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










Base de datos
Intervalo de año de publicación
1.
J Pharmacol Exp Ther ; 371(1): 95-105, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31366602

RESUMEN

Extended half-life (EHL) factor therapies are needed to reduce the burden of prophylaxis and improve treatment adherence in patients with hemophilia. BAX 826 is a novel polysialylated full-length recombinant factor VIII [polysialyic acid (PSA) rFVIII] with improved pharmacokinetics (PK), prolonged pharmacology, and maintained safety attributes to enable longer-acting rFVIII therapy. In factor VIII (FVIII)-deficient hemophilic mice, PSArFVIII showed a substantially higher mean residence time (>2-fold) and exposure (>3-fold), and prolonged efficacy in tail-bleeding experiments (48 vs. 30 hours) compared with unmodified recombinant FVIII (rFVIII), as well as a potentially favorable immunogenicity profile. Reduced binding to a scavenger receptor (low-density lipoprotein receptor-related protein 1) and von Willebrand factor (VWF) as well as a largely VWF-independent circulation time in mice provide a rationale for prolonged BAX 826 activity. The significantly improved PK profile versus rFVIII was confirmed in cynomolgus monkeys [mean residence time: 23.4 vs. 10.1 hours; exposure (area under the curve from time 0 to infinity): 206 vs. 48.2 IU/ml⋅h] and is in line with results from rodent studies. Finally, safety and toxicity evaluations did not indicate increased thrombogenic potential, and repeated administration of BAX 826 to monkeys and rats was well tolerated. The favorable profile and mechanism of this novel experimental therapeutic demonstrated all of the requirements for an EHL-rFVIII candidate, and thus BAX 826 was entered into clinical assessment for the treatment of hemophilia A. SIGNIFICANCE STATEMENT: Prolongation of FVIII half-life aims to reduce the burden of prophylaxis and improve treatment outcomes in patients with hemophilia. This study shows that polysialylation of PSArFVIII resulted in prolongations of rFVIII circulation time and procoagulant activity, together with a favorable nonclinical safety profile of the experimental therapeutic.


Asunto(s)
Factor VIII/uso terapéutico , Hemofilia A/tratamiento farmacológico , Absorción Fisiológica , Animales , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Factor VIII/efectos adversos , Factor VIII/farmacocinética , Femenino , Semivida , Humanos , Macaca fascicularis , Masculino , Ácido N-Acetilneuramínico/química , Unión Proteica , Ratas , Receptores Depuradores/metabolismo , Factor de von Willebrand/metabolismo
2.
Pharm Res ; 36(5): 77, 2019 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-30937539

RESUMEN

PURPOSE: To explore how the natural heterogeneity of human coagulation factor VIII (FVIII) and the processing of its B-domain specifically modulate protein aggregation. METHODS: Recombinant FVIII (rFVIII) molecular species containing 70% or 20% B-domain, and B-domain-deleted rFVIII (BDD-rFVIII), were separated from full-length recombinant FVIII (FL-rFVIII). Purified human plasma-derived FVIII (pdFVIII) was used as a comparator. Heterogeneity and aggregation of the various rFVIII molecular species, FL-rFVIII and pdFVIII were analysed by SDS-PAGE, dynamic light scattering, high-performance size-exclusion chromatography and flow cytometry-based particle analysis. RESULTS: FL-rFVIII and pdFVIII were heterogeneous in nature and demonstrated similar resistance to aggregation under physical stress. Differences were observed between these and among rFVIII molecular species. FVIII molecular species exhibited diverging aggregation pathways dependent on B-domain content. The propensity to form aggregates increased with decreasing proportions of B-domain, whereas the opposite was observed for oligomer formation. Development of cross-ß sheet-containing aggregates in BDD-rFVIII induced effective homologous seeding and faster aggregation. Naturally heterogeneous FL-rFVIII and pdFVIII displayed the lowest propensity to aggregate in all experiments. CONCLUSIONS: These results demonstrate that pdFVIII and FL-rFVIII have similar levels of molecular heterogeneity, and suggest that heterogeneity and the B-domain are involved in stabilising FVIII by modulating its aggregation pathway.


Asunto(s)
Factor VIII/química , Fragmentos de Péptidos/química , Cromatografía Líquida de Alta Presión , Electroforesis en Gel de Poliacrilamida , Factor VIII/aislamiento & purificación , Humanos , Espectrometría de Masas , Fragmentos de Péptidos/aislamiento & purificación , Agregado de Proteínas , Estabilidad Proteica , Elementos Estructurales de las Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación
3.
Lab Anim ; 53(1): 43-52, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30419767

RESUMEN

Genetically engineered mouse models are used to investigate beneficial treatment in haemophilia by comparison with wild-type mice. It has been recognized that wild-type and haemophilic mice of different genetic backgrounds show different bleeding phenotypes. We assessed ex-vivo coagulation parameters in nine wild-type substrains of 129S1/Sv, BALB/c and C57BL/6 mice applying thromboelastography (TEG), activated partial thromboplastin time (aPTT), prothrombin time (PT) and fibrinogen levels. The comprehensive ex-vivo data are discussed in view of results from a tail-tip bleeding assay. Time to first clot formation ( R-time) showed higher within-substrain (CV range: 28-54%) and higher between-substrain (median range: 25.53-42.60 min) variation for BALB/c than for C57BL/6 mice (CV range: 14-31%; median range: 22.45-24.93 min). Median R-time for 129S1/Sv mice was 30.42 min (CV: 33%). No distinct strain differences were observed for maximum amplitude (MA), aPTT, or PT, but males generally showed higher MA and shorter aPTT than females. Males of all substrains had higher fibrinogen levels than females. The heightened in-vivo variability (CV range: 81-171%; median range: 36.00-469.50 mg) in the tail-tip bleeding assay and increased blood loss in wild-type C57BL/6 male mice was not reflected in ex-vivo coagulation parameters. In general, ex-vivo coagulation results appeared consistent within substrains, but showed substrain and sex differences of variable magnitudes. We conclude that alignment of the mouse substrain genetic background to the experimental model is critical to reduce data variability and animal numbers.


Asunto(s)
Coagulación Sanguínea/fisiología , Ratones/fisiología , Fenotipo , Animales , Coagulación Sanguínea/genética , Femenino , Fibrinógeno/análisis , Masculino , Ratones/genética , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Tiempo de Tromboplastina Parcial , Tiempo de Protrombina , Factores Sexuales , Tromboelastografía
4.
Int J Hematol ; 98(5): 525-32, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24061775

RESUMEN

Baxter has developed a new recombinant factor IX (rFIX) drug product (BAX326) for treating patients with hemophilia B, or congenital FIX deficiency. An extensive preclinical program evaluated the pharmacokinetics, efficacy, and safety of BAX326 in different species. The efficacy of BAX326 was tested in three mouse models of primary pharmacodynamics: tail-tip bleeding, carotid occlusion, and thrombelastography. The pharmacokinetics was evaluated after a single intravenous bolus injection in mice, rats, and macaques. Toxicity was assessed in rats and macaques, safety pharmacology in rabbits and macaques, and immunogenicity in mice. BAX326 was shown to be efficacious in all three primary pharmacodynamic studies (P ≤ 0.0076). Hemostatic efficacy was dose related and similar for the three lots tested. Pharmacokinetic results showed that rFIX activity and rFIX antigen concentrations declined in a bi-phasic manner, similar to a previously licensed rFIX product. BAX326 was well tolerated in rabbits and macaques at all dose levels; no thrombogenic events and no adverse clinical, respiratory, or cardiovascular effects occurred. BAX326 was also shown to have a similar immunogenicity profile to the comparator rFIX product in mice. These results demonstrate that BAX326 has a favorable preclinical safety and efficacy profile, predictive of a comparable effect to that of the previously licensed rFIX in humans.


Asunto(s)
Factor IX/farmacología , Hemofilia B/tratamiento farmacológico , Proteínas Recombinantes/farmacología , Animales , Tiempo de Sangría , Coagulación Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Factor IX/administración & dosificación , Factor IX/efectos adversos , Hemofilia B/sangre , Humanos , Macaca , Masculino , Ratones , Conejos , Ratas , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/efectos adversos , Tromboelastografía , Resultado del Tratamiento
5.
Vaccine ; 30(43): 6127-35, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22884662

RESUMEN

BACKGROUND: Influenza pandemic preparedness involves priming of the population with pre-pandemic vaccines. Such vaccines should be well tolerated and induce a long-lasting immunological memory that can effectively be boosted with a single dose of pandemic vaccine once available. The presented studies assessed different prime-boost regimens with a Vero cell-derived whole virus non-adjuvanted H5N1 vaccine. METHODS: In one study, 281 healthy adult (18-59 years) and 280 elderly (≥ 60 years) subjects received two vaccinations, 21 days apart, with Vero cell-derived whole virus non-adjuvanted H5N1 vaccine (7.5 µg HA Antigen A/Vietnam/1203/2004) followed by a 6, 12-15, or 24 month booster (7.5 or 3.75µg A/Indonesia/05/2005 or A/Vietnam/1203/2004). In the other study, 230 healthy adults (18-59 years) received single dose priming (7.5 µg A/Vietnam/1203/2004) followed by a 12 month booster (7.5 or 3.75 µg A/Indonesia/05/2005). Antibody responses were assessed by microneutralization (MN) and single radial hemolysis (SRH) assay. Vaccine safety was assessed throughout. RESULTS: Two dose priming was equally immunogenic in adults and the elderly: >72% of subjects in each population achieved MN titers ≥ 1:20 after the second vaccination. Booster vaccinations at 6, 12-15, and 24 months induced substantial antibody increases to both strains: after a 7.5 µg A/Indonesia/05/2005 booster, 93-95% of adults and 72-84% of the elderly achieved MN titers ≥ 1:20 against this strain. Homologous and heterologous booster responses were higher in the 7.5µg dose group than in the 3.75 µg dose group. Booster responses following single dose priming were similar; a 7.5 µg booster dose induced homologous MN titers ≥ 1:20 in 93% of subjects. CONCLUSIONS: A Vero cell derived whole virus non-adjuvanted H5N1 influenza vaccine is well tolerated and induces long-lasting cross-clade immunological memory that can be effectively boosted 1-2 years after two dose or single dose priming, supporting its suitability for pre-pandemic vaccination.


Asunto(s)
Reacciones Cruzadas/inmunología , Memoria Inmunológica , Subtipo H5N1 del Virus de la Influenza A/inmunología , Vacunas contra la Influenza/inmunología , Adulto , Animales , Anticuerpos Antivirales/sangre , Formación de Anticuerpos , Chlorocebus aethiops , Femenino , Humanos , Inmunización Secundaria , Masculino , Persona de Mediana Edad , Pruebas de Neutralización , Células Vero , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...