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










Base de datos
Intervalo de año de publicación
1.
Exp Ther Med ; 18(5): 3271-3280, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31602200

RESUMEN

Sepsis is a highly complex and often fatal syndrome which varies widely in its clinical manifestations, and therapies that target the underlying uncontrolled immune status in sepsis are needed. The failure of preclinical approaches to provide significant sepsis survival benefit in the clinic is often attributed to inappropriate animal disease models. It has been demonstrated that high mobility group box protein 1 (HMGB1) blockade can reduce inflammation, mortality and morbidity in experimental sepsis without promoting immunosuppression. Within this study, we explored the use of ovine anti-HMGB1 antibodies in a model of ovine septic shock incorporating intensive care supports (OSSICS). Results: Septic sheep exhibited elevated levels of HMGB1 within 12 h after the induction of sepsis. In this study, sepsis was induced in six anaesthetized adult Border Leicester × Merino ewes via intravenous instillation of E. coli and sheep monitored according to intensive care unit standard protocols for 26 h, with the requirement for noradrenaline as the primary endpoint. Septic sheep exhibited a hyperdynamic circulation, renal dysfunction, deranged coagulation profile and severe metabolic acidosis. Sheep were assigned a severity of illness score, which increased over time. While a therapeutic effect of intravenous anti-HMGB1 antibody could not be observed in this model due to limited animal numbers, a reduced bacterial dose induced a septic syndrome of much lower severity. With modifications including a reduced bacterial dose, a longer timeframe and broad spectrum antibiotics, the OSSICS model may become a robust tool for preclinical assessment of sepsis therapeutics.

2.
Mol Ther Methods Clin Dev ; 11: 65-72, 2018 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-30397628

RESUMEN

Pre-existing neutralizing antibody (NAb) against adeno-associated virus (AAV) commonly found in primates is a major host barrier that can severely compromise in vivo gene transfer by AAV vectors. To achieve proof-of-concept success in clinical development of recombinant AAV (rAAV)-based in vivo gene therapy, it is crucial to consider the potential interference of NAb and to enroll serologically compatible study subjects. In this study, we report a large AAV NAb dataset comprising multiple large animal species and AAV serotypes and compare two NAb assays based on in vitro or in vivo transduction inhibition, respectively. Together with previously published AAV seroepidemiology studies, these data can serve as a reference for selecting suitable serotypes, study subjects of large animal species, and potentially human patients for rAAV treatment. In addition, we modeled the intrathalamus rAAV9 delivery in the presence of circulating anti-AAV9 NAb generated by either pre-immunization or passive transfer of NAb-positive large animal serum to mice. The data showed that circulating NAb may not be the sole determinant to inhibit brain transduction. Other aspects of pre-existing AAV immunity following natural infection or rAAV administration may be further studied to establish a more accurate inclusion criterion for clinical studies employing intraparenchymal rAAV9 injections.

3.
J Nanobiotechnology ; 16(1): 38, 2018 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-29653579

RESUMEN

BACKGROUND: Multidrug resistance-associated protein 1 (MRP1) overexpression plays a major role in chemoresistance in glioblastoma multiforme (GBM) contributing to its notorious deadly nature. Although MRP1-siRNA transfection to GBM in vitro has been shown to sensitise the cells to drug, MRP1 silencing in vivo and the phenotypic influence on the tumour and normal tissues upon MRP1 down-regulation have not been established. Here, porous silicon nanoparticles (pSiNPs) that enable high-capacity loading and delivery of siRNA are applied in vitro and in vivo. RESULT: We established pSiNPs with polyethyleneimine (PEI) capping that enables high-capacity loading of siRNA (92 µg of siRNA/mg PEI-pSiNPs), and optimised release profile (70% released between 24 and 48 h). These pSiNPs are biocompatible, and demonstrate cellular uptake and effective knockdown of MRP1 expression in GBM by 30%. Also, siRNA delivery was found to significantly reduce GBM proliferation as an associated effect. This effect is likely mediated by the attenuation of MRP1 transmembrane transport, followed by cell cycle arrest. MRP1 silencing in GBM tumour using MRP1-siRNA loaded pSiNPs was demonstrated in mice (82% reduction at the protein level 48 h post-injection), and it also produced antiproliferative effect in GBM by reducing the population of proliferative cells. These results indicate that in vitro observations are translatable in vivo. No histopathological signs of acute damage were observed in other MRP1-expressing organs despite collateral downregulations. CONCLUSIONS: This study proposes the potential of efficient MRP1-siRNA delivery by using PEI-capped pSiNPs in achieving a dual therapeutic role of directly attenuating the growth of GBM while sensitising residual tumour cells to the effects of chemotherapy post-resection.


Asunto(s)
Silenciador del Gen , Glioblastoma/patología , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo , Nanopartículas/química , Polietileneimina/química , ARN Interferente Pequeño/administración & dosificación , Silicio/química , Animales , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Ratones Desnudos , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/antagonistas & inhibidores , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/genética , Nanopartículas/ultraestructura , Especificidad de Órganos , Fenotipo , Porosidad , Propionatos/farmacología , Quinolinas/farmacología
4.
Hum Gene Ther ; 29(6): 663-673, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29207890

RESUMEN

Huntington's disease (HD) is a fatal neurodegenerative disease caused by a genetic expansion of the CAG repeat region in the huntingtin (HTT) gene. Studies in HD mouse models have shown that artificial miRNAs can reduce mutant HTT, but evidence for their effectiveness and safety in larger animals is lacking. HD transgenic sheep express the full-length human HTT with 73 CAG repeats. AAV9 was used to deliver unilaterally to HD sheep striatum an artificial miRNA targeting exon 48 of the human HTT mRNA under control of two alternative promoters: U6 or CßA. The treatment reduced human mutant (m) HTT mRNA and protein 50-80% in the striatum at 1 and 6 months post injection. Silencing was detectable in both the caudate and putamen. Levels of endogenous sheep HTT protein were not affected. There was no significant loss of neurons labeled by DARPP32 or NeuN at 6 months after treatment, and Iba1-positive microglia were detected at control levels. It is concluded that safe and effective silencing of human mHTT protein can be achieved and sustained in a large-animal brain by direct delivery of an AAV carrying an artificial miRNA.


Asunto(s)
Proteína Huntingtina/metabolismo , Enfermedad de Huntington/genética , Enfermedad de Huntington/patología , MicroARNs/metabolismo , Proteínas Mutantes/metabolismo , Neostriado/metabolismo , Animales , Animales Modificados Genéticamente , Dependovirus/genética , Modelos Animales de Enfermedad , Electrólitos/metabolismo , Vectores Genéticos/metabolismo , Genoma Viral , Humanos , Inmunoensayo , Inyecciones , Riñón/fisiopatología , Hígado/fisiopatología , MicroARNs/genética , Microglía/metabolismo , Neuronas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ovinos
5.
Sci Rep ; 7(1): 5850, 2017 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-28724977

RESUMEN

Sepsis remains a significant health burden and a major clinical need exists for therapeutics to dampen the excessive and uncontrolled immune activation. Nuclear protein high mobility group box protein 1 (HMGB1) is released following cell death and is a late mediator in sepsis pathogenesis. While approaches targeting HMGB1 have demonstrated reduced mortality in pre-clinical models of sepsis, the impact of HMGB1 blockade on the complex septic inflammatory milieu and the development of subsequent immunosuppression remain enigmatic. Analysis of plasma samples obtained from septic shock patients established an association between increased HMGB1 and non-survival, higher APACHE II scores, and increased pro-inflammatory cytokine responses. Pre-clinically, administration of neutralising ovine anti-HMGB1 polyclonal antibodies improved survival in murine endotoxaemia and caecal ligation and puncture-induced sepsis models, and altered early cytokine profiles to one which corresponded to patterns observed in the surviving patient cohort. Additionally, anti-HMGB1 treated murine sepsis survivors were significantly more resistant to secondary bacterial infection and exhibited altered innate immune cell phenotypes and cytokine responses. These findings demonstrate that anti-HMGB1 antibodies alter inflammation in murine sepsis models and reduce sepsis mortality without potentiating immunosuppression.


Asunto(s)
Citocinas/sangre , Proteína HMGB1/sangre , Inflamación/patología , Terapia Molecular Dirigida , Sepsis/sangre , Anciano , Animales , Ciego/patología , Estudios de Cohortes , Células Dendríticas/metabolismo , Células Dendríticas/patología , Modelos Animales de Enfermedad , Endotoxemia/sangre , Endotoxemia/patología , Femenino , Humanos , Inflamación/sangre , Ligadura , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Pseudomonas aeruginosa/fisiología , Punciones , Ovinos , Choque Séptico/mortalidad , Análisis de Supervivencia , Resultado del Tratamiento
6.
Sci Rep ; 7: 43030, 2017 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-28223686

RESUMEN

The pronounced cachexia (unexplained wasting) seen in Huntington's disease (HD) patients suggests that metabolic dysregulation plays a role in HD pathogenesis, although evidence of metabolic abnormalities in HD patients is inconsistent. We performed metabolic profiling of plasma from presymptomatic HD transgenic and control sheep. Metabolites were quantified in sequential plasma samples taken over a 25 h period using a targeted LC/MS metabolomics approach. Significant changes with respect to genotype were observed in 89/130 identified metabolites, including sphingolipids, biogenic amines, amino acids and urea. Citrulline and arginine increased significantly in HD compared to control sheep. Ten other amino acids decreased in presymptomatic HD sheep, including branched chain amino acids (isoleucine, leucine and valine) that have been identified previously as potential biomarkers of HD. Significant increases in urea, arginine, citrulline, asymmetric and symmetric dimethylarginine, alongside decreases in sphingolipids, indicate that both the urea cycle and nitric oxide pathways are dysregulated at early stages in HD. Logistic prediction modelling identified a set of 8 biomarkers that can identify 80% of the presymptomatic HD sheep as transgenic, with 90% confidence. This level of sensitivity, using minimally invasive methods, offers novel opportunities for monitoring disease progression in HD patients.


Asunto(s)
Biomarcadores/metabolismo , Enfermedad de Huntington/patología , Metabolómica , Aminoácidos de Cadena Ramificada/metabolismo , Animales , Animales Modificados Genéticamente/genética , Área Bajo la Curva , Arginina/análogos & derivados , Arginina/metabolismo , Citrulina/metabolismo , Genotipo , Enfermedad de Huntington/metabolismo , Enfermedad de Huntington/veterinaria , Modelos Logísticos , Curva ROC , Ovinos
7.
Sci Rep ; 6: 29154, 2016 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-27380890

RESUMEN

Passive immunotherapy may have particular benefits for the treatment of severe influenza infection in at-risk populations, however little is known of the impact of passive immunotherapy on the formation of memory responses to the virus. Ideally, passive immunotherapy should attenuate the severity of infection while still allowing the formation of adaptive responses to confer protection from future exposure. In this study, we sought to determine if administration of influenza-specific ovine polyclonal antibodies could inhibit adaptive immune responses in a murine model of lethal influenza infection. Ovine polyclonal antibodies generated against recombinant PR8 (H1N1) hemagglutinin exhibited potent prophylactic capacity and reduced lethality in an established influenza infection, particularly when administered intranasally. Surviving mice were also protected against reinfection and generated normal antibody and cytotoxic T lymphocyte responses to the virus. The longevity of ovine polyclonal antibodies was explored with a half-life of over two weeks following a single antibody administration. These findings support the development of an ovine passive polyclonal antibody therapy for treatment of severe influenza infection which does not affect the formation of subsequent acquired immunity to the virus.


Asunto(s)
Inmunidad Adaptativa , Anticuerpos Antivirales/inmunología , Inmunoterapia , Subtipo H1N1 del Virus de la Influenza A/fisiología , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/terapia , Animales , Reacciones Cruzadas/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Cinética , Ratones Endogámicos BALB C , Pruebas de Neutralización , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/virología , Ovinos
8.
Biomaterials ; 74: 217-30, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26466356

RESUMEN

Advanced biosensors in future medicine hinge on the evolvement of biomaterials. Porous silicon (pSi), a generally biodegradable and biocompatible material that can be fabricated to include environment-responsive optical characteristics, is an excellent candidate for in vivo biosensors. However, the feasibility of using this material as a subcutaneously implanted optical biosensor has never been demonstrated. Here, we investigated the stability and biocompatibility of a thermally-hydrocarbonised (THC) pSi optical rugate filter, and demonstrated its optical functionality in vitro and in vivo. We first compared pSi films with different surface chemistries and observed that the material was cytotoxic despite the outstanding stability of the THC pSi films. We then showed that the cytotoxicity correlates with reactive oxygen species levels, which could be mitigated by pre-incubation of THC pSi (PITHC pSi). PITHC pSi facilitates normal cellular phenotypes and is biocompatible in vivo. Importantly, the material also possesses optical properties capable of responding to microenvironmental changes that are readable non-invasively in cell culture and subcutaneous settings. Collectively, we demonstrate, for the first time, that PITHC pSi rugate filters are both biocompatible and optically functional for lab-on-a-chip and subcutaneous biosensing scenarios. We believe that this study will deepen our understanding of cell-pSi interactions and foster the development of implantable biosensors.


Asunto(s)
Técnicas Biosensibles , Hidrocarburos/química , Silicio/química , Piel , Células 3T3 , Animales , Ratones , Microscopía Electrónica de Rastreo , Porosidad
9.
J Immunother ; 38(1): 12-23, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25415284

RESUMEN

Cancer immunotherapy has long been used in the treatment of metastatic melanoma, and an anti-CTLA-4 monoclonal antibody treatment has recently been approved by the US Food and Drug Administration. Targeted therapies such as small molecule kinase inhibitors targeting deregulated mitogen-activated protein kinase (MAPK) signaling have markedly improved melanoma control in up to 50% of metastatic disease patients and have likewise been recently approved. Combination therapies for melanoma have been proposed as a way to exploit the high-level but short-term responses associated with kinase inhibitor therapies and the low-level but longer-term responses associated with immunotherapy. Cancer immunotherapy now includes adoptive transfer of autologous tumor-specific chimeric antigen receptor (CAR) T cells and this mode of therapy is a candidate for combination with small molecule drugs. This paper describes CART cells that target GD2-expressing melanoma cells and investigates the effects of approved MAPK pathway-targeted therapies for melanoma [vemurafenib (Vem), dabrafenib (Dab), and trametinib (Tram)] on the viability, activation, proliferation, and cytotoxic T lymphocyte activity of these CAR T cells, as well as on normal peripheral blood mononuclear cells. We report that, although all these drugs lead to inhibition of stimulated T cells at high concentrations in vitro, only Vem inhibited T cells at concentrations equivalent to reported plasma concentrations in treated patients. Although the combination of Dab and Tram also resulted in inhibition of T-cell effector functions at some therapeutic concentrations, Dab itself had little adverse effect on CAR T-cell function. These findings may have implications for novel therapeutic combinations of adoptive CAR T-cell immunotherapy and MAPK pathway inhibitors.


Asunto(s)
Antineoplásicos/farmacología , Activación de Linfocitos/efectos de los fármacos , Melanoma/inmunología , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Antígenos de Linfocitos T/inmunología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citometría de Flujo , Humanos , Imidazoles/farmacología , Inmunoterapia/métodos , Técnicas In Vitro , Indoles/farmacología , Leucocitos Mononucleares/inmunología , Activación de Linfocitos/inmunología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Oximas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Piridonas/farmacología , Pirimidinonas/farmacología , Sulfonamidas/farmacología , Linfocitos T Citotóxicos , Transfección , Vemurafenib
10.
EJNMMI Res ; 4(1): 2, 2014 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-24387284

RESUMEN

BACKGROUND: The lupus-associated (La)-specific murine monoclonal antibody DAB4 (APOMAB®) specifically binds dead cancer cells. Using DAB4, we examined La expression in human lung cancer samples to assess its suitability as a cancer-selective therapeutic target. We evaluated the safety and effectiveness of radioimmunotherapy (RIT) using DAB4 radiolabeled with Lutetium-177 (177Lu) in the murine Lewis Lung (LL2) carcinoma model, and determined whether combining RIT with DNA-damaging cisplatin-based chemotherapy, a PARP inhibitor (PARPi), or both alters treatment responses. METHODS: The expression of La mRNA in human lung cancer samples was analysed using the online database Oncomine, and the protein expression of La was examined using a TissueFocus Cancer Survey Tissue Microarray. The binding of DAB4 to cisplatin-treated LL2 cells was assessed in vitro. LL2 tumour-bearing mice were administered escalating doses of 177Lu-DAB4 alone or in combination with chemotherapy, and tumour growth and survival measured. Biodistribution analysis was used to determine tissue uptake of 177Lu-DAB4 or its isotype control (177Lu-Sal5), when delivered alone or after chemotherapy. PARPi (rucaparib; AG-014699) was combined with chemotherapy and the effects of combined treatment on tumour growth, tumour cell DNA damage and death, and intratumoural DAB4 binding were also analysed. The effect of the triple combination of PARPi, chemotherapy and 177Lu-DAB4 on tumour growth and survival of LL2 tumour-bearing mice was tested. RESULTS: La was over-expressed at both mRNA and protein levels in surgical specimens of human lung cancer and the over-expression of La mRNA conferred a poorer prognosis. DAB4 bound specifically to cisplatin-induced dead LL2 cells in vitro. An anti-tumour dose response was observed when escalating doses of 177Lu-DAB4 were delivered in vivo, with supra-additive responses observed when chemotherapy was combined with 177Lu-DAB4. Combining PARPi with chemotherapy was more effective than chemotherapy alone with increased tumour cell DNA damage and death, and intratumoural DAB4 binding. The combination of PARPi, chemotherapy and 177Lu-DAB4 was well-tolerated and maximised tumour growth delay. CONCLUSIONS: The La antigen represents a dead cancer cell-specific target in lung cancer, and DAB4 specifically targeted tumour tissue in vivo, particularly after chemotherapy. Tumour uptake of DAB4 increased further after the combination of PARPi and chemotherapy, which generated new dead tumour cell-binding targets. Consequently, combining 177Lu-DAB4 with PARPi and chemotherapy produced the greatest anti-tumour response. Therefore, the triple combination of PARPi, chemotherapy and RIT may have broad clinical utility.

11.
PLoS One ; 8(7): e68895, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23894371

RESUMEN

Passive immunotherapies utilising polyclonal antibodies could have a valuable role in preventing and treating infectious diseases such as influenza, particularly in pandemic situations but also in immunocompromised populations such as the elderly, the chronically immunosuppressed, pregnant women, infants and those with chronic diseases. The aim of this study was to optimise current methods used to generate ovine polyclonal antibodies. Polyclonal antibodies to baculovirus-expressed recombinant influenza haemagglutinin from A/Puerto Rico/8/1934 H1N1 (PR8) were elicited in sheep using various immunisation regimens designed to investigate the priming immunisation route, adjuvant formulation, sheep age, and antigen dose, and to empirically ascertain which combination maximised antibody output. The novel adjuvant CoVaccine HT™ was compared to Freund's adjuvant which is currently the adjuvant of choice for commercial production of ovine polyclonal Fab therapies. CoVaccine HT™ induced significantly higher titres of functional ovine anti-haemagglutinin IgG than Freund's adjuvant but with fewer side effects, including reduced site reactions. Polyclonal hyperimmune sheep sera effectively neutralised influenza virus in vitro and, when given before or after influenza virus challenge, prevented the death of infected mice. Neither the age of the sheep nor the route of antigen administration appeared to influence antibody titre. Moreover, reducing the administrated dose of haemagglutinin antigen minimally affected antibody titre. Together, these results suggest a cost effective way of producing high and sustained yields of functional ovine polyclonal antibodies specifically for the prevention and treatment of globally significant diseases.


Asunto(s)
Adyuvantes Inmunológicos , Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Adyuvante de Freund/inmunología , Subtipo H1N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Envejecimiento/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Relación Dosis-Respuesta Inmunológica , Femenino , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Vacunas contra la Influenza/administración & dosificación , Inyecciones Intraperitoneales , Inyecciones Subcutáneas , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/prevención & control , Ovinos , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/inmunología
12.
J Virol ; 85(7): 3385-96, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21248035

RESUMEN

Fowlpox virus (FWPV) is a double-stranded DNA virus long used as a live-attenuated vaccine against poultry diseases, but more recent interest has focused on its use as a mammalian vaccine vector. Here, in a mouse model system using FWPV encoding the nominal target antigen chicken ovalbumin (OVA) (FWPV(OVA)), we describe for the first time some of the fundamental processes by which FWPV engages both the innate and adaptive immune systems. We show that Toll-like receptor 7 (TLR7) and TLR9 are important for type I interferon secretion by dendritic cells, while TLR9 is solely required for proinflammatory cytokine secretion. Despite this functional role for TLR7 and TLR9 in vitro, only the adapter protein myeloid differentiation primary response gene 88 (MyD88) was shown to be essential for the formation of adaptive immunity to FWPV(OVA) in vivo. The dependence on MyD88 was confined only to the T-cell compartment and was not related to its contribution to TLR signaling, dendritic cell maturation, or the capture and presentation of FWPV-derived OVA antigen. We demonstrate that this is not by means of mediating T-cell-dependent interleukin-1 (IL-1) signaling, but rather, we suggest that MyD88 functions to support T-cell-specific IL-18 receptor signaling, which in turn is essential for the formation of adaptive immunity to FWPV-encoded OVA.


Asunto(s)
Virus de la Viruela de las Aves de Corral/inmunología , Interleucina-18/metabolismo , Glicoproteínas de Membrana/inmunología , Factor 88 de Diferenciación Mieloide/metabolismo , Linfocitos T/inmunología , Receptor Toll-Like 7/inmunología , Receptor Toll-Like 9/inmunología , Vacunas Virales/inmunología , Animales , Pollos , Virus de la Viruela de las Aves de Corral/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ovalbúmina/genética , Ovalbúmina/inmunología , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Vacunas Virales/genética
13.
Clin Vaccine Immunol ; 17(11): 1679-86, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20810681

RESUMEN

Recombinant fowlpox viruses (rFPV) and ovine atadenoviruses (rOAdV) are being developed as safe, nonpathogenic, prophylactic and therapeutic vaccine vectors. There is scope, however, to improve the limited immune responses elicited by each of these vaccine vectors. Using previously determined and optimized routes of administration and viral doses, we characterized the primary adaptive immune responses elicited by recombinant variants of each virus. We demonstrate the contrasting nature of the response elicited by each recombinant virus. Whereas rFPV generates predominately cell-mediated immunity to our nominal target antigen, ovalbumin (OVA), rOAdV drives strong humoral responses. By defining the time taken to achieve maximal cytotoxic T cell responses and by studying the different patterns and kinetics of major histocompatibility complex class I-restricted OVA antigen expression postimmunization, we proposed a heterologous prime-boost regimen of immunization with rOAdV followed by rFPV. The subsequent experimental results showed that this approach produced robust cell-mediated and humoral immune responses against OVA that, importantly, were accompanied by weak anti-viral vector antibody responses. These results, therefore, represent a novel and potentially clinically applicable way to achieve broadly based and effective immunity to the antigens encoded by vectored vaccines.


Asunto(s)
Anticuerpos Antivirales/sangre , Atadenovirus/genética , Virus de la Viruela de las Aves de Corral/genética , Inmunización Secundaria/métodos , Linfocitos T Citotóxicos/inmunología , Vacunación/métodos , Vacunas Virales/inmunología , Animales , Anticuerpos Antivirales/inmunología , Portadores de Fármacos , Vectores Genéticos , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/genética , Ovalbúmina/inmunología , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas Virales/administración & dosificación
14.
Cancer Biol Ther ; 10(7): 715-27, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20676039

RESUMEN

The Src/Abl tyrosine kinase inhibitor dasatinib is an approved chronic myeloid leukemia treatment and is under investigation for solid tumor therapy. Members of the Src family of kinases (SFKs) are involved in the process of metastasis and dasatinib inhibits the migration and invasiveness of human melanoma cell lines in vitro. SFKs are also involved in immune function and angiogenesis, which both contribute to As active and passive immunotherapies continue to be investigated in metastatic melanoma, we investigated possible interactions between kinase inhibitors and immunotherapies. A murine syngenic model of metastatic melanoma in which B16F10 cells expressed ovalbumin (B16-OVA) was employed and the active immunotherapy comprised immunization with an OVA-expressing recombinant fowlpox virus (FPVOVA).Dasatinib did not affect B16-OVA viability, proliferation, migration or soft agar colony formation. However, depending on drug dose and schedule, differences in the metastatic behavior of B16-OVA were observed in vivo after dasatinib therapy. At a dose of 5 mg/kg/day given before tumor challenge, dasatinib therapy reduced the number of pulmonary metastases. Conversely, a higher dose (25 mg/kg/day), did not affect the number of pulmonary metastases and increased the number of extra-pulmonary metastases. Finally, immunization of B16-OVA-bearing mice with FPVOVA reduced the number of lung metastases. Prior treatment of these mice with dasatinib 5 mg/kg/day did not affect the incidence of lung metastases. Although the mechanisms by which dasatinib alters the metastatic behavior of B16-OVA cells in vivo remain to be determined, we hypothesize that dasatinib acts via multiple tumor-extrinsic processes that include immune function and neoangiogenesis.


Asunto(s)
Inmunoterapia/métodos , Neoplasias Pulmonares/prevención & control , Melanoma Experimental/terapia , Pirimidinas/farmacología , Tiazoles/farmacología , Animales , Línea Celular Tumoral , Dasatinib , Relación Dosis-Respuesta a Droga , Virus de la Viruela de las Aves de Corral/genética , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Melanoma Experimental/inmunología , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ovalbúmina/genética , Ovalbúmina/inmunología , Ovalbúmina/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Factores de Tiempo
15.
Expert Rev Anticancer Ther ; 10(6): 917-34, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20553215

RESUMEN

The immune system has an intricate and complex relationship with tumorigenesis; while it has the capacity to identify and eliminate cancerous cells, the emergence of a tumor signifies its failure to do this. Thus, the immune-tumor interplay is paradoxical as through initial suppression of tumor growth, an immunologically silent or even suppressive tumor evolves. Furthermore, certain immune processes, such as chronic inflammation and immunosuppression, can facilitate malignant progression. Nevertheless, immunotherapeutic approaches can manipulate the immune milieu to improve the therapeutic outcomes of cancer treatments. Furthermore, particular conventional cancer therapies also have immunostimulatory properties in their own right. An in-depth understanding of the intimate involvement of the immune system in tumorigenesis and the potential to manipulate this interaction to improve disease outcomes will enable the development of new and broadly effective cancer therapies.


Asunto(s)
Sistema Inmunológico/fisiología , Inmunoterapia , Neoplasias/metabolismo , Humanos
16.
J Virol ; 84(13): 6549-63, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20410285

RESUMEN

Type I interferons (IFNs) are considered to be important mediators of innate immunity due to their inherent antiviral activity, ability to drive the transcription of a number of genes involved in viral clearance, and their role in the initiation of innate and adaptive immune responses. Due to the central role of type I IFNs, we sought to determine their importance in the generation of immunity to a recombinant vaccine vector fowlpox virus (FPV). In analyzing the role of type I IFNs in immunity to FPV, we show that they are critical to the secretion of a number of innate and proinflammatory cytokines, including type I IFNs themselves as well as interleukin-12 (IL-12), tumor necrosis factor-alpha (TNF-alpha), IL-6, and IL-1beta, and that deficiency leads to enhanced virus-mediated antigen expression. Interestingly, however, type I IFNs were not required for adaptive immune responses to recombinant FPV even though plasmacytoid dendritic cells (pDCs), the primary producers of type I IFNs, have been shown to be requisite for this to occur. Furthermore, we provide evidence that the importance of pDCs may lie in their ability to capture and present virally derived antigen to T cells rather than in their capacity as professional type I IFN-producing cells.


Asunto(s)
Inmunidad Adaptativa , Citocinas/inmunología , Células Dendríticas/inmunología , Virus de la Viruela de las Aves de Corral/inmunología , Interferón Tipo I/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos Virales/inmunología , Ratones , Ratones Endogámicos C57BL , Linfocitos T/inmunología , Vacunas Sintéticas/inmunología , Vacunas Virales/inmunología
17.
Exp Hematol ; 37(12): 1435-44, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19786067

RESUMEN

OBJECTIVE: Dasatinib (SPRYCEL, BMS-354825) is a small molecule Src/Abl tyrosine kinase inhibitor approved for the treatment of chronic myeloid leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia. However, kinases inhibited by dasatinib are also involved in the induction and regulation of innate immunity. The purpose of this study was to evaluate the effect of dasatinib on cytokine secretion in response to toll-like receptor (TLR) stimulation. MATERIALS AND METHODS: Dasatinib-treated mice were administered intraperitoneally with lipopolysaccharide (LPS) and serum cytokine (tumor necrosis factor-alpha [TNF-alpha], interleukin [IL]-10, and IL-6) levels and neutrophil accumulation in the lungs were analyzed. Cytokine secretions (TNF-alpha and IL-6) from TLR3-, TLR4-, and TLR9-stimulated RAW264.7, as well as TLR4- and TLR9-stimulated bone marrow-derived macrophages (BMDM) were also evaluated. RESULTS: Dasatinib-treated mice had reduced serum levels of TNF-alpha in response to LPS administration; however, other inflammatory hallmarks of systemic LPS administration, such as secretion of IL-6 and accumulation of neutrophils in the lung, were unaffected. In contrast to the reduced TNF-alpha levels, dasatinib treatment increased serum levels of IL-10 following LPS administration. The production of TNF-alpha was also impaired in vitro in response to TLR3, TLR4, and TLR9 stimulation of the mouse macrophage cell line RAW264.7, as well as TLR4 and TLR9 stimulation of BMDM; IL-6 production was also impaired in dasatinib-treated BMDM. CONCLUSIONS: These findings further support the ability of dasatinib to modulate the host immune response and highlights scope for off-target applications of dasatinib for the control of TNF-alpha-mediated inflammatory disorders.


Asunto(s)
Macrófagos/efectos de los fármacos , Pirimidinas/farmacología , Tiazoles/farmacología , Receptores Toll-Like/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Línea Celular , Células Cultivadas , Dasatinib , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Inyecciones Intraperitoneales , Interleucina-10/sangre , Interleucina-10/metabolismo , Interleucina-6/sangre , Interleucina-6/metabolismo , Lipopolisacáridos/administración & dosificación , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Receptor Toll-Like 3/metabolismo , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 9/metabolismo , Factor de Necrosis Tumoral alfa/sangre
18.
Exp Hematol ; 37(2): 256-65, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19056158

RESUMEN

OBJECTIVE: Dasatinib (BMS-354825) is a small molecule Src/Abl tyrosine kinase inhibitor approved for the treatment of chronic myeloid leukemia and Philadelphia chromosome-positive acute lymphoblastic leukemia. Members of the Src family of kinases are involved in the induction of innate and adaptive immunity. The purpose of this study was to evaluate the inhibitory action of dasatinib on antigen-specific CD8(+) and CD4(+) T-cell function, as well as natural killer (NK) cell cytotoxicity. MATERIALS AND METHODS: To assess dasatinib-mediated inhibition of antigen-specific T-cell proliferation, transgenic CD4(+) and CD8(+) T cells specific for ovalbumin were utilized. Endogenous CD4(+) and CD8(+) T-cell responses were determined following immunization of dasatinib-treated or control mice with a nonreplicating recombinant virus. Clearance of the RMA-S cells, a major histocompatibility complex (MHC) class I-deficient thymoma sensitive to NK-cell lysis, was analyzed in mice undergoing dasatinib treatment. RESULTS: Dasatinib inhibited antigen-specific proliferation of murine CD4(+) and CD8(+) transgenic T cells in vitro and in vivo. Endogenous antigen-specific helper T-cell recall responses and induction of T-cell-mediated cytotoxicity following immunization with a nonreplicating recombinant virus were also inhibited. So to was the ability of NK cells to eliminate MHC class I-deficient cells in vivo. CONCLUSIONS: These findings suggest that dasatinib has the potential to modulate the host immune response at clinical doses and highlights scope for off target applications, e.g., therapeutic immunosuppression in the context of autoimmune pathogenesis and allogeneic tissue transplantation.


Asunto(s)
Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Virus de la Viruela de las Aves de Corral/inmunología , Células Asesinas Naturales/inmunología , Ovalbúmina/inmunología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Tiazoles/farmacología , Animales , Antígenos/genética , Proliferación Celular/efectos de los fármacos , Dasatinib , Virus de la Viruela de las Aves de Corral/genética , Inmunidad Celular/efectos de los fármacos , Inmunidad Celular/genética , Inmunización , Ratones , Ratones Transgénicos , Ovalbúmina/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Transducción Genética
20.
Expert Rev Vaccines ; 6(4): 559-78, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17669010

RESUMEN

While vaccination continues to be the most successful interventionist health policy to date, infectious disease remains a significant cause of death worldwide. A primary reason that vaccination is not able to generate effective immunity is a lack of appropriate adjuvants capable of initiating the desired immune response. Adjuvant combinations can potentially overcome this problem; however, the possible permutations to consider, which include the route and kinetics of vaccination, as well as combinations of adjuvants, are practically limitless. This review aims to summarize the current understanding of adjuvants and related immunological processes and how this knowledge can and has been applied to the strategic selection of adjuvant combinations as components of vaccines against human infectious disease.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Vacunas/inmunología , Compuestos de Alumbre/administración & dosificación , Animales , Células Dendríticas/fisiología , Sistemas de Liberación de Medicamentos , Emulsiones , Vectores Genéticos , Humanos , Sistema Inmunológico/fisiología , Inmunidad Innata , Inmunidad Mucosa , Memoria Inmunológica , Saponinas/administración & dosificación , Vacunas de ADN/inmunología , Virión/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...