Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Pharmacokinet Pharmacodyn ; 50(6): 461-474, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37535240

RESUMEN

Avalglucosidase alfa (AVAL) was approved in the United States (2021) for patients with late-onset Pompe disease (LOPD), aged ≥ 1 year. In the present study, pharmacokinetic (PK) simulations were conducted to propose alternative dosing regimens for pediatric LOPD patients based on a bodyweight cut-off. Population PK (PopPK) analysis was performed using nonlinear mixed effect modeling approach on pooled data from three clinical trials with LOPD patients, and a phase 2 study (NCT03019406) with infantile-onset Pompe disease (IOPD: 1-12 years) patients. A total of 2257 concentration-time points from 91 patients (LOPD, n = 75; IOPD, n = 16) were included in the analysis. The model was bodyweight dependent allometric scaling with time varying bodyweight included on clearance and distribution volume. Simulations were performed for two dosing regimens (20 mg/kg or 40 mg/kg) with different bodyweight cut-off (25, 30, 35 and 40 kg) by generating virtual pediatric (1-17 years) and adult patients. Corresponding simulated individual exposures (maximal concentration, Cmax and area under the curve in the 2-week dosing interval, AUC2W), and distributions were calculated. It was found that dosing of 40 mg/kg and 20 mg/kg in pediatric patients < 30 kg and ≥ 30 kg, respectively, achieved similar AVAL exposure (based on AUC2W) to adult patients receiving 20 mg/kg. PK simulations conducted on the basis of this model provided supporting data for the currently approved US labelling for dosing adapted bodyweight in LOPD patients ≥ 1 year by USFDA.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II , Adulto , Humanos , Niño , Estados Unidos , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Enfermedad del Almacenamiento de Glucógeno Tipo II/inducido químicamente , Enfermedad del Almacenamiento de Glucógeno Tipo II/epidemiología , alfa-Glucosidasas/efectos adversos , alfa-Glucosidasas/metabolismo , Peso Corporal , Cinética
2.
Mol Genet Metab ; 137(1-2): 228-240, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35718712

RESUMEN

Alglucosidase alpha is an orphan drug approved for enzyme replacement therapy (ERT) in Pompe disease (PD); however, its efficacy is limited in skeletal muscle because of a partial blockage of autophagic flux that hinders intracellular trafficking and enzyme delivery. Adjunctive therapies that enhance autophagic flux and protect mitochondrial integrity may alleviate autophagic blockage and oxidative stress and thereby improve ERT efficacy in PD. In this study, we compared the benefits of ERT combined with a ketogenic diet (ERT-KETO), daily administration of an oral ketone precursor (1,3-butanediol; ERT-BD), a multi-ingredient antioxidant diet (ERT-MITO; CoQ10, α-lipoic acid, vitamin E, beetroot extract, HMB, creatine, and citrulline), or co-therapy with the ketone precursor and multi-ingredient antioxidants (ERT-BD-MITO) on skeletal muscle pathology in GAA-KO mice. We found that two months of 1,3-BD administration raised circulatory ketone levels to ≥1.2 mM, attenuated autophagic buildup in type 2 muscle fibers, and preserved muscle strength and function in ERT-treated GAA-KO mice. Collectively, ERT-BD was more effective vs. standard ERT and ERT-KETO in terms of autophagic clearance, dampening of oxidative stress, and muscle maintenance. However, the addition of multi-ingredient antioxidants (ERT-BD-MITO) provided the most consistent benefits across all outcome measures and normalized mitochondrial protein expression in GAA-KO mice. We therefore conclude that nutritional co-therapy with 1,3-butanediol and multi-ingredient antioxidants may provide an alternative to ketogenic diets for inducing ketosis and enhancing autophagic flux in PD patients.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II , Ácido Tióctico , Ratones , Animales , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Creatina/metabolismo , Citrulina , alfa-Glucosidasas/genética , alfa-Glucosidasas/uso terapéutico , alfa-Glucosidasas/metabolismo , Terapia de Reemplazo Enzimático , Músculo Esquelético/metabolismo , Proteínas Mitocondriales/metabolismo , Vitamina E/farmacología , Cetonas/metabolismo , Cetonas/farmacología , Cetonas/uso terapéutico
3.
Ital J Pediatr ; 48(1): 48, 2022 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-35346323

RESUMEN

BACKGROUND: Pompe disease (PD) is a disorder of glycogen metabolism conditioning a progressive and life conditioning myopathy. Enzyme replacement therapy (ERT) is currently the best treatment option for PD, but is not resolutive. While other potential therapeutic approaches have been reported before, these have never been tried as co- treatments. L-alanine oral supplementation (LAOS) has been proven to reduce muscle breakdown: we hereby report the first case of supplementation on a PD patient on ERT. CASE PRESENTATION: F. is a 9 y.o. infantile onset Pompe Disease (IOPD) girl ERT-treated since age 1 developing a progressive myopathy. We started her on LAOS and performed assessments at baseline, 6 and 9 months. At baseline, F.'s weight, height and BMI were within normal ranges, while body composition showed low fat mass -FM and high resting energy expenditure-REE levels. After LAOS, a progressive FM increase and REE reduction could be observed both at 6 and 9 months. CONCLUSIONS: ERT is not curative for PD patients thus additional treatments could be considered to improve outcomes. Our patient showed physical signs of inability to accumulate energy when exclusively on ERT, while FM increase and REE reduction occurred when supplemented with LAOS, likely reflecting anabolic pathways' implementation. This is the first case reporting potential LAOS benefits in PD-on ERT patients. Longitudinal case control studies are yet needed to evaluate possible efficacy of combined LAOS And ERT treatment in PD patients.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II , Alanina/uso terapéutico , Suplementos Dietéticos , Terapia de Reemplazo Enzimático , Femenino , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Humanos , Lactante , alfa-Glucosidasas/metabolismo , alfa-Glucosidasas/uso terapéutico
4.
J Neuromuscul Dis ; 8(3): 401-417, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33720849

RESUMEN

BACKGROUND: Metabolic myopathies are a heterogenous group of muscle diseases typically characterized by exercise intolerance, myalgia and progressive muscle weakness. Effective treatments for some of these diseases are available, but while our understanding of the pathogenesis of metabolic myopathies related to glycogen storage, lipid metabolism and ß-oxidation is well established, evidence linking treatments with the precise causative genetic defect is lacking. OBJECTIVE: The objective of this study was to collate all published evidence on pharmacological therapies for the aforementioned metabolic myopathies and link this to the genetic mutation in a format amenable to databasing for further computational use in line with the principles of the "treatabolome" project. METHODS: A systematic literature review was conducted to retrieve all levels of evidence examining the therapeutic efficacy of pharmacological treatments on metabolic myopathies related to glycogen storage and lipid metabolism. A key inclusion criterion was the availability of the genetic variant of the treated patients in order to link treatment outcome with the genetic defect. RESULTS: Of the 1,085 articles initially identified, 268 full-text articles were assessed for eligibility, of which 87 were carried over into the final data extraction. The most studied metabolic myopathies were Pompe disease (45 articles), multiple acyl-CoA dehydrogenase deficiency related to mutations in the ETFDH gene (15 articles) and systemic primary carnitine deficiency (8 articles). The most studied therapeutic management strategies for these diseases were enzyme replacement therapy, riboflavin, and carnitine supplementation, respectively. CONCLUSIONS: This systematic review provides evidence for treatments of metabolic myopathies linked with the genetic defect in a computationally accessible format suitable for databasing in the treatabolome system, which will enable clinicians to acquire evidence on appropriate therapeutic options for their patient at the time of diagnosis.


Asunto(s)
Glucógeno/metabolismo , Metabolismo de los Lípidos , Errores Innatos del Metabolismo/tratamiento farmacológico , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Humanos , Deficiencia Múltiple de Acil Coenzima A Deshidrogenasa/tratamiento farmacológico , Debilidad Muscular , Mutación
5.
Rev. Enferm. UERJ (Online) ; 28: e54407, jan.-dez. 2020.
Artículo en Inglés, Portugués | LILACS, BDENF | ID: biblio-1146478

RESUMEN

Objetivo: investigar o conhecimento e práticas da equipe de enfermagem em relação ao cuidado à criança com Doença de Pompe em terapia intensiva. Método: trata-se de um estudo descritivo com abordagem qualitativa. A coleta de dados foi realizada com entrevista semiestruturada com enfermeiras e técnicas em enfermagem que atuavam na Unidade de Terapia Intensiva Neonatal de um hospital do Rio Grande do Sul, após aprovação pelo Comitê de Ética em Pesquisa. Os dados foram analisados pela análise de conteúdo. Resultados: as profissionais enfatizaram experiências que superam procedimentos técnicos, na busca de fornecer um cuidado integral qualificado e seguro, para proporcionar vivências mais próximas de um lar para a criança e familiares. Conclusão: a equipe de enfermagem possui conhecimentos para o cuidado e atua de forma multiprofissional. Conclui-se que estudos relacionados às doenças raras oferecem subsídios para qualificar o cuidado de enfermagem.


Objective: to investigate nursing team knowledge and practices regarding care for children with Pompe disease in intensive care. Method: in this qualitative, descriptive study, data were collected by semi-structured interviews of nurses and nursing technicians working in the neonatal intensive care unit of a hospital in Rio Grande do Sul, after ethics committee approval. Data were subjected to content analysis. Result: the nurses emphasized experiences that go beyond technical procedures, in the endeavor to provide safe and qualified comprehensive care in order to provide experiences closer to a home for the children and their families. Conclusion: the nursing team was knowledgeable about care and worked in multidisciplinary manner. It was concluded that studies relating to rare disease offer input to inform nursing care.


Objetivo: investigar el conocimiento y las prácticas del equipo de enfermería sobre el cuidado de niños con enfermedad de Pompe en cuidados intensivos. Método: en este estudio cualitativo descriptivo, los datos fueron recolectados mediante entrevistas semiestructuradas a enfermeras y técnicos de enfermería que laboran en la unidad de cuidados intensivos neonatales de un hospital de Rio Grande do Sul, previa aprobación del comité de ética. Los datos se sometieron a análisis de contenido. Resultado: las enfermeras destacaron experiencias que van más allá de los procedimientos técnicos, en el afán de brindar una atención integral segura y calificada con el fin de brindar experiencias más cercanas a un hogar para los niños y sus familias. Conclusión: el equipo de enfermería tenía conocimiento del cuidado y trabajaba de manera multidisciplinar. Se concluyó que los estudios relacionados con las enfermedades raras ofrecen información para informar la atención de enfermería.


Asunto(s)
Humanos , Femenino , Recién Nacido , Adulto , Unidades de Cuidado Intensivo Neonatal , Enfermedad del Almacenamiento de Glucógeno Tipo II/enfermería , Cuidado Intensivo Neonatal , Competencia Clínica , Atención de Enfermería , Grupo de Enfermería , Brasil , Enfermedades Raras/enfermería , Investigación Cualitativa , Enfermeros no Diplomados , Enfermeras y Enfermeros
6.
Neuromuscul Disord ; 30(11): 904-914, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33127291

RESUMEN

To address progressive respiratory muscle weakness in late-onset Pompe disease (LOPD), we developed a 12-week respiratory muscle training (RMT) program. In this exploratory, double-blind, randomized control trial, 22 adults with LOPD were randomized to RMT or sham-RMT. The primary outcome was maximum inspiratory pressure (MIP). Secondary and exploratory outcomes included maximum expiratory pressure (MEP), peak cough flow, diaphragm ultrasound, polysomnography, patient-reported outcomes, and measures of gross motor function. MIP increased 7.6 cmH2O (15.9) in the treatment group and 2.7 cmH2O (7.6) in the control group (P = 0.4670). MEP increased 14.0 cmH2O (25.9) in the treatment group and 0.0 cmH2O (12.0) in the control group (P = 0.1854). The only statistically significant differences in secondary/exploratory outcomes were improvements in time to climb 4 steps (P = 0.0346) and daytime sleepiness (P = 0.0160). The magnitude of changes in MIP and MEP in the treatment group were consistent with our pilot findings but did not achieve statistical significance in comparison to controls. Explanations for this include inadequate power and baseline differences in subject characteristics between groups. Additionally, control group subjects appeared to exhibit an active response to sham-RMT and therefore sham-RMT may not be an optimal control condition for RMT in LOPD.


Asunto(s)
Ejercicios Respiratorios/métodos , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Músculos Respiratorios/fisiopatología , Adulto , Anciano , Diafragma/fisiopatología , Método Doble Ciego , Femenino , Humanos , Inhalación/fisiología , Masculino , Persona de Mediana Edad , Debilidad Muscular/fisiopatología , Pruebas de Función Respiratoria , Ultrasonografía
7.
J Pediatr Rehabil Med ; 13(1): 71-80, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32176666

RESUMEN

BACKGROUND: Respiratory muscle weakness is a primary cause of morbidity and mortality in patients with Pompe disease. We previously described the effects of our 12-week respiratory muscle training (RMT) regimen in 8 adults with late-onset Pompe disease [1] and 2 children with infantile-onset Pompe disease [2]. CASE REPORT: Here we describe repeat enrollment by one of the pediatric participants who completed a second 12-week RMT regimen after 7 months of detraining. We investigated the effects of two 12-week RMT regimens (RMT #1, RMT #2) using a single-participant A-B-A experimental design. Primary outcome measures were maximum inspiratory pressure (MIP) and maximum expiratory pressure (MEP). Effect sizes for changes in MIP and MEP were determined using Cohen's d statistic. Exploratory outcomes targeted motor function. RELEVANCE: From pretest to posttest, RMT #2 was associated with a 25% increase in MIP and a 22% increase in MEP, corresponding with very large effect sizes (d= 2.92 and d= 2.65, respectively). Following two 12-week RMT regimens over 16 months, MIP increased by 69% and MEP increased by 97%, corresponding with very large effect sizes (d= 3.57 and d= 5.10, respectively). MIP and MEP were largely stable over 7 months of detraining between regimens. Magnitude of change was greater for RMT #1 relative to RMT #2.


Asunto(s)
Ejercicios Respiratorios/métodos , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Enfermedad del Almacenamiento de Glucógeno Tipo II/rehabilitación , Insuficiencia Respiratoria/fisiopatología , Insuficiencia Respiratoria/rehabilitación , Femenino , Enfermedad del Almacenamiento de Glucógeno Tipo II/complicaciones , Humanos , Lactante , Fuerza Muscular/fisiología , Insuficiencia Respiratoria/complicaciones , Músculos Respiratorios/fisiopatología , Retratamiento , Resultado del Tratamiento
8.
Biochim Biophys Acta Mol Basis Dis ; 1866(5): 165662, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-31917327

RESUMEN

Pompe disease (PD) is an autosomal recessive muscular disorder caused by deficiency of the glycogen hydrolytic enzyme acid α-glucosidase (GAA). The enzyme replacement therapy, currently the only available therapy for PD patients, is efficacious in improving cardiomyopathy in the infantile form, but not equally effective in the late onset cases with involvement of skeletal muscle. Correction of the skeletal muscle phenotype has indeed been challenging, probably due to concomitant dysfunctional autophagy. The increasing attention to the pathogenic mechanisms of PD and the search of new therapeutic strategies prompted us to generate and characterize a novel transient PD model, using zebrafish. Our model presented increased glycogen content, markedly altered motor behavior and increased lysosome content, in addition to altered expression of the autophagy-related transcripts and proteins Beclin1, p62 and Lc3b. Furthermore, the model was used to assess the beneficial effects of 3-bromopyruvic acid (3-BrPA). Treatment with 3-BrPA induced amelioration of the model phenotypes regarding glycogen storage, motility behavior and autophagy-related transcripts and proteins. Our zebrafish PD model recapitulates most of the defects observed in human patients, proving to be a powerful translational model. Moreover, 3-BrPA unveiled to be a promising compound for treatment of conditions with glycogen accumulation.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Glucógeno/metabolismo , Hexoquinasa/antagonistas & inhibidores , Piruvatos/farmacología , Animales , Animales Modificados Genéticamente , Autofagia/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Técnicas de Silenciamiento del Gen , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Glucólisis/efectos de los fármacos , Hexoquinasa/metabolismo , Humanos , Lisosomas , Microscopía Electrónica , Morfolinos/administración & dosificación , Morfolinos/genética , Actividad Motora/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Músculo Esquelético/ultraestructura , Piruvatos/uso terapéutico , Pez Cebra , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo , alfa-Glucosidasas/genética , alfa-Glucosidasas/metabolismo
9.
Neurol Sci ; 41(4): 859-868, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31811531

RESUMEN

Aerobic exercise, training to sustain motor ability, and respiratory rehabilitation may improve general functioning and quality of life (QoL) in neuromuscular disorders. Patients with late-onset Pompe disease (LOPD) typically show progressive muscle weakness, respiratory dysfunction and minor cardiac involvement. Characteristics and modalities of motor and respiratory rehabilitation in LOPD are not well defined and specific guidelines are lacking. Therefore, we evaluated the role of physical activity, therapeutic exercise, and pulmonary rehabilitation programs in order to promote an appropriate management of motor and respiratory dysfunctions and improve QoL in patients with LOPD. We propose two operational protocols: one for an adapted physical activity (APA) plan and the other for an individual rehabilitation plan, particularly focused on therapeutic exercise (TE) and respiratory rehabilitation.


Asunto(s)
Ejercicios Respiratorios/métodos , Terapia por Ejercicio/métodos , Ejercicio Físico , Enfermedad del Almacenamiento de Glucógeno Tipo II/rehabilitación , Enfermedades Musculares/rehabilitación , Adolescente , Adulto , Edad de Inicio , Ciclismo , Niño , Protocolos Clínicos , Ejercicio Físico/fisiología , Enfermedad del Almacenamiento de Glucógeno Tipo II/complicaciones , Humanos , Ejercicios de Estiramiento Muscular , Enfermedades Musculares/etiología , Entrenamiento de Fuerza/métodos
10.
Hum Mol Genet ; 28(23): 3880-3894, 2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31518394

RESUMEN

Pompe disease (OMIM # 232300) is a glycogen storage disease caused by autosomal recessive mutations of the gene encoding alpha-1,4-glucosidase (GAA; EC 3.2.1.20). Despite the relatively effective employment of enzyme replacement therapy, some critical medical issues still exist in patients with this disease, including the persistence of abnormalities in the central nervous system (CNS), probably because of the inability of the recombinant GAA to pass through the blood-brain barrier. To address this issue, identification of more therapeutic agents that target the CNS of patients with Pompe disease may be required. In this study, we derived neuronal cells from Pompe disease-induced pluripotent stem cells (Pom-iPSCs) and proved that they are able to recapitulate the hallmark cellular and biochemical phenotypes of Pompe disease. Using the Pom-iPSC-derived neurons as an in vitro drug-testing model, we then identified three compounds, ebselen, wortmannin and PX-866, with therapeutic potential to alleviate Pompe disease-associated pathological phenotypes in the neurons derived from Pom-iPSCs. We confirmed that all three compounds were able to enhance the GAA activity in the Pom-iPSC-derived neurons. Moreover, they were able to enhance the GAA activity in several important internal organs of GAA-deficient mice when co-injected with recombinant human GAA, and we found that intraperitoneal injection of ebselen was able to promote the GAA activity of the GAA-heterozygous mouse brain. Our results prove the usefulness of Pom-iPSC-derived neuronal populations for identifying new compounds with therapeutic potential.


Asunto(s)
Azoles/administración & dosificación , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Compuestos de Organoselenio/administración & dosificación , alfa-Glucosidasas/metabolismo , Animales , Azoles/farmacología , Barrera Hematoencefálica , Encéfalo/metabolismo , Técnicas de Cultivo de Célula , Células Cultivadas , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Femenino , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Gonanos/farmacología , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Inyecciones Intraperitoneales , Isoindoles , Masculino , Ratones , Células-Madre Neurales/efectos de los fármacos , Compuestos de Organoselenio/farmacología , Wortmanina/farmacología , alfa-Glucosidasas/genética
11.
Mol Genet Metab ; 127(4): 346-354, 2019 08.
Artículo en Español | MEDLINE | ID: mdl-31303277

RESUMEN

INTRODUCTION: Morbidity and mortality in adults with late-onset Pompe disease (LOPD) results primarily from persistent progressive respiratory muscle weakness despite treatment with enzyme replacement therapy (ERT). To address this need, we have developed a 12-week respiratory muscle training (RMT) program that provides calibrated, individualized, and progressive pressure-threshold resistance against inspiration and expiration. Our previous results suggest that our RMT regimen is safe, well-tolerated, and results in large increases in respiratory muscle strength. We now conduct an exploratory double-blind, randomized control trial (RCT) to determine: 1) utility and feasibility of sham-RMT as a control condition, 2) the clinically meaningful outcome measures for inclusion in a future efficacy trial. This manuscript provides comprehensive information regarding the design and methods used in our trial and will aid in the reporting and interpretation of our future findings. METHODS: Twenty-eight adults with LOPD will be randomized (1:1) in blocks of 4 to RMT (treatment) or sham-RMT (control). Assessments will be conducted at pretest, posttest, 3-months detraining, and 6-months detraining. The primary outcome is maximum inspiratory pressure (MIP). Secondary outcomes include maximum expiratory pressure (MEP), 6-min walk test (6MWT), Gait, Stairs, Gowers, and Chair test (GSGC), peak cough flow (PCF), and patient-reported life activity/social participation (Rasch-built Pompe-specific Activity scale [R-Pact]). Exploratory outcomes include quantitative measures from polysomnography; patient reported measures of fatigue, daytime sleepiness, and sleep quality; and ultrasound measures of diaphragm thickness. This research will use a novel tool to provide automated data collection and user feedback, and improve control over dose. ETHICS AND DISSEMINATION: The results of this clinical trial will be promptly analyzed and submitted for publication. Results will also be made available on clinicaltrials.gov. ClinicalTrials.gov: NCT02801539, R21AR069880.


Asunto(s)
Ejercicios Respiratorios , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Método Doble Ciego , Humanos , Calidad de Vida , Ensayos Clínicos Controlados Aleatorios como Asunto , Pruebas de Función Respiratoria , Músculos Respiratorios/fisiología , Adulto Joven
12.
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
13.
Cardiol Young ; 29(3): 425-427, 2019 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-30678746

RESUMEN

Infantile-onset Pompe disease is a lysosomal storage disorder characterised with hypertrophic cardiomyopathy, respiratory insufficiency, and hypotonia. Dilated cardiomyopathy is an extremely rare and curable complication of nutritional hypocalcaemic rickets. A 3-month-old female infant was referred to our paediatric ICU with a 4-day history of fatigue, tachypnoea, tachycardia, hypoxia, and respiratory failure. According to the laboratory, radiology, and echocardiography findings, she was first diagnosed with nutritional hypocalcaemic rickets-related dilated cardiomyopathy, but vitamin D and elementary calcium supplementation unmasked the underlying infantile-onset Pompe disease. Nutritional hypocalcaemic rickets and infantile-onset Pompe disease must always be kept in mind among the causes of concomitant dilated cardiomyopathy and hypertrophic cardiomyopathy.


Asunto(s)
Cardiomiopatía Dilatada/etiología , Enfermedad del Almacenamiento de Glucógeno Tipo II/complicaciones , Hipocalcemia/complicaciones , Raquitismo/complicaciones , Cardiomiopatía Dilatada/diagnóstico , Ecocardiografía , Electrocardiografía , Femenino , Humanos , Lactante
14.
Acta Myol ; 38(4): 215-232, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31970320

RESUMEN

Muscle and lower motor neuron diseases share a common denominator of perturbed muscle function, most often related to wasting and weakness of muscles. This leads to a number of challenges, such as restricted mobility and respiratory difficulties. Currently there is no cure for these diseases. The purpose of this review is to present research that examines the effects of exercise in muscle and lower motor neuron diseases. Evidence indicates that moderate intensity aerobic- and strength exercise is advantageous for patients with muscle diseases, without causing harmful exercise-induced muscle damage. On the contrary, motor neuron diseases show a rather blunted response from exercise training. High-intensity training is a modality that seems safe and a promising exercise method, which may circumvent neural fatigue and provide effect to patients with motor neuron disease. Although we have come far in changing the view on exercise therapy in neuromuscular diseases to a positive one, much knowledge is still needed on what dose of time, intensity and duration should be implemented for different disease and how we should provide exercise therapy to very weak, non-ambulatory and wheelchair bound patients.


Asunto(s)
Terapia por Ejercicio , Ejercicio Físico , Enfermedad de la Neurona Motora/terapia , Enfermedades Musculares/terapia , Terapia por Estimulación Eléctrica , Ejercicio Físico/fisiología , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Enfermedad del Almacenamiento de Glucógeno Tipo V/terapia , Humanos , Miopatías Mitocondriales/terapia , Distrofias Musculares/terapia , Entrenamiento de Fuerza
15.
J Neurol ; 266(1): 133-147, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30430231

RESUMEN

BACKGROUND: In patients with late-onset Pompe disease, progressive respiratory muscle weakness with predominantly diaphragmatic involvement is a frequent finding at later stages of the disease. Respiratory muscle training (RMT) is an established therapy option for patients with several neuromuscular disorders including Duchenne muscular dystrophy. Forced voluntary muscle contractions of inspiration and/or expiration muscles enhance ventilation by increasing respiratory coordination, endurance, and strength. Efficacy of RMT in LOPD is rarely examined, and the clinical studies performed are difficult to compare because of different training programs and protocols. This impedes a useful statement and recommendation about the safety and efficacy of respiratory muscle training. METHODS: We conducted a monocentric unblinded single-arm pilot study in patients with LOPD to evaluate the safety and efficacy of inspiratory muscle training (IMT). The primary objective was to determine the efficacy of a 6-week repetitive IMT with a gradual increase of inspiratory resistance, measured by MIP (maximum inspiratory pressure) in the upright position. For statistical analysis, we used an A-B-C single subject design. The 6-week training-period A was followed by a 6-week non-training period B and an optional training period of 40 weeks in period C. The total study duration for the periods A, B and C was 52 weeks. Throughout the study, spirometry assessments (FCV, FEV1) and measurements of respiratory strength (MIP, MEP) were performed at defined time points, as well as capillary oximetry and capnometry, motor function test and patient's questionnaires for quality of life and dyspnea, measured by St. George's Respiratory Questionnaire (SGRQ) and MMRC-Dyspnea scale. For the cross-sectional comparison, a paired two-sided t test, and for the longitudinal comparison, a two-sample, two-sided t test were used. When data were not normally distributed, a Wilcoxon-Mann-Whitney test was added. Finally, the annual decline in FVC and FEV1 before and after IMT was compared. FINDINGS: 11 subjects were included in this pilot study. Overall, IMT was well tolerated. In four subjects, a total of six adverse events related to the study procedures were noticed. Training compliance was excellent in the first weeks of training, but declined continuously in the extension period. There was a significant increase in our primary outcome measure MIP within the 6-week period of frequent IMT with a mean of 15.7% (p =0.024; d =0.402). A significant increase was also seen after week 52 by a mean of + 26.4% (mean + 13.4 cmH2O, p =0.001, d =0.636). In the 6-week non-training interim-period (period B), the values remained stable, and there was no clinically meaningful decline in secondary outcome measures. The increase in MIP did not have any effect on secondary outcome measures like spirometry tests (FVC, FEV1), capillary blood gas analysis, motor function tests, patient's perceived quality of life or any significant change in dyspnea score. CONCLUSIONS: Frequent IMT improves MIP and thereby stabilizes and decelerates the decline of the diaphragm strength. The gradual increase of inspiratory resistance is well tolerated without any increase of side effects, as long as IMT is supervised and resistance is individually adjusted to the patient's perceived grade of exhaustion. Although we could not detect a significant impact on secondary outcome measures, IMT should be offered to all LOPD patients, especially to those who demonstrate a progressive decline in respiratory muscle function or are unable to receive ERT.


Asunto(s)
Ejercicios Respiratorios , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , Inhalación , Análisis de los Gases de la Sangre , Ejercicios Respiratorios/efectos adversos , Ejercicios Respiratorios/métodos , Estudios Transversales , Disnea/fisiopatología , Femenino , Enfermedad del Almacenamiento de Glucógeno Tipo II/fisiopatología , Humanos , Inhalación/fisiología , Estudios Longitudinales , Masculino , Persona de Mediana Edad , Actividad Motora , Fatiga Muscular , Debilidad Muscular/fisiopatología , Debilidad Muscular/terapia , Proyectos Piloto , Estudios Prospectivos , Calidad de Vida , Músculos Respiratorios/fisiopatología , Espirometría , Resultado del Tratamiento
17.
J Med Chem ; 60(23): 9462-9469, 2017 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-29112434

RESUMEN

The highly stereocontrolled de novo synthesis of l-NBDNJ (the unnatural enantiomer of the iminosugar drug Miglustat) and a preliminary evaluation of its chaperoning potential are herein reported. l-NBDNJ is able to enhance lysosomal α-glucosidase levels in Pompe disease fibroblasts, either when administered singularly or when coincubated with the recombinant human α-glucosidase. In addition, differently from its d-enantiomer, l-NBDNJ does not act as a glycosidase inhibitor.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Activación Enzimática/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , alfa-Glucosidasas/metabolismo , 1-Desoxinojirimicina/síntesis química , 1-Desoxinojirimicina/química , 1-Desoxinojirimicina/farmacología , Regulación Alostérica/efectos de los fármacos , Línea Celular , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Fibroblastos/enzimología , Fibroblastos/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Enfermedad del Almacenamiento de Glucógeno Tipo II/metabolismo , Humanos , Lisosomas/efectos de los fármacos , Lisosomas/enzimología , Lisosomas/metabolismo , Modelos Moleculares , Estereoisomerismo
18.
Echocardiography ; 34(4): 621-624, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28266734

RESUMEN

Classic infantile-onset Pompe disease (IOPD), characterized by predominantly cardiac involvement, used to be considered uniformly lethal within months. The availability of enzyme replacement therapy (ERT) has transformed the course of the disease. Decrease in ventricular hypertrophy and improvement in ventricular function have been suggested as proof for efficacy. We report the cardiac response to ERT of a child with IOPD and severe hypertrophic cardiomyopathy. The myocardial hypertrophy resolved. Change in ejection fraction, however, was slow. We discuss the potential benefit of other parameters beyond ejection to assess cardiac function in IOPD, including speckle tracking-based strain.


Asunto(s)
Cardiomiopatía Hipertrófica/complicaciones , Cardiomiopatía Hipertrófica/tratamiento farmacológico , Terapia de Reemplazo Enzimático/métodos , Enfermedad del Almacenamiento de Glucógeno Tipo II/complicaciones , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , Acarbosa/uso terapéutico , Cardiomiopatía Hipertrófica/diagnóstico por imagen , Ecocardiografía , Ecocardiografía Doppler , Femenino , Glucosidasas/uso terapéutico , Enfermedad del Almacenamiento de Glucógeno Tipo II/diagnóstico por imagen , Inhibidores de Glicósido Hidrolasas/uso terapéutico , Humanos , Lactante , Resultado del Tratamiento
19.
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
20.
Hum Gene Ther Clin Dev ; 27(4): 152-159, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27855487

RESUMEN

Neuromuscular disorders such as Pompe disease (glycogen storage disease, type II), result in early and potentially irreversible cellular damage with a very limited opportunity for intervention in the newborn period. Pompe disease is due to deficiency in acid α-glucosidase (GAA) leading to lysosomal accumulation of glycogen in all cell types, abnormal myofibrillogenesis, respiratory insufficiency, neurological deficits, and reduced contractile function in striated muscle. Previous studies have shown that fetal delivery of recombinant adeno-associated virus (rAAV) encoding GAA to the peritoneal cavity of Gaa-/- mice resulted in high-level transduction of the diaphragm. While progression of other genetic disorders may occur later in life, the potential of fetal gene delivery to avoid the onset of irreversible damage suggests it is an attractive option for many inherited diseases. In this study, rhesus monkey fetuses were administered 4.5 × 1012 particles of rAAV type 1 expressing human GAA (rAAV1-CMV-hGAA), human α-1-antitrypsin (rAAV1-CBA-hAAT), or human mini-dystrophin (rAAV1-CMV-miniDMD) in the late first trimester using an established intraperitoneal ultrasound-guided approach. Fetuses were monitored sonographically and newborns delivered at term for postnatal studies. All animals remained healthy during the study period (growth, hematology, and clinical chemistry), with no evidence of adverse effects. Tissues were collected at a postnatal age of 3 months (∼7 months post-fetal gene transfer) for immunohistochemistry (IHC) and quantitative PCR. Both the diaphragm and peritoneum from vector-treated animals were strongly positive for expression of human GAA, AAT, or dystrophin by IHC, similar to findings when reporter genes were used. Protein expression in the diaphragm and peritoneum correlated with high vector copy numbers detected by real-time PCR. Other anatomical areas were negative, although the liver showed minimal evidence of human GAA, AAT, and DMD, vector genomes. In summary, delivery of rAAV vectors provided stable transduction of the muscular component of the diaphragm without any evidence of adverse effects.


Asunto(s)
Proteínas Portadoras/genética , Dependovirus/genética , Distrofina/genética , Terapia Genética , Vectores Genéticos/administración & dosificación , Enfermedad del Almacenamiento de Glucógeno Tipo II/terapia , alfa-Glucosidasas/genética , Adolescente , Animales , Niño , Preescolar , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Diafragma , Evaluación Preclínica de Medicamentos , Femenino , Técnicas de Transferencia de Gen , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Humanos , Macaca mulatta , Masculino , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA