Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros

Bases de datos
Tipo de estudio
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Blood ; 113(16): 3682-9, 2009 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-19109232

RESUMEN

Continuous expression of activated factor VII (FVIIa) via gene transfer is a potential therapeutic approach for hemophilia patients with or without inhibitory antibodies to human factor VIII (FVIII) or IX (FIX). Here, we investigate whether gene transfer of an engineered canine FVIIa (cFVIIa) transgene can affect hemostasis in a canine model of hemophilia, a good predictor of efficacy of hemophilia treatments. Purified recombinant cFVIIa exhibited 12-fold higher tissue factor-dependent activity than purified recombinant zymogen cFVII. Subsequently, we generated a serotype 8 recombinant adeno-associated viral vector expressing cFVIIa from a liver-specific promoter. Vector delivery via the portal vein in hemophilia A and B dogs was well tolerated, and long-term expression of cFVIIa resulted in a shortening of the prothrombin time, partial correction of the whole blood clotting time and thromboelastography parameters, and a complete absence of spontaneous bleeding episodes. No evidence of hepatotoxicity, thrombotic complications, or inhibitory immune response was found. These data provide the first evidence for in vivo efficacy and safety of continuously expressed FVIIa as a FVIII/FIX-bypassing agent in a large animal model of hemophilia, avoiding the risk of inhibitor formation associated with bolus FVIII or FIX infusion.


Asunto(s)
Dependovirus , Expresión Génica , Terapia Genética , Hemofilia A/terapia , Animales , Inhibidores de Factor de Coagulación Sanguínea/genética , Inhibidores de Factor de Coagulación Sanguínea/metabolismo , Modelos Animales de Enfermedad , Perros , Factor VIIa , Hemofilia A/genética , Hemofilia A/metabolismo , Hemorragia/genética , Hemorragia/metabolismo , Hemorragia/terapia , Humanos , Hígado/metabolismo , Especificidad de Órganos , Regiones Promotoras Genéticas , Tiempo de Protrombina , Tromboelastografía
2.
Thromb Res ; 120(2): 269-80, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17095052

RESUMEN

INTRODUCTION: Gene therapy could prevent bleeding in hemophilia. However, antibodies could inhibit coagulation, while cytotoxic T lymphocytes could destroy modified cells. The immaturity of the newborn immune system might prevent these immune responses from occurring after neonatal gene therapy. MATERIALS AND METHODS: Newborn dogs, cats, or mice were injected intravenously with a retroviral vector expressing human Factor IX. Plasma was evaluated for antigen and anti-human Factor IX antibodies. Cytotoxic T lymphocyte responses were evaluated indirectly by analysis of retroviral vector RNA in liver. Lymphocytes were evaluated for cytokine secretion and the ability to suppress an immune response to human Factor IX in mice. RESULTS AND CONCLUSIONS: Hemophilia B dogs that achieved 942+/-500 ng/ml (19% normal) or 5+/-0.4 ng/ml (0.1% normal) of human Factor IX in plasma only bled 0 or 1.2 times per year, respectively, and were tolerant to infusion of human Factor IX. Normal cats expressed human Factor IX at 118+/-29 ng/ml (2% normal) in plasma without antibody formation. However, plasma human Factor IX disappeared at late times in 1 of 4 cats, which was probably due to a cytotoxic T lymphocyte response that destroyed cells with high expression. C3H mice were tolerant to human Factor IX after neonatal gene therapy, which may involve clonal deletion of human Factor IX-responsive cells. These data demonstrate that neonatal gene therapy does not induce antibodies to human Factor IX in dogs, cats, or mice. The putative cytotoxic T lymphocyte response in one cat requires further study.


Asunto(s)
Factor IX/genética , Técnicas de Transferencia de Gen , Animales , Animales Recién Nacidos , Gatos , Citocinas/biosíntesis , Enfermedades de los Perros/sangre , Enfermedades de los Perros/genética , Enfermedades de los Perros/terapia , Perros , Factor IX/inmunología , Factor IX/metabolismo , Terapia Genética , Vectores Genéticos , Hemofilia B/sangre , Hemofilia B/genética , Hemofilia B/terapia , Hemofilia B/veterinaria , Humanos , Tolerancia Inmunológica , Ratones , Ratones Endogámicos C3H , Proteínas Recombinantes/sangre , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Retroviridae/genética , Linfocitos T Citotóxicos/inmunología
3.
ILAR J ; 50(2): 144-67, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19293459

RESUMEN

Dogs with hemophilia A, hemophilia B, von Willebrand disease (VWD), and factor VII deficiency faithfully recapitulate the severe bleeding phenotype that occurs in humans with these disorders. The first rational approach to diagnosing these bleeding disorders became possible with the development of reliable assays in the 1940s through research that used these dogs. For the next 60 years, treatment consisted of replacement of the associated missing or dysfunctional protein, first with plasma-derived products and subsequently with recombinant products. Research has consistently shown that replacement products that are safe and efficacious in these dogs prove to be safe and efficacious in humans. But these highly effective products require repeated administration and are limited in supply and expensive; in addition, plasma-derived products have transmitted bloodborne pathogens. Recombinant proteins have all but eliminated inadvertent transmission of bloodborne pathogens, but the other limitations persist. Thus, gene therapy is an attractive alternative strategy in these monogenic disorders and has been actively pursued since the early 1990s. To date, several modalities of gene transfer in canine hemophilia have proven to be safe, produced easily detectable levels of transgene products in plasma that have persisted for years in association with reduced bleeding, and correctly predicted the vector dose required in a human hemophilia B liver-based trial. Very recently, however, researchers have identified an immune response to adeno-associated viral gene transfer vector capsid proteins in a human liver-based trial that was not present in preclinical testing in rodents, dogs, or nonhuman primates. This article provides a review of the strengths and limitations of canine hemophilia, VWD, and factor VII deficiency models and of their historical and current role in the development of improved therapy for humans with these inherited bleeding disorders.


Asunto(s)
Modelos Animales de Enfermedad , Deficiencia del Factor VII/terapia , Terapia Genética/métodos , Hemofilia A/terapia , Hemofilia B/terapia , Proteínas/administración & dosificación , Enfermedades de von Willebrand/terapia , Animales , Coagulación Sanguínea/fisiología , Perros , Deficiencia del Factor VII/genética , Vectores Genéticos , Hemofilia A/genética , Hemofilia B/genética , Trasplante de Hígado/métodos , Enfermedades de von Willebrand/genética , Factor de von Willebrand/metabolismo
4.
Blood ; 105(8): 3079-86, 2005 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-15637142

RESUMEN

Adeno-associated virus 8 (AAV8), a new member of the AAV family isolated from nonhuman primates, is an attractive candidate for hepatic gene transfer applications because of 10- to 100-fold improved transduction efficiency in mouse liver models. Additionally, AAV8 has lesser frequency of pre-existing immunity in humans. These properties could solve some of the problems associated with AAV2 vectors. The benefits of AAV8 demonstrated in mouse models, however, have not been confirmed in larger animals. In this study, we evaluate the efficacy and safety of AAV2/8 vector in both naive and AAV2-pretreated hemophilia B dogs. Two naive hemophilia B dogs that received a single intraportal administration of AAV2/8 vector have achieved sustained expression of 10% and 26% of normal levels of canine factor IX (cFIX) for more than a year. In an AAV2-pretreated hemophilia B dog, cFIX expression increased from less than 1% to 16% of normal levels when treated with an AAV2/8 vector, and a high level of expression has lasted for more than 2 years. No significant liver toxicity or cFIX-specific antibodies have been detected in these animals. Studies here have demonstrated the safety and improved efficacy of AAV2/8 vector in large-animal models for liver-directed gene therapy.


Asunto(s)
Dependovirus/genética , Factor IX/genética , Terapia Genética/métodos , Hemofilia B/terapia , Animales , Anticuerpos , Modelos Animales de Enfermedad , Perros , Factor IX/inmunología , Factor IX/metabolismo , Vectores Genéticos , Hemofilia B/genética , Hemofilia B/inmunología , Hemorragia/prevención & control , Hígado/fisiología , Sistema Porta
5.
Blood ; 102(7): 2403-11, 2003 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-12805062

RESUMEN

Many approaches for treating hemophilia via gene transfer have been attempted in large animal models but all have potential drawbacks. Recombinant adenoviral vectors offer high-efficiency transfer of an episomal vector but have been plagued by the cytotoxicity/immunogenicity of early-generation vectors that contain viral genes. In our current study, we have used a nonintegrating helper-dependent (HD) adenoviral vector for liver-directed gene transfer to achieve hemostatic correction in a dog with hemophilia B. We measured plasma canine factor IX (cFIX) concentrations at a therapeutic range for up to 2.5 months and normalization of the whole blood clotting time (WBCT) for about a month. This was followed by a decrease and stabilized partial correction for 4.5 months. Hepatic gene transfer of a slightly lower dose of the HD vector resulted in WBCTs that were close to normal for 2 weeks, suggesting a dose threshold effect in dogs. In sharp contrast to other studies using first- or second-generation adenoviral vectors, we observed no vector-related elevation of liver enzymes, no fall in platelet counts, and normal liver histology. Taken together, this study demonstrates that injection of an adenoviral HD vector results in complete but transient phenotypic correction of FIX deficiency in canine models with no detectable toxicity.


Asunto(s)
Adenoviridae/genética , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Hemofilia B/terapia , Hígado/fisiología , Animales , ADN Recombinante , Perros , Vectores Genéticos/toxicidad , Hemofilia B/genética , Hígado/citología , Mutación Missense , Fenotipo , Trombocitopenia/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA