Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 9 de 9
1.
Arch Dis Child ; 105(4): 332-338, 2020 04.
Article En | MEDLINE | ID: mdl-31484632

PURPOSE: Individuals with X-linked myotubular myopathy (XLMTM) who survive infancy require extensive supportive care, including ventilator assistance, wheelchairs and feeding tubes. Half die before 18 months of age. We explored respiratory support and associated mortality risk in RECENSUS, particularly among patients ≤5 years old who received respiratory support at birth; this subgroup closely matches patients in the ASPIRO trial of gene therapy for XLMTM. DESIGN: RECENSUS is an international, retrospective study of patients with XLMTM. Descriptive and time-to-event analyses examined survival on the basis of age, respiratory support, tracheostomy use, predicted mutational effects and life-sustaining care. RESULTS: Outcomes for 145 patients were evaluated. Among 126 patients with respiratory support at birth, mortality was 47% overall and 59% among those ≤5 years old. Median survival time was shorter for patients ≤5 years old than for those >5 years old (2.2 years (IQR 0.7-5.6) vs 30.2 years (IQR 19.4-30.2)). The most common cause of death was respiratory failure (66.7%). Median survival time was longer for patients with a tracheostomy than for those without (22.8 years (IQR 8.7-30.2) vs 1.8 years (IQR 0.2-not estimable)). The proportion of patients living without a tracheostomy was 50% at age 6 months and 28% at age 2 years. Median survival time was longer with provision of life-sustaining care than without (19.4 years (IQR 3.1-not estimable) vs 0.2 years (IQR 0.1-2.1)). CONCLUSIONS: High mortality, principally due to respiratory failure, among patients with XLMTM ≤5 years old despite respiratory support underscores the need for early diagnosis, informed decision-making and disease-modifying therapies. TRIAL REGISTRATION NUMBER: NCT02231697.


Myopathies, Structural, Congenital/mortality , Respiration, Artificial/statistics & numerical data , Age Factors , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Myopathies, Structural, Congenital/physiopathology , Myopathies, Structural, Congenital/therapy , Premature Birth/epidemiology , Retrospective Studies
2.
Muscle Nerve ; 57(4): 550-560, 2018 04.
Article En | MEDLINE | ID: mdl-29149770

INTRODUCTION: X-linked myotubular myopathy (XLMTM), characterized by severe hypotonia, weakness, respiratory distress, and early mortality, is rare and natural history studies are few. METHODS: RECENSUS is a multicenter chart review of male XLMTM patients characterizing disease burden and unmet medical needs. Data were collected between September 2014 and June 2016. RESULTS: Analysis included 112 patients at six clinical sites. Most recent patient age recorded was ≤18 months for 40 patients and >18 months for 72 patients. Mean (SD) age at diagnosis was 3.7 (3.7) months and 54.3 (77.1) months, respectively. Mortality was 44% (64% ≤18 months; 32% >18 months). Premature delivery occurred in 34/110 (31%) births. Nearly all patients (90%) required respiratory support at birth. In the first year of life, patients underwent an average of 3.7 surgeries and spent 35% of the year in the hospital. DISCUSSION: XLMTM is associated with high mortality, disease burden, and healthcare utilization. Muscle Nerve 57: 550-560, 2018.


Myopathies, Structural, Congenital/mortality , Premature Birth/epidemiology , Respiration, Artificial/statistics & numerical data , Surgical Procedures, Operative/statistics & numerical data , Adolescent , Age Factors , Child , Child, Preschool , Humans , Infant , Infant, Newborn , Male , Mortality , Retrospective Studies , Young Adult
3.
Clin Genet ; 92(6): 616-623, 2017 Dec.
Article En | MEDLINE | ID: mdl-28436997

Dilated cardiomyopathy (DCM) is one of the leading causes of heart failure with high morbidity and mortality. More than 40 genes have been reported to cause DCM. To provide new insights into the pathophysiology of dilated cardiomyopathy, a next-generation sequencing (NGS) workflow based on a panel of 48 cardiomyopathies-causing genes was used to analyze a cohort of 222 DCM patients. Truncating variants were detected on 63 unrelated DCM cases (28.4%). Most of them were identified, as expected, on TTN (29 DCM probands), but truncating variants were also identified on myofibrillar myopathies causing genes in 17 DCM patients (7.7% of the DCM cohort): 10 variations on FLNC and 7 variations on BAG3 . This study confirms that truncating variants on myofibrillar myopathies causing genes are frequently associated with dilated cardiomyopathies and also suggest that FLNC mutations could be considered as a common cause of dilated cardiomyopathy. Molecular approaches that would allow to detect systematically truncating variants in FLNC and BAG3 into genetic testing should significantly increase test sensitivity, thereby allowing earlier diagnosis and therapeutic intervention for many patients with dilated cardiomyopathy.


Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Cardiomyopathy, Dilated/diagnosis , Connectin/genetics , Filamins/genetics , Mutation , Myopathies, Structural, Congenital/diagnosis , Adult , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/mortality , Cardiomyopathy, Dilated/physiopathology , Cohort Studies , Female , France , Gene Expression , High-Throughput Nucleotide Sequencing , Humans , Infant, Newborn , Male , Middle Aged , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/mortality , Myopathies, Structural, Congenital/physiopathology , Pedigree , Survival Analysis
4.
Mol Ther ; 25(4): 839-854, 2017 04 05.
Article En | MEDLINE | ID: mdl-28237839

X-linked myotubular myopathy (XLMTM) results from MTM1 gene mutations and myotubularin deficiency. Most XLMTM patients develop severe muscle weakness leading to respiratory failure and death, typically within 2 years of age. Our objective was to evaluate the efficacy and safety of systemic gene therapy in the p.N155K canine model of XLMTM by performing a dose escalation study. A recombinant adeno-associated virus serotype 8 (rAAV8) vector expressing canine myotubularin (cMTM1) under the muscle-specific desmin promoter (rAAV8-cMTM1) was administered by simple peripheral venous infusion in XLMTM dogs at 10 weeks of age, when signs of the disease are already present. A comprehensive analysis of survival, limb strength, gait, respiratory function, neurological assessment, histology, vector biodistribution, transgene expression, and immune response was performed over a 9-month study period. Results indicate that systemic gene therapy was well tolerated, prolonged lifespan, and corrected the skeletal musculature throughout the body in a dose-dependent manner, defining an efficacious dose in this large-animal model of the disease. These results support the development of gene therapy clinical trials for XLMTM.


Dependovirus/genetics , Genetic Therapy , Genetic Vectors/genetics , Muscle, Skeletal/metabolism , Myopathies, Structural, Congenital/genetics , Animals , Biopsy , Dependovirus/classification , Disease Models, Animal , Disease Progression , Dogs , Gait , Gene Expression , Genetic Therapy/adverse effects , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/adverse effects , Genetic Vectors/pharmacokinetics , Immunity, Cellular , Immunity, Humoral , Kaplan-Meier Estimate , Muscle Strength , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscle, Skeletal/ultrastructure , Myopathies, Structural, Congenital/diagnosis , Myopathies, Structural, Congenital/mortality , Myopathies, Structural, Congenital/therapy , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Recovery of Function , Reflex , Respiratory Function Tests , Tissue Distribution , Transgenes/genetics , Transgenes/immunology , Treatment Outcome
5.
Am J Phys Med Rehabil ; 93(11 Suppl 3): S97-107, 2014 Nov.
Article En | MEDLINE | ID: mdl-25313664

The development of clinical vectors to correct genetic mutations that cause inherited myopathies and related disorders of skeletal muscle is advancing at an impressive rate. Adeno-associated virus vectors are attractive for clinical use because (1) adeno-associated viruses do not cause human disease and (2) these vectors are able to persist for years. New vectors are now becoming available as gene therapy delivery tools, and recent preclinical experiments have demonstrated the feasibility, safety, and efficacy of gene therapy with adeno-associated virus for long-term correction of muscle pathology and weakness in myotubularin-deficient canine and murine disease models. In this review, recent advances in the application of gene therapies to treat inherited muscle disorders are presented, including Duchenne muscular dystrophy and x-linked myotubular myopathy. Potential areas for therapeutic synergies between rehabilitation medicine and genetics are also discussed.


Genetic Therapy/methods , Muscular Dystrophy, Duchenne/therapy , Myopathies, Structural, Congenital/therapy , Physical and Rehabilitation Medicine/trends , Regenerative Medicine/trends , Animals , Cohort Studies , Combined Modality Therapy , Disease Models, Animal , Dogs , Forecasting , Genetic Vectors , Humans , Muscular Diseases/diagnosis , Muscular Diseases/therapy , Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/mortality , Myopathies, Structural, Congenital/diagnosis , Myopathies, Structural, Congenital/mortality , Physical and Rehabilitation Medicine/methods , Prognosis , Regenerative Medicine/methods , Survival Analysis , Treatment Outcome
7.
Sci Transl Med ; 6(220): 220ra10, 2014 Jan 22.
Article En | MEDLINE | ID: mdl-24452262

Loss-of-function mutations in the myotubularin gene (MTM1) cause X-linked myotubular myopathy (XLMTM), a fatal, congenital pediatric disease that affects the entire skeletal musculature. Systemic administration of a single dose of a recombinant serotype 8 adeno-associated virus (AAV8) vector expressing murine myotubularin to Mtm1-deficient knockout mice at the onset or at late stages of the disease resulted in robust improvement in motor activity and contractile force, corrected muscle pathology, and prolonged survival throughout a 6-month study. Similarly, single-dose intravascular delivery of a canine AAV8-MTM1 vector in XLMTM dogs markedly improved severe muscle weakness and respiratory impairment, and prolonged life span to more than 1 year in the absence of toxicity or a humoral or cell-mediated immune response. These results demonstrate the therapeutic efficacy of AAV-mediated gene therapy for myotubular myopathy in small- and large-animal models, and provide proof of concept for future clinical trials in XLMTM patients.


Disease Models, Animal , Genetic Therapy/methods , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/therapy , Animals , Dependovirus/genetics , Diaphragm , Dogs , Genetic Vectors , Genotype , HEK293 Cells , Humans , Male , Mice , Mice, Knockout , Muscle Contraction , Muscle Weakness , Mutation , Myopathies, Structural, Congenital/mortality , Protein Tyrosine Phosphatases, Non-Receptor/genetics
9.
Hum Mutat ; 15(5): 393-409, 2000.
Article En | MEDLINE | ID: mdl-10790201

X-linked myotubular myopathy (XLMTM; MIM# 310400) is a severe congenital muscle disorder caused by mutations in the MTM1 gene. This gene encodes a dual-specificity phosphatase named myotubularin, defining a large gene family highly conserved through evolution (which includes the putative anti-phosphatase Sbf1/hMTMR5). We report 29 mutations in novel cases, including 16 mutations not described before. To date, 198 mutations have been identified in unrelated families, accounting for 133 different disease-associated mutations which are widespread throughout the gene. Most point mutations are truncating, but 26% (35/133) are missense mutations affecting residues conserved in the Drosophila ortholog and in the homologous MTMR1 gene. Three recurrent mutations affect 17% of the patients, and a total of 21 different mutations were found in several independent families. The frequency of female carriers appears higher than expected (only 17% are de novo mutations). While most truncating mutations cause the severe and early lethal phenotype, some missense mutations are associated with milder forms and prolonged survival (up to 54 years).


Mutation , Myopathies, Structural, Congenital/genetics , Protein Tyrosine Phosphatases/genetics , X Chromosome , Alternative Splicing , DNA Transposable Elements , Female , Humans , Male , Middle Aged , Multigene Family , Mutation, Missense , Myopathies, Structural, Congenital/mortality , Polymorphism, Genetic , Protein Tyrosine Phosphatases, Non-Receptor , Sequence Deletion , Survival Analysis
...