Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
AIDS ; 32(17): 2533-2545, 2018 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-30289805

RESUMEN

OBJECTIVE: The efficacy of therapeutic vaccines against HIV-1 infection has been modest. New inerts to redirect responses to vulnerable sites are urgently needed to improve these results. DESIGN: We performed the first-in-human clinical trial with naked mRNA (iHIVARNA) combining a dendritic cell activation strategy (TriMix:CD40L+CD70+caTLR4 RNA) with a novel HIV immunogen sequences (HTI immunogen). METHODS: A dose escalation, phase I clinical trial was performed in 21 chronic HIV-1-infected patients under ART who received three intranodal doses of mRNA (weeks 0, 2 and 4) as follow: TriMix-100 g, TriMix-300 g, TriMix-300 g with HTI-300 g, TriMix-300 g with HTI-600 g, TriMix-300 g with HTI-900 g. Primary end-point was safety and secondary-exploratory end-points were immunogenicity, changes in viral reservoir and transcriptome. RESULTS: Overall, the vaccine was secure and well tolerated. There were 31 grade 1/2 and 1 grade 3 adverse events, mostly unrelated to the vaccination. Patients who received the highest dose showed a moderate increase in T-cell responses spanning HTI sequence at week 8. In addition, the proportion of responders receiving any dose of HTI increased from 31% at w0 to 80% postvaccination. The intervention had no impact on caHIV-DNA levels, however, caHIV-RNA expression and usVL were transiently increased at weeks 5 and 6 in the highest dose of iHIVARNA, and these changes were positively correlated with HIV-1-specific-induced immune responses. CONCLUSION: This phase I dose-escalating trial showed that iHIVARNA administration was safe and well tolerated, induced moderate HIV-specific T-cell responses and transiently increased different viral replication readouts. These data support further exploration of iHIVARNA in a phase II study. CLINICALTRIALS. GOV IDENTIFIER: NCT02413645.


Asunto(s)
Vacunas contra el SIDA/administración & dosificación , Células Dendríticas/inmunología , Infecciones por VIH/terapia , ARN Mensajero/administración & dosificación , Adulto , Antirretrovirales/administración & dosificación , Terapia Combinada/métodos , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Resultado del Tratamiento
2.
J Leukoc Biol ; 82(1): 93-105, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17449724

RESUMEN

CD4(+)CD25(+) regulatory T cells (Treg) have been described as an important hurdle for immunotherapy. Engagement of glucocorticoid-induced TNF receptor-related protein (GITR) has emerged recently as an important mechanism to control the suppression of CD4(+)CD25(+) Treg. Furthermore, it has been documented extensively that GITR ligation is costimulatory for naive and activated T cells in the murine setting. However, little is known about the role of the human GITR ligand (huGITRL). We wanted to explore whether huGITRL could enhance antigen-specific T cell priming by dendritic cells (DC). First, we confirmed the endogenous expression of GITRL on HUVEC. We also detected GITRL expression on EBV-B cell lines, whereas no GITRL expression was observed on human monocyte-derived DC. Electroporation of GITRL mRNA in monocyte-derived DC resulted in a strong and long-lasting surface expression of GITRL. In contrast to data obtained in mice, no significant abrogation of Treg suppression by GITRL-expressing human DC was observed. Consistent with our mouse data, we showed that huGITRL is costimulatory for responder T cells. Furthermore, we found that GITRL-expressing DC primed increased numbers of Melan-A-specific CD8(+) T cells. We conclude that although huGITRL is not capable of alleviating Treg suppression of responder T cells, huGITRL overexpression on monocyte-derived DC enhances their capacity to induce antigen-specific T cell responses. Thus, GITRL incorporation in DC might improve the antitumor immune response after vaccination.


Asunto(s)
Células Dendríticas/inmunología , Linfocitos T Reguladores/inmunología , Factores de Necrosis Tumoral/inmunología , Presentación de Antígeno , Células Cultivadas , Células Dendríticas/química , Electroporación , Endotelio Vascular/citología , Humanos , Inmunidad , Células Mieloides , Especificidad del Receptor de Antígeno de Linfocitos T , Factores de Necrosis Tumoral/genética , Cordón Umbilical/citología
3.
Mol Ther ; 12(5): 922-32, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16257383

RESUMEN

We previously described mRNA electroporation as an efficient gene delivery method to introduce tumor-antigens (Ag) into murine immature dendritic cells (DC). Here, we further optimize the protocol and evaluate the capacity of mRNA-electroporated DC as a vaccine for immunotherapy. First, the early DC maturation kinetics and the effect of different lipopolysaccharide incubation periods on the phenotypic maturation profile of DC are determined. Next, we show that either immature or mature DC are equally well electroporated and express and present the transgene at a comparable level after electroporation. We point out that the mRNA electroporation results in a negative effect on the interleukin (IL)-12p70, IL-6, and tumor necrosis factor-alpha secretion after maturation. Nevertheless, mRNA-electroporated DC induce an effective cytotoxic T lymphocyte (CTL) response in vivo. Mature electroporated DC are significantly more potent in eliciting an Ag-specific CD8+ CTL response compared to their immature electroporated counterparts. In addition, a significant improvement in CTL response is obtained both in the primary and in the memory effector phases when CD4+CD25+ regulatory T cells (Treg) are depleted in vivo prior to immunization. These findings are further substantiated in tumor protection experiments and hold convincing evidence for the merit of Treg cell depletion prior to immunization with mRNA-electroporated DC.


Asunto(s)
Presentación de Antígeno , Células Dendríticas/inmunología , Electroporación , Técnicas de Transferencia de Gen , ARN Mensajero/genética , Linfocitos T/inmunología , Animales , Animales Modificados Genéticamente , Antígenos de Neoplasias , Antígeno CD24 , Células Cultivadas , Citocinas/metabolismo , Femenino , Inmunoterapia , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Neoplasias/terapia , Receptores de Interleucina-2 , Transducción Genética , Vacunas de ADN
4.
Mol Ther ; 10(4): 768-79, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15451461

RESUMEN

The use of tumor antigen-loaded dendritic cells (DC) is one of the most promising approaches to inducing a tumor-specific immune response. We compared electroporation of mRNA to lentiviral transduction for the delivery of tumor antigens to human monocyte-derived and murine bone marrow-derived DC. Both lentiviral transduction and mRNA electroporation induced eGFP expression in on average 81% of human DC. For murine DC, eGFP mRNA electroporation (62%) proved to be more efficient than lentiviral transduction (47%). When we used tNGFR as a transgene we observed lentiviral pseudotransduction that overestimated lentiviral efficiency. Neither gene transfer method had an adverse effect on viability, phenotype, or allostimulatory capacity of either human or murine DC. Yet, the mRNA-electroporated DC showed a reduced production of IL-12p70 compared to their lentivirally transduced and unmodified counterparts. Human Ii80MAGE-A3-modified DC and murine Ii80tOVA-modified DC were able to present antigenic epitopes in the context of MHC class I and class II. Both types of modified murine DC were able to induce OVA-specific cytotoxic T cells in vivo; however, the mRNA-electroporated DC were less potent. Our data indicate that this may be related to their impaired IL-12 production.


Asunto(s)
Vacunas contra el Cáncer/genética , Células Dendríticas/inmunología , Electroporación , Lentivirus/genética , ARN Mensajero/genética , Transducción Genética/métodos , Animales , Presentación de Antígeno/inmunología , Antígenos de Neoplasias/análisis , Antígenos de Neoplasias/genética , Línea Celular , Trasplante de Células , Citocinas/biosíntesis , Femenino , Proteínas Fluorescentes Verdes/análisis , Proteínas Fluorescentes Verdes/genética , Antígenos de Histocompatibilidad Clase I/análisis , Antígenos de Histocompatibilidad Clase II/análisis , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas de Neoplasias/análisis , Proteínas de Neoplasias/genética , Neoplasias/terapia , Receptor de Factor de Crecimiento Nervioso/análisis , Receptor de Factor de Crecimiento Nervioso/genética
5.
J Leukoc Biol ; 76(5): 1028-38, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15316032

RESUMEN

It has been postulated that low-level interleukin (IL)-12 production of antigen-presenting cells is associated with the risk of developing atopic asthma. To study the relationship between IL-12 production capacity of dendritic cells (DCs) and development of T helper type 2 (Th2) responses in the lung, we genetically engineered DCs to constutively overexpress bioactive IL-12. Retrovirally mediated overexpression of IL-12 in DCs strongly polarized naive ovalbumin (OVA)-specific CD4+ T cells toward Th1 effector cells in vitro. After intratracheal injection, OVA-pulsed IL-12-overexpressing DCs failed to induce Th2 responses in vivo and no longer primed mice for Th2-dependent eosinophilic airway inflammation upon OVA aerosol challenge, readily observed in mice immunized with sham-transfected, OVA-pulsed DCs. Analysis of a panel of cytokines and chemokines in the lung demonstrated that the lack of Th2 sensitization was accompanied by increased production of the Th1 cytokine interferon-gamma (IFN-gamma), chemokines induced by IFN-gamma, and the immunoregulatory cytokine IL-10. When Th2 priming was induced using OVA/alum prior to intratracheal DC administration, DCs constitutively expressing IL-12 were no longer capable of preventing eosinophilic airway inflammation and even enhanced it. These data show directly that high-level expression of IL-12 in DCs prevents the development of Th2 sensitization. Enhancing IL-12 production in DCs should be seen as a primary prevention strategy for atopic disorders. Enhancing IL-12 production in DCs is less likely to be of benefit in already Th2-sensitized individuals.


Asunto(s)
Células Dendríticas/inmunología , Inmunización , Interleucina-12/inmunología , Pulmón/inmunología , Células TH1/inmunología , Células Th2/inmunología , Animales , Antígenos/inmunología , Antígenos/farmacología , Asma/inmunología , Asma/fisiopatología , Pruebas de Provocación Bronquial , Células Cultivadas , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Eosinófilos/efectos de los fármacos , Eosinófilos/inmunología , Femenino , Vectores Genéticos/genética , Interferón gamma/inmunología , Interleucina-10/inmunología , Interleucina-12/biosíntesis , Interleucina-12/genética , Pulmón/fisiopatología , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/inmunología , Ovalbúmina/farmacología , Retroviridae/genética , Células TH1/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética
6.
J Gene Med ; 5(8): 654-67, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12898635

RESUMEN

BACKGROUND: Dendritic cells (DC) are the professional antigen-presenting cells of the immune system, fully equipped to prime naive T cells and thus essential components for cancer immunotherapy. METHODS: We tested the influence of several elements (cPPT, trip, WPRE, SIN) on the transduction efficiency of human DC. Human and murine DC were transduced with tNGFR-encoding lentiviruses to assess the effect of transduction on phenotype and function. Human DC were transduced with lentiviruses encoding huIi80MAGE-A3 and murine DC with huIi80tOVA to test antigen presentation. RESULTS: A self-inactivating (SIN) lentiviral vector containing the trip element was most efficient in transducing human DC. The transduction of DC with trip/SIN tNGFR encoding lentiviral vectors at MOI 15 resulted in stable gene expression in up to 94.6% (murine) and 88.2% (human) of the mature DC, without perturbing viability, phenotype and function. Human huIi80MAGE-A3-transduced DC were able to stimulate MAGE-A3-specific CD4(+) and CD8(+) T cell clones and could prime both MAGE-A3-specific CD4(+) and CD8(+) T cells in vitro. Murine huIi80tOVA-transduced DC were able to present OVA peptides in the context of MHC class I and class II in vitro and induced a strong OVA-specific cytotoxic T lymphocyte response in vivo, that was protective against subsequent challenge with OVA-expressing tumor cells. CONCLUSIONS: We show that, using lentiviral vectors, efficient gene transfer in human and murine DC can be obtained and that these DC can elicit antigen-specific immune responses in vitro and in vivo. The composition of the transfer vector has a major impact on the transduction efficiency.


Asunto(s)
Células Dendríticas/fisiología , Técnicas de Transferencia de Gen , Lentivirus/genética , Proteínas de Neoplasias , Neoplasias/terapia , Transducción Genética , Animales , Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Antígenos de Neoplasias/metabolismo , Línea Celular , Células Dendríticas/virología , Femenino , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Humanos , Interleucina-12/metabolismo , Lentivirus/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias/genética , Neoplasias/inmunología , Tasa de Supervivencia , Linfocitos T/inmunología , Linfocitos T/metabolismo
7.
Blood ; 101(4): 1469-76, 2003 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-12393481

RESUMEN

Dendritic cells (DCs) genetically engineered to overexpress CD95 (Fas) ligand (CD95L-DC) were proposed as tools to induce peripheral tolerance to alloantigens. Herein, we observed that CD95L-DC obtained after retroviral gene transfer in bone marrow (BM) precursors derived from CD95-deficient (lpr/lpr) mice elicit much stronger allospecific type 1 helper T-cell and cytotoxic T-cell activities than control DCs upon injection in vivo, although they induce lower T-cell responses in vitro. Indeed, a single injection of CD95L-DC prepared from C57BL/6 mice was sufficient to prime bm13 recipients for acute rejection of C57BL/6 skin allografts that were otherwise tolerated in the context of this single weak major histocompatibility complex (MHC) class I incompatibility. Massive neutrophil infiltrates depending on interleukin (IL)-1 signaling were observed at sites of CD95L-DC injection. Experiments in IL-1 receptor-deficient mice or in animals injected with depleting anti-Gr1 monoclonal antibody (mAb) established that neutrophil recruitment is required for the development of vigorous T-cell responses after injection of CD95L-DC in vivo.


Asunto(s)
Células Dendríticas/inmunología , Linfocitos T/inmunología , Receptor fas/genética , Animales , Anticuerpos Monoclonales/farmacología , Rechazo de Injerto/inmunología , Tolerancia Inmunológica , Interferón gamma/biosíntesis , Interleucina-2/biosíntesis , Interleucina-4/biosíntesis , Interleucina-5/biosíntesis , Isoantígenos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Receptores de Interleucina-1/deficiencia , Retroviridae/genética , Trasplante de Piel , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Transfección , Receptor fas/inmunología , Receptor fas/fisiología
8.
Cancer Gene Ther ; 9(9): 787-97, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12189529

RESUMEN

Recently, human dendritic cells (DCs) pulsed with mRNA encoding a broad range of tumor antigens have proven to be potent activators of a primary anti-tumor-specific T-cell response in vitro. The aim of this study was to improve the mRNA pulsing of murine DC. Compared to a standard lipofection protocol and passive pulsing, electroporation was, in our hands, the most efficient method. The optimal conditions to electroporate murine bone marrow-derived DCs with mRNA were determined using enhanced green fluorescent protein and a truncated form of the nerve growth factor receptor. We could obtain high transfection efficiencies around 70-80% with a mean fluorescence intensity of 100-200. A maximal expression level was reached 3 hours after electroporation. A clear dose-response effect was seen depending on the amount of mRNA used. Importantly, the electroporation process did not affect the viability nor the allostimulatory capacity or phenotype of the DC. To study the capacity of mRNA-electroporated DCs to present antigen in the context of MHC classes I and II, we made use of chimeric constructs of ovalbumin. The dose-dependent response effect and the duration of presentation were also determined. Together, these results demonstrate that mRNA electroporation is a useful method to generate genetically modified murine DC, which can be used for preclinical studies testing immunotherapeutic approaches.


Asunto(s)
Presentación de Antígeno/inmunología , Células Dendríticas/inmunología , Electroporación/métodos , Técnicas de Transferencia de Gen , Linfoma/terapia , ARN Mensajero/genética , Animales , División Celular , Pollos , Cartilla de ADN/química , Femenino , Citometría de Flujo , Genes MHC Clase I/genética , Genes MHC Clase II/genética , Proteínas Fluorescentes Verdes , Humanos , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Activación de Linfocitos , Linfoma/inmunología , Linfoma/metabolismo , Linfoma/patología , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/metabolismo , Fragmentos de Péptidos/inmunología , Reacción en Cadena de la Polimerasa , Receptor de Factor de Crecimiento Nervioso/genética , Receptor de Factor de Crecimiento Nervioso/metabolismo , Linfocitos T/inmunología , Transcripción Genética , Ubiquitina/farmacología
9.
Transplantation ; 73(1 Suppl): S27-30, 2002 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11810058

RESUMEN

Dendritic cells (DC) are the most potent presenters of alloantigens and therefore are responsible for the induction of allograft rejection. Genetic modifications of DC allowing the expression of a tolerogenic molecule may render them immunosuppressive. We transduced bone marrow-derived DC with recombinant MFG retrovirus encoding either viral interleukin (vIL)-10 or Fas ligand (FasL) to induce transplantation tolerance. Up to 10 ng/ml of bioactive vIL-10 was produced by DC after transfer of the corresponding gene. Although the inhibitory properties of vIL-10-transduced DC were revealed in vitro in a mixed lymphocyte culture, no clear down-regulation of the allogeneic response was observed in vivo after single or multiple injections of those DC overexpressing vIL-10. When we transduced wild-type bone marrow-derived DC with recombinant MFG retrovirus encoding murine FasL, cells quickly died, probably because of suicidal or fratricidal Fas-dependent death. Indeed, only DC from Fas-deficient lpr mice survived to FasL gene transfer. Those FasL-transduced lpr DC exhibited a strong cytotoxic activity against Fas-positive targets in vitro. DC overexpressing FasL did not behave as immunosuppressive DC in vivo. The subcutaneous injection of FasL+ lpr DC in MHC class II-disparate mice hyperactivated the allospecific proliferation of T cells in the draining lymph nodes compared with mice treated with control-transduced DC. These results argue against the development of FasL+ DC or vIL-10-secreting DC as immunosuppressive tools in vivo. The alternative pathways of T-cell activation triggered by these genetically modified DC need to be investigated.


Asunto(s)
Células Dendríticas/fisiología , Interleucina-10/genética , Glicoproteínas de Membrana/genética , Retroviridae/genética , Transducción Genética , Tolerancia al Trasplante , Animales , Células Dendríticas/inmunología , Proteína Ligando Fas , Humanos
10.
Cancer Immun ; 2: 2, 2002 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-12747747

RESUMEN

Recent studies have shown the importance of triggering CD40 molecules to enhance the efficiency of dendritic cells (DCs) as antigen-presenting cells (APCs). The P198 and P1A tumor antigens, which are expressed by mastocytoma P815, have been assessed for their immunogenicity using different modes of immunization. We measured CTL responses induced in vivo with antigenic peptides P198 and P1A loaded onto bone marrow-derived DCs that had matured as a consequence of CD40-CD40L interactions. CD40L-transfected 3T3 fibroblasts were used as a source of CD40L signal. Our results show that this mode of DC activation considerably improves their ability to induce CTLs against P198 and P1A antigens in vivo as compared to untreated DCs. We also show that immunizations carried out with CD40L-activated DCs loaded with the P1A peptide induce a very efficient protection against a lethal challenge with P815 tumor cells, which express P1A. Our results indicate that the efficiency of DC-based vaccines used in clinical trials of cancer immunotherapy could be increased significantly by triggering DCs via CD40 prior to immunization.


Asunto(s)
Antígenos CD40/inmunología , Células Dendríticas/inmunología , Vacunación/métodos , Células 3T3 , Animales , Antígenos de Neoplasias/inmunología , Ligando de CD40/genética , Ligando de CD40/inmunología , Técnicas de Cocultivo , Pruebas Inmunológicas de Citotoxicidad , Citotoxicidad Inmunológica/inmunología , Células Dendríticas/citología , Células Dendríticas/trasplante , Masculino , Ratones , Ratones Endogámicos DBA , Bazo/citología , Bazo/inmunología , Linfocitos T Citotóxicos/inmunología , Transfección , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA