Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Pharm Res ; 32(3): 941-54, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25208876

RESUMEN

PURPOSE: Mucopolysaccharidosis I is a genetic disorder caused by alpha-L-iduronidase deficiency. Its primary treatment is enzyme replacement therapy (ERT), which has limitations such as a high cost and a need for repeated infusions over the patient's lifetime. Considering that nanotechnological approaches may enhance enzyme delivery to organs and can reduce the dosage thereby enhancing ERT efficiency and/or reducing its cost, we synthesized laronidase surface-functionalized lipid-core nanocapsules (L-MLNC). METHODS: L-MLNCs were synthesized by using a metal complex. Size distributions were evaluated by laser diffraction and dynamic light scattering. The kinetic properties, cytotoxicity, cell uptake mechanisms, clearance profile and biodistribution were evaluated. RESULTS: Size distributions showed a D[4,3] of 134 nm and a z-average diameter of 71 nm. L-MLNC enhanced the Vmax and Kcat in comparison with laronidase. L-MLNC is not cytotoxic, and nanocapsule uptake by active transport is not only mediated by mannose-6-phosphate receptors. The clearance profile is better for L-MLNC than for laronidase. A biodistribution analysis showed enhanced enzyme activity in different organs within 4 h and 24 h for L-MLNC. CONCLUSIONS: The use of lipid-core nanocapsules as building blocks to synthesize surface-functionalized nanocapsules represents a new platform for producing decorated soft nanoparticles that are able to modify drug biodistribution.


Asunto(s)
Terapia de Reemplazo Enzimático , Fibroblastos/efectos de los fármacos , Iduronidasa/química , Lípidos/química , Mucopolisacaridosis I/tratamiento farmacológico , Nanocápsulas , Animales , Área Bajo la Curva , Transporte Biológico , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Química Farmacéutica , Terapia de Reemplazo Enzimático/efectos adversos , Fibroblastos/metabolismo , Fibroblastos/patología , Iduronidasa/administración & dosificación , Iduronidasa/genética , Iduronidasa/farmacocinética , Iduronidasa/toxicidad , Inyecciones Intravenosas , Tasa de Depuración Metabólica , Ratones Noqueados , Mucopolisacaridosis I/enzimología , Nanomedicina , Tamaño de la Partícula , Tecnología Farmacéutica/métodos , Distribución Tisular
2.
Bioconjug Chem ; 24(1): 97-104, 2013 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-23249376

RESUMEN

The chronic administration of recombinant fusion proteins in preclinical animal models may generate an immune response and the formation of antidrug antibodies (ADA). Such ADAs could alter the plasma pharmacokinetics of the fusion protein, and mask any underlying toxicity of the recombinant fusion protein. In the present study, a model IgG-enzyme fusion protein was evaluated with chronic dosing of rhesus monkeys. The IgG domain of the fusion protein is a genetically engineered monoclonal antibody (mAb) against the human insulin receptor (HIR), which is shown to cross-react with the primate insulin receptor. The enzyme domain of the fusion protein is human iduronidase (IDUA), the lysosomal enzyme mutated in Mucopolysaccharidosis Type I (MPSI). MPSI affects the brain, but enzyme replacement therapy is not effective for the brain, because IDUA does not cross the blood-brain barrier (BBB). The HIRMAb domain of the fusion protein acts as a molecular Trojan horse to deliver the IDUA across the BBB. The HIRMAb-IDUA fusion protein was administered to rhesus monkeys with weekly intravenous infusions of 3-30 mg/kg for 6 months, and the pharmacokinetics, immune response, and tissue toxicology were assessed. The pharmacokinetics of plasma clearance of the fusion protein was determined with measurements of plasma IDUA enzyme activity. ADAs formed during the course of the 6 months of treatment, as determined by a sandwich ELISA. However, the plasma clearance of the fusion protein at the start and end of the 6-month study was comparable at all drug doses. Fusion protein administration for 6 months showed no evidence of chronic tissue toxicity. These studies demonstrate that the immune response produced with chronic treatment of primates with an IgG-enzyme fusion protein has no effect on the pharmacokinetics of plasma clearance of the fusion protein.


Asunto(s)
Formación de Anticuerpos , Iduronidasa/inmunología , Inmunoglobulina G/inmunología , Macaca mulatta/inmunología , Receptor de Insulina/inmunología , Proteínas Recombinantes de Fusión/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/toxicidad , Humanos , Iduronidasa/administración & dosificación , Iduronidasa/farmacocinética , Iduronidasa/toxicidad , Inmunoglobulina G/administración & dosificación , Inmunoglobulina G/toxicidad , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/toxicidad
3.
Drug Metab Dispos ; 40(10): 2021-5, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22822036

RESUMEN

Hurler's syndrome, or mucopolysaccharidosis type I, is a lysosomal storage disorder caused by mutations in the gene encoding the lysosomal enzyme iduronidase (IDUA). The disease affects both peripheral tissues and the central nervous system (CNS). Recombinant IDUA treatment does not affect the CNS, because IDUA does not cross the blood-brain barrier (BBB). To enable BBB penetration, human IDUA was re-engineered as an IgG-IDUA fusion protein, where the IgG domain is a genetically engineered monoclonal antibody (MAb) against the human insulin receptor (HIR). The HIRMAb penetrates the brain from the blood via transport on the endogenous BBB insulin receptor and acts as a molecular Trojan horse to deliver the fused IDUA to the brain. Before human testing, the HIRMAb-IDUA fusion protein was evaluated in a 6-month weekly dosing toxicology study at doses of 0, 3, 9, and 30 mg/kg/week of the fusion protein administered to 40 rhesus monkeys. The focus of the present study is the effect of chronic high dose administration of this fusion protein on plasma glucose and long-term glycemic control. The results show that the HIRMAb has weak insulin agonist activity and causes hypoglycemia at the high dose, 30 mg/kg, after intravenous infusion in normal saline. When dextrose is added to the saline infusion solution, no hypoglycemia is observed at any dose. An intravenous glucose tolerance test performed at the end of the 6 months of chronic treatment showed no change in glucose tolerance at any dose of the HIRMAb-IDUA fusion protein.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Glucemia/efectos de los fármacos , Iduronidasa/administración & dosificación , Receptor de Insulina/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/administración & dosificación , Animales , Anticuerpos Monoclonales/toxicidad , Barrera Hematoencefálica/metabolismo , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Terapia de Reemplazo Enzimático , Femenino , Glucosa/administración & dosificación , Prueba de Tolerancia a la Glucosa , Humanos , Hipoglucemia/sangre , Hipoglucemia/inducido químicamente , Hipoglucemia/prevención & control , Iduronidasa/toxicidad , Infusiones Intravenosas , Macaca mulatta , Masculino , Mucopolisacaridosis I/sangre , Mucopolisacaridosis I/tratamiento farmacológico , Mucopolisacaridosis I/enzimología , Receptor de Insulina/inmunología , Proteínas Recombinantes de Fusión/toxicidad , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA