Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 25(7)2024 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-38612407

RESUMEN

A small fraction of people vaccinated with mRNA-lipid nanoparticle (mRNA-LNP)-based COVID-19 vaccines display acute or subacute inflammatory symptoms whose mechanism has not been clarified to date. To better understand the molecular mechanism of these adverse events (AEs), here, we analyzed in vitro the vaccine-induced induction and interrelations of the following two major inflammatory processes: complement (C) activation and release of proinflammatory cytokines. Incubation of Pfizer-BioNTech's Comirnaty and Moderna's Spikevax with 75% human serum led to significant increases in C5a, sC5b-9, and Bb but not C4d, indicating C activation mainly via the alternative pathway. Control PEGylated liposomes (Doxebo) also induced C activation, but, on a weight basis, it was ~5 times less effective than that of Comirnaty. Viral or synthetic naked mRNAs had no C-activating effects. In peripheral blood mononuclear cell (PBMC) cultures supplemented with 20% autologous serum, besides C activation, Comirnaty induced the secretion of proinflammatory cytokines in the following order: IL-1α < IFN-γ < IL-1ß < TNF-α < IL-6 < IL-8. Heat-inactivation of C in serum prevented a rise in IL-1α, IL-1ß, and TNF-α, suggesting C-dependence of these cytokines' induction, although the C5 blocker Soliris and C1 inhibitor Berinert, which effectively inhibited C activation in both systems, did not suppress the release of any cytokines. These findings suggest that the inflammatory AEs of mRNA-LNP vaccines are due, at least in part, to stimulation of both arms of the innate immune system, whereupon C activation may be causally involved in the induction of some, but not all, inflammatory cytokines. Thus, the pharmacological attenuation of inflammatory AEs may not be achieved via monotherapy with the tested C inhibitors; efficacy may require combination therapy with different C inhibitors and/or other anti-inflammatory agents.


Asunto(s)
COVID-19 , Inactivadores del Complemento , Nanopartículas , Humanos , Liposomas , Vacunas contra la COVID-19/efectos adversos , Leucocitos Mononucleares , Citocinas , Factor de Necrosis Tumoral alfa , Vacuna BNT162 , Activación de Complemento , Lípidos
2.
J Immunol ; 202(2): 451-459, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30559321

RESUMEN

The intracellular pathway of cross-presentation, which allows MHC class I-restricted presentation of peptides derived from exogenous Ags, remains poorly defined and may vary with the nature of the exogenous Ag and the type of APC. It can be cytosolic, characterized by proteasome and TAP dependency, or vacuolar, usually believed to be proteasome and TAP independent. Cross-presentation is particularly effective with long synthetic peptides, and we previously reported that the HLA-A2-restricted cross-presentation of a long peptide derived from melanoma Ag gp100 by human monocyte-derived immature dendritic cells occurred in a vacuolar pathway, making use of newly synthesized HLA-A2 molecules that follow a nonclassical secretion route. In this article, we show that the HLA-A1-restricted cross-presentation of a long peptide derived from tumor Ag MAGE-A3 by human monocyte-derived immature dendritic cells also follows a vacuolar pathway. However, as opposed to the HLA-A2-restricted peptide, cross-presentation of the HLA-A1-restricted peptide is TAP dependent. We show that this paradoxical TAP-dependency is indirect and reflects the need for TAP to load HLA-A1 molecules with peptides in the endoplasmic reticulum, to allow them to escape the endoplasmic reticulum and reach the vacuole, where peptide exchange with the cross-presented peptide likely occurs. Our results confirm and extend the involvement of the vacuolar pathway in the cross-presentation of long peptides, and indicate that TAP-dependency can no longer be used as a key criterion to distinguish the cytosolic from the vacuolar pathway of cross-presentation. They also stress the existence of an alternative secretory route for MHC class I, which will be worthy of further studies.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Antígenos de Neoplasias/metabolismo , Células Dendríticas/inmunología , Retículo Endoplásmico/metabolismo , Antígeno HLA-A1/metabolismo , Proteínas de Neoplasias/metabolismo , Linfocitos T Citotóxicos/inmunología , Vacuolas/metabolismo , Presentación de Antígeno , Línea Celular , Reactividad Cruzada , Citosol/metabolismo , Antígeno HLA-A2/metabolismo , Humanos , Péptidos/metabolismo , Antígeno gp100 del Melanoma/metabolismo
3.
Cancer Immunol Immunother ; 69(12): 2589-2598, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32591862

RESUMEN

BACKGROUND: Autologous monocyte-derived mRNA co-electroporated dendritic cells with mRNA encoding CD40 ligand (CD40L), CD70 and a constitutively activated TLR4 (caTLR4) (referred to as TriMixDC-MEL) have anti-tumor activity in advanced melanoma patients. We investigated the safety and activity of adjuvant TriMixDC-MEL in stage III/IV melanoma patients. MATERIALS AND METHODS: Forty-one patients were randomly assigned to treatment with TriMixDC-MEL (n = 21) and standard follow-up (n = 20). "Cross-over" was allowed at the time of non-salvageable recurrence. The primary endpoint was the percentage of patients alive and disease-free at 1-year. For a subset of patients, (formalin-fixed paraffin-embedded), tumor tissue samples were available for mRNA expression profiling and PD-L1 immunohistochemical staining. RESULTS: Baseline characteristics were well balanced. One-year after randomization, 71% of patients in the study arm were alive and free of disease compared to 35% in the control arm. After a median follow-up of 53 months (range 3-67), 23 patients experienced a non-salvageable melanoma recurrence (TriMixDC-Mel arm n = 9 and control arm n = 14).The median time to non-salvageable recurrence was superior in the TriMixDC-MEL arm (median 8 months (range 1-6) vs. not reached; log-rank p 0.044). TriMixDC-MEL-related adverse events (AE) consisted of transient local skin reactions, flu-like symptoms and post-infusion chills. No grade ≥ 3 AE's occurred. The mRNA expression profiling revealed four genes (STAT2, TPSAB1, CD9 and CSF2) as potential predictive biomarkers. CONCLUSION: TriMixDC-MEL id/iv as adjuvant therapy is tolerable and may improve the 1-year disease-free survival rate. Combination of optimized autologous monocyte-derived DC-formulations warrants further investigation in combination with currently approved adjuvant therapy options.


Asunto(s)
Células Dendríticas/trasplante , Melanoma/terapia , Recurrencia Local de Neoplasia/epidemiología , ARN Mensajero/inmunología , Neoplasias Cutáneas/terapia , Adulto , Anciano , Anciano de 80 o más Años , Ligando CD27/genética , Ligando CD27/inmunología , Ligando de CD40/genética , Ligando de CD40/inmunología , Terapia Combinada/métodos , Células Dendríticas/metabolismo , Supervivencia sin Enfermedad , Electroporación , Femenino , Estudios de Seguimiento , Humanos , Inmunoterapia/métodos , Masculino , Melanoma/inmunología , Melanoma/mortalidad , Melanoma/secundario , Persona de Mediana Edad , Recurrencia Local de Neoplasia/prevención & control , Estadificación de Neoplasias , ARN Mensajero/genética , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/mortalidad , Neoplasias Cutáneas/patología , Procedimientos Quirúrgicos Operativos , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Trasplante Autólogo/métodos , Adulto Joven
4.
Int J Cancer ; 145(4): 1111-1124, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-30719701

RESUMEN

No curative treatment options are available for advanced hepatocellular carcinoma (HCC). Anti-PD1 antibody therapy can induce tumor regression in 20% of advanced HCC patients, demonstrating that co-inhibitory immune checkpoint blockade has therapeutic potential for this type of cancer. However, whether agonistic targeting of co-stimulatory receptors might be able to stimulate anti-tumor immunity in HCC is as yet unknown. We investigated whether agonistic targeting of the co-stimulatory receptor GITR could reinvigorate ex vivo functional responses of tumor-infiltrating lymphocytes (TIL) freshly isolated from resected tumors of HCC patients. In addition, we compared GITR expression between TIL and paired samples of leukocytes isolated from blood and tumor-free liver tissues, and studied the effects of combined GITR and PD1 targeting on ex vivo TIL responses. In all three tissue compartments, CD4+ FoxP3+ regulatory T cells (Treg) showed higher GITR- expression than effector T-cell subsets. The highest expression of GITR was found on CD4+ FoxP3hi CD45RA- activated Treg in tumors. Recombinant GITR-ligand as well as a humanized agonistic anti-GITR antibody enhanced ex vivo proliferative responses of CD4+ and CD8+ TIL to tumor antigens presented by mRNA-transfected autologous B-cell blasts, and also reinforced proliferation, IFN-γ secretion and granzyme B production in stimulations of TIL with CD3/CD28 antibodies. Combining GITR ligation with anti-PD1 antibody nivolumab further enhanced tumor antigen-specific responses of TIL in some, but not all, HCC patients, compared to either single treatment. In conclusion, agonistic targeting of GITR can enhance functionality of HCC TIL, and may therefore be a promising strategy for single or combinatorial immunotherapy in HCC.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Carcinoma Hepatocelular/inmunología , Proteína Relacionada con TNFR Inducida por Glucocorticoide/inmunología , Neoplasias Hepáticas/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos T Reguladores/inmunología , Anticuerpos Monoclonales/inmunología , Linfocitos B/inmunología , Línea Celular Tumoral , Femenino , Humanos , Inmunoterapia/métodos , Masculino , Persona de Mediana Edad , ARN Mensajero/inmunología
5.
J Transl Med ; 17(1): 242, 2019 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-31345237

RESUMEN

BACKGROUND: Current human influenza vaccines lack the adaptability to match the mutational rate of the virus and therefore require annual revisions. Because of extensive manufacturing times and the possibility that antigenic alterations occur during viral vaccine strain production, an inherent risk exists for antigenic mismatch between the new influenza vaccine and circulating viruses. Targeting more conserved antigens such as nucleoprotein (NP) could provide a more sustainable vaccination strategy by inducing long term and heterosubtypic protection against influenza. We previously demonstrated that intranodal mRNA injection can induce potent antigen-specific T-cell responses. In this study, we investigated whether intranodal administration of mRNA encoding NP can induce T-cell responses capable of protecting against a heterologous influenza virus challenge. METHODS: BALB/c mice were immunized in the inguinal lymph nodes with different vaccination regimens of mRNA encoding NP. Immune responses were compared with NP DNA vaccination via IFN-γ ELISPOT and in vivo cytotoxicity. For survival experiments, mice were prime-boost vaccinated with 17 µg NP mRNA and infected with 1LD50 of H1N1 influenza virus 8 weeks after boost. Weight was monitored and viral titers, cytokines and immune cell populations in the bronchoalveolar lavage, and IFN-γ responses in the spleen were analyzed. RESULTS: Our results demonstrate that NP mRNA induces superior systemic T-cell responses against NP compared to classical DNA vaccination. These responses were sustained for several weeks even at low vaccine doses. Upon challenge infection, vaccination with NP mRNA resulted in reduced lung viral titers and improved recovery from infection. Finally, we show that vaccination with NP mRNA affects the immune response in infected lungs by lowering immune cell infiltration while increasing the fraction of T cells, monocytes and MHC II+ alveolar macrophages within immune infiltrates. This change was associated with altered levels of both pro- and anti-inflammatory cytokines. CONCLUSIONS: These findings suggest that intranodal vaccination with NP mRNA induces cross-strain immunity against influenza, but also highlight a paradox of influenza immunity, whereby robust immune responses can provide protection, but can also transiently exacerbate symptoms during infection.


Asunto(s)
Vacunas contra la Influenza/inmunología , Nucleoproteínas/administración & dosificación , Infecciones por Orthomyxoviridae/prevención & control , ARN Mensajero/administración & dosificación , Animales , Anticuerpos Antivirales/inmunología , Antígenos/química , Lavado Broncoalveolar , Perros , Femenino , Humanos , Subtipo H3N2 del Virus de la Influenza A , Interferón gamma/inmunología , Interferón gamma/metabolismo , Células de Riñón Canino Madin Darby , Ratones , Ratones Endogámicos BALB C , Plásmidos , Linfocitos T/citología
6.
Diabetologia ; 61(8): 1804-1810, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29789879

RESUMEN

AIMS/HYPOTHESIS: The initial avascular period following islet transplantation seriously compromises graft function and survival. Enhancing graft revascularisation to improve engraftment has been attempted through virus-based delivery of angiogenic triggers, but risks associated with viral vectors have hampered clinical translation. In vitro transcribed mRNA transfection circumvents these risks and may be used for improving islet engraftment. METHODS: Mouse and human pancreatic islet cells were transfected with mRNA encoding the angiogenic growth factor vascular endothelial growth factor A (VEGF-A) before transplantation under the kidney capsule in mice. RESULTS: At day 7 post transplantation, revascularisation of grafts transfected with Vegf-A (also known as Vegfa) mRNA was significantly higher compared with non-transfected or Gfp mRNA-transfected controls in mouse islet grafts (2.11- and 1.87-fold, respectively) (vessel area/graft area, mean ± SEM: 0.118 ± 0.01 [n = 3] in Vegf-A mRNA transfected group (VEGF) vs 0.056 ± 0.01 [n = 3] in no RNA [p < 0.05] vs 0.063 ± 0.02 [n = 4] in Gfp mRNA transfected group (GFP) [p < 0.05]); EndoC-bH3 grafts (2.85- and 2.48-fold. respectively) (0.085 ± 0.02 [n = 4] in VEGF vs 0.030 ± 0.004 [n = 4] in no RNA [p < 0.05] vs 0.034 ± 0.01 [n = 5] in GFP [p < 0.05]); and human islet grafts (3.17- and 3.80-fold, respectively) (0.048 ± 0.013 [n = 3] in VEGF vs 0.015 ± 0.0051 [n = 4] in no RNA [p < 0.01] vs 0.013 ± 0.0046 [n = 4] in GFP [p < 0.01]). At day 30 post transplantation, human islet grafts maintained a vascularisation benefit (1.70- and 1.82-fold, respectively) (0.049 ± 0.0042 [n = 8] in VEGF vs 0.029 ± 0.0052 [n = 5] in no RNA [p < 0.05] vs 0.027 ± 0.0056 [n = 4] in GFP [p < 0.05]) and a higher beta cell volume (1.64- and 2.26-fold, respectively) (0.0292 ± 0.0032 µl [n = 7] in VEGF vs 0.0178 ± 0.0021 µl [n = 5] in no RNA [p < 0.01] vs 0.0129 ± 0.0012 µl [n = 4] in GFP [p < 0.001]). CONCLUSIONS/INTERPRETATION: Vegf-A mRNA transfection before transplantation provides a promising and safe strategy to improve engraftment of islets and other cell-based implants.


Asunto(s)
Células Secretoras de Insulina/citología , Islotes Pancreáticos/citología , Neovascularización Fisiológica , ARN Mensajero/genética , Transfección , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Supervivencia Celular , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/trasplante , Trasplante de Islotes Pancreáticos , Ratones
7.
Int J Cancer ; 143(3): 686-698, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29464699

RESUMEN

Improved understanding of cancer immunology has provided insight into the phenomenon of frequent tumor recurrence after initially successful immunotherapy. A delicate balance exists between the capacity of the immune system to control tumor growth and various resistance mechanisms that arise to avoid or even counteract the host's immune system. These resistance mechanisms include but are not limited to (i) adaptive expression of inhibitory checkpoint molecules in response to the proinflammatory environment and (ii) amplification of cancer stem cells, a small fraction of tumor cells possessing the capacity for self-renewal and mediating treatment resistance and formation of metastases after long periods of clinical remission. Several individual therapeutic agents have so far been developed to revert T-cell exhaustion or disrupt the cross-talk between cancer stem cells and the tumor-promoting microenvironment. Here, we demonstrate that a three-arm combination therapy-consisting of an mRNA-based vaccine to induce antigen-specific T-cell responses, monoclonal antibodies blocking inhibitory checkpoint molecules (PD-1, TIM-3, LAG-3), and antibodies targeting IL-6 and TGF-ß-improves the therapeutic outcome in subcutaneous TC-1 tumors and significantly prolongs survival of treated mice. Our findings point to a need for a rational development of multidimensional anticancer therapies, aiming at the induction of tumor-specific immunity and simultaneously targeting multiple resistance mechanisms.


Asunto(s)
Antineoplásicos Inmunológicos/farmacología , Interleucina-6/antagonistas & inhibidores , Neoplasias/genética , Neoplasias/metabolismo , ARN Mensajero/genética , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Antineoplásicos Inmunológicos/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Inmunoterapia , Interleucina-6/metabolismo , Melanoma Experimental , Ratones , Neoplasias/patología , Neoplasias/terapia , Recurrencia , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
8.
Eur J Immunol ; 45(12): 3351-61, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26377033

RESUMEN

In situ modification of antigen-presenting cells garnered interest in cancer immunotherapy. Therefore, we developed APC-targeted lentiviral vectors (LVs). Unexpectedly, these LVs were inferior vaccines to broad tropism LVs. Since IL-12 is a potent mediator of antitumor immunity, we evaluated whether this proinflammatory cytokine could enhance antitumor immunity of an APC-targeted LV-based vaccine. Therefore, we compared subcutaneous administration of broad tropism LVs (VSV-G-LV) with APC-targeted LVs (DC2.1-LV)-encoding enhanced GFP and ovalbumin, or IL-12 and ovalbumin in mice. We show that codelivery of IL-12 by VSV-G-LVs or DC2.1-LVs augments CD4(+) or CD8(+) T-cell proliferation, respectively. Furthermore, we demonstrate that codelivery of IL-12 enhances the CD4(+) TH 1 profile irrespective of its delivery mode, while an increase in cytotoxic and therapeutic CD8(+) T cells was only induced upon VSV-G-LV injection. While codelivery of IL-12 by DC2.1-LVs did not enhance CD8(+) T-cell performance, it increased expression of inhibitory checkpoint markers Lag3, Tim3, and PD-1. Finally, the discrepancy between CD4(+) T-cell stimulation with and without functional CD8(+) T-cell stimulation by VSV-G- and DC2.1-LVs is partly explained by the observation that IL-12 relieves CD8(+) T cells from CD4(+) T-cell help, implying that a T(H)1 profile is of minor importance for antitumor immunotherapy if IL-12 is exogenously delivered.


Asunto(s)
Interleucina-12/genética , Lentivirus/genética , Transducción Genética , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Femenino , Células HEK293 , Humanos , Activación de Linfocitos , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos C57BL , Proteínas del Envoltorio Viral/genética
9.
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
10.
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
11.
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
12.
Cancer Immunol Immunother ; 63(10): 1023-36, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24947180

RESUMEN

Multiple myeloma (MM) is characterized by a malignant proliferation of plasma cells in the bone marrow with associated organ damage. Although the prognosis of MM has improved recently, the disease remains incurable for the large majority of patients. The eradication of residual disease in the bone marrow is a main target on the road toward cure. Immune cells play a role in the control of cancer and can be tools to attack residual MM cells. However, the myeloma-associated immune deficiency is a major hurdle to immunotherapy. We evaluated ex vivo the effects of low doses of the immunomodulatory drugs (IMiDs) lenalidomide and pomalidomide on several immune cell types from MM patients after autologous stem cell transplantation and with low tumor burden. We observed that these drugs increased CD4(+) and CD8(+) T-cell proliferation and cytokine production, enhanced the lytic capacity of cytotoxic T lymphocytes and reduced the suppressive effects of regulatory T cells on CD8(+) T-cell responses. In addition, we found that functional dendritic cells (DCs) can be generated from mononuclear cells from MM patients. The presence of IMiDs improved the quality of antigen-specific T cells induced or expanded by these DCs as evidenced by a higher degree of T-cell polyfunctionality. Our results provide a rationale for the design of early phase clinical studies to assess the efficacy of DC-based immunotherapy in combination with posttransplant maintenance treatment with IMiDs in MM.


Asunto(s)
Células Dendríticas/inmunología , Trasplante de Células Madre Hematopoyéticas/métodos , Factores Inmunológicos/uso terapéutico , Inmunoterapia Adoptiva/métodos , Mieloma Múltiple/terapia , Talidomida/análogos & derivados , Adulto , Inhibidores de la Angiogénesis/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Proliferación Celular , Dexametasona/administración & dosificación , Doxorrubicina/administración & dosificación , Femenino , Humanos , Inmunomodulación , Lenalidomida , Masculino , Melfalán/administración & dosificación , Persona de Mediana Edad , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/inmunología , Talidomida/uso terapéutico , Acondicionamiento Pretrasplante/métodos , Trasplante Autólogo , Vincristina/administración & dosificación
13.
J Virol ; 87(20): 11304-8, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23864630

RESUMEN

Antigen-presenting cells are a heterogeneous group of cells that are characterized by their functional specialization. Consequently, targeting specific antigen-presenting cell subsets offers opportunities to induce distinct T cell responses. Here we report on the generation and use of nanobodies (Nbs) to target lentivectors specifically to human lymph node-resident myeloid dendritic cells, demonstrating that Nbs represent a powerful tool to redirect lentivectors to human antigen-presenting cell subsets.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Vectores Genéticos/metabolismo , Lentivirus/genética , Lentivirus/metabolismo , Anticuerpos de Dominio Único/metabolismo , Animales , Humanos , Ganglios Linfáticos/inmunología
14.
J Immunol ; 187(9): 4517-29, 2011 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-21948987

RESUMEN

Sirtuins are a unique class of NAD(+)-dependent deacetylases that regulate diverse biological functions such as aging, metabolism, and stress resistance. Recently, it has been shown that sirtuins may have anti-inflammatory activities by inhibiting proinflammatory transcription factors such as NF-κB. In contrast, we report in this study that pharmacological inhibition of sirtuins dampens adaptive Th2 responses and subsequent allergic inflammation by interfering with lung dendritic cell (DC) function in a mouse model of airway allergy. Using genetic engineering, we demonstrate that sirtuin 1 represses the activity of the nuclear receptor peroxisome proliferator-activated receptor-γ in DCs, thereby favoring their maturation toward a pro-Th2 phenotype. This study reveals a previously unappreciated function of sirtuin 1 in the regulation of DC function and Th2 responses, thus shedding new light on our current knowledge on the regulation of inflammatory processes by sirtuins.


Asunto(s)
Asma/inmunología , Células Dendríticas/inmunología , PPAR gamma/antagonistas & inhibidores , Sirtuina 1/fisiología , Células Th2/inmunología , Animales , Asma/enzimología , Asma/patología , Hiperreactividad Bronquial/genética , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/patología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Movimiento Celular/genética , Movimiento Celular/inmunología , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/patología , Femenino , Inmunofenotipificación , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , PPAR gamma/metabolismo , Sirtuina 1/antagonistas & inhibidores , Células Th2/enzimología , Células Th2/patología
15.
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
16.
Clin Immunol ; 142(3): 252-68, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22177848

RESUMEN

In a phase I/IIa clinical trial, 17 HIV-1 infected patients, stable on cART, received 4 vaccinations with autologous dendritic cells electroporated with mRNA encoding Tat, Rev and Nef, after which cART was interrupted. Vaccination was safe and feasible. During the analytical treatment interruption (ATI), no serious adverse events were observed. Ninety-six weeks following ATI, 6/17 patients remained off therapy. Although induced and/or enhanced CD4(+) and CD8(+) T-cell responses specific for the immunogens were observed in most of the patients, we found no correlation with the number of weeks off cART. Moreover, CD4(+) T-cell counts, plasma viral load and the time remaining off cART following ATI did not differ from historical control data. To conclude, the vaccine was safe, well tolerated and resulted in vaccine-specific immune responses. Since no correlation with clinical parameters could be found, these results warrant further research in order to optimize the efficacy of vaccine-induced T-cell responses.


Asunto(s)
Vacunas contra el SIDA/inmunología , Células Dendríticas/inmunología , Infecciones por VIH/terapia , VIH-1/inmunología , Inmunización , Adulto , Anciano , Células Cultivadas , Productos del Gen rev/inmunología , Productos del Gen tat/inmunología , Infecciones por VIH/inmunología , Humanos , Masculino , Persona de Mediana Edad , Productos del Gen nef del Virus de la Inmunodeficiencia Humana/inmunología
17.
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
18.
J Immunol ; 182(2): 860-70, 2009 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19124729

RESUMEN

A20 is a zinc finger protein with ubiquitin-modifying activity. A20 has been described as negatively regulating signaling induced by the TNF receptor and TLR family in a number of cell types, including mouse bone marrow-derived dendritic cells (DCs). However, the expression and effect of A20 in activated human monocyte-derived DCs have not been previously evaluated. We report that DCs activated with the TLR3 ligand poly(I:C) up-regulate A20. Down-regulating A20 demonstrated its role in the functional activation of DCs. A20 down-regulated DCs showed higher activation of the transcription factors NF-kappaB and activator protein-1, which resulted in increased and sustained production of IL-6, IL-10, and IL-12p70. We additionally silenced the immunosuppressive cytokine IL-10 and demonstrated that IL-10 inhibits T cell proliferation. We further demonstrated that A20 down-regulated DCs skew naive CD4+ T cells toward IFN-gamma producing Th1 cells, a process which is dependent on IL-12p70 and which is unaffected by IL-10. Furthermore, A20 and/or IL-10 down-regulated DCs had an enhanced capacity to prime Melan-A/MART-1 specific CD8+ T cells. Finally, we demonstrated that potent T cell stimulatory DCs are generated by the simultaneous delivery of poly(I:C12U), A20, or A20/IL-10 small interfering RNA and Ag-encoding mRNA, introducing a one step approach to improve DC-based vaccines. Together these findings demonstrate that A20 negatively regulates NF-kappaB and activator protein-1 in DCs and that down-regulation of A20 results in DCs with enhanced T cell stimulatory capacity.


Asunto(s)
Adyuvantes Inmunológicos/genética , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Células Dendríticas/trasplante , Regulación hacia Abajo/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Nucleares/genética , ARN Bicatenario/fisiología , Regulación hacia Arriba/inmunología , Adyuvantes Inmunológicos/biosíntesis , Adyuvantes Inmunológicos/fisiología , Linfocitos T CD4-Positivos/inmunología , Células Cultivadas , Técnicas de Cocultivo , Proteínas de Unión al ADN , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Regulación hacia Abajo/genética , Humanos , Interleucina-10/biosíntesis , Interleucina-12/biosíntesis , Interleucina-6/biosíntesis , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Monocitos/inmunología , Monocitos/metabolismo , FN-kappa B/antagonistas & inhibidores , FN-kappa B/biosíntesis , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/biosíntesis , Interferencia de ARN/inmunología , ARN Interferente Pequeño/fisiología , Factor de Transcripción AP-1/antagonistas & inhibidores , Factor de Transcripción AP-1/biosíntesis , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa , Regulación hacia Arriba/genética
19.
J Immunol ; 181(10): 7230-42, 2008 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-18981145

RESUMEN

Dendritic cells (DCs) are professional APCs that have a unique capacity to initiate primary immune responses, including tolerogenic responses. We have genetically engineered bone marrow-derived DCs to express the immunosuppressive cytokine IL-10 and tested the ability of these cells to control experimental asthma. A single intratracheal injection of OVA-pulsed IL-10-transduced DCs (OVA-IL-10-DCs) to naive mice before OVA sensitization and challenge prevented all of the cardinal features of airway allergy, namely, eosinophilic airway inflammation, airway hyperreactivity, and production of mucus, Ag-specific Igs, and IL-4. OVA-IL-10-DCs also reversed established experimental asthma and had long-lasting and Ag-specific effects. We furthermore showed, by using IL-10-deficient mice, that host IL-10 is required for mediating the immunomodulatory effects of OVA-IL-10-DCs and demonstrated a significant increase in the percentage of OVA-specific CD4(+)CD25(+)Foxp3(+)IL-10(+) regulatory T cells in the mediastinal lymph nodes of OVA-IL-10-DC-injected mice. Finally, adoptive transfer of CD4(+) mediastinal lymph node T cells from mice injected with OVA-IL-10-DCs protected OVA-sensitized recipients from airway eosinophilia upon OVA provocation. Our study describes a promising strategy to induce long-lasting Ag-specific tolerance in airway allergy.


Asunto(s)
Asma/metabolismo , Células Dendríticas/metabolismo , Ingeniería Genética/métodos , Tolerancia Inmunológica , Interleucina-10/metabolismo , Traslado Adoptivo , Animales , Antígenos/inmunología , Apoptosis/inmunología , Asma/inmunología , Asma/terapia , Células Cultivadas , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Inmunoterapia/métodos , Interleucina-10/genética , Ratones , Ratones Noqueados , Ovalbúmina/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T Reguladores/inmunología , Transducción Genética
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA