Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
JCI Insight ; 4(5)2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30843882

RESUMEN

Pompe disease is a rare inherited disorder of lysosomal glycogen metabolism due to acid α-glucosidase (GAA) deficiency. Enzyme replacement therapy (ERT) using alglucosidase alfa, a recombinant human GAA (rhGAA), is the only approved treatment for Pompe disease. Although alglucosidase alfa has provided clinical benefits, its poor targeting to key disease-relevant skeletal muscles results in suboptimal efficacy. We are developing an rhGAA, ATB200 (Amicus proprietary rhGAA), with high levels of mannose-6-phosphate that are required for efficient cellular uptake and lysosomal trafficking. When administered in combination with the pharmacological chaperone AT2221 (miglustat), which stabilizes the enzyme and improves its pharmacokinetic properties, ATB200/AT2221 was substantially more potent than alglucosidase alfa in a mouse model of Pompe disease. The new investigational therapy is more effective at reversing the primary abnormality - intralysosomal glycogen accumulation - in multiple muscles. Furthermore, unlike the current standard of care, ATB200/AT2221 dramatically reduces autophagic buildup, a major secondary defect in the diseased muscles. The reversal of lysosomal and autophagic pathologies leads to improved muscle function. These data demonstrate the superiority of ATB200/AT2221 over the currently approved ERT in the murine model.


Asunto(s)
Terapia de Reemplazo Enzimático/métodos , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , alfa-Glucosidasas/farmacología , alfa-Glucosidasas/uso terapéutico , 1-Desoxinojirimicina/análogos & derivados , Animales , Modelos Animales de Enfermedad , Femenino , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Masculino , Manosafosfatos/metabolismo , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Ratas , Ratas Sprague-Dawley , alfa-Glucosidasas/sangre , alfa-Glucosidasas/genética
2.
Anal Chem ; 89(16): 8288-8295, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28686011

RESUMEN

Gaucher disease (GD) is caused by mutations in the GBA1 gene that encodes the lysosomal enzyme acid ß-glucosidase (GCase). Reduced GCase activity primarily leads to the accumulation of two substrates, glucosylceramide (GlcCer) and glucosylsphingosine (GlcSph). Current treatment options have not been shown to ameliorate the neurological pathology observed in the most severe forms of GD, clearly representing an unmet medical need. To better understand the relationship between GlcCer and GlcSph accumulation and ultimately their connection with the progression of neurological pathology, we developed LC-MS/MS methods to quantify GlcCer and GlcSph in mouse brain tissue. A significant challenge in developing these methods was the chromatographic separation of GlcCer and GlcSph from the far more abundant isobaric galactosyl epimers naturally occurring in white matter. After validation of both methods, we evaluated the levels of both substrates in five different GD mouse models, and found significant elevation of brain GlcSph in all five, while GlcCer was elevated in only one of the five models. In addition, we measured GlcCer and GlcSph levels in the brains of wild-type mice after administration of the GCase inhibitor conduritol ß-epoxide (CBE), as well as the nonlysosomal ß-glucosidase (GBA2) inhibitor N-butyldeoxygalactonojirimycin (NB-DGJ). Inhibition of GCase by CBE resulted in elevation of both sphingolipids; however, inhibition of GBA2 by NB-DGJ resulted in elevation of GlcCer only. Taken together, these data support the idea that GlcSph is a more selective and sensitive biomarker than GlcCer for neuronopathic GD in preclinical models.


Asunto(s)
Biomarcadores/análisis , Enfermedad de Gaucher/metabolismo , Glucosilceramidas/análisis , Psicosina/análogos & derivados , Animales , Biomarcadores/metabolismo , Encéfalo/metabolismo , Cromatografía Liquida , Glucosilceramidasa/antagonistas & inhibidores , Glucosilceramidas/metabolismo , Ratones Endogámicos C57BL , Psicosina/análisis , Psicosina/metabolismo , Espectrometría de Masas en Tándem , beta-Glucosidasa/antagonistas & inhibidores
3.
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
4.
Genet Med ; 19(4): 430-438, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27657681

RESUMEN

PURPOSE: Fabry disease is an X-linked lysosomal storage disorder caused by mutations in the α-galactosidase A gene. Migalastat, a pharmacological chaperone, binds to specific mutant forms of α-galactosidase A to restore lysosomal activity. METHODS: A pharmacogenetic assay was used to identify the α-galactosidase A mutant forms amenable to migalastat. Six hundred Fabry disease-causing mutations were expressed in HEK-293 (HEK) cells; increases in α-galactosidase A activity were measured by a good laboratory practice (GLP)-validated assay (GLP HEK/Migalastat Amenability Assay). The predictive value of the assay was assessed based on pharmacodynamic responses to migalastat in phase II and III clinical studies. RESULTS: Comparison of the GLP HEK assay results in in vivo white blood cell α-galactosidase A responses to migalastat in male patients showed high sensitivity, specificity, and positive and negative predictive values (≥0.875). GLP HEK assay results were also predictive of decreases in kidney globotriaosylceramide in males and plasma globotriaosylsphingosine in males and females. The clinical study subset of amenable mutations (n = 51) was representative of all 268 amenable mutations identified by the GLP HEK assay. CONCLUSION: The GLP HEK assay is a clinically validated method of identifying male and female Fabry patients for treatment with migalastat.Genet Med 19 4, 430-438.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Enfermedad de Fabry/genética , Mutación , alfa-Galactosidasa/genética , 1-Desoxinojirimicina/administración & dosificación , 1-Desoxinojirimicina/farmacología , Bioensayo , Línea Celular , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Enfermedad de Fabry/tratamiento farmacológico , Femenino , Células HEK293 , Humanos , Leucocitos/efectos de los fármacos , Leucocitos/enzimología , Masculino , Valor Predictivo de las Pruebas , Estudios de Validación como Asunto
5.
PLoS One ; 10(8): e0134341, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26252393

RESUMEN

UNLABELLED: Migalastat HCl (AT1001, 1-Deoxygalactonojirimycin) is an investigational pharmacological chaperone for the treatment of α-galactosidase A (α-Gal A) deficiency, which leads to Fabry disease, an X-linked, lysosomal storage disorder. The currently approved, biologics-based therapy for Fabry disease is enzyme replacement therapy (ERT) with either agalsidase alfa (Replagal) or agalsidase beta (Fabrazyme). Based on preclinical data, migalastat HCl in combination with agalsidase is expected to result in the pharmacokinetic (PK) enhancement of agalsidase in plasma by increasing the systemic exposure of active agalsidase, thereby leading to increased cellular levels in disease-relevant tissues. This Phase 2a study design consisted of an open-label, fixed-treatment sequence that evaluated the effects of single oral doses of 150 mg or 450 mg migalastat HCl on the PK and tissue levels of intravenously infused agalsidase (0.2, 0.5, or 1.0 mg/kg) in male Fabry patients. As expected, intravenous administration of agalsidase alone resulted in increased α-Gal A activity in plasma, skin, and peripheral blood mononuclear cells (PBMCs) compared to baseline. Following co-administration of migalastat HCl and agalsidase, α-Gal A activity in plasma was further significantly increased 1.2- to 5.1-fold compared to agalsidase administration alone, in 22 of 23 patients (95.6%). Importantly, similar increases in skin and PBMC α-Gal A activity were seen following co-administration of migalastat HCl and agalsidase. The effects were not related to the administered migalastat HCl dose, as the 150 mg dose of migalastat HCl increased α-Gal A activity to the same extent as the 450 mg dose. Conversely, agalsidase had no effect on the plasma PK of migalastat. No migalastat HCl-related adverse events or drug-related tolerability issues were identified. TRIAL REGISTRATION: ClinicalTrials.gov NCT01196871.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Enfermedad de Fabry/tratamiento farmacológico , Enfermedad de Fabry/enzimología , Isoenzimas/uso terapéutico , alfa-Galactosidasa/metabolismo , 1-Desoxinojirimicina/administración & dosificación , 1-Desoxinojirimicina/sangre , 1-Desoxinojirimicina/farmacocinética , 1-Desoxinojirimicina/uso terapéutico , Administración Oral , Adulto , Área Bajo la Curva , Demografía , Enfermedad de Fabry/sangre , Humanos , Bombas de Infusión , Isoenzimas/administración & dosificación , Masculino , Persona de Mediana Edad , Proteínas Recombinantes , Piel/enzimología , alfa-Galactosidasa/administración & dosificación , alfa-Galactosidasa/sangre , alfa-Galactosidasa/uso terapéutico
6.
Mol Ther ; 23(7): 1169-1181, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25915924

RESUMEN

Fabry disease is an X-linked lysosomal storage disorder caused by mutations in the gene that encodes α-galactosidase A and is characterized by pathological accumulation of globotriaosylceramide and globotriaosylsphingosine. Earlier, the authors demonstrated that oral coadministration of the pharmacological chaperone AT1001 (migalastat HCl; 1-deoxygalactonojirimycin HCl) prior to intravenous administration of enzyme replacement therapy improved the pharmacological properties of the enzyme. In this study, the authors investigated the effects of coformulating AT1001 with a proprietary recombinant human α-galactosidase A (ATB100) into a single intravenous formulation. AT1001 increased the physical stability and reduced aggregation of ATB100 at neutral pH in vitro, and increased the potency for ATB100-mediated globotriaosylceramide reduction in cultured Fabry fibroblasts. In Fabry mice, AT1001 coformulation increased the total exposure of active enzyme, and increased ATB100 levels in cardiomyocytes, cardiac vascular endothelial cells, renal distal tubular epithelial cells, and glomerular cells, cell types that do not show substantial uptake with enzyme replacement therapy alone. Notably, AT1001 coformulation also leads to greater tissue globotriaosylceramide reduction when compared with ATB100 alone, which was positively correlated with reductions in plasma globotriaosylsphingosine. Collectively, these data indicate that intravenous administration of ATB100 coformulated with AT1001 may provide an improved therapy for Fabry disease and thus warrants further investigation.


Asunto(s)
Enfermedad de Fabry/tratamiento farmacológico , Chaperonas Moleculares/administración & dosificación , Oligopéptidos/administración & dosificación , alfa-Galactosidasa/administración & dosificación , Animales , Modelos Animales de Enfermedad , Combinación de Medicamentos , Terapia de Reemplazo Enzimático , Enfermedad de Fabry/patología , Fibroblastos/efectos de los fármacos , Humanos , Ratones , Mutación , Especificidad por Sustrato
7.
Mol Ther ; 23(7): 1138-1148, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25881001

RESUMEN

Lysosomal storage disorders (LSDs) are a group of inborn metabolic diseases caused by mutations in genes that encode proteins involved in different lysosomal functions, in most instances acidic hydrolases. Different therapeutic approaches have been developed to treat these disorders. Pharmacological chaperone therapy (PCT) is an emerging approach based on small-molecule ligands that selectively bind and stabilize mutant enzymes, increase their cellular levels, and improve lysosomal trafficking and activity. Compared to other approaches, PCT shows advantages, particularly in terms of oral administration, broad biodistribution, and positive impact on patients' quality of life. After preclinical in vitro and in vivo studies, PCT is now being translated in the first clinical trials, either as monotherapy or in combination with enzyme replacement therapy, for some of the most prevalent LSDs. For some LSDs, the results of the first clinical trials are encouraging and warrant further development. Future research in the field of PCT will be directed toward the identification of novel chaperones, including new allosteric drugs, and the exploitation of synergies between chaperone treatment and other therapeutic approaches.


Asunto(s)
Estabilidad de Enzimas/efectos de los fármacos , Enfermedades por Almacenamiento Lisosomal/terapia , Chaperonas Moleculares/uso terapéutico , Humanos , Ligandos , Enfermedades por Almacenamiento Lisosomal/genética , Lisosomas/efectos de los fármacos , Lisosomas/genética , Lisosomas/patología , Biosíntesis de Proteínas/efectos de los fármacos , Pliegue de Proteína/efectos de los fármacos , Proteolisis/efectos de los fármacos
8.
PLoS One ; 9(7): e102092, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25036864

RESUMEN

Pompe disease is an inherited lysosomal storage disorder that results from a deficiency in acid α-glucosidase (GAA) activity due to mutations in the GAA gene. Pompe disease is characterized by accumulation of lysosomal glycogen primarily in heart and skeletal muscles, which leads to progressive muscle weakness. We have shown previously that the small molecule pharmacological chaperone AT2220 (1-deoxynojirimycin hydrochloride, duvoglustat hydrochloride) binds and stabilizes wild-type as well as multiple mutant forms of GAA, and can lead to higher cellular levels of GAA. In this study, we examined the effect of AT2220 on mutant GAA, in vitro and in vivo, with a primary focus on the endoplasmic reticulum (ER)-retained P545L mutant form of human GAA (P545L GAA). AT2220 increased the specific activity of P545L GAA toward both natural (glycogen) and artificial substrates in vitro. Incubation with AT2220 also increased the ER export, lysosomal delivery, proteolytic processing, and stability of P545L GAA. In a new transgenic mouse model of Pompe disease that expresses human P545L on a Gaa knockout background (Tg/KO) and is characterized by reduced GAA activity and elevated glycogen levels in disease-relevant tissues, daily oral administration of AT2220 for 4 weeks resulted in significant and dose-dependent increases in mature lysosomal GAA isoforms and GAA activity in heart and skeletal muscles. Importantly, oral administration of AT2220 also resulted in significant glycogen reduction in disease-relevant tissues. Compared to daily administration, less-frequent AT2220 administration, including repeated cycles of 4 or 5 days with AT2220 followed by 3 or 2 days without drug, respectively, resulted in even greater glycogen reductions. Collectively, these data indicate that AT2220 increases the specific activity, trafficking, and lysosomal stability of P545L GAA, leads to increased levels of mature GAA in lysosomes, and promotes glycogen reduction in situ. As such, AT2220 may warrant further evaluation as a treatment for Pompe disease.


Asunto(s)
1-Desoxinojirimicina/farmacología , Glucano 1,4-alfa-Glucosidasa/genética , Glucano 1,4-alfa-Glucosidasa/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Glucógeno/metabolismo , Lisosomas/efectos de los fármacos , Mutación , 1-Desoxinojirimicina/administración & dosificación , 1-Desoxinojirimicina/farmacocinética , Administración Oral , Animales , Biocatálisis/efectos de los fármacos , Disponibilidad Biológica , Células COS , Chlorocebus aethiops , Modelos Animales de Enfermedad , Retículo Endoplásmico/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Estabilidad de Enzimas/efectos de los fármacos , Técnicas de Inactivación de Genes , Glucano 1,4-alfa-Glucosidasa/biosíntesis , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Humanos , Isoenzimas/biosíntesis , Isoenzimas/genética , Isoenzimas/metabolismo , Lisosomas/metabolismo , Ratones , Ratones Transgénicos , Proteínas Mutantes/biosíntesis , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Transporte de Proteínas/efectos de los fármacos , Proteolisis/efectos de los fármacos
9.
Bioorg Med Chem Lett ; 24(14): 3001-5, 2014 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-24894562

RESUMEN

Mutations in the gene that encodes the lysosomal enzyme acid ß-glucosidase lead to reduced cellular activity and accumulation of glycosphingolipid substrates, biochemical hallmarks of the lysosomal storage disorder Gaucher disease (GD). Recently such mutations have been identified as risk factors for Parkinson's disease (PD) and related disorders. Both gain-of-function (due to toxic cellular accumulation of mutant enzyme) and loss-of-function (due to accumulation of lipid substrates) hypotheses have been put forth to address the biochemical link between GD and PD. Similarly, links between Alzheimer's disease and other lysosomal enzyme deficiencies have begun to emerge. The use of pharmacological chaperones to restore the cellular trafficking and activity of mutant lysosomal enzymes may offer a novel approach to treat these debilitating neurodegenerative diseases.


Asunto(s)
Lisosomas/efectos de los fármacos , Enfermedades Neurodegenerativas/tratamiento farmacológico , Descubrimiento de Drogas , Glucosilceramidasa/genética , Glucosilceramidasa/metabolismo , Humanos , Lisosomas/enzimología , Lisosomas/metabolismo , Mutación , Enfermedades Neurodegenerativas/metabolismo
10.
PLoS One ; 8(3): e57631, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23472096

RESUMEN

Fabry disease (FD) results from mutations in the gene (GLA) that encodes the lysosomal enzyme α-galactosidase A (α-Gal A), and involves pathological accumulation of globotriaosylceramide (GL-3) and globotriaosylsphingosine (lyso-Gb3). Migalastat hydrochloride (GR181413A) is a pharmacological chaperone that selectively binds, stabilizes, and increases cellular levels of α-Gal A. Oral administration of migalastat HCl reduces tissue GL-3 in Fabry transgenic mice, and in urine and kidneys of some FD patients. A liquid chromatography-tandem mass spectrometry method was developed to measure lyso-Gb3 in mouse tissues and human plasma. Oral administration of migalastat HCl to transgenic mice reduced elevated lyso-Gb3 levels up to 64%, 59%, and 81% in kidney, heart, and skin, respectively, generally equal to or greater than observed for GL-3. Furthermore, baseline plasma lyso-Gb3 levels were markedly elevated in six male FD patients enrolled in Phase 2 studies. Oral administration of migalastat HCl (150 mg QOD) reduced urine GL-3 and plasma lyso-Gb3 in three subjects (range: 15% to 46% within 48 weeks of treatment). In contrast, three showed no reductions in either substrate. These results suggest that measurement of tissue and/or plasma lyso-Gb3 is feasible and may be warranted in future studies of migalastat HCl or other new potential therapies for FD.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Enfermedad de Fabry/genética , Glucolípidos/metabolismo , Esfingolípidos/metabolismo , Esfingosina/metabolismo , Trihexosilceramidas/metabolismo , 1-Desoxinojirimicina/farmacología , Administración Oral , Animales , Enfermedad de Fabry/sangre , Enfermedad de Fabry/tratamiento farmacológico , Glucolípidos/sangre , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Reproducibilidad de los Resultados , Esfingolípidos/sangre , Trihexosilceramidas/sangre , alfa-Galactosidasa/genética
11.
J Med Chem ; 56(7): 2705-25, 2013 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-23363020

RESUMEN

Lysosomal enzymes are responsible for the degradation of a wide variety of glycolipids, oligosaccharides, proteins, and glycoproteins. Inherited mutations in the genes that encode these proteins can lead to reduced stability of newly synthesized lysosomal enzymes. While often catalytically competent, the mutated enzymes are unable to efficiently pass the quality control mechanisms of the endoplasmic reticulum, resulting in reduced lysosomal trafficking, substrate accumulation, and cellular dysfunction. Pharmacological chaperones (PCs) are small molecules that bind and stabilize mutant lysosomal enzymes, thereby allowing proper cellular translocation. Such compounds have been shown to increase enzyme activity and reduce substrate burden in a number of preclinical models and clinical studies. In this Perspective, we review several of the lysosomal diseases for which PCs have been studied and the SAR of the various classes of molecules.


Asunto(s)
Enfermedades por Almacenamiento Lisosomal/tratamiento farmacológico , Chaperonas Moleculares/uso terapéutico , Línea Celular , Glucolípidos/metabolismo , Humanos , Relación Estructura-Actividad
12.
PLoS One ; 7(7): e40776, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22815812

RESUMEN

Pompe disease is an inherited lysosomal storage disease that results from a deficiency in the enzyme acid α-glucosidase (GAA), and is characterized by progressive accumulation of lysosomal glycogen primarily in heart and skeletal muscles. Recombinant human GAA (rhGAA) is the only approved enzyme replacement therapy (ERT) available for the treatment of Pompe disease. Although rhGAA has been shown to slow disease progression and improve some of the pathophysiogical manifestations, the infused enzyme tends to be unstable at neutral pH and body temperature, shows low uptake into some key target tissues, and may elicit immune responses that adversely affect tolerability and efficacy. We hypothesized that co-administration of the orally-available, small molecule pharmacological chaperone AT2220 (1-deoxynojirimycin hydrochloride, duvoglustat hydrochloride) may improve the pharmacological properties of rhGAA via binding and stabilization. AT2220 co-incubation prevented rhGAA denaturation and loss of activity in vitro at neutral pH and 37°C in both buffer and blood. In addition, oral pre-administration of AT2220 to rats led to a greater than two-fold increase in the circulating half-life of intravenous rhGAA. Importantly, co-administration of AT2220 and rhGAA to GAA knock-out (KO) mice resulted in significantly greater rhGAA levels in plasma, and greater uptake and glycogen reduction in heart and skeletal muscles, compared to administration of rhGAA alone. Collectively, these preclinical data highlight the potentially beneficial effects of AT2220 on rhGAA in vitro and in vivo. As such, a Phase 2 clinical study has been initiated to investigate the effects of co-administered AT2220 on rhGAA in Pompe patients.


Asunto(s)
1-Desoxinojirimicina/uso terapéutico , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Glucógeno/metabolismo , Proteínas Recombinantes/metabolismo , alfa-Glucosidasas/metabolismo , 1-Desoxinojirimicina/administración & dosificación , 1-Desoxinojirimicina/farmacología , Animales , Tampones (Química) , Modelos Animales de Enfermedad , Activación Enzimática/efectos de los fármacos , Estabilidad de Enzimas/efectos de los fármacos , Semivida , Humanos , Ratones , Ratones Noqueados , Desnaturalización Proteica/efectos de los fármacos , Ratas , Proteínas Recombinantes/sangre , alfa-Glucosidasas/administración & dosificación , alfa-Glucosidasas/sangre
13.
J Neurosci ; 32(15): 5223-36, 2012 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-22496568

RESUMEN

Alterations in the lipid composition of endosomal-lysosomal membranes may constitute an early event in Alzheimer's disease (AD) pathogenesis. In this study, we investigated the possibility that GM2 ganglioside accumulation in a mouse model of Sandhoff disease might be associated with the accumulation of intraneuronal and extracellular proteins commonly observed in AD. Our results show intraneuronal accumulation of amyloid-ß peptide (Aß)-like, α-synuclein-like, and phospho-tau-like immunoreactivity in the brains of ß-hexosaminidase knock-out (HEXB KO) mice. Biochemical and immunohistochemical analyses confirmed that at least some of the intraneuronal Aß-like immunoreactivity (iAß-LIR) represents amyloid precursor protein C-terminal fragments (APP-CTFs) and/or Aß. In addition, we observed increased levels of Aß40 and Aß42 peptides in the lipid-associated fraction of HEXB KO mouse brains, and intraneuronal accumulation of ganglioside-bound Aß (GAß) immunoreactivity in a brain region-specific manner. Furthermore, α-synuclein and APP-CTFs and/or Aß were found to accumulate in different regions of the substantia nigra, indicating different mechanisms of accumulation or turnover pathways. Based on the localization of the accumulated iAß-LIR to endosomes, lysosomes, and autophagosomes, we conclude that a significant accumulation of iAß-LIR may be associated with the lysosomal-autophagic turnover of Aß and fragments of APP-containing Aß epitopes. Importantly, intraneuronal GAß immunoreactivity, a proposed prefibrillar aggregate found in AD, was found to accumulate throughout the frontal cortices of postmortem human GM1 gangliosidosis, Sandhoff disease, and Tay-Sachs disease brains. Together, these results establish an association between the accumulation of gangliosides, autophagic vacuoles, and the intraneuronal accumulation of proteins associated with AD.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Gangliósidos/metabolismo , Hexosaminidasa B/genética , Lisosomas/fisiología , Enfermedad de Sandhoff/patología , Adulto , Animales , Western Blotting , Química Encefálica/genética , Química Encefálica/fisiología , Preescolar , Gangliósido G(M2)/metabolismo , Humanos , Inmunohistoquímica , Lactante , Metabolismo de los Lípidos , Bulbo Raquídeo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Médula Espinal/metabolismo , Sustancia Negra/metabolismo , Adulto Joven , alfa-Sinucleína/metabolismo , Proteínas tau/metabolismo
14.
Mol Ther ; 20(4): 717-26, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22215019

RESUMEN

Fabry disease is an X-linked lysosomal storage disorder (LSD) caused by mutations in the gene (GLA) that encodes the lysosomal hydrolase α-galactosidase A (α-Gal A), and is characterized by pathological accumulation of the substrate, globotriaosylceramide (GL-3). Regular infusion of recombinant human α-Gal A (rhα-Gal A), termed enzyme replacement therapy (ERT), is the primary treatment for Fabry disease. However, rhα-Gal A has low physical stability, a short circulating half-life, and variable uptake into different disease-relevant tissues. We hypothesized that coadministration of the orally available, small molecule pharmacological chaperone AT1001 (GR181413A, 1-deoxygalactonojirimycin, migalastat hydrochloride) may improve the pharmacological properties of rhα-Gal A via binding and stabilization. AT1001 prevented rhα-Gal A denaturation and activity loss in vitro at neutral pH and 37 °C. Coincubation of Fabry fibroblasts with rhα-Gal A and AT1001 resulted in up to fourfold higher cellular α-Gal A and ~30% greater GL-3 reduction compared to rhα-Gal A alone. Furthermore, coadministration of AT1001 to rats increased the circulating half-life of rhα-Gal A by >2.5-fold, and in GLA knockout mice resulted in up to fivefold higher α-Gal A levels and fourfold greater GL-3 reduction than rhα-Gal A alone. Collectively, these data highlight the potentially beneficial effects of AT1001 on rhα-Gal A, thus warranting clinical investigation.


Asunto(s)
Terapia de Reemplazo Enzimático/métodos , Enfermedad de Fabry/tratamiento farmacológico , Oligopéptidos/uso terapéutico , Proteínas Recombinantes/uso terapéutico , alfa-Galactosidasa/uso terapéutico , Animales , Western Blotting , Enfermedad de Fabry/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Ratones , Ratas , Trihexosilceramidas/metabolismo
16.
Assay Drug Dev Technol ; 9(3): 213-35, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21612550

RESUMEN

Many human diseases result from mutations in specific genes. Once translated, the resulting aberrant proteins may be functionally competent and produced at near-normal levels. However, because of the mutations, the proteins are recognized by the quality control system of the endoplasmic reticulum and are not processed or trafficked correctly, ultimately leading to cellular dysfunction and disease. Pharmacological chaperones (PCs) are small molecules designed to mitigate this problem by selectively binding and stabilizing their target protein, thus reducing premature degradation, facilitating intracellular trafficking, and increasing cellular activity. Partial or complete restoration of normal function by PCs has been shown for numerous types of mutant proteins, including secreted proteins, transcription factors, ion channels, G protein-coupled receptors, and, importantly, lysosomal enzymes. Collectively, lysosomal storage disorders (LSDs) result from genetic mutations in the genes that encode specific lysosomal enzymes, leading to a deficiency in essential enzymatic activity and cellular accumulation of the respective substrate. To date, over 50 different LSDs have been identified, several of which are treated clinically with enzyme replacement therapy or substrate reduction therapy, although insufficiently in some cases. Importantly, a wide range of in vitro assays are now available to measure mutant lysosomal enzyme interaction with and stabilization by PCs, as well as subsequent increases in cellular enzyme levels and function. The application of these assays to the identification and characterization of candidate PCs for mutant lysosomal enzymes will be discussed in this review. In addition, considerations for the successful in vivo use and development of PCs to treat LSDs will be discussed.


Asunto(s)
Bioensayo/métodos , Diseño de Fármacos , Enfermedades por Almacenamiento Lisosomal/tratamiento farmacológico , Chaperonas Moleculares/química , Chaperonas Moleculares/uso terapéutico , Tecnología Farmacéutica/métodos , Animales , Humanos
17.
Hum Mutat ; 32(8): 965-77, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21598360

RESUMEN

Fabry disease is caused by mutations in the gene (GLA) that encodes α-galactosidase A (α-Gal A). The iminosugar AT1001 (GR181413A, migalastat hydrochloride, 1-deoxygalactonojirimycin) is a pharmacological chaperone that selectively binds and stabilizes α-Gal A, increasing total cellular levels and activity for some mutant forms (defined as "responsive"). In this study, we developed a cell-based assay in cultured HEK-293 cells to identify mutant forms of α-Gal A that are responsive to AT1001. Concentration-dependent increases in α-Gal A activity in response to AT1001 were shown for 49 (60%) of 81 mutant forms. The responses of α-Gal A mutant forms were generally consistent with the responses observed in male Fabry patient-derived lymphoblasts. Importantly, the HEK-293 cell responses of 19 α-Gal A mutant forms to a clinically achievable concentration of AT1001 (10 µM) were generally consistent with observed increases in α-Gal A activity in peripheral blood mononuclear cells from male Fabry patients orally administered AT1001 during Phase 2 clinical studies. This indicates that the cell-based responses can identify mutant forms of α-Gal A that are likely to respond to AT1001 in vivo. Thus, the HEK-293 cell-based assay may be a useful aid in the identification of Fabry patients with AT1001-responsive mutant forms.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Enfermedad de Fabry/genética , Proteínas Mutantes/análisis , alfa-Galactosidasa/genética , 1-Desoxinojirimicina/metabolismo , 1-Desoxinojirimicina/farmacología , Bioensayo , Activación Enzimática/efectos de los fármacos , Enfermedad de Fabry/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Humanos , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Masculino , Proteínas Mutantes/metabolismo , Mutación Puntual/genética , Conformación Proteica , alfa-Galactosidasa/química , alfa-Galactosidasa/metabolismo
18.
J Pharmacol Exp Ther ; 335(3): 520-32, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20826565

RESUMEN

Heterozygous null mutations in the melanocortin-4 receptor (MC4R) cause early-onset obesity in humans, indicating that metabolic homeostasis is sensitive to quantitative variation in MC4R function. Most of the obesity-causing MC4R mutations functionally characterized so far lead to intracellular retention of receptors by the cell's quality control system. Thus, recovering cell surface expression of mutant MC4Rs could have a beneficial therapeutic value. We tested a pharmacological chaperone approach to restore cell surface expression and function of 10 different mutant forms of human melanocortin-4 receptor found in obese patients. Five cell-permeant MC4R-selective ligands were tested and displayed pharmacological chaperone activities, restoring cell surface targeting and function of the receptors with distinct efficacy profiles for the different mutations. Such mutation-specific efficacies suggested a structure-activity relationship between compounds and mutant receptor conformations that may open a path toward personalized therapy. In addition, one of the five pharmacological chaperones restored function to most of the mutant receptors tested. Combined with its ability to reach the central nervous system and its selectivity for the MC4R, this pharmacological chaperone may represent a candidate for the development of a targeted therapy suitable for a large subset of patients with MC4R-deficient obesity.


Asunto(s)
Mutación Missense/fisiología , Obesidad/tratamiento farmacológico , Obesidad/genética , Pliegue de Proteína/efectos de los fármacos , Receptor de Melanocortina Tipo 4/antagonistas & inhibidores , Edad de Inicio , Animales , Sitios de Unión , Encéfalo/metabolismo , Membrana Celular/metabolismo , AMP Cíclico/metabolismo , Endocitosis/efectos de los fármacos , Células HEK293 , Humanos , Cinética , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Estructura Molecular , Preparaciones Farmacéuticas/sangre , Preparaciones Farmacéuticas/química , Preparaciones Farmacéuticas/metabolismo , Farmacocinética , Transporte de Proteínas/efectos de los fármacos , Receptor de Melanocortina Tipo 4/agonistas , Receptor de Melanocortina Tipo 4/química , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Transfección , alfa-MSH/análogos & derivados , alfa-MSH/farmacología
19.
FEBS J ; 277(7): 1618-38, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20148966

RESUMEN

Gaucher disease is caused by mutations in the gene that encodes the lysosomal enzyme acid beta-glucosidase (GCase). We have shown previously that the small molecule pharmacological chaperone isofagomine (IFG) binds and stabilizes N370S GCase, resulting in increased lysosomal trafficking and cellular activity. In this study, we investigated the effect of IFG on L444P GCase. Incubation of Gaucher patient-derived lymphoblastoid cell lines (LCLs) or fibroblasts with IFG led to approximately 3.5- and 1.3-fold increases in L444P GCase activity, respectively, as measured in cell lysates. The effect in fibroblasts was increased approximately 2-fold using glycoprotein-enrichment, GCase-immunocapture, or by incubating cells overnight in IFG-free media prior to assay, methods designed to maximize GCase activity by reducing IFG carryover and inhibition in the enzymatic assay. IFG incubation also increased the lysosomal trafficking and in situ activity of L444P GCase in intact cells, as measured by reduction in endogenous glucosylceramide levels. Importantly, this reduction was seen only following three-day incubation in IFG-free media, underscoring the importance of IFG removal to restore lysosomal GCase activity. In mice expressing murine L444P GCase, oral administration of IFG resulted in significant increases (2- to 5-fold) in GCase activity in disease-relevant tissues, including brain. Additionally, eight-week IFG administration significantly lowered plasma chitin III and IgG levels, and 24-week administration significantly reduced spleen and liver weights. Taken together, these data suggest that IFG can increase the lysosomal activity of L444P GCase in cells and tissues. Moreover, IFG is orally available and distributes into multiple tissues, including brain, and may thus merit therapeutic evaluation for patients with neuronopathic and non-neuronopathic Gaucher disease.


Asunto(s)
Enfermedad de Gaucher/genética , Iminopiranosas/química , Enfermedades por Almacenamiento Lisosomal/genética , Mutación , beta-Glucosidasa/genética , Animales , Relación Dosis-Respuesta a Droga , Fibroblastos/metabolismo , Glucosilceramidasa/metabolismo , Humanos , Masculino , Ratones , Microscopía Confocal/métodos , Chaperonas Moleculares/metabolismo , Ratas , Ratas Sprague-Dawley
20.
Mol Ther ; 18(1): 23-33, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19773742

RESUMEN

Fabry disease is an X-linked lysosomal storage disorder caused by a deficiency in alpha-galactosidase A (alpha-Gal A) activity and subsequent accumulation of the substrate globotriaosylceramide (GL-3), which contributes to disease pathology. The pharmacological chaperone (PC) DGJ (1-deoxygalactonojirimycin) binds and stabilizes alpha-Gal A, increasing enzyme levels in cultured cells and in vivo. The ability of DGJ to reduce GL-3 in vivo was investigated using transgenic (Tg) mice that express a mutant form of human alpha-Gal A (R301Q) on a knockout background (Tg/KO), which leads to GL-3 accumulation in disease-relevant tissues. Four-week daily oral administration of DGJ to Tg/KO mice resulted in significant and dose-dependent increases in alpha-Gal A activity, with concomitant GL-3 reduction in skin, heart, kidney, brain, and plasma; 24-week administration resulted in even greater reductions. Compared to daily administration, less frequent DGJ administration, including repeated cycles of 4 days with DGJ followed by 3 days without or every other day with DGJ, resulted in even greater GL-3 reductions that were comparable to those obtained with Fabrazyme. Collectively, these data indicate that oral administration of DGJ increases mutant alpha-Gal A activity and reduces GL-3 in disease-relevant tissues in Tg/KO mice, and thus merits further evaluation as a treatment for Fabry disease.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Enfermedad de Fabry/tratamiento farmacológico , Trihexosilceramidas/metabolismo , 1-Desoxinojirimicina/uso terapéutico , Animales , Western Blotting , Modelos Animales de Enfermedad , Enfermedad de Fabry/genética , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , alfa-Galactosidasa/antagonistas & inhibidores , alfa-Galactosidasa/genética , alfa-Galactosidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...