Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Mol Genet Metab ; 120(4): 370-377, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28189603

RESUMEN

BACKGROUND: Long-chain fatty acid oxidation disorders (LC-FAOD) lead to accumulation of high concentrations of potentially toxic fatty acid intermediates. Newborn screening and early intervention have reduced mortality, but most patients continue to experience frequent hospitalizations and significant morbidity despite treatment. The deficient energy state can cause serious liver, muscle, and heart disease, and may be associated with an increased risk of sudden death. Triheptanoin is a medium odd-chain fatty acid. Anaplerotic metabolites of triheptanoin have the potential to replace deficient tricarboxylic acid (TCA) cycle intermediates, resulting in net glucose production as a novel energy source for the treatment of LC-FAOD. STUDY DESIGN: A single-arm, open-label, multicenter Phase 2 safety and efficacy study evaluated patients with severe LC-FAOD evidenced by ongoing related musculoskeletal, cardiac, and/or hepatic events despite treatment. After a four-week run-in on current regimen, investigational triheptanoin (UX007) was titrated to a target dose of 25-35% of total daily caloric intake. Patients were evaluated on several age/condition-eligible endpoints, including submaximal exercise tests to assess muscle function/endurance (12-minute walk test; 12MWT) and exercise tolerance (cycle ergometry), and health related quality of life (HR-QoL). Results through 24weeks of treatment are presented; total study duration is 78weeks. RESULTS: Twenty-nine patients (0.8 to 58years) were enrolled; most qualified based on severe musculoskeletal disease. Twenty-five patients (86%) completed the 24-week treatment period. At Week 18, eligible patients (n=8) demonstrated a 28% increase (LS mean=+181.9 meters; p=0.087) from baseline (673.4meters) in 12MWT distance. At Week 24, eligible patients (n=7) showed a 60% increase in watts generated (LS mean=+409.3W; p=0.149) over baseline (744.6W) for the exercise tolerance test. Improvements in exercise tests were supported by significant improvements from baseline in the adult (n=5) self-reported SF-12v2 physical component summary score (LS mean=+8.9; p<0.001). No difference from baseline was seen in pediatric parent-reported (n=5) scores (SF-10) at Week 24. Eighteen patients (62%) had treatment-related adverse events, predominantly gastrointestinal (55%), mild-to-moderate in severity, similar to that seen with prior treatment with medium chain triglyceride (MCT) oil. One patient experienced a treatment-related serious adverse event of gastroenteritis. One patient discontinued from study due to diarrhea of moderate severity; the majority of patients (25/29; 86%) elected to continue treatment in the extension period. CONCLUSIONS: In patients with severe LC-FAOD, UX007 interim study results demonstrated improved exercise endurance and tolerance, and were associated with positive changes in self-reported HR-QoL.


Asunto(s)
Ácidos Grasos/toxicidad , Errores Innatos del Metabolismo Lipídico/tratamiento farmacológico , Resistencia Física/efectos de los fármacos , Triglicéridos/administración & dosificación , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Errores Innatos del Metabolismo Lipídico/metabolismo , Errores Innatos del Metabolismo Lipídico/fisiopatología , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Calidad de Vida , Resultado del Tratamiento , Triglicéridos/farmacología , Prueba de Paso , Adulto Joven
3.
Mol Genet Metab ; 102(3): 326-38, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21176882

RESUMEN

The Research Challenges in CNS Manifestations of Inborn Errors of Metabolism workshop was designed to address challenges in translating potential therapies for these rare disorders, and to highlight novel therapeutic strategies and innovative approaches to CNS delivery, assessment of effects and directions for the future in the treatment of these diseases. Therapies for the brain in inborn errors represent some of the greatest challenges to translational research due to the special properties of the brain, and of inborn errors themselves. This review covers the proceedings of this workshop as submitted by participants. Scientific, ethical and regulatory issues are discussed, along with ways to measure outcomes and the conduct of clinical trials. Participants included regulatory and funding agencies, clinicians, scientists, industry and advocacy groups.


Asunto(s)
Investigación Biomédica , Sistema Nervioso Central , Errores Innatos del Metabolismo/diagnóstico , Errores Innatos del Metabolismo/terapia , Animales , Investigación Biomédica/ética , Investigación Biomédica/tendencias , Sistema Nervioso Central/patología , Ensayos Clínicos como Asunto/ética , Humanos , Errores Innatos del Metabolismo/fisiopatología , Enfermedades Raras/terapia
4.
J Inherit Metab Dis ; 32 Suppl 1: S253-8, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19562502

RESUMEN

Intravenous enzyme replacement therapy with recombinant human α-L-iduronidase (rhIDU) is used weekly to treat mucopolysaccharidosis (MPS) I. We tested continuous administration of rhIDU at two dosing levels (0.58 mg/kg per week and 2 mg/kg per week) in MPS I dogs, and compared the efficacy of continuous infusion with the clinically used 0.58 mg/kg weekly three-hour infusion. Peak plasma concentrations of rhIDU were much higher in weekly-treated dogs (mean 256 units/ml) than steady-state concentrations in dogs treated with continuous infusion (mean 1.97 units/ml at 0.58 mg/kg per week; 8.44 units/ml at 2 mg/kg per week). Dogs receiving continuous IV rhIDU, even at a higher (2 mg/kg per week) dose, had consistently lower iduronidase levels in tissues than dogs receiving a weekly (0.58 mg/kg per week) dose. GAG storage was also less improved by continuous intravenous infusion. Adverse events were similar in all dosing groups. We found that continuous administration of 2 mg/kg per week rhIDU to MPS I dogs was insufficient to achieve GAG storage reduction comparable to 0.58 mg/kg weekly dosing.


Asunto(s)
Terapia de Reemplazo Enzimático/métodos , Iduronidasa/administración & dosificación , Mucopolisacaridosis I/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Perros , Terapia de Reemplazo Enzimático/instrumentación , Glicosaminoglicanos/metabolismo , Humanos , Iduronidasa/sangre , Bombas de Infusión , Infusiones Intravenosas , Mucopolisacaridosis I/metabolismo , Proteínas Recombinantes/administración & dosificación , Resultado del Tratamiento
5.
Gene Ther ; 13(11): 917-25, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16482204

RESUMEN

Mucopolysaccharidosis type I (MPS I) is caused by an inherited deficiency of alpha-L-iduronidase (IDUA). The result is a progressive, lysosomal storage disease with central nervous system (CNS) as well as systemic involvement. To target gene therapy to the CNS, recombinant adeno-associated virus (AAV) vectors carrying IDUA sequence were administered to MPS I mice via injection into cerebrospinal fluid. In contrast to intravenous administration, this intrathecal administration was effective in generating widespread IDUA activity in the brain, with the cerebellum and olfactory bulbs having highest activities. In general, IDUA levels correlated with vector dose, although this correlation was obscured in cerebellum by particularly high variability. High doses of vector (4 x 10(10) particles) provided IDUA levels approaching or exceeding normal levels in the brain. Histopathology indicated that the number of cells with storage vacuoles was reduced extensively or was eliminated entirely. Elimination of storage material in Purkinje cells was particularly dramatic. A lower vector dose (2 x 10(9) particles) reduced both the number of storage cells and the extent of storage per cell, but the effect was not complete. Some perivascular cells with storage persisted, and this cell type appeared to be more resistant to treatment than neurons or glial cells. We conclude that intrathecal administration of AAV-IDUA delivers vector to brain cells, and that this route of administration is both minimally invasive and effective.


Asunto(s)
Encéfalo/metabolismo , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Iduronidasa/genética , Mucopolisacaridosis I/terapia , Animales , Encéfalo/ultraestructura , Técnica del Anticuerpo Fluorescente Indirecta , Expresión Génica , Iduronidasa/análisis , Iduronidasa/metabolismo , Inyecciones Espinales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Mucopolisacaridosis I/metabolismo , Mucopolisacaridosis I/patología
6.
Mol Genet Metab ; 86(1-2): 141-9, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16006167

RESUMEN

Central nervous system disease can have devastating consequences in the severe or Hurler form of mucopolysaccharisosis I (MPS I). Intravenously administered recombinant human alpha-L-iduronidase (rhIDU) is not expected to reach and treat the brain disease due to the blood-brain barrier. To determine whether administration of rhIDU into the cerebrospinal fluid could successfully treat the brain, we studied intraventricular administration of rhIDU in rats. RhIDU was stereotactically administered directly to the lateral ventricle of the intact rat brain and the brain tissues assessed by enzyme assays, immunofluorescence and confocal microscopy 30 min, 24 h, or 7 days later. Quantitation of activity revealed that rhIDU was widely distributed throughout the brain following injection into the lateral ventricle, with activities increased by a factor of 3.3 higher than control in most samples 30 min-24 h after injection and highest levels on the side of injection. The enzyme crossed the ependymal lining of the ventricle and entered neurons into lysosomal-like vesicles. The enzyme was able to diffuse through brain tissue as demonstrated by a decreasing signal gradient from 0.2 to 4.8 mm from the ventricle surface. The largest amount of rhIDU, as detected by immunostaining, was observed 24 h after injection and decreased approximately 50% during the first 7 days. Although the immunostaining decreased with time, specific vesicular staining was still detectable 28 days after injection. The data suggest that rhIDU given into the ventricle can diffuse, penetrate at least several millimeters of brain tissue and be taken up into neurons and glial cells.


Asunto(s)
Iduronidasa/farmacocinética , Animales , Difusión , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Iduronidasa/administración & dosificación , Inyecciones Intraventriculares , Masculino , Microscopía Confocal , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacocinética
7.
Mol Genet Metab ; 83(1-2): 163-74, 2004.
Artículo en Inglés | MEDLINE | ID: mdl-15464431

RESUMEN

Enzyme replacement therapy (ERT) has been developed for several lysosomal storage disorders, including mucopolysaccharidosis I (MPS I), and is effective at reducing lysosomal storage in many tissues and in ameliorating clinical disease. However, intravenous ERT does not adequately treat storage disease in the central nervous system (CNS), presumably due to effects of the blood-brain barrier on enzyme distribution. To circumvent this barrier, we studied whether intrathecal (IT) recombinant human alpha-L-iduronidase (rhIDU) could penetrate and treat the brain and meninges. An initial dose-response study showed that doses of 0.46-4.14 mg of IT rhIDU successfully penetrated the brain of normal dogs and reached tissue levels 5.6 to 18.9-fold normal overall and 2.7 to 5.9-fold normal in deep brain sections lacking CSF contact. To assess the efficacy and safety in treating lysosomal storage disease, four weekly doses of approximately 1 mg of IT rhIDU were administered to MPS I-affected dogs resulting in a mean 23- and 300-fold normal levels of iduronidase in total brain and meninges, respectively. Quantitative glycosaminoglycan (GAG) analysis showed that the IT treatment reduced mean total brain GAG to normal levels and achieved a 57% reduction in meningeal GAG levels accompanied by histologic improvement in lysosomal storage in all cell types. The dogs did develop a dose-dependent immune response against the recombinant human protein and a meningeal lymphocytic/plasmacytic infiltrate. The IT route of ERT administration may be an effective way to treat the CNS disease in MPS I and could be applicable to other lysosomal storage disorders.


Asunto(s)
Encéfalo/patología , Iduronidasa/farmacología , Meninges/patología , Mucopolisacaridosis I/tratamiento farmacológico , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Perros , Relación Dosis-Respuesta a Droga , Glicosaminoglicanos/metabolismo , Humanos , Iduronidasa/administración & dosificación , Iduronidasa/efectos adversos , Sistema Inmunológico/efectos de los fármacos , Inyecciones Espinales , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Lisosomas/patología , Meninges/efectos de los fármacos , Meninges/metabolismo , Mucopolisacaridosis I/patología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/genética , Proteínas Recombinantes/farmacología , Valores de Referencia , Distribución Tisular
8.
Hum Mutat ; 24(3): 199-207, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15300847

RESUMEN

Mucopolysaccharidosis type I (MPS I) is an autosomal recessive lysosomal storage disorder caused by a deficiency of alpha-L-iduronidase (IDUA). Mutations in the gene are responsible for the enzyme deficiency, which leads to the intralysosomal storage of the partially degraded glycosaminoglycans dermatan sulfate and heparan sulfate. Molecular characterization of MPS I patients has resulted in the identification of over 70 distinct mutations in the IDUA gene. The high degree of molecular heterogeneity reflects the wide clinical variability observed in MPS I patients. Six novel mutations, c.1087C>T (p.R363C), c.1804T>A (p.F602I), c.793G>C, c.712T>A (p.L238Q), c.1727+2T>A, and c.1269C>G (p.S423R), in a total of 14 different mutations, and 13 different polymorphic changes, including the novel c.246C>G (p.H82Q), were identified in a cohort of 10 MPS I patients enrolled in a clinical trial of enzyme-replacement therapy. Five novel amino acid substitutions and c.236C>T (p.A79V) were engineered into the wild-type IDUA cDNA and expressed. A p.G265R read-through mutation, arising from the c.793G>C splice mutation, was also expressed. Each mutation reduced IDUA protein and activity levels to varying degrees with the processing of many of the mutant forms also affected by IDUA. The varied properties of the expressed mutant forms of IDUA reflect the broad range of biochemical and clinical phenotypes of the 10 patients in this study. IDUA kinetic data derived from each patient's cultured fibroblasts, in combination with genotype data, was used to predict disease severity. Finally, residual IDUA protein concentration in cultured fibroblasts showed a weak correlation to the degree of immune response to enzyme-replacement therapy in each patient.


Asunto(s)
Iduronidasa/genética , Mucopolisacaridosis I/genética , Mutación , Sustitución de Aminoácidos , Animales , Células CHO , Línea Celular/enzimología , Codón/genética , Estudios de Cohortes , Cricetinae , Cricetulus , Análisis Mutacional de ADN , ADN Complementario/genética , Exones/genética , Fibroblastos/enzimología , Humanos , Iduronidasa/química , Iduronidasa/deficiencia , Iduronidasa/metabolismo , Iduronidasa/uso terapéutico , Cinética , Mucopolisacaridosis I/tratamiento farmacológico , Mutagénesis Sitio-Dirigida , Mutación Missense , Fenotipo , Mutación Puntual , Polimorfismo Genético , Proteínas Recombinantes de Fusión/metabolismo
9.
Proc Natl Acad Sci U S A ; 101(3): 829-34, 2004 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-14715900

RESUMEN

Immune responses can interfere with the effective use of therapeutic proteins to treat genetic deficiencies and have been challenging to manage. To address this problem, we adapted and studied methods of immune tolerance used in canine organ transplantation research to soluble protein therapeutics. A tolerization regimen was developed that prevents a strong antibody response to the enzyme alpha-l-iduronidase during enzyme replacement therapy of a canine model of the lysosomal storage disorder mucopolysaccharidosis I. The tolerizing regimen consists of a limited 60-day course of cyclosporin A and azathioprine combined with weekly i.v. infusions of low-dose recombinant human alpha-l-iduronidase. The canines tolerized with this regimen maintain a reduced immune response for up to 6 months despite weekly therapeutic doses of enzyme in the absence of immunosuppressive drugs. Successful tolerization depended on high plasma levels of cyclosporin A combined with azathioprine. In addition, the induction of tolerance may require mannose 6-phosphate receptor-mediated uptake because alpha-l-iduronidase and alpha-glucosidase induced tolerance with the drug regimen whereas ovalbumin and dephosphorylated alpha-l-iduronidase did not. This tolerization method should be applicable to the treatment of other lysosomal storage disorders and provides a strategy to consider for other nontoleragenic therapeutic proteins and autoimmune diseases.


Asunto(s)
Iduronidasa/inmunología , Iduronidasa/uso terapéutico , Terapia de Inmunosupresión/métodos , Mucopolisacaridosis I/tratamiento farmacológico , Mucopolisacaridosis I/inmunología , Animales , Azatioprina/administración & dosificación , Ciclosporina/administración & dosificación , Ciclosporina/sangre , Modelos Animales de Enfermedad , Perros , Humanos , Iduronidasa/administración & dosificación , Tolerancia Inmunológica , Inmunosupresores/administración & dosificación , Inmunosupresores/sangre , Mucopolisacaridosis I/metabolismo , Receptor IGF Tipo 2/metabolismo , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/uso terapéutico
10.
Mol Genet Metab ; 72(3): 199-208, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11243725

RESUMEN

Enzyme replacement therapy (ERT) has long been considered an approach to treating lysosomal storage disorders caused by deficiency of lysosomal enzymes. ERT is currently used to treat Gaucher disease and is being developed for several lysosomal storage disorders now that recombinant sources of the enzymes have become available. We have continued development of ERT for mucopolysaccharidosis I (MPS I) using the feline model. Recombinant alpha-L-iduronidase was administered intravenously at low dose (approximately 0.1 mg/kg or 25,000 units/kg) to four cats and high dose (0.5 mg/kg or 125,000 units/kg) to two cats on a weekly basis for 3- or 6-month terms. Clinical examinations showed distinct clearing of corneal clouding in one cat although clinical effects in the others were not evident. Biochemical studies of the cats showed that the enzyme was distributed to a variety of tissues although the liver and spleen contained the highest enzyme activities. Glycosaminoglycan storage was decreased in liver and spleen, and the histologic appearance improved in liver, spleen, and renal cortex. Enzyme was not consistently detected in cerebral cortex, brainstem, or cerebellum and the histologic appearance and ganglioside profiles did not improve. A variety of other tissues showed low variable uptake of enzyme and no distinct improvement. IgG antibodies to alpha-L-iduronidase were observed in five cats with higher titers noted when higher doses were administered. Mild complement activation occurred in three cats. Enzyme replacement therapy was effective in reversing storage in some tissues at the biochemical and histologic level in MPS I cats but an improved tissue distribution and prevention of a significant immune response could make the therapy more effective.


Asunto(s)
Iduronidasa/uso terapéutico , Mucopolisacaridosis I/tratamiento farmacológico , Animales , Encéfalo/metabolismo , Gatos , Glicosaminoglicanos/metabolismo , Iduronidasa/administración & dosificación , Iduronidasa/genética , Iduronidasa/metabolismo , Inmunoglobulina G/biosíntesis , Infusiones Intravenosas , Corteza Renal/metabolismo , Corteza Renal/ultraestructura , Hígado/metabolismo , Hígado/ultraestructura , Mucopolisacaridosis I/patología , Proteínas Recombinantes/uso terapéutico , Bazo/metabolismo , Bazo/ultraestructura , Distribución Tisular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...