Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Intervalo de año de publicación
1.
Neuromuscul Disord ; 29(3): 167-186, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30770310

RESUMEN

This multicenter/multinational, open-label, ascending-dose study (NCT01898364) evaluated safety, tolerability, pharmacokinetics, pharmacodynamics, and exploratory efficacy of repeat-dose avalglucosidase alfa (neoGAA), a second-generation, recombinant acid α-glucosidase replacement therapy, in late-onset Pompe disease (LOPD). Patients ≥18 years, alglucosidase alfa naïve (Naïve) or previously receiving alglucosidase alfa for ≥9 months (Switch), with baseline FVC ≥50% predicted and independently ambulatory, received every-other-week avalglucosidase alfa 5, 10, or 20 mg/kg over 24 weeks. 9/10 Naïve and 12/14 Switch patients completed the study. Avalglucosidase alfa was well-tolerated; no deaths/life-threatening serious adverse events (SAEs). One Naïve patient withdrew for study drug-related SAEs (respiratory distress/chest discomfort). Infusion-associated reactions (IARs) affected 8 patients. Most treatment-emergent AEs/IARs were non-serious with mild-to-moderate intensity. At screening, 5 Switch patients tested positive for anti-avalglucosidase alfa antibodies; on-treatment, 2 Switch and 9 Naïve patients seroconverted. Post-infusion, avalglucosidase alfa plasma concentrations declined monoexponentially (t1/2z∼1.0 h). AUC was 5-6 × higher in the 20 vs 5 mg/kg group. Pharmacokinetics were similar between Switch and Naïve groups and over time. Baseline quadriceps muscle glycogen was low (∼6%) in most patients, generally remaining unchanged thereafter. Exploratory efficacy parameters (pulmonary function/functional capacity) generally remained stable or improved. Avalglucosidase alfa's well-tolerated safety profile and exploratory efficacy results support further avalglucosidase alfa development.


Asunto(s)
Terapia de Reemplazo Enzimático , Glucano 1,4-alfa-Glucosidasa/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , alfa-Glucosidasas/farmacología , Adulto , Terapia de Reemplazo Enzimático/efectos adversos , Femenino , Glucano 1,4-alfa-Glucosidasa/farmacología , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Humanos , Masculino , Persona de Mediana Edad , Resultado del Tratamiento , alfa-Glucosidasas/efectos adversos , alfa-Glucosidasas/efectos de los fármacos , alfa-Glucosidasas/farmacocinética
2.
Mol Ther ; 25(5): 1199-1208, 2017 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-28341561

RESUMEN

Duvoglustat HCl (AT2220, 1-deoxynojirimycin) is an investigational pharmacological chaperone for the treatment of acid α-glucosidase (GAA) deficiency, which leads to the lysosomal storage disorder Pompe disease, which is characterized by progressive accumulation of lysosomal glycogen primarily in heart and skeletal muscles. The current standard of care is enzyme replacement therapy with recombinant human GAA (alglucosidase alfa [AA], Genzyme). Based on preclinical data, oral co-administration of duvoglustat HCl with AA increases exposure of active levels in plasma and skeletal muscles, leading to greater substrate reduction in muscle. This phase 2a study consisted of an open-label, fixed-treatment sequence that evaluated the effect of single oral doses of 50 mg, 100 mg, 250 mg, or 600 mg duvoglustat HCl on the pharmacokinetics and tissue levels of intravenously infused AA (20 mg/kg) in Pompe patients. AA alone resulted in increases in total GAA activity and protein in plasma compared to baseline. Following co-administration with duvoglustat HCl, total GAA activity and protein in plasma were further increased 1.2- to 2.8-fold compared to AA alone in all 25 Pompe patients; importantly, muscle GAA activity was increased for all co-administration treatments from day 3 biopsy specimens. No duvoglustat-related adverse events or drug-related tolerability issues were identified.


Asunto(s)
1-Desoxinojirimicina/uso terapéutico , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Lisosomas/enzimología , Músculo Esquelético/efectos de los fármacos , alfa-Glucosidasas/farmacocinética , Administración Oral , Adulto , Esquema de Medicación , Sinergismo Farmacológico , Quimioterapia Combinada , Terapia de Reemplazo Enzimático/métodos , Femenino , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Humanos , Infusiones Intravenosas , Lisosomas/patología , Masculino , Persona de Mediana Edad , Músculo Esquelético/enzimología , Músculo Esquelético/patología , Seguridad del Paciente , Resultado del Tratamiento , alfa-Glucosidasas/sangre
3.
J Pharmacol Exp Ther ; 360(2): 313-323, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27856936

RESUMEN

Pompe disease is a rare neuromuscular disorder caused by an acid α-glucosidase (GAA) deficiency resulting in glycogen accumulation in muscle, leading to myopathy and respiratory weakness. Reveglucosidase alfa (BMN 701) is an insulin-like growth factor 2-tagged recombinant human acid GAA (rhGAA) that enhances rhGAA cellular uptake via a glycosylation-independent insulin-like growth factor 2 binding region of the cation-independent mannose-6-phosphate receptor (CI-MPR). The studies presented here evaluated the effects of Reveglucosidase alfa treatment on glycogen clearance in muscle relative to rhGAA, as well as changes in respiratory function and glycogen clearance in respiratory-related tissue in a Pompe mouse model (GAAtm1Rabn/J). In a comparison of glycogen clearance in muscle with Reveglucosidase alfa and rhGAA, Reveglucosidase alfa was more effective than rhGAA with 2.8-4.7 lower EC50 values, probably owing to increased cellular uptake. The effect of weekly intravenous administration of Reveglucosidase alfa on respiratory function was monitored in Pompe and wild-type mice using whole body plethysmography. Over 12 weeks of 20-mg/kg Reveglucosidase alfa treatment in Pompe mice, peak inspiratory flow (PIF) and peak expiratory flow (PEF) stabilized with no compensation in respiratory rate and inspiratory time during hypercapnic and recovery conditions compared with vehicle-treated Pompe mice. Dose-related decreases in glycogen levels in both ambulatory and respiratory muscles generally correlated to changes in respiratory function. Improvement of murine PIF and PEF were similar in magnitude to increases in maximal inspiratory and expiratory pressure observed clinically in late onset Pompe patients treated with Reveglucosidase alfa (Byrne et al., manuscript in preparation).


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Receptor IGF Tipo 2/metabolismo , Proteínas Recombinantes/farmacología , Respiración/efectos de los fármacos , alfa-Glucosidasas/farmacología , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Humanos , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapéutico , Factores de Tiempo , alfa-Glucosidasas/metabolismo , alfa-Glucosidasas/farmacocinética , alfa-Glucosidasas/uso terapéutico
4.
J Nanosci Nanotechnol ; 15(4): 2657-66, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26353478

RESUMEN

Glycogenosis type II, or Pompe Disease, is a lysosomal storage disease caused by the deficiency of acid alpha-glucosidase (GAA), leading to glycogen accumulation in muscles. A recombinant human GAA (rhGAA, Myozyme®) is currently used for enzyme replacement therapy. Despite its efficacy in most of patients, some of them show a diminished response to the treatment with rapidly progressive clinical deterioration, due to immuno-mediated enzyme inactivation. To demonstrate that Nanoparticles (NPs) could be profitably exploited to carry macromolecules, PLGA NPs loaded with rhGAA (GAA-NPs) were prepared by double emulsion solvent evaporation. Their surface morphology, particle size, zeta-potential and biochemical activity were assessed. "Pulse and chase" experiments were made by administrating GAA-NPs on patients' fibroblasts. Biochemical activity tests showed a more efficient cellular uptake of rhGAA loaded to NPs and a more significant stability of the enzyme (up to 7 days) in vitro, if compared to the same amount of rhGAA free enzyme. This data allows to envision in vivo experiments, in significant animal models, to further characterize lysosomal enzyme loaded-NPs' efficacy and toxicity.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II , Ácido Láctico/química , Lisosomas/metabolismo , Nanopartículas/química , Ácido Poliglicólico/química , ARN/química , alfa-Glucosidasas/química , Células Cultivadas , Sistemas de Liberación de Medicamentos , Fibroblastos , Humanos , Ácido Láctico/farmacocinética , Ácido Poliglicólico/farmacocinética , Copolímero de Ácido Poliláctico-Ácido Poliglicólico , ARN/farmacocinética , alfa-Glucosidasas/farmacocinética
5.
Mol Genet Metab ; 107(3): 469-79, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23041258

RESUMEN

BACKGROUND: Aerobic exercise may be used in conjunction with enzyme replacement therapy (ERT) to attenuate cardiovascular deconditioning, skeletal muscle wasting, and loss of motor function in Pompe disease (glycogen storage disease type II; GSDII), but the effects on lysosomal glycogen content and macroautophagy have not been defined to date. PURPOSE: The main objectives of this study were to determine if acute aerobic exercise enhances 24-h uptake of recombinant human enzyme (rhGAA; Myozyme® [aim 1]) and if endurance training improves disease pathology when combined with ERT [aim 2] in Pompe mice. METHODS: For the first aim in our study, Pompe mutant mice (6(neo)/6(neo)) were grouped into ERT (Myozyme® injection only [40 mg/kg]) and ERT+EX (Myozyme® injection followed by 90 min treadmill exercise) cohorts, and enzyme uptake was assessed in the heart and quadriceps 24h post injection. For the second aim of our study, mutant mice were randomized into control, endurance-trained, enzyme-treated, or combination therapy groups. Exercised animals underwent 14 weeks of progressive treadmill training with or without biweekly Myozyme® injections (40 mg/kg) and tissues were harvested 1 week post last treatment. RESULTS: Myozyme® uptake (GAA activity) was not improved in ERT+EX over ERT alone at 24-h post injection. Endurance exercise training, with or without ERT, improved aerobic capacity and normalized grip strength, motor function, and lean mass (P<0.05), but did not reduce glycogen content or normalize macroautophagy beyond traditional enzyme replacement therapy. CONCLUSIONS: Endurance training is beneficial as an adjunctive therapy to ERT in Pompe disease, although it works by mechanisms independent of a reduction in glycogen content.


Asunto(s)
Terapia de Reemplazo Enzimático , Terapia por Ejercicio , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Glucógeno/metabolismo , Músculo Esquelético/enzimología , alfa-Glucosidasas/uso terapéutico , Animales , Ejercicio Físico , Femenino , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Corazón/efectos de los fármacos , Humanos , Infusiones Intravenosas , Masculino , Ratones , Ratones Transgénicos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/patología , Condicionamiento Físico Animal , alfa-Glucosidasas/farmacocinética
6.
Mol Genet Metab ; 104(4): 552-5, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21967859

RESUMEN

Enzyme replacement therapy (ERT) with alglucosidase alfa was registered as a treatment for Pompe disease in 2006. It is as yet unknown whether ERT can be safely applied during pregnancy and lactation. A primiparous 40-year-old woman diagnosed with Pompe disease continued receiving ERT during pregnancy and lactation. Before pregnancy, she had moderate limb-girdle weakness and used nocturnal ventilation. During pregnancy, her clinical condition remained fairly stable until the 25th gestational week. Thereafter she experienced more problems with mobility and respiration. Fetal growth was normal as monitored by regular ultrasound investigations. A healthy boy was born at a gestational age of 37 weeks and 5 days by elective Cesarean section. There were no maternal complications and the child developed normally. One year after delivery the mother's physical condition was similar as prior to her pregnancy. Pharmacokinetic studies following enzyme infusion showed that alglucosidase alfa was secreted into the breast milk. Activity levels in the milk (245 nmol/ml.h) peaked at 2.5h after the end of the infusion; which was 2h later than in the plasma (80 µmol/ml.h). Twenty-four hours after start of the infusion, the enzyme activity in the breast milk was back to the pre-infusion level. In this case report, the continuation of treatment with alglucosidase alfa during pregnancy and lactation has been safe for the mother and the child.


Asunto(s)
Terapia de Reemplazo Enzimático , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Complicaciones del Embarazo/tratamiento farmacológico , alfa-Glucosidasas/uso terapéutico , Adulto , Femenino , Humanos , Recién Nacido , Masculino , Leche Humana/enzimología , Embarazo , Resultado del Tratamiento , alfa-Glucosidasas/efectos adversos , alfa-Glucosidasas/farmacocinética
7.
Curr Opin Investig Drugs ; 5(10): 1101-10, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15535432

RESUMEN

Genzyme General is developing recombinant human alpha-glucosidase, produced in mammalian cell culture, as a potential treatment for Pompe disease. By July 2004, enrollment was completed in two clinical trials and an observational study in adults. Genzyme was planning to file for regulatory approval in Europe during 2004, followed by filings in the US and Japan in mid-2005.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , alfa-Glucosidasas/uso terapéutico , Animales , Ensayos Clínicos como Asunto , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Humanos , Proteínas Recombinantes/química , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/uso terapéutico , Relación Estructura-Actividad , Resultado del Tratamiento , alfa-Glucosidasas/química , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/farmacocinética
8.
An. otorrinolaringol. mex ; 44(3): 134-7, jun.-ago. 1999. ilus, tab, graf
Artículo en Español | LILACS | ID: lil-276925

RESUMEN

Antecedentes e hipótesis. La estreptomicina (STP)es un antibiótico aminoglicósido que se usa en el tratamiento de la tuberculosis, con efectos tóxicos importantes sobre la función vestibular. Los antecedentes farmacocinéticos muestran que la STP se acumula en un principio en la parte perilinfática y por mecanismos aún no descritos pasa a la endolinfa donde su principal blanco de acción son los cilios sensorios de las células pilosas vestibulares. Consideramos importante averiguar que parte de la molécula de STP es la responsable de su toxicidad vestibular, y tenemos evidencias que señalan la parte estreptidina (STD)como la porción activa, por lo que se propone que la STP actúa fragmentada por acción de una alfa-glucosidasa que hidroliza el enlace -1,4 con la liberación de la STD y que esté presente ya sea en el vestíbulo, en el torrente sanguíneo o en ambos. Método. Se determinó la actividad de alfa-glucosidasa en el suero y en homogeneizado vestibular de la rata usando p-nitrofenil--d-glucósido como sustrato; siguiendo el p-nitrofenilo (pNP)liberado espectrofotométricamente a 400 nm. Resultados. Se encontró alfa-glucosidasa tanto en el suero como en el vestíbulo de la rata con actividades específicas de 0.035 y 1.3 nmol de pNP/mg proteína/min respectivamente. Conclusión. La actividad encontrada en vestíbulo es semejante a la de la alfa-glucosidasa lisosomal del hígado de la rata (1.5 nmol de pNP/mg proteína/min)por lo que es posible que la alfa-glucosidasa presente en el suero y en el vestíbulo pueda romper a la STP, liberando la parte STD que actuaría sobre el oído


Asunto(s)
Animales , Ratas , alfa-Glucosidasas/farmacocinética , Antibacterianos/toxicidad , Hidrólisis , Vestíbulo del Laberinto/efectos de los fármacos , Ratas Long-Evans/lesiones , Estreptomicina/efectos adversos
9.
Hum Mol Genet ; 7(11): 1695-702, 1998 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-9736771

RESUMEN

Glycogen storage disease type II (GSD II) is an autosomal recessive disorder caused by defects in the lysosomal acid alpha-glucosidase (GAA) gene. We investigated the feasibility of using a recombinant adenovirus containing the human GAA gene under the control of the cytomegalovirus promoter (AdCMV-GAA) to correct the enzyme deficiency in different cultured cells from patients with the infantile form of GSD II. In GAA-deficient fibroblasts infected with AdCMV-GAA, transduction and transcription of the human transgene resulted in de novo synthesis of GAA protein. The GAA enzyme activity was corrected from the deficient level to 12 times the activity of normal cells. The transduced cells overexpressed the 110 kDa precursor form of GAA, which was secreted into the culture medium and was taken up by recipient cells. The recombinant GAA protein was correctly processed and was active on both an artificial substrate 4-methylumbelliferyl-alpha-D-glucopyranoside (4MUG) and glycogen. In GAA-deficient muscle cells, a significant increase in cellular enzyme level, approximately 20-fold higher than in normal cells, was also observed after viral treatment. The transduced muscle cells were also able to efficiently secrete the recombinant GAA. Moreover, transfer of the human transgene resulted in normalization of cellular glycogen content with clearance of glycogen from lysosomes, as assessed by electron microscopy, in differentiated myotubes. These results demonstrate phenotypic correction of cultured skeletal muscle from a patient with infantile-onset GSD II using a recombinant adenovirus. We conclude that adenovirus-mediated gene transfer might be a suitable model system for further in vivo studies on delivering GAA to GSD II muscle, not only by direct cell targeting but also by a combination of secretion and uptake mechanisms.


Asunto(s)
Adenoviridae/genética , Técnicas de Transferencia de Gen , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , alfa-Glucosidasas/genética , alfa-Glucosidasas/metabolismo , Western Blotting , Células Cultivadas , Fibroblastos/metabolismo , Terapia Genética/métodos , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Humanos , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacocinética , Transducción Genética , alfa-Glucosidasas/farmacocinética
10.
C R Seances Soc Biol Fil ; 185(5): 345-56, 1991.
Artículo en Francés | MEDLINE | ID: mdl-1806194

RESUMEN

In vivo injection, into emerging bees, of a quantity of trehalose sufficient to double the initial natural haemolymph level of the sugar, leads first to a reduction in the maximal velocity (Vm) of the alpha-glucosidase activity, after 2 hours, followed by an increase in Vm at 6 hours, then a return to normal. The substrate constant (KR) shows minor and insignificant fluctuations about the initial value, and the Hill coefficient (h) a slight, but significant, rise 2 h and 8 h after injection. The simultaneous injection of a protein synthesis inhibitor (ISP) reduces the changes in Vm and h, and lowers value of the ratio Vm/KR from 6 h to 16 h after injection. The overall pattern of results prompts the hypothesis of an intermediary ISP-sensitive induction of an inhibitor of the enzyme-substrate complex, preceding the phase of induction or liberation of active enzyme molecules whose aggregation is subject to a succession of rearrangements alternately favourable to sucrose (lesser specificity) or to trehalose (heightened specificity).


Asunto(s)
Abejas/enzimología , Hemolinfa/enzimología , Trehalosa/farmacología , alfa-Glucosidasas/farmacocinética , Animales , Inhibidores de la Síntesis de la Proteína/farmacología , alfa-Glucosidasas/sangre , alfa-Glucosidasas/metabolismo
11.
C R Seances Soc Biol Fil ; 182(3): 249-55, 1988.
Artículo en Francés | MEDLINE | ID: mdl-2976291

RESUMEN

Neutral maltase from human granulocytes has a different substrate specificity from the human neutral maltase of kidney, though it has been reported that these two enzymes are immunologically similar. We report here that human granulocyte neutral maltase is similar to the neutral maltase from rat's kidney as regards the substrate specificity and the inhibition by Tris and maltodextrins. We also report a different thermal stability that might imply some structural differences between the two enzymes.


Asunto(s)
Granulocitos/enzimología , Riñón/enzimología , alfa-Glucosidasas/farmacocinética , Animales , Humanos , Masculino , Polisacáridos/farmacología , Ratas , Ratas Endogámicas , Especificidad por Sustrato , Temperatura , Factores de Tiempo , Trometamina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...