Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
1.
Hum Mutat ; 43(4): 511-528, 2022 04.
Article in English | MEDLINE | ID: mdl-35165973

ABSTRACT

DMD pathogenic variants for Duchenne and Becker muscular dystrophy are detectable with high sensitivity by standard clinical exome analyses of genomic DNA. However, up to 7% of DMD mutations are deep intronic and analysis of muscle-derived RNA is an important diagnostic step for patients who have negative genomic testing but abnormal dystrophin expression in muscle. In this study, muscle biopsies were evaluated from 19 patients with clinical features of a dystrophinopathy, but negative clinical DMD mutation analysis. Reverse transcription-polymerase chain reaction or high-throughput RNA sequencing methods identified 19 mutations with one of three pathogenic pseudoexon types: deep intronic point mutations, deletions or insertions, and translocations. In association with point mutations creating intronic splice acceptor sites, we observed the first examples of DMD pseudo 3'-terminal exon mutations causing high efficiency transcription termination within introns. This connection between splicing and premature transcription termination is reminiscent of U1 snRNP-mediating telescripting in sustaining RNA polymerase II elongation across large genes, such as DMD. We propose a novel classification of three distinct types of mutations identifiable by muscle RNA analysis, each of which differ in potential treatment approaches. Recognition and appropriate characterization may lead to therapies directed toward full-length dystrophin expression for some patients.


Subject(s)
Dystrophin , Muscular Dystrophy, Duchenne , Dystrophin/genetics , Humans , Introns/genetics , Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Mutation , RNA Splice Sites
2.
Muscle Nerve ; 65(3): 266-277, 2022 03.
Article in English | MEDLINE | ID: mdl-34878187

ABSTRACT

Duchenne muscular dystrophy (DMD) is associated with progressive muscle weakness, loss of ambulation (LOA), and early mortality. In this review we have synthesized published data on the clinical course of DMD by genotype. Using a systematic search implemented in Medline and Embase, 53 articles were identified that describe the clinical course of DMD, with pathogenic variants categorizable by exon skip or stop-codon readthrough amenability and outcomes presented by age. Outcomes described included those related to ambulatory, cardiac, pulmonary, or cognitive function. Estimates of the mean (95% confidence interval) age at LOA ranged from 9.1 (8.7-9.6) years among 90 patients amenable to skipping exon 53 to 11.5 (9.5-13.5) years among three patients amenable to skipping exon 8. Although function worsened with age, the impact of genotype was less clear for other outcomes (eg, forced vital capacity and left ventricular ejection fraction). Understanding the distribution of pathogenic variants is important for studies in DMD, as this research suggests major differences in the natural history of disease. In addition, specific details of the use of key medications, including corticosteroids, antisense oligonucleotides, and cardiac medications, should be reported.


Subject(s)
Muscular Dystrophy, Duchenne , Child , Dystrophin/genetics , Genotype , Humans , Stroke Volume , Ventricular Function, Left
3.
N Engl J Med ; 378(7): 625-635, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29443664

ABSTRACT

BACKGROUND: Nusinersen is an antisense oligonucleotide drug that modulates pre-messenger RNA splicing of the survival motor neuron 2 ( SMN2) gene. It has been developed for the treatment of spinal muscular atrophy (SMA). METHODS: We conducted a multicenter, double-blind, sham-controlled, phase 3 trial of nusinersen in 126 children with SMA who had symptom onset after 6 months of age. The children were randomly assigned, in a 2:1 ratio, to undergo intrathecal administration of nusinersen at a dose of 12 mg (nusinersen group) or a sham procedure (control group) on days 1, 29, 85, and 274. The primary end point was the least-squares mean change from baseline in the Hammersmith Functional Motor Scale-Expanded (HFMSE) score at 15 months of treatment; HFMSE scores range from 0 to 66, with higher scores indicating better motor function. Secondary end points included the percentage of children with a clinically meaningful increase from baseline in the HFMSE score (≥3 points), an outcome that indicates improvement in at least two motor skills. RESULTS: In the prespecified interim analysis, there was a least-squares mean increase from baseline to month 15 in the HFMSE score in the nusinersen group (by 4.0 points) and a least-squares mean decrease in the control group (by -1.9 points), with a significant between-group difference favoring nusinersen (least-squares mean difference in change, 5.9 points; 95% confidence interval, 3.7 to 8.1; P<0.001). This result prompted early termination of the trial. Results of the final analysis were consistent with results of the interim analysis. In the final analysis, 57% of the children in the nusinersen group as compared with 26% in the control group had an increase from baseline to month 15 in the HFMSE score of at least 3 points (P<0.001), and the overall incidence of adverse events was similar in the nusinersen group and the control group (93% and 100%, respectively). CONCLUSIONS: Among children with later-onset SMA, those who received nusinersen had significant and clinically meaningful improvement in motor function as compared with those in the control group. (Funded by Biogen and Ionis Pharmaceuticals; CHERISH ClinicalTrials.gov number, NCT02292537 .).


Subject(s)
Oligonucleotides, Antisense/therapeutic use , Oligonucleotides/therapeutic use , Spinal Muscular Atrophies of Childhood/drug therapy , Age of Onset , Child , Child, Preschool , Double-Blind Method , Female , Humans , Infant , Injections, Spinal , Least-Squares Analysis , Male , Motor Skills , Oligonucleotides/adverse effects , Oligonucleotides, Antisense/adverse effects , Spinal Muscular Atrophies of Childhood/physiopathology
4.
Muscle Nerve ; 64(2): 153-155, 2021 08.
Article in English | MEDLINE | ID: mdl-33959970

ABSTRACT

Implementation of newborn screening for spinal muscular atrophy (SMA) in 33 US states and increased genetic carrier screening have led to an increase in early, presymptomatic diagnosis of SMA. Early treatment is critically important and is recommended for presymptomatic infants with two to four copies of survival motor neuron 2. Currently, no specific treatment recommendations exist for preterm infants with SMA. The US Food and Drug Administration does not recommend using onasemnogene abeparvovec-xioi in preterm infants. Some insurance companies interpret "preterm" to be less than 40 weeks gestational age (GA) instead of the commonly accepted 37 weeks GA, which can be a barrier to treatment access. Given the risk of rapid decline in some infants, we recommend treatment of preterm infants when they reach 37 weeks GA, based on the definitions of term GA from the World Health Organization and Centers for Disease Control and Prevention, assuming all other treatment criteria are met.


Subject(s)
Biological Products/therapeutic use , Muscular Atrophy, Spinal/drug therapy , Spinal Muscular Atrophies of Childhood/drug therapy , Time Factors , Genetic Therapy/methods , Humans , Infant , Infant, Newborn , Infant, Premature , Male , Motor Neurons , Muscular Atrophy, Spinal/diagnosis , Muscular Atrophy, Spinal/genetics , Neonatal Screening , Spinal Muscular Atrophies of Childhood/diagnosis
5.
Muscle Nerve ; 63(6): 902-905, 2021 06.
Article in English | MEDLINE | ID: mdl-33675054

ABSTRACT

INTRODUCTION: This retrospective study reports our tertiary care center's experience with intrathecal nusinersen administration in children and adults with spinal muscular atrophy (SMA). METHODS: We reviewed safety monitoring laboratory results and need for procedural sedation and fluoroscopy-guidance in all SMA patients receiving nusinersen between February 2017 and March 2020. RESULTS: Fifty-eight patients ages 1 mo- 56 y received 494 nusinersen doses. There were 166 laboratory abnormalities in 45 patients. Most were either mild (145 [87.3%]) or were transient proteinuria (18 [10.8%]). None altered nusinersen treatment. Twenty-eight patients required either general anesthesia (75 doses) or anxiolysis with oral midazolam (133 doses, including 6 patients [23 doses] with SMA type I). Eight patients with complicated spines (45 doses) required fluoroscopic guidance. One treatment-related serious adverse event (emesis leading to intubation) occurred during general anesthesia. Two children had asymptomatic increased intracranial pressure. No patients discontinued treatment due to adverse events. DISCUSSION: Intrathecal nusinersen is generally safe and well-tolerated, including in patients requiring oral anxiolysis, general sedation, and fluoroscopic guidance. Frequent serial laboratory monitoring did not identify any persistent significantly abnormal findings or alter treatment.


Subject(s)
Laboratories, Hospital , Muscular Atrophy, Spinal/drug therapy , Oligonucleotides/adverse effects , Adolescent , Adult , Child , Child, Preschool , Female , Humans , Infant , Injections, Spinal , Male , Middle Aged , Oligonucleotides/administration & dosage , Retrospective Studies , Young Adult
6.
N Engl J Med ; 377(18): 1723-1732, 2017 11 02.
Article in English | MEDLINE | ID: mdl-29091570

ABSTRACT

BACKGROUND: Spinal muscular atrophy is an autosomal recessive neuromuscular disorder that is caused by an insufficient level of survival motor neuron (SMN) protein. Nusinersen is an antisense oligonucleotide drug that modifies pre-messenger RNA splicing of the SMN2 gene and thus promotes increased production of full-length SMN protein. METHODS: We conducted a randomized, double-blind, sham-controlled, phase 3 efficacy and safety trial of nusinersen in infants with spinal muscular atrophy. The primary end points were a motor-milestone response (defined according to results on the Hammersmith Infant Neurological Examination) and event-free survival (time to death or the use of permanent assisted ventilation). Secondary end points included overall survival and subgroup analyses of event-free survival according to disease duration at screening. Only the first primary end point was tested in a prespecified interim analysis. To control the overall type I error rate at 0.05, a hierarchical testing strategy was used for the second primary end point and the secondary end points in the final analysis. RESULTS: In the interim analysis, a significantly higher percentage of infants in the nusinersen group than in the control group had a motor-milestone response (21 of 51 infants [41%] vs. 0 of 27 [0%], P<0.001), and this result prompted early termination of the trial. In the final analysis, a significantly higher percentage of infants in the nusinersen group than in the control group had a motor-milestone response (37 of 73 infants [51%] vs. 0 of 37 [0%]), and the likelihood of event-free survival was higher in the nusinersen group than in the control group (hazard ratio for death or the use of permanent assisted ventilation, 0.53; P=0.005). The likelihood of overall survival was higher in the nusinersen group than in the control group (hazard ratio for death, 0.37; P=0.004), and infants with a shorter disease duration at screening were more likely than those with a longer disease duration to benefit from nusinersen. The incidence and severity of adverse events were similar in the two groups. CONCLUSIONS: Among infants with spinal muscular atrophy, those who received nusinersen were more likely to be alive and have improvements in motor function than those in the control group. Early treatment may be necessary to maximize the benefit of the drug. (Funded by Biogen and Ionis Pharmaceuticals; ENDEAR ClinicalTrials.gov number, NCT02193074 .).


Subject(s)
Oligonucleotides, Antisense/therapeutic use , Oligonucleotides/therapeutic use , Spinal Muscular Atrophies of Childhood/drug therapy , Age of Onset , Disease-Free Survival , Double-Blind Method , Female , Humans , Infant , Injections, Spinal , Male , Motor Skills , Oligonucleotides/adverse effects , Oligonucleotides, Antisense/adverse effects , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Respiration, Artificial , Spinal Muscular Atrophies of Childhood/genetics , Spinal Muscular Atrophies of Childhood/mortality , Spinal Muscular Atrophies of Childhood/physiopathology , Survival Analysis , Survival of Motor Neuron 2 Protein/genetics , Survival of Motor Neuron 2 Protein/metabolism
7.
Muscle Nerve ; 62(6): 705-709, 2020 12.
Article in English | MEDLINE | ID: mdl-32893348

ABSTRACT

Childhood onset chronic inflammatory demyelinating polyneuropathy (CIDP) often requires long-term immunomodulatory therapy. We report a comprehensive review of our treatment of pediatric CIDP with a focus on high-dose weekly corticosteroids ("pulse oral corticosteroids"), a treatment method that is not commonly reported. We retrospectively reviewed medical records of pediatric patients with CIDP treated at our center between 2000 and 2018 for whom we had at least 12 mo follow-up. Here, we describe the demographics, disease course, treatment regimens, and long-term outcomes of these patients. Twenty-five patients were identified for analysis. Pulse oral corticosteroid monotherapy was the predominant maintenance treatment in 56% of patients. Patients were followed for a median of 4 y. Side effects were seen in a minority of patients. The probability of a normal exam or being off treatment at last follow-up was similar regardless of predominant maintenance therapy. Pulse oral corticosteroid therapy is a safe and effective long-term treatment option in children with CIDP.


Subject(s)
Glucocorticoids/administration & dosage , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/drug therapy , Prednisone/administration & dosage , Administration, Oral , Adolescent , Adrenal Cortex Hormones/administration & dosage , Child , Child, Preschool , Duration of Therapy , Female , Humans , Immunoglobulins, Intravenous/therapeutic use , Immunologic Factors/therapeutic use , Infant , Irritable Mood , Maintenance Chemotherapy , Male , Polyradiculoneuropathy, Chronic Inflammatory Demyelinating/physiopathology , Pulse Therapy, Drug , Sleep Initiation and Maintenance Disorders/chemically induced , Weight Gain , Young Adult
8.
Muscle Nerve ; 62(1): 41-45, 2020 07.
Article in English | MEDLINE | ID: mdl-32329920

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic has resulted in the reorganization of health-care settings affecting clinical care delivery to patients with Duchenne and Becker muscular dystrophy (DBMD) as well as other inherited muscular dystrophies. The magnitude of the impact of this public health emergency on the care of patients with DBMD is unclear as they are suspected of having an increased risk for severe manifestations of COVID-19. In this article, the authors discuss their consensus recommendations pertaining to care of these patients during the pandemic. We address issues surrounding corticosteroid and exon-skipping treatments, cardiac medications, hydroxychloroquine use, emergency/respiratory care, rehabilitation management, and the conduct of clinical trials. We highlight the importance of collaborative treatment decisions between the patient, family, and health-care provider, considering any geographic or institution-specific policies and precautions for COVID-19. We advocate for continuing multidisciplinary care for these patients using telehealth.


Subject(s)
Betacoronavirus , Coronavirus Infections/complications , Disease Management , Muscular Dystrophy, Duchenne/therapy , Pandemics , Pneumonia, Viral/complications , COVID-19 , Coronavirus Infections/epidemiology , Humans , Muscular Dystrophy, Duchenne/complications , Pneumonia, Viral/epidemiology , SARS-CoV-2
9.
Muscle Nerve ; 62(1): 46-49, 2020 07.
Article in English | MEDLINE | ID: mdl-32329921

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic has resulted in reorganization of healthcare settings affecting the delivery of clinical care to patients with spinal muscular atrophy (SMA). There is a concern that patients with SMA may be at increased risk of manifesting severe symptoms of COVID-19. Currently approved therapies for SMA improve survival and motor function; however, their delivery requires an increased exposure to the health system and a dedicated healthcare team. In this study, we discuss consensus recommendations pertaining to care of SMA patients during the pandemic. We highlight that SMA treatments should not be perceived as elective. Decisions regarding the delay of treatments should be made with consideration of the potential risks of COVID-19 exposure and the risk of that delay. We emphasize the importance of collaborative treatment decisions between the patient, family, and healthcare provider, considering any geographic- or institution-specific policies and precautions for COVID-19.


Subject(s)
Betacoronavirus , Consensus , Coronavirus Infections/complications , Delivery of Health Care/methods , Disease Management , Muscular Atrophy, Spinal/therapy , Pandemics , Pneumonia, Viral/complications , COVID-19 , Coronavirus Infections/epidemiology , Humans , Muscular Atrophy, Spinal/complications , Pneumonia, Viral/epidemiology , SARS-CoV-2
10.
Ann Neurol ; 83(6): 1105-1124, 2018 06.
Article in English | MEDLINE | ID: mdl-29691892

ABSTRACT

OBJECTIVE: Comprehensive clinical characterization of congenital titinopathy to facilitate diagnosis and management of this important emerging disorder. METHODS: Using massively parallel sequencing we identified 30 patients from 27 families with 2 pathogenic nonsense, frameshift and/or splice site TTN mutations in trans. We then undertook a detailed analysis of the clinical, histopathological and imaging features of these patients. RESULTS: All patients had prenatal or early onset hypotonia and/or congenital contractures. None had ophthalmoplegia. Scoliosis and respiratory insufficiency typically developed early and progressed rapidly, whereas limb weakness was often slowly progressive, and usually did not prevent independent walking. Cardiac involvement was present in 46% of patients. Relatives of 2 patients had dilated cardiomyopathy. Creatine kinase levels were normal to moderately elevated. Increased fiber size variation, internalized nuclei and cores were common histopathological abnormalities. Cap-like regions, whorled or ring fibers, and mitochondrial accumulations were also observed. Muscle magnetic resonance imaging showed gluteal, hamstring and calf muscle involvement. Western blot analysis showed a near-normal sized titin protein in all samples. The presence of 2 mutations predicted to impact both N2BA and N2B cardiac isoforms appeared to be associated with greatest risk of cardiac involvement. One-third of patients had 1 mutation predicted to impact exons present in fetal skeletal muscle, but not included within the mature skeletal muscle isoform transcript. This strongly suggests developmental isoforms are involved in the pathogenesis of this congenital/early onset disorder. INTERPRETATION: This detailed clinical reference dataset will greatly facilitate diagnostic confirmation and management of patients, and has provided important insights into disease pathogenesis. Ann Neurol 2018;83:1105-1124.


Subject(s)
Cardiomyopathy, Dilated/congenital , Connectin/genetics , Muscle Proteins/genetics , Muscle, Skeletal/pathology , Female , Humans , Male , Mutation/genetics , Phenotype , Protein Isoforms/genetics
11.
Muscle Nerve ; 59(6): 650-657, 2019 06.
Article in English | MEDLINE | ID: mdl-30706490

ABSTRACT

INTRODUCTION: Glucocorticosteroids (GC) are effective in slowing weakness in boys with Duchenne muscular dystrophy (DMD). METHODS: This is a multisite, 1-year, open-label trial of twice-weekly prednisolone (5 mg/kg/dose) in infants/young boys (0.4-2.4 years) with DMD. We compared changes in Bayley III Scales of Infant Development (Bayley-III) with untreated boys followed for 1 year (historical control cohort [HCC]). Twenty-three of 25 participants completed the study. RESULTS: Treated boys gained an average of 0.5 points on the Bayley-III gross motor scaled score (GMSS) compared with the HCC who, on average, declined 1.3 points (P = 0.03). All boys maintained linear growth, and none developed Cushingoid features. Excessive weight gain occurred in 13 of 23 (56%) boys. DISCUSSION: This study provides evidence that twice-weekly GC is well tolerated in infants and young boys with DMD and improves GMSS. Excessive weight gain is a potential risk. Longer follow-up is required to determine whether early GC initiation is feasible in most infants/boys with DMD. Muscle Nerve 59:650-657, 2019.


Subject(s)
Glucocorticoids/administration & dosage , Muscular Dystrophy, Duchenne/drug therapy , Prednisolone/administration & dosage , Case-Control Studies , Child, Preschool , Cohort Studies , Disease Progression , Humans , Infant , Male , Muscle Weakness/physiopathology , Muscular Dystrophy, Duchenne/physiopathology , Weight Gain
12.
Ann Neurol ; 82(6): 883-891, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29149772

ABSTRACT

OBJECTIVE: Infantile-onset spinal muscular atrophy (SMA) is the most common genetic cause of infant mortality, typically resulting in death preceding age 2. Clinical trials in this population require an understanding of disease progression and identification of meaningful biomarkers to hasten therapeutic development and predict outcomes. METHODS: A longitudinal, multicenter, prospective natural history study enrolled 26 SMA infants and 27 control infants aged <6 months. Recruitment occurred at 14 centers over 21 months within the NINDS-sponsored NeuroNEXT (National Network for Excellence in Neuroscience Clinical Trials) Network. Infant motor function scales (Test of Infant Motor Performance Screening Items [TIMPSI], The Children's Hospital of Philadelphia Infant Test for Neuromuscular Disorders, and Alberta Infant Motor Score) and putative physiological and molecular biomarkers were assessed preceding age 6 months and at 6, 9, 12, 18, and 24 months with progression, correlations between motor function and biomarkers, and hazard ratios analyzed. RESULTS: Motor function scores (MFS) and compound muscle action potential (CMAP) decreased rapidly in SMA infants, whereas MFS in all healthy infants rapidly increased. Correlations were identified between TIMPSI and CMAP in SMA infants. TIMPSI at first study visit was associated with risk of combined endpoint of death or permanent invasive ventilation in SMA infants. Post-hoc analysis of survival to combined endpoint in SMA infants with 2 copies of SMN2 indicated a median age of 8 months at death (95% confidence interval, 6, 17). INTERPRETATION: These data of SMA and control outcome measures delineates meaningful change in clinical trials in infantile-onset SMA. The power and utility of NeuroNEXT to provide "real-world," prospective natural history data sets to accelerate public and private drug development programs for rare disease is demonstrated. Ann Neurol 2017;82:883-891.


Subject(s)
Spinal Muscular Atrophies of Childhood/blood , Spinal Muscular Atrophies of Childhood/diagnosis , Biomarkers/blood , Child, Preschool , Cohort Studies , Female , Humans , Infant , Longitudinal Studies , Male , Prospective Studies , Spinal Muscular Atrophies of Childhood/genetics , Survival of Motor Neuron 1 Protein/blood , Survival of Motor Neuron 1 Protein/genetics , Survival of Motor Neuron 2 Protein/blood , Survival of Motor Neuron 2 Protein/genetics
13.
Am J Respir Crit Care Med ; 196(4): 512-519, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28636407

ABSTRACT

Development of novel therapeutics for treatment of Duchenne muscular dystrophy (DMD) has led to clinical trials that include pulmonary endpoints that allow assessment of respiratory muscle status, especially in nonambulatory subjects. Parent Project Muscular Dystrophy (PPMD) convened a workshop in Bethesda, Maryland, on April 14 and 15, 2016, to summarize published respiratory data in DMD and give guidance to clinical researchers assessing the effect of interventions on pulmonary outcomes in DMD.


Subject(s)
Lung/physiopathology , Muscular Dystrophy, Duchenne/physiopathology , Respiratory Muscles/physiopathology , Humans
14.
Neurogenetics ; 17(3): 173-8, 2016 07.
Article in English | MEDLINE | ID: mdl-27094857

ABSTRACT

Exome sequencing is an effective way to identify genetic causes of etiologically heterogeneous conditions such as developmental delay and intellectual disabilities. Using exome sequencing, we have identified four patients with similar phenotypes of developmental delay, intellectual disability, failure to thrive, hypotonia, ataxia, and tooth enamel defects who all have the same de novo R331W missense variant in C-terminal binding protein 1 (CTBP1). CTBP1 is a transcriptional regulator critical for development by coordinating different regulatory pathways. The R331W variant found in these patients is within the C-terminal portion of the PLDLS (Pro-Leu-Asp-Leu-Ser) binding cleft, which is the domain through which CTBP1, interacts with chromatin-modifying enzymes and mediates chromatin-dependent gene repression pathways. This is the first report of mutations within CTBP1 in association with any human disease.


Subject(s)
Alcohol Oxidoreductases/genetics , Ataxia/genetics , DNA-Binding Proteins/genetics , Dental Enamel/pathology , Developmental Disabilities/genetics , Muscle Hypotonia/genetics , Mutation, Missense , Adult , Ataxia/complications , Child , Developmental Disabilities/complications , Female , Humans , Intellectual Disability/complications , Intellectual Disability/genetics , Male , Muscle Hypotonia/complications , Exome Sequencing , Young Adult
15.
Muscle Nerve ; 54(2): 186-91, 2016 08.
Article in English | MEDLINE | ID: mdl-27170260

ABSTRACT

New developments in the rapid diagnosis and treatment of boys with Duchenne muscular dystrophy (DMD) have led to growing enthusiasm for instituting DMD newborn screening (NBS) in the United States. Our group has been interested in developing clinical guidance to be implemented consistently in specialty care clinics charged with the care of presymptomatically identified newborns referred after DMD-NBS. We reviewed the existing literature covering patient-centered clinical follow-up after NBS, educational material from public health and advocacy sites, and federal recommendations on effective NBS follow-up. We discussed the review as a group and added our own experience to develop materials suitable for initial parent and primary care provider education. These materials and a series of templates for subspecialist encounters could be used to provide consistent care across centers and serve as the basis for ongoing quality improvement. Muscle Nerve 54: 186-191, 2016.


Subject(s)
Muscular Dystrophy, Duchenne/diagnosis , Neonatal Screening/methods , Female , Follow-Up Studies , Humans , Infant, Newborn , Male
16.
Muscle Nerve ; 54(4): 681-9, 2016 10.
Article in English | MEDLINE | ID: mdl-26930423

ABSTRACT

INTRODUCTION: Outcomes sensitive to change over time in non-ambulatory boys/men with Duchenne muscular dystrophy (DMD) are not well-established. METHODS: Subjects (n = 91; 16.8 ± 4.5 years old) were assessed at baseline and 6-month intervals for 2 years. We analyzed all subjects using an intent-to-treat model and a subset of stronger subjects with Brooke Scale score ≤4, using repeated measures. RESULTS: Eight patients (12-33 years old) died during the study. Sixty-six completed 12-month follow-up, and 51 completed 24-month follow-up. Those taking corticosteroids performed better at baseline, but rates of decline were similar. Forced vital capacity percent predicted (FVC% predicted) declined significantly only after 2 years. However, Brooke and Egen Klassifikation (EK) Scale scores, elbow flexion, and grip strength declined significantly over both 1 and 2 years. CONCLUSION: Brooke and EK Scale scores, elbow flexion, and grip strength were outcomes most responsive to change. FVC% predicted was responsive to change over 2 years. Corticosteroids benefited non-ambulatory DMD subjects but did not affect decline rates of measures tested here. Muscle Nerve 54: 681-689, 2016.


Subject(s)
Mobility Limitation , Muscular Dystrophy, Duchenne/diagnosis , Muscular Dystrophy, Duchenne/physiopathology , Patient Participation/methods , Adolescent , Adrenal Cortex Hormones/therapeutic use , Adult , Child , Follow-Up Studies , Hand Strength/physiology , Humans , Male , Muscular Dystrophy, Duchenne/drug therapy , Range of Motion, Articular/physiology , Vital Capacity/physiology , Young Adult
17.
J Paediatr Child Health ; 52(9): 896-900, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27650145

ABSTRACT

AIM: To develop and evaluate an online educational package instructing paediatricians and trainees in the diagnosis and management of a first unprovoked seizure in children. METHODS: The E-learning content was created following a comprehensive literature review that referenced current international guidelines. Rigorous consultation with local paediatric neurologists, paediatricians and epilepsy nurses was undertaken. A series of learning modules was created and sequenced to reflect steps needed to achieve optimal diagnosis and management in a real-life situation of a child presenting with a paroxysmal event. Paediatric registrars and advanced trainees from the Sydney Children's Hospitals Network were assessed before and after using the E-learning Resource. Measures included general epilepsy knowledge, case-based scenario knowledge; self-rated measures of satisfaction with instruction and confidence regarding clinical approach to the child with first unprovoked seizure; and open ended questions evaluating the usefulness of the E-learning resource. RESULTS: Performance on measures of general epilepsy knowledge and on the seizure-related case scenarios improved significantly following completion of the E-learning as did self-rated satisfaction with instruction and confidence across all aspects of managing first seizure. CONCLUSIONS: The E-learning resource has been validated as a useful educational resource regarding the first afebrile unprovoked seizure for paediatricians.


Subject(s)
Computer-Assisted Instruction/methods , Education, Medical, Continuing/methods , Education, Medical, Graduate/methods , Pediatrics/education , Seizures , Adult , Child , Clinical Competence , Diagnosis, Differential , Female , Humans , Male , Middle Aged , New South Wales , Seizures/diagnosis , Seizures/etiology , Seizures/therapy
18.
FASEB J ; 28(5): 2047-61, 2014 May.
Article in English | MEDLINE | ID: mdl-24500923

ABSTRACT

Duchenne muscular dystrophy in boys progresses rapidly to severe impairment of muscle function and death in the second or third decade of life. Current supportive therapy with corticosteroids results in a modest increase in strength as a consequence of a general reduction in inflammation, albeit with potential untoward long-term side effects and ultimate failure of the agent to maintain strength. Here, we demonstrate that alternative approaches that rescue defective autophagy in mdx mice, a model of Duchenne muscular dystrophy, with the use of rapamycin-loaded nanoparticles induce a reproducible increase in both skeletal muscle strength and cardiac contractile performance that is not achievable with conventional oral rapamycin, even in pharmacological doses. This increase in physical performance occurs in both young and adult mice, and, surprisingly, even in aged wild-type mice, which sets the stage for consideration of systemic therapies to facilitate improved cell function by autophagic disposal of toxic byproducts of cell death and regeneration.


Subject(s)
Autophagy/drug effects , Immunosuppressive Agents/administration & dosage , Myocardium/metabolism , Nanoparticles/chemistry , Sirolimus/administration & dosage , Adrenal Cortex Hormones/therapeutic use , Animals , Cell Death , Creatine Kinase/metabolism , Drug Delivery Systems , Fibrosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Strength , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/pathology , Myocardial Contraction , Regeneration , Tissue Distribution
19.
Muscle Nerve ; 52(3): 334-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25704979

ABSTRACT

INTRODUCTION: Quantitative muscle ultrasound (QUS) in boys with Duchenne muscular dystrophy (DMD) shows increased echointensity as muscle is replaced with fat and fibrosis. Studies of quantitative ultrasound in infants/young boys with DMD over time have not been reported. METHODS: We used calibrated muscle backscatter (cMB), a reproducible measure of ultrasound echointensity, to quantify muscle pathology in 5 young boys with DMD (ages 0.5-2.8 years) over 17-29 months. We compared the results with repeated assessments of function (n = 4) and with muscle ultrasound images from a cross-section of 6 male controls (0.6-3.1 years). RESULTS: cMB in boys with DMD increased (worsened) over time (P < 0.001), whereas function improved. After age 2 years, cMB in most (4 of 5) boys with DMD was higher than in any control. CONCLUSIONS: QUS measures disease progression in young boys with DMD despite functional improvements. QUS could be employed as an outcome measure for serial assessment of young boys with DMD.


Subject(s)
Muscular Dystrophy, Duchenne/diagnostic imaging , Quadriceps Muscle/diagnostic imaging , Arm , Case-Control Studies , Child, Preschool , Disease Progression , Humans , Infant , Male , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/physiopathology , Quadriceps Muscle/physiopathology , Ultrasonography
20.
Muscle Nerve ; 52(4): 592-7, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25702806

ABSTRACT

INTRODUCTION: Electrical impedance myography (EIM) is a non-invasive, painless, objective technique to quantify muscle pathology. METHODS: We measured EIM in 8 arm and leg muscles in 61 boys with Duchenne muscular dystrophy (DMD) and 31 healthy boys, ages 3-12 years, at 5 centers. We determined the reliability of EIM and compared results in boys with DMD to controls and to 6-minute walk distance (6MWD), North Star Ambulatory Assessment (NSAA), timed functional tests (TFTs), and strength (hand-held dynamometry). RESULTS: EIM was well tolerated and had good inter- and intrarater reliability (intraclass correlation coefficient 0.81-0.96). The averaged EIM phase value from all muscles was higher (P < 0.001) in controls (10.45 ± 2.29) than boys with DMD (7.31 ± 2.23), and correlated (P ≤ 0.001) with 6MWD (r = 0.55), NSAA (r = 0.66), TFTs (r = -0.56), and strength (r = 0.44). CONCLUSION: EIM is a reliable and valid measure of disease severity in DMD. Longitudinal studies comparing EIM with other assessments over time in DMD are warranted.


Subject(s)
Electric Impedance , Muscle Strength/physiology , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Age Factors , Child , Child, Preschool , Female , Humans , Male , Muscle Strength Dynamometer , Myography , ROC Curve , Reproducibility of Results , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL