Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Nov 1.
Published in final edited form as: Immunol Lett. 2014 Jul 27;162(1 0 0):59–67. doi: 10.1016/j.imlet.2014.07.004

Targeting human dendritic cells in situ to improve vaccines

Kartik Sehgal 1, Kavita M Dhodapkar 2, Madhav V Dhodapkar 1
PMCID: PMC4506641  NIHMSID: NIHMS616926  PMID: 25072116

Abstract

Dendritic cells (DCs) provide a critical link between innate and adaptive immunity. The potent antigen presenting properties of DCs makes them a valuable target for the delivery of immunogenic cargo. Recent clinical studies describing in situ DC targeting with antibodymediated targeting of DC receptor through DEC-205 provide new opportunities for the clinical application of DC-targeted vaccines. Further advances with nanoparticle vectors which can encapsulate antigens and adjuvants within the same compartment and be targeted against diverse DC subsets also represent an attractive strategy for targeting DCs. This review provides a brief summary of the rationale behind targeting dendritic cells in situ, the existing pre-clinical and clinical data on these vaccines and challenges faced by the next generation DC-targeted vaccines.

Keywords: Dendritic cell, targeted vaccines, nanoparticles

1. Introduction

Vaccines represent one of the major success stories of modern medicine [1]. However in spite of considerable effort, it has proven harder to develop effective vaccines against certain pathogens (such as human immune deficiency virus and tuberculosis), and chronic diseases (such as cancer) wherein strong cell-mediated immunity is desired [2-4]. The major goal of vaccination against these conditions is generation of high avidity antigen-specific CD8+ T cells capable of cytotoxic T lymphocyte (CTL) response and generation of long-lived memory cells [4,5].

Dendritic cells (DCs) are specialized antigen-presenting cells (APCs) that play a central role in initiating and regulating immunity [6]. DCs efficiently capture both foreign and self-antigens from the environment and process and present them to T cells [6]. They induce differential immune responses according to the accompanying stimulus and thus regulate development of immunity or tolerance [7,8]. Owing to their potent antigen presentation capacity and ability to generate distinct T cell responses, they have received particular attention in the field of immunotherapy.

2. Dendritic cells as potent antigen presenting cells

Dendritic cell regulate innate as well as acquired immunity and serve as a bridge between these two arms. They possess intrinsic specialized features which make them particularly efficient to capture, process and present antigens [9]. Firstly, DCs are present at the self-environment intersection (i.e. skin and mucosal surfaces) and hence strategically located to encounter pathogens and other foreign material. Secondly, they have specialized uptake receptors and downstream endocytic system for antigen processing and presentation (classical MHC molecules I and II for presentation of peptides, and CD1d system for presentation of lipid antigens). The specialized surface or intracellular receptors, called pattern recognition receptors (PRRs), include C-lectin type receptors (CLRs), Toll-like receptors (TLRs), NOD-like receptors (NLRs), RIG-1 like receptors (RLRs) and helicases [7,10,11]. Thirdly, they undergo a process called maturation on exposure to a wide range of stimuli or ‘danger signals’ (bacterial lipopolysaccharide, viral nucleic acids etc.) which are recognized by TLRs, NLRs and RLRs. It is now well appreciated that vaccine adjuvants act by inducing DC maturation, which improves antigen processing and presentation [9]. Several TLR agonists [Poly I:C (TLR3 agonist), MPLA (TLR4 agonist), CpG ODN (TLR9 agonist) and Resiquimod/ R848 (TLR7/8 agonist)] have thus been administered along with vaccines to deliver concomitant DC activation signals. Lastly, they comprise of multiple subsets with distinct location, phenotype and function, and differential expression of specialized receptors [12,13]. These receptors can be used to target specific subsets through incorporation of monoclonal antibodies in the vaccines [14,15]. These subsets respond uniquely to different stimuli and thus contribute to the generation of a broad spectrum of immune responses.

3. Diversity and biology of human dendritic cell subsets

Human dendritic cells have been typically divided into blood and cutaneous subsets for classification purposes, largely because these compartments are easier to study in humans. Blood DCs are further sub-classified into three categories- BDCA2 (CD303)+ plasmacytoid, BDCA1 (CD1c)+ myeloid and BDCA3 (CD141)+ myeloid DCs [16-19]. Cutaneous DCs comprise of epidermal (Langerhans cells) and dermal (CD14+ DCs and CD1a+ myeloid) DCs [16]. Another distinct category, inflammatory DCs are putatively derived from monocytes unlike the above mentioned DC subsets which are derived from bone marrow precursors [16,20]. These inflammatory DCs have distinct functions, dependent upon the inflammatory environment [16,21]. The properties of different DC subsets have been succinctly described in reviews [3,16,22,23], with some key features described below and in Table 1.

Table 1. Major human dendritic cell subsets.

Blood DCs Cutaneous DCs Inflammatory
DCs
Myeloid DCs
Type I
Myeloid DCs
Type II
Plasmacytoid
DCs
Langerhans Cells CD14+ DCs
Phenotype BDCA1 (CD1c)+ CD11c+ CD13+ CD33+ CD11b+ BDCA3 (CD141)+ CD11c+ CD13+ CD33+ CD11b+ BDCA2 (CD303)+ BDCA4 (CD304)+ CD123 (IL-3R α)+ ILT7+ Langerin+, CD1a+ CD14+, CD11c+ CD14+, CD11c+
Location Blood, tissue & lymphoid organs Blood, tissue & lymphoid organs Blood, tissue & lymphoid organs Epidermis & stratified squamous epithelia Dermis & non-lymphoid organs Site of inflammation
TLRs TLR 1-8, 10 TLR 1, 2, 3, 6, 8, 10 TLR 7, 9 TLR 1, 2, 3, 6, 10 TLR 2, 4, 5, 6, 8, 10
Other receptors Dectin-1, Dectin-2, DCIR, Mannose receptor CLEC9A, XCR1, NECL2 DCIR, CD32, Siglec-H, BST-2 DCIR, Dectin-1, Dectin-2 DC-SIGN, LOX-1, Dectin-1, DCIR SIRPα, FcεR
Major cytokines produced on activation TNF-α, IL-8, IL-10,IL-23 IFN-λ, TNF-α, CXCL10 IFN-α, IL-6, IL12 p40 IL-15 IL-1β, IL-6, IL-8, IL-10, IL-12, GM-CSF, MCP, TGF- β IL-1β, TNF-α, IL-6, IL-23
Major function Antigen-presentation Antigen-presentation/cross-presentation Anti-viral response, Maintenance of tolerance Tolerance/immune-regulation; epithelial homeostasis Formation of follicular helper T cells, B cell help Depends upon inflammatory environment
Predominant immune response Th2, Th1, Th17 Th1 Type I IFN, Tregs Th22, Th1 Tfh
*

Th= T helper response, Tfh= T follicular helper response

Myeloid DCs (MDCs) are the major antigen-presenting cells. Out of the BDCA1+ and BDCA3+ MDCs, the latter constitutes a minor, yet significant subset with superior cross antigen-presentation capacity [24-27]. Plasmacytoid dendritic cells (PDCs), on the other hand, secrete large amounts of interferon-alpha on exposure to viruses [28,29] as well as maintain tolerance against self-antigens [30,31]. This may explain why their dysfunction has been linked to the pathogenesis of autoimmune conditions such as systemic lupus erythematosus and immune thrombocytopenic purpura [32,33]. Langerhans cells (LCs) display a striking duality of function. They can prime T cell immunity as well as induce regulatory and IL-22 secreting T cells [34-36]. Therefore the role of LCs has evolved in recent years to include their tolerogenic function and broader roles in epithelial homeostasis [16]. Dermal CD14+ DCs, on the other hand, primarily stimulate humoral immunity [34,36-38].

4. Human versus mouse dendritic cell subsets

The human counterparts for the two most studied mouse DC subsets - CD8α+ and CD8α- DCs are BDCA3+ MDCs [26,27] and BDCA1+ MDCs respectively. Plasmacytoid DCs, on the other hand, are shared by both human and murine immune system. Although the majority of TLRs and CLRs on the major DC subsets are common in both human and mouse counterparts, clear differences exist. TLR9 which is found in all murine major DC subsets, is expressed only by PDCs in humans [12]. Other examples include CLRs DC-SIGN [39] and DC-ASPGR [40] whose biology differ between murine and human DCs.

The major murine DC subsets - CD8α+/DEC205+ and CD8α-/DCIR+ DCs show a remarkable division of labor in terms of their predominant response. While CD8α+ DCs efficiently cross-prime CD8+ T cell immunity through MHC class I antigen presentation [41], CD8α- DCs stimulate predominant CD4+ T cell response through MHC class II presentation [42]. This has been explained partly by some inherent characteristics of CD8α+ DCs- high endosomal pH, low antigen degradation, high antigen export to cytosol and more pre-synthesized stores of MHC class I molecules [22,43]. In humans, BDCA3+ MDCs were initially described to have superior cross-presentation capability than other DC subsets, however the cross-presentation capacity is not restricted to this subset [27]. Chatterjee et al found that cross-presentation capacity of human DCs was highly influenced by antigen delivery and whether antigens were delivered to early or late endosomes [44]. While late endosomal delivery through DEC205 maintained the superiority of BDCA3+ MDCs over BDCA1+ MDCs, this was eliminated on antigen delivery to early endosomes through CD40 or CD11c [45]. Another study showed all freshly isolated tonsilresident DC subsets – BDCA1+ MDCs, BDCA3+ MDCs and PDCs- possessed similar antigen cross-presentation capacity [46]. All three DC subsets could export proteins into cytosol efficiently and both BDCA1+ and BDCA3+ MDCs displayed similar phagosomal pH and production of reactive oxygen species. These findings are supported by numerous other studies where other DC subsets – PDCs [47-49], BDCA1+ MDCs [26,50], Langerhans cells [34,35] and CD1a+ DCs in skin-draining lymph nodes [51] cross-primed efficient CD8+ T cell immunity in culture.

5. Ex vivo as opposed to in situ dendritic cell targeting

Efficient and specific delivery of antigens to dendritic cells is the cornerstone for generating strong immune responses. Two major strategies have been utilized to engage dendritic cells [52]. The first approach involves ex vivo loading of autologous DCs with antigens/adjuvants and re-injecting them into patients while the second one targets DCs in situ through vaccine conjugated to DC receptor-specific monoclonal antibodies. Most studies to date have focused on adoptive transfer of DCs and found it to be safe and immunogenic [52-54]. Two broad approaches have been tried, injection of naturally occurring DCs, or differentiation of DCs from progenitors ex vivo, before adoptive transfer. Adoptive transfer of naturally occurring DCs is best exemplified by Sipuleucel-T therapy, which involves isolation and ex-vivo culture of patient’s APCs with prostatic acid phosphatase (PAP) and GM-CSF fusion protein. GM-CSF is added in addition to PAP antigen to promote activation of APCs, manifest as increased expression of HLA class II, co-stimulatory molecules and secretion of cytokines [55]. A large, randomized, double-blind, placebo-controlled phase III trial (IMPACT study) showed a median survival benefit of 4.1 months following Sipuleucel-T, leading to FDA approval for treatment of asymptomatic or minimally symptomatic patients with metastatic prostate cancer [56]. Ex-vivo vaccines were also tested in melanoma using patients’ plasmacytoid dendritic cells loaded with tumor antigen-associated peptides. Specific CD4+ and CD8+ T cells were generated in addition to a much desirable interferon signature [57]. However, unlike the above two studies which employed naturally occurring DCs, majority of the work with ex-vivo DC vaccines utilized monocyte-derived dendritic cells (Mo-DCs), which are not physiological DCs. These studies included treatment of patients with melanoma [58], breast [59] and ovarian cancer [60] and HIV infection [61,62], as well generation of tolerogenic response in autoimmune conditions such as rheumatoid arthritis and multiple sclerosis [63,64]. Of note, decrease in HIV viral load was reported in two studies after injection of DCs loaded ex vivo with chemically inactivated autologous virus [61,62]. A summary of clinical trials using ex vivo DC vaccines is provided in a recent review [52]. Widespread application of adoptive DC transfer has been limited by cost, labor requirements and technical complexity of the procedure [13,65]. Targeting dendritic cells in situ will circumvent these problems and provide readily available off-the-shelf products. Moreover, after ex vivo injections, DCs need to migrate to lymph nodes, while in case of in situ targeting, vaccines can be directly targeted to desired DC subsets present in desired locations [12].

6. Antibody-based targeting – lessons learnt from preclinical and early clinical studies targeting DEC205

The pioneering studies in the field of in situ DC targeting by Steinman and Nussenzweig laboratories through anti-DEC205 antibody constructs laid the groundwork for the clinical development of these vaccines [15,66,67]. A critical finding was the generation of tolerance when antigens were targeted to steady state DCs [15]. Application of adjuvants along with targeted vaccines, however, led to the generation of protective antigen-specific cellular immunity. This led to further testing of vaccines targeting DCs via antibodies to generate protective and therapeutic immunity against chronic infections and cancer (Table 2) [9].

Table 2. DC targeted antibody-based vaccines.

Antigen (Ag) Target Adjuvant Key findings Ref.
Animal in vitro and in vivo

Model Ag:
• OVA DEC-205 CD40 Ab Enhanced resistance to rapidly growing tumor and mucosal viral infection. [105,106]
CpG Ab-Ag-adjuvant conjugates more effective than Ab-Ag conjugates + soluble adjuvant or Ab-free Ag-adjuvant conjugates. [107]
Clec9A/DNGR-1 CD40 Ab /Poly IC Promoted tumor immunity. No adjuvant led to Treg differentiation, addition of Poly I:C led to Th1 & curdlan to Th17 polarization [76] [108]
DC-SIGN CD40 Ab Protective against infection with OVA expressing L. monocytogenes [109]
Dectin-1 Poly IC Preferential induction of CD4 T cell and Ab response on targeting to CD8a- DCs. [106]
Mannose R CpG CpG enhanced effector T cell immunity and Ag-specific protective tumor immunity [110]
BST-2 Poly IC / R848 Targeted to PDCs. Protective immunity against subsequent viral infection & tumor growth [80,111,112]
Siglec-H - / CpG Targeted to PDCs or cDCs. Inferior to BST-2 & DEC-205 targeting [111,113]
• TT & KLH DC-SIGN - Inhibited tumor growth in NOD/SCID mice [39]
Infectious Ag:
• HIV gag p24 DEC-205 Poly IC
Poly dA:dT
Most effective adjuvant and matured DCs by inducing Type I IFN response
Type I IFN production & improved IFN-y+ CD8 & CD4 T cell response
[68,70,114-116]
Clec9A/ DNGR-1 Poly IC / CD40 Ab Comparable stimulation of Th1 & CD8 immunity by CD8a+ DC targeting by Clec9A, Langerin & DEC205. Greater than targeting to CD8a- DCs through DCIR. [69]
• Dengue NS1 DEC-205 Poly IC Protection from lethal intracranial challenge with DENV2 NGC strain. In comparison, DCIR targeting led to Ab response only [71]
• Leishmania LmSTI1a DEC-205 PolyICLC & CD40 Ab Multi-epitope Th1 CD4+ T cell immunity, protective against L. major challenge. [117]
• Mycobacterial ESX Ag DEC-205 Poly IC Targeted to airway CD205+ DCs. Significant protection against virulent M.tuberculosis [118]
• EBNA1 DEC-205 PolyICLC Targeted to CD141+ cDCs. Protective against autologous EBV-transformed B cells [119]
• Influenza HA1 LOX-1 - Ag-specific IFN-y producing CD4+ T cells in non-human primates [40]
Tumor Ag:
• HER2/neu DEC-205 PolyICLC Protective against neu-expressing mammary tumor challenge [120]
• PSA fusion protein LOX-1 - Ag-specific IFN-y producing CD4+ T cells in non-human primates [40]

Human in vitro

Model Ag:
• TT DC-SIGN - Targeted delivery induced durable T cell responses in vaccinated donors [75]
• KLH Clec9A Poly IC & R848 Induced recall CD4 T cell response [93]
DCIR CpG Targeting to PDCs caused proliferation of PBLs [121]
Infectious Ag:
• HIV gag p24 DEC-205 - Superior to DC-SIGN/ CD209 targeting [122]
• 5 HIV peptide regions CD40 CD40 Ab Broad repertoire of multifunctional CD8 T cells generated with cytotoxic effects, ability to kill autologous target cells & suppress viral replication in vitro [123]
Tumor Ag:
•gp100/ pmel17 DC-SIGN Poly IC & R848 Equally efficient as cell-penetrating peptide PolyR at cross-presenting antigens [124]
Mannose R CD40L Cytotoxicity towards gp100 (+) HLA-matched melanoma targets [125]
• MART-1 Dectin-1 - Targeted to IFNDCs efficiently cross-presented Ag to stimulate functionally active CD8+ T cell responses. [78]
DCIR CL075, poly IC, LPS or CD40L Targeted to ex-vivo generated DCs, skin Langerhans cells, and blood MDCs & PDCs. CL075 (TLR7/8) agonist found to be the most potent adjuvant, especially when combined with CD40L [35]
• hCG beta Mannose R CD40L or poly IC & R848 CTLs capable of killing human cancer cell lines. [126,127]

Clinical trials

Tumor Ag:
• NY-ESO1 DEC-205 Resiquimod + Poly-ICLC Phase I trial reported robust humoral and cellular immunity against NY-ESO1. Stabilization of disease in 13 out of 48 patients, with tumor regression in 2 patients [81]
• hCG beta Mannose R Poly-ICLC + Resiquimod Phase I studies in patients with advanced epithelial malignancies. Humoral and T-cell responses seen. [128]
*

Ab = Antibody, Mannose R = Mannose receptor, CD40L = CD40 Ligand

Both these principles have been evaluated in numerous preclinical studies. Targeting DCs through DEC205 in the presence of adjuvants (TLR3, TLR7/8 or CD40 ligands) led to protective immunity against pathogens (HIV [68-70], tuberculosis and dengue [71]) and cancer. On the other hand, DEC205 targeting in the absence of DC activators leads to tolerance in experimental models of type 1 diabetes mellitus and experimental allergic encephalomyelitis [72-74]. Similarly, targeting through other DC surface receptors such as DC-SIGN [75], CLEC9A [69,76], DCIR [35,77], Dectin-1 [78] and Langerin [69,79] along with adjuvants stimulated integrated humoral and cellular immune responses.

It is important to note the plasticity of DC subsets which are capable of generating differential immune responses when targeted through different DC receptors [42,67]. In one study, targeting human DCs in the absence of adjuvant through DC-ASPGR led to the generation of IL-10 producing suppressive CD4+ T cells, while targeting through LOX-1 led to stimulation of IFN-γ producing CD4+ T cells [40]. In another study, where vaccines were targeted to both conventional and plasmacytoid murine DCs, Siglec-H targeting was found inferior to initiate either MHC-I or MHC-II antigen presentation, compared to BST-2 or DEC205 targeting [80].

Recently, in situ DC targeting through soluble antigen-DC receptor antibody construct was tested in a phase I clinical trial using CDX-1401 [81]. This vaccine (CDX1401, Celldex Therapeutics, Hampton, NJ, USA) consisted of a human anti-DEC205 monoclonal antibody fused to full-length tumor antigen NY-ESO-1 and was administered along with TLR agonists resiquimod (TLR 7/8 agonist) and Hiltonol (polyICLC, TLR3 agonist). Intracutaneous injection (combination of intradermal and subcutaneous injection) along with topical or subcutaneous administration of adjuvants led to generation of robust humoral and cellular immunity against NY-ESO-1. This was observed even in patients where NY-ESO-1 expression was not present in the patient tumor. Thirteen out of forty-eight patients had stabilization of disease with a median duration of 6.7 months (2.4+ to 13.4 months). Additionally, two patients experienced tumor regression. The vaccine did not result in any Grade 3/4 or dose-limiting toxicities. This first in-human study of a protein- antibody construct vaccine targeting DCs demonstrated that these vaccines are immunogenic, safe and well-tolerated. Of note, 6 out of 8 patients (75%) who received immune-checkpoint inhibitors within 3 months of receiving CDX-1401 had objective clinical responses. Of these patients, clinical responses were observed in 4 of 6 melanoma patients who received Ipilumimab following CDX-1401. These findings are encouraging but preliminary and need to be confirmed in the context of formal clinical trial testing this combination. These data nonetheless provide support for clinical studies to test whether combining DC targeted vaccines with strategies such as immune check-point inhibitors will lead to improved efficacy compared to immune checkpoint inhibitors alone. This approach may be particularly relevant for patients lacking immunity to tumor antigens at baseline prior to checkpoint blockade[82].

7. Emerging approaches – Nanoparticles

Another approach for in situ targeting that is approaching the clinic is to encapsulate antigens and adjuvants within delivery vehicles [12]. This will also eliminate the requirement for systemic administration of adjuvants and the consequent untoward systemic effects. Co-delivering antigens and adjuvants within the same compartment will also ensure that only the APC exposed to antigen receives the activation signal. This would prevent the dual problems of T cell anergy in the absence of co-stimulation and non-specific activation of APCs which have not seen the antigen. It would also allow delivery of high dose of immunogenic cargo, all within the same vector [65].

Nanoparticles (NP) are rapidly emerging as the new vehicles for delivering vaccines [83]. These include polymeric particles, liposomes, virus-like particles (VLPs), nanocrystals and immune-stimulating complexes (ISCOMs). These particles are efficiently taken up by DCs because of their size and particulate structure which resembles pathogens. They can induce long-lasting immune responses by delivering antigens in a slow and sustained manner [84]. Importantly, their release properties can be easily controlled by modulating their physic-chemical properties. Of these, poly-lactic-co-glycolic acid (PLGA) nanoparticles have received the most attention because of their production from a biodegradable, FDA approved polymer. Liposomes and virus-like particles have also been extensively studied, but their clinical application may be limited by the stability issues with liposomes and vector immunogenicity issues with VLPs [12]. While this review focuses on DC-targeted nanoparticle-based vaccines, recent reviews [83,85,86] provide excellent summaries of bioengineering issues with nanoparticles. Although DC-targeted NPs have not been tested in the clinic, the use of NPs as vaccine-delivery vehicles has already reached the clinic. For example, in a phase I/II clinical trial involving stage II-IV melanoma patients, VLPs loaded with Melan-A/Mart-1 peptide along with CpG led to tumorspecific CD8 T cell responses in 14 out of 22 patients [87].

8. Preclinical data with nanoparticle-based DC-targeted approaches

Nanoparticles can be decorated on their surface with antibodies or carbohydrate ligands that bind specifically to DC receptors. While polymer nanoparticles and liposomes can be coated with antibodies by PEGylation or avidin-biotin interactions, virus-like particles can also be engineered to express receptor ligands. In one of the earlier studies, Cruz et al demonstrated that DC-SIGN targeted PLGA nanoparticles, but not microparticles specifically delivered antigens to human dendritic cells in vitro [88]. Consequently, only targeted nanoparticles were able to improve antigen presentation and T cell response. Mannan bound PLGA nanoparticles were also found to improve antigen-specific CD4+ and CD8+ T cell responses in mouse in vitro and in vivo systems [89]. Interestingly, both of these studies were able to achieve these results in the absence of TLR agonists, which is consistent with the possibility that NPs themselves provide an activation signal to DCs.

The role of TLR agonists was evaluated by Tacken et al by targeting through DC-SIGN in human and DEC205 in mouse studies [90]. Co-encapsulating TLR3 and TLR7/8 ligands (poly IC and resiquimod/ R848 respectively) with the antigen in PLGA nanoparticles in this study did improve the generation of CTL responses. Of note, targeted delivery of TLR agonists reduced their dose requirement by 100 fold and was associated with decreased serum cytokine storm and related toxicities in vivo, compared to administration of soluble adjuvants. Similar results were achieved with mannose-targeted liposomes which showed higher anti-tumor therapeutic efficacy in vivo compared to non-targeted liposomes, thereby allowing use of lesser quantities of both TLR ligands and peptide epitopes [91]. On comparison between PLGA NP coated with either DC receptor-specific antibodies or carbohydrate ligands, targeting through former was shown to be more efficient to target dendritic cells and induce immune responses [92]. Targeting to specific human dendritic cell subsets has also been evaluated. BDCA3+ MDCs targeted via PLGA NP through CLEC9A efficiently presented melanoma-associated antigens to CD4+ T cells as well as cross-presented them to CD8+ T cells [93]. Human plasmacytoid dendritic cells also cross-presented antigens delivered via PLGA NP co-encapsulating R848 and targeted through DEC205, DCIR, BDCA-2 or FcyR CD32. Notably, the presence of TLR agonist led to robust type I interferon secretion, a desirable effect in immune activation [94]. A summary of selected studies where nanoparticle-based vaccines were actively targeted to dendritic cells is provided in Table 3.

Table 3. DC targeted nanoparticle-based vaccines.

Vector Antigen (Ag) Target Targeting moiety Key Findings Ref.
Animal in vitro and in vivo

Polymer Particles Model Ag:
• OVA DEC205 Monoclonal Ab 100 fold less adjuvant required, as compared to soluble forms. Reduced serum cytokine storm and related toxicity [90]
DC-SIGN PMAM dendrimers Robust CD4+ & CD8+ T cell responses when loaded to BMDCs derived from DC-SIGN-transgenic mice [129]
Mannose R Mannan Enhanced CD4+ and CD8+ T cell responses in vitro and in vivo [89]
Mannosylated-alginate Targeted nanogel delivery induced more efficient Th1 response in vitro [130]

Liposomes Model Ag:
• OVA DEC205 Monoclonal Ab Strong Ag-specific CTL response in splenic T cells and marked protection against tumor growth. LPS or IFN-y used as adjuvant. [131]
DC-SIGN Glycans Only glycan-modified non-PEGylated liposomes could bind to DC-SIGN [132]
Mannose R Glycan ligands Enhanced cross-presentation and Th1 skewing. Found to be independent of TLR-mediated signaling [133]
TLR5 Flagellin-related peptides Induced DC maturation and Ag-specific CD8 & humoral immunity, which significantly inhibited tumor growth/metastasis &induced complete tumor regression in majority of mice tumor models [134]
Infectious Ag:
• HIV gp120 18IIIB Mannose R Oligomannose – neoglycolipid These coated liposomes were proposed as adjuvants. Induced CD8+ CTL response. [135]
• Neisseria PorA Mannose R Ligands On subcutaneous immunization, Increased localization in draining lymph nodes and improved bactericidal Ab response [136]
Tumor Ag:
• ErbB2 p63-71 Mannose R Mannosylated ligands Mannose-targeted liposomes had higher anti-tumor therapeutic efficiency allowing use of lesser quantities of both TLR ligands & peptide epitopes. TLR2/6 agonists found to be more efficient than TLR1/2 agonist for tumor eradication [91]
• HPV16 E7 Mannose R Mannan Coating of DOTAP liposomes with mannan significantly enhanced both preventive & therapeutic anti-tumor effects in vivo [137]

Viruses /Virus-likeparticles Model Ag:
• OVA DC-SIGN Engineered Sindbis viral glycoprotein Specific transduction and maturation of DCs by lentivector. Generated Ag-specific CD8+T cells and significant Ab response. Protected against tumor growth and induced regression of established tumors [138]
Tumor Ag:
• TRP2180-188 CD40 CD40L extracellular domain Targeted adenovector enhanced both transduction & maturation of DCs. Improved CD8+ T cell immunity and therapeutic efficacy in a melanoma model [139]

Human in vitro

Polymer Particles Model Ag:
• TT DC-SIGN Monoclonal Ab Nanoparticles, but not microparticles improved antigen presentation. No adjuvant used [88]
Tumor Ag:
• gp100 DC-SIGN Monoclonal Ab / carbohydrate ligands Targeting antigens and adjuvants within the same particles enhanced CD8+ T cell stimulation potential. Receptor-specific antibodies more effective than carbohydrates [90,92]
Clec9A Monoclonal Ab Cross-presented by BDCA3+ DCs. Adjuvants R848 & poly IC [93]
DEC205, DCIR, BDCA2, FcR CD32 Monoclonal Ab Delivered to plasmacytoid dendritic cells. Adjuvant R848. Triggered robust Type I IFN response [94]

Liposomes Tumor Ag:
• NY-ESO-1 Fcy-R Fc fragment from IgG Co-encapsulated with adjuvants Palm-IL-1 & MAP-IFN-y, generated potent immunological responses [140]
•LHRH Fcy-R Fc fragment from IgG Enhanced immune response compared to non-targeted NP & soluble peptides [141]

Viruses /Virus-like particles Infectious Ag:
• Haem-influenza M1 CD40 Monoclonal Ab Fv fragment Superior ability to activate antigen-specific cytotoxic T lymphocyte response, compared to non-targeted adenoviral vectors [142]
*

Fcy= constant fragment of IgG, Fv= variable fragment.

9. Unmet needs for nanoparticle-based strategies

As discussed earlier, an important aspect of DC biology is the presence of distinct subsets specialized for distinct effects on the immune system. However in terms of in situ DC targeting in humans, questions regarding the optimal DC subset, target receptor and adjuvant still remain unanswered [12]. It is notable that recent studies have challenged the superiority of BDCA3+ MDCs over other DC subsets to cross-present antigens in humans [45,46]. It is also increasingly appreciated that generation of optimal T helper-1 (Th-1) immunity may require the engagement of multiple DC subsets [95-97]. This is supported by the finding that the yellow fever vaccine 17D, one of the most effective vaccines in recent history, activates multiple TLRs in DC subsets [98]. In a recent study, we have shown that combinatorial targeting of BDCA3 and DC-SIGN+ DCs via NPs was superior to targeting either subset alone. The mechanism underlying this synergy involved IL15-dependent DC-DC crosstalk [99]. Therefore, active targeting of nanoparticle-based vaccines to a single DC subset, though effective in the pre-clinical studies, may deprive the resultant immune response of the benefit of cross-talk between different DC subsets. One possible strategy to target multiple DC subsets in situ is to target receptors such as CD40 or CD32, which are expressed by multiple DC subsets and also mediate DC maturation [100-102].

The ideal DC activation signal or TLR ligand for these vaccines also remains to be defined. This may depend on the specific DC subset being targeted, as different subsets express different TLRs. Importantly, prior studies have shown that co-encapsulating more than one TLR agonist within NP significantly improved CTL responses, when compared to single agonist vaccines [103,104]. The ideal approach would be to generate a nanoparticle-based platform targeting combination of DC subsets which yield synergistic effects.

One potential advantage of NP platform is the potential flexibility in terms of antigen loading. This is potentially very valuable in the setting of cancer as antigenic peptides specific for mutations could be loaded onto NPs. Ultimately, this should pave the way for development of truly personalized cancer vaccines.

10. Conclusions

To conclude, active targeting of dendritic cells in situ is emerging as an attractive approach to generate strong protective cellular immunity against chronic infectious diseases and cancer. The recently reported phase I trial of human in-situ CDX-1401 has laid the foundation for clinical application of these vaccines. The observed clinical responses in patients receiving immune checkpoint blockade following the vaccine suggest that combining these vaccines with immune-checkpoint blockade (such as anti-CTLA4, anti-PD1) may be of therapeutic benefit in human cancer. Nanoparticles are also emerging as attractive vehicles to target antigens to DCs and recent data suggest that combinatorial targeting of multiple DC subsets may significantly enhance the efficacy of DC targeting. The development of such combinatorial approaches would allow us to harness the full potential of the human immune system in the fight against cancer and chronic infections [9].

Highlights.

  • The specialized antigen-presentation capability of dendritic cells and the plasticity of different DC subsets make them valuable targets for immunotherapy.

  • The success of recently reported phase I trial of NY-ESO1-anti-DEC205 antibody vaccine has set the stage for further clinical testing of in-situ DC-targeted vaccines.

  • Nanoparticles also represent an attractive strategy for targeting DCs in situ.

Acknowledgments

MVD and KMD are supported by funds from NIH (CA135110, CA106802, AI079222), Dana Foundation, and Multiple Myeloma Research Foundation.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.Bachmann MF, Jennings GT. Vaccine delivery: a matter of size, geometry, kinetics and molecular patterns. Nature reviews Immunology. 2010;10:787–796. doi: 10.1038/nri2868. [DOI] [PubMed] [Google Scholar]
  • 2.Nabel GJ. Designing tomorrow's vaccines. The New England journal of medicine. 2013;368:551–560. doi: 10.1056/NEJMra1204186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Palucka K, Banchereau J. Human dendritic cell subsets in vaccination. Curr Opin Immunol. 2013;25:396–402. doi: 10.1016/j.coi.2013.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Palucka K, Banchereau J. Dendritic-cell-based therapeutic cancer vaccines. Immun. 2013;39:38–48. doi: 10.1016/j.immuni.2013.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Appay V, Douek DC, Price DA. CD8+ T cell efficacy in vaccination and disease. Nature medicine. 2008;14:623–628. doi: 10.1038/nm.f.1774. [DOI] [PubMed] [Google Scholar]
  • 6.Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–252. doi: 10.1038/32588. [DOI] [PubMed] [Google Scholar]
  • 7.Mazzoni A, Segal DM. Controlling the Toll road to dendritic cell polarization. Journal of leukocyte biology. 2004;75:721–730. doi: 10.1189/jlb.1003482. [DOI] [PubMed] [Google Scholar]
  • 8.Kwissa M, Kasturi SP, Pulendran B. The science of adjuvants. Expert review of vaccines. 2007;6:673–684. doi: 10.1586/14760584.6.5.673. [DOI] [PubMed] [Google Scholar]
  • 9.Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature. 2007;449:419–426. doi: 10.1038/nature06175. [DOI] [PubMed] [Google Scholar]
  • 10.Desmet CJ, Ishii KJ. Nucleic acid sensing at the interface between innate and adaptive immunity in vaccination. Nature reviews Immunology. 2012;12:479–491. doi: 10.1038/nri3247. [DOI] [PubMed] [Google Scholar]
  • 11.Figdor CG, van Kooyk Y, Adema GJ. C-type lectin receptors on dendritic cells and Langerhans cells. Nat Rev Immunol. 2002;2:77–84. doi: 10.1038/nri723. [DOI] [PubMed] [Google Scholar]
  • 12.Kreutz M, Tacken PJ, Figdor CG. Targeting dendritic cells--why bother? Blood. 2013;121:2836–2844. doi: 10.1182/blood-2012-09-452078. [DOI] [PubMed] [Google Scholar]
  • 13.Radford KJ, Caminschi I. New generation of dendritic cell vaccines. Human vaccines & immunotherapeutics. 2013;9:259–264. doi: 10.4161/hv.22487. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Mahnke K, Guo M, Lee S, Sepulveda H, Swain SL, Nussenzweig M, et al. The dendritic cell receptor for endocytosis, DEC-205, can recycle and enhance antigen presentation via major histocompatibility complex class II-positive lysosomal compartments. The Journal of cell biology. 2000;151:673–684. doi: 10.1083/jcb.151.3.673. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Hawiger D, Inaba K, Dorsett Y, Guo M, Mahnke K, Rivera M, et al. Dendritic cells induceperipheral T cell unresponsiveness under steady state conditions in vivo. The Journal of experimental medicine. 2001;194:769–779. doi: 10.1084/jem.194.6.769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Collin M, McGovern N, Haniffa M. Human dendritic cell subsets. Immunology. 2013;140:22–30. doi: 10.1111/imm.12117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Dzionek A, Fuchs A, Schmidt P, Cremer S, Zysk M, Miltenyi S, et al. 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. doi: 10.4049/jimmunol.165.11.6037. [DOI] [PubMed] [Google Scholar]
  • 18.MacDonald KP, Munster DJ, Clark GJ, Dzionek A, Schmitz J, Hart DN. Characterization of human blood dendritic cell subsets. Blood. 2002;100:4512–4520. doi: 10.1182/blood-2001-11-0097. [DOI] [PubMed] [Google Scholar]
  • 19.Ziegler-Heitbrock L, Ancuta P, Crowe S, Dalod M, Grau V, Hart DN, et al. Nomenclature of monocytes and dendritic cells in blood. Blood. 2010;116:e74–80. doi: 10.1182/blood-2010-02-258558. [DOI] [PubMed] [Google Scholar]
  • 20.Doulatov S, Notta F, Eppert K, Nguyen LT, Ohashi PS, Dick JE. Revised map of the human progenitor hierarchy shows the origin of macrophages and dendritic cells in early lymphoid development. Nature immunology. 2010;11:585–593. doi: 10.1038/ni.1889. [DOI] [PubMed] [Google Scholar]
  • 21.Hansel A, Gunther C, Ingwersen J, Starke J, Schmitz M, Bachmann M, et al. Human slan (6-sulfo LacNAc) dendritic cells are inflammatory dermal dendritic cells in psoriasis and drive strong TH17/TH1 T-cell responses. The Journal of allergy and clinical immunology. 2011;127:787–794. e781–789. doi: 10.1016/j.jaci.2010.12.009. [DOI] [PubMed] [Google Scholar]
  • 22.Segura E, Amigorena S. Cross-presentation by human dendritic cell subsets. Immunology letters. 2014;158:73–78. doi: 10.1016/j.imlet.2013.12.001. [DOI] [PubMed] [Google Scholar]
  • 23.Ueno H, Klechevsky E, Schmitt N, Ni L, Flamar AL, Zurawski S, et al. Targeting human dendritic cell subsets for improved vaccines. Semin Immunol. 2011;23:21–27. doi: 10.1016/j.smim.2011.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.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;207:1261–1271. doi: 10.1084/jem.20092618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.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;207:2703–2717. doi: 10.1084/jem.20092720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.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. The Journal of experimental medicine. 2010;207:1247–1260. doi: 10.1084/jem.20092140. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Bachem A, Guttler 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. The Journal of experimental medicine. 2010;207:1273–1281. doi: 10.1084/jem.20100348. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Colonna M, Krug A, Cella M. Interferon-producing cells: on the front line in immune responses against pathogens. Current opinion in immunology. 2002;14:373–379. doi: 10.1016/s0952-7915(02)00349-7. [DOI] [PubMed] [Google Scholar]
  • 29.Siegal FP, Kadowaki N, Shodell M, Fitzgerald-Bocarsly PA, Shah K, Ho S, et al. The nature of the principal type 1 interferon-producing cells in human blood. Science. 1999;284:1835–1837. doi: 10.1126/science.284.5421.1835. [DOI] [PubMed] [Google Scholar]
  • 30.Moseman EA, Liang X, Dawson AJ, Panoskaltsis-Mortari A, Krieg AM, Liu YJ, et al. Human plasmacytoid dendritic cells activated by CpG oligodeoxynucleotides induce the generation of CD4+CD25+ regulatory T cells. J Immunol. 2004;173:4433–4442. doi: 10.4049/jimmunol.173.7.4433. [DOI] [PubMed] [Google Scholar]
  • 31.Ito T, Yang M, Wang YH, Lande R, Gregorio J, Perng OA, et al. Plasmacytoid dendritic cells prime IL-10-producing T regulatory cells by inducible costimulator ligand. The Journal of experimental medicine. 2007;204:105–115. doi: 10.1084/jem.20061660. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Gilliet M, Cao W, Liu YJ. Plasmacytoid dendritic cells: sensing nucleic acids in viral infection and autoimmune diseases. Nature reviews Immunology. 2008;8:594–606. doi: 10.1038/nri2358. [DOI] [PubMed] [Google Scholar]
  • 33.Sehgal K, Guo X, Koduru S, Shah A, Lin A, Yan X, et al. Plasmacytoid dendritic cells, interferon signaling, and FcgammaR contribute to pathogenesis and therapeutic response in childhood immune thrombocytopenia. Science translational medicine. 2013;5:193ra189. doi: 10.1126/scitranslmed.3006277. [DOI] [PubMed] [Google Scholar]
  • 34.Klechevsky E, Morita R, Liu M, Cao Y, Coquery S, Thompson-Snipes L, et al. Functional specializations of human epidermal Langerhans cells and CD14+ dermal dendritic cells. Immunity. 2008;29:497–510. doi: 10.1016/j.immuni.2008.07.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Klechevsky E, Flamar AL, Cao Y, Blanck JP, Liu M, O'Bar A, et al. Cross-priming CD8+ T cells by targeting antigens to human dendritic cells through DCIR. Blood. 2010;116:1685–1697. doi: 10.1182/blood-2010-01-264960. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Banchereau J, Thompson-Snipes L, Zurawski S, Blanck JP, Cao Y, Clayton S, et al. The differential production of cytokines by human Langerhans cells and dermal CD14(+) DCs controls CTL priming. Blood. 2012;119:5742–5749. doi: 10.1182/blood-2011-08-371245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Schmitt N, Bustamante J, Bourdery L, Bentebibel SE, Boisson-Dupuis S, Hamlin F, et al. IL-12 receptor beta1 deficiency alters in vivo T follicular helper cell response in humans. Blood. 2013;121:3375–3385. doi: 10.1182/blood-2012-08-448902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Matthews K, Chung NP, Klasse PJ, Moore JP, Sanders RW. Potent induction of antibody-secreting B cells by human dermal-derived CD14+ dendritic cells triggered by dual TLR ligation. J Immunol. 2012;189:5729–5744. doi: 10.4049/jimmunol.1200601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kretz-Rommel A, Qin F, Dakappagari N, Torensma R, Faas S, Wu D, et al. In vivo targeting of antigens to human dendritic cells through DC-SIGN elicits stimulatory immune responses and inhibits tumor growth in grafted mouse models. J Immunother. 2007;30:715–726. doi: 10.1097/CJI.0b013e318135472c. [DOI] [PubMed] [Google Scholar]
  • 40.Li D, Romain G, Flamar AL, Duluc D, Dullaers M, Li XH, et al. Targeting self- and foreign antigens to dendritic cells via DC-ASGPR generates IL-10-producing suppressive CD4+ T cells. The Journal of experimental medicine. 2012;209:109–121. doi: 10.1084/jem.20110399. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Shortman K, Heath WR. The CD8+ dendritic cell subset. Immunological reviews. 2010;234:18–31. doi: 10.1111/j.0105-2896.2009.00870.x. [DOI] [PubMed] [Google Scholar]
  • 42.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;315:107–111. doi: 10.1126/science.1136080. [DOI] [PubMed] [Google Scholar]
  • 43.Savina A, Peres A, Cebrian I, Carmo N, Moita C, Hacohen N, et al. The small GTPase Rac2 controls phagosomal alkalinization and antigen crosspresentation selectively in CD8(+) dendritic cells. Immunity. 2009;30:544–555. doi: 10.1016/j.immuni.2009.01.013. [DOI] [PubMed] [Google Scholar]
  • 44.Chatterjee B, Smed-Sorensen A, Cohn L, Chalouni C, Vandlen R, Lee BC, et al. Internalization and endosomal degradation of receptor-bound antigens regulate the efficiency of cross presentation by human dendritic cells. Blood. 2012;120:2011–2020. doi: 10.1182/blood-2012-01-402370. [DOI] [PubMed] [Google Scholar]
  • 45.Cohn L, Chatterjee B, Esselborn F, Smed-Sorensen A, Nakamura N, Chalouni C, et al. Antigen delivery to early endosomes eliminates the superiority of human blood BDCA3+ dendritic cells at cross presentation. J Exp Med. 2013;210:1049–1063. doi: 10.1084/jem.20121251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.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. doi: 10.1084/jem.20121103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Hoeffel G, Ripoche AC, Matheoud D, Nascimbeni M, Escriou N, Lebon P, et al. Antigen crosspresentation by human plasmacytoid dendritic cells. Immunity. 2007;27:481–492. doi: 10.1016/j.immuni.2007.07.021. [DOI] [PubMed] [Google Scholar]
  • 48.Di Pucchio T, Chatterjee B, Smed-Sorensen A, Clayton S, Palazzo A, Montes M, et al. Direct proteasome-independent cross-presentation of viral antigen by plasmacytoid dendritic cells on major histocompatibility complex class I. Nature immunology. 2008;9:551–557. doi: 10.1038/ni.1602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Tel J, Lambeck AJ, Cruz LJ, Tacken PJ, de Vries IJ, Figdor CG. Human plasmacytoid dendritic cells phagocytose, process, and present exogenous particulate antigen. J Immunol. 2010;184:4276–4283. doi: 10.4049/jimmunol.0903286. [DOI] [PubMed] [Google Scholar]
  • 50.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;186:6207–6217. doi: 10.4049/jimmunol.1002632. [DOI] [PubMed] [Google Scholar]
  • 51.van de Ven R, van den Hout MF, Lindenberg JJ, Sluijter BJ, van Leeuwen PA, Lougheed SM, et al. Characterization of four conventional dendritic cell subsets in human skin-draining lymph nodes in relation to T-cell activation. Blood. 2011;118:2502–2510. doi: 10.1182/blood-2011-03-344838. [DOI] [PubMed] [Google Scholar]
  • 52.Palucka K, Banchereau J. Cancer immunotherapy via dendritic cells. Nature reviews Cancer. 2012;12:265–277. doi: 10.1038/nrc3258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Vacchelli E, Vitale I, Eggermont A, Fridman WH, Fucikova J, Cremer I, et al. Trial watch: Dendritic cell-based interventions for cancer therapy. Oncoimmunology. 2013;2:e25771. doi: 10.4161/onci.25771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Schuler G. Dendritic cells in cancer immunotherapy. European journal of immunology. 2010;40:2123–2130. doi: 10.1002/eji.201040630. [DOI] [PubMed] [Google Scholar]
  • 55.Gomella LG, Gelpi-Hammerschmidt F, Kundavram C. Practical guide to immunotherapy in castration resistant prostate cancer: the use of sipuleucel-T immunotherapy. The Canadian journal of urology. 2014;21:48–56. [PubMed] [Google Scholar]
  • 56.Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, et al. Sipuleucel-Timmunotherapy for castration-resistant prostate cancer. The New England journal of medicine. 2010;363:411–422. doi: 10.1056/NEJMoa1001294. [DOI] [PubMed] [Google Scholar]
  • 57.Tel J, Aarntzen EH, Baba T, Schreibelt G, Schulte BM, Benitez-Ribas D, et al. Natural human plasmacytoid dendritic cells induce antigen-specific T-cell responses in melanoma patients. Cancer research. 2013;73:1063–1075. doi: 10.1158/0008-5472.CAN-12-2583. [DOI] [PubMed] [Google Scholar]
  • 58.Aarntzen EH, De Vries IJ, Lesterhuis WJ, Schuurhuis D, Jacobs JF, Bol K, et al. Targeting CD4(+) Thelper cells improves the induction of antitumor responses in dendritic cell-based vaccination. Cancer research. 2013;73:19–29. doi: 10.1158/0008-5472.CAN-12-1127. [DOI] [PubMed] [Google Scholar]
  • 59.Sharma A, Koldovsky U, Xu S, Mick R, Roses R, Fitzpatrick E, et al. HER-2 pulsed dendritic cell vaccine can eliminate HER-2 expression and impact ductal carcinoma in situ. Cancer. 2012;118:4354–4362. doi: 10.1002/cncr.26734. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Kandalaft LE, Powell DJ, Jr, Chiang CL, Tanyi J, Kim S, Bosch M, et al. Autologous lysate-pulsed dendritic cell vaccination followed by adoptive transfer of vaccine-primed ex vivo co-stimulated T cells in recurrent ovarian cancer. Oncoimmunology. 2013;2:e22664. doi: 10.4161/onci.22664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Lu W, Arraes LC, Ferreira WT, Andrieu JM. Therapeutic dendritic-cell vaccine for chronic HIV-1 infection. Nature medicine. 2004;10:1359–1365. doi: 10.1038/nm1147. [DOI] [PubMed] [Google Scholar]
  • 62.Garcia F, Climent N, Guardo AC, Gil C, Leon A, Autran B, et al. A dendritic cell-based vaccine elicits T cell responses associated with control of HIV-1 replication. Science translational medicine. 2013;5:166ra162. doi: 10.1126/scitranslmed.3004682. [DOI] [PubMed] [Google Scholar]
  • 63.Gross CC, Wiendl H. Dendritic cell vaccination in autoimmune disease. Current opinion in rheumatology. 2013;25:268–274. doi: 10.1097/BOR.0b013e32835cb9f2. [DOI] [PubMed] [Google Scholar]
  • 64.Gross CC, Jonuleit H, Wiendl H. Fulfilling the dream: tolerogenic dendritic cells to treat multiple sclerosis. European journal of immunology. 2012;42:569–572. doi: 10.1002/eji.201242402. [DOI] [PubMed] [Google Scholar]
  • 65.Paulis LE, Mandal S, Kreutz M, Figdor CG. Dendritic cell-based nanovaccines for cancer immunotherapy. Curr Opin Immunol. 2013;25:389–395. doi: 10.1016/j.coi.2013.03.001. [DOI] [PubMed] [Google Scholar]
  • 66.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. The Journal of experimental medicine. 2002;196:1627–1638. doi: 10.1084/jem.20021598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Soares H, Waechter H, Glaichenhaus N, Mougneau E, Yagita H, Mizenina O, et al. A subset of dendritic cells induces CD4+ T cells to produce IFN-gamma by an IL-12-independent but CD70-dependent mechanism in vivo. J Exp Med. 2007;204:1095–1106. doi: 10.1084/jem.20070176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Cheong C, Choi JH, Vitale L, He LZ, Trumpfheller C, Bozzacco L, et al. Improved cellular and humoral immune responses in vivo following targeting of HIV Gag to dendritic cells within human anti-human DEC205 monoclonal antibody. Blood. 2010;116:3828–3838. doi: 10.1182/blood-2010-06-288068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.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. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:2384–2389. doi: 10.1073/pnas.1019547108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Flynn BJ, Kastenmuller 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. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:7131–7136. doi: 10.1073/pnas.1103869108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Henriques HR, Rampazo EV, Goncalves AJ, Vicentin EC, Amorim JH, Panatieri RH, et al. Targeting the non-structural protein 1 from dengue virus to a dendritic cell population confers protective immunity to lethal virus challenge. PLoS neglected tropical diseases. 2013;7:e2330. doi: 10.1371/journal.pntd.0002330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hawiger D, Masilamani RF, Bettelli E, Kuchroo VK, Nussenzweig MC. Immunological unresponsiveness characterized by increased expression of CD5 on peripheral T cells induced by dendritic cells in vivo. Immun. 2004;20:695–705. doi: 10.1016/j.immuni.2004.05.002. [DOI] [PubMed] [Google Scholar]
  • 73.Stern JN, Keskin DB, Kato Z, Waldner H, Schallenberg S, Anderson A, et al. Promoting tolerance to proteolipid protein-induced experimental autoimmune encephalomyelitis through targeting dendritic cells. Proceedings of the National Academy of Sciences of the United States of America. 2010;107:17280–17285. doi: 10.1073/pnas.1010263107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Mukherjee G, Geliebter A, Babad J, Santamaria P, Serreze DV, Freeman GJ, et al. DEC-205-mediated antigen targeting to steady-state dendritic cells induces deletion of diabetogenic CD8(+) T cells independently of PD-1 and PD-L1. International immunology. 2013;25:651–660. doi: 10.1093/intimm/dxt031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Dakappagari N, Maruyama T, Renshaw M, Tacken P, Figdor C, Torensma R, et al. Internalizing antibodies to the C-type lectins, L-SIGN and DC-SIGN, inhibit viral glycoprotein binding and deliver antigen to human dendritic cells for the induction of T cell responses. J Immunol. 2006;176:426–440. doi: 10.4049/jimmunol.176.1.426. [DOI] [PubMed] [Google Scholar]
  • 76.Sancho D, Mourao-Sa 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. The Journal of clinical investigation. 2008;118:2098–2110. doi: 10.1172/JCI34584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Meyer-Wentrup F, Cambi A, Joosten B, Looman MW, de Vries IJ, Figdor CG, et al. DCIR is endocytosed into human dendritic cells and inhibits TLR8-mediated cytokine production. Journal of leukocyte biology. 2009;85:518–525. doi: 10.1189/jlb.0608352. [DOI] [PubMed] [Google Scholar]
  • 78.Ni L, Gayet I, Zurawski S, Duluc D, Flamar AL, Li XH, et al. Concomitant activation and antigen uptake via human dectin-1 results in potent antigen-specific CD8+ T cell responses. J Immunol. 2010;185:3504–3513. doi: 10.4049/jimmunol.1000999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Flacher V, Sparber F, Tripp CH, Romani N, Stoitzner P. Targeting of epidermal Langerhans cells with antigenic proteins: attempts to harness their properties for immunotherapy. Cancer immunology, immunotherapy : CII. 2009;58:1137–1147. doi: 10.1007/s00262-008-0563-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Moffat JM, Segura E, Khoury G, Caminschi I, Cameron PU, Lewin SR, et al. Targeting antigen to bone marrow stromal cell-2 expressed by conventional and plasmacytoid dendritic cells elicits efficient antigen presentation. European journal of immunology. 2013;43:595–605. doi: 10.1002/eji.201242799. [DOI] [PubMed] [Google Scholar]
  • 81.Dhodapkar MV, Sznol M, Zhao B, Wang D, Carvajal RD, Keohan ML, et al. Induction of Antigen-Specific Immunity with a Vaccine Targeting NY-ESO-1 to the Dendritic Cell Receptor DEC-205. Sci Transl Med. 2014;6:232ra251. doi: 10.1126/scitranslmed.3008068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Dhodapkar KM, Gettinger SN, Das R, Zebroski H, Dhodapkar MV. SOX2-specific adaptive immunity and response to immunotherapy in non-small cell lung cancer. Oncoimmunology. 2013;2:e25205. doi: 10.4161/onci.25205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Gregory AE, Titball R, Williamson D. Vaccine delivery using nanoparticles. Frontiers in cellular and infection microbiology. 2013;3:13. doi: 10.3389/fcimb.2013.00013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Demento SL, Cui W, Criscione JM, Stern E, Tulipan J, Kaech SM, et al. Role of sustained antigen release from nanoparticle vaccines in shaping the T cell memory phenotype. Biomaterials. 2012;33:4957–4964. doi: 10.1016/j.biomaterials.2012.03.041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Smith DM, Simon JK, Baker JR., Jr Applications of nanotechnology for immunology. Nature reviews Immunology. 2013;13:592–605. doi: 10.1038/nri3488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Silva JM, Videira M, Gaspar R, Preat V, Florindo HF. Immune system targeting by biodegradable nanoparticles for cancer vaccines. Journal of controlled release : official journal of the Controlled Release Society. 2013;168:179–199. doi: 10.1016/j.jconrel.2013.03.010. [DOI] [PubMed] [Google Scholar]
  • 87.Speiser DE, Schwarz K, Baumgaertner P, Manolova V, Devevre E, Sterry W, et al. Memory and effector CD8 T-cell responses after nanoparticle vaccination of melanoma patients. J Immunother. 2010;33:848–858. doi: 10.1097/CJI.0b013e3181f1d614. [DOI] [PubMed] [Google Scholar]
  • 88.Cruz LJ, Tacken PJ, Fokkink R, Joosten B, Stuart MC, Albericio F, et al. Targeted PLGA nano- but not microparticles specifically deliver antigen to human dendritic cells via DC-SIGN in vitro. Journal of controlled release : official journal of the Controlled Release Society. 2010;144:118–126. doi: 10.1016/j.jconrel.2010.02.013. [DOI] [PubMed] [Google Scholar]
  • 89.Hamdy S, Haddadi A, Shayeganpour A, Samuel J, Lavasanifar A. Activation of antigen-specific T cell-responses by mannan-decorated PLGA nanoparticles. Pharmaceutical research. 2011;28:2288–2301. doi: 10.1007/s11095-011-0459-9. [DOI] [PubMed] [Google Scholar]
  • 90.Tacken PJ, Zeelenberg IS, Cruz LJ, van Hout-Kuijer MA, van de Glind G, Fokkink RG, et al. Targeted delivery of TLR ligands to human and mouse dendritic cells strongly enhances adjuvanticity. Blood. 2011;118:6836–6844. doi: 10.1182/blood-2011-07-367615. [DOI] [PubMed] [Google Scholar]
  • 91.Thomann JS, Heurtault B, Weidner S, Braye M, Beyrath J, Fournel S, et al. Antitumor activity of liposomal ErbB2/HER2 epitope peptide-based vaccine constructs incorporating TLR agonists and mannose receptor targeting. Biomaterials. 2011;32:4574–4583. doi: 10.1016/j.biomaterials.2011.03.015. [DOI] [PubMed] [Google Scholar]
  • 92.Cruz LJ, Tacken PJ, Pots JM, Torensma R, Buschow SI, Figdor CG. Comparison of antibodies and carbohydrates to target vaccines to human dendritic cells via DC-SIGN. Biomaterials. 2012;33:4229–4239. doi: 10.1016/j.biomaterials.2012.02.036. [DOI] [PubMed] [Google Scholar]
  • 93.Schreibelt G, Klinkenberg LJ, Cruz LJ, Tacken PJ, Tel J, Kreutz M, et al. The C-type lectin receptor CLEC9A mediates antigen uptake and (cross-)presentation by human blood BDCA3+ myeloid dendritic cells. Blood. 2012;119:2284–2292. doi: 10.1182/blood-2011-08-373944. [DOI] [PubMed] [Google Scholar]
  • 94.Tel J, Sittig SP, Blom RA, Cruz LJ, Schreibelt G, Figdor CG, et al. Targeting uptake receptors on human plasmacytoid dendritic cells triggers antigen cross-presentation and robust type I IFN secretion. J Immunol. 2013;191:5005–5012. doi: 10.4049/jimmunol.1300787. [DOI] [PubMed] [Google Scholar]
  • 95.Piccioli D, Sammicheli C, Tavarini S, Nuti S, Frigimelica E, Manetti AG, et al. Human plasmacytoid dendritic cells are unresponsive to bacterial stimulation and require a novel type of cooperation with myeloid dendritic cells for maturation. Blood. 2009;113:4232–4239. doi: 10.1182/blood-2008-10-186890. [DOI] [PubMed] [Google Scholar]
  • 96.Kastenmuller 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;121:1782–1796. doi: 10.1172/JCI45416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Oh JZ, Kurche JS, Burchill MA, Kedl RM. TLR7 enables cross-presentation by multiple dendritic cell subsets through a type I IFN-dependent pathway. Blood. 2011;118:3028–3038. doi: 10.1182/blood-2011-04-348839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Querec T, Bennouna S, Alkan S, Laouar Y, Gorden K, Flavell R, et al. Yellow fever vaccine YF-17D activates multiple dendritic cell subsets via TLR2, 7, 8, and 9 to stimulate polyvalent immunity. J Exp Med. 2006;203:413–424. doi: 10.1084/jem.20051720. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Sehgal K, Ragheb R, F T, Dhodapkar M, Dhodapkar K. Nanoparticle-mediated combinatorial targeting of multiple human dendritic cell (DC) subsets leads to enhanced T cell activation via IL15-dependent DC cross-talk. J Immunol. 2014 doi: 10.4049/jimmunol.1400489. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Schjetne KW, Fredriksen AB, Bogen B. Delivery of antigen to CD40 induces protective immune responses against tumors. J Immunol. 2007;178:4169–4176. doi: 10.4049/jimmunol.178.7.4169. [DOI] [PubMed] [Google Scholar]
  • 101.Dhodapkar KM, Banerjee D, Connolly J, Kukreja A, Matayeva E, Veri MC, et al. Selective blockade of the inhibitory Fc{gamma} receptor (Fc{gamma}RIIB) in human dendritic cells and monocytes induces a type I interferon response program. J Exp Med. 2007;204:1359–1369. doi: 10.1084/jem.20062545. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Dhodapkar KM, Kaufman JL, Ehlers M, Banerjee DK, Bonvini E, Koenig S, et al. Selective blockade of inhibitory Fc gamma receptor enables human dendritic cell maturation with IL-12p70 production and immunity to antibody-coated tumor cells. Proc Natl Acad Sci U S A. 2005;102:2910–2915. doi: 10.1073/pnas.0500014102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Flemming A. Vaccines: nano-adjuvant: double TLR stimulation is the key. Nature reviews Drug discovery. 2011;10:258. doi: 10.1038/nrd3426. [DOI] [PubMed] [Google Scholar]
  • 104.Kasturi SP, Skountzou I, Albrecht RA, Koutsonanos D, Hua T, Nakaya HI, et al. Programming the magnitude and persistence of antibody responses with innate immunity. Nature. 2011;470:543–547. doi: 10.1038/nature09737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.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. The Journal of experimental medicine. 2004;199:815–824. doi: 10.1084/jem.20032220. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.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;177:2276–2284. doi: 10.4049/jimmunol.177.4.2276. [DOI] [PubMed] [Google Scholar]
  • 107.Kreutz M, Giquel B, Hu Q, Abuknesha R, Uematsu S, Akira S, et al. Antibody-antigen-adjuvant conjugates enable co-delivery of antigen and adjuvant to dendritic cells in cis but only have partial targeting specificity. PloS one. 2012;7:e40208. doi: 10.1371/journal.pone.0040208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.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. European journal of immunology. 2010;40:1255–1265. doi: 10.1002/eji.201040419. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Hesse C, Ginter W, Forg T, Mayer CT, Baru AM, Arnold-Schrauf C, et al. In vivo targeting of human DC-SIGN drastically enhances CD8(+) T-cell-mediated protective immunity. European journal of immunology. 2013;43:2543–2553. doi: 10.1002/eji.201343429. [DOI] [PubMed] [Google Scholar]
  • 110.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;178:6259–6267. doi: 10.4049/jimmunol.178.10.6259. [DOI] [PubMed] [Google Scholar]
  • 111.Loschko J, Heink S, Hackl D, Dudziak D, Reindl W, Korn T, et al. Antigen targeting to plasmacytoid dendritic cells via Siglec-H inhibits Th cell-dependent autoimmunity. J Immunol. 2011;187:6346–6356. doi: 10.4049/jimmunol.1102307. [DOI] [PubMed] [Google Scholar]
  • 112.Loschko J, Schlitzer A, Dudziak D, Drexler I, Sandholzer N, Bourquin C, et al. Antigen delivery to plasmacytoid dendritic cells via BST2 induces protective T cell-mediated immunity. J Immunol. 2011;186:6718–6725. doi: 10.4049/jimmunol.1004029. [DOI] [PubMed] [Google Scholar]
  • 113.Zhang J, Raper A, Sugita N, Hingorani R, Salio M, Palmowski MJ, et al. Characterization of Siglec-H as a novel endocytic receptor expressed on murine plasmacytoid dendritic cell precursors. Blood. 2006;107:3600–3608. doi: 10.1182/blood-2005-09-3842. [DOI] [PubMed] [Google Scholar]
  • 114.Trumpfheller C, Caskey M, Nchinda G, Longhi MP, Mizenina O, Huang Y, et al. The microbial mimic poly IC induces durable and protective CD4+ T cell immunity together with a dendritic cell targeted vaccine. Proceedings of the National Academy of Sciences of the United States of America. 2008;105:2574–2579. doi: 10.1073/pnas.0711976105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Longhi MP, Trumpfheller C, Idoyaga J, Caskey M, Matos I, Kluger C, et al. Dendritic cells require a systemic type I interferon response to mature and induce CD4+ Th1 immunity with poly IC as adjuvant. The Journal of experimental medicine. 2009;206:1589–1602. doi: 10.1084/jem.20090247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Barbuto S, Idoyaga J, Vila-Perello M, Longhi MP, Breton G, Steinman RM, et al. Induction of innate and adaptive immunity by delivery of poly dA:dT to dendritic cells. Nature chemical biology. 2013;9:250–256. doi: 10.1038/nchembio.1186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Matos I, Mizenina O, Lubkin A, Steinman RM, Idoyaga J. Targeting Antigens to Dendritic Cells In Vivo Induces Protective Immunity. PloS one. 2013;8:e67453. doi: 10.1371/journal.pone.0067453. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Dong H, Stanek O, Salvador FR, Langer U, Morillon E, Ung C, et al. Induction of protective immunity against Mycobacterium tuberculosis by delivery of ESX antigens into airway dendritic cells. Mucosal immunology. 2013;6:522–534. doi: 10.1038/mi.2012.92. [DOI] [PubMed] [Google Scholar]
  • 119.Meixlsperger S, Leung CS, Ramer PC, Pack M, Vanoaica LD, Breton G, et al. CD141+ dendritic cells produce prominent amounts of IFN-alpha after dsRNA recognition and can be targeted via DEC-205 in humanized mice. Blood. 2013;121:5034–5044. doi: 10.1182/blood-2012-12-473413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Wang B, Zaidi N, He LZ, Zhang L, Kuroiwa JM, Keler T, et al. Targeting of the non-mutated tumor antigen HER2/neu to mature dendritic cells induces an integrated immune response that protects against breast cancer in mice. Breast cancer research : BCR. 2012;14:R39. doi: 10.1186/bcr3135. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Meyer-Wentrup F, Benitez-Ribas D, Tacken PJ, Punt CJ, Figdor CG, de Vries IJ, et al. Targeting DCIR on human plasmacytoid dendritic cells results in antigen presentation and inhibits IFN-alpha production. Blood. 2008;111:4245–4253. doi: 10.1182/blood-2007-03-081398. [DOI] [PubMed] [Google Scholar]
  • 122.Bozzacco L, Trumpfheller C, Siegal FP, Mehandru S, Markowitz M, Carrington M, et al. DEC-205 receptor on dendritic cells mediates presentation of HIV gag protein to CD8+ T cells in a spectrum of human MHC I haplotypes. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:1289–1294. doi: 10.1073/pnas.0610383104. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Flamar AL, Xue Y, Zurawski SM, Montes M, King B, Sloan L, et al. Targeting concatenated HIV antigens to human CD40 expands a broad repertoire of multifunctional CD4+ and CD8+ T cells. AIDS. 2013;27:2041–2051. doi: 10.1097/QAD.0b013e3283624305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Tacken PJ, Joosten B, Reddy A, Wu D, Eek A, Laverman P, et al. No advantage of cell-penetrating peptides over receptor-specific antibodies in targeting antigen to human dendritic cells for cross-presentation. J Immunol. 2008;180:7687–7696. doi: 10.4049/jimmunol.180.11.7687. [DOI] [PubMed] [Google Scholar]
  • 125.Ramakrishna V, Treml JF, Vitale L, Connolly JE, O'Neill T, Smith PA, et al. Mannose receptor targeting of tumor antigen pmel17 to human dendritic cells directs anti-melanoma T cell responses via multiple HLA molecules. J Immunol. 2004;172:2845–2852. doi: 10.4049/jimmunol.172.5.2845. [DOI] [PubMed] [Google Scholar]
  • 126.He LZ, Ramakrishna V, Connolly JE, Wang XT, Smith PA, Jones CL, et al. A novel human cancer vaccine elicits cellular responses to the tumor-associated antigen, human chorionic gonadotropin beta. Clinical cancer research : an official journal of the American Association for Cancer Research. 2004;10:1920–1927. doi: 10.1158/1078-0432.ccr-03-0264. [DOI] [PubMed] [Google Scholar]
  • 127.Ramakrishna V, Vasilakos JP, Tario JD, Jr, Berger MA, Wallace PK, Keler T. Toll-like receptor activation enhances cell-mediated immunity induced by an antibody vaccine targeting human dendritic cells. Journal of translational medicine. 2007;5:5. doi: 10.1186/1479-5876-5-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Morse MA, Chapman R, Powderly J, Blackwell K, Keler T, Green J, et al. Phase I study utilizing a novel antigen-presenting cell-targeted vaccine with Toll-like receptor stimulation to induce immunity to self-antigens in cancer patients. Clinical cancer research : an official journal of the American Association for Cancer Research. 2011;17:4844–4853. doi: 10.1158/1078-0432.CCR-11-0891. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Garcia-Vallejo JJ, Ambrosini M, Overbeek A, van Riel WE, Bloem K, Unger WW, et al. Multivalent glycopeptide dendrimers for the targeted delivery of antigens to dendritic cells. Molecular immunology. 2013;53:387–397. doi: 10.1016/j.molimm.2012.09.012. [DOI] [PubMed] [Google Scholar]
  • 130.Thomann-Harwood LJ, Kaeuper P, Rossi N, Milona P, Herrmann B, McCullough KC. Nanogel vaccines targeting dendritic cells: contributions of the surface decoration and vaccine cargo on cell targeting and activation. Journal of controlled release : official journal of the Controlled Release Society. 2013;166:95–105. doi: 10.1016/j.jconrel.2012.11.015. [DOI] [PubMed] [Google Scholar]
  • 131.van Broekhoven CL, Parish CR, Demangel C, Britton WJ, Altin JG. Targeting dendritic cells with antigen-containing liposomes: a highly effective procedure for induction of antitumor immunity and for tumor immunotherapy. Cancer research. 2004;64:4357–4365. doi: 10.1158/0008-5472.CAN-04-0138. [DOI] [PubMed] [Google Scholar]
  • 132.Joshi MD, Unger WW, van Beelen AJ, Bruijns SC, Litjens M, van Bloois L, et al. DC-SIGN mediated antigen-targeting using glycan-modified liposomes: formulation considerations. International journal of pharmaceutics. 2011;416:426–432. doi: 10.1016/j.ijpharm.2011.02.055. [DOI] [PubMed] [Google Scholar]
  • 133.Singh SK, Streng-Ouwehand I, Litjens M, Kalay H, Burgdorf S, Saeland E, et al. Design of neo-glycoconjugates that target the mannose receptor and enhance TLR-independent cross-presentation and Th1 polarization. European journal of immunology. 2011;41:916–925. doi: 10.1002/eji.201040762. [DOI] [PubMed] [Google Scholar]
  • 134.Faham A, Altin JG. Antigen-containing liposomes engrafted with flagellin-related peptides are effective vaccines that can induce potent antitumor immunity and immunotherapeutic effect. J Immunol. 2010;185:1744–1754. doi: 10.4049/jimmunol.1000027. [DOI] [PubMed] [Google Scholar]
  • 135.Fukasawa M, Shimizu Y, Shikata K, Nakata M, Sakakibara R, Yamamoto N, et al. Liposome oligomannose-coated with neoglycolipid, a new candidate for a safe adjuvant for induction of CD8+ cytotoxic T lymphocytes. FEBS letters. 1998;441:353–356. doi: 10.1016/s0014-5793(98)01577-4. [DOI] [PubMed] [Google Scholar]
  • 136.Arigita C, Bevaart L, Everse LA, Koning GA, Hennink WE, Crommelin DJ, et al. Liposomal meningococcal B vaccination: role of dendritic cell targeting in the development of a protective immune response. Infection and immunity. 2003;71:5210–5218. doi: 10.1128/IAI.71.9.5210-5218.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Cui Z, Han SJ, Huang L. Coating of mannan on LPD particles containing HPV E7 peptide significantly enhances immunity against HPV-positive tumor. Pharmaceutical research. 2004;21:1018–1025. doi: 10.1023/b:pham.0000029292.66792.4f. [DOI] [PubMed] [Google Scholar]
  • 138.Yang L, Yang H, Rideout K, Cho T, Joo KI, Ziegler L, et al. Engineered lentivector targeting of dendritic cells for in vivo immunization. Nature biotechnology. 2008;26:326–334. doi: 10.1038/nbt1390. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Hangalapura BN, Oosterhoff D, de Groot J, Boon L, Tuting T, van den Eertwegh AJ, et al. Potent antitumor immunity generated by a CD40-targeted adenoviral vaccine. Cancer research. 2011;71:5827–5837. doi: 10.1158/0008-5472.CAN-11-0804. [DOI] [PubMed] [Google Scholar]
  • 140.Cruz LJ, Rueda F, Simon L, Cordobilla B, Albericio F, D JC. Liposomes containing NY-ESO-1/tetanus toxoid and adjuvant peptides targeted to human dendritic cells via the Fc receptor for cancer vaccines. Nanomedicine (Lond) 2013 doi: 10.2217/NNM.13.66. [DOI] [PubMed] [Google Scholar]
  • 141.Cruz LJ, Rueda F, Cordobilla B, Simon L, Hosta L, Albericio F, et al. Targeting nanosystems to human DCs via Fc receptor as an effective strategy to deliver antigen for immunotherapy. Molecular pharmaceutics. 2011;8:104–116. doi: 10.1021/mp100178k. [DOI] [PubMed] [Google Scholar]
  • 142.Brandao JG, Scheper RJ, Lougheed SM, Curiel DT, Tillman BW, Gerritsen WR, et al. CD40-targeted adenoviral gene transfer to dendritic cells through the use of a novel bispecific single-chain Fv antibody enhances cytotoxic T cell activation. Vaccine. 2003;21:2268–2272. doi: 10.1016/s0264-410x(03)00050-1. [DOI] [PubMed] [Google Scholar]

RESOURCES