Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Viruses ; 14(4)2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35458477

RESUMEN

Swine influenza (SI) is a major respiratory disease of swine; SI is due to the influenza A virus of swine (IAV-S), a highly contagious virus with zoonotic potential. The intensity of IAV-S surveillance varies among countries because it is not a reportable disease and causes limited mortality in swine. Although Asia accounts for half of all pig production worldwide, SI is not well managed in those countries. Rigorously managing SI on pig farms could markedly reduce the economic losses, the likelihood of novel reassortants among IAV-S, and the zoonotic IAV-S infections in humans. Vaccination of pigs is a key control measure for SI, but its efficacy relies on the optimal antigenic matching of vaccine strains with the viral strains circulating in the field. Here, we phylogenetically reviewed the genetic diversity of the hemagglutinin gene among IAVs-S that have circulated in Asia during the last decade. This analysis revealed the existence of country-specific clades in both the H1 and H3 subtypes and cross-border transmission of IAVs-S. Our findings underscore the importance of choosing vaccine antigens for each geographic region according to both genetic and antigenic analyses of the circulating IAV-S to effectively manage SI in Asia.


Asunto(s)
Virus de la Influenza A , Gripe Humana , Infecciones por Orthomyxoviridae , Enfermedades de los Porcinos , Animales , Variación Genética , Hemaglutininas/genética , Humanos , Subtipo H3N2 del Virus de la Influenza A/genética , Virus de la Influenza A/genética , Filogenia , Porcinos
2.
Expert Opin Biol Ther ; 19(7): 721-733, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31286786

RESUMEN

Objective: We report the characterization of MCLA-117, a novel T cell-redirecting antibody for acute myeloid leukaemia (AML) treatment targeting CD3 on T cells and CLEC12A on leukaemic cells. In AML, CLEC12A is expressed on blasts and leukaemic stem cells. Methods: The functional capacity of MCLA-117 to redirect resting T cells to eradicate CLEC12APOS tumor cells was studied using human samples, including primary AML samples. Results: Within the normal hematopoietic compartment, MCLA-117 binds to cells expressing CD3 and CLEC12A but not to early myeloid progenitors or hematopoietic stem cells. MCLA-117 induces T cell activation (EC50 = 44 ng/mL), T cell proliferation, mild pro-inflammatory cytokine release, and redirects T cells to lyse CLEC12APOS target cells (EC50 = 68 ng/mL). MCLA-117-induced targeting of normal CD34POS cells co-cultured with T cells spares erythrocyte and megakaryocyte differentiation as well as preserves mono-myelocytic lineage development. In primary AML patient samples with autologous T cells, MCLA-117 robustly induced AML blast killing (23-98%) at low effector-to-target ratios (1:3-1:97). Conclusion: These findings demonstrate that MCLA-117 efficiently redirects T cells to kill tumour cells while sparing the potential of the bone marrow to develop the full hematological compartment and support further clinical evaluation as a potentially potent treatment option for AML.


Asunto(s)
Anticuerpos Biespecíficos/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Linfocitos T/inmunología , Animales , Anticuerpos Biespecíficos/metabolismo , Anticuerpos Biespecíficos/farmacocinética , Complejo CD3/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Citocinas/análisis , Citocinas/metabolismo , Células HL-60 , Semivida , Humanos , Lectinas Tipo C/inmunología , Leucemia Mieloide Aguda/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Receptores Mitogénicos/inmunología , Linfocitos T/metabolismo
3.
Clin Cancer Res ; 23(15): 4107-4118, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28280089

RESUMEN

Purpose: Older acute myeloid leukemia (AML) patients have a poor prognosis; therefore, novel therapies are needed. Allogeneic natural killer (NK) cells have been adoptively transferred with promising clinical results. Here, we report the first-in-human study exploiting a unique scalable NK-cell product generated ex vivo from CD34+ hematopoietic stem and progenitor cells (HSPC) from partially HLA-matched umbilical cord blood units.Experimental Design: Ten older AML patients in morphologic complete remission received an escalating HSPC-NK cell dose (between 3 and 30 × 106/kg body weight) after lymphodepleting chemotherapy without cytokine boosting.Results: HSPC-NK cell products contained a median of 75% highly activated NK cells, with <1 × 104 T cells/kg and <3 × 105 B cells/kg body weight. HSPC-NK cells were well tolerated, and neither graft-versus-host disease nor toxicity was observed. Despite no cytokine boosting being given, transient HSPC-NK cell persistence was clearly found in peripheral blood up to 21% until day 8, which was accompanied by augmented IL15 plasma levels. Moreover, donor chimerism up to 3.5% was found in bone marrow. Interestingly, in vivo HSPC-NK cell maturation was observed, indicated by the rapid acquisition of CD16 and KIR expression, while expression of most activating receptors was sustained. Notably, 2 of 4 patients with minimal residual disease (MRD) in bone marrow before infusion became MRD negative (<0.1%), which lasted for 6 months.Conclusions: These findings indicate that HSPC-NK cell adoptive transfer is a promising, potential "off-the-shelf" translational immunotherapy approach in AML. Clin Cancer Res; 23(15); 4107-18. ©2017 AACR.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Trasplante de Células Madre Hematopoyéticas/métodos , Leucemia Mieloide Aguda/terapia , Anciano , Antígenos CD34/genética , Antígenos CD34/inmunología , Trasplante de Células Madre de Sangre del Cordón Umbilical/efectos adversos , Femenino , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Humanos , Interleucina-15/sangre , Células Asesinas Naturales/trasplante , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/patología , Masculino , Regresión Neoplásica Espontánea/patología , Pronóstico
4.
J Immunother ; 37(5): 267-77, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24810638

RESUMEN

Autologous, patient-specific, monocyte-derived dendritic cell (MoDC) vaccines have been successfully applied in the clinical studies so far. However, the routine application of this strategy has been hampered by the difficulties in generating sufficient numbers of DC and the poor DC vaccine quality because of pathology or prior treatment received by the patients. The immunotherapeutic potential of other subsets of DC has not been thoroughly investigated because of their rarity in tissues and difficulties associated with their ex vivo generation. The high expansion and differentiation potential of CD34 hematopoietic progenitor cells (HPC), isolated from umbilical cord blood (UCB), into different DC subsets make them an attractive alternative DC source for cancer immunotherapy. Therefore, the aim of this study was to generate a large number of different DC subsets from CD34 HPC and evaluate their functionality in comparison with MoDC. Our culture protocol generated a clinically relevant number of mature CD1a myeloid DC and CD207 Langerhans cells (LC)-like DC subsets from CD34 HPC with >95% purity. Both DC subsets exhibited a cytokine profile that favors cytotoxic T-cell responses. Furthermore, UCB-DC and UCB-LC demonstrated superior induction of proliferation of both allogeneic as well as viral antigen-specific CD8 T cells, both in vitro and in vivo. Additional studies revealed that UCC-DC and UCB-LC can efficiently expand minor histocompatibility antigen (MiHA) HA-1-specific cytotoxic T cells in the peripheral blood of leukemia patients and prime MiHA HA-1-specific and HA-2-specific cytotoxic T cells in vitro. These preclinical findings support the pharmaceutical development of the described culture protocol for clinical evaluation.


Asunto(s)
Vacunas contra el Cáncer , Células Dendríticas/fisiología , Neoplasias Hematológicas/terapia , Células Madre Hematopoyéticas/fisiología , Inmunoterapia/métodos , Monocitos/fisiología , Linfocitos T Citotóxicos/inmunología , Antígenos CD/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Citocinas/metabolismo , Células Dendríticas/trasplante , Neoplasias Hematológicas/inmunología , Humanos , Activación de Linfocitos , Antígenos de Histocompatibilidad Menor/inmunología
5.
Stem Cells Dev ; 23(9): 955-67, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24325394

RESUMEN

The superiority of dendritic cells (DCs) as antigen-presenting cells has been exploited in numerous clinical trials, where generally monocyte-derived DCs (Mo-DCs) are injected to induce immunity in patients with cancer or infectious diseases. Despite promising expansion of antigen-specific T cells, the clinical responses following vaccination have been limited, indicating that further improvements of DC vaccine potency are necessary. Pre-clinical studies suggest that vaccination with combination of primary DC subsets, such as myeloid and plasmacytoid blood DCs (mDCs and pDCs, respectively), may result in stronger clinical responses. However, it is a challenge to obtain high enough numbers of primary DCs for immunotherapy, since their frequency in blood is very low. We therefore explored the possibility to generate them from hematopoietic progenitor cells (HPCs). Here, we show that by inhibiting the aryl hydrocarbon receptor with its antagonist StemRegenin 1 (SR1), clinical-scale numbers of functional BDCA2(+)BDCA4(+) pDCs, BDCA1(+) mDCs, and BDCA3(+)DNGR1(+) mDCs can be efficiently generated from human CD34(+) HPCs. The ex vivo-generated DCs were phenotypically and functionally comparable to peripheral blood DCs. They secreted high levels of pro-inflammatory cytokines such as interferon (IFN)-α, interleukin (IL)-12, and tumor necrosis factor (TNF)-α and upregulated co-stimulatory molecules and maturation markers following stimulation with Toll-like receptor (TLR) ligands. Further, they induced potent allogeneic T-cell responses and activated antigen-experienced T cells. These findings demonstrate that SR1 can be exploited to generate high numbers of functional pDCs and mDCs from CD34(+) HPCs, providing an alternative option to Mo-DCs for immunotherapy of patients with cancer or infections.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Células Dendríticas/metabolismo , Sangre Fetal/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Mieloides/metabolismo , Células Plasmáticas/metabolismo , Purinas/farmacología , Receptores de Hidrocarburo de Aril/antagonistas & inhibidores , Antígenos de Diferenciación/metabolismo , Células Dendríticas/citología , Femenino , Sangre Fetal/citología , Células Madre Hematopoyéticas/citología , Humanos , Masculino , Células Mieloides/citología , Células Plasmáticas/citología , Receptores de Hidrocarburo de Aril/metabolismo
6.
PLoS One ; 8(6): e64384, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23755121

RESUMEN

Natural killer (NK) cell-based adoptive immunotherapy is an attractive adjuvant treatment option for patients with acute myeloid leukemia. Recently, we reported a clinical-grade, cytokine-based culture method for the generation of NK cells from umbilical cord blood (UCB) CD34⁺ hematopoietic progenitor cells with high yield, purity and in vitro functionality. The present study was designed to evaluate the in vivo anti-leukemic potential of UCB-NK cells generated with our GMP-compliant culture system in terms of biodistribution, survival and cytolytic activity following adoptive transfer in immunodeficient NOD/SCID/IL2Rg(null) mice. Using single photon emission computed tomography, we first demonstrated active migration of UCB-NK cells to bone marrow, spleen and liver within 24 h after infusion. Analysis of the chemokine receptor expression profile of UCB-NK cells matched in vivo findings. Particularly, a firm proportion of UCB-NK cells functionally expressed CXCR4, what could trigger BM homing in response to its ligand CXCL12. In addition, high expression of CXCR3 and CCR6 supported the capacity of UCB-NK cells to migrate to inflamed tissues via the CXCR3/CXCL10-11 and CCR6/CCL20 axis. Thereafter, we showed that low dose IL-15 mediates efficient survival, expansion and maturation of UCB-NK cells in vivo. Most importantly, we demonstrate that a single UCB-NK cell infusion combined with supportive IL-15 administration efficiently inhibited growth of human leukemia cells implanted in the femur of mice, resulting in significant prolongation of mice survival. These preclinical studies strongly support the therapeutic potential of ex vivo-generated UCB-NK cells in the treatment of myeloid leukemia after immunosuppressive chemotherapy.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Células Asesinas Naturales/trasplante , Leucemia Mieloide Aguda/terapia , Traslado Adoptivo , Animales , Médula Ósea/patología , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Rastreo Celular , Células Cultivadas , Sangre Fetal/citología , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Interleucina-15/farmacología , Interleucina-15/fisiología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Leucemia Mieloide Aguda/inmunología , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Trasplante de Neoplasias , Especificidad de Órganos , Receptores Mensajeros de Linfocitos/metabolismo , Transcriptoma
7.
J Gene Med ; 14(6): 416-27, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22228547

RESUMEN

The ability of dendritic cells (DCs) to orchestrate innate and adaptive immune responses has been exploited to develop potent anti-cancer immunotherapies. Recent clinical trials exploring the efficacy of ex vivo modified autologous DC-based vaccines have reported some promising results. However, in vitro generation of autologous DCs for clinical administration, their loading with tumor associated antigens (TAAs) and their activation, is laborious and expensive, and, as a result of inter-individual variability in the personalized vaccines, remains poorly standardized. An attractive alternative approach is to load resident DCs in vivo by targeted delivery of TAAs, using viral vectors and activating them simultaneously. To this end, we have constructed genetically-modified adenoviral (Ad) vectors and bispecific adaptor molecules to retarget Ad vectors encoding TAAs to the CD40 receptor on DCs. Pre-clinical human and murine studies conducted so far have clearly demonstrated the suitability of a 'two-component' (i.e. Ad and adaptor molecule) configuration for targeted modification of DCs in vivo for cancer immunotherapy. This review summarizes recent progress in the development of CD40-targeted Ad-based cancer vaccines and highlights pre-clinical issues in the clinical translation of this approach.


Asunto(s)
Antígenos CD40 , Vacunas contra el Cáncer/genética , Células Dendríticas/inmunología , Neoplasias/terapia , Adenoviridae/genética , Animales , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Antígenos CD40/genética , Antígenos CD40/inmunología , Vacunas contra el Cáncer/inmunología , Técnicas de Transferencia de Gen , Vectores Genéticos , Humanos , Inmunoterapia , Activación de Linfocitos , Ratones
8.
Cancer Res ; 71(17): 5827-37, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21747119

RESUMEN

In situ delivery of tumor-associated antigen (TAA) genes into dendritic cells (DC) has great potential as a generally applicable tumor vaccination approach. Although adenoviruses (Ad) are an attractive vaccine vehicle in this regard, Ad-mediated transduction of DCs is hampered by the lack of expression of the Ad receptor CAR on the DC surface. DC activation also requires interaction of CD40 with its ligand CD40L to generate protective T-cell-mediated tumor immunity. Therefore, to create a strategy to target Ads to DCs in vivo, we constructed a bispecific adaptor molecule with the CAR ectodomain linked to the CD40L extracellular domain via a trimerization motif (CFm40L). By targeting Ad to CD40 with the use of CFm40L, we enhanced both transduction and maturation of cultured bone marrow-derived DCs. Moreover, we improved transduction efficiency of DCs in lymph node and splenic cell suspensions in vitro and in skin and vaccination site-draining lymph nodes in vivo. Furthermore, CD40 targeting improved the induction of specific CD8(+) T cells along with therapeutic efficacy in a mouse model of melanoma. Taken together, our findings support the use of CD40-targeted Ad vectors encoding full-length TAA for in vivo targeting of DCs and high-efficacy induction of antitumor immunity.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/inmunología , Antígenos CD40/inmunología , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Melanoma Experimental/prevención & control , Neoplasias Cutáneas/prevención & control , Linfocitos T/inmunología , Adenoviridae , Animales , Antígenos de Carbohidratos Asociados a Tumores/genética , Ligando de CD40/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Ganglios Linfáticos/inmunología , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Neoplasias Cutáneas/inmunología , Transducción Genética
9.
Vaccine ; 29(12): 2313-21, 2011 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-21272606

RESUMEN

Adenovirus (Ad)-based vaccines are considered for cancer immunotherapy, yet, detailed knowledge on their mechanism of action and optimal delivery route for anti-tumor efficacy is lacking. Here, we compared the anti-tumor efficacy of an Ad-based melanoma vaccine after intradermal, intravenous, intranasal or intraperitoneal delivery in the B16F10 melanoma model. The intradermal route induced superior systemic anti-melanoma immunity which was MyD88 signaling-dependent. Predominant transduction of non-professional antigen-presenting cells at the dermal vaccination sites and draining lymph nodes, suggested a role for cross-presentation, which was confirmed in vitro. We conclude that the dermis provides an optimal route of entry for Ad-based vaccines for high-efficacy systemic anti-tumor immunization and that this immunization likely involves cross-priming events in the draining lymph nodes.


Asunto(s)
Vacunas contra el Cáncer/administración & dosificación , Reactividad Cruzada , Melanoma Experimental/terapia , Factor 88 de Diferenciación Mieloide/metabolismo , Adenoviridae/genética , Animales , Células Presentadoras de Antígenos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Células Dendríticas/inmunología , Inyecciones Intradérmicas , Inyecciones Intravenosas , Ganglios Linfáticos/inmunología , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
10.
J Immunother ; 33(7): 706-15, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20664356

RESUMEN

Targeted delivery of tumor antigen genes to dendritic cells (DCs) using adenoviral (Ad) vectors holds great potential for cancer immunotherapy. We previously showed that CD40 targeting of Ad vectors enhanced specific transduction of DC in human skin, while simultaneously ensuring their stable maturation and superior allogeneic T-cell stimulatory capacity. In this study, we evaluated whether CD40-targeted Ad encoding the full-length melanoma antigen recognized by T cells-1 (CD40-Ad-MART-1) could be used to efficiently and selectively transduce conventional and plasmacytoid DC to prime melanoma-specific CD8(+) T-effector cells in human melanoma-draining sentinel lymph nodes (SLNs). CD40 targeting of Ad was achieved using a bispecific fusion protein, binding and neutralizing the Ad fiber knob through soluble coxsackie and adenovirus receptor while retargeting the virus to hCD40 through the tumor necrosis factor-like domain of mCD40L. Selective transduction of conventional and plasmacytoid DC subsets by CD40-Ad was observed in suspensions of human melanoma-draining SLN. Moreover, CD40-Ad-MART-1 enhanced the expansion of functional MART-1-specific CD8(+) T cells from SLN with concomitant decreases in CD4:CD8 T-cell ratios and CD4(+)CD25(hi)FoxP3(+) regulatory T-cell rates. Additional studies revealed that transduction and activation of monocyte-derived DCs with CD40-Ad-MART-1 significantly enhanced their priming efficiency of functional CD8(+) effector T cells with high avidity. These findings provide preclinical evidence of possible efficacy of this approach for cancer immunotherapy.


Asunto(s)
Adenoviridae/genética , Antígenos CD40/metabolismo , Células Dendríticas/metabolismo , Inmunoterapia , Melanoma/terapia , Linfocitos T Citotóxicos/metabolismo , Antígenos CD4/biosíntesis , Ligando de CD40/genética , Ligando de CD40/inmunología , Ligando de CD40/metabolismo , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Células Dendríticas/patología , Factores de Transcripción Forkhead/biosíntesis , Humanos , Interferón gamma/metabolismo , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Células K562 , Ganglios Linfáticos/patología , Antígeno MART-1/genética , Antígeno MART-1/inmunología , Antígeno MART-1/metabolismo , Melanoma/inmunología , Ingeniería de Proteínas , Proteínas Recombinantes de Fusión/genética , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología , Transducción Genética
11.
Dis Aquat Organ ; 78(1): 13-22, 2007 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-18159668

RESUMEN

A clinical infection in post-larval (glass) European eels Anguilla anguilla was successfully induced after artificial bath immersion with Herpesvirus anguillae (HVA), isolated from diseased European eel. HVA caused a clinical infection after 7 d post-inoculation (pi); virus was detected by polymerase chain reaction (PCR) from Day 1 pi; virus isolation was positive from Day 7 pi, and HVA antigen was detected by immunohistochemistry in gills and stomach from Day 4 pi. Tissue changes were found by histological examination in gills and skin from Day 4 pi. In general, there was good correlation in the timing of the clinical signs, PCR, virus isolation, immunohistochemistry and histopathology results, although PCR, histopathology and immunohistochemistry were the first positive tests. HVA was first detected in skin and stomach, followed by gills, and later heart and intestine, whereas HVA was detected persistently in gills and skin. Koch's postulates were fulfilled. For diagnosis of HVA infections, clinical pathology combined with virus isolation and/or PCR are recommended.


Asunto(s)
Anguilla , Enfermedades de los Peces/virología , Infecciones por Herpesviridae/veterinaria , Herpesviridae/crecimiento & desarrollo , Animales , ADN Viral/química , ADN Viral/genética , Enfermedades de los Peces/patología , Branquias/patología , Branquias/virología , Herpesviridae/genética , Infecciones por Herpesviridae/patología , Infecciones por Herpesviridae/virología , Inmunohistoquímica/veterinaria , Reacción en Cadena de la Polimerasa/veterinaria , Piel/patología , Piel/virología , Estómago/patología , Estómago/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...