1,131
Views
36
CrossRef citations to date
0
Altmetric
Special Focus Review

Multifunctional dendritic cell-targeting polymeric microparticles

Engineering new vaccines for type 1 diabetes

, &
Pages 37-44 | Received 25 Jun 2010, Accepted 05 Jul 2010, Published online: 01 Jan 2011

Type 1 diabetes (T1D) is an autoimmune disease characterized by T-cell mediated destruction of insulin-producing β-cells.Citation1 Approximately 13,000 new cases of T1D are diagnosed each year in US children, with a prevalence of about 2 cases per 1,000 individuals, and associated treatment and care costs are large.Citation2 Following clinical diagnosis, many T1D patients maintain functional pancreatic β-cell mass for some time.Citation3 This therapeutic window has motivated attempts to halt autoimmune inflammation of pancreatic islets before too many islets are destroyed to maintain glucose regulation. Schemes involving broad immunosuppressionCitation4 or antigen non-specific immunomodulationCitation5 have been explored for T1D treatment, however, only antigen-specific approaches hold the unique promise of long-lasting disease remission in the absence of adverse side effects.Citation6 Therefore, efforts have focused on manipulating the body’s most efficient antigen-presenting cell, dendritic cells (DCs), in T1D and other autoimmune diseases.Citation7 Early and ongoing efforts involving DCs therapeutically focus on the exogenous generation of DCs for administration as a cellular vaccine.Citation7,Citation8 In fact, an exogenously manipulated DC-based vaccine is currently being investigated in clinical trials for application in T1D.Citation9 However, it is generally accepted that cell-based vaccines for T1D are primarily intended to provide proof-of-concept, as several factors limit this approach.Citation10 In particular, dissemination and lymph node homing of exogenously delivered DCs is inefficientCitation11 and treatment involves isolation and storage of DCs under stringent manufacturing standards, amounting to high costs which prohibit widespread application.Citation12,Citation13

An attractive alternative strategy involves the in vivo targeting of DCs with injectable polymeric, biodegradable microparticles delivering a payload of vaccine components and immunomodulatory factors. Microparticle systems may be easily administered in a single injection to patients with extended delivery of both prime and boost doses using timed-release materials.Citation14,Citation15 Furthermore, polymeric microparticle strategies greatly simplifies issues related to manufacturing, storage and shipping, as microparticle encapsulation affords stability for off-the-shelf availability.Citation16,Citation17 Microparticles can be engineered to be multifunctional and modular, where features of particular interest are: (1) targeting to DCs, (2) providing a depot for antigen, (3) providing controlled delivery and subcellular targeting of adjuvants, immunosuppressants, chemokines or other conditioning factors. Conceptually, microparticle systems can thus be designed to attract DCs and precursors into an injection site, provide signals to promote differentiation into DCs, promote uptake of antigen and provided immunosuppressive/tolerogenic conditioning of DCs to induce specific tolerance. In practice, the amalgamation of these design principles are just beginning to be assembled for application toward T1D. The goal of this review is in part to showcase recent microparticle-based efforts toward immunosuppression and T1D mitigation in particular, but also to draw on the broader literature of DC-targeting of microparticles, largely applied to infection or cancer, as instructive examples to highlight the repertoire of tools available for the scientist/clinician investigating T1D therapies.

Tolerogenic Dendritic Cells

The rationale for therapeutic interest in DCs is founded in their physiological roles. Dendritic cells are phagocytes and the most efficient antigen presenting cell.Citation18 Moreover, DCs are central regulators of the immune system, processing and presenting antigen to direct induction and expansion of specific T-cell subsets (e.g., Th1, Th2, Th17, regulatory T-cells), promoting either antigen-specific tolerance or immunity.Citation19,Citation20 Key characteristics of DCs include: (1) their ability to uptake and transport antigen from peripheral tissues to T-cell zones in secondary lymphoid organs, (2) activation marked by upregulation of major histocompatibility complex II (MHC II), co-stimulatory surface molecules (e.g., CD80, CD83, CD86), (3) Ag processing and presentation on both MHC II and MHC I complexes and (4) T-cell interaction and stimulation.Citation18,Citation20 Furthermore, DCs play a critical role in both the establishment of central tolerance and the maintenance of peripheral tolerance. Mechanisms by which DCs facilitate immune protection in homeostasis include induction of T-cell deletion, anergy or regulatory T-cells (Tregs) for peripheral tolerance.Citation7,Citation21-Citation24 General (but not requisite) characteristics of tolerance-promoting/tolerogenic DCs that have been reported include low expression of stimulatory and co-stimulatory molecules, low production of inflammatory cytokines (e.g., IL-12), and increased production of immunosuppressive cytokines (e.g., IL-10); with various reports of specific markers such as high levels of inhibitory molecules (e.g., programmed death ligand, immunoglobulin-like transcript-3) and indoleamine 2,3-dioxygenase (IDO).Citation21,Citation25-Citation29 There is enormous potential use of tolerogenic DCs as therapeutic agents to mitigate autoimmune diseases and transplant rejection.Citation7,Citation25 Of particular interest for durable antigen-specific tolerance are tolerogenic DCs which can induce CD4+CD25+Foxp3+ Tregs. These regulatory T-cells have the ability to suppress the actions of effector T-cells and also impair the activation of DCs.Citation23,Citation24 A number of biological and pharmacological agents have been shown to induce DCs capable of generating CD4+CD25+Foxp3+ Tregs and halt or reverse autoimmunity in animal models.Citation7,Citation22 Conceivably, any of these factors or combination of factors are readily incorporated into microparticle formulations, some of which have already been reported, as discussed below.

Polymeric Microparticles for Targeted Antigen Delivery

Numerous polymeric biomaterials have been investigated in particle-based drug delivery vehicles to deliver immunomodulatory, functionally-active proteins, peptides, nucleic acids, antigens and adjuvants.Citation30-Citation36 Of these, microparticles fabricated using biodegradable poly(lactide-glycolide) (PLGA) have been the most investigated vehicle for delivering immunotherapeutics and the influence of PLGA microparticles on DCs has been characterized.Citation14,Citation37-Citation43 Many biomaterial options exist for particle formulation, with various advantages.Citation44 However, PLGA, is appealing in terms of translation, as it has been approved by the US Food and Drug Administration for numerous devices including resorbable surgical sutures and drug delivery products, and is degraded in the body into natural products of lactic and glycolic acid. By altering the composition, polymeric microparticles can be designed to provide a tailored initial burst of the encapsulated immuno-modulatory molecule followed by sustained release, permitting the design of a one-time drug administration with prime and boost doses.Citation14

The most common technique to fabricate polymeric microparticles encapsulating molecules is the double-emulsion solvent-evaporation method.Citation14,Citation30,Citation45 Using this technique, a hydrophilic molecule (e.g., protein, peptide, nucleic acid) to be encapsulated is placed in aqueous solution while the polymer and a hydrophobic molecule (e.g., immunosuppressive drugs) to be incorporated is dissolved in an organic solution. The two phases are emulsified by sonication or homogenization and this primary emulsion is then added to a second larger, bulk aqueous phase and again mixed by homogenization to form a water-in-oil-in-water double emulsion. The organic solvent is then evaporated and the polymer-containing droplets harden to form microparticles, which are then isolated by filtration or centrifugation. Finally, the particles are lyophilized to remove water from the interior aqueous phase to result in a dry suspension of the encapsulated material within the polymer matrix. Additionally, microparticles can be surface-modified. For example, surface immobilization of a polycationic polymer allows surface-loading of nucleic acids or cell surface receptor-targeting molecules such as antibodies can be immobilized to promote DC interactions.

Microparticles of appropriate size (generally 1–5 μm) are too large to be taken up by cells through pinocytosis/endocytosis (by which nanoparticles are taken up), but are phagocytosed efficiently by specialized phagocytes.Citation30,Citation46 This size exclusion of particulate uptake by cells provides a simple, but powerful passive targeting mechanism to deliver a payload of antigen and immunomodulatory factors to phagocytic cells such as DCs.Citation47-Citation49 Protein-based antigen choices for microparticle formulations include whole protein antigens (e.g., insulinCitation50-Citation52) or specific antigenic peptides.Citation53-Citation55 An advantage to whole protein antigens is it is not necessary to determine the immunodominant peptides, which may vary from patient to patient, while use of defined antigenic peptides carries less risk of an adverse immune response. Protein degradation within DC endosomes involves acidic hydrolysis, reactive oxygen species and enzymatic degradation, the mechanisms and consequences of the timing of which are being uncovered.Citation56 Critically, encapsulation of antigen in microparticles (e.g., PLGA) provides the advantages of enhancing and prolonging antigen presentation on both MHC-II and MHC-I molecules, requiring orders of magnitude less antigen compared to a soluble dose and providing antigen presentation for days-to-weeks, compared to hours-to-days for soluble antigen.Citation47,Citation48,Citation57,Citation58 Loading of microparticle encapsulated antigen onto MHC-II complexes occurs in phagosomes.Citation59 On the other hand, loading onto MHC-I molecules through cross-presentation of exogenously-delivered antigen occurs via endosomal escape into the cytosol.Citation57,Citation58 Microparticles of PLGA have the demonstrated ability to provide effective cross-presentation, shown to require active proteasome, indicating access to the cytosol,Citation60 possibly because polymer degradation acidifies the endosome, increasing the osmotic pressure to the point of leakage,Citation58,Citation61 or through an alternate MHC-I processing pathway.Citation62,Citation63 The molecular weight of the PLGA used to form microparticles can direct the kinetics of delivery to the endosome and subsequent release to the cytoplasm, with the lower molecular weight polymer (6 kDa) delivering an encapsulate after 24 h, while endosomal release generated from the higher molecular weight polymer (60 kDa) was delayed to day 5.Citation64

Microparticle-associated endosomal escape has also been capitalized upon to effect non-viral delivery of nucleic acids to the cytosol for subsequent nuclear localization.Citation30 Microparticle incorporation of nucleic acids such as DNA plasmids, antisense oligodeoxynucleotides and small interfering RNA has been well-established, and provides protection from degradation by nucleases, as well as effective nucleotide delivery for gene expression or knockdown.Citation65-Citation67 Gene delivery for expression of antigenic proteins is also an established means of establishing an antigen depot, generally referred to as DNA vaccines.Citation68 As applied to autoimmunity, DNA-based vaccines with genes encoding for autoantigen have been referred to as an “inverse vaccination”, and are being investigated clinically for multiple sclerosis and T1D.Citation69 In cases where endogenous antigen presentation by DCs is considered favorable (i.e., MHC-I loading for presentation to CD8+ T-cells), DNA-based vaccines are expected to benefit from incorporation into microparticles for targeting and delivery. A number of polymeric microparticle systems have been developed to deliver DNA to cells, primarily incorporating polycationic molecules to facilitate DNA loading and nuclear translocation.Citation70,Citation71

Taken together, polymeric microparticles have demonstrated versatility to provide an antigen depot for vaccines and biomaterial properties are easily tailored to suit the design constraints set by the form of the antigen. An antigen depot can be passively targeted to DCs through microparticle size, and subcellular sites can be further targeted for payload delivery to the phagosome and cytosol as well as translocation into the nucleus. However, issues remain related to reduced protein stability when encapsulated in acidic microparticles such as PLGA. Denaturation of protein antigen is expected to reduce conformationally-dependent antibody responses, whereas, T-cell mediated responses, relying on recognition of processed antigen, are not as sensitive to a harsh carrier environment.Citation72,Citation73 In order to improve encapsulated protein stability, the introduction of protein stabilizers has been explored.Citation74

Tuning Microparticle Properties

Controlling the loading and degradation rate of polymeric systems is an important parameter that can affect immune responses. For instance, the level of antigen loading into polymeric microparticles is important as antigen dosing at both the high (e.g., in autoimmune encephalomyelitisCitation75) and low extremes have been shown to either maintain ignorance or to induce tolerance.Citation76-Citation79 Tuning the rate of polymer degradation in antigen-loaded microparticles is an important parameter, optimization of which currently requires empirical determination for a given application. For example, in the case of antigenic peptide loading into PLGA microparticles for rabies vaccination, the faster-releasing formulation was found to provide protection superior to complete Freund’s adjuvant.Citation80 In contrast, slower-degrading ovalbumin-loaded PLGA microparticles induced longer-lasting anti-ovalbumin antibody titers.Citation81 On the other hand, low levels of prolonged antigen delivery are capable of inducing an “exhausted” T-cell phenotype, and in some (location-dependent) cases, tolerance.Citation82,Citation83 It should be noted, however, that composition of the PLGA copolymer in itself could potentially affect DC response, as PLGA microparticles with a 50:50 ratio of lactide to glycolide been reported to maintain DC immaturity,Citation43 while a 75:25 ratio has been reported to induce DC activation.Citation41 It has been postulated that this effect may be explained by the fact that the higher lactide-containing polymers possess an increased surface hydrophobicity, which may conceivably present hydrophobic moieties interacting with receptors involved in danger signal recognition.Citation84 However, this issue remains to be resolved, as conflicting evidence regarding this trend has been reported.Citation85

Because PLGA degrades relatively slowly, even at lysosomal pH, it has been posited that newer materials for microparticle vaccine systems which have been designed to primarily release their payload only when in phagosolysosomes, will prove advantageous.Citation44 For example, DC-targeting microparticles fabricated with materials incorporating pH-responsive elements such as cleavable acetal linkagesCitation86,Citation87 and orthoestersCitation88 have been investigated. These materials capitalize on the fact that extracellular pH is an average of 7.4, while phagosolysomes reach ~pH 5,Citation89,Citation90 and have been used to provide high levels of DC-mediated CD8+ T-cell stimulation in vitro and vivo.Citation86-Citation88 Additionally, reduction-oxidation states can be exploited for timed payload delivery given that the endosomal compartment is reductive while the lysosomal compartment is increasingly oxidative, compared to an extracellular environment that is only mildly oxidative.Citation44,Citation89 This concept has been utilized to deliver contents to the early endosome using reduction-sensitive materials,Citation91 while oxidation-sensitive particles have been used to generate humoral and cellular immunity to particle-loaded ovalbumin.Citation92 Furthermore, biomaterial schemes have been developed to facilitate endosomal escape through use of these same endosomal triggers of pH and reduction state to generate membranolytic products with demonstrated ability for antigen delivery and cross-presentation.Citation91,Citation93

Finally, reports indicate that particle size can modulate tissue targeting, cellular uptake specificity and adjuvancy. For example, despite a presumed loss in phagocyte-targeting specificity, subcutaneous injection of very small sized nanoparticles (20–45 nm) have been shown to be driven to lymph nodes through interstitial fluid flow and taken up by lymph node-resident DCs.Citation92,Citation94-Citation96 Additionally, it has been reported that 40–100 nm inert particles injected subcutaneously were taken up more efficiently by DCs, while 1 μm particles were taken up more by macrophages, and that 40 nm antigen-loaded particles were more immunogenic.Citation96 On the other hand, phagocyte targeting has been shown to also be dependent on the route of administration, as intraperitoneal injection of PLGA microparticles favored the uptake and migration to lymph nodes by macrophages while subcutaneous injection favored DCs.Citation39 PLGA microparticles size (a range of 1–14 μm) has been demonstrated to direct adjuvancy of antigen-loaded PLGA microparticles delivered orally, with an optimal particle size of 4 μm.Citation97 Interplay between the parameters of particle size and receptor targeting can also be a factor, receptor-targeted nanoparticles (0.2 μm) but not microparticles (2 μm) have demonstrated increased specificity of antigen delivery to DCs, in vitro.Citation98

Dendritic Cell Receptor Targeting

The targeting of antigens to specific DC receptors has been investigated primarily utilizing soluble antigens. Normally, endocytosed soluble antigens are cross-presented by DCs less efficiently than particulate antigens which are taken up by phagocytosis,Citation99 however, DC receptor-targeting has been shown to facilitate cross-presentation.Citation100,Citation101 This is an important consideration given that cross-presentation by DCs can enable peripheral CD8+ T-cell cross-tolerance.Citation102 Ligation of antigens to molecules targeting DC upatke receptors, for example, the dendritic and epithelial cell 205 receptor (DEC-205,Citation103) can increase uptake efficiency by approximately 100-fold.Citation101,Citation104,Citation105 Approaches for targeting DC receptors have generally involved either natural receptor ligands or the use of antibodies raised against specific receptors. Ligands for DC receptors that have been investigated for soluble antigen delivery include heat shock proteins,Citation106 bacterial toxins,Citation107 sugar residuesCitation108 and CD40-ligand.Citation109 For example, targeting C-type lectin receptors has been accomplished through incorporation of specific sugar residues such as mannose/mannan.Citation110 Additionally, antigen targeting to DC receptors through the use of receptor-specific antibodies has been widely investigated.Citation108 Dendritic cell surface receptors which have been antibody-targeted for antigen delivery include the integrin CD11c/CD18,Citation111 Fc receptors,Citation112,Citation113 and the C-type lectin receptors: mannose receptor,Citation114,Citation115 DEC-205Citation100,Citation116 and DC-SIGN.Citation117,Citation118

Notably, immunogenic applications benefit from targeting antigen to a receptor which provides concomitant DC activation (e.g., CD40-ligandCitation119), however a tolerogenic application will require targeting DC receptors which do not activate DCs. Fortunately, certain receptors have the potential for DC targeting reportedly without activation. For example, targeting soluble antigen to DEC-205 did not activate DCs, and in fact, lead to DC-induced peripheral T-cell unresponsiveness in the absence of an additional stimulatory signal.Citation101,Citation120,Citation121 In fact, targeting antigen to DEC-205 has demonstrated the ability to induce tolerance in animal models of T1D.Citation122,Citation123 Additionally, inhibitory receptors such as the Fc receptor, CD32b,Citation124,Citation125 or DC inhibitory receptor-2 (DCIR2/Clec4a2)Citation126,Citation127 may present a potential way to simultaneously target and provide inhibitory signals to DCs.

A number of these targeting approaches described for soluble antigen delivery have also been applied toward DC-targeting of microparticles. For example, mannose/mannan has also been surface bound to microparticles, demonstrating a dose-dependent response in DC uptake.Citation128 In this study, Wattendorf et al. also investigated targeting microparticles to DC integrins by incorporating the RGD adhesive peptide. Surface-modified microparticles with RGD peptide also increased DC uptake (this aspect is corroborated by othersCitation129) and combining both mannan and RGD provided cumulative effects on microparticle uptake by DCs. It was also found that these ligand-modified microparticles did not activate DCs, as determined by expression of MHC-II, CD86 and CD83.Citation128 Furthermore, microparticles have been surface modified to target DEC-205,Citation86,Citation98 demonstrating increased receptor-mediated uptake by DCs, migration to lymph nodes and stimulation of naïve T-cells in vivo.Citation86 Additionally, other microparticle modification approaches to target DCs include surface-tethering antibodies targeting CD40, and the αVβ3, αVβ5 and CD11c integrins, reporting differential levels of DC activation for each.Citation130,Citation131 This is an informative comparison, as the functional consequences of DC integrin binding is not well understood,Citation132,Citation133 and are currently under investigation.Citation134

Incorporating Immunomodulatory Signals into Microparticles

Immunogenic applications of microparticle-based vaccines have established precedent for the incorporation of immunomodulatory factors. In particular, incorporation of adjuvants in polymeric microparticles have ranged from the well-established (e.g., alumCitation135) to the well-defined (e.g., toll-like receptor ligands: CpG,Citation136,Citation137 poly(I:C),Citation87 monophosphoryl lipid ACitation138,Citation139). Within these types of combined antigen/adjuvant microparticle systems, co-encapsulation has been shown to be more beneficial than co-administration, by sustaining immunomodulatory signaling after antigen collection.Citation136,Citation140 Notably, the use of polymeric microparticles can facilitate a mechanistic understanding of DC endocytic processes. For example, the requirement that both antigen and TLR ligand need to be colocalized in the same endosome in order distinguish non-self-antigen from self antigen has recently been determined.Citation141 Together, these data begin to reveal to a set of design principles for immunogenic microparticle-based vaccine applications. Whether or not these design principles will have an effective tolerogenic/immunosuppressive counterpart remains to be seen.

In addition to stimulatory signals, a promising approach is the incorporation of factors such as chemokines and growth factors to both attract DCs and condition the local injection microenvironment. For example, chemokines such as the peptide N-formyl-Nle-Leu-Phe-Nle-Tyr-Lys,Citation142 macrophage inflammatory protein-3α (MIP-3α; CCL20),Citation142,Citation143 CCL19,Citation144 CCL21Citation144 and granulocyte macrophage colony-stimulating factor (GM-CSF)Citation145,Citation146 have all been investigated in biomaterial timed-release systems consisting of microparticles and/or hydrogels, demonstrating sustained ability to attract DCs. GM-CSF is a particularly interesting case, as use has been FDA-approved, and its effects are pleiotropic by: serving as a chemoattractant for both DCs and monocytes, promoting monocyte differentiation into DCs and increasing DC endocytic activity.Citation145,Citation146 Furthermore, treatment with GM-CSF in NOD mice has been shown to be able to delay T1D through acting on DCs and expanding Foxp3+ Tregs.Citation147

Compared to investigations with stimulatory molecules, incorporation of immunosuppressive molecules into microparticles has only recently begun to be explored. In particular, incorporation of rapamycin-loaded microparticles targeted to DCs has demonstrated generation of DCs maintained in an immature state,Citation148-Citation150 that resist maturation,Citation150 with lowered expression of ICAM-1,Citation149 with high levels of TGFβ1 produced,Citation150 and which have reduced T-cell stimulatory capacity.Citation148,Citation150 Additionally, microparticles with encapsulated IL-10 plasmid targeted to DCs generates DCs which elicit only weak T-cell stimulatory capacity,Citation151 and which expand Foxp3+ Tregs.Citation152

Dendritic Cell-Targeting Microparticle-Based Vaccine for T1D

Other types of particle-based approaches have been explored, such as the recent investigation using T-cell-targeting nanoparticles with tethered peptide-MHC molecules.Citation153 However, there is apparently only one study which has reported DC-targeting microparticle-based vaccines for T1D amelioration, led by the Giannoukakis group at the University of Pittsburgh School of Medicine. This line of research began with the demonstration that injection of NOD DCs treated ex vivo with antisense oligonucleotides knocking down expression of the costimulatory molecules CD40, CD80 and CD86, was able to significantly delay the incidence of diabetes in NOD mice.Citation154 This was subsequently linked to the increased prevalence of regulatory CD4+CD25+ T-cells through DC production of IL-7.Citation155 Remarkably, this approach is currently being investigated in a phase I clinical trial, in which antisense-treated autologous DCs are being administered to established T1D adult patients in order to establish safety (ClinicalTrials.gov Identifier: NCT00445913). Despite the promise of this study, anticipating the limitations of cell-based vaccinations, the Pittsburgh group is also pursuing a microparticle-based vaccine for T1D targeting DCs in vivo. This approach relies on the demonstrated abilityCitation67 of microparticle-encapsulated antisense oligonucleotides to effect gene knockdown in DCs. In the study by Phillips et al. polymeric microparticles carrying antisense oligonucleotides to CD40, CD80 and CD86 were delivered to a subcutaneous site proximal to the pancreatic lymph nodes in NOD mice.Citation10 The injected microparticle formulation augmented Foxp3+ Tregs and provoked hypo-responsiveness to β-cell antigen without compromising global immune responses to alloantigen. A fraction of the microparticles were found to accumulate primarily within the pancreatic lymph nodes and to a lesser extent the spleen after injection subcutaneously or intraperitoneally, indicating uptake and migration by phagocytes. Most importantly, the microparticle formulation both prevented and reversed new-onset T1D.Citation10 Notably, this formulation did not employ the use of a delivered antigen, which raises questions regarding the antigen-specific nature of the immune response. Additionally, it’s not clear if the microparticle system used (ProMaxxCitation156) is optimal for loading and extended delivery of antisense oligonucleotides, given that eight injections were necessary to effect reversal.

Concluding Remarks

Collectively, from these examples we can see that the advantages of microparticle-based vaccines delineated in the literature with immunogenic applications are beginning to be appreciated by the autoimmunity/immunosuppression community. Critically, the advantages of in vivo targeting of DCs/phagocytes, antigenic depot, incorporation of additional immunomodulatory factors and sustained release are all retained in these new applications, opening up a multitude of new avenues for research in T1D. Given the number of antigens, targeting molecules, immunomodulatory agents, chemokines and growth factors that may be of interest for delivery to DCs, the large number of potentially useful microparticle formulations is staggering. Advancement in key areas would enable transformative gains. Primarily, the identification of predictive in vitro markers is critical.Citation157 Additionally, development of a high-throughput screening methodology for the efficient, systematic examination of DC responses to microparticle formulationsCitation45 is also important, representing a step toward personalized vaccines.

References

  • Atkinson MA, Eisenbarth GS. Type 1 diabetes: new perspectives on disease pathogenesis and treatment. Lancet 2001; 358:221 - 9
  • Haller MJ, Atkinson MA, Schatz D. Type 1 diabetes mellitus: etiology, presentation and management. Pediatr Clin North Am 2005; 52:1553 - 78
  • bdul-Rasoul M, Habib H, Al-Khouly M. ‘The honeymoon phase in children with type 1 diabetes mellitus: frequency, duration and influential factors. Pediatr Diabetes 2006; 7:101 - 7
  • Lipton R, LaPorte RE, Becker DJ, Dorman JS, Orchard TJ, Atchison J, et al. Cyclosporin therapy for prevention and cure of IDDM. Epidemiological perspective of benefits and risks. Diabetes Care 1990; 13:776 - 84
  • Chatenoud L, Bluestone JA. CD3-specific antibodies: a portal to the treatment of autoimmunity. Nat Rev Immunol 2007; 7:622 - 32
  • Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature 2007; 449:419 - 26
  • Hilkens CM, Isaacs JD, Thomson AW. Development of dendritic cell-based immunotherapy for autoimmunity. Int Rev Immunol 2010; 29:156 - 83
  • Palucka K, Ueno H, Fay J, Banchereau J. Harnessing dendritic cells to generate cancer vaccines. Ann N Y Acad Sci 2009; 1174:88 - 98
  • http://clinicaltrials.gov/ ClinicalTrials.gov Identifier: NCT00445913; Autologous Dendritic Cell Therapy for Type 1 Diabetes Suppression: A Safety Study 2010.
  • Phillips B, Nylander K, Harnaha J, Machen J, Lakomy R, Styche A, et al. A microsphere-based vaccine prevents and reverses new-onset autoimmune diabetes. Diabetes 2008; 57:1544 - 55
  • MartIn-Fontecha A, Sebastiani S, Hopken UE, Uguccioni M, Lipp M, Lanzavecchia A, et al. Regulation of dendritic cell migration to the draining lymph node: impact on T lymphocyte traffic and priming. J Exp Med 2003; 198:615 - 21
  • Giannoukakis N, Phillips B, Trucco M. Toward a cure for type 1 diabetes mellitus: diabetes-suppressive dendritic cells and beyond. Pediatr Diabetes 2008; 9:4 - 13
  • Tacken PJ. de V, Torensma RI, Figdor CG. Dendritic-cell immunotherapy: from ex vivo loading to in vivo targeting. Nat Rev Immunol 2007; 7:790 - 802
  • Cohen S, Yoshioka T, Lucarelli M, Hwang LH, Langer R. Controlled delivery systems for proteins based on poly(lactic/glycolic acid) microspheres. Pharm Res 1991; 8:713 - 20
  • Zhao Z, Leong KW. Controlled delivery of antigens and adjuvants in vaccine development. J Pharm Sci 1996; 85:1261 - 70
  • Pean JM, Venier-Julienne MC, Boury F, Menei P, Denizot B, Benoit JP. NGF release from poly(D,L-lactide-co-glycolide) microspheres. Effect of some formulation parameters on encapsulated NGF stability. J Control Release 1998; 56:175 - 87
  • Schaefer MJ, Singh J. Effect of additives on stability of etoposide in PLGA microspheres. Drug Dev Ind Pharm 2001; 27:345 - 50
  • Steinman RM. Dendritic cells in vivo: a key target for a new vaccine science. Immunity 2008; 29:319 - 24
  • Steinman RM, Nussenzweig MC. Dendritic cells: features and functions. Immunol Rev 1980; 53:127 - 47
  • Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, et al. Immunobiology of dendritic cells. Annu Rev Immunol 2000; 18:767 - 811
  • Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic cells. Annu Rev Immunol 2003; 21:685 - 711
  • Morelli AE, Thomson AW. Dendritic cells: regulators of alloimmunity and opportunities for tolerance induction. Immunol Rev 2003; 196:125 - 46
  • Wing K, Sakaguchi S. Regulatory T cells exert checks and balances on self tolerance and autoimmunity. Nat Immunol 2010; 11:7 - 13
  • Mellanby RJ, Thomas DC, Lamb J. Role of regulatory T-cells in autoimmunity. Clin Sci (Lond) 2009; 116:639 - 49
  • Adler HS, Steinbrink K. Tolerogenic dendritic cells in health and disease: friend and foe!. Eur J Dermatol 2007; 17:476 - 91
  • Adorini L, Penna G. Dendritic cell tolerogenicity: a key mechanism in immunomodulation by vitamin D receptor agonists. Hum Immunol 2009; 70:345 - 52
  • Chang CC, Ciubotariu R, Manavalan JS, Yuan J, Colovai AI, Piazza F, et al. Tolerization of dendritic cells by T(S) cells: the crucial role of inhibitory receptors ILT3 and ILT4. Nat Immunol 2002; 3:237 - 43
  • Keir ME, Francisco LM, Sharpe AH. PD-1 and its ligands in T-cell immunity. Curr Opin Immunol 2007; 19:309 - 14
  • Huang L, Baban B, Johnson BA III, Mellor AL. Dendritic cells, indoleamine 2,3 dioxygenase and acquired immune privilege. Int Rev Immunol 2010; 29:133 - 55
  • Singh M, Chakrapani A, O’Hagan D. Nanoparticles and microparticles as vaccine-delivery systems. Expert Rev Vaccines 2007; 6:797 - 808
  • Shi Y, Huang G. Recent developments of biodegradable and biocompatible materials based micro/nanoparticles for delivering macromolecular therapeutics. Crit Rev Ther Drug Carrier Syst 2009; 26:29 - 84
  • Newman KD, Samuel J, Kwon G. Ovalbumin peptide encapsulated in poly(d,l lactic-co-glycolic acid) microspheres is capable of inducing a T helper type 1 immune response. J Control Release 1998; 54:49 - 59
  • Newman KD, Sosnowski DL, Kwon GS, Samuel J. Delivery of MUC1 mucin peptide by Poly(d,l-lactic-co-glycolic acid) microspheres induces type 1 T helper immune responses. J Pharm Sci 1998; 87:1421 - 7
  • Tamber H, Johansen P, Merkle HP, Gander B. Formulation aspects of biodegradable polymeric microspheres for antigen delivery. Adv Drug Deliv Rev 2005; 57:357 - 76
  • Lutolf MP, Hubbell JA. Synthetic biomaterials as instructive extracellular microenvironments for morphogenesis in tissue engineering. Nat Biotechnol 2005; 23:47 - 55
  • Peppas NA, Langer R. New challenges in biomaterials. Science 1994; 263:1715 - 20
  • Johansen P, Martinez Gomez JM, Gander B. Development of synthetic biodegradable microparticulate vaccines: a roller coaster story. Expert Rev Vaccines 2007; 6:471 - 4
  • Elamanchili P, Diwan M, Cao M, Samuel J. Characterization of poly(D,L-lactic-co-glycolic acid) based nanoparticulate system for enhanced delivery of antigens to dendritic cells. Vaccine 2004; 22:2406 - 12
  • Newman KD, Elamanchili P, Kwon GS, Samuel J. Uptake of poly(D,L-lactic-co-glycolic acid) microspheres by antigen-presenting cells in vivo. J Biomed Mater Res 2002; 60:480 - 6
  • Wang D, Molavi O, Lutsiak ME, Elamanchili P, Kwon GS, Samuel J. Poly(D,L-lactic-co-glycolic acid) microsphere delivery of adenovirus for vaccination. J Pharm Pharm Sci 2007; 10:217 - 30
  • Yoshida M, Mata J, Babensee JE. Effect of poly(lactic-co-glycolic acid) contact on maturation of murine bone marrow-derived dendritic cells. J Biomed Mater Res A 2007; 80:7 - 12
  • Yoshida M, Babensee JE. Molecular aspects of microparticle phagocytosis by dendritic cells. J Biomater Sci Polym Ed 2006; 17:893 - 907
  • Fischer S, Uetz-von AE, Waeckerle-Men Y, Groettrup M, Merkle HP, Gander B. The preservation of phenotype and functionality of dendritic cells upon phagocytosis of polyelectrolyte-coated PLGA microparticles. Biomaterials 2007; 28:994 - 1004
  • Hubbell JA, Thomas SN, Swartz MA. Materials engineering for immunomodulation. Nature 2009; 462:449 - 60
  • Acharya AP, Clare-Salzler MJ, Keselowsky BG. A high-throughput microparticle microarray platform for dendritic cell-targeting vaccines. Biomaterials 2009; 30:4168 - 77
  • Mailander V, Landfester K. Interaction of nanoparticles with cells. Biomacromolecules 2009; 10:2379 - 400
  • Waeckerle-Men Y, Allmen EU, Gander B, Scandella E, Schlosser E, Schmidtke G, et al. Encapsulation of proteins and peptides into biodegradable poly (D,L-lactide-co-glycolide) microspheres prolongs and enhances antigen presentation by human dendritic cells. Vaccine 2006; 24:1847 - 57
  • Waeckerle-Men Y, Gander B, Groettrup M. Delivery of tumor antigens to dendritic cells using biodegradable microspheres. Methods Mol Med 2005; 109:35 - 46
  • Waeckerle-Men Y, Groettrup M. PLGA microspheres for improved antigen delivery to dendritic cells as cellular vaccines. Adv Drug Deliv Rev 2005; 57:475 - 82
  • Hamishehkar H, Emami J, Najafabadi AR, Gilani K, Minaiyan M, Hassanzadeh K, et al. Pharmacokinetics and pharmacodynamics of controlled release insulin loaded PLGA microcapsules using dry powder inhaler in diabetic rats. Biopharm Drug Dispos 2010; 31:189 - 201
  • Hamishehkar H, Emami J, Najafabadi AR, Gilani K, Minaiyan M, Mahdavi H, et al. The effect of formulation variables on the characteristics of insulin-loaded poly(lactic-co-glycolic acid) microspheres prepared by a single phase oil in oil solvent evaporation method. Colloids Surf B Biointerfaces 2009; 74:340 - 9
  • Sheshala R, Peh KK, Darwis Y. Preparation, characterization, and in vivo evaluation of insulin-loaded PLA-PEG microspheres for controlled parenteral drug delivery. Drug Dev Ind Pharm 2009; 35:1364 - 74
  • Goncharova E, Ryzhikov E, Poryvaev V, Bulychev L, Karpyshev N, Maksyutov A, et al. Intranasal immunization with inactivated tick-borne encephalitis virus and the antigenic peptide 89-119 protects mice against intraperitoneal challenge. Int J Med Microbiol 2006; 296:195 - 201
  • Cui C, Stevens VC, Schwendeman SP. Injectable polymer microspheres enhance immunogenicity of a contraceptive peptide vaccine. Vaccine 2007; 25:500 - 9
  • Goldberg J, Shrikant P, Mescher MF. In vivo augmentation of tumor-specific CTL responses by class I/peptide antigen complexes on microspheres (large multivalent immunogen). J Immunol 2003; 170:228 - 35
  • Savina A, Amigorena S. Phagocytosis and antigen presentation in dendritic cells. Immunol Rev 2007; 219:143 - 56
  • Gerelchuluun T, Lee YH, Lee YR, Im SA, Song S, Park JS, et al. Dendritic cells process antigens encapsulated in a biodegradable polymer, poly(D,L-lactide-co-glycolide), via an alternate class I MHC processing pathway. Arch Pharm Res 2007; 30:1440 - 6
  • Shen H, Ackerman AL, Cody V, Giodini A, Hinson ER, Cresswell P, et al. Enhanced and prolonged cross-presentation following endosomal escape of exogenous antigens encapsulated in biodegradable nanoparticles. Immunology 2006; 117:78 - 88
  • Jones KS. Biomaterials as vaccine adjuvants. Biotechnol Prog 2008; 24:807 - 14
  • Men Y, Audran R, Thomasin C, Eberl G, Demotz S, Merkle HP, et al. MHC class I- and class II-restricted processing and presentation of microencapsulated antigens. Vaccine 1999; 17:1047 - 56
  • Heit A, Schmitz F, Haas T, Busch DH, Wagner H. Antigen co-encapsulated with adjuvants efficiently drive protective T cell immunity. Eur J Immunol 2007; 37:2063 - 74
  • Gerelchuluun T, Lee YH, Lee YR, Im SA, Song S, Park JS, et al. Dendritic cells process antigens encapsulated in a biodegradable polymer, poly(D,L-lactide-co-glycolide), via an alternate class I MHC processing pathway. Arch Pharm Res 2007; 30:1440 - 6
  • Shen Z, Reznikoff G, Dranoff G, Rock KL. Cloned dendritic cells can present exogenous antigens on both MHC class I and class II molecules. J Immunol 1997; 158:2723 - 30
  • Newman KD, Kwon GS, Miller GG, Chlumecky V, Samuel J. Cytoplasmic delivery of a macromolecular fluorescent probe by poly(d, l-lactic-co-glycolic acid) microspheres. J Biomed Mater Res 2000; 50:591 - 7
  • Caputo A, Sparnacci K, Ensoli B, Tondelli L. Functional polymeric nano/microparticles for surface adsorption and delivery of protein and DNA vaccines. Curr Drug Deliv 2008; 5:230 - 42
  • Lewis KJ, Irwin WJ, Akhtar S. Development of a sustained-release biodegradable polymer delivery system for site-specific delivery of oligonucleotides: characterization of P(LA-GA) copolymer microspheres in vitro. J Drug Target 1998; 5:291 - 302
  • Singh A, Nie H, Ghosn B, Qin H, Kwak LW, Roy K. Efficient modulation of T-cell response by dual-mode, single-carrier delivery of cytokine-targeted siRNA and DNA vaccine to antigen-presenting cells. Mol Ther 2008; 16:2011 - 21
  • Kutzler MA, Weiner DB. DNA vaccines: ready for prime time?. Nat Rev Genet 2008; 9:776 - 88
  • Steinman L. Inverse vaccination, the opposite of Jenner’s concept, for therapy of autoimmunity. J Intern Med 2010; 267:441 - 51
  • Jechlinger W. Optimization and delivery of plasmid DNA for vaccination. Expert Rev Vaccines 2006; 5:803 - 25
  • Wagstaff KM, Jans DA. Nucleocytoplasmic transport of DNA: enhancing non-viral gene transfer. Biochem J 2007; 406:185 - 202
  • Jiang W, Gupta RK, Deshpande MC, Schwendeman SP. Biodegradable poly(lactic-co-glycolic acid) microparticles for injectable delivery of vaccine antigens. Adv Drug Deliv Rev 2005; 57:391 - 410
  • Bramwell VW, Perrie Y. Particulate delivery systems for vaccines: what can we expect?. J Pharm Pharmacol 2006; 58:717 - 28
  • Son S, Lee WR, Joung YK, Kwon MH, Kim YS, Park KD. Optimized stability retention of a monoclonal antibody in the PLGA nanoparticles. Int J Pharm 2009; 368:178 - 85
  • Critchfield JM, Racke MK, Zuniga-Pflucker JC, Cannella B, Raine CS, Goverman J, et al. T cell deletion in high antigen dose therapy of autoimmune encephalomyelitis. Science 1994; 263:1139 - 43
  • Kurts C, Sutherland RM, Davey G, Li M, Lew AM, Blanas E, et al. CD8 T cell ignorance or tolerance to islet antigens depends on antigen dose. Proc Natl Acad Sci USA 1999; 96:12703 - 7
  • Lim DG, Hollsberg P, Hafler DA. Strength of prior stimuli determines the magnitude of secondary responsiveness in CD8+ T cells. Cell Immunol 2002; 217:36 - 46
  • Firpo EJ, Kong RK, Zhou Q, Rudensky AY, Roberts JM, Franza BR. Antigen-specific dose-dependent system for the study of an inheritable and reversible phenotype in mouse CD4+ T cells. Immunology 2002; 107:480 - 8
  • Piaggio E, Hartemann-Heurtier A, Cabarrocas J, Desbois S, Mars LT, Zappulla JP, et al. Maintaining or breaking CD8+ T-cell tolerance to beta islet cell antigens: lessons from transgenic mouse models. J Autoimmun 2004; 22:115 - 20
  • Ertl HC, Varga I, Xiang ZQ, Kaiser K, Stephens L, Otvos L Jr.. Poly (DL-lactide-co-glycolide) microspheres as carriers for peptide vaccines. Vaccine 1996; 14:879 - 85
  • Coombes AG, Lavelle EC, Jenkins PG, Davis SS. Single dose, polymeric, microparticle-based vaccines: the influence of formulation conditions on the magnitude and duration of the immune response to a protein antigen. Vaccine 1996; 14:1429 - 38
  • Yang TC, Millar J, Groves T, Grinshtein N, Parsons R, Takenaka S, et al. The CD8+ T cell population elicited by recombinant adenovirus displays a novel partially exhausted phenotype associated with prolonged antigen presentation that nonetheless provides long-term immunity. J Immunol 2006; 176:200 - 10
  • Strobel S. Immunity induced after a feed of antigen during early life: oral tolerance v. sensitisation. Proc Nutr Soc 2001; 60:437 - 42
  • Reddy ST, Swartz MA, Hubbell JA. Targeting dendritic cells with biomaterials: developing the next generation of vaccines. Trends Immunol 2006; 27:573 - 9
  • Jilek S, Ulrich M, Merkle HP, Walter E. Composition and surface charge of DNA-loaded microparticles determine maturation and cytokine secretion in human dendritic cells. Pharm Res 2004; 21:1240 - 7
  • Kwon YJ, James E, Shastri N, Frechet JM. In vivo targeting of dendritic cells for activation of cellular immunity using vaccine carriers based on pH-responsive microparticles. Proc Natl Acad Sci USA 2005; 102:18264 - 8
  • Heffernan MJ, Kasturi SP, Yang SC, Pulendran B, Murthy N. The stimulation of CD8+ T cells by dendritic cells pulsed with polyketal microparticles containing ion-paired protein antigen and poly(inosinic acid)-poly(cytidylic acid). Biomaterials 2009; 30:910 - 8
  • Wang C, Ge Q, Ting D, Nguyen D, Shen HR, Chen J, et al. Molecularly engineered poly(ortho ester) microspheres for enhanced delivery of DNA vaccines. Nat Mater 2004; 3:190 - 6
  • Watts C. Phagosome neutrality in host defense. Cell 2006; 126:17 - 9
  • Lee S, Yang SC, Heffernan MJ, Taylor WR, Murthy N. Polyketal microparticles: a new delivery vehicle for superoxide dismutase. Bioconjug Chem 2007; 18:4 - 7
  • Cerritelli S, Velluto D, Hubbell JA. PEG-SS-PPS: reduction-sensitive disulfide block copolymer vesicles for intracellular drug delivery. Biomacromolecules 2007; 8:1966 - 72
  • Reddy ST, van’d V, Simeoni E, Angeli V, Randolph GJ, O’Neil CP, et al. Exploiting lymphatic transport and complement activation in nanoparticle vaccines. Nat Biotechnol 2007; 25:1159 - 64
  • Flanary S, Hoffman AS, Stayton PS. Antigen delivery with poly(propylacrylic acid) conjugation enhances MHC-1 presentation and T-cell activation. Bioconjug Chem 2009; 20:241 - 8
  • Swartz MA, Hubbell JA, Reddy ST. Lymphatic drainage function and its immunological implications: from dendritic cell homing to vaccine design. Semin Immunol 2008; 20:147 - 56
  • Reddy ST, Rehor A, Schmoekel HG, Hubbell JA, Swartz MA. In vivo targeting of dendritic cells in lymph nodes with poly(propylene sulfide) nanoparticles. J Control Release 2006; 112:26 - 34
  • Fifis T, Gamvrellis A, Crimeen-Irwin B, Pietersz GA, Li J, Mottram PL, et al. Size-dependent immunogenicity: therapeutic and protective properties of nano-vaccines against tumors. J Immunol 2004; 173:3148 - 54
  • Uchida T, Goto S. Oral delivery of poly(lactide-co-glycolide) microspheres containing ovalbumin as vaccine formulation: particle size study. Biol Pharm Bull 1994; 17:1272 - 6
  • 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. J Control Release 2010;
  • Reis e Sousa. Germain RN. Major histocompatibility complex class I presentation of peptides derived from soluble exogenous antigen by a subset of cells engaged in phagocytosis. J Exp Med 1995; 182:841 - 51
  • 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
  • Hawiger D, Inaba K, Dorsett Y, Guo M, Mahnke K, Rivera M, et al. Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivo. J Exp Med 2001; 194:769 - 79
  • Lutz MB, Kurts C. Induction of peripheral CD4+ T-cell tolerance and CD8+ T-cell cross-tolerance by dendritic cells. Eur J Immunol 2009; 39:2325 - 30
  • Jiang W, Swiggard WJ, Heufler C, Peng M, Mirza A, Steinman RM, et al. The receptor DEC-205 expressed by dendritic cells and thymic epithelial cells is involved in antigen processing. Nature 1995; 375:151 - 5
  • Trumpfheller C, Finke JS, Lopez CB, Moran TM, Moltedo B, Soares H, et al. Intensified and protective CD4+ T cell immunity in mice with anti-dendritic cell HIV gag fusion antibody vaccine. J Exp Med 2006; 203:607 - 17
  • 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 IFNgamma by an IL-12-independent but CD70-dependent mechanism in vivo. J Exp Med 2007; 204:1095 - 106
  • Suzue K, Young RA. Adjuvant-free hsp70 fusion protein system elicits humoral and cellular immune responses to HIV-1 p24. J Immunol 1996; 156:873 - 9
  • Guermonprez P, Khelef N, Blouin E, Rieu P, Ricciardi-Castagnoli P, Guiso N, et al. The adenylate cyclase toxin of Bordetella pertussis binds to target cells via the alpha(M)beta(2) integrin (CD11b/CD18). J Exp Med 2001; 193:1035 - 44
  • Tacken PJ, Torensma R, Figdor CG. Targeting antigens to dendritic cells in vivo. Immunobiology 2006; 211:599 - 608
  • Zhang L, Tang Y, Akbulut H, Zelterman D, Linton PJ, Deisseroth AB. An adenoviral vector cancer vaccine that delivers a tumor-associated antigen/CD40-ligand fusion protein to dendritic cells. Proc Natl Acad Sci USA 2003; 100:15101 - 6
  • Apostolopoulos V, Pietersz GA, Gordon S, Martinez-Pomares L, McKenzie IF. Aldehyde-mannan antigen complexes target the MHC class I antigen-presentation pathway. Eur J Immunol 2000; 30:1714 - 23
  • Berry JD, Licea A, Popkov M, Cortez X, Fuller R, Elia M, et al. Rapid monoclonal antibody generation via dendritic cell targeting in vivo. Hybrid Hybridomics 2003; 22:23 - 31
  • Schuurhuis DH, Ioan-Facsinay A, Nagelkerken B, van Schip JJ, Sedlik C, Melief CJ, et al. Antigen-antibody immune complexes empower dendritic cells to efficiently prime specific CD8+ CTL responses in vivo. J Immunol 2002; 168:2240 - 6
  • Kalergis AM, Ravetch JV. Inducing tumor immunity through the selective engagement of activating Fcgamma receptors on dendritic cells. J Exp Med 2002; 195:1653 - 9
  • 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 - 52
  • 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
  • Boscardin SB, Hafalla JC, Masilamani RF, Kamphorst AO, Zebroski HA, Rai U, et al. Antigen targeting to dendritic cells elicits long-lived T cell help for antibody responses. J Exp Med 2006; 203:599 - 606
  • Tacken PJ, de V, Gijzen KI, Joosten B, Wu D, Rother RP, et al. Effective induction of naive and recall T-cell responses by targeting antigen to human dendritic cells via a humanized anti-DC-SIGN antibody. Blood 2005; 106:1278 - 85
  • Engering A, Geijtenbeek TB, van Vliet SJ, Wijers M, van LE, Demaurex N, et al. The dendritic cell-specific adhesion receptor DC-SIGN internalizes antigen for presentation to T cells. J Immunol 2002; 168:2118 - 26
  • Tillman BW, de Gruijl TD, Luykx-de Bakker SA, Scheper RJ, Pinedo HM, Curiel TJ, et al. Maturation of dendritic cells accompanies high-efficiency gene transfer by a CD40-targeted adenoviral vector. J Immunol 1999; 162:6378 - 83
  • 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
  • Mahnke K, Qian Y, Knop J, Enk AH. Induction of CD4+/CD25+ regulatory T cells by targeting of antigens to immature dendritic cells. Blood 2003; 101:4862 - 9
  • Bruder D, Westendorf AM, Hansen W, Prettin S, Gruber AD, Qian Y, et al. On the edge of autoimmunity: T-cell stimulation by steady-state dendritic cells prevents autoimmune diabetes. Diabetes 2005; 54:3395 - 401
  • Mukhopadhaya A, Hanafusa T, Jarchum I, Chen YG, Iwai Y, Serreze DV, et al. Selective delivery of beta cell antigen to dendritic cells in vivo leads to deletion and tolerance of autoreactive CD8+ T cells in NOD mice. Proc Natl Acad Sci USA 2008; 105:6374 - 9
  • Nimmerjahn F, Ravetch JV. Fcgamma receptors: old friends and new family members. Immunity 2006; 24:19 - 28
  • Ravetch JV, Lanier LL. Immune inhibitory receptors. Science 2000; 290:84 - 9
  • Yamazaki S, Dudziak D, Heidkamp GF, Fiorese C, Bonito AJ, Inaba K, et al. CD8+ CD205+ splenic dendritic cells are specialized to induce Foxp3+ regulatory T cells. J Immunol 2008; 181:6923 - 33
  • Fujikado N, Saijo S, Yonezawa T, Shimamori K, Ishii A, Sugai S, et al. Dcir deficiency causes development of autoimmune diseases in mice due to excess expansion of dendritic cells. Nat Med 2008; 14:176 - 80
  • Wattendorf U, Coullerez G, Voros J, Textor M, Merkle HP. Mannose-based molecular patterns on stealth microspheres for receptor-specific targeting of human antigen-presenting cells. Langmuir 2008; 24:11790 - 802
  • Faraasen S, Voros J, Csucs G, Textor M, Merkle HP, Walter E. Ligand-specific targeting of microspheres to phagocytes by surface modification with poly(L-lysine)-grafted poly(ethylene glycol) conjugate. Pharm Res 2003; 20:237 - 46
  • Kempf M, Mandal B, Jilek S, Thiele L, Voros J, Textor M, et al. Improved stimulation of human dendritic cells by receptor engagement with surface-modified microparticles. J Drug Target 2003; 11:11 - 8
  • Regner M, Martinez X, Belnoue E, Sun CM, Boisgerault F, Lambert PH, et al. Partial activation of neonatal CD11c+ dendritic cells and induction of adult-like CD8+ cytotoxic T cell responses by synthetic microspheres. J Immunol 2004; 173:2669 - 74
  • Acharya AP, Dolgova NV, Clare-Salzler MJ, Keselowsky BG. Adhesive substrate-modulation of adaptive immune responses. Biomaterials 2008; 29:4736 - 50
  • Acharya AP, Dolgova NV, Xia CQ, Clare-Salzler MJ, Keselowsky BG. Adhesive substrates modulate activation and stimulatory capacity of non-obese diabetic mouse-derived dendritic cells. Acta Biomater 2010; 7:180 - 92
  • Acharya AP, Dolgova NV, Xia CQ, Clare-Salzler MJ, Becker ML, Moore NM, et al. The modulation of dendritic cell integrin binding and activation by RGD-peptide density gradient substrates. Biomaterials 2010; 31:7444 - 54
  • Raghuvanshi RJ, Mistra A, Talwar GP, Levy RJ, Labhasetwar V. Enhanced immune response with a combination of alum and biodegradable nanoparticles containing tetanus toxoid. J Microencapsul 2001; 18:723 - 32
  • Schlosser E, Mueller M, Fischer S, Basta S, Busch DH, Gander B, et al. TLR ligands and antigen need to be coencapsulated into the same biodegradable microsphere for the generation of potent cytotoxic T lymphocyte responses. Vaccine 2008; 26:1626 - 37
  • Vollmer J, Weeratna R, Payette P, Jurk M, Schetter C, Laucht M, et al. Characterization of three CpG oligodeoxynucleotide classes with distinct immunostimulatory activities. Eur J Immunol 2004; 34:251 - 62
  • Elamanchili P, Lutsiak CM, Hamdy S, Diwan M, Samuel J. “Pathogen-mimicking” nanoparticles for vaccine delivery to dendritic cells. J Immunother 2007; 30:378 - 95
  • Hamdy S, Molavi O, Ma Z, Haddadi A, Alshamsan A, Gobti Z, et al. Co-delivery of cancer-associated antigen and Toll-like receptor 4 ligand in PLGA nanoparticles induces potent CD8+ T cell-mediated anti-tumor immunity. Vaccine 2008; 26:5046 - 57
  • Malyala P, Chesko J, Ugozzoli M, Goodsell A, Zhou F, Vajdy M, et al. The potency of the adjuvant, CpG oligos, is enhanced by encapsulation in PLG microparticles. J Pharm Sci 2008; 97:1155 - 64
  • Blander JM, Medzhitov R. Toll-dependent selection of microbial antigens for presentation by dendritic cells. Nature 2006; 440:808 - 12
  • Zhao X, Jain S, Benjamin LH, Gonzalez S, Irvine DJ. Directed cell migration via chemoattractants released from degradable microspheres. Biomaterials 2005; 26:5048 - 63
  • Singh A, Suri S, Roy K. In-situ crosslinking hydrogels for combinatorial delivery of chemokines and siRNA-DNA carrying microparticles to dendritic cells. Biomaterials 2009; 30:5187 - 200
  • Hori Y, Winans AM, Huang CC, Horrigan EM, Irvine DJ. Injectable dendritic cell-carrying alginate gels for immunization and immunotherapy. Biomaterials 2008; 29:3671 - 82
  • Ali OA, Mooney DJ. Sustained GM-CSF and PEI condensed pDNA presentation increases the level and duration of gene expression in dendritic cells. J Control Release 2008; 132:273 - 8
  • Ali OA, Huebsch N, Cao L, Dranoff G, Mooney DJ. Infection-mimicking materials to program dendritic cells in situ. Nat Mater 2009; 8:151 - 8
  • Cheatem D, Ganesh BB, Gangi E, Vasu C, Prabhakar BS. Modulation of dendritic cells using granulocyte-macrophage colony-stimulating factor (GM-CSF) delays type 1 diabetes by enhancing CD4+CD25+ regulatory T cell function. Clin Immunol 2009; 131:260 - 70
  • Jhunjhunwala S, Raimondi G, Thomson AW, Little SR. Delivery of rapamycin to dendritic cells using degradable microparticles. J Control Release 2009; 133:191 - 7
  • Das S, Haddadi A, Veniamin S, Samuel J. Delivery of rapamycin-loaded nanoparticle down regulates ICAM-1 expression and maintains an immunosuppressive profile in human CD34+ progenitor-derived dendritic cells. J Biomed Mater Res A 2008; 85:983 - 92
  • Haddadi A, Elamanchili P, Lavasanifar A, Das S, Shapiro J, Samuel J. Delivery of rapamycin by PLGA nanoparticles enhances its suppressive activity on dendritic cells. J Biomed Mater Res A 2008; 84:885 - 98
  • Jia L, Kovacs JR, Zheng Y, Gawalt ES, Shen H, Meng WS. Attenuated alloreactivity of dendritic cells engineered with surface-modified microspheres carrying a plasmid encoding interleukin-10. Biomaterials 2006; 27:2076 - 82
  • Jia L, Kovacs JR, Zheng Y, Shen H, Gawalt ES, Meng WS. Expansion of Foxp3-expressing regulatory T cells in vitro by dendritic cells modified with polymeric particles carrying a plasmid encoding interleukin-10. Biomaterials 2008; 29:1250 - 61
  • Tsai S, Shameli A, Yamanouchi J, Clemente-Casares X, Wang J, Serra P, et al. Reversal of autoimmunity by boosting memory-like autoregulatory T cells. Immunity 2010; 32:568 - 80
  • Machen J, Harnaha J, Lakomy R, Styche A, Trucco M, Giannoukakis N. Antisense oligonucleotides down-regulating costimulation confer diabetes-preventive properties to nonobese diabetic mouse dendritic cells. J Immunol 2004; 173:4331 - 41
  • Harnaha J, Machen J, Wright M, Lakomy R, Styche A, Trucco M, et al. Interleukin-7 is a survival factor for CD4+ CD25+ T-cells and is expressed by diabetes-suppressive dendritic cells. Diabetes 2006; 55:158 - 70
  • Mandal TK. Inhaled insulin for diabetes mellitus. Am J Health Syst Pharm 2005; 62:1359 - 64
  • Poland GA, Ovsyannikova IG, Jacobson RM, Smith DI. Heterogeneity in vaccine immune response: the role of immunogenetics and the emerging field of vaccinomics. Clin Pharmacol Ther 2007; 82:653 - 64

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.