Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 12(1): 4445, 2021 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-34290245

RESUMO

Immune checkpoint inhibitors demonstrate clinical activity in many tumor types, however, only a fraction of patients benefit. Combining CD137 agonists with these inhibitors increases anti-tumor activity preclinically, but attempts to translate these observations to the clinic have been hampered by systemic toxicity. Here we describe a human CD137xPD-L1 bispecific antibody, MCLA-145, identified through functional screening of agonist- and immune checkpoint inhibitor arm combinations. MCLA-145 potently activates T cells at sub-nanomolar concentrations, even under suppressive conditions, and enhances T cell priming, differentiation and memory recall responses. In vivo, MCLA-145 anti-tumor activity is superior to immune checkpoint inhibitor comparators and linked to recruitment and intra-tumor expansion of CD8 + T cells. No graft-versus-host-disease is observed in contrast to other antibodies inhibiting the PD-1 and PD-L1 pathway. Non-human primates treated with 100 mg/kg/week of MCLA-145 show no adverse effects. The conditional activation of CD137 signaling by MCLA-145, triggered by neighboring cells expressing >5000 copies of PD-L1, may provide both safety and potency advantages.


Assuntos
Ligante 4-1BB/agonistas , Anticorpos Biespecíficos/farmacologia , Antígeno B7-H1/antagonistas & inibidores , Linfócitos T CD8-Positivos/efeitos dos fármacos , Inibidores de Checkpoint Imunológico/farmacologia , Ligante 4-1BB/imunologia , Animais , Anticorpos Biespecíficos/imunologia , Antígeno B7-H1/imunologia , Linfócitos T CD8-Positivos/imunologia , Epitopos , Humanos , Inibidores de Checkpoint Imunológico/imunologia , Tolerância Imunológica/efeitos dos fármacos , Memória Imunológica/efeitos dos fármacos , Imunoterapia , Ativação Linfocitária/efeitos dos fármacos
2.
Molecules ; 24(9)2019 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-31083610

RESUMO

Optimal targeting of nanoparticles (NP) to dendritic cells (DCs) receptors to deliver cancer-specific antigens is key to the efficient induction of anti-tumour immune responses. Poly (lactic-co-glycolic acid) (PLGA) nanoparticles containing tètanus toxoid and gp100 melanoma-associated antigen, toll-like receptor adjuvants were targeted to the DC-SIGN receptor in DCs by specific humanized antibodies or by ICAM3-Fc fusion proteins, which acts as the natural ligand. Despite higher binding and uptake efficacy of anti-DC-SIGN antibody-targeted NP vaccines than ICAM3-Fc ligand, no difference were observed in DC activation markers CD80, CD83, CD86 and CCR7 induced. DCs loaded with NP coated with ICAM3-Fc appeared more potent in activating T cells via cross-presentation than antibody-coated NP vaccines. This fact could be very crucial in the design of new cancer vaccines.


Assuntos
Vacinas Anticâncer/metabolismo , Células Dendríticas/metabolismo , Molécula 3 de Adesão Intercelular/metabolismo , Nanopartículas/química , Vacinas Anticâncer/química , Células Cultivadas , Humanos , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Leucócitos/metabolismo , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Receptores de IgG/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
3.
Nanomedicine (Lond) ; 12(5): 491-510, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28181470

RESUMO

AIM: Dendritic cells rapidly capture nanoparticles and induce a potent cellular immune response. It is yet unknown whether the immunological response induced by slow release of encapsulated versus soluble antigen and adjuvant is superior. MATERIALS & METHODS: The kinetics of poly(lactic-co-glycolic acid) PLGA nanoparticles antigen release was studied by the DQ-bovine serum albumin (BSA) self-quenching antigen model. The immunological response induced was evaluated by means of dendritic cell activation/maturation markers, cytokine production and their ability to drive antigen-specific T-cell proliferation. RESULTS & CONCLUSION: PLGA-encapsulated antigen and adjuvant showed an enhanced T-cell response when compared with soluble vaccine components by increasing antigenicity and adjuvanticity. Although the kinetic profile followed the same pattern, encapsulation increased strength and duration of the response.


Assuntos
Células Dendríticas/imunologia , Imunidade Celular/efeitos dos fármacos , Imunogenicidade da Vacina/imunologia , Nanopartículas/administração & dosagem , Linfócitos T/imunologia , Animais , Antígenos/química , Antígenos/imunologia , Bovinos , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Humanos , Imunogenicidade da Vacina/efeitos dos fármacos , Ácido Láctico/administração & dosagem , Ácido Láctico/química , Ácido Láctico/imunologia , Nanopartículas/química , Ácido Poliglicólico/administração & dosagem , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Soroalbumina Bovina/química , Soroalbumina Bovina/imunologia , Linfócitos T/efeitos dos fármacos
4.
Mol Pharm ; 11(12): 4299-313, 2014 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-25290882

RESUMO

Dendritic cells (DCs) are the most potent antigen-presenting cells (APCs), involved in the induction of immunity and currently exploited for antitumor immunotherapies. An optimized noninvasive imaging modality capable of determining and quantifying DC-targeted nanoparticle (NP) trajectories could provide valuable information regarding therapeutic vaccine outcome. Here, targeted poly(d,l-lactide-co-glycolide) nanoparticles (PLGA NPs) recognizing DC receptors were equipped with superparamagnetic iron oxide particles (SPIO) or gold nanoparticles with fluorescently labeled antigen. The fluorescent label allowed for rapid analysis and quantification of DC-specific uptake of targeted PLGA NPs in comparison to uptake by other cells. Transmission electron microscopy (TEM) showed that a fraction of the encapsulated antigen reached the lysosomal compartment of DCs, where SPIO and gold were already partially released. However, part of the PLGA NPs localized within the cytoplasm, as confirmed by confocal microscopy. DCs targeted with NPs carrying SPIO or fluorescent antigen were detected within lymph nodes as early as 1 h after injection by magnetic resonance imaging (MRI). Despite the fact that targeting did not markedly affect PLGA NP biodistribution on organism and tissue level, it increased delivery of NPs to DCs residing in peripheral lymph nodes and resulted in enhanced T cell proliferation. In conclusion, two imaging agents within a single carrier allows tracking of targeted PLGA NPs at the subcellular, cellular, and organismal levels, thereby facilitating the rational design of in vivo targeted vaccination strategies.


Assuntos
Portadores de Fármacos/química , Nanopartículas/química , Nanoestruturas/química , Animais , Células Cultivadas , Meios de Contraste , Células Dendríticas/imunologia , Humanos , Camundongos , Microscopia Eletrônica de Transmissão , Vacinas/imunologia
5.
Blood ; 121(15): 2836-44, 2013 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-23390195

RESUMO

Vaccination is among the most efficient forms of immunotherapy. Although sometimes inducing lifelong protective B-cell responses, T-cell-mediated immunity remains challenging. Targeting antigen to dendritic cells (DCs) is an extensively explored concept aimed at improving cellular immunity. The identification of various DC subsets with distinct functional characteristics now allows for the fine-tuning of targeting strategies. Although some of these DC subsets are regarded as superior for (cross-) priming of naive T cells, controversies still remain about which subset represents the best target for immunotherapy. Because targeting the antigen alone may not be sufficient to obtain effective T-cell responses, delivery systems have been developed to target multiple vaccine components to DCs. In this Perspective, we discuss the pros and cons of targeting DCs: if targeting is beneficial at all and which vaccine vehicles and immunization routes represent promising strategies to reach and activate DCs.


Assuntos
Linfócitos B/imunologia , Células Dendríticas/imunologia , Imunidade Celular/imunologia , Linfócitos T/imunologia , Antígenos/imunologia , Humanos , Imunoterapia/métodos , Modelos Imunológicos , Vacinas/administração & dosagem , Vacinas/imunologia
6.
Nanomedicine (Lond) ; 7(10): 1591-610, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23148541

RESUMO

Current retroviral treatments have reduced AIDS to a chronic disease for most patients. However, given drug-related side effects, the emergence of drug-resistant strains and the persistence of viral replication, the development of alternative treatments is a pressing need. This review focuses on recent developments in HIV immunotherapy treatments, with particular emphasis on current vaccination strategies for optimizing the induction of an effective immune response by the recruitment of dendritic cells. In addition to cell-based therapies, targeted strategies aiming to deliver synthetic HIV peptides to dendritic cell-specific receptors in vivo will be discussed.


Assuntos
Células Dendríticas/química , Antígenos HIV/imunologia , HIV-1/imunologia , Vacinas contra a AIDS/imunologia , Reações Cruzadas , Humanos
7.
Eur J Immunol ; 42(8): 1989-98, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22653683

RESUMO

DCs are regarded as key APCs that initiate humoral and cellular immune responses. Consequently, targeted delivery of Ag toward DC-specific receptors enhances vaccine efficacy. DC-SIGN is a C-type lectin receptor that facilitates DC-specific delivery of Ag. This is accomplished by conjugating Ag to receptor-specific Ab or carbohydrate ligands that bind to its carbohydrate recognition domain. Here, we investigated the fate of DC-SIGN following receptor triggering with Ab. Both whole and single-chain Ab induced rapid internalization of about half of the surface receptor molecules. Biochemical studies showed that about half of the receptor molecules were still intracellular after 3 h, while minimal or no resurfacing of internalized or newly synthesized unbound DC-SIGN molecules was observed. Prolonged exposure of DCs to DC-SIGN Ab, but not carbohydrate ligands, resulted in reduced receptor expression levels, which lasted up to 2 days following removal of the Ab. In addition, exposure to DC-SIGN Ab reduced the ability of the receptor to internalize. Consequently, DC-SIGN showed a poor ability to accumulate targeting Abs within DCs. Vaccine efficacy may therefore be enhanced by strategies increasing the amount of Ag entering via a single receptor molecule, such as the use of targeting moieties allowing DC-SIGN recycling or Ab-coated vaccine carriers.


Assuntos
Anticorpos/metabolismo , Antígenos/metabolismo , Carboidratos/imunologia , Moléculas de Adesão Celular/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Lectinas Tipo C/metabolismo , Receptores de Superfície Celular/metabolismo , Anticorpos/imunologia , Apresentação de Antígeno , Células Cultivadas , Humanos , Ligantes , Transporte Proteico , Vacinas/imunologia
8.
Methods Enzymol ; 509: 143-63, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22568905

RESUMO

Dendritic cells (DCs) are key players in the initiation of adaptive immune responses and are currently exploited in immunotherapy for treatment of cancer and infectious diseases. Development of targeted nanodelivery systems carrying vaccine components, including antigens and adjuvants, to DCs in vivo represents a promising strategy to enhance immune responses. Delivering particulate vaccines specifically to DCs and preventing nonspecific uptake by other endocytotic cells are challenging. Size represents a critical parameter determining whether particulate vaccines can penetrate lymph nodes and reach resident DCs. Specific delivery is further enhanced by actively targeting DC-specific receptors. This chapter discusses the rationale for the use of particle-based vaccines and provides an overview of antigen-delivery vehicles currently under investigation. In addition, we discuss how vaccine delivery systems may be developed, focusing on liposomes, PLGA polymers, and gold nanoparticles, to obtain safe and efficacious vaccines.


Assuntos
Células Dendríticas/imunologia , Imunoterapia/métodos , Nanocápsulas/química , Adjuvantes Imunológicos/química , Sequência de Aminoácidos , Animais , Anticorpos Imobilizados/química , Antígenos/química , Antígenos/imunologia , Ouro/química , Humanos , Fragmentos Fc das Imunoglobulinas/química , Ácido Láctico , Lipossomos/química , Dados de Sequência Molecular , Nanoconjugados/química , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Vacinas Sintéticas/química
9.
Cancer Immunol Immunother ; 61(11): 2003-11, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22527252

RESUMO

PURPOSE: Keyhole limpet hemocyanin (KLH) attracts biomedical interest because of its remarkable immunostimulatory properties. Currently, KLH is used as vaccine adjuvant, carrier protein for haptens and as local treatment for bladder cancer. Since a quantitative human anti-KLH assay is lacking, it has not been possible to monitor the dynamics of KLH-specific antibody (Ab) responses after in vivo KLH exposure. We designed a quantitative assay to measure KLH-specific Abs in humans and retrospectively studied the relation between vaccination parameters and the vaccine-induced anti-KLH Ab responses. EXPERIMENTAL DESIGN: Anti-KLH Abs were purified from pooled serum of melanoma patients who have responded to KLH as a vaccine adjuvant. Standard isotype-specific calibration curves were generated to measure KLH-specific Ab responses in individual serum samples using ELISA. RESULTS: KLH-specific IgM, IgA, IgG and all IgG-subclasses were accurately measured at concentrations as low as 20 µg/ml. The intra- and inter-assay coefficients of variation of this ELISA were below 6.7 and 9.9 %, respectively. Analyses of 128 patients demonstrated that mature DC induced higher levels of KLH-specific IgG compared to immature DC, prior infusion with anti-CD25 abolished IgG and IgM production and patients with locoregional disease developed more robust IgG responses than advanced metastatic melanoma patients. CONCLUSIONS: We present the first quantitative assay to measure KLH-specific Abs in human serum, which now enables monitoring both the dynamics and absolute concentrations of humoral immune responses in individuals exposed to KLH. This assay may provide a valuable biomarker for the immunogenicity and clinical effectiveness of KLH-containing vaccines and therapies.


Assuntos
Anticorpos/sangue , Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Ensaio de Imunoadsorção Enzimática , Hemocianinas/imunologia , Melanoma/terapia , Adjuvantes Imunológicos/uso terapêutico , Vacinas Anticâncer/uso terapêutico , Feminino , Humanos , Imunidade Humoral , Imunoterapia , Masculino , Melanoma/imunologia , Reprodutibilidade dos Testes , Estudos Retrospectivos
10.
Biomaterials ; 33(16): 4229-39, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22410170

RESUMO

Vaccine efficacy is improved upon specific delivery to professional antigen (Ag) presenting cells, such as dendritic cells (DCs). Antigenicity and adjuvanticity of vaccine components can be enhanced by encapsulation within nanoparticle (NP) vaccine carriers that are targeted to the human DC-specific C-type lectin receptor DC-SIGN. Here we used two strategies to target vaccines components to DC-SIGN: 1) carbohydrates as natural receptor ligands and 2) receptor-specific antibodies (Abs). To determine the optimal targeting strategy, we coated NP vaccines harboring MHC class I or II-restricted Ags and the TLR ligands (TLRLs) poly I:C and resiquimod with either the DC-SIGN ligands Lewis-X (Le(x)), mannosylated lipoarabinomannan (ManLAM), glycosylated HIV protein gp120, or three distinct DC-SIGN Abs. Although, because of their lower MW, surface coating of NP vaccines with carbohydrates resulted in a higher number of surface molecules per NP than coating with Abs, NP vaccines carrying Abs were more effectively bound and internalized by human DCs than carriers harboring Le(x), ManLAM or gp120. Furthermore, NP vaccines harboring TLRLs triggered significant induction of DC maturation markers when compared to those without TLRLs, irrespective of the targeting moiety. Ab- and gp120-mediated targeting induced equally high levels of proinflammatory cytokines and increased presentation of the MHC class I-restricted epitope. By contrast, presentation of the MHC class II-restricted epitope was more efficient upon Ab-mediated targeting than when using gp120, Le(x) or ManLAM. From these findings we conclude that receptor-specific Abs are more effective than carbohydrates for DC-targeted vaccination strategies.


Assuntos
Anticorpos/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Carboidratos/imunologia , Moléculas de Adesão Celular/imunologia , Lectinas Tipo C/imunologia , Receptores de Superfície Celular/imunologia , Vacinas/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Cinética , Ativação Linfocitária , Receptores Toll-Like/imunologia
11.
Blood ; 119(10): 2284-92, 2012 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-22234694

RESUMO

CLEC9A is a recently discovered C-type lectin receptor involved in sensing necrotic cells. In humans, this receptor is selectively expressed by BDCA3(+) myeloid dendritic cells (mDCs), which have been proposed to be the main human cross-presenting mDCs and may represent the human homologue of murine CD8(+) DCs. In mice, it was demonstrated that antigens delivered with antibodies to CLEC9A are presented by CD8(+) DCs to both CD4(+) and CD8(+) T cells and induce antitumor immunity in a melanoma model. Here we assessed the ability of CLEC9A to mediate antigen presentation by human BDCA3(+) mDCs, which represent < 0.05% of peripheral blood leukocytes. We demonstrate that CLEC9A is only expressed on immature BDCA3(+) mDCs and that cell surface expression is lost after TLR-mediated maturation. CLEC9A triggering via antibody binding rapidly induces receptor internalization but does not affect TLR-induced cytokine production or expression of costimulatory molecules. More importantly, antigens delivered via CLEC9A antibodies to BDCA3(+) mDCs are presented by both MHC class I (cross-presentation) and MHC class II to antigen-specific T cells. We conclude that CLEC9A is a promising target for in vivo antigen delivery in humans to increase the efficiency of vaccines against infectious or malignant diseases.


Assuntos
Apresentação de Antígeno/imunologia , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Lectinas Tipo C/imunologia , Receptores Mitogênicos/imunologia , Antígenos CD/imunologia , Antígenos CD/metabolismo , Antígenos de Superfície/imunologia , Antígenos de Superfície/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Células Cultivadas , Células Dendríticas/metabolismo , Endocitose/imunologia , Citometria de Fluxo , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Lectinas Tipo C/metabolismo , Antígenos de Histocompatibilidade Menor , Células Mieloides/imunologia , Células Mieloides/metabolismo , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo , Receptores Mitogênicos/metabolismo , Trombomodulina , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo
12.
Blood ; 118(26): 6836-44, 2011 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-21967977

RESUMO

Effective vaccines consist of 2 components: immunodominant antigens and effective adjuvants. Whereas it has been demonstrated that targeted delivery of antigens to dendritic cells (DCs) improves vaccine efficacy, we report here that co-targeting of TLR ligands (TLRLs) to DCs strongly enhances adjuvanticity and immunity. We encapsulated ligands for intracellular TLRs within biodegradable nanoparticles coated with Abs recognizing DC-specific receptors. Targeted delivery of TLRLs to human DCs enhanced the maturation and production of immune stimulatory cytokines and the Ag-specific activation of naive CD8(+) T cells. In vivo studies demonstrated that nanoparticles carrying Ag induced cytotoxic T-lymphocyte responses at 100-fold lower adjuvant dose when TLRLs were co-encapsulated instead of administered in soluble form. Moreover, the efficacy of these targeted TLRLs reduced the serum cytokine storm and related toxicity that is associated with administration of soluble TLRLs. We conclude that the targeted delivery of adjuvants may improve the efficacy and safety of DC-based vaccines.


Assuntos
Células Dendríticas/imunologia , Ligantes , Receptores Toll-Like/imunologia , Vacinas/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Células Cultivadas , Citocinas/sangue , Citocinas/imunologia , Citocinas/metabolismo , Citotoxicidade Imunológica/imunologia , Células Dendríticas/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Feminino , Citometria de Fluxo , Humanos , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Monócitos/imunologia , Monócitos/metabolismo , Nanopartículas/administração & dosagem , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Fatores de Tempo , Receptores Toll-Like/metabolismo , Vacinas/administração & dosagem
13.
Blood ; 118(15): 4111-9, 2011 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-21860028

RESUMO

Targeting antigens to dendritic cell (DC)-specific receptors, such as DC-SIGN, induces potent T cell-mediated immune responses. DC-SIGN is a transmembrane C-type lectin receptor with a long extracellular neck region and a carbohydrate recognition domain (CRD). Thus far, only antibodies binding the CRD have been used to target antigens to DC-SIGN. We evaluated the endocytic pathway triggered by antineck antibodies as well as their intracellular routing and ability to induce CD8(+) T-cell activation. In contrast to anti-CRD antibodies, antineck antibodies induced a clathrin-independent mode of DC-SIGN internalization, as demonstrated by the lack of colocalization with clathrin and the observation that silencing clathrin did not affect antibody internalization in human DCs. Interestingly, we observed that anti-neck and anti-CRD antibodies were differentially routed within DCs. Whereas anti-CRD antibodies were mainly routed to late endosomal compartments, anti-neck antibodies remained associated with early endosomal compartments positive for EEA-1 and MHC class I for up to 2 hours after internalization. Finally, cross-presentation of protein antigen conjugated to antineck antibodies was approximately 1000-fold more effective than nonconjugated antigen. Our studies demonstrate that anti-neck antibodies trigger a distinct mode of DC-SIGN internalization that shows potential for targeted vaccination strategies.


Assuntos
Apresentação de Antígeno/fisiologia , Linfócitos T CD8-Positivos/imunologia , Moléculas de Adesão Celular/imunologia , Apresentação Cruzada/fisiologia , Células Dendríticas/imunologia , Endossomos/imunologia , Lectinas Tipo C/imunologia , Lisossomos/imunologia , Receptores de Superfície Celular/imunologia , Animais , Anticorpos/genética , Anticorpos/imunologia , Antígenos/genética , Antígenos/imunologia , Células CHO , Moléculas de Adesão Celular/genética , Cricetinae , Cricetulus , Endossomos/genética , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Imunidade Celular/fisiologia , Lectinas Tipo C/genética , Lisossomos/genética , Camundongos , Camundongos Transgênicos , Estrutura Terciária de Proteína , Receptores de Superfície Celular/genética , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/imunologia
14.
Biomaterials ; 32(28): 6791-803, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21724247

RESUMO

Targeted delivery of nanoparticles (NPs) carrying vaccine components to dendritic cells (DCs) is a promising strategy to initiate antigen-specific immune responses. Improving the interactions between nanoparticle-carried ligands and receptors on DCs is a major challenge. These NPs are generally coated with poly(ethylene glycol) (PEG), to shield non-specific interactions, and antibodies, to facilitate specific delivery to DC surface receptors. We have devised a strategy to covalently link PEG molecules of various chain length (Mw 2000-20000 g/moL) to poly(lactic-co-)glycolic acid (PLGA) NP vaccines. We coated these NPs with various antibodies recognizing the DC-specific receptor DC-SIGN to study the effects of shielding and antibody type on antibody--receptor interactions. Chemical attachment of PEG to the particle surface was followed by detailed zeta potential, DLS and NMR studies, and analyzed by analytical chemistry. Increasing the PEG chain length increased particle size and polydispersity index and reduced the intracellular degradation rate of encapsulated antigens. Binding and uptake of NPs by human DCs was affected by both PEG chain length and antibody type. NPs coated with PEG-3000 had the optimal chain length for antibody--receptor interactions and induction of antigen-specific T-cell responses. Interestingly, clear differences were observed upon targeting distinct epitopes of the same receptor. Binding and uptake of NPs carrying antibodies recognizing the carbohydrate recognition domain of DC-SIGN was enhanced when compared to those carrying antibodies recognizing the receptor's neck region. In conclusion, our data show that PEG chains cannot be extended beyond a certain length for shielding purposes without compromising the efficacy of targeted delivery. Thereby, the implications of our findings are not limited to the future design of nanovaccines specifically targeted to DC-SIGN, but apply to the general design of targeted nanocarriers.


Assuntos
Anticorpos/metabolismo , Células Dendríticas/imunologia , Portadores de Fármacos/química , Nanopartículas/química , Polietilenoglicóis/química , Vacinas/química , Vacinas/imunologia , Anticorpos/química , Moléculas de Adesão Celular/imunologia , Células Dendríticas/citologia , Portadores de Fármacos/metabolismo , Humanos , Lectinas Tipo C/imunologia , Teste de Materiais , Estrutura Molecular , Nanopartículas/ultraestrutura , Tamanho da Partícula , Receptores de Superfície Celular/imunologia , Propriedades de Superfície , Linfócitos T/imunologia , Linfócitos T/fisiologia
15.
Mol Pharm ; 8(2): 520-31, 2011 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-21381651

RESUMO

Dendritic cells (DCs) are key players in the initiation of adaptive immune responses and are currently exploited in immunotherapy against cancer and infectious diseases. The targeted delivery of nanovaccine particles (NPs) to DCs in vivo is a promising strategy to enhance immune responses. Here, targeted nanovaccine carriers were generated that allow multimodal imaging of nanocarrier-DC interactions from the subcellular to the organism level. These carriers were made of biodegradable poly(D,L-lactide-co-glycolide) harboring superparamagnetic iron oxide particles (SPIO) and fluorescently labeled antigen in a single particle. Targeted delivery was facilitated by coating the NPs with antibodies recognizing the DC-specific receptor DC-SIGN. The fluorescent label allowed for rapid analysis and quantification of specific versus nonspecific uptake of targeted NPs by DCs compared to other blood cells. In addition, it showed that part of the encapsulated antigen reached the lysosomal compartment of DCs within 24 h. Moreover, the presence of fluorescent label did not prevent the antigen from being presented to antigen-specific T cells. The incorporated SPIO was applied to track the NPs at subcellular cell organel level using transmission electron microscopy (TEM). NPs were found within endolysosomal compartments, where part of the SPIO was already released within 24 h. Furthermore, part of the NPs seemed to localize within the cytoplasm. Ex vivo loading of DCs with NPs resulted in efficient labeling and detection by MRI and did not abolish cell migration within collagen scaffolds. In conclusion, incorporation of two imaging agents within a single carrier allows tracking of targeted nanovaccines on a subcellular, cellular and possibly organism level, thereby facilitating rational design of in vivo targeted vaccination strategies.


Assuntos
Moléculas de Adesão Celular/imunologia , Células Dendríticas/imunologia , Portadores de Fármacos , Lectinas Tipo C/imunologia , Imageamento por Ressonância Magnética , Nanopartículas de Magnetita/administração & dosagem , Receptores de Superfície Celular/imunologia , Vacinas de Subunidades Antigênicas/imunologia , Apresentação de Antígeno , Células Sanguíneas/imunologia , Moléculas de Adesão Celular/metabolismo , Células Dendríticas/metabolismo , Compostos Férricos/química , Citometria de Fluxo , Humanos , Ácido Láctico/química , Lectinas Tipo C/metabolismo , Nanopartículas de Magnetita/química , Nanopartículas de Magnetita/ultraestrutura , Nanotecnologia , Fragmentos de Peptídeos/imunologia , Ácido Poliglicólico/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Receptores de Superfície Celular/metabolismo , Linfócitos T/imunologia
16.
Eur J Immunol ; 41(4): 1014-23, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21413003

RESUMO

DEC-205 is a type I C-type lectin receptor (CLR) that is expressed on various APC subsets and has been suggested to bind necrotic and apoptotic cells. Here we study DEC-205 characteristics in plasmacytoid DCs (pDCs) obtained from healthy individuals and assess its ability to mediate antigen presentation by isolating sufficient numbers of pDCs from apheresis material obtained from stage III/IV melanoma patients. The results demonstrate that DEC-205 is expressed on human pDCs. Internalization of DEC-205 after antibody ligation is clathrin- and dynamin-dependent as it is blocked by hypertonic shock or by inhibition of dynamin activity. Antibody targeting to DEC-205 does not affect TLR-induced expression levels of co-stimulatory and MHC molecules, but clearly impairs TLR-induced IFN-α secretion by 40%. We observed that TLR-mediated signaling increases DEC-205 expression levels without affecting receptor internalization. Moreover, human pDCs retained the capacity to present antigens via DEC-205 following TLR activation.


Assuntos
Apresentação de Antígeno , Antígenos CD/imunologia , Células Dendríticas/imunologia , Lectinas Tipo C/imunologia , Receptores de Superfície Celular/imunologia , Diferenciação Celular , Proliferação de Células , Separação Celular , Células Cultivadas , Células Dendríticas/citologia , Humanos , Interferon-alfa/biossíntese , Interferon-alfa/imunologia , Antígenos de Histocompatibilidade Menor , Receptores Toll-Like/imunologia
17.
Semin Immunol ; 23(1): 12-20, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21269839

RESUMO

During the past decade, the immunotherapeutic potential of ex vivo generated professional antigen presenting dendritic cells (DCs) has been explored in the clinic. Albeit safe, clinical results have thus far been limited. A major disadvantage of current cell-based dendritic cell (DC) therapies, preventing universal implementation of this form of immunotherapy, is the requirement that vaccines need to be tailor made for each individual. Targeted delivery of antigens to DC surface receptors in vivo would circumvent this laborious and expensive ex vivo culturing steps involved with these cell-based therapies. In addition, the opportunity to target natural and often rare DC subsets in vivo might have advantages over loading more artificial ex vivo cultured DCs. Preclinical studies show targeting antigens to DCs effectively induces humoral responses, while cellular responses are induced provided a DC maturation or activation stimulus is co-administered. Here, we discuss strategies to target antigens to distinct DC subsets and to simultaneously employ adjuvants to activate these cells to induce immunity.


Assuntos
Antígenos/imunologia , Células Dendríticas/imunologia , Vacinas/economia , Vacinas/imunologia , Adjuvantes Imunológicos , Animais , Apresentação de Antígeno/imunologia , Humanos , Receptores de Superfície Celular/imunologia , Receptores de Superfície Celular/metabolismo
18.
J Immunol ; 184(8): 4276-83, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20304825

RESUMO

Plasmacytoid dendritic cells (pDCs) play a major role in shaping both innate and adaptive immune responses, mainly via their production of large amounts of type I IFNs. pDCs are considered to primarily present endogenous Ags and are thought not to participate in the uptake and presentation of Ags from the extracellular environment, in contrast to their myeloid counterparts, which efficiently endocytose extracellular particulates. In this study, we show that human pDCs are able to phagocytose and process particulate forms of Ag entrapped in poly(lactic-coglycolic acid) microparticles. Furthermore, pDCs were also able to sense TLR ligands (TLR-Ls) incorporated in these particles, resulting in rapid pDC activation and high IFN-alpha secretion. Combining a tetanus toxoid peptide and TLR-Ls (CpG C and R848) in these microparticles resulted in efficient pDC activation and concomitant Ag-specific T cell stimulation. Moreover, particulate Ag was phagocytosed and presented more efficiently than soluble Ag, indicating that microparticles can be exploited to facilitate efficient delivery of antigenic cargo and immunostimulatory molecules to pDCs. Together, our results show that in addition to their potency to stimulate innate immunity, pDCs can polarize adaptive immune responses against exogenous particulate Ag. These results may have important consequences for the development of new immunotherapeutic strategies exploiting Ag and TLR-Ls encapsulated in microparticles to target APC subsets.


Assuntos
Apresentação de Antígeno/imunologia , Micropartículas Derivadas de Células/imunologia , Células Dendríticas/imunologia , Glicolatos/imunologia , Fagocitose/imunologia , Soroalbumina Bovina/imunologia , Animais , Cápsulas , Bovinos , Diferenciação Celular/imunologia , Micropartículas Derivadas de Células/metabolismo , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Epitopos de Linfócito T/imunologia , Glicolatos/metabolismo , Humanos , Interferon-alfa/biossíntese , Ácido Láctico , Ligantes , Ativação Linfocitária/imunologia , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Soroalbumina Bovina/metabolismo , Toxoide Tetânico/imunologia , Toxoide Tetânico/metabolismo , Receptores Toll-Like/metabolismo
19.
J Control Release ; 144(2): 118-26, 2010 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-20156497

RESUMO

Vaccine efficacy is strongly enhanced by antibody-mediated targeting of vaccine components to dendritic cells (DCs), which are professional antigen presenting cells. However, the options to link antigens or immune modulators to a single antibody are limited. Here, we engineered versatile nano- and micrometer-sized slow-release vaccine delivery vehicles that specifically target human DCs to overcome this limitation. The nano- (NPs) and microparticles (MPs), with diameters of approximately 200nm and 2microm, consist of a PLGA core coated with a polyethylene glycol-lipid layer carrying the humanized targeting antibody hD1, which does not interact with complement or Fc receptors and recognizes the human C-type lectin receptor DC-SIGN on DCs. We studied how these particles interact with human DCs and blood cells, as well as the kinetics of PLGA-encapsulated antigen degradation within DCs. Encapsulation of antigen resulted in almost 38% degradation for both NPs and MPs 6days after particle ingestion by DCs, compared to 94% when nonencapsulated, soluble antigen was used. In contrast to the MPs, which were taken up rather nonspecifically, the NPs effectively targeted human DCs. Consequently, targeted delivery only improved antigen presentation of NPs and induced antigen-dependent T cell responses at 10-100 fold lower concentrations than nontargeted NPs.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Células Dendríticas/imunologia , Sistemas de Liberação de Medicamentos , Lectinas Tipo C/imunologia , Anticorpos/imunologia , Anticorpos/metabolismo , Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos/imunologia , Antígenos/metabolismo , Moléculas de Adesão Celular , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Humanos , Ácido Láctico , Lectinas Tipo C/metabolismo , Polietilenoglicóis/metabolismo , Ácido Poliglicólico , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Receptores de Superfície Celular , Vacinas/imunologia , Vacinas/metabolismo
20.
J Immunol ; 181(8): 5219-24, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18832675

RESUMO

Human plasmacytoid dendritic cells (pDCs)(2) exploit Ag uptake receptors like CD32a for internalization of exogenous Ags. Activation of pDC by TLR9 ligand CpG-C induces strong maturation. Surprisingly, we observed that CpG-C-stimulated pDCs showed impaired Ag-specific T cell proliferation whereas the induction of allogeneic T cell proliferation was not affected. We demonstrated that signals from TLR9 caused a rapid down-regulation of the capacity of pDC to take-up Ab-Ag complexes without altering their CD32a expression, thus explaining the reduced Ag presentation. The recent contrasting biological responses that were observed upon TLR9 ligation in pDCs prompted us to study the effect of several TLR9 ligands. We observed that type I IFN-inducer CpG-A, localizing in the early endosomal compartment, did not affect CD32a function, whereas CpGs localizing in the late endosomes and inducing pDC maturation clearly inhibited CD32a-mediated Ag uptake and presentation. We conclude that TLR9 ligands not only determine the type of response, i.e., type I IFN production (innate immunity) or maturation (adaptive immunity), but also directly affect Ag presentation capacity of pDCs. We hypothesize that pDC, once activated via TLR9-ligands reaching the late endosomes, can only present initially sampled Ags and thus are protected from uptake and processing of additional potential self-Ags.


Assuntos
Apresentação de Antígeno/imunologia , Complexo Antígeno-Anticorpo/imunologia , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Plasmócitos/imunologia , Receptores de IgG/imunologia , Receptor Toll-Like 9/imunologia , Apresentação de Antígeno/efeitos dos fármacos , Complexo Antígeno-Anticorpo/metabolismo , Antígenos/imunologia , Antígenos/metabolismo , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Endossomos/imunologia , Endossomos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/fisiologia , Interferon Tipo I/imunologia , Interferon Tipo I/metabolismo , Ligantes , Oligodesoxirribonucleotídeos/farmacologia , Plasmócitos/citologia , Plasmócitos/metabolismo , Receptores de IgG/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo , Receptor Toll-Like 9/agonistas , Receptor Toll-Like 9/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...