategy, as recently shown for the induction of HIV responses using DEC-205 targeting in non-human primates.Citation71 Finally, translating promising findings from preclinical animal models into effective human vaccines remains a major challenge. The profound advantages of targeting DEC-205 in mice were more modest in non-human primate studies.Citation71,Citation72 However, the first proof-of-concept clinical trials using DEC-205 to target DC in vivo in healthy volunteers are underwayCitation72 and the results are eagerly anticipated.

Abbreviations:
Ag=

antigen

CTL=

cytotoxic T lymphocytes

DC=

dendritic cells

cDC=

conventional DC

pDC=

plasmacytoid DC

mAb=

monoclonal antibodies

Acknowledgment

We would like to thank Prof W.R. Heath and Dr M. O’Keeffe for constructive criticism of this manuscript. K.J.R. is the recipient of NHMRC Career Development Fellowship, NHMRC project grant 604306 and Prostate Cancer Foundation of Australia project grant. I.C. was supported by the National Health and Medical Research Council of Australia (NHMRC) project grants 575546 and 1003355. This work was made possible through Victorian State Government Operational Infrastructure Support and Australian Government NHMRC IRIIS.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

References

  • Asselin-Paturel C, Boonstra A, Dalod M, Durand I, Yessaad N, Dezutter-Dambuyant C, et al. Mouse type I IFN-producing cells are immature APCs with plasmacytoid morphology. Nat Immunol 2001; 2:1144 - 50; http://dx.doi.org/10.1038/ni736; PMID: 11713464
  • Hochrein H, Shortman K, Vremec D, Scott B, Hertzog P, O’Keeffe M. Differential production of IL-12, IFN-alpha, and IFN-gamma by mouse dendritic cell subsets. J Immunol 2001; 166:5448 - 55; PMID: 11313382
  • Liu YJ. IPC: professional type 1 interferon-producing cells and plasmacytoid dendritic cell precursors. Annu Rev Immunol 2005; 23:275 - 306; http://dx.doi.org/10.1146/annurev.immunol.23.021704.115633; PMID: 15771572
  • Heath WR, Carbone FR. Dendritic cell subsets in primary and secondary T cell responses at body surfaces. Nat Immunol 2009; 10:1237 - 44; http://dx.doi.org/10.1038/ni.1822; PMID: 19915624
  • Shortman K, Heath WR. The CD8+ dendritic cell subset. Immunol Rev 2010; 234:18 - 31; http://dx.doi.org/10.1111/j.0105-2896.2009.00870.x; PMID: 20193009
  • Allan RS, Waithman J, Bedoui S, Jones CM, Villadangos JA, Zhan Y, et al. Migratory dendritic cells transfer antigen to a lymph node-resident dendritic cell population for efficient CTL priming. Immunity 2006; 25:153 - 62; http://dx.doi.org/10.1016/j.immuni.2006.04.017; PMID: 16860764
  • Sixt M, Kanazawa N, Selg M, Samson T, Roos G, Reinhardt DP, et al. The conduit system transports soluble antigens from the afferent lymph to resident dendritic cells in the T cell area of the lymph node. Immunity 2005; 22:19 - 29; http://dx.doi.org/10.1016/j.immuni.2004.11.013; PMID: 15664156
  • Wilson NS, Young LJ, Kupresanin F, Naik SH, Vremec D, Heath WR, et al. Normal proportion and expression of maturation markers in migratory dendritic cells in the absence of germs or Toll-like receptor signaling. Immunol Cell Biol 2008; 86:200 - 5; http://dx.doi.org/10.1038/sj.icb.7100125; PMID: 18026177
  • Bedoui S, Whitney PG, Waithman J, Eidsmo L, Wakim L, Caminschi I, et al. Cross-presentation of viral and self antigens by skin-derived CD103+ dendritic cells. Nat Immunol 2009; 10:488 - 95; http://dx.doi.org/10.1038/ni.1724; PMID: 19349986
  • Caminschi I, Maraskovsky E, Heath WR. Targeting Dendritic Cells in vivo for Cancer Therapy. Front Immunol 2012; 3:13; http://dx.doi.org/10.3389/fimmu.2012.00013; PMID: 22566899
  • Henri S, Poulin LF, Tamoutounour S, Ardouin L, Guilliams M, de Bovis B, et al. CD207+ CD103+ dermal dendritic cells cross-present keratinocyte-derived antigens irrespective of the presence of Langerhans cells. J Exp Med 2010; 207:S1-6 189 - 206; http://dx.doi.org/10.1084/jem.20091964; PMID: 20038600
  • Shortman K, Naik SH. Steady-state and inflammatory dendritic-cell development. Nat Rev Immunol 2007; 7:19 - 30; http://dx.doi.org/10.1038/nri1996; PMID: 17170756
  • Xu Y, Zhan Y, Lew AM, Naik SH, Kershaw MH. Differential development of murine dendritic cells by GM-CSF versus Flt3 ligand has implications for inflammation and trafficking. J Immunol 2007; 179:7577 - 84; PMID: 18025203
  • Robbins SH, Walzer T, Dembélé D, Thibault C, Defays A, Bessou G, et al. Novel insights into the relationships between dendritic cell subsets in human and mouse revealed by genome-wide expression profiling. Genome Biol 2008; 9:R17; http://dx.doi.org/10.1186/gb-2008-9-1-r17; PMID: 18218067
  • Hildner K, Edelson BT, Purtha WE, Diamond M, Matsushita H, Kohyama M, et al. Batf3 deficiency reveals a critical role for CD8alpha+ dendritic cells in cytotoxic T cell immunity. Science 2008; 322:1097 - 100; http://dx.doi.org/10.1126/science.1164206; PMID: 19008445
  • Mashayekhi M, Sandau MM, Dunay IR, Frickel EM, Khan A, Goldszmid RS, et al. CD8α(+) dendritic cells are the critical source of interleukin-12 that controls acute infection by Toxoplasma gondii tachyzoites. Immunity 2011; 35:249 - 59; http://dx.doi.org/10.1016/j.immuni.2011.08.008; PMID: 21867928
  • Piva L, Tetlak P, Claser C, Karjalainen K, Renia L, Ruedl C. Cutting edge: Clec9A+ dendritic cells mediate the development of experimental cerebral malaria. J Immunol 2012; 189:1128 - 32; http://dx.doi.org/10.4049/jimmunol.1201171; PMID: 22732587
  • Edelson BT, Wumesh KC, Juang R, Kohyama M, Benoit LA, Klekotka PA, et al. Peripheral CD103+ dendritic cells form a unified subset developmentally related to CD8alpha+ conventional dendritic cells. J Exp Med 2010; 207:823 - 36; http://dx.doi.org/10.1084/jem.20091627; PMID: 20351058
  • Crozat K, Tamoutounour S, Vu Manh TP, Fossum E, Luche H, Ardouin L, et al. Cutting edge: expression of XCR1 defines mouse lymphoid-tissue resident and migratory dendritic cells of the CD8α+ type. J Immunol 2011; 187:4411 - 5; http://dx.doi.org/10.4049/jimmunol.1101717; PMID: 21948982
  • Ginhoux F, Liu K, Helft J, Bogunovic M, Greter M, Hashimoto D, et al. The origin and development of nonlymphoid tissue CD103+ DCs. J Exp Med 2009; 206:3115 - 30; http://dx.doi.org/10.1084/jem.20091756; PMID: 20008528
  • Edelson BT, Bradstreet TR, Kc W, Hildner K, Herzog JW, Sim J, et al. Batf3-dependent CD11b(low/-) peripheral dendritic cells are GM-CSF-independent and are not required for Th cell priming after subcutaneous immunization. PLoS ONE 2011; 6:e25660; http://dx.doi.org/10.1371/journal.pone.0025660; PMID: 22065991
  • Tussiwand R, Lee WL, Murphy TL, Mashayekhi M, Kc W, Albring JC, et al. Compensatory dendritic cell development mediated by BATF-IRF interactions. Nature 2012; http://dx.doi.org/10.1038/nature11531; PMID: 22992524
  • Bachem A, Güttler S, Hartung E, Ebstein F, Schaefer M, Tannert A, 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 - 81; http://dx.doi.org/10.1084/jem.20100348; PMID: 20479115
  • Crozat K, Guiton R, Contreras V, Feuillet V, Dutertre CA, Ventre E, et al. The XC chemokine receptor 1 is a conserved selective marker of mammalian cells homologous to mouse CD8alpha+ dendritic cells. J Exp Med 2010; 207:1283 - 92; http://dx.doi.org/10.1084/jem.20100223; PMID: 20479118
  • Jongbloed SL, Kassianos AJ, McDonald KJ, Clark GJ, Ju X, Angel CE, et al. Human CD141+ (BDCA-3)+ dendritic cells (DCs) represent a unique myeloid DC subset that cross-presents necrotic cell antigens. J Exp Med 2010; 207:1247 - 60; http://dx.doi.org/10.1084/jem.20092140; PMID: 20479116
  • Poulin LF, Salio M, Griessinger E, Anjos-Afonso F, Craciun L, Chen JL, et al. Characterization of human DNGR-1+ BDCA3+ leukocytes as putative equivalents of mouse CD8alpha+ dendritic cells. J Exp Med 2010; 207:1261 - 71; http://dx.doi.org/10.1084/jem.20092618; PMID: 20479117
  • Lauterbach H, Bathke B, Gilles S, Traidl-Hoffmann C, Luber CA, Fejer G, et al. Mouse CD8alpha+ DCs and human BDCA3+ DCs are major producers of IFN-lambda in response to poly IC. J Exp Med 2010; 207:2703 - 17; http://dx.doi.org/10.1084/jem.20092720; PMID: 20975040
  • Ahrens S, Zelenay S, Sancho D, Hanč P, Kjær S, Feest C, et al. F-actin is an evolutionarily conserved damage-associated molecular pattern recognized by DNGR-1, a receptor for dead cells. Immunity 2012; 36:635 - 45; http://dx.doi.org/10.1016/j.immuni.2012.03.008; PMID: 22483800
  • Sancho D, Joffre OP, Keller AM, Rogers NC, Martínez D, Hernanz-Falcón P, et al. Identification of a dendritic cell receptor that couples sensing of necrosis to immunity. Nature 2009; 458:899 - 903; http://dx.doi.org/10.1038/nature07750; PMID: 19219027
  • Zhang JG, Czabotar PE, Policheni AN, Caminschi I, Wan SS, Kitsoulis S, et al. The dendritic cell receptor Clec9A binds damaged cells via exposed actin filaments. Immunity 2012; 36:646 - 57; http://dx.doi.org/10.1016/j.immuni.2012.03.009; PMID: 22483802
  • Dorner BG, Dorner MB, Zhou X, Opitz C, Mora A, Güttler S, et al. Selective expression of the chemokine receptor XCR1 on cross-presenting dendritic cells determines cooperation with CD8+ T cells. Immunity 2009; 31:823 - 33; http://dx.doi.org/10.1016/j.immuni.2009.08.027; PMID: 19913446
  • Galibert L, Diemer GS, Liu Z, Johnson RS, Smith JL, Walzer T, et al. Nectin-like protein 2 defines a subset of T-cell zone dendritic cells and is a ligand for class-I-restricted T-cell-associated molecule. J Biol Chem 2005; 280:21955 - 64; http://dx.doi.org/10.1074/jbc.M502095200; PMID: 15781451
  • Bachem A, Hartung E, Güttler S, Mora A, Zhou X, Hegemann A, et al. Expression of XCR1 Characterizes the Batf3-Dependent Lineage of Dendritic Cells Capable of Antigen Cross-Presentation. Front Immunol 2012; 3:214; http://dx.doi.org/10.3389/fimmu.2012.00214; PMID: 22826713
  • Caminschi I, Proietto AI, Ahmet F, Kitsoulis S, Shin Teh J, Lo JC, et al. The dendritic cell subtype-restricted C-type lectin Clec9A is a target for vaccine enhancement. Blood 2008; 112:3264 - 73; http://dx.doi.org/10.1182/blood-2008-05-155176; PMID: 18669894
  • Huysamen C, Willment JA, Dennehy KM, Brown GD. CLEC9A is a novel activation C-type lectin-like receptor expressed on BDCA3+ dendritic cells and a subset of monocytes. J Biol Chem 2008; 283:16693 - 701; http://dx.doi.org/10.1074/jbc.M709923200; PMID: 18408006
  • Poulin LF, Reyal Y, Uronen-Hansson H, Schraml BU, Sancho D, Murphy KM, et al. DNGR-1 is a specific and universal marker of mouse and human Batf3-dependent dendritic cells in lymphoid and nonlymphoid tissues. Blood 2012; 119:6052 - 62; http://dx.doi.org/10.1182/blood-2012-01-406967; PMID: 22442345
  • Sancho D, Mourão-Sá D, Joffre OP, Schulz O, Rogers NC, Pennington DJ, et al. Tumor therapy in mice via antigen targeting to a novel, DC-restricted C-type lectin. J Clin Invest 2008; 118:2098 - 110; http://dx.doi.org/10.1172/JCI34584; PMID: 18497879
  • Haniffa M, Shin A, Bigley V, McGovern N, Teo P, See P, et al. Human tissues contain CD141hi cross-presenting dendritic cells with functional homology to mouse CD103+ nonlymphoid dendritic cells. Immunity 2012; 37:60 - 73; http://dx.doi.org/10.1016/j.immuni.2012.04.012; PMID: 22795876
  • Mittag D, Proietto AI, Loudovaris T, Mannering SI, Vremec D, Shortman K, et al. Human dendritic cell subsets from spleen and blood are similar in phenotype and function but modified by donor health status. J Immunol 2011; 186:6207 - 17; http://dx.doi.org/10.4049/jimmunol.1002632; PMID: 21515786
  • Lesterhuis WJ, Aarntzen EH, De Vries IJ, Schuurhuis DH, Figdor CG, Adema GJ, et al. Dendritic cell vaccines in melanoma: from promise to proof?. Crit Rev Oncol Hematol 2008; 66:118 - 34; http://dx.doi.org/10.1016/j.critrevonc.2007.12.007; PMID: 18262431
  • Vulink A, Radford KJ, Melief C, Hart DN. Dendritic cells in cancer immunotherapy. Adv Cancer Res 2008; 99:363 - 407; http://dx.doi.org/10.1016/S0065-230X(07)99006-5; PMID: 18037410
  • Kroczek RA, Henn V. The Role of XCR1 and its Ligand XCL1 in Antigen Cross-Presentation by Murine and Human Dendritic Cells. Front Immunol 2012; 3:14; http://dx.doi.org/10.3389/fimmu.2012.00014; PMID: 22566900
  • Joffre OP, Sancho D, Zelenay S, Keller AM, Reis e Sousa C. Efficient and versatile manipulation of the peripheral CD4+ T-cell compartment by antigen targeting to DNGR-1/CLEC9A. Eur J Immunol 2010; 40:1255 - 65; http://dx.doi.org/10.1002/eji.201040419; PMID: 20333625
  • Lahoud MH, Ahmet F, Kitsoulis S, Wan SS, Vremec D, Lee CN, et al. Targeting antigen to mouse dendritic cells via Clec9A induces potent CD4 T cell responses biased toward a follicular helper phenotype. J Immunol 2011; 187:842 - 50; http://dx.doi.org/10.4049/jimmunol.1101176; PMID: 21677141
  • Belyakov IM, Ahlers JD. What role does the route of immunization play in the generation of protective immunity against mucosal pathogens?. J Immunol 2009; 183:6883 - 92; http://dx.doi.org/10.4049/jimmunol.0901466; PMID: 19923474
  • Lambert PH, Laurent PE. Intradermal vaccine delivery: will new delivery systems transform vaccine administration?. Vaccine 2008; 26:3197 - 208; http://dx.doi.org/10.1016/j.vaccine.2008.03.095; PMID: 18486285
  • Bonifaz L, Bonnyay D, Mahnke K, Rivera M, Nussenzweig MC, Steinman RM. Efficient targeting of protein antigen to the dendritic cell receptor DEC-205 in the steady state leads to antigen presentation on major histocompatibility complex class I products and peripheral CD8+ T cell tolerance. J Exp Med 2002; 196:1627 - 38; http://dx.doi.org/10.1084/jem.20021598; PMID: 12486105
  • Bonifaz LC, Bonnyay DP, Charalambous A, Darguste DI, Fujii S, Soares H, et al. In vivo targeting of antigens to maturing dendritic cells via the DEC-205 receptor improves T cell vaccination. J Exp Med 2004; 199:815 - 24; http://dx.doi.org/10.1084/jem.20032220; PMID: 15024047
  • Carter RW, Thompson C, Reid DM, Wong SY, Tough DF. Preferential induction of CD4+ T cell responses through in vivo targeting of antigen to dendritic cell-associated C-type lectin-1. J Immunol 2006; 177:2276 - 84; PMID: 16887988
  • Dudziak D, Kamphorst AO, Heidkamp GF, Buchholz VR, Trumpfheller C, Yamazaki S, et al. Differential antigen processing by dendritic cell subsets in vivo. Science 2007; 315:107 - 11; http://dx.doi.org/10.1126/science.1136080; PMID: 17204652
  • Caminschi I, Vremec D, Ahmet F, Lahoud MH, Villadangos JA, Murphy KM, et al. Antibody responses initiated by Clec9A-bearing dendritic cells in normal and Batf3(-/-) mice. Mol Immunol 2012; 50:9 - 17; http://dx.doi.org/10.1016/j.molimm.2011.11.008; PMID: 22209163
  • Tagliani E, Guermonprez P, Sepúlveda J, López-Bravo M, Ardavín C, Amigorena S, et al. Selection of an antibody library identifies a pathway to induce immunity by targeting CD36 on steady-state CD8 alpha+ dendritic cells. J Immunol 2008; 180:3201 - 9; PMID: 18292544
  • Wei H, Wang S, Zhang D, Hou S, Qian W, Li B, et al. Targeted delivery of tumor antigens to activated dendritic cells via CD11c molecules induces potent antitumor immunity in mice. Clin Cancer Res 2009; 15:4612 - 21; http://dx.doi.org/10.1158/1078-0432.CCR-08-3321; PMID: 19584156
  • Lahoud MH, Proietto AI, Ahmet F, Kitsoulis S, Eidsmo L, Wu L, et al. The C-type lectin Clec12A present on mouse and human dendritic cells can serve as a target for antigen delivery and enhancement of antibody responses. J Immunol 2009; 182:7587 - 94; http://dx.doi.org/10.4049/jimmunol.0900464; PMID: 19494282
  • He LZ, Crocker A, Lee J, Mendoza-Ramirez J, Wang XT, Vitale LA, et al. Antigenic targeting of the human mannose receptor induces tumor immunity. J Immunol 2007; 178:6259 - 67; PMID: 17475854
  • Nchinda G, Kuroiwa J, Oks M, Trumpfheller C, Park CG, Huang Y, et al. The efficacy of DNA vaccination is enhanced in mice by targeting the encoded protein to dendritic cells. J Clin Invest 2008; 118:1427 - 36; http://dx.doi.org/10.1172/JCI34224; PMID: 18324335
  • Idoyaga J, Lubkin A, Fiorese C, Lahoud MH, Caminschi I, Huang Y, et al. Comparable T helper 1 (Th1) and CD8 T-cell immunity by targeting HIV gag p24 to CD8 dendritic cells within antibodies to Langerin, DEC205, and Clec9A. Proc Natl Acad Sci USA 2011; 108:2384 - 9; http://dx.doi.org/10.1073/pnas.1019547108; PMID: 21262813
  • den Haan JM, Lehar SM, Bevan MJ. CD8(+) but not CD8(-) dendritic cells cross-prime cytotoxic T cells in vivo. J Exp Med 2000; 192:1685 - 96; http://dx.doi.org/10.1084/jem.192.12.1685; PMID: 11120766
  • Pooley JL, Heath WR, Shortman K. Cutting edge: intravenous soluble antigen is presented to CD4 T cells by CD8- dendritic cells, but cross-presented to CD8 T cells by CD8+ dendritic cells. J Immunol 2001; 166:5327 - 30; PMID: 11313367
  • Zhao X, Deak E, Soderberg K, Linehan M, Spezzano D, Zhu J, et al. Vaginal submucosal dendritic cells, but not Langerhans cells, induce protective Th1 responses to herpes simplex virus-2. J Exp Med 2003; 197:153 - 62; http://dx.doi.org/10.1084/jem.20021109; PMID: 12538655
  • Kamphorst AO, Guermonprez P, Dudziak D, Nussenzweig MC. Route of antigen uptake differentially impacts presentation by dendritic cells and activated monocytes. J Immunol 2010; 185:3426 - 35; http://dx.doi.org/10.4049/jimmunol.1001205; PMID: 20729332
  • Blander JM, Medzhitov R. Toll-dependent selection of microbial antigens for presentation by dendritic cells. Nature 2006; 440:808 - 12; http://dx.doi.org/10.1038/nature04596; PMID: 16489357
  • Hou B, Reizis B, DeFranco AL. Toll-like receptors activate innate and adaptive immunity by using dendritic cell-intrinsic and -extrinsic mechanisms. Immunity 2008; 29:272 - 82; http://dx.doi.org/10.1016/j.immuni.2008.05.016; PMID: 18656388
  • Kastenmüller K, Wille-Reece U, Lindsay RW, Trager LR, Darrah PA, Flynn BJ, et al. Protective T cell immunity in mice following protein-TLR7/8 agonist-conjugate immunization requires aggregation, type I IFN, and multiple DC subsets. J Clin Invest 2011; 121:1782 - 96; http://dx.doi.org/10.1172/JCI45416; PMID: 21540549
  • Spörri R, Reis e Sousa C. Inflammatory mediators are insufficient for full dendritic cell activation and promote expansion of CD4+ T cell populations lacking helper function. Nat Immunol 2005; 6:163 - 70; http://dx.doi.org/10.1038/ni1162; PMID: 15654341
  • Burgdorf S, Schölz C, Kautz A, Tampé R, Kurts C. Spatial and mechanistic separation of cross-presentation and endogenous antigen presentation. Nat Immunol 2008; 9:558 - 66; http://dx.doi.org/10.1038/ni.1601; PMID: 18376402
  • Caskey M, Lefebvre F, Filali-Mouhim A, Cameron MJ, Goulet JP, Haddad EK, et al. Synthetic double-stranded RNA induces innate immune responses similar to a live viral vaccine in humans. J Exp Med 2011; 208:2357 - 66; http://dx.doi.org/10.1084/jem.20111171; PMID: 22065672
  • Edwards AD, Diebold SS, Slack EM, Tomizawa H, Hemmi H, Kaisho T, et al. Toll-like receptor expression in murine DC subsets: lack of TLR7 expression by CD8 alpha+ DC correlates with unresponsiveness to imidazoquinolines. Eur J Immunol 2003; 33:827 - 33; http://dx.doi.org/10.1002/eji.200323797; PMID: 12672047
  • Kassianos AJ, Hardy MY, Ju X, Vijayan D, Ding Y, Vulink AJ, et al. Human CD1c (BDCA-1)+ myeloid dendritic cells secrete IL-10 and display an immuno-regulatory phenotype and function in response to Escherichia coli. Eur J Immunol 2012; 42:1512 - 22; http://dx.doi.org/10.1002/eji.201142098; PMID: 22678905
  • Galluzzi L, Senovilla L, Zitvogel L, Kroemer G. The secret ally: immunostimulation by anticancer drugs. Nat Rev Drug Discov 2012; 11:215 - 33; http://dx.doi.org/10.1038/nrd3626; PMID: 22301798
  • Flynn BJ, Kastenmüller K, Wille-Reece U, Tomaras GD, Alam M, Lindsay RW, et al. Immunization with HIV Gag targeted to dendritic cells followed by recombinant New York vaccinia virus induces robust T-cell immunity in nonhuman primates. Proc Natl Acad Sci USA 2011; 108:7131 - 6; http://dx.doi.org/10.1073/pnas.1103869108; PMID: 21467219
  • Trumpfheller C, Longhi MP, Caskey M, Idoyaga J, Bozzacco L, Keler T, et al. Dendritic cell-targeted protein vaccines: a novel approach to induce T-cell immunity. J Intern Med 2012; 271:183 - 92; http://dx.doi.org/10.1111/j.1365-2796.2011.02496.x; PMID: 22126373

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.