Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Dis Model Mech ; 16(12)2023 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-38050706

RESUMO

Duchenne muscular dystrophy (DMD), caused by mutations in the dystrophin (DMD) gene, is associated with fatal muscle degeneration and atrophy. Patients with DMD have progressive reductions in skeletal muscle strength and resistance to eccentric muscle stretch. Using the DE50-MD dog model of DMD, we assessed tibiotarsal joint (TTJ) flexor and extensor force dynamics, and the resistance of dystrophic muscle to eccentric stretch. Male DE50-MD and wild-type (WT) dogs were analysed every 3 months until 18 months of age. There was an age-associated decline in eccentric contraction resistance in DE50-MD TTJ flexors that discriminated, with high statistical power, WT from DE50-MD individuals. For isometric contraction, at the majority of timepoints, DE50-MD dogs had lower maximum absolute and relative TTJ flexor force, reduced TTJ muscle contraction times and prolonged relaxation compared to those in WT dogs. Cranial tibial muscles, the primary TTJ flexor, of 18-month-old DE50-MD dogs had significant numbers of regenerating fibres as expected, but also fewer type I fibres and more hybrid fibres than those in WT dogs. We conclude that these parameters, in particular, the eccentric contraction decrement, could be used as objective outcome measures for pre-clinical assessment in DE50-MD dogs.


Assuntos
Distrofia Muscular de Duchenne , Humanos , Cães , Masculino , Animais , Lactente , Distrofia Muscular de Duchenne/genética , Músculo Esquelético , Distrofina/genética , Contração Muscular/fisiologia , Força Muscular/fisiologia , Mutação
2.
Arch Immunol Ther Exp (Warsz) ; 71(1): 24, 2023 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-37999748

RESUMO

Duchenne muscular dystrophy (DMD) is a lethal X-linked disease caused by mutations in the dystrophin gene, leading to muscle degeneration and wasting. Electromyography (EMG) is an objective electrophysiological biomarker of muscle fiber function in muscular dystrophies. A novel, DT-DEC01 therapy, consisting of Dystrophin Expressing Chimeric (DEC) cells created by fusing allogeneic myoblasts from normal donors with autologous myoblasts from DMD-affected patients, was assessed for safety and preliminary efficacy in boys of age 6-15 years old (n = 3). Assessments included EMG testing of selected muscles of upper (deltoideus, biceps brachii) and lower (rectus femoris and gastrocnemius) extremities at the screening visit and at 3, 6, and 12 months following systemic-intraosseous administration of a single low dose of DT-DEC01 therapy (Bioethics Committee approval no. 46/2019). No immunosuppression was administered. Safety of DT-DEC01 was confirmed by the lack of therapy-related Adverse Events or Serious Adverse Events up to 22 months following DT-DEC01 administration. EMG of selected muscles of both, ambulatory and non-ambulatory patients confirmed preliminary efficacy of DT-DEC01 therapy by an increase in motor unit potentials (MUP) duration, amplitudes, and polyphasic MUPs at 12 months. This study confirmed EMG as a reliable and objective biomarker of functional assessment in DMD patients after intraosseous administration of the novel DT-DEC01 therapy.


Assuntos
Distrofia Muscular de Duchenne , Masculino , Humanos , Criança , Adolescente , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/genética , Distrofina/genética , Músculo Esquelético , Biomarcadores , Terapia Baseada em Transplante de Células e Tecidos
3.
Int J Mol Sci ; 24(14)2023 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-37511179

RESUMO

Duchenne muscular dystrophy (DMD) is a yet incurable rare genetic disease that affects the skeletal and cardiac muscles, leading to progressive muscle wasting and premature death. DMD is caused by the lack of dystrophin, a muscle protein essential for the biochemical support and integrity of muscle fibers. Gene replacement strategies for Duchenne muscular dystrophy (DMD) employing the adeno-associated virus (AAV) face the challenge imposed by the limited packaging capacity of AAV, only allowing the accommodation of a short version of dystrophin (µDys) that is still far removed from correcting human disease. The need to develop strategies leading to the expression of a best performing dystrophin variant led to only few studies reporting on the use of dual vectors, but none reported on a method to assess in vivo transgene reconstitution efficiency, the degree of which directly affects the use of safe AAV dosing. We report here on the generation of a dual AAV vector approach for the expression of a larger dystrophin version (quasidystrophin) based on homologous recombination, and the development of a methodology employing a strategic droplet digital PCR design, to determine the recombination efficiency as well as the occurrence of unwanted concatemerization events or aberrant expression from the single vectors. We demonstrated that, upon systemic delivery in the dystrophic D2.B10-Dmdmdx/J (DBA2mdx) mice, our dual AAV approach led to high transgene reconstitution efficiency and negligible Inverted Terminal Repeats (ITR)-dependent concatemerization, with consequent remarkable protein restoration in muscles and improvement of muscle pathology. This evidence supports the suitability of our system for gene therapy application and the potential of this methodology to assess and improve the feasibility for therapeutic translation of multiple vector approaches.


Assuntos
Distrofia Muscular de Duchenne , Camundongos , Humanos , Animais , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/metabolismo , Distrofina/genética , Distrofina/metabolismo , Dependovirus/genética , Dependovirus/metabolismo , Músculo Esquelético/metabolismo , Camundongos Endogâmicos mdx , Vetores Genéticos/genética
4.
Neurology ; 100(9): e975-e984, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-36849458

RESUMO

BACKGROUND AND OBJECTIVES: The slow and variable disease progression of Becker muscular dystrophy (BMD) urges the development of biomarkers to facilitate clinical trials. We explored changes in 3 muscle-enriched biomarkers in serum of patients with BMD over 4-year time and studied associations with disease severity, disease progression, and dystrophin levels in BMD. METHODS: We quantitatively measured creatine kinase (CK) using the International Federation of Clinical Chemistry reference method, creatine/creatinineratio (Cr/Crn) using liquid chromatography-tandem mass spectrometry, and myostatin with ELISA in serum and assessed functional performance using the North Star Ambulatory Assessment (NSAA), 10-meter run velocity (TMRv), 6-Minute Walking Test (6MWT), and forced vital capacity in a 4-year prospective natural history study. Dystrophin levels were quantified in the tibialis anterior muscle using capillary Western immunoassay. The correlation between biomarkers, age, functional performance, mean annual change, and prediction of concurrent functional performance was analyzed using linear mixed models. RESULTS: Thirty-four patients with 106 visits were included. Eight patients were nonambulant at baseline. Cr/Crn and myostatin were highly patient specific (intraclass correlation coefficient for both = 0.960). Cr/Crn was strongly negatively correlated, whereas myostatin was strongly positively correlated with the NSAA, TMRv, and 6MWT (Cr/Crn rho = -0.869 to -0.801 and myostatin rho = 0.792 to 0.842, all p < 0.001). CK showed a negative association with age (p = 0.0002) but was not associated with patients' performance. Cr/Crn and myostatin correlated moderately with the average annual change of the 6MWT (rho = -0.532 and 0.555, p = 0.02). Dystrophin levels did not correlate with the selected biomarkers nor with performance. Cr/Crn, myostatin, and age could explain up to 75% of the variance of concurrent functional performance of the NSAA, TMRv, and 6MWT. DISCUSSION: Both Cr/Crn and myostatin could potentially serve as monitoring biomarkers in BMD, as higher Cr/Crn and lower myostatin were associated with lower motor performance and predictive of concurrent functional performance when combined with age. Future studies are needed to more precisely determine the context of use of these biomarkers.


Assuntos
Distrofia Muscular de Duchenne , Humanos , Distrofia Muscular de Duchenne/diagnóstico , Distrofina , Creatina , Creatinina , Miostatina , Estudos Prospectivos , Biomarcadores , Creatina Quinase , Progressão da Doença
5.
Sci Transl Med ; 15(677): eabo1815, 2023 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-36599002

RESUMO

Duchenne muscular dystrophy (DMD) is a progressive muscle wasting disease caused by the absence of dystrophin, a membrane-stabilizing protein encoded by the DMD gene. Although mouse models of DMD provide insight into the potential of a corrective therapy, data from genetically homologous large animals, such as the dystrophin-deficient golden retriever muscular dystrophy (GRMD) model, may more readily translate to humans. To evaluate the clinical translatability of an adeno-associated virus serotype 9 vector (AAV9)-microdystrophin (µDys5) construct, we performed a blinded, placebo-controlled study in which 12 GRMD dogs were divided among four dose groups [control, 1 × 1013 vector genomes per kilogram (vg/kg), 1 × 1014 vg/kg, and 2 × 1014 vg/kg; n = 3 each], treated intravenously at 3 months of age with a canine codon-optimized microdystrophin construct, rAAV9-CK8e-c-µDys5, and followed for 90 days after dosing. All dogs received prednisone (1 milligram/kilogram) for a total of 5 weeks from day -7 through day 28. We observed dose-dependent increases in tissue vector genome copy numbers; µDys5 protein in multiple appendicular muscles, the diaphragm, and heart; limb and respiratory muscle functional improvement; and reduction of histopathologic lesions. As expected, given that a truncated dystrophin protein was generated, phenotypic test results and histopathologic lesions did not fully normalize. All administrations were well tolerated, and adverse events were not seen. These data suggest that systemically administered AAV-microdystrophin may be dosed safely and could provide therapeutic benefit for patients with DMD.


Assuntos
Distrofia Muscular Animal , Distrofia Muscular de Duchenne , Animais , Cães , Humanos , Recém-Nascido , Camundongos , Distrofina/genética , Distrofina/metabolismo , Terapia Genética , Coração , Músculo Esquelético/metabolismo , Músculos/metabolismo , Distrofia Muscular Animal/genética , Distrofia Muscular Animal/terapia , Distrofia Muscular Animal/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia
6.
Methods Mol Biol ; 2587: 67-103, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36401025

RESUMO

Duchenne muscular dystrophy (DMD) is a lethal muscle disease caused by dystrophin deficiency. Patients gradually lose motor function, become wheelchair-bound, and die from respiratory and/or cardiac muscle failure. Dystrophin-null dogs have been used as a large animal model for DMD since 1988 and are considered an excellent bridge between rodent models and human patients. While numerous protocols have been published for studying muscle and heart physiology in mice, few such protocols exist for studying skeletal muscle contractility, heart function, and whole-body activity in dogs. Over the last 20 years, we have developed and adapted an array of assays to evaluate whole-body movement, gait, single muscle force, whole limb torque, cardiac electrophysiology, and hemodynamic function in normal and dystrophic dogs. In this chapter, we present detailed working protocols for these assays and lessons we learned during the development and use of these protocols.


Assuntos
Distrofina , Distrofia Muscular de Duchenne , Humanos , Cães , Animais , Camundongos , Coração , Músculo Esquelético , Miocárdio
7.
Methods Mol Biol ; 2587: 303-338, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36401036

RESUMO

Assessing histological changes is essential for characterizing muscle disease progression and for studying the response to therapies in Duchenne muscular dystrophy (DMD), an X-linked progressive muscle-wasting disease caused by the loss of the dystrophin protein. Canine models are by far the best-characterized large animal models for DMD. In this chapter, we describe methods for muscle tissue collection and storage, hematoxylin and eosin staining for studying general muscle morphology, and special staining protocols for evaluating fibrosis, calcification, and neuronal nitric oxide synthase (nNOS) activity. We also provide immunofluorescence staining protocols that are often used to characterize the expression and localization of dystrophin and components of the dystrophin-associated glycoprotein complex. Lastly, we presented immunohistochemical staining protocols that we use to assess muscle inflammation and immune responses.


Assuntos
Distrofina , Distrofia Muscular de Duchenne , Cães , Animais , Distrofina/genética , Distrofina/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/metabolismo , Terapia Genética , Músculos/metabolismo
8.
Methods Mol Biol ; 2587: 353-375, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36401038

RESUMO

The immune response is a primary hurdle in the development of gene therapy for neuromuscular diseases. Both innate and adaptive immune responses have been observed in human trials. The canine model is an excellent platform to understand immunological consequences of gene therapy. Over the last several decades, we have conducted gene replacement and gene repair therapies in the canine model of Duchenne muscular dystrophy (DMD) using adeno-associated virus (AAV)-mediated expression of the highly abbreviated micro-dystrophin gene, the larger mini-dystrophin gene, and the Cas9-based CRISPR genome editing machinery. We have evaluated the innate, humoral, and cellular immune responses to the AAV vector and the transgene product. In this chapter, we share our experience in collecting and processing of the dog blood samples for immunological assays, and our protocols for quantitative evaluation of cytokines and chemokines, antibodies, and T-cell responses.


Assuntos
Distrofina , Distrofia Muscular de Duchenne , Humanos , Cães , Animais , Distrofina/genética , Distrofina/metabolismo , Vetores Genéticos/genética , Terapia Genética/métodos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Distrofia Muscular de Duchenne/metabolismo , Imunidade Humoral
9.
Lima; IETSI; abr. 2022.
Não convencional em Espanhol | BRISA/RedTESA | ID: biblio-1552390

RESUMO

ANTECEDENTES: El presente documento expone la actualización del Dictamen Preliminar de Evaluación de Tecnología Sanitaria N° 056-SDEPFyOTS-DETS-IETSI-2017 "Eficacia de ataluren en pacientes con diagnóstico de distrofia muscular de Duchenne portadores de una mutación sin sentido en el gen de distrofina - actualización" (IETSI-EsSalud 2017). El Dictamen Preliminar de Evaluación de Tecnología Sanitaria N° 056-SDEPFyOTSDETS-IETSI-2017 corresponde a la primera actualización del Dictamen Preliminar de Evaluación de Tecnología Sanitaria N° 020-SDEPFyOTS-DETS-IETSI-2016 "Seguridad y eficacia de ataluren en pacientes con diagnóstico de distrofia muscular de Duchenne portadores de una mutación sin sentido en el gen de distrofina" (IETSI-EsSalud 2016). ASPECTOS GENERALES Los aspectos generales de la DMD se han descrito previamente en el Dictamen Preliminar de Evaluación de Tecnología Sanitaria N° 020-SDEPFyOTS-DETS-1 ETSI2016 (IETSI-EsSalud 2016). Brevemente, la DMD es un trastorno genético ligado al cromosoma X y es la forma más común de distrofia muscular en la infancia y afecta a uno de cada 3500 a 6000 varones recién nacidos (Beytía, Vry, and Kirschner 2012; Mah et al. 2014). Hay aproximadamente 50000 casos de DMD en todo el mundo (Guiraud et al. 2015). La enfermedad es causada por mutaciones del gen DMD que codifica la distrofina, una proteína que protege la integridad de las células musculares (AbdulRazak, Malerba, and Dicksona 2016; Mah 2016; Shimizu-Motohashi et al. 2016). La pérdida de distrofina en pacientes con DMD altera la membrana y las fibras musculares. Esto conduce a debilidad muscular progresiva, alteración de la marcha, retraso en el desarrollo motor, hipertrofia de la pantorrilla y niveles elevados de creatina quinasa (un biomarcador que refleja el daño muscular) (Mah 2016). METODOLOGÍA: Se realizó una búsqueda sistemática de literatura con el objetivo de actualizar el Dictamen Preliminar de Evaluación de Tecnología Sanitaria N° 056-SDEPFyOTSDETS-IETSI-2017 "Eficacia de ataluren en pacientes con diagnóstico de distrofia muscular de Duchenne portadores de una mutación sin sentido en el gen de distrofina - actualización" (IETSI-EsSalud 2017), Se utilizaron las bases de datos PubMed, Cochrane Library y LILACS. Las búsquedas se limitaron a estudios publicados a partir de enero de 2016. Asimismo, se realizó una búsqueda dentro de bases de datos pertenecientes a grupos que realizan ETS y guías de práctica clínica (GPC), incluyendo el SMC, el National Institute for Health and Care Excellence (NICE), la Canadian Agency for Drugs and Technologies in Health (CADTH), la Haute Autorité de Santé (HAS), el Institute for Quality and Efficiency in Health Care (IQWiG), el Instituto de Evaluación Tecnológica en Salud de Colombia (IETS), la Comissáo Nacional de Incorporagáo de Tecnologias no Sistema Único de Saúde (CONITEC), entre otros; además de la Base Regional de Informes de Evaluación de Tecnologías en Salud de las Américas (BRISA) y páginas web de sociedades especializadas en el manejo de la DMD como American Academy of Neurology (AAN), DMD Care Considerations Working Group (CCWG), y American Academy of Pediatrics (AAP). Se hizo una búsqueda adicional en la página web del registro de ensayos clínicos administrado por la Biblioteca Nacional de Medicina de los Estados Unidos (https://clinicaltrials.qov/)e International Clinical Trial Registry Platform (ICTRP) (https://apps.who.int/trialsearch/),para identificar ensayos clínicos en curso o cuyos resultados no hayan sido publicados para, de este modo, disminuir el riesgo de sesgo de publicación. RESULTADOS: Publicaciones no incluidas en la evaluación de la evidencia: Los siguientes documentos se excluyeron por tratarse de consensos de expertos: Adult North Star Network (ANSN): Consensus Guideline For The Standard Of Care Of Adults With Duchenne Muscular Dystrophy (Quinlivan et al. 2021). Neurology Care, Diagnostics, and Emerging Therapies of the Patient With Duchenne Muscular Dystrophy (Leigh et al. 2018). Review and management of Duchenne muscular dystrophy, part 1: diagnosis, and neuromuscular, rehabilitation, endocrine, and gastrointestinal and nutritional management (Birnkrant et al. 2018). Evaluaciones de Tecnologías Sanitarias Publicaciones incluidas en la evaluación de la evidencia: Ataluren 125mg, 250mg, and 1,000mg granules for oral suspension (Translarna®). SMC2327 (SMC 2021). Informe de Intervenciones no Favorables 2018. Condición de Salud: Distrofia Muscular de Duchenne. Tecnología Sanitaria Evaluada: Ataluren (MINSAL Chile 2018). Publicaciones No incluidas en la evaluación de la evidencia La siguiente: ETS se excluyó por haberse realizado antes de la publicación del ECA de fase 3 de ataluren, estudio 020: Emerging Drugs for Duchenne Muscular Dystrophy (CADTH 2017). La siguiente ETS se excluyó por evaluar la extensión de la indicación de ataluren para incluir niños de 2 a 5 años, población que no es el objeto de este dictamen: Transparency Committee Opinion 23 October 2019: Ataluren Translarna 125 mg, 250 mg, 1000 mg, granules for oral suspensión (HAS 2019). Revisiones sistemáticas Publicaciones incluidas en la evaluación de la evidencia: Meta-analyses of ataluren randomized controlled trials in nonsense mutation Duchenne muscular dystrophy (Campbell et al. 2020). Restorative treatments of dystrophin expression in Duchenne muscular dystrophy: A systematic review (Pascual-Morena et al. 2020). Estudios observacionales Publicaciones incluidas en la evaluación de la evidencia: Safety and effectiveness of ataluren: comparison of results from the STRIDE Registry and CINRG DMD Natural History Study (Mercuri et al. 2020). Ataluren delays loss of ambulation and respiratory decline in nonsense mutation Duchenne muscular dystrophy patients (McDonald et al. 2022). Ensayos clínicos en curso o no publicados registrados Se identificó un ECA, de fase 3, doble ciego, controlado con placebo, de 72 semanas, en curso (estudio 041; identificador de ClinicalTrials.gov:NCT03179631). La fecha estimada de finalización del estudio es julio de 2023. CONCLUSIONES: Este documento tuvo como objetivo actualizar el Dictamen Preliminar de Evaluación de Tecnología Sanitaria N° 056-SDEPFyOTS-DETS-IETSI-2017 "Eficacia de ataluren en pacientes con diagnóstico de distrofia muscular de Duchenne portadores de una mutación sin sentido en el gen de distrofina - actualización" (IETSI-EsSalud 2017), publicado en octubre de 2017. La búsqueda de evidencia científica permitió identificar dos ETS realizadas por el SMC de Escocia (SMC 2021) y el Ministerio de Salud de Chile (MINSAL Chile 2018), dos RS con MA (Campbell et al. 2020; Pascual-Morena et al. 2020), y dos estudios observacionales (Mercuri et al. 2020; McDonald et al. 2022). Las dos ETS identificadas meta-analizaron los resultados de dos ECA (estudios 007 y 020) y concluyeron que alectinib no brinda un efecto clínicamente relevante en términos de eficacia (cambio en la 6MWD) en la población objeto del dictamen. La única ETS que emitió una recomendación, desfavoreció el uso de ataluren (MINSAL Chile). Las dos MA de ECA (estudios 007 y 020) reportaron resultados estadísticamente significativos y favorables para ataluren, sin embargo, la calidad de la evidencia se vio afectada principalmente por la imprecisión de los resultados y el pequeño tamaño del efecto, que no fue clínicamente relevante según las consideraciones de la EMA. Los estudios observacionales incluidos en este dictamen representan evidencia de baja calidad y sus resultados deben considerarse exploratorios. Dada la falta de un proceso de aleatorización, como en el caso de los ECA, la evidencia de los estudios observacionales no permite establecer una relación causal entre ataluren y los efectos reportados. En consecuencia, los resultados no pueden atribuirse de forma fiable al uso de ataluren. Por todo lo expuesto, el 'ETS' no aprueba el uso de ataluren en pacientes con DMD portadores de una mutación sin sentido en el gen de distrofina.


Assuntos
Humanos , Proteínas Ribossômicas/uso terapêutico , Distrofina/genética , Distrofia Muscular de Duchenne/tratamento farmacológico , Eficácia , Análise Custo-Benefício/economia
10.
PLoS One ; 16(6): e0253882, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34170974

RESUMO

INTRODUCTION: The aim of this study was to report 36-month longitudinal changes using the North Star Ambulatory Assessment (NSAA) in ambulant patients affected by Duchenne muscular dystrophy amenable to skip exons 44, 45, 51 or 53. MATERIALS AND METHODS: We included 101 patients, 34 had deletions amenable to skip exon 44, 25 exon 45, 19 exon 51, and 28 exon 53, not recruited in any ongoing clinical trials. Five patients were counted to skip exon 51 and 53 since they had a single deletion of exon 52. RESULTS: The difference between subgroups (skip 44, 45, 51 and 53) was significant at 12 (p = 0.043), 24 (p = 0.005) and 36 months (p≤0.001). DISCUSSION: Mutations amenable to skip exons 53 and 51 had lower baseline values and more negative changes than the other subgroups while those amenable to skip exon 44 had higher scores both at baseline and at follow up. CONCLUSION: Our results confirm different progression of disease in subgroups of patients with deletions amenable to skip different exons. This information is relevant as current long term clinical trials are using the NSAA in these subgroups of mutations.


Assuntos
Distrofina/genética , Distrofia Muscular de Duchenne/genética , Mutação/genética , Criança , Progressão da Doença , Éxons/genética , Seguimentos , Humanos , Estudos Longitudinais , Masculino , Homens , Distrofia Muscular de Duchenne/patologia , Índice de Gravidade de Doença , Caminhada/fisiologia
11.
Am J Med Genet A ; 185(8): 2541-2545, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34018669

RESUMO

Prenatal detection of structural variants of uncertain significance, including copy number variants (CNV), challenges genetic counseling, and creates ambiguity for expectant parents. In Duchenne muscular dystrophy, variant classification and phenotypic severity of CNVs are currently assessed by familial segregation, prediction of the effect on the reading frame, and precedent data. Delineation of pathogenicity by familial segregation is limited by time and suitable family members, whereas analytical tools can rapidly delineate potential consequences of variants. We identified a duplication of uncertain significance encompassing a portion of the dystrophin gene (DMD) in an unaffected mother and her male fetus. Using long-read whole genome sequencing and alignment of short reads, we rapidly defined the precise breakpoints of this variant in DMD and could provide timely counseling. The benign nature of the variant was substantiated, more slowly, by familial segregation to a healthy maternal uncle. We find long-read whole genome sequencing of clinical utility in a prenatal setting for accurate and rapid characterization of structural variants, specifically a duplication involving DMD.


Assuntos
Variações do Número de Cópias de DNA , Distrofina/genética , Testes Genéticos/métodos , Distrofia Muscular de Duchenne/diagnóstico , Distrofia Muscular de Duchenne/genética , Diagnóstico Pré-Natal/métodos , Adulto , Pontos de Quebra do Cromossomo , Duplicação Cromossômica , Cromossomos Humanos X , Hibridização Genômica Comparativa , Éxons , Feminino , Estudos de Associação Genética/métodos , Predisposição Genética para Doença , Humanos , Masculino , Linhagem , Gravidez , Análise de Sequência de DNA
15.
JAMA Neurol ; 77(9): 1122-1131, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32539076

RESUMO

Importance: Micro-dystrophin gene transfer shows promise for treating patients with Duchenne muscular dystrophy (DMD) using recombinant adeno-associated virus serotype rh74 (rAAVrh74) and codon-optimized human micro-dystrophin driven by a skeletal and cardiac muscle-specific promoter with enhanced cardiac expression (MHCK7). Objective: To identify the 1-year safety and tolerability of intravenous rAAVrh74.MHCK7.micro-dystrophin in patients with DMD. Design, Setting, and Participants: This open-label, phase 1/2a nonrandomized controlled trial was conducted at the Nationwide Children's Hospital in Columbus, Ohio. It began on November 2, 2017, with a planned duration of follow-up of 3 years, ending in March 2021. The first 4 patients who met eligibility criteria were enrolled, consisting of ambulatory male children with DMD without preexisting AAVrh74 antibodies and a stable corticosteroid dose (≥12 weeks). Interventions: A single dose of 2.0 × 1014 vg/kg rAAVrh74.MHCK7.micro-dystrophin was infused through a peripheral limb vein. Daily prednisolone, 1 mg/kg, started 1 day before gene delivery (30-day taper after infusion). Main Outcomes and Measures: Safety was the primary outcome. Secondary outcomes included micro-dystrophin expression by Western blot and immunohistochemistry. Functional outcomes measured by North Star Ambulatory Assessment (NSAA) and serum creatine kinase were exploratory outcomes. Results: Four patients were included (mean [SD] age at enrollment, 4.8 [1.0] years). All adverse events (n = 53) were considered mild (33 [62%]) or moderate (20 [38%]), and no serious adverse events occurred. Eighteen adverse events were considered treatment related, the most common of which was vomiting (9 of 18 events [50%]). Three patients had transiently elevated γ-glutamyltransferase, which resolved with corticosteroids. At 12 weeks, immunohistochemistry of gastrocnemius muscle biopsy specimens revealed robust transgene expression in all patients, with a mean of 81.2% of muscle fibers expressing micro-dystrophin with a mean intensity of 96% at the sarcolemma. Western blot showed a mean expression of 74.3% without fat or fibrosis adjustment and 95.8% with adjustment. All patients had confirmed vector transduction and showed functional improvement of NSAA scores and reduced creatine kinase levels (posttreatment vs baseline) that were maintained for 1 year. Conclusions and Relevance: This trial showed rAAVrh74.MHCK7.micro-dystrophin to be well tolerated and have minimal adverse events; the safe delivery of micro-dystrophin transgene; the robust expression and correct localization of micro-dystrophin protein; and improvements in creatine kinase levels and NSAA scores. These findings suggest that rAAVrh74.MHCK7.micro-dystrophin can provide functional improvement that is greater than that observed under standard of care. Trial Registration: ClinicalTrials.gov Identifier: NCT03375164.


Assuntos
Distrofina , Terapia Genética/métodos , Distrofia Muscular de Duchenne/terapia , Avaliação de Resultados em Cuidados de Saúde , Criança , Pré-Escolar , Dependovirus , Distrofina/genética , Seguimentos , Técnicas de Transferência de Genes , Terapia Genética/efeitos adversos , Vetores Genéticos , Humanos , Masculino , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Projetos Piloto
16.
Exp Cell Res ; 392(2): 112033, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32360435

RESUMO

AAV-delivered microdystrophin genes hold great promise for Duchenne muscular dystrophy (DMD) treatment. It is anticipated that the optimization of engineered dystrophin genes will be required to increase the efficacy and reduce the immunogenicity of transgenic proteins. An in vitro system is required for the efficacy testing of genetically engineered dystrophin genes. We report here on the proof of concept for an in vitro assay based on the assessment of sarcolemma damage after repetitively applied electrical stimuli. The primary cell culture of myoblasts was established from wild-type C57BL/10ScSnJ and dystrophin-deficient mdx mice. The preparation parameters and the differentiation of contractile myotubes were optimized. DAPI and TO-PRO-3 dyes were used to assess myotubular membrane permeability in response to electrical pulse stimulation (EPS). Myotubes derived from mdx mice exhibited a greater increase in membrane damage, as assessed by TO-PRO-3-measured permeability after EPS, than was exhibited by the healthy control myotubes. AAV-DJ particles carrying the microdystrophin gene were used to transduce mdx-derived differentiated myotubes. Microdystrophin delivery ameliorated the disease phenotype and reduced the EPS-induced membrane damage to a level comparable to that of the healthy controls. Thus, the in vitro system was shown to be capable of supporting studies on DMD gene therapy.


Assuntos
Dependovirus/genética , Distrofina/genética , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Mioblastos/patologia , Animais , Diferenciação Celular , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Contração Muscular , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/genética , Mioblastos/metabolismo
17.
J Manag Care Spec Pharm ; 26(4): 361-366, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32223597

RESUMO

DISCLOSURES: Funding for this summary was contributed by Arnold Ventures, Commonwealth Fund, California Health Care Foundation, National Institute for Health Care Management (NIHCM), New England States Consortium Systems Organization, Blue Cross Blue Shield of Massachusetts, Harvard Pilgrim Health Care, Kaiser Foundation Health Plan, and Partners HealthCare to the Institute for Clinical and Economic Review (ICER), an independent organization that evaluates the evidence on the value of health care interventions. ICER's annual policy summit is supported by dues from Aetna, America's Health Insurance Plans, Anthem, Allergan, Alnylam, AstraZeneca, Biogen, Blue Shield of CA, Cambia Health Services, CVS, Editas, Express Scripts, Genentech/Roche, GlaxoSmithKline, Harvard Pilgrim, Health Care Service Corporation, Health Partners, Johnson & Johnson (Janssen), Kaiser Permanente, LEO Pharma, Mallinckrodt, Merck, Novartis, National Pharmaceutical Council, Premera, Prime Therapeutics, Regeneron, Sanofi, Spark Therapeutics, and United Healthcare. Agboola, Fluetsch, Rind, and Pearson are employed by ICER. Lin reports support from ICER during work on this economic model and grants from Mount Zion Health Fund, National Institutes of Health (National Cancer Institute and National Heart, Lung, and Blood Institute), and the Tobacco-Related Diseases Research Program, unrelated to this work. Walton reports support from ICER for work on this economic model and unrelated consulting fees from Baxter.


Assuntos
Distrofina/genética , Imunossupressores/uso terapêutico , Distrofia Muscular de Duchenne/tratamento farmacológico , Oligonucleotídeos Antissenso/uso terapêutico , Pregnenodionas/uso terapêutico , Análise Custo-Benefício , Éxons/efeitos dos fármacos , Éxons/genética , Humanos , Imunossupressores/economia , Modelos Econômicos , Morfolinos/economia , Morfolinos/farmacologia , Morfolinos/uso terapêutico , Distrofia Muscular de Duchenne/economia , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/imunologia , Oligonucleotídeos/economia , Oligonucleotídeos/farmacologia , Oligonucleotídeos/uso terapêutico , Oligonucleotídeos Antissenso/economia , Oligonucleotídeos Antissenso/farmacologia , Prednisona/economia , Prednisona/uso terapêutico , Pregnenodionas/economia , Ensaios Clínicos Controlados Aleatórios como Assunto , Resultado do Tratamento
18.
Sci Rep ; 9(1): 6078, 2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30988454

RESUMO

Antisense oligonucleotide (AO)-mediated splice modulation has been established as a therapeutic approach for tackling genetic diseases. Recently, Exondys51, a drug that aims to correct splicing defects in the dystrophin gene was approved by the US Food and Drug Administration (FDA) for the treatment of Duchenne muscular dystrophy (DMD). However, Exondys51 has relied on phosphorodiamidate morpholino oligomer (PMO) chemistry which poses challenges in the cost of production and compatibility with conventional oligonucleotide synthesis procedures. One approach to overcome this problem is to construct the AO with alternative nucleic acid chemistries using solid-phase oligonucleotide synthesis via standard phosphoramidite chemistry. 2'-Fluoro (2'-F) is a potent RNA analogue that possesses high RNA binding affinity and resistance to nuclease degradation with good safety profile, and an approved drug Macugen containing 2'-F-modified pyrimidines was approved for the treatment of age-related macular degeneration (AMD). In the present study, we investigated the scope of 2'-F nucleotides to construct mixmer and gapmer exon skipping AOs with either 2'-O-methyl (2'-OMe) or locked nucleic acid (LNA) nucleotides on a phosphorothioate (PS) backbone, and evaluated their efficacy in inducing exon-skipping in mdx mouse myotubes in vitro. Our results showed that all AOs containing 2'-F nucleotides induced efficient exon-23 skipping, with LNA/2'-F chimeras achieving better efficiency than the AOs without LNA modification. In addition, LNA/2'-F chimeric AOs demonstrated higher exonuclease stability and lower cytotoxicity than the 2'-OMe/2'-F chimeras. Overall, our findings certainly expand the scope of constructing 2'-F modified AOs in splice modulation by incorporating 2'-OMe and LNA modifications.


Assuntos
Fibras Musculares Esqueléticas/efeitos dos fármacos , Distrofia Muscular de Duchenne/terapia , Oligonucleotídeos Antissenso/farmacologia , Splicing de RNA/efeitos dos fármacos , Animais , Células Cultivadas , Técnicas de Química Sintética/economia , Técnicas de Química Sintética/métodos , Química Farmacêutica/economia , Química Farmacêutica/métodos , Avaliação Pré-Clínica de Medicamentos , Distrofina/genética , Distrofina/metabolismo , Éxons/efeitos dos fármacos , Éxons/genética , Terapia Genética/economia , Terapia Genética/métodos , Humanos , Camundongos , Camundongos Endogâmicos mdx , Morfolinos/economia , Morfolinos/uso terapêutico , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular de Duchenne/genética , Oligonucleotídeos/química , Oligonucleotídeos/economia , Oligonucleotídeos/farmacologia , Oligonucleotídeos/uso terapêutico , Oligonucleotídeos Antissenso/química , Oligonucleotídeos Antissenso/economia , Oligonucleotídeos Antissenso/uso terapêutico
19.
Arch Pathol Lab Med ; 143(2): 197-205, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30168727

RESUMO

CONTEXT.­: Duchenne muscular dystrophy is a rare, progressive, and fatal neuromuscular disease caused by dystrophin protein loss. Common investigational treatment approaches aim at increasing dystrophin expression in diseased muscle. Some clinical trials include assessments of novel dystrophin production as a surrogate biomarker of efficacy, which may predict a clinical benefit from treatment. OBJECTIVES.­: To establish an immunofluorescent scanning and digital image analysis workflow that provides an objective approach for staining intensity assessment of the immunofluorescence dystrophin labeling and determination of the percentage of biomarker-positive fibers in muscle cryosections. DESIGN.­: Optimal and repeatable digital image capture was achieved by a rigorously qualified fluorescent scanning process. After scanning qualification, the MuscleMap (Flagship Biosciences, Westminster, Colorado) algorithm was validated by comparing high-power microscopic field total and dystrophin-positive fiber counts obtained by trained pathologists to data derived by MuscleMap. Next, the algorithm was tested on whole-slide images of immunofluorescent-labeled muscle sections from Duchenne muscular dystrophy, Becker muscular dystrophy, and control patients. RESULTS.­: When used under the guidance of a trained pathologist, the digital image analysis tool met predefined validation criteria and demonstrated functional and statistical equivalence with manual assessment. This work is the first, to our knowledge, to qualify and validate immunofluorescent scanning and digital tissue image-analysis workflow, respectively, with the rigor required to support the clinical trial environments. CONCLUSIONS.­: MuscleMap enables analysis of all fibers within an entire muscle biopsy section and provides data on a fiber-by-fiber basis. This will allow future clinical trials to objectively investigate myofibers' dystrophin expression at a greater level of consistency and detail.


Assuntos
Distrofina/análise , Interpretação de Imagem Assistida por Computador/métodos , Distrofia Muscular de Duchenne/diagnóstico , Adolescente , Biópsia , Criança , Pré-Escolar , Feminino , Secções Congeladas , Humanos , Masculino , Pessoa de Meia-Idade , Músculo Esquelético/patologia
20.
Methods Mol Biol ; 1828: 219-228, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30171544

RESUMO

Duplications of one or more dystrophin exons that disrupt the reading frame account for about 15% of all Duchenne cases, and like the more common genomic deletions, most pathogenic duplications of single or multiple dystrophin exons are also amenable to targeted exon skipping. However, additional considerations must be taken into account: (1) skipping of all duplicated exons, and, flanking exons as necessary, will frequently be required to restore the reading frame and generate an in-frame Becker muscular dystrophy-like mRNA, (2) the phosphorodiamidate morpholino oligomer chemistry is more effective than the 2'-O-methyl modified oligonucleotides at inducing multiple exon skipping, and (2) the apparent efficiency of exon skipping can be confounded by the choice of RT-PCR system. Standard RT-PCR systems can preferentially amplify the shorter amplicons, implying more efficient exon skipping than may actually be induced. Unless high fidelity RT-PCR systems are used, strand slippage during annealing/elongation steps will generate normal length transcripts that are artifacts of the amplification.


Assuntos
Distrofina/genética , Éxons , Engenharia Genética , Linhagem Celular , Humanos , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Oligonucleotídeos Antissenso , Fases de Leitura , Deleção de Sequência , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA