Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Cancer Immunol Immunother ; 64(3): 381-8, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25548092

RESUMEN

PURPOSE: Melanoma patients with a high risk of recurrence may benefit from immunotherapy with mRNA-electroporated autologous monocyte-derived dendritic cells (DCs). Further benefit may be found in combining DC-therapy with interferon alfa-2b. PATIENTS AND METHODS: The long-term clinical outcome of AJCC stage III/IV melanoma patients who had no evidence of disease at the time of treatment with autologous mRNA-electroporated DCs in a single-center pilot clinical trial was analyzed. Antigen loading was accomplished by co-electroporation of mRNA encoding a fusion protein between MAGE-A1, -A3, -C2, Tyrosinase, MelanA/MART-1, or gp100, and an HLA class II-targeting sequence. DCs were administered by 4-6 bi-weekly intradermal injections. IFN-α-2b (5 MIU TIW) was initiated either at recurrence (cohort 1), concomitant with DCs (cohorts 2 and 3), or following the fourth DC administration (cohort 4). RESULTS: Thirty melanoma patients were recruited between April 2006 and June 2009. DC-related adverse events included grade 2 local injection site reactions in all patients, grade 2 fever and flu-like symptoms in one patient, and skin depigmentation in seven patients. After a median follow-up of over 6 years, the median relapse-free survival is 22 months (95% CI 12-32 months). Twelve patients have died. The median overall survival has not been reached; the 2-year and 4-year survival rates are 93 and 70%, respectively. CONCLUSIONS: Adjuvant therapy following the resection of melanoma metastases with autologous mRNA-electroporated DCs, combined with interferon alfa-2b, is tolerable and results in encouraging long-term overall survival rates justifying further evaluation in a randomized clinical trial.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Células Dendríticas/inmunología , Inmunoterapia Adoptiva/métodos , Melanoma/terapia , ARN Mensajero/administración & dosificación , Neoplasias Cutáneas/terapia , Adulto , Anciano , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Electroporación , Femenino , Humanos , Interferón alfa-2 , Interferón-alfa/administración & dosificación , Interferón-alfa/inmunología , Antígeno MART-1/genética , Masculino , Melanoma/inmunología , Melanoma/cirugía , Antígenos Específicos del Melanoma/genética , Persona de Mediana Edad , Monofenol Monooxigenasa/genética , Metástasis de la Neoplasia , Proyectos Piloto , ARN Mensajero/genética , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/cirugía , Resultado del Tratamiento , Melanoma Cutáneo Maligno
2.
J Immunol ; 191(4): 1976-83, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23842750

RESUMEN

Regulatory T cells (Tregs) counteract anticancer immune responses through a number of mechanisms, limiting dendritic cell (DC)-based anticancer immunotherapy. In this study, we investigated the influence of various DC activation stimuli on the Treg functionality. We compared DCs activated by electroporation with mRNA encoding constitutively active TLR4 (caTLR4) and CD40 ligand (DiMix-DCs), or these factors together with mRNA encoding the costimulatory molecule CD70 (TriMix-DCs) with DCs maturated in the presence of a mixture of inflammatory cytokines (DCs maturated with a combination of the cytokines IL-1ß, IL-6, TNF-α, and PGE2) for their ability to counteract Tregs on different levels. We first demonstrated that there was no difference in the extent of Treg induction starting from CD4(+)CD25(-) T cells under the influence of the different DC maturation stimuli. Second, we showed that both DiMix- and TriMix-DCs could partly alleviate Treg inhibition of CD8(+) T cells. Third, we observed that CD8(+) T cells that had been precultured with DiMix-DCs or TriMix-DCs were partially protected against subsequent Treg suppression. Finally, we showed that Tregs cocultured in the presence of TriMix-DCs, but not DiMix-DCs, partially lost their suppressive capacity. This was accompanied by a decrease in CD27 and CD25 expression on Tregs, as well as an increase in the expression of T-bet and secretion of IFN-γ, TNF-α, and IL-10, suggesting a shift of the Treg phenotype toward a Th1 phenotype. In conclusion, these data suggest that TriMix-DCs are not only able to suppress Treg functions, but moreover could be able to reprogram Tregs to Th1 cells under certain circumstances.


Asunto(s)
Ligando CD27/fisiología , Ligando de CD40/fisiología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Tolerancia Inmunológica/inmunología , Linfopoyesis/fisiología , Linfocitos T Reguladores/inmunología , Receptor Toll-Like 4/fisiología , Ligando CD27/genética , Linfocitos T CD4-Positivos/citología , Ligando de CD40/genética , Diferenciación Celular/efectos de los fármacos , División Celular , Células Cultivadas , Técnicas de Cocultivo , Citocinas/farmacología , Células Dendríticas/metabolismo , Electroporación , Humanos , Inmunofenotipificación , Activación de Linfocitos , MAP Quinasa Quinasa 1/genética , MAP Quinasa Quinasa 1/fisiología , Monocitos/citología , Monocitos/efectos de los fármacos , ARN Mensajero/administración & dosificación , ARN Mensajero/genética , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T Reguladores/citología , Células TH1/inmunología , Receptor Toll-Like 4/genética
3.
Cancer Immunol Immunother ; 63(9): 959-67, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24878889

RESUMEN

Since decades, the main goal of tumor immunologists has been to increase the capacity of the immune system to mediate tumor regression. In this regard, one of the major focuses of cancer immunotherapy has been the design of vaccines promoting strong tumor-specific cytotoxic T lymphocyte responses in cancer patients. Here, dendritic cells (DCs) play a pivotal role as they are regarded as nature's adjuvant and as such have become the natural agents for antigen delivery in order to finally elicit strong T cell responses (Villadangos and Schnorrer in Nat Rev Immunol 7:543-555, 2007; Melief in Immunity 29:372-383, 2008; Palucka and Banchereau in Nat Rev Cancer 12:265-277, 2012; Vacchelli et al. in Oncoimmunology 2:e25771, 2013; Galluzzi et al. in Oncoimmunology 1:1111-1134, 2012). Therefore, many investigators are actively pursuing the use of DCs as an efficient way of inducing anticancer immune responses. Nowadays, DCs can be generated at a large scale in closed systems, yielding sufficient numbers of cells for clinical application. In addition, with the identification of tumor-associated antigens, which are either selectively or preferentially expressed by tumors, a whole range of strategies using DCs for immunotherapy have been designed and tested in clinical studies. Despite the evidence that DCs loaded with tumor-associated antigens can elicit immune responses in vivo, clinical responses remained disappointingly low. Therefore, optimization of the cellular product and route of administration was urgently needed. Here, we review the path we have followed in the development of TriMixDC-MEL, a potent DC-based cellular therapy, discussing its development as well as further modifications and applications.


Asunto(s)
Células Dendríticas/inmunología , Inmunoterapia Adoptiva/métodos , Melanoma/inmunología , Melanoma/terapia , Animales , Humanos
4.
Eur J Immunol ; 42(6): 1417-28, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22678898

RESUMEN

Antitumor cytolytic T lymphocytes (CTLs) recognize peptides derived from cellular proteins and presented on MHC class I. One category of peptides recognized by these CTLs is derived from proteins encoded by "cancer-germline" genes, which are specifically expressed in tumors, and therefore represent optimal targets for cancer immunotherapy. Here, we identify an antigenic peptide, which is derived from the MAGE-A1-encoded protein (160-169) and presented to CTLs by HLA-B*44:02. Although this peptide is encoded by MAGE-A1, processed endogenously and presented by tumor cells, the corresponding synthetic peptide is hardly able to sensitize target cells to CTL recognition when pulsed exogenously. Endogenous processing and presentation of this peptide is strictly dependent on the presence of tapasin, which is believed to help peptide loading by stabilizing a peptide-receptive form of HLA-B*44:02. Exogenous loading of the peptide can be dramatically improved by paraformaldehyde fixation of surface molecules or by peptide loading at acidic pH. Either strategy allows efficient exogenous loading of the peptide, presumably by generating or stabilizing a peptide-receptive, empty conformation of the HLA. Altogether, our results indicate a potential drawback of short peptide-based vaccination strategies and offer possible solutions regarding the use of problematic epitopes such as the one described here.


Asunto(s)
Antígeno HLA-B44/inmunología , Antígenos Específicos del Melanoma/inmunología , Proteínas de Transporte de Membrana/fisiología , Linfocitos T Citotóxicos/inmunología , Animales , Presentación de Antígeno , Células COS , Línea Celular Tumoral , Chlorocebus aethiops , Humanos , Concentración de Iones de Hidrógeno
5.
Mol Ther ; 20(5): 1063-74, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22371843

RESUMEN

It is generally thought that dendritic cells (DCs) loaded with full-length tumor antigen could improve immunotherapy by stimulating broad T-cell responses and by allowing treatment irrespective of the patient's human leukocyte antigen (HLA) type. To investigate this, we determined the specificity of T cells from melanoma patients treated with DCs loaded with mRNA encoding a full-length tumor antigen fused to a signal peptide and an HLA class II sorting signal, allowing presentation in HLA class I and II. In delayed-type hypersensitive (DTH)-biopsies and blood, we found functional CD8(+) and CD4(+) T cells recognizing novel treatment-antigen-derived epitopes, presented by several HLA types. Additionally, we identified a CD8(+) response specific for the signal peptide incorporated to elicit presentation by HLA class II and a CD4(+) response specific for the fusion region of the signal peptide and one of the antigens. This demonstrates that the fusion proteins contain newly created immunogenic sequences and provides evidence that ex vivo-generated mRNA-modified DCs can induce effector CD8(+) and CD4(+) T cells from the naive T-cell repertoire of melanoma patients. Thus, this work provides definitive proof that DCs presenting the full antigenic spectrum of tumor antigens can induce T cells specific for novel epitopes and can be administered to patients irrespective of their HLA type.


Asunto(s)
Antígenos de Neoplasias/inmunología , Células Dendríticas/trasplante , Antígenos HLA-D/inmunología , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoterapia Adoptiva/métodos , Melanoma/terapia , ARN Mensajero/inmunología , Neoplasias Cutáneas/terapia , Secuencia de Aminoácidos , Presentación de Antígeno , Antígenos de Neoplasias/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Electroporación , Antígenos HLA-D/genética , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Activación de Linfocitos , Melanoma/inmunología , Melanoma/patología , Datos de Secuencia Molecular , Señales de Clasificación de Proteína , ARN Mensajero/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/patología , Transfección
6.
Cancer Immunol Immunother ; 61(7): 1033-43, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22159452

RESUMEN

Dendritic cells (DCs) electroporated with mRNA encoding CD70, CD40L and a constitutively active toll-like receptor 4 (TriMix-DC) have an increased T-cell stimulatory capacity. In a prospective phase IB clinical trial, we treated melanoma patients with intradermal and intravenous injections of autologous TriMix-DC co-electroporated with mRNA encoding full-length MAGE-A3, MAGE-C2, tyrosinase and gp100. We report here the immunological and clinical results obtained in one patient with a particularly favorable outcome. This patient had stage IV-M1c melanoma with documented progression during dacarbazine chemotherapy and received 5 TriMix-DC injections. Following DC therapy, a broad CD8(+) T-cell response against multiple epitopes derived from all four treatment antigens was found in the blood and among T cells derived from DTH biopsy. In addition, CD4(+) T cells recognizing different MAGE-A3-derived epitopes were detected in DTH-derived cells. A spontaneous anti-MAGE-C2 CD8(+) T-cell response was present prior to TriMix-DC therapy and increased during treatment. The tumor response was assessed with 18-fluorodeoxyglucose-positron emission/computed tomography. We documented a partial tumor response according to RECIST criteria with a marked reduction in (18)F-FDG-uptake by lung, lymph node and bone metastases. The patient remains free from progression after 12 months of follow-up. This case report indicates that administration of autologous TriMix-DC by the combined intradermal and intravenous route can mediate a durable objective tumor response accompanied by a broad T-cell response in a chemorefractory stage IV-M1c melanoma patient.


Asunto(s)
Células Dendríticas/inmunología , Helio/administración & dosificación , Inmunoterapia Adoptiva/métodos , Melanoma/inmunología , Melanoma/terapia , Nitrógeno/administración & dosificación , Oxígeno/administración & dosificación , Linfocitos T/inmunología , Secuencia de Aminoácidos , Ligando CD27/biosíntesis , Ligando CD27/genética , Ligando CD27/inmunología , Ligando de CD40/biosíntesis , Ligando de CD40/genética , Ligando de CD40/inmunología , Células Dendríticas/patología , Electroporación/métodos , Humanos , Hipersensibilidad Tardía/inmunología , Masculino , Melanoma/patología , Persona de Mediana Edad , Datos de Secuencia Molecular , Estadificación de Neoplasias , Estudios Prospectivos , ARN Mensajero/administración & dosificación , ARN Mensajero/genética , Receptor Toll-Like 4/biosíntesis , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
7.
Mol Ther ; 19(5): 841-53, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21468005

RESUMEN

Cancer immunotherapy aims to establish immune-mediated control of tumor growth by priming T-cell responses to target tumor-associated antigens. Three signals are required for T-cell activation: (i) presentation of cognate antigen in self MHC molecules; (ii) costimulation by membrane-bound receptor-ligand pairs; and (iii) soluble factors to direct polarization of the ensuing immune response. The ability of dendritic cells (DCs) to provide all three signals required for T-cell activation makes them an ideal cancer vaccine platform. Several strategies have been developed to enhance and control antigen presentation, costimulation, and cytokine production. In this review, we discuss progress toward developing DC-based cancer vaccines by genetic modification using RNA, DNA, and recombinant viruses. Furthermore, the ability of DC-based vaccines to activate natural killer (NK) and B-cells, and the impact of gene modification strategies on these populations is described. Clinical trials using gene-modified DCs have shown modest results, therefore, further considerations for DC manipulation to enhance their clinical efficacy are also discussed.


Asunto(s)
Células Dendríticas/citología , Inmunoterapia/métodos , Neoplasias/terapia , Presentación de Antígeno , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Humanos , Activación de Linfocitos , Neoplasias/inmunología
8.
Clin Cancer Res ; 15(10): 3366-75, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19417017

RESUMEN

PURPOSE: A critical factor determining the effectiveness of currently used dendritic cell (DC)-based vaccines is the DC activation or maturation status. We have recently shown that the T-cell stimulatory capacity of DCs pulsed with tumor-antigen-derived peptides can be considerably increased by activating the DCs through electroporation with mRNA encoding CD40 ligand, CD70, and a constitutively active Toll-like receptor 4 (TriMix DCs). Here, we investigate whether TriMix DCs can be coelectroporated with whole tumor-antigen-encoding mRNA. EXPERIMENTAL DESIGN: The T-cell stimulatory capacity of TriMix DCs pulsed with the immunodominant MelanA-A2 peptide and that of TriMix DCs coelectroporated with MelanA mRNA were compared in vitro. TriMix DCs were also coelectroporated with mRNA encoding Mage-A3, Mage-C2, tyrosinase, or gp100. The capacity of these DCs to stimulate tumor-antigen-specific T cells in melanoma patients was investigated both in vitro before vaccination and after DC vaccination. RESULTS: Like peptide-pulsed TriMix DCs, TriMix DCs coelectroporated with MelanA mRNA are very potent in inducing MelanA-specific CD8(+) T cells in vitro. These T cells have an activated phenotype, show cytolytic capacity, and produce inflammatory cytokines in response to specific stimulation. TriMix DCs coelectroporated with tyrosinase are able to stimulate tyrosinase-specific CD8(+) T cells in vitro from the blood of nonvaccinated melanoma patients. Furthermore, TriMix DCs coelectroporated with Mage-A3, Mage-C2, or tyrosinase are able to induce antigen-specific CD8(+) T cells through therapeutic DC vaccination. CONCLUSIONS: TriMix DCs coelectroporated with whole tumor-antigen mRNA stimulate antigen-specific T cells in vitro and induce antigen-specific T-cell responses in melanoma patients through vaccination. Therefore, they represent a promising new approach for antitumor immunotherapy.


Asunto(s)
Antígenos de Neoplasias/inmunología , Células Dendríticas/inmunología , Electroporación/métodos , Melanoma/inmunología , ARN Mensajero/genética , Antígenos de Neoplasias/genética , Ligando CD27/genética , Ligando CD27/inmunología , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Citocinas/metabolismo , Células Dendríticas/citología , Células Dendríticas/metabolismo , Femenino , Citometría de Flujo , Humanos , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Antígeno MART-1 , Masculino , Melanoma/patología , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Monofenol Monooxigenasa/genética , Monofenol Monooxigenasa/inmunología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Estadificación de Neoplasias , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Vacunación/métodos , Antígeno gp100 del Melanoma
9.
Mol Ther ; 16(6): 1170-80, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18431362

RESUMEN

The effectiveness of the dendritic cell (DC) vaccination protocols that are currently in use could be improved by providing the DCs with a more potent maturation signal. We therefore investigated whether the T-cell stimulatory capacity of human monocyte-derived DCs could be increased by co-electroporation with different combinations of CD40L, CD70, and constitutively active toll-like receptor 4 (caTLR4) encoding mRNA. We show that immature DCs electroporated with CD40L and/or caTLR4 mRNA, but not those electroporated with CD70 mRNA, acquire a mature phenotype along with an enhanced secretion of several cytokines/chemokines. Moreover, these DCs are very potent in inducing naive CD4(+) T cells to differentiate into interferon-gamma (IFN-gamma)-secreting type 1 T helper (Th1) cells. Further, we assessed the capacity of the electroporated DCs to activate naive HLA-A2-restricted MelanA-specific CD8(+) T cells without the addition of any exogenous cytokines. When all three molecules were combined, a >500-fold increase in MelanA-specific CD8(+) T cells was observed when compared with immature DCs, and a >200-fold increase when compared with cytokine cocktail-matured DCs. In correlation, we found a marked increase in cytolytic and IFN-gamma/tumor necrosis factor-alpha (TNF-alpha) secreting CD8(+) T cells. Our data indicate that immature DCs genetically modified to express stimulating molecules can induce tumor antigen-specific T cells in vitro and could prove to be a significant improvement over DCs matured with the methods currently in use.


Asunto(s)
Ligando CD27/metabolismo , Ligando de CD40/metabolismo , Células Dendríticas/citología , Electroporación/métodos , Linfocitos T/citología , Receptor Toll-Like 4/metabolismo , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Antígeno HLA-A2/metabolismo , Humanos , Interferón gamma/metabolismo , Células K562 , Modelos Biológicos , Células TH1/citología , Factor de Necrosis Tumoral alfa/metabolismo
10.
Methods Mol Biol ; 423: 155-63, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18370196

RESUMEN

Antigen-loaded dendritic cells (DCs) have been intensively investigated as potential cellular antitumor vaccines. Several recent reports have indicated that loading DCs with whole tumor derived mRNA or defined tumor-antigen-encoding mRNA represents an effective nonviral strategy to stimulate T cell responses both for in vitro and in vivo models. Here, we describe the electroporation method as a tool for introducing in vitro transcribed capped mRNA into human DCs for tumor vaccination. We use MART-1/Melan-A as a model tumor-associated antigen for the generation of a DC-based vaccine against melanoma cancer. In addition to efficient antigen loading, it is important to obtain a maximal number of potent antigen-presenting cells. Another prerequisite for the development of a DC-based cancer vaccine is to obtain mature DCs. In this chapter, we describe the basic techniques required for the successful genetic modification of DCs by using the mRNA electroporation method.


Asunto(s)
Antígenos de Neoplasias/genética , Vacunas contra el Cáncer/administración & dosificación , Células Dendríticas/inmunología , ARN Mensajero/administración & dosificación , ARN Mensajero/genética , Secuencia de Bases , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Separación Celular , Cartilla de ADN/genética , Electroquimioterapia/métodos , Electroporación/métodos , Humanos , Antígeno MART-1 , Melanoma/genética , Melanoma/inmunología , Melanoma/terapia , Mutagénesis Sitio-Dirigida , Proteínas de Neoplasias/genética , Plásmidos/genética , Caperuzas de ARN/genética , Linfocitos T/inmunología
11.
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
12.
J Leukoc Biol ; 78(4): 898-908, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16037410

RESUMEN

Dendritic cells (DC) are professional antigen-presenting cells that are used in vaccine approaches to cancer. Classically, mature monocyte-derived DC are generated in vitro in the presence of interleukin (IL)-4, granulocyte macrophage-colony stimulating factor, and inflammatory cytokines (G4-DC). Recently, it has been described that DC can also be generated in the presence of IL-3 and interferon (IFN)-beta and that these DC are efficiently matured using polyriboinosinic polyribocytidylic acid (I3-DC). In this study, a series of in vitro experiments was performed to compare side-by-side I3-DC and G4-DC as vaccine adjuvants. Phenotypic characterization of the DC revealed differences in the expression of the monocyte marker CD14 and the maturation marker CD83. Low expression of CD14 and high expression of CD83 characterized G4-DC, whereas I3-DC displayed intermediate expression of CD14 and CD83. Both types of DC were as potent in the induction of allogeneic T cell proliferation. Upon CD40 ligation, G4-DC produced lower amounts of IFN-alpha and pulmonary and activation-regulated chemokine, similar amounts of IL-6, macrophage-inflammatory protein (MIP)-1alpha, and MIP-1beta, and higher amounts of IL-12 p70, tumor necrosis factor alpha, and MIP-3beta than I3-DC. We further evaluated whether the DC could be frozen/thawed without loss of cell number, viability, phenotype, and function. After freezing/thawing, 56.0% +/- 9.0% of I3-DC and 77.0% +/- 3.0% of G4-DC (n=9) were recovered as viable cells, displaying the same phenotype as their fresh counterparts. Finally, in vitro stimulations showed that fresh and frozen peptide-loaded I3-DC are more potent inducers of Melan-A-specific CD8(+) T cell responses than G4-DC. The antigen-specific T cells were functional as shown in cytotoxicity and IFN-gamma secretion assay.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Células Dendríticas/fisiología , Interferón beta/farmacología , Interleucina-3/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Diferenciación Celular/fisiología , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/inmunología , Humanos , Técnicas In Vitro , Interferón gamma/metabolismo , Fenotipo
13.
Cancer Res ; 63(17): 5587-94, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-14500399

RESUMEN

For the induction of an optimal immune response against cancer or infections not only CD8(+) CTLs but also CD4(+) T helper cells must be induced, in particular IFN-gamma-secreting type 1 T helper cells. Several strategies have been explored to target tumor-associated antigens to the HLA class II processing compartments. We engineered a genetic construct encoding an invariant chain (Ii) protein where the CLIP region has been replaced by sequences encoding HLA class II-restricted MAGE-A3 epitopes. Monocyte-derived dendritic cells (DCs) were electroporated with in vitro transcribed mRNA encoding a modified Ii protein containing the HLA-DP4-restricted MAGE-A3 epitope. The capacity of these electroporated DCs to stimulate a MAGE-A3-specific T-cell clone was compared at different stages of DC maturation with the T-cell stimulatory capacity of DCs pulsed with the synthetic peptide. After electroporation, the T-cell stimulatory capacity was shown to be high and long lasting, whereas the stimulatory capacity of peptide-pulsed DCs decreased rapidly. Upon coculture with epitope-specific T cells, electroporated immature DCs expressed enhanced levels of costimulatory molecules, HLA class II molecules, and CD83, suggesting the induction of maturation. The electroporated DCs can be frozen and thawed without losing their capability to stimulate the specific T-cell clone in vitro, and they are able to stimulate unprimed CD4(+) T cells specific to the HLA-DP4-restricted MAGE-A3 epitope in vitro. Similar results were obtained with a recombinant Ii containing the MAGE-A3 epitope presented in the context of HLA-DR13.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos de Diferenciación de Linfocitos B/inmunología , Antígenos de Neoplasias/inmunología , Células Dendríticas/inmunología , Antígenos HLA-DP/inmunología , Antígenos HLA-DR/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Proteínas de Neoplasias , Secuencia de Aminoácidos , Presentación de Antígeno/genética , Antígenos de Diferenciación de Linfocitos B/genética , Antígenos de Neoplasias/genética , Linfocitos T CD4-Positivos/inmunología , Electroporación , Epítopos/genética , Epítopos/inmunología , Epítopos de Linfocito T/inmunología , Antígenos HLA-DP/genética , Cadenas beta de HLA-DP , Antígenos HLA-DR/genética , Subtipos Serológicos HLA-DR , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Inmunofenotipificación , Activación de Linfocitos/inmunología , Datos de Secuencia Molecular , ARN Mensajero/genética , ARN Mensajero/inmunología
14.
Methods Mol Biol ; 1428: 115-23, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27236795

RESUMEN

Dendritic cells (DCs) are the orchestrators of the immune system and are frequently used in clinical trials in order to boost the immune system in cancer patients. Among several available techniques for DC modification, mRNA electroporation is an interesting technique due to the favorable characteristics of mRNA. Antigen expression level and duration can be increased by multiple optimizations of an antigen-encoding mRNA template. Here, we describe different molecular modifications to a WT1-encoding mRNA construct in order to increase antigen expression and the subsequent introduction of mRNA into DCs.


Asunto(s)
Células Dendríticas/metabolismo , Ingeniería Genética/métodos , ARN Mensajero/metabolismo , Proteínas WT1/genética , Electroporación , Humanos , Biosíntesis de Proteínas , Caperuzas de ARN/metabolismo , ARN Mensajero/química , ARN Mensajero/genética , Proteínas WT1/metabolismo
15.
Expert Rev Vaccines ; 14(2): 161-76, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25196947

RESUMEN

Cancer immunotherapy has been proposed as a powerful treatment modality. Active immunotherapy aspires to stimulate the patient's immune system, particularly T cells. These cells can recognize and kill cancer cells and can form an immunological memory. Dendritic cells (DCs) are the professional antigen-presenting cells of our immune system. They take up and process antigens to present them to T cells. Consequently, DCs have been investigated as a means to stimulate cancer-specific T-cell responses. An efficient strategy to program DCs is the use of mRNA, a well-defined and safe molecule that can be easily generated at high purity. Importantly, vaccines consisting of mRNA-modified DCs showed promising results in clinical trials. Therefore, we will introduce cancer immunotherapy and DCs and give a detailed overview on the application of mRNA to generate cancer-fighting DC vaccines.


Asunto(s)
Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Inmunoterapia Activa , Neoplasias/inmunología , ARN Mensajero/inmunología , Presentación de Antígeno , Humanos , Memoria Inmunológica , Neoplasias/terapia , Linfocitos T/inmunología
16.
Mol Med Rep ; 12(2): 2443-50, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25936433

RESUMEN

The aim of the present study was to enhance the efficiency of leukemia immunotherapy by increasing the antigen-specific cytotoxic T lymphocyte-inducing ability of leukemia cells. The leukemic plasmacytoid dendritic cell line PMDC05 containing the HLA-A02/24 antigen, which was previously established in our laboratory (Laboratory of Hematology and Oncology, Graduate School of Health Sciences, Niigata University, Niigata, Japan), was used in the present study. It exhibited higher expression levels of CD80 following transduction with lentiviruses encoding the CD80 gene. This CD80-expressing PMDC05 was named PMDC11. In order to establish a more potent antigen-presenting cell for cellular immunotherapy of tumors or severe infections, PMDC11 cells were transduced with a constitutively active (ca) toll-like receptor 4 (TLR4) gene using the Tet-On system (caTLR4-PMDC11). CD8(+) T cells from healthy donors with HLA-A02 were co-cultured with mutant WT1 peptide-pulsed PMDC11, lipopolysaccharide (LPS)-stimulated PMDC11 or caTLR4-PMDC11 cells. Interleukin (IL)-2 (50 IU/ml) and IL-7 (10 ng/ml) were added on day three of culture. Priming with mutant WT1 peptide-pulsed PMDC11, LPS-stimulated PMDC11 or caTLR4-PMDC11 cells was conducted once per week and two thirds of the IL-2/IL-7 containing medium was replenished every 3-4 days. Immediately prior to the priming with these various PMDC11 cells, the cultured cells were analyzed for the secretion of interferon (IFN)-γ in addition to the percentage and number of CD8(+)/WT1 tetramer(+) T cells using flow cytometry. caTLR4-PMDC11 cells were observed to possess greater antigen-presenting abilities compared with those of PMDC11 or LPS-stimulated PMDC11 cells in a mixed leukocyte culture. CD8 T cells positive for the WT1 tetramer were generated following 3-4 weeks of culture and CD8(+)/WT1 tetramer+ T cells were markedly increased in caTLR4-PMDC11-primed CD8(+) T cell culture compared with PMDC11 or LPS-stimulated PMDC11-primed CD8(+) T cell culture. These CD8(+) T cells co-cultured with caTLR4-PMDC11 cells were demonstrated to secrete IFN-γ and to be cytotoxic to WT1-expressing target cells. These data suggested that the antigen-specific cytotoxic T lymphocyte (CTL)-inducing ability of PMDC11 was potentiated via transduction of the caTLR4 gene. The present study also suggested that caTLR4-PMDC11 cells may be applied as potent antigen-presenting cells for generating antigen-specific CTLs in adoptive cellular immunotherapy against tumors and severe viral infections.


Asunto(s)
Antígenos de Neoplasias/genética , Células Dendríticas/inmunología , Regulación Leucémica de la Expresión Génica , Linfocitos T Citotóxicos/inmunología , Receptor Toll-Like 4/genética , Transducción Genética , Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Antígeno B7-1/genética , Antígeno B7-1/inmunología , Línea Celular Tumoral , Técnicas de Cocultivo , Células Dendríticas/efectos de los fármacos , Células Dendríticas/patología , Vectores Genéticos/inmunología , Vectores Genéticos/metabolismo , Antígenos HLA-A/genética , Antígenos HLA-A/inmunología , Humanos , Inmunoterapia/métodos , Interferón gamma/biosíntesis , Interferón gamma/inmunología , Interleucina-2/farmacología , Interleucina-7/farmacología , Lentivirus/genética , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/inmunología , Leucemia Linfocítica Crónica de Células B/patología , Leucemia Linfocítica Crónica de Células B/terapia , Prueba de Cultivo Mixto de Linfocitos , Transducción de Señal , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/efectos de los fármacos , Receptor Toll-Like 4/inmunología , Proteínas WT1/farmacología
17.
Hum Gene Ther ; 15(6): 562-73, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15212715

RESUMEN

In this study, we compared dendritic cells (DCs) differentiated from positively selected monocytes (CD14-DCs) to DCs differentiated from adherence-selected monocytes (adh-DCs) with emphasis on lentiviral transduction. Using a second-generation, triple-helix containing, self-inactivating lentiviral vector at a multiplicity of infection (MOI) of 15, we observed enhanced transduction of CD14-DCs (72.8 +/- 5.3%, mean fluorescence intensity [MFI] = 166 +/- 76) compared to adh-DCs (32.3 +/- 13.1%, MFI = 119 +/- 76, n = 5). More importantly, the efficiency to transduce adh-DCs was significantly increased when monocytes were incubated with antiCD14 antibody coupled beads, anti-CD14 antibodies, or lipopolysaccharide (LPS), reaching transduction efficiencies up to 86.6%, 53.3%, and 80.9%, respectively. We showed that this enhanced transduction was correlated to an activation of the monocytes, characterized by the up regulation of the cytokines interleukin (IL)-1beta and tumor necrosis factor (TNF)-alpha and the de novo synthesis of IL-6 and IL-10. However, the enhanced transduction of immature CD14-DCs was not correlated with a progression in the cell cycle from G(0) to G(1). We further showed that CD14-DCs were phenotypically comparable to adh-DCs. Functional analysis revealed that there were no differences in allostimulatory capacity, production of IL-12 p70 on CD40 ligation or expression of IL-1beta, IL-6, IL-8, IL-10, IL-12, and TNF-alpha as evaluated by reverse transcriptase-polymerase chain reaction (RT-PCR). Finally, we showed that lentivirally transduced CD14-DCs were equally capable as adh-DCs in stimulating MAGE-A3 antigen-specific CD4(+) and CD8(+) T cells in vitro.


Asunto(s)
Células Dendríticas/inmunología , Técnicas de Transferencia de Gen , Vectores Genéticos , Lentivirus/genética , Receptores de Lipopolisacáridos/metabolismo , Monocitos/inmunología , Transducción Genética , Anticuerpos Monoclonales , Antígenos de Neoplasias/metabolismo , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Ciclo Celular , Diferenciación Celular/efectos de los fármacos , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/citología , Humanos , Inmunofenotipificación , Leucocitos Mononucleares/metabolismo , Receptores de Lipopolisacáridos/genética , Lipopolisacáridos/farmacología , Activación de Linfocitos , Monocitos/citología , Proteínas de Neoplasias/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
18.
Cancer Gene Ther ; 10(9): 696-706, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12944989

RESUMEN

Genetically modified dendritic cells (DC) constitute a promising approach in cancer immunotherapy. Viral gene delivery systems have been shown to be very efficient strategies, but safety concerns for their clinical use in immunotherapy remain an important issue. Recently, the technique of mRNA electroporation was described as a very efficient tool for the genetic modification of human monocyte-derived DC. Here, we show that transgene expression can be modulated by varying the amount of mRNA used for electroporation. We document that CD40 ligation leads to a significant production of IL-12 by the electroporated DC, although the level of IL-12 production is somewhat lower than for non- or mock-electroporated DC. Furthermore, we show that the electroporated DC can be frozen and thawed without loss of viability or function and that Influenza virus Matrix Protein 1 mRNA electroporated DC are capable of inducing a memory cytotoxic T lymphocyte response and are more potent in doing so than mRNA-pulsed DC. Similar results were obtained with MelanA/MART-1 mRNA electroporated DC. These results clearly indicate that mRNA-electroporated DC represent powerful candidates for use as tumor vaccines and could constitute an improvement compared with vaccines using peptide-pulsed DC.


Asunto(s)
Células Dendríticas/inmunología , Proteínas de Neoplasias/inmunología , Linfocitos T/inmunología , Proteínas de la Matriz Viral/inmunología , Presentación de Antígeno , Antígenos de Neoplasias , Antígenos CD40/metabolismo , Vacunas contra el Cáncer/inmunología , Línea Celular , Supervivencia Celular , Ensayos Clínicos como Asunto , Criopreservación , Células Dendríticas/citología , Electroporación , Humanos , Memoria Inmunológica , Inmunoterapia , Interferón gamma/inmunología , Interleucina-12/biosíntesis , Interleucina-12/inmunología , Antígeno MART-1 , Proteínas de Neoplasias/genética , Orthomyxoviridae/genética , Orthomyxoviridae/inmunología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transgenes/genética , Proteínas de la Matriz Viral/genética
19.
Methods Mol Biol ; 1139: 3-15, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24619665

RESUMEN

Dendritic cells (DC) are key players in several types of cancer vaccines. Large numbers of DC can easily be generated in closed systems from the monocyte fraction of the peripheral blood. They are the professional antigen-presenting cells, and electroporation of mRNA-encoding tumor antigens is a very efficient and a relatively simple way to load the DC with antigen. The co-electroporation of a tumor antigen of choice and the combination of 3 costimulatory molecules, including CD70, caTLR4, and CD40L (TriMix-DC), leads to fully potent antigen-presenting DC able to generate a broad immune response.Here we describe the in vitro transcription of the mRNA and the subsequent generation and electroporation of autologous DC used for the treatment of melanoma patients.


Asunto(s)
Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Diferenciación Celular , Células Dendríticas/citología , Células Dendríticas/inmunología , Electroporación/métodos , ARN Mensajero/metabolismo , Ligando CD27/genética , Ligando CD27/inmunología , Ligando de CD40/genética , Ligando de CD40/inmunología , Células Dendríticas/trasplante , Humanos , Melanoma/inmunología , Melanoma/prevención & control , ARN Mensajero/genética , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Transcripción Genética
20.
Biomed Res Int ; 2013: 976383, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23509826

RESUMEN

Treatment of melanoma patients with mRNA electroporated dendritic cells (TriMixDC-MEL) stimulates T-cell responses against the presented tumor-associated antigens (TAAs). In the current clinical trials, melanoma patients with systemic metastases are treated, requiring priming and/or expansion of preexisting TAA-specific T cells that are able to migrate to both the skin and internal organs. We monitored the presence of TAA-specific CD8(+) T cells infiltrating the skin at sites of intradermal TriMixDC-MEL injection (SKILs) and within the circulation of melanoma patients treated in two clinical trials. In 10 out of fourteen (71%) patients screened, CD8(+) T cells recognizing any of the four TAA presented by TriMixDC-MEL cellular vaccine were found in both compartments. In total, 30 TAA-specific T-cell responses were detected among the SKILs and 29 among peripheral blood T cells, of which 24 in common. A detailed characterization of the antigen specificity of CD8(+) T-cell populations in four patients indicates that the majority of the epitopes detected were only recognized by CD8(+) T cells derived from either skin biopsies or peripheral blood, indicating that some compartmentalization occurs after TriMix-DC therapy. To conclude, functional TAA-specific CD8(+) T cells distribute both to the skin and peripheral blood of patients after TriMixDC-MEL therapy.


Asunto(s)
Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/uso terapéutico , Células Dendríticas/trasplante , Melanoma/terapia , Neoplasias Cutáneas/terapia , Biopsia , Movimiento Celular , Electroporación , Epítopos/inmunología , Humanos , Metástasis de la Neoplasia , Proyectos Piloto , Recurrencia , Piel/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA