51
|
Lavezzi SM, Rocchetti M, Bettica P, Petrini S, De Nicolao G. Assessing drug effect from distributional data: A population approach with application to Duchenne Muscular Dystrophy treatment. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2019; 178:329-342. [PMID: 31416560 DOI: 10.1016/j.cmpb.2019.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 05/13/2019] [Accepted: 06/03/2019] [Indexed: 06/10/2023]
Abstract
BACKGROUND AND OBJECTIVE In Duchenne Muscular Dystrophy (DMD) treatment, muscle fiber size can be considered as an indicator of muscle health and function. In particular, the statistical distribution of fibers cross-sectional areas (CSAs) has been used as quantitative efficacy endpoint. For each patient, assessment of treatment effect relies on the comparison of pre- and post-treatment biopsies. Since biopsies provide "distributional data", i.e. empirical distributions of fibers CSA, the comparison must be carried out between the empirical pre- and post-treatment distributions. METHODS Here, distributional fiber CSA data are analyzed by means of a hierarchical statistical model based on the population approach, considering both the single patient and the population level. RESULTS The proposed method was used to assess the histological clinical effects of Givinostat, a compound under study for DMD treatment. At the single patient level, a two-component Gaussian mixture adequately represents pre- and post-treatment distributions of log-transformed CSAs; drug effect is described via a dose-dependent multiplicative increase of muscle fiber size. The single patient model was also validated via muscle composition data. At the patient population level, typical model parameters and inter-patient variabilities were obtained. CONCLUSIONS The proposed methodological approach completely characterizes fiber CSA distributions and quantifies drug effect on muscle fiber size, both at the single patient and at the patient population level. This approach might be applied also in other contexts, where outcomes measured in terms of distributional data are to be assessed.
Collapse
Affiliation(s)
- S M Lavezzi
- Dipartimento di Ingegneria Industriale e dell'Informazione, Università degli Studi di Pavia, via Ferrata 5, Pavia 27100, Italy.
| | - M Rocchetti
- Independent Consultant, via Marcantonio Colonna 43, Milan 20149, Italy
| | - P Bettica
- Italfarmaco S.p.A., via dei Lavoratori 54, Cinisello Balsamo, Milan 20092, Italy
| | - S Petrini
- Confocal Microscopy Core Facility Research Center, Bambino Gesù Children's Hospital, Viale San Paolo 15, Rome 00146, Italy
| | - G De Nicolao
- Dipartimento di Ingegneria Industriale e dell'Informazione, Università degli Studi di Pavia, via Ferrata 5, Pavia 27100, Italy
| |
Collapse
|
52
|
Hoskens J, Goemans N, Feys H, De Waele L, Van den Hauwe M, Klingels K. Normative data and percentile curves for the three-minute walk test and timed function tests in healthy Caucasian boys from 2.5 up to 6 years old. Neuromuscul Disord 2019; 29:585-600. [DOI: 10.1016/j.nmd.2019.06.597] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 06/06/2019] [Accepted: 06/25/2019] [Indexed: 10/26/2022]
|
53
|
Flotats-Bastardas M, Ebrahimi-Fakhari D, Bernert G, Ziegler A, Schlachter K, Poryo M, Hahn A, Meyer S. [Non-ambulatory patients with Duchenne muscular dystrophy : Recommendations for monitoring disease progression and course of treatment]. DER NERVENARZT 2019; 90:817-823. [PMID: 31270551 DOI: 10.1007/s00115-019-0754-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a severe X‑linked recessive neuromuscular disorder. In children without corticosteroid therapy, progressive muscular weakness is associated with loss of ambulation on average by the age of 9.5 years. OBJECTIVE, MATERIAL AND METHODS On the basis of current guidelines, a group of experts in this field defined a number of clinical parameters and examinations that should be performed on a regular basis to assess changes over time in non-ambulant patients. RESULTS AND CONCLUSION To assess function of the upper extremities the Brooke upper extremity functional rating scale or the performance of upper limb test should be used. For assessment of pulmonary function measurement of forced vital capacity (FVC) is recommended. The extent of cardiac involvement can best be evaluated using cardiac magnetic resonance imaging (MRI), measurement of the ejection fraction (EF) and the left ventricular shortening fraction (LVSF) by echocardiography. The pediatric quality of life inventory should be used for assessment of quality of life. In addition, the body mass index (BMI), the number of infections and need for in-hospital treatment as well as early detection of orthopedic problems, most importantly the development of scoliosis should be monitored. After transition from pediatric to adult care DMD patients should be primarily cared for by adult neurologists and specialists in pulmonary and cardiac medicine.
Collapse
Affiliation(s)
- Marina Flotats-Bastardas
- Klinik für Allgemeine Pädiatrie und Neonatologie, Sektion Neuropädiatrie, Universitätsklinikum des Saarlandes, Geb. 9, 66421, Homburg, Deutschland
| | - Daniel Ebrahimi-Fakhari
- Klinik für Allgemeine Pädiatrie und Neonatologie, Sektion Neuropädiatrie, Universitätsklinikum des Saarlandes, Geb. 9, 66421, Homburg, Deutschland
| | - Günther Bernert
- Sozialmedizinisches Zentrum Süd, Kaiser-Franz-Josef-Spital mit Gottfried von Preyer'schem Kinderspital, Wien, Österreich
| | - Andreas Ziegler
- Sektion für Neuropädiatrie und Stoffwechselmedizin, Zentrum für Kinder- und Jugendmedizin, Universitätsklinikum Heidelberg, Heidelberg, Deutschland
| | - Kurt Schlachter
- Klinik für Kinder- und Jugendheilkunde, Landeskrankenhaus Bregenz, Bregenz, Österreich
| | - Martin Poryo
- Klinik für Pädiatrische Kardiologie, Universitätsklinikum des Saarlandes, Homburg, Deutschland
| | - Andreas Hahn
- Abteilung Kinderneurologie, Sozialpädiatrie und Epileptologie, Universitätskinderklinik Gießen, Gießen, Deutschland
| | - Sascha Meyer
- Klinik für Allgemeine Pädiatrie und Neonatologie, Sektion Neuropädiatrie, Universitätsklinikum des Saarlandes, Geb. 9, 66421, Homburg, Deutschland.
| |
Collapse
|
54
|
Alfano LN, Charleston JS, Connolly AM, Cripe L, Donoghue C, Dracker R, Dworzak J, Eliopoulos H, Frank DE, Lewis S, Lucas K, Lynch J, Milici AJ, Flynt A, Naughton E, Rodino-Klapac LR, Sahenk Z, Schnell FJ, Young GD, Mendell JR, Lowes LP. Long-term treatment with eteplirsen in nonambulatory patients with Duchenne muscular dystrophy. Medicine (Baltimore) 2019; 98:e15858. [PMID: 31261494 PMCID: PMC6617421 DOI: 10.1097/md.0000000000015858] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
This analysis aims to describe the outcomes of two nonambulatory patients with Duchenne muscular dystrophy (DMD) who participated in two clinical studies. The two consecutive trials of eteplirsen (studies 201 and 202) were conducted in patients with DMD (N = 12) and confirmed genetic mutations amenable to exon 51 skipping.In study 201, 12 patients were randomized to receive once-weekly, double-blind intravenous infusions of eteplirsen 30 or 50 mg/kg or placebo for 24 weeks; patients then received open-label eteplirsen during weeks 25 through 28. All 12 patients continued onto open-label extension study 202 and received long-term treatment with eteplirsen. We compared cardiac, pulmonary, and upper limb function and dystrophin production in the nonambulatory twin patients versus the 10 ambulatory patients through 240 combined treatment weeks.Ten study patients remained ambulatory through both studies, while the identical twin patients both experienced early, rapid loss of ambulation. The twin patients had greater disease severity at baseline (6-minute walk test [6MWT], 330 and 256 m) versus the other patients (n = 10; 6MWT range, 341-418 m). They maintained cardiac and upper limb function through combined week 240, with outcomes similar to those of the patients who remained ambulatory. Dystrophin production was confirmed following eteplirsen treatment.Despite the loss of ambulation, other markers of disease progression remained relatively stable in the eteplirsen-treated twin patients and were similar to those of the ambulatory patients.
Collapse
Affiliation(s)
- Lindsay N. Alfano
- Pediatrics, Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH
| | | | - Anne M. Connolly
- Currently: Pediatrics, Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH
- Department of Neurology, Washington University School of Medicine, St. Louis, MO
| | - Linda Cripe
- Pediatrics, Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH
| | | | - Robert Dracker
- Summerwood Pediatrics/Infusacare Medical Services, PC, Liverpool, NY
| | | | | | | | - Sarah Lewis
- Pediatrics, Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH
| | | | | | | | | | | | - Louise R. Rodino-Klapac
- Pediatrics, Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH
- Currently: Sarepta Therapeutics, Inc., Cambridge, MA, USA
| | - Zarife Sahenk
- Pediatrics, Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH
| | | | | | - Jerry R. Mendell
- Pediatrics, Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH
| | - Linda P. Lowes
- Pediatrics, Center for Gene Therapy, Nationwide Children's Hospital, Columbus, OH
| |
Collapse
|
55
|
Yin L, Xie ZY, Xu HY, Zheng SS, Wang ZX, Xiao JX, Yuan Y. T2 Mapping and Fat Quantification of Thigh Muscles in Children with Duchenne Muscular Dystrophy. Curr Med Sci 2019; 39:138-145. [PMID: 30868504 DOI: 10.1007/s11596-019-2012-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 12/12/2018] [Indexed: 11/24/2022]
Abstract
Quantitative magnetic resonance image (MRI) in individual muscles may be useful for monitoring disease progression in Duchenne muscular dystrophy (DMD). The purpose of this study was to measure T2 relaxation time of thigh muscles in children with DMD and healthy boys, and to correlate the T2 relaxation time of muscles with the fat fraction (FF) at quantitative magnetic resonance and results of clinical assessment. Thirty-two boys with DMD and 18 healthy boys were evaluated with T2 mapping and three-point Dixon MRI. Age, body mass index (BMI), muscle strength assessment, timed functional tests (time to walk or run 10 metres, rise from the floor and ascend four stairs), and the North Star Ambulatory Assessment (NSAA) were evaluated. Spearman's correlation was used to assess the relationships between FF and clinical assessments and T2 relaxation time. The mean T2 relaxation time of thigh muscles in DMD was significantly longer than that in the control group (P<0.05), except for the gracilis (P=0.952). The gracilis, sartorius and adductor longus were relatively spared by fatty infiltration in DMD patients. The T2 relaxation time was correlated significantly with the mean FF in all muscles. Age, BMI, total muscle strength score, timed functional tests and NSAA were significantly correlated with the overall mean T2 relaxation time. T2 mapping may prove clinically useful in monitoring muscle changes as a result of the disease process and in predicting the outcome of DMD patients.
Collapse
Affiliation(s)
- Liang Yin
- Department of Radiology, Peking University First Hospital, Beijing, 100034, China
| | - Zhi-Ying Xie
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China
| | - Hai-Yan Xu
- Department of Radiology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Sui-Sheng Zheng
- Department of Radiology, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zhao-Xia Wang
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China
| | - Jiang-Xi Xiao
- Department of Radiology, Peking University First Hospital, Beijing, 100034, China.
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China.
| |
Collapse
|
56
|
Roy B, Darras BT, Zaidman CM, Wu JS, Kapur K, Rutkove SB. Exploring the relationship between electrical impedance myography and quantitative ultrasound parameters in Duchenne muscular dystrophy. Clin Neurophysiol 2019; 130:515-520. [PMID: 30772764 DOI: 10.1016/j.clinph.2019.01.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/10/2018] [Accepted: 01/13/2019] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Quantitative ultrasound (QUS), including grayscale level analysis (GLA) and quantitative backscatter analysis (QBA), and electrical impedance myography (EIM) have been proposed as biomarkers in Duchenne muscular dystrophy (DMD). However, the relationship between these methods has not been assessed. METHODS QUS values (including GLA and QBA) and several EIM measures were recorded from six muscles in 36 DMD and 29 healthy boys between ages 5 and 13 years at baseline, 6-months, and 12-months. RESULTS In the DMD boys, a moderate correlation was noted between QUS and EIM parameters, with the strongest correlations being identified for averaged muscle values. Of the individual muscles, biceps brachii and deltoid showed the strongest correlations. For example, in biceps, the QBA/EIM correlation coefficient (Spearman rho) was ≥0.70 (p < 0.01). Importantly, changes in QUS values over 12 months also correlated moderately with changes in EIM parameters and EIM/QBA rho values mostly varied between -0.53 and -0.70 (p ≤ 0.02). No significant correlations were identified in the healthy boys. CONCLUSIONS A moderate correlation of QUS with EIM in DMD boys suggests that the two technologies provide related data but are sensitive to different pathological features of muscle. SIGNIFICANCE The use of both technologies jointly in assessing DMD progression and response to therapy should be considered.
Collapse
Affiliation(s)
- Bhaskar Roy
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | | | - Jim S Wu
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Kush Kapur
- Boston Children's Hospital, Boston, MA, USA
| | | |
Collapse
|
57
|
Luiz LC, Marson FAL, Bresciani Almeida CC, Toro AADC, Nucci A, Ribeiro JD. Analysis of motor and respiratory function in Duchenne muscular dystrophy patients. Respir Physiol Neurobiol 2019; 262:1-11. [PMID: 30660861 DOI: 10.1016/j.resp.2019.01.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 06/28/2018] [Accepted: 01/17/2019] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Duchenne muscular dystrophy(DMD) shows motor and respiratory impairment. METHODS 19 DMD patients (DMDG) (nine ambulatory and 10 non-ambulatory) were evaluated through motor function measure (MFM), 6-minute walk test (6MWT), respiratory muscle strength, cough peak flow, spirometry and volumetric capnography (VCap) tools. Control group that performed spirometry and VCap (CG1-n = 17) were different from those that performed the 6MWT (CG2-n = 8). RESULTS The follow tools were assessed (p < 0.05): (i) MFM: Ambulatory patients showed higher values than non-ambulatory patients; (ii) 6MWT: DMDG walked a shorter distance and showed higher respiratory rate at rest and heart rate (HR) at rest than CG2; (iii) Spirometry: DMDG and non-ambulatory patients had minor values achieved in spirometry when compared with CG1 and ambulatory patients, respectively; (iv) VCap: DMDG when compared with CG1 showed: (<11 years-old) lower values in VCap parameters; (>11 years-old): higher HR and lower slope 2. There was correlation between spirometry, mainly for zFEV1/FVC, and MFM. CONCLUSION DMDG showed motor (MFM/6MWT) and respiratory (spirometry/VCap) deterioration when compared with CG. Non-ambulatory condition was associated with worse MFM and spirometry.
Collapse
Affiliation(s)
- Lívia Cocato Luiz
- Department of Pediatrics, School of Medical Sciences, University of Campinas, 13081-970, P.O. Box: 6111 Campinas, São Paulo, Brazil.
| | - Fernando Augusto Lima Marson
- Department of Pediatrics, School of Medical Sciences, University of Campinas, 13081-970, P.O. Box: 6111 Campinas, São Paulo, Brazil; Department of Medical Genetics and Genomic Medicine, School of Medical Sciences, University of Campinas, 13081-970, P.O. Box: 6111 Campinas, São Paulo, Brazil; Laboratory of Pulmonary Physiology, Center for Pediatrics Investigation, School of Medical Sciences, University of Campinas, 13081-970, P.O. Box: 6111 Campinas, São Paulo, Brazil.
| | - Celize Cruz Bresciani Almeida
- Department of Pediatrics, School of Medical Sciences, University of Campinas, 13081-970, P.O. Box: 6111 Campinas, São Paulo, Brazil.
| | | | - Anamarli Nucci
- Department of Neurology, School of Medical Sciences, University of Campinas, 13081-970, P.O. Box: 6111 Campinas, São Paulo, Brazil.
| | - José Dirceu Ribeiro
- Department of Pediatrics, School of Medical Sciences, University of Campinas, 13081-970, P.O. Box: 6111 Campinas, São Paulo, Brazil; Laboratory of Pulmonary Physiology, Center for Pediatrics Investigation, School of Medical Sciences, University of Campinas, 13081-970, P.O. Box: 6111 Campinas, São Paulo, Brazil.
| |
Collapse
|
58
|
Progression of Ankle Plantarflexion Contractures and Functional Decline in Duchenne Muscular Dystrophy: Implications for Physical Therapy Management. Pediatr Phys Ther 2019; 31:61-66. [PMID: 30557283 DOI: 10.1097/pep.0000000000000553] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE This study characterizes the progressive loss of ankle dorsiflexion range of motion in boys with Duchenne muscular dystrophy (DMD), the relationship to functional decline, and the implications for physical therapy management. METHODS Longitudinal data for 332 boys with DMD were extracted from medical records and analyzed. Summary statistics for age, number of visits, ankle dorsiflexion measures, and North Star Ambulatory Assessment (NSAA) scores were computed. RESULTS Ankle dorsiflexion motion ranged from -32.5 to 25 degrees. Progression of ankle contractures is demonstrated by a trend line: slope -1.43 per year. NSAA score was estimated to decline approximately 0.23 points per 1 degree of ankle dorsiflexion lost. CONCLUSIONS The results of this study describe the progression of ankle contractures and functional decline in DMD. The findings may help inform decisions regarding interventions to support participants with DMD and their families.
Collapse
|
59
|
Arora H, Willcocks RJ, Lott DJ, Harrington AT, Senesac CR, Zilke KL, Daniels MJ, Xu D, Tennekoon GI, Finanger EL, Russman BS, Finkel RS, Triplett WT, Byrne BJ, Walter GA, Sweeney HL, Vandenborne K. Longitudinal timed function tests in Duchenne muscular dystrophy: ImagingDMD cohort natural history. Muscle Nerve 2018; 58:631-638. [PMID: 29742798 DOI: 10.1002/mus.26161] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 05/03/2018] [Accepted: 05/05/2018] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Tests of ambulatory function are common clinical trial endpoints in Duchenne muscular dystrophy (DMD). Using these tests, the ImagingDMD study has generated a large data set that can describe the contemporary natural history of DMD in 5-12.9-year-olds. METHODS Ninety-two corticosteroid-treated boys with DMD and 45 controls participated in this longitudinal study. Participants performed the 6-minute walk test (6MWT) and timed function tests (TFT: 10-m walk/run, climbing 4 stairs, supine to stand). RESULTS Boys with DMD had impaired functional performance even at 5-6.9 years old. Boys older than 7 had significant declines in function over 1 year for 10-m walk/run and 6MWT. Eighty percent of participants could perform all functional tests at 9 years old. TFTs appear to be slightly more responsive and predictive of disease progression than the 6MWT in 7-12.9 year olds. DISCUSSION This study provides insight into the contemporary natural history of key functional endpoints in DMD. Muscle Nerve 58: 631-638, 2018.
Collapse
Affiliation(s)
- Harneet Arora
- Department of Physical Therapy, University of Florida, Box 100154, UFHSC, Gainesville, Florida, 32610, USA
| | - Rebecca J Willcocks
- Department of Physical Therapy, University of Florida, Box 100154, UFHSC, Gainesville, Florida, 32610, USA
| | - Donovan J Lott
- Department of Physical Therapy, University of Florida, Box 100154, UFHSC, Gainesville, Florida, 32610, USA
| | - Ann T Harrington
- The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Claudia R Senesac
- Department of Physical Therapy, University of Florida, Box 100154, UFHSC, Gainesville, Florida, 32610, USA
| | | | - Michael J Daniels
- Department of Statistics, University of Florida, Gainesville, Florida, USA
| | - Dandan Xu
- Department of Statistics & Data Sciences, The University of Texas at Austin, Austin, Texas, USA
| | - Gihan I Tennekoon
- The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | | | | | | | - William T Triplett
- Department of Physical Therapy, University of Florida, Box 100154, UFHSC, Gainesville, Florida, 32610, USA
| | - Barry J Byrne
- Department of Pediatrics and Molecular Genetics and Microbiology, Powell Gene Therapy Center, University of Florida, Gainesville, Florida, USA
| | - Glenn A Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, Florida, USA
| | - H Lee Sweeney
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, Florida, USA
| | - Krista Vandenborne
- Department of Physical Therapy, University of Florida, Box 100154, UFHSC, Gainesville, Florida, 32610, USA
| |
Collapse
|
60
|
McDonald CM, Wong B, Flanigan KM, Wilson R, de Kimpe S, Lourbakos A, Lin Z, Campion G. Placebo-controlled Phase 2 Trial of Drisapersen for Duchenne Muscular Dystrophy. Ann Clin Transl Neurol 2018; 5:913-926. [PMID: 30128316 PMCID: PMC6093847 DOI: 10.1002/acn3.579] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 04/11/2018] [Accepted: 04/14/2018] [Indexed: 12/25/2022] Open
Abstract
Objective This double‐blind, randomized, placebo‐controlled Phase 2 study (NCT01462292) assessed the 24‐week efficacy, safety, tolerability, and pharmacokinetics of two different subcutaneous drisapersen doses, and the 24‐week off‐dose persistent effect, in ambulant Duchenne muscular dystrophy (DMD) patients. Methods Male DMD patients (≥5 years; time to rise from floor ≤15 s) were randomized to drisapersen 3 mg/kg/week, 6 mg/kg/week or placebo. The primary efficacy endpoint was change from baseline in 6‐minute walking distance (6MWD) at week 24. Secondary endpoints included changes in timed function tests, muscle strength, and pulmonary function tests. Results Fifty‐one patients were randomized to placebo (N = 16), drisapersen 3 mg/kg/week (N = 17) or 6 mg/kg/week (N = 18). All but 2 patients had baseline rise from floor time <7 s. This study was exploratory and not prospectively powered; however, a difference in mean 6MWD versus placebo in favor of drisapersen 6 mg/kg/week was observed at week 24 (27.1 m; P = 0.069) and maintained 24 weeks off‐treatment (27.9 m; P = 0.177). The 3 mg/kg/week group showed no statistically significant difference in mean 6MWD versus placebo. For some secondary endpoints, a more positive response in favor of drisapersen 6 mg/kg/week compared to placebo was shown. Drisapersen had a long half‐life with steady state reached after approximately 36 weeks. Most common adverse events in both drisapersen groups were related to injection site reactions and subclinical proteinuria. Interpretation Drisapersen 6 mg/kg/week for 24 weeks resulted in a treatment benefit in 6MWD, largely maintained 24 weeks off‐treatment. This study provided insights for further studies to optimize dosage regimen.
Collapse
Affiliation(s)
| | - Brenda Wong
- Cincinnati Children's Hospital Cincinnati Ohio
| | | | | | | | | | | | | | | |
Collapse
|
61
|
Abstract
Duchenne muscular dystrophy (DMD) is the most common form of muscular dystrophy in childhood. Mutations of the DMD gene destabilize the dystrophin associated glycoprotein complex in the sarcolemma. Ongoing mechanical stress leads to unregulated influx of calcium ions into the sarcoplasm, with activation of proteases, release of proinflammatory cytokines, and mitochondrial dysfunction. Cumulative damage and reparative failure leads to progressive muscle necrosis, fibrosis, and fatty replacement. Although there is presently no cure for DMD, scientific advances have led to many potential disease-modifying treatments, including dystrophin replacement therapies, upregulation of compensatory proteins, anti-inflammatory agents, and other cellular targets. Recently approved therapies include ataluren for stop codon read-through and eteplirsen for exon 51 skipping of eligible individuals. The purpose of this chapter is to summarize the clinical features of DMD, to describe current outcome measures used in clinical studies, and to highlight new emerging therapies for affected individuals.
Collapse
|
62
|
Goemans N, Mercuri E, Belousova E, Komaki H, Dubrovsky A, McDonald CM, Kraus JE, Lourbakos A, Lin Z, Campion G, Wang SX, Campbell C. A randomized placebo-controlled phase 3 trial of an antisense oligonucleotide, drisapersen, in Duchenne muscular dystrophy. Neuromuscul Disord 2017; 28:4-15. [PMID: 29203355 DOI: 10.1016/j.nmd.2017.10.004] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 09/23/2017] [Accepted: 10/17/2017] [Indexed: 01/16/2023]
Abstract
This 48-week, randomized, placebo-controlled phase 3 study (DMD114044; NCT01254019) evaluated efficacy and safety of subcutaneous drisapersen 6 mg/kg/week in 186 ambulant boys aged ≥5 years, with Duchenne muscular dystrophy (DMD) resulting from an exon 51 skipping amenable mutation. Drisapersen was generally well tolerated, with injection-site reactions and renal events as most commonly reported adverse events. A nonsignificant treatment difference (P = 0.415) in the change from baseline in six-minute walk distance (6MWD; primary efficacy endpoint) of 10.3 meters in favor of drisapersen was observed at week 48. Key secondary efficacy endpoints (North Star Ambulatory Assessment, 4-stair climb ascent velocity, and 10-meter walk/run velocity) gave consistent findings. Lack of statistical significance was thought to be largely due to greater data variability and subgroup heterogeneity. The increased standard deviation alone, due to less stringent inclusion/exclusion criteria, reduced the statistical power from pre-specified 90% to actual 53%. Therefore, a post-hoc analysis was performed in 80 subjects with a baseline 6MWD 300-400 meters and ability to rise from floor. A statistically significant improvement in 6MWD of 35.4 meters (P = 0.039) in favor of drisapersen was observed in this subpopulation. Results suggest that drisapersen could have benefit in a less impaired population of DMD subjects.
Collapse
Affiliation(s)
- Nathalie Goemans
- Department of Pediatrics and Child Neurology, University Hospitals Leuven, Leuven, Belgium.
| | | | - Elena Belousova
- Research and Clinical Institute of Pediatrics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Hirofumi Komaki
- Department of Child Neurology, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Alberto Dubrovsky
- Fundacion Cenit, Instituto de Neurociencias, Fundación Favaloro, Buenos Aires, Argentina
| | - Craig M McDonald
- School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - John E Kraus
- GlaxoSmithKline, Research Triangle Park, NC, USA
| | | | | | | | | | - Craig Campbell
- Paediatric Neurology, Schulich School of Medicine, Western University, London, Canada
| | | |
Collapse
|
63
|
Timed function tests, motor function measure, and quantitative thigh muscle MRI in ambulant children with Duchenne muscular dystrophy: A cross-sectional analysis. Neuromuscul Disord 2017; 28:16-23. [PMID: 29174526 DOI: 10.1016/j.nmd.2017.10.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 09/08/2017] [Accepted: 10/17/2017] [Indexed: 12/25/2022]
Abstract
The development of new therapeutic agents for the treatment of Duchenne muscular dystrophy has put a focus on defining outcome measures most sensitive to capture treatment effects. This cross-sectional analysis investigates the relation between validated clinical assessments such as the 6-minute walk test, motor function measure and quantitative muscle MRI of thigh muscles in ambulant Duchenne muscular dystrophy patients, aged 6.5 to 10.8 years (mean 8.2, SD 1.1). Quantitative muscle MRI included the mean fat fraction using a 2-point Dixon technique, and transverse relaxation time (T2) measurements. All clinical assessments were highly significantly inter-correlated with p < 0.001. The strongest correlation with the motor function measure and its D1-subscore was shown by the 6-minute walk test. Clinical assessments showed no correlation with age. Importantly, quantitative muscle MRI values significantly correlated with all clinical assessments with the extensors showing the strongest correlation. In contrast to the clinical assessments, quantitative muscle MRI values were highly significantly correlated with age. In conclusion, the motor function measure and timed function tests measure disease severity in a highly comparable fashion and all tests correlated with quantitative muscle MRI values quantifying fatty muscle degeneration.
Collapse
|
64
|
Wilson K, Faelan C, Patterson-Kane JC, Rudmann DG, Moore SA, Frank D, Charleston J, Tinsley J, Young GD, Milici AJ. Duchenne and Becker Muscular Dystrophies: A Review of Animal Models, Clinical End Points, and Biomarker Quantification. Toxicol Pathol 2017; 45:961-976. [PMID: 28974147 DOI: 10.1177/0192623317734823] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) are neuromuscular disorders that primarily affect boys due to an X-linked mutation in the DMD gene, resulting in reduced to near absence of dystrophin or expression of truncated forms of dystrophin. Some newer therapeutic interventions aim to increase sarcolemmal dystrophin expression, and accurate dystrophin quantification is critical for demonstrating pharmacodynamic relationships in preclinical studies and clinical trials. Current challenges with measuring dystrophin include the variation in protein expression within individual muscle fibers and across whole muscle samples, the presence of preexisting dystrophin-positive revertant fibers, and trace amounts of residual dystrophin. Immunofluorescence quantification of dystrophin can overcome many of these challenges, but manual quantification of protein expression may be complicated by variations in the collection of images, reproducible scoring of fluorescent intensity, and bias introduced by manual scoring of typically only a few high-power fields. This review highlights the pathology of DMD and BMD, discusses animal models of DMD and BMD, and describes dystrophin biomarker quantitation in DMD and BMD, with several image analysis approaches, including a new automated method that evaluates protein expression of individual muscle fibers.
Collapse
Affiliation(s)
- Kristin Wilson
- 1 Flagship Biosciences, Inc., Westminster, Colorado, USA
| | - Crystal Faelan
- 1 Flagship Biosciences, Inc., Westminster, Colorado, USA
| | | | | | - Steven A Moore
- 2 Department of Pathology, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, USA
| | - Diane Frank
- 3 Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Jay Charleston
- 3 Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA
| | - Jon Tinsley
- 4 Summit Therapeutics, Abingdon, United Kingdom
| | - G David Young
- 1 Flagship Biosciences, Inc., Westminster, Colorado, USA
| | | |
Collapse
|
65
|
|
66
|
Hamuro L, Chan P, Tirucherai G, AbuTarif M. Developing a Natural History Progression Model for Duchenne Muscular Dystrophy Using the Six-Minute Walk Test. CPT Pharmacometrics Syst Pharmacol 2017; 6:596-603. [PMID: 28643370 PMCID: PMC5613187 DOI: 10.1002/psp4.12220] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/06/2017] [Accepted: 06/08/2017] [Indexed: 01/17/2023] Open
Abstract
The 6-minute walk test (6MWT) is used as a clinical endpoint to evaluate drug efficacy in Duchenne Muscular Dystrophy (DMD) trials. A model was developed using digitized 6MWT data that estimated two slopes and two intercepts to characterize 6MWT improvement during development and 6MWT decline. Mean baseline 6MWT was 362 (±87) meters. The model predicted an improvement at a rate of 20 meters/year (95% confidence interval (CI) = 9.4-30) up until 10 years old (95% CI = 6.78-13.1), and then a decline at a rate of 85 meters/year (95% CI = 72-98). Interpatient slope variability for improvement and decline were similar at 21.9 percentage of coefficient of variation (%CV) and 23.3%CV, respectively. Model simulations using age demographics from a previous DMD natural history study could reasonably predict the trend in improvement and decline in the 6MWT. This model can be used to quantitate individual patient trajectories, identify prognostic factors for disease progression, and evaluate drug effect.
Collapse
Affiliation(s)
- Lora Hamuro
- Department of Clinical Pharmacology and PharmacometricsBristol‐Myers SquibbPrincetonNew JerseyUSA
| | - Phyllis Chan
- Department of Clinical Pharmacology and PharmacometricsBristol‐Myers SquibbPrincetonNew JerseyUSA
| | - Giridhar Tirucherai
- Department of Clinical Pharmacology and PharmacometricsBristol‐Myers SquibbPrincetonNew JerseyUSA
| | - Malaz AbuTarif
- Department of Clinical Pharmacology and PharmacometricsBristol‐Myers SquibbPrincetonNew JerseyUSA
| |
Collapse
|
67
|
Reliability and validity analyses of the North Star Ambulatory Assessment in Brazilian Portuguese. Neuromuscul Disord 2017. [DOI: 10.1016/j.nmd.2017.05.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
68
|
Burakiewicz J, Sinclair CDJ, Fischer D, Walter GA, Kan HE, Hollingsworth KG. Quantifying fat replacement of muscle by quantitative MRI in muscular dystrophy. J Neurol 2017; 264:2053-2067. [PMID: 28669118 PMCID: PMC5617883 DOI: 10.1007/s00415-017-8547-3] [Citation(s) in RCA: 154] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/12/2017] [Accepted: 06/13/2017] [Indexed: 12/15/2022]
Abstract
The muscular dystrophies are rare orphan diseases, characterized by progressive muscle weakness: the most common and well known is Duchenne muscular dystrophy which affects young boys and progresses quickly during childhood. However, over 70 distinct variants have been identified to date, with different rates of progression, implications for morbidity, mortality, and quality of life. There are presently no curative therapies for these diseases, but a range of potential therapies are presently reaching the stage of multi-centre, multi-national first-in-man clinical trials. There is a need for sensitive, objective end-points to assess the efficacy of the proposed therapies. Present clinical measurements are often too dependent on patient effort or motivation, and lack sensitivity to small changes, or are invasive. Quantitative MRI to measure the fat replacement of skeletal muscle by either chemical shift imaging methods (Dixon or IDEAL) or spectroscopy has been demonstrated to provide such a sensitive, objective end-point in a number of studies. This review considers the importance of the outcome measures, discusses the considerations required to make robust measurements and appropriate quality assurance measures, and draws together the existing literature for cross-sectional and longitudinal cohort studies using these methods in muscular dystrophy.
Collapse
Affiliation(s)
- Jedrzej Burakiewicz
- Department of Radiology, C. J. Gorter Center for High Field MRI, Leiden University Medical Centre, Leiden, The Netherlands
| | - Christopher D J Sinclair
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, London, UK.,Neuroradiological Academic Unit, UCL Institute of Neurology, London, UK
| | - Dirk Fischer
- Division of Neuropaediatrics, University of Basel Children's Hospital, Spitalstrasse 33, Postfach, Basel, 4031, Switzerland.,Department of Neurology, University of Basel Hospital, Petersgraben 4, Basel, 4031, Switzerland
| | - Glenn A Walter
- Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, 32610, USA
| | - Hermien E Kan
- Department of Radiology, C. J. Gorter Center for High Field MRI, Leiden University Medical Centre, Leiden, The Netherlands
| | - Kieren G Hollingsworth
- Newcastle Magnetic Resonance Centre, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, UK.
| |
Collapse
|
69
|
Nunes MF, Hukuda ME, Favero FM, Oliveira AB, Voos MC, Caromano FA. Relationship between muscle strength and motor function in Duchenne muscular dystrophy. ARQUIVOS DE NEURO-PSIQUIATRIA 2017; 74:530-5. [PMID: 27487372 DOI: 10.1590/0004-282x20160085] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 05/05/2016] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To investigate the relationship between muscle strength and motor function and between these variables and age. METHOD Muscle strength was measured by Medical Research Council (MRC) scale and motor function, by Motor Function Measure (MFM), in 40 non-ambulatory patients. Spearman tests investigated the relationships between muscle strength, motor function and age. RESULTS Total MRC and MFM scores were strongly related to each other (r = 0.94; p < 0.001), but not to age (r = -0.19, r = -0.31, respectively; p > 0.05). Strong and moderate relationships between partial muscle strength and motor function scores were found. Higher correlation coefficients were found between total scores and Dimensions 2 (axial/ proximal control) and 3 (distal control) of MFM. CONCLUSION Muscle strength and motor function are strongly correlated and seem to decrease proportionally in DMD.
Collapse
Affiliation(s)
- Milene F Nunes
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Fisioterapia, Fonoaudiologia e Terapia Ocupacional, São Paulo SP, Brasil
| | - Michele E Hukuda
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Fisioterapia, Fonoaudiologia e Terapia Ocupacional, São Paulo SP, Brasil
| | - Francis M Favero
- Universidade Federal de São Paulo, Faculdade de Medicina, Departamento de Neurologia, São Paulo SP, Brasil
| | - Acary B Oliveira
- Universidade Federal de São Paulo, Faculdade de Medicina, Departamento de Neurologia, São Paulo SP, Brasil
| | - Mariana C Voos
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Fisioterapia, Fonoaudiologia e Terapia Ocupacional, São Paulo SP, Brasil
| | - Fátima A Caromano
- Universidade de São Paulo, Faculdade de Medicina, Departamento de Fisioterapia, Fonoaudiologia e Terapia Ocupacional, São Paulo SP, Brasil
| |
Collapse
|
70
|
Verma S, Lin J, Travers C, McCracken C, Shah D. Quantitative electromyography in ambulatory boys with Duchenne muscular dystrophy. Muscle Nerve 2017; 56:1168-1171. [PMID: 28457006 DOI: 10.1002/mus.25678] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 04/06/2017] [Accepted: 04/24/2017] [Indexed: 11/07/2022]
Abstract
INTRODUCTION This study's objective was to evaluate quantitative electromyography (QEMG) using multiple-motor-unit (multi-MUP) analysis in Duchenne muscular dystrophy (DMD). METHODS Ambulatory DMD boys, aged 5-15 years, were evaluated with QEMG at 6-month intervals over 14 months. EMG was performed in the right biceps brachii (BB) and tibialis anterior (TA) muscles. Normative QEMG data were obtained from age-matched healthy boys. Wilcoxon signed-rank tests were performed. RESULTS Eighteen DMD subjects were enrolled, with a median age of 7 (interquartile range 7-10) years. Six-month evaluations were performed on 14 subjects. QEMG showed significantly abnormal mean MUP duration in BB and TA muscles, with no significant change over 6 months. CONCLUSIONS QEMG is a sensitive electrophysiological marker of myopathy in DMD. Preliminary data do not reflect a significant change in MUP parameters over a 6-month interval; long-term follow-up QEMG studies are needed to understand its role as a biomarker for disease progression. Muscle Nerve 56: 1361-1364, 2017.
Collapse
Affiliation(s)
- Sumit Verma
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, 30307, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA.,Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Jenny Lin
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, 30307, USA.,Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA.,Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | - Curtis Travers
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, 30307, USA
| | - Courtney McCracken
- Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, 30307, USA
| | - Durga Shah
- Department of Rehabilitation, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
71
|
Zaidman CM, Wu JS, Kapur K, Pasternak A, Madabusi L, Yim S, Pacheck A, Szelag H, Harrington T, Darras BT, Rutkove SB. Quantitative muscle ultrasound detects disease progression in Duchenne muscular dystrophy. Ann Neurol 2017; 81:633-640. [PMID: 28241384 DOI: 10.1002/ana.24904] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 02/21/2017] [Accepted: 02/22/2017] [Indexed: 01/19/2023]
Abstract
OBJECTIVE We assessed changes in quantitative muscle ultrasound data in boys with Duchenne muscular dystrophy (DMD) and healthy controls to determine whether ultrasound can serve as a biomarker of disease progression. Two approaches were used: gray scale level (GSL), measured from the ultrasound image, and quantitative backscatter analysis (QBA), measured directly from the received echoes. METHODS GSL and QBA were obtained from 6 unilateral arm/leg muscles in 36 boys with DMD and 28 healthy boys (age = 2-14 years) for up to 2 years. We used a linear mixed effects model with random intercept and slope terms to compare trajectories of GSL, QBA, and functional assessments. We analyzed separately a subset of boys who initiated corticosteroids. RESULTS Compared to healthy boys, increasing GSL in DMD boys >7.0 years old was first identified at 6 months (eg, anterior forearm slope difference of 1.16 arbitrary units/mo, p = 0.004, 95% confidence interval [CI] = 0.38-1.94); in boys ≤ 7 years old, differences in GSL first appeared at 12 months (0.82 arbitrary units/mo, p = 0.04, 95% CI = 0.075-1.565, in rectus femoris). QBA performed similarly to GSL (eg, DMD boys > 7 years old: 0.41dB/mo, p = 0.01, 95% CI = 0.096-0.72, in anterior forearm at 6 months). Ultrasound identified differences earlier than functional measures including 6-minute walk and supine-to-stand tests. However, neither QBA nor GSL showed an effect of corticosteroid initiation. INTERPRETATION QBA performs similarly to GSL, and both appear more sensitive than functional assessments for detecting muscle deterioration in DMD. Additional studies will be required to determine whether quantitative muscle ultrasound can detect therapeutic efficacy. Ann Neurol 2017;81:633-640.
Collapse
Affiliation(s)
- Craig M Zaidman
- Department of Neurology, Washington University in St Louis, St Louis, MO
| | - Jim S Wu
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Kush Kapur
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Amy Pasternak
- Department of Physical Therapy Services and Occupational Therapy Services, Boston Children's Hospital, Boston, MA
| | - Lavanya Madabusi
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Sung Yim
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA
| | - Adam Pacheck
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA
| | - Heather Szelag
- Department of Neurology, Washington University in St Louis, St Louis, MO
| | - Tim Harrington
- Department of Neurology, Washington University in St Louis, St Louis, MO
| | - Basil T Darras
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Seward B Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center, Boston, MA
| |
Collapse
|
72
|
Rutkove SB, Kapur K, Zaidman CM, Wu JS, Pasternak A, Madabusi L, Yim S, Pacheck A, Szelag H, Harrington T, Darras BT. Electrical impedance myography for assessment of Duchenne muscular dystrophy. Ann Neurol 2017; 81:622-632. [PMID: 28076894 DOI: 10.1002/ana.24874] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 12/24/2016] [Accepted: 12/26/2016] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Sensitive, objective, and easily applied methods for evaluating disease progression and response to therapy are needed for clinical trials in Duchenne muscular dystrophy (DMD). In this study, we evaluated whether electrical impedance myography (EIM) could serve this purpose. METHODS In this nonblinded study, 36 boys with DMD and 29 age-similar healthy boys underwent multifrequency EIM measurements for up to 2 years on 6 muscles unilaterally along with functional assessments. A linear mixed-effects model with random intercept and slope terms was used for the analysis of multifrequency EIM values and functional measures. Seven DMD boys were initiated on corticosteroids; these data were analyzed using a piecewise linear mixed-effects model. RESULTS In boys > 7.0 years old, a significant difference in the slope of EIM phase ratio trajectories in the upper extremity was observed by 6 months of -0.074/month, p = 0.023, 95% confidence interval (CI) = -0.013, -0.14; at 2 years, this difference was -0.048/month, p < 0.0001, 95% CI = -0.028, -0.068. In boys ≤ 7.0 years old, differences appeared at 6 months in gastrocnemius (EIM phase slope = -0.83 °/kHz/mo, p = 0.007, 95% CI = -0.26, -1.40). EIM outcomes showed significant differences earlier than functional tests. Initiation of corticosteroids significantly improved the slope of EIM phase ratio (0.057/mo, p = 0.00019, 95% CI = 0.028, 0.086) and EIM phase slope (0.14 °/kHz/mo, p = 0.013, 95% CI = 0.028, 0.25), consistent with corticosteroids' known clinical benefit. INTERPRETATION EIM detects deterioration in muscles of both younger and older boys by 6 months; it also identifies the therapeutic effect of corticosteroid initiation. Because EIM is rapid to apply, painless, and requires minimal operator training, the technique deserves to be further evaluated as a biomarker in DMD clinical therapeutic trials. Ann Neurol 2017;81:622-632.
Collapse
Affiliation(s)
- Seward B Rutkove
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Kush Kapur
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Craig M Zaidman
- Department of Neurology, Washington University in St Louis, St Louis, MO
| | - Jim S Wu
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Amy Pasternak
- Department of Physical Therapy Services and Occupational Therapy Services, Boston Children's Hospital, Boston, MA
| | - Lavanya Madabusi
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Sung Yim
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Adam Pacheck
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA
| | - Heather Szelag
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Tim Harrington
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Basil T Darras
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
73
|
Alfano LN, Miller NF, Berry KM, Yin H, Rolf KE, Flanigan KM, Mendell JR, Lowes LP. The 100-meter timed test: Normative data in healthy males and comparative pilot outcome data for use in Duchenne muscular dystrophy clinical trials. Neuromuscul Disord 2017; 27:452-457. [DOI: 10.1016/j.nmd.2017.02.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 01/12/2017] [Accepted: 02/15/2017] [Indexed: 11/27/2022]
|
74
|
Ryder S, Leadley RM, Armstrong N, Westwood M, de Kock S, Butt T, Jain M, Kleijnen J. The burden, epidemiology, costs and treatment for Duchenne muscular dystrophy: an evidence review. Orphanet J Rare Dis 2017; 12:79. [PMID: 28446219 PMCID: PMC5405509 DOI: 10.1186/s13023-017-0631-3] [Citation(s) in RCA: 337] [Impact Index Per Article: 42.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 04/12/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Duchenne Muscular Dystrophy (DMD) is a rapidly progressive, lethal neuromuscular disorder, present from birth, which occurs almost exclusively in males. We have reviewed contemporary evidence of burden, epidemiology, illness costs and treatment patterns of DMD. This systematic review adhered to published methods with information also sought from the web and contacting registries. Searches were carried out from 2005 to June 2015. The population of interest was individuals with clearly defined DMD or their carers. RESULTS Nine thousand eight hundred fifty titles were retrieved from searches. Fifty-eight studies were reviewed with three assessed as high, 33 as medium and 22 as low quality. We found two studies reporting birth and four reporting point prevalence, three reporting mortality, 41 reporting severity and/or progression, 18 reporting treatment patterns, 12 reporting quality of life, two reporting utility measures, three reporting costs of illness and three treatment guidelines. Birth prevalence ranged from 15.9 to 19.5 per 100,000 live births. Point prevalence per 100,000 males was for France, USA, UK and Canada, 10.9, 1.9, 2.2 and 6.1 respectively. A study of adult DMD patients at a centre in France found median survival for those born between 1970 and 1994 was 40.95 years compared to 25.77 years for those born between 1955 and 1969. Loss of ambulation occurred at a median age of 12 and ventilation starts at about 20 years. There was international variation in use of corticosteroids, scoliosis surgery, ventilation and physiotherapy. The economic cost of DMD climbs dramatically with disease progression - rising as much as 5.7 fold from the early ambulatory phase to the non-ambulatory phase in Germany. CONCLUSIONS This is the first systematic review of treatment, progression, severity and quality of life in DMD. It also provides the most recent description of the burden, epidemiology, illness costs and treatment patterns in DMD. There are evidence gaps, particularly in prevalence and mortality. People with DMD seem to be living longer, possibly due to corticosteroid use, cardiac medical management and ventilation. Future research should incorporate registry data to improve comparability across time and between countries and to investigate the quality of life impact as the condition progresses.
Collapse
Affiliation(s)
- S Ryder
- Kleijnen Systematic Reviews Ltd., Unit 6, Escrick Business Park, Riccall Road, Escrick, York, YO19 6FD, UK.
| | - R M Leadley
- Kleijnen Systematic Reviews Ltd., Unit 6, Escrick Business Park, Riccall Road, Escrick, York, YO19 6FD, UK
| | - N Armstrong
- Kleijnen Systematic Reviews Ltd., Unit 6, Escrick Business Park, Riccall Road, Escrick, York, YO19 6FD, UK
| | - M Westwood
- Kleijnen Systematic Reviews Ltd., Unit 6, Escrick Business Park, Riccall Road, Escrick, York, YO19 6FD, UK
| | - S de Kock
- Kleijnen Systematic Reviews Ltd., Unit 6, Escrick Business Park, Riccall Road, Escrick, York, YO19 6FD, UK
| | - T Butt
- BioMarin Europe Ltd., 164 Shaftesbury Ave, London, WC2H 8HL, UK
| | - M Jain
- BioMarin Europe Ltd., 164 Shaftesbury Ave, London, WC2H 8HL, UK
| | - J Kleijnen
- School for Public Health and Primary Care, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
75
|
Wong BL, Rybalsky I, Shellenbarger KC, Tian C, McMahon MA, Rutter MM, Sawnani H, Jefferies JL. Long-Term Outcome of Interdisciplinary Management of Patients with Duchenne Muscular Dystrophy Receiving Daily Glucocorticoid Treatment. J Pediatr 2017; 182:296-303.e1. [PMID: 28043681 DOI: 10.1016/j.jpeds.2016.11.078] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 10/12/2016] [Accepted: 11/29/2016] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To evaluate clinical outcomes and steroid side effects in a cohort of patients with Duchenne muscular dystrophy (DMD) treated with long-term daily glucocorticoid therapy. Although daily glucocorticoid therapy has been shown to extend ambulatory function in DMD, less frequent dosing is often used because of side effect concerns. STUDY DESIGN Retrospective study of 97 patients with DMD aged 10 to <16 years treated with daily glucocorticoid (89% on deflazacort) for a mean of 8.5 years. Outcome measures were motor, pulmonary, and cardiac function, and scoliosis. Side effects were growth failure and weight gain, facial fullness, blood pressure, bone health, cataracts, gastrointestinal symptoms, behavior, hypertrichosis, and need for medication interventions. RESULTS For 13- to 16-year-old patients, 40% could rise from the floor and 50% could perform the 30-foot run test. Forced vital capacity for the entire cohort was well preserved. Thirteen percent of younger (10- to <13-year-old) and 21% of older patients had findings of left ventricle systolic dysfunction. Six percent (all aged 16 years) developed scoliosis (Cobb angle >20 degrees). Eighty-six percent had normal weight velocities; 30% had no increased facial fullness; 72% had short stature; and 19% had asymptomatic cataracts. Asymptomatic spine compression deformities were noted in 76% and long bone fractures in 30%. One patient stopped glucocorticoid because of behavioral concerns. CONCLUSIONS With evidence for improved outcomes and manageable side effects, we recommend use of daily glucocorticoid therapy for patients with DMD with anticipatory management of side effects and a coordinated interdisciplinary care approach.
Collapse
Affiliation(s)
- Brenda L Wong
- Comprehensive Neuromuscular Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH.
| | - Irina Rybalsky
- Comprehensive Neuromuscular Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Karen C Shellenbarger
- Comprehensive Neuromuscular Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Cuixia Tian
- Comprehensive Neuromuscular Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Mary A McMahon
- Comprehensive Neuromuscular Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Division of Physical Medicine and Rehabilitation, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Meilan M Rutter
- Comprehensive Neuromuscular Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Division of Endocrinology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Hemant Sawnani
- Comprehensive Neuromuscular Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; Division of Pulmonary Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - John L Jefferies
- Comprehensive Neuromuscular Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH; The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
76
|
Bell JM, Shields MD, Watters J, Hamilton A, Beringer T, Elliott M, Quinlivan R, Tirupathi S, Blackwood B, Cochrane Neuromuscular Group. Interventions to prevent and treat corticosteroid-induced osteoporosis and prevent osteoporotic fractures in Duchenne muscular dystrophy. Cochrane Database Syst Rev 2017; 1:CD010899. [PMID: 28117876 PMCID: PMC6464928 DOI: 10.1002/14651858.cd010899.pub2] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Corticosteroid treatment is considered the 'gold standard' for Duchenne muscular dystrophy (DMD); however, it is also known to induce osteoporosis and thus increase the risk of vertebral fragility fractures. Good practice in the care of those with DMD requires prevention of these adverse effects. Treatments to increase bone mineral density include bisphosphonates and vitamin D and calcium supplements, and in adolescents with pubertal delay, testosterone. Bone health management is an important part of lifelong care for patients with DMD. OBJECTIVES To assess the effects of interventions to prevent or treat osteoporosis in children and adults with DMD taking long-term corticosteroids; to assess the effects of these interventions on the frequency of vertebral fragility fractures and long-bone fractures, and on quality of life; and to assess adverse events. SEARCH METHODS On 12 September 2016, we searched the Cochrane Neuromuscular Specialised Register, CENTRAL, MEDLINE, Embase, and CINAHL Plus to identify potentially eligible trials. We also searched the Web of Science ISI Proceedings (2001 to September 2016) and three clinical trials registries to identify unpublished studies and ongoing trials. We contacted correspondence authors of the included studies in the review to obtain information on unpublished studies or work in progress. SELECTION CRITERIA We considered for inclusion in the review randomised controlled trials (RCTs) and quasi-RCTs involving any bone health intervention for corticosteroid-induced osteoporosis and fragility fractures in children, adolescents, and adults with a confirmed diagnosis of DMD. The interventions might have included oral and intravenous bisphosphonates, vitamin D supplements, calcium supplements, dietary calcium, testosterone, and weight-bearing activity. DATA COLLECTION AND ANALYSIS Two review authors independently assessed reports and selected potential studies for inclusion, following standard Cochrane methodology. We contacted study authors to obtain further information for clarification on published work, unpublished studies, and work in progress. MAIN RESULTS We identified 18 potential studies, of which two, currently reported only as abstracts, met the inclusion criteria for this review. Too little information was available for us to present full results or adequately assess risk of bias. The participants were children aged five to 15 years with DMD, ambulant and non-ambulant. The interventions were risedronate versus no treatment in one trial (13 participants) and whole-body vibration versus a placebo device in the second (21 participants). Both studies reported improved bone mineral density with the active treatments, with no improvement in the control groups, but the abstracts did not compare treatment and control conditions. All children tolerated whole-body vibration treatment. No study provided information on adverse events. Two studies are ongoing: one investigating whole-body vibration, the other investigating zoledronic acid. AUTHORS' CONCLUSIONS We know of no high-quality evidence from RCTs to guide use of treatments to prevent or treat corticosteroid-induced osteoporosis and reduce the risk of fragility fractures in children and adults with DMD; only limited results from two trials reported in abstracts were available. We await formal trial reports. Findings from two ongoing relevant studies and two trials, for which only abstracts are available, will be important in future updates of this review.
Collapse
Affiliation(s)
- Jennifer M Bell
- Queen's University BelfastCentre for Experimental Medicine, School of Medicine, Dentistry and Biomedical SciencesRoom 02.041, 2nd FloorMulhouse, Grosvenor RoadBelfastNorthern IrelandUKBT12 6BJ
| | - Michael D Shields
- Queen's University BelfastCentre for Experimental Medicine, School of Medicine, Dentistry and Biomedical SciencesRoom 02.041, 2nd FloorMulhouse, Grosvenor RoadBelfastNorthern IrelandUKBT12 6BJ
| | - Janet Watters
- Belfast Health and Social Care TrustGP Out of Hours ServiceBelfastNorthern IrelandUK
| | - Alistair Hamilton
- Belfast Health and Social Care TrustWithers Orthopaedic CentreMusgrave Park Hospital, Royal Group of Hospitals,Stockman's LaneBelfastNorthern IrelandUK
| | - Timothy Beringer
- Belfast Health and Social Care TrustDepartment of Care for the ElderyFlorence Elliot CentreRoyal Victoria HospitalBelfastNorthern IrelandUKBT12 6BA
| | - Mark Elliott
- Musgrave Park Hospital, Belfast Health and Social Care TrustBelfastUK
| | - Rosaline Quinlivan
- UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery and Great Ormond StreetMRC Centre for Neuromuscular Diseases and Dubowitz Neuromuscular CentrePO Box 114LondonUKWC1B 3BN
| | - Sandya Tirupathi
- Royal Belfast Hospital for Sick ChildrenPaediatric Neurology180 Falls RoadBelfastUKBT12 6BE
| | - Bronagh Blackwood
- Queen's University BelfastCentre for Experimental Medicine, School of Medicine, Dentistry and Biomedical SciencesRoom 02.041, 2nd FloorMulhouse, Grosvenor RoadBelfastNorthern IrelandUKBT12 6BJ
| | | |
Collapse
|
77
|
Goemans N, Tulinius M, Kroksmark AK, Wilson R, van den Hauwe M, Campion G. Comparison of ambulatory capacity and disease progression of Duchenne muscular dystrophy subjects enrolled in the drisapersen DMD114673 study with a matched natural history cohort of subjects on daily corticosteroids. Neuromuscul Disord 2016; 27:203-213. [PMID: 28169120 DOI: 10.1016/j.nmd.2016.11.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 11/17/2016] [Accepted: 11/21/2016] [Indexed: 01/16/2023]
Abstract
Duchenne muscular dystrophy is a rare genetic disorder with life-limiting pathology. Drisapersen induces exon 51 skipping, thereby producing a shorter but functional dystrophin protein. The longest available data are from an open-label extension study (PRO051-02) treating 12 boys with drisapersen (6 mg/kg/week subcutaneously). The median change (range) from baseline to week 177 in six-minute walking distance (6MWD) was 8 (-263, 163) metres. The current analysis aimed to put the results from PRO051-02 in the context of natural progression by comparing the functional trajectory of drisapersen-treated subjects to a matched natural history (NH) cohort, treated by standard of care. Subjects were matched individually by age and 6MWD, as the primary analysis, and by age and rise from floor (RFF), as sensitivity analysis. A total of 75 NH subjects were available for 6MWD analysis, of which matching was possible for 9 ambulant drisapersen-treated subjects. None of the 6 "stable" (baseline 6MWD ≥330 metres) drisapersen-treated subjects lost ambulation vs 4 out of 10 matched NH subjects over a comparable timeframe (~3.4 years), compared with 2 out of 3 ambulant "in decline" drisapersen-treated subjects vs all 6 matched NH subjects. A total of 79 NH subjects were available for RFF analysis. For continuous ambulatory subjects (N = 4), the RFF decline was more pronounced in the NH cohort than in the drisapersen-treated subjects. In conclusion, a comparison of ambulant drisapersen-treated subjects with matched NH subjects showed a difference in functional trajectories over a timeframe of up to 3.4 years in favour of drisapersen.
Collapse
Affiliation(s)
- Nathalie Goemans
- Department of Paediatrics and Child Neurology, University Hospitals Leuven, Leuven, Belgium.
| | - Mar Tulinius
- The Queen Silvia Children's Hospital, Gothenburg, Sweden
| | | | | | - Marleen van den Hauwe
- Department of Paediatrics and Child Neurology, University Hospitals Leuven, Leuven, Belgium
| | | |
Collapse
|
78
|
Mylius CF, Paap D, Takken T. Reference value for the 6-minute walk test in children and adolescents: a systematic review. Expert Rev Respir Med 2016; 10:1335-1352. [DOI: 10.1080/17476348.2016.1258305] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
79
|
Goemans NM, Tulinius M, van den Hauwe M, Kroksmark AK, Buyse G, Wilson RJ, van Deutekom JC, de Kimpe SJ, Lourbakos A, Campion G. Long-Term Efficacy, Safety, and Pharmacokinetics of Drisapersen in Duchenne Muscular Dystrophy: Results from an Open-Label Extension Study. PLoS One 2016; 11:e0161955. [PMID: 27588424 PMCID: PMC5010191 DOI: 10.1371/journal.pone.0161955] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 08/13/2016] [Indexed: 01/16/2023] Open
Abstract
Background Drisapersen induces exon 51 skipping during dystrophin pre-mRNA splicing and allows synthesis of partially functional dystrophin in Duchenne muscular dystrophy (DMD) patients with amenable mutations. Methods This 188-week open-label extension of the dose-escalation study assessed the long-term efficacy, safety, and pharmacokinetics of drisapersen (PRO051/GSK2402968), 6 mg/kg subcutaneously, in 12 DMD subjects. Dosing was once weekly for 72 weeks. All subjects had a planned treatment interruption (weeks 73–80), followed by intermittent dosing (weeks 81–188). Results Subjects received a median (range) total dose of 5.93 (5.10 to 6.02) mg/kg drisapersen. After 177 weeks (last efficacy assessment), median (mean [SD]) six-minute walk distance (6MWD) improved by 8 (-24.5 [161]) meters for the 10 subjects able to complete the 6MWD at baseline (mean age [SD]: 9.5 [1.9] years). These statistics include 2 subjects unable to complete the test at later visits and who scored “zero”. When only the 8 ambulant subjects at week 177 were taken into account, a median (mean [SD]) increase of 64 (33 [121]) meters in 6MWD was observed. Of 7 subjects walking ≥330 m at extension baseline, 5 walked farther at week 177. Of 3 subjects walking <330 m, 2 lost ambulation, while 1 declined overall but walked farther at some visits. Over the 188 weeks, the most common adverse events were injection-site reactions, raised urinary α1-microglobulin and proteinuria. Dystrophin expression was detected in all muscle biopsies obtained at week 68 or 72. Conclusion Drisapersen was generally well tolerated over 188 weeks. Possible renal effects, thrombocytopenia and injection-site reactions warrant continued monitoring. Improvements in the 6MWD at 12 weeks were sustained after 3.4 years of dosing for most patients. For a small, uncontrolled study, the outcomes are encouraging, as natural history studies would anticipate a decline of over 100 meters over a 3-year period in a comparable cohort. Trial Registration ClinicalTrials.gov NCT01910649
Collapse
|
80
|
Bello L, Campadello P, Barp A, Fanin M, Semplicini C, Sorarù G, Caumo L, Calore C, Angelini C, Pegoraro E. Functional changes in Becker muscular dystrophy: implications for clinical trials in dystrophinopathies. Sci Rep 2016; 6:32439. [PMID: 27582364 PMCID: PMC5007528 DOI: 10.1038/srep32439] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 08/05/2016] [Indexed: 01/16/2023] Open
Abstract
We performed a 1-year longitudinal study of Six Minute Walk Test (6MWT), North Star Ambulatory Assessment (NSAA), and timed function tests in Becker muscular dystrophy (BMD). Skeletal muscle dystrophin was quantified by immunoblot. We grouped deletions ending on exon 45 ("del 45-x", n = 28) or 51 ("del x-51", n = 10); isolated exon 48 deletion ("del 48", n = 10); and other mutations (n = 21). Only patients in the "del 45-x" or "other" groups became non-ambulatory (n = 5, log-rank p = n.s.) or unable to run (n = 22, p < 0.001). All measures correlated positively with dystrophin quantity and negatively with age, and were significantly more impaired in the "del 45-x" and "other" groups. After one year, NSAA score decreased significantly (-0.9 ± 1.6, p < 0.001); in the "del 45-x" group, both NSAA (-1.3 ± 1.7, p = 0.001) and 6MWT (-12 ± 31 m, p = 0.059) decreased. We conclude that patients with "del x-51" or "del 48" mutations have mild or asymptomatic BMD, while "del 45-x" mutations cause comparatively severe weakness, and functional deterioration in 1 year. Furthermore, exon 51 skipping could be more effective than exon 45 skipping in Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Luca Bello
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Paola Campadello
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Andrea Barp
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Marina Fanin
- Department of Neurosciences, University of Padova, Padova, Italy
| | | | - Gianni Sorarù
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Luca Caumo
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Chiara Calore
- Department of Cardiac, Thoracic and Vascular Sciences, Cardiology Clinic, University of Padova, Padova, Italy
| | - Corrado Angelini
- Department of Neurosciences, University of Padova, Padova, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padova, Padova, Italy
| |
Collapse
|
81
|
Mercuri E, Signorovitch JE, Swallow E, Song J, Ward SJ. Categorizing natural history trajectories of ambulatory function measured by the 6-minute walk distance in patients with Duchenne muscular dystrophy. Neuromuscul Disord 2016; 26:576-83. [PMID: 27423700 PMCID: PMC5026045 DOI: 10.1016/j.nmd.2016.05.016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Revised: 05/16/2016] [Accepted: 05/24/2016] [Indexed: 11/22/2022]
Abstract
High variability in patients' changes in 6 minute walk distance (6MWD) over time has complicated clinical trials of treatment efficacy in Duchenne muscular dystrophy (DMD). We assessed whether boys with DMD could be grouped into classes that shared similar ambulatory function trajectories as measured by 6MWD. Ambulatory boys aged 5 years or older with genetically confirmed DMD who were enrolled in a natural history study at 11 care centers throughout Italy were included. For each boy, standardized assessments of 6MWD were available at annual intervals spanning 3 years. Trajectories of 6MWD vs. age and trajectories of 6MWD vs. time from enrollment were examined using latent class analysis. A total of 96 boys were included. At enrollment, the mean age was 8.3 years (mean 6MWD: 374 meters). After accounting for age, baseline 6MWD, and steroid use, four latent trajectory classes were identified as explaining 3-year 6MWD outcomes significantly better than a single average trajectory. Patient trajectories of 6MWD change from enrollment were categorized as having fast decline (n = 25), moderate decline (n = 19), stable function (n = 37), and improving function (n = 15) during the 3-year follow-up. After accounting for trajectory classes, the standard deviation of variation in 6MWD was reduced by approximately 40%. The natural history of ambulatory function in DMD may be composed of distinct trajectory classes. The extent to which trajectories are associated with novel and established prognostic factors warrants further study. Reducing unexplained variation in patient outcomes could help to further improve DMD clinical trial design and analysis.
Collapse
Affiliation(s)
- Eugenio Mercuri
- Paediatric Neurology Unit, Catholic University, Rome, Italy.
| | - James Edward Signorovitch
- Analysis Group, Inc., 111 Huntington Ave, 10th Floor, Boston, MA, USA; The TAP Collaboration, One Broadway, 14th Floor, Cambridge, MA, USA
| | - Elyse Swallow
- Analysis Group, Inc., 111 Huntington Ave, 10th Floor, Boston, MA, USA
| | - Jinlin Song
- Analysis Group, Inc., 111 Huntington Ave, 10th Floor, Boston, MA, USA
| | - Susan J Ward
- The TAP Collaboration, One Broadway, 14th Floor, Cambridge, MA, USA
| |
Collapse
|
82
|
Mercuri E, Coratti G, Messina S, Ricotti V, Baranello G, D’Amico A, Pera MC, Albamonte E, Sivo S, Mazzone ES, Arnoldi MT, Fanelli L, De Sanctis R, Romeo DM, Vita GL, Battini R, Bertini E, Muntoni F, Pane M. Revised North Star Ambulatory Assessment for Young Boys with Duchenne Muscular Dystrophy. PLoS One 2016; 11:e0160195. [PMID: 27494024 PMCID: PMC4975396 DOI: 10.1371/journal.pone.0160195] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/16/2016] [Indexed: 11/18/2022] Open
Abstract
The advent of therapeutic approaches for Duchenne muscular dystrophy (DMD) has highlighted the need to identify reliable outcome measures for young boys with DMD. The aim of this study was to develop a revised version of the North Star Ambulatory Assessment (NSAA) suitable for boys between the age of 3 and 5 years by identifying age appropriate items and revising the scoring system accordingly. Using the scale in 171 controls between the age of 2.9 and 4.8 years, we identified items that were appropriate at different age points. An item was defined as age appropriate if it was completed, achieving a full score, by at least 85% of the typically developing boys at that age. At 3 years (±3months) there were only 8 items that were age appropriate, at 3 years and 6 months there were 13 items while by the age of 4 years all 17 items were appropriate. A revised version of the scale was developed with items ordered according to the age when they could be reliably performed. The application of the revised version of the scale to data collected in young DMD boys showed that very few of the DMD boys were able to complete with a full score all the age appropriate items. In conclusion, our study suggests that a revised version of the NSAA can be used in boys from the age of 3 years to obtain information on how young DMD boys acquire new abilities and how this correlates with their peers.
Collapse
Affiliation(s)
- Eugenio Mercuri
- Department of Paediatric Neurology, Catholic University, and Nemo Roma center for neuromuscular disorders, Rome, Italy
| | - Giorgia Coratti
- Department of Paediatric Neurology, Catholic University, and Nemo Roma center for neuromuscular disorders, Rome, Italy
| | - Sonia Messina
- Department of Neurosciences and Nemo and Clinical Center, Psychiatry and Anaesthesiology, University of Messina, Messina, Italy
| | - Valeria Ricotti
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College, London, United Kingdom
| | - Giovanni Baranello
- Developmental Neurology Unit, Istituto Neurologico “Besta”, Milan, Italy
| | - Adele D’Amico
- Unit of Neuromuscular and Neurodegenerative Diseases, Department of Neurosciences, Bambino Gesù Children's Hospital, Rome, Italy
| | - Maria Carmela Pera
- Department of Paediatric Neurology, Catholic University, and Nemo Roma center for neuromuscular disorders, Rome, Italy
| | - Emilio Albamonte
- Department of Paediatric Neurology, Catholic University, and Nemo Roma center for neuromuscular disorders, Rome, Italy
| | - Serena Sivo
- Department of Paediatric Neurology, Catholic University, and Nemo Roma center for neuromuscular disorders, Rome, Italy
| | - Elena Stacy Mazzone
- Department of Paediatric Neurology, Catholic University, and Nemo Roma center for neuromuscular disorders, Rome, Italy
| | | | - Lavinia Fanelli
- Department of Paediatric Neurology, Catholic University, and Nemo Roma center for neuromuscular disorders, Rome, Italy
| | - Roberto De Sanctis
- Department of Paediatric Neurology, Catholic University, and Nemo Roma center for neuromuscular disorders, Rome, Italy
| | - Domenico M Romeo
- Department of Paediatric Neurology, Catholic University, and Nemo Roma center for neuromuscular disorders, Rome, Italy
| | - Gian Luca Vita
- Department of Neurosciences and Nemo and Clinical Center, Psychiatry and Anaesthesiology, University of Messina, Messina, Italy
| | - Roberta Battini
- Department of Developmental Neuroscience, Stella Maris Institute, Pisa, Italy
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Diseases, Department of Neurosciences, Bambino Gesù Children's Hospital, Rome, Italy
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, Institute of Child Health, University College, London, United Kingdom
| | - Marika Pane
- Department of Paediatric Neurology, Catholic University, and Nemo Roma center for neuromuscular disorders, Rome, Italy
| |
Collapse
|
83
|
Hafner P, Bonati U, Rubino D, Gocheva V, Zumbrunn T, Gueven N, Fischer D. Treatment with L-citrulline and metformin in Duchenne muscular dystrophy: study protocol for a single-centre, randomised, placebo-controlled trial. Trials 2016; 17:389. [PMID: 27488051 PMCID: PMC4973063 DOI: 10.1186/s13063-016-1503-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 07/17/2016] [Indexed: 12/19/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is an X-linked recessive disease that affects 1 in 3500–6000 male births. Despite broad research aiming to improve muscle function as well as heart and brain function, sufficient therapeutic efficacy has not yet been achieved and current therapeutic management is still supportive. In a recent pilot trial, oral treatment with l-arginine and metformin showed consistent changes of muscular metabolism both in vitro and in vivo by raising NO levels and expression of mitochondrial proteins in the skeletal muscle tissue of patients with DMD. This randomised, double-blind, placebo-controlled trial aims to demonstrate the superiority of l-citrulline and metformin therapy over placebo in DMD patients with regard to the Motor Function Measure (MFM) D1 subscore (primary endpoint) as well as additional clinical and subclinical tests. Methods/Design A total of 40–50 ambulant patients with DMD will be recruited at the outpatient department of the University of Basel Children’s Hospital (Switzerland), as well as from the DMD patient registries of Switzerland, Germany and Austria. Patients will be randomly allocated to one of the two arms of the study and will receive either a combination of l-citrulline and metformin or placebo for 26 weeks. Co-medication with glucocorticoids is allowed. The primary endpoint is the change of the MFM D1 subscore from baseline to week 26 under l-citrulline and metformin therapy. Secondary endpoints will include the motor function measure (MFM) and its items and subscores, the 6-minute walking test, timed function tests and quantitative muscle testing. Furthermore, quantitative muscle MRI assessment to evaluate the muscle fat fraction as well as safety and biomarker laboratory analyses from blood will be included. For comparison, muscle metabolism and mitochondrial function will be analysed in 10–20 healthy age-matched male children. Discussion The aim of this study is to test if a 6-month treatment of a combination of l-citrulline and metformin is more effective than placebo in preventing loss of motor function and muscle degeneration in DMD. The MFM D1 subscore is used as a clinical outcome measure and a quantitative muscle MRI assessment as the surrogate outcome measure of fatty muscle degeneration. Trial registration ClinicalTrials.gov: NCT01995032. Registered on 20 November 2013. Electronic supplementary material The online version of this article (doi:10.1186/s13063-016-1503-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Patricia Hafner
- Division of Neuropaediatrics, University of Basel Children's Hospital, Basel, Switzerland. .,Division of Neurology, Medical University Clinic, Kantonsspital Baselland, Bruderholz, Switzerland. .,Division of Neuropaediatrics, University Children's Hospital, Spitalstrasse 33, Postfach, Basel, 4056, Switzerland.
| | - Ulrike Bonati
- Division of Neuropaediatrics, University of Basel Children's Hospital, Basel, Switzerland
| | - Daniela Rubino
- Division of Neuropaediatrics, University of Basel Children's Hospital, Basel, Switzerland
| | - Vanya Gocheva
- Division of Neuropaediatrics, University of Basel Children's Hospital, Basel, Switzerland
| | - Thomas Zumbrunn
- Department of Clinical research, Clinical Trial Unit, University of Basel Hospital, Basel, Switzerland
| | - Nuri Gueven
- Pharmacy, School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Dirk Fischer
- Division of Neuropaediatrics, University of Basel Children's Hospital, Basel, Switzerland.,Division of Neurology, Medical University Clinic, Kantonsspital Baselland, Bruderholz, Switzerland.,Department of Neurology, University of Basel Hospital, Basel, Switzerland
| |
Collapse
|
84
|
Heberer K, Fowler E, Staudt L, Sienko S, Buckon CE, Bagley A, Sison-Williamson M, McDonald CM, Sussman MD. Hip kinetics during gait are clinically meaningful outcomes in young boys with Duchenne muscular dystrophy. Gait Posture 2016; 48:159-164. [PMID: 27267770 DOI: 10.1016/j.gaitpost.2016.05.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 05/19/2016] [Accepted: 05/23/2016] [Indexed: 02/02/2023]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked genetic neuromuscular disorder characterized by progressive proximal to distal muscle weakness. The success of randomized clinical trials for novel therapeutics depends on outcome measurements that are sensitive to change. As the development of motor skills may lead to functional improvements in young boys with DMD, their inclusion may potentially confound clinical trials. Three-dimensional gait analysis is an under-utilized approach that can quantify joint moments and powers, which reflect functional muscle strength. In this study, gait kinetics, kinematics, spatial-temporal parameters, and timed functional tests were quantified over a one-year period for 21 boys between 4 and 8 years old who were enrolled in a multisite natural history study. At baseline, hip moments and powers were inadequate. Between the two visits, 12 boys began a corticosteroid regimen (mean duration 10.8±2.4 months) while 9 boys remained steroid-naïve. Significant between-group differences favoring steroid use were found for primary kinetic outcomes (peak hip extensor moments (p=.007), duration of hip extensor moments (p=.007), peak hip power generation (p=.028)), and spatial-temporal parameters (walking speed (p=.016) and cadence (p=.021)). Significant between-group differences were not found for kinematics or timed functional tests with the exception of the 10m walk test (p=.03), which improves in typically developing children within this age range. These results indicate that hip joint kinetics can be used to identify weakness in young boys with DMD and are sensitive to corticosteroid intervention. Inclusion of gait analysis may enhance detection of a treatment effect in clinical trials particularly for young boys with more preserved muscle function.
Collapse
Affiliation(s)
- Kent Heberer
- Department of Orthopaedics, University of California, Los Angeles, 1000 Veteran Ave., Rehab. Building 22-64, Los Angeles, CA 90025, United States.
| | - Eileen Fowler
- Department of Orthopaedics, University of California, Los Angeles, 1000 Veteran Ave., Rehab. Building 22-64, Los Angeles, CA 90025, United States
| | - Loretta Staudt
- Department of Orthopaedics, University of California, Los Angeles, 1000 Veteran Ave., Rehab. Building 22-64, Los Angeles, CA 90025, United States
| | - Susan Sienko
- Department of Clinical Research, Shriners Hospitals for Children Portland, 3101 SW Sam Jackson Park Road, Portland, OR 97239, United States
| | - Cathleen E Buckon
- Department of Clinical Research, Shriners Hospitals for Children Portland, 3101 SW Sam Jackson Park Road, Portland, OR 97239, United States
| | - Anita Bagley
- Motion Analysis Laboratory, Shriners Hospitals for Children Northern California, 2425 Stockton Blvd., Sacramento, CA 95817, United States
| | - Mitell Sison-Williamson
- Motion Analysis Laboratory, Shriners Hospitals for Children Northern California, 2425 Stockton Blvd., Sacramento, CA 95817, United States
| | - Craig M McDonald
- Motion Analysis Laboratory, Shriners Hospitals for Children Northern California, 2425 Stockton Blvd., Sacramento, CA 95817, United States; University of California Davis Medical Center, 2315 Stockton Blvd., Sacramento, CA 95817, United States
| | - Michael D Sussman
- Department of Clinical Research, Shriners Hospitals for Children Portland, 3101 SW Sam Jackson Park Road, Portland, OR 97239, United States
| |
Collapse
|
85
|
Bello L, Morgenroth LP, Gordish-Dressman H, Hoffman EP, McDonald CM, Cirak S. DMD genotypes and loss of ambulation in the CINRG Duchenne Natural History Study. Neurology 2016; 87:401-9. [PMID: 27343068 DOI: 10.1212/wnl.0000000000002891] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 04/22/2016] [Indexed: 01/16/2023] Open
Abstract
OBJECTIVE To correlate time to loss of ambulation (LoA) and different truncating DMD gene mutations in a large, prospective natural history study of Duchenne muscular dystrophy (DMD), with particular attention to mutations amenable to emerging molecular treatments. METHODS We analyzed data from the Cooperative International Neuromuscular Research Group Duchenne Natural History Study for participants with DMD single- or multi-exon deletions or duplications with defined exon boundaries (n = 186), or small mutations identified by sequencing (n = 26, including 16 nonsense point mutations). We performed a time-to-event analysis of LoA, a strong indicator of overall disease severity, adjusting for glucocorticoid treatment and genetic modifiers. RESULTS Participants with deletions amenable to skipping of exon 44 had later LoA (median 14.8 years, hazard ratio 0.31, 95% confidence interval 0.14-0.69, p = 0.004). Age at LoA did not differ significantly in participants with deletions amenable to exon 45, 51, and 53 skipping, duplications, and small rearrangements. Nonsense mutation DMD also showed a typical median age at LoA (11.1 years), with a few outliers (ambulatory around or after 16 years of age) carrying stop codons within in-frame exons, more often situated in the rod domain. CONCLUSIONS As exon 44 skipping-amenable DMD has a later LoA, mutation-specific randomization and selection of placebo groups are essential for the success of clinical trials.
Collapse
Affiliation(s)
- Luca Bello
- From the Children's National Health System (L.B., L.P.M., H.G.-D., E.P.H., S.C.), Washington, DC; Department of Neuroscience (L.B.), University of Padova, Italy; University of California Davis Medical Center (C.M.M.), Sacramento; and Institute of Human Genetics (S.C.), University Children's Hospital, and Center of Molecular Medicine, University of Cologne, Germany
| | - Lauren P Morgenroth
- From the Children's National Health System (L.B., L.P.M., H.G.-D., E.P.H., S.C.), Washington, DC; Department of Neuroscience (L.B.), University of Padova, Italy; University of California Davis Medical Center (C.M.M.), Sacramento; and Institute of Human Genetics (S.C.), University Children's Hospital, and Center of Molecular Medicine, University of Cologne, Germany
| | - Heather Gordish-Dressman
- From the Children's National Health System (L.B., L.P.M., H.G.-D., E.P.H., S.C.), Washington, DC; Department of Neuroscience (L.B.), University of Padova, Italy; University of California Davis Medical Center (C.M.M.), Sacramento; and Institute of Human Genetics (S.C.), University Children's Hospital, and Center of Molecular Medicine, University of Cologne, Germany
| | - Eric P Hoffman
- From the Children's National Health System (L.B., L.P.M., H.G.-D., E.P.H., S.C.), Washington, DC; Department of Neuroscience (L.B.), University of Padova, Italy; University of California Davis Medical Center (C.M.M.), Sacramento; and Institute of Human Genetics (S.C.), University Children's Hospital, and Center of Molecular Medicine, University of Cologne, Germany
| | - Craig M McDonald
- From the Children's National Health System (L.B., L.P.M., H.G.-D., E.P.H., S.C.), Washington, DC; Department of Neuroscience (L.B.), University of Padova, Italy; University of California Davis Medical Center (C.M.M.), Sacramento; and Institute of Human Genetics (S.C.), University Children's Hospital, and Center of Molecular Medicine, University of Cologne, Germany
| | | | | |
Collapse
|
86
|
Cossu G, Previtali SC, Napolitano S, Cicalese MP, Tedesco FS, Nicastro F, Noviello M, Roostalu U, Natali Sora MG, Scarlato M, De Pellegrin M, Godi C, Giuliani S, Ciotti F, Tonlorenzi R, Lorenzetti I, Rivellini C, Benedetti S, Gatti R, Marktel S, Mazzi B, Tettamanti A, Ragazzi M, Imro MA, Marano G, Ambrosi A, Fiori R, Sormani MP, Bonini C, Venturini M, Politi LS, Torrente Y, Ciceri F. Intra-arterial transplantation of HLA-matched donor mesoangioblasts in Duchenne muscular dystrophy. EMBO Mol Med 2016; 7:1513-28. [PMID: 26543057 PMCID: PMC4693504 DOI: 10.15252/emmm.201505636] [Citation(s) in RCA: 120] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Intra‐arterial transplantation of mesoangioblasts proved safe and partially efficacious in preclinical models of muscular dystrophy. We now report the first‐in‐human, exploratory, non‐randomized open‐label phase I–IIa clinical trial of intra‐arterial HLA‐matched donor cell transplantation in 5 Duchenne patients. We administered escalating doses of donor‐derived mesoangioblasts in limb arteries under immunosuppressive therapy (tacrolimus). Four consecutive infusions were performed at 2‐month intervals, preceded and followed by clinical, laboratory, and muscular MRI analyses. Two months after the last infusion, a muscle biopsy was performed. Safety was the primary endpoint. The study was relatively safe: One patient developed a thalamic stroke with no clinical consequences and whose correlation with mesoangioblast infusion remained unclear. MRI documented the progression of the disease in 4/5 patients. Functional measures were transiently stabilized in 2/3 ambulant patients, but no functional improvements were observed. Low level of donor DNA was detected in muscle biopsies of 4/5 patients and donor‐derived dystrophin in 1. Intra‐arterial transplantation of donor mesoangioblasts in human proved to be feasible and relatively safe. Future implementation of the protocol, together with a younger age of patients, will be needed to approach efficacy.
Collapse
Affiliation(s)
- Giulio Cossu
- Institute of Inflammation and Repair, University of Manchester, Manchester, UK
| | - Stefano C Previtali
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sara Napolitano
- HSR/TIGET Pediatric Clinical Research Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy Hematology and BMT Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Pia Cicalese
- HSR/TIGET Pediatric Clinical Research Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy Hematology and BMT Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Francesca Nicastro
- Laboratory of Analysis and Rehabilitation of Motor Function, Division of Neurosciences, IRCCS San Raffaele Scientific Institute, Milan, Italy Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maddalena Noviello
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Urmas Roostalu
- Institute of Inflammation and Repair, University of Manchester, Manchester, UK
| | | | - Marina Scarlato
- Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Claudia Godi
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy Neuroradiology Department and Neuroradiology Research Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Serena Giuliani
- Hematology and BMT Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Ciotti
- Hematology and BMT Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rossana Tonlorenzi
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Isabella Lorenzetti
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Cristina Rivellini
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sara Benedetti
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Roberto Gatti
- Laboratory of Analysis and Rehabilitation of Motor Function, Division of Neurosciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sarah Marktel
- Hematology and BMT Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Benedetta Mazzi
- Immunogenetics Laboratory, Department of Immunohematology & Blood Transfusion, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Tettamanti
- Laboratory of Analysis and Rehabilitation of Motor Function, Division of Neurosciences, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Martina Ragazzi
- Department of Cell and Developmental Biology, University College London, London, UK
| | | | | | | | - Rossana Fiori
- Unit of Anesthesiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Chiara Bonini
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Venturini
- Department of Radiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Letterio S Politi
- Neuroradiology Department and Neuroradiology Research Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Yvan Torrente
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Fabio Ciceri
- HSR/TIGET Pediatric Clinical Research Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
87
|
Mazzone ES, Coratti G, Sormani MP, Messina S, Pane M, D'Amico A, Colia G, Fanelli L, Berardinelli A, Gardani A, Lanzillotta V, D’Ambrosio P, Petillo R, Cavallaro F, Frosini S, Bello L, Bonfiglio S, De Sanctis R, Rolle E, Forcina N, Magri F, Vita G, Palermo C, Donati MA, Procopio E, Arnoldi MT, Baranello G, Mongini T, Pini A, Battini R, Pegoraro E, Torrente Y, Previtali SC, Bruno C, Politano L, Comi GP, D’Angelo MG, Bertini E, Mercuri E. Timed Rise from Floor as a Predictor of Disease Progression in Duchenne Muscular Dystrophy: An Observational Study. PLoS One 2016; 11:e0151445. [PMID: 26982196 PMCID: PMC4794120 DOI: 10.1371/journal.pone.0151445] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Accepted: 02/29/2016] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The role of timed items, and more specifically, of the time to rise from the floor, has been reported as an early prognostic factor for disease progression and loss of ambulation. The aim of our study was to investigate the possible effect of the time to rise from the floor test on the changes observed on the 6MWT over 12 months in a cohort of ambulant Duchenne boys. SUBJECTS AND METHODS A total of 487 12-month data points were collected from 215 ambulant Duchenne boys. The age ranged between 5.0 and 20.0 years (mean 8.48 ±2.48 DS). RESULTS The results of the time to rise from the floor at baseline ranged from 1.2 to 29.4 seconds in the boys who could perform the test. 49 patients were unable to perform the test at baseline and 87 at 12 month The 6MWT values ranged from 82 to 567 meters at baseline. 3 patients lost the ability to perform the 6mwt at 12 months. The correlation between time to rise from the floor and 6MWT at baseline was high (r = 0.6, p<0.01). CONCLUSIONS Both time to rise from the floor and baseline 6MWT were relevant for predicting 6MWT changes in the group above the age of 7 years, with no interaction between the two measures, as the impact of time to rise from the floor on 6MWT change was similar in the patients below and above 350 m. Our results suggest that, time to rise from the floor can be considered an additional important prognostic factor of 12 month changes on the 6MWT and, more generally, of disease progression.
Collapse
Affiliation(s)
- Elena S. Mazzone
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| | - Giorgia Coratti
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| | - Maria Pia Sormani
- Biostatistics Unit, Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Sonia Messina
- Department of Neurosciences and Nemo and Clinical Center, Psychiatry and Anaesthesiology, University of Messina, Messina, Italy
| | - Marika Pane
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| | - Adele D'Amico
- Unit of Neuromuscular and Neurodegenerative Diseases, Department of Neurosciences, Bambino Gesù Children's Hospital, Rome, Italy
| | - Giulia Colia
- Unit of Neuromuscular and Neurodegenerative Diseases, Department of Neurosciences, Bambino Gesù Children's Hospital, Rome, Italy
| | - Lavinia Fanelli
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| | | | - Alice Gardani
- Child Neurology and Psychiatry Unit, “C. Mondino” Foundation, Pavia, Italy
| | | | - Paola D’Ambrosio
- Cardiomiologia e genetica medica, Dipartimento di Medicina Sperimentale, Seconda Università di Napoli, Napoli, Italy
| | - Roberta Petillo
- Cardiomiologia e genetica medica, Dipartimento di Medicina Sperimentale, Seconda Università di Napoli, Napoli, Italy
| | - Filippo Cavallaro
- Department of Neurosciences and Nemo and Clinical Center, Psychiatry and Anaesthesiology, University of Messina, Messina, Italy
| | - Silvia Frosini
- Department of Developmental Neuroscience, Stella Maris Institute, Pisa,Italy
| | - Luca Bello
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Serena Bonfiglio
- Child Neurology and Psychiatry Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | | | - Enrica Rolle
- Neuromuscular Center, SG. Battista Hospital, University of Turin, Turin, Italy
| | - Nicola Forcina
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| | - Francesca Magri
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCSS Foudation, Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gianluca Vita
- Department of Neurosciences and Nemo and Clinical Center, Psychiatry and Anaesthesiology, University of Messina, Messina, Italy
| | - Concetta Palermo
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| | | | - Elena Procopio
- Metabolic and Neuromuscular Unit, Meyer Hospital, Florence, Italy
| | | | - Giovanni Baranello
- Developmental Neurology Unit, Istituto Neurologico “Besta” Milan, Milan, Italy
| | - Tiziana Mongini
- Neuromuscular Center, SG. Battista Hospital, University of Turin, Turin, Italy
| | - Antonella Pini
- Child Neurology and Psychiatry Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Roberta Battini
- Department of Developmental Neuroscience, Stella Maris Institute, Pisa,Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Yvan Torrente
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCSS Foudation, Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano C. Previtali
- Neuromuscular repair unit, Inspe and division of neuroscience, IRCSS San Raffaele Scientific Institute, Milan, Italy
| | - Claudio Bruno
- Neuromuscular Disease Unit, G. Gaslini Institute, Genoa, Italy
| | - Luisa Politano
- Cardiomiologia e genetica medica, Dipartimento di Medicina Sperimentale, Seconda Università di Napoli, Napoli, Italy
| | - Giacomo P. Comi
- Dino Ferrari Centre, Neuroscience Section, Department of Pathophysiology and Transplantation (DEPT), University of Milan, Neurology Unit, IRCSS Foudation, Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Grazia D’Angelo
- Neuromuscular Disorders Unit, Scientific Institute IRCCS E. Medea, 23842, Bosisio Parini, Lecco, Italy
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Diseases, Department of Neurosciences, Bambino Gesù Children's Hospital, Rome, Italy
| | - Eugenio Mercuri
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| |
Collapse
|
88
|
The Performance of the Upper Limb scores correlate with pulmonary function test measures and Egen Klassifikation scores in Duchenne muscular dystrophy. Neuromuscul Disord 2016; 26:264-71. [PMID: 27056113 DOI: 10.1016/j.nmd.2016.02.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Revised: 01/18/2016] [Accepted: 02/29/2016] [Indexed: 11/22/2022]
Abstract
The Performance of the Upper Limb scale was developed as an outcome measure specifically for ambulant and non-ambulant patients with Duchenne muscular dystrophy and is implemented in clinical trials needing longitudinal data. The aim of this study is to determine whether this novel tool correlates with functional ability using pulmonary function test, cardiac function test and Egen Klassifikation scale scores as clinical measures. In this cross-sectional study, 43 non-ambulatory Duchenne males from ages 10 to 30 years and on long-term glucocorticoid treatment were enrolled. Cardiac and pulmonary function test results were analyzed to assess cardiopulmonary function, and Egen Klassifikation scores were analyzed to assess functional ability. The Performance of the Upper Limb scores correlated with pulmonary function measures and had inverse correlation with Egen Klassifikation scores. There was no correlation with left ventricular ejection fraction and left ventricular dysfunction. Body mass index and decreased joint range of motion affected total Performance of the Upper Limb scores and should be considered in clinical trial designs.
Collapse
|
89
|
Messina S, Vita GL, Sframeli M, Mondello S, Mazzone E, D'Amico A, Berardinelli A, La Rosa M, Bruno C, Distefano MG, Baranello G, Barcellona C, Scutifero M, Marcato S, Palmieri A, Politano L, Morandi L, Mongini T, Pegoraro E, D'Angelo MG, Pane M, Rodolico C, Minetti C, Bertini E, Vita G, Mercuri E. Health-related quality of life and functional changes in DMD: A 12-month longitudinal cohort study. Neuromuscul Disord 2016; 26:189-96. [PMID: 26916554 PMCID: PMC4819956 DOI: 10.1016/j.nmd.2016.01.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 01/21/2016] [Accepted: 01/25/2016] [Indexed: 11/26/2022]
Abstract
At baseline, the PedsQLTM inventories correlated with almost all the functional measures. There was a significant decrease between baseline and 12 months on PedsQLTM GCS. This decrement paralleled with the decrement in the functional outcome measures. PedsQLTM correlates with the level of impairment. This correlations were not confirmed when 12 month changes are considered.
In Duchenne muscular dystrophy (DMD) little has been reported on the association between clinical outcome measures and patient health-related quality of life (HRQOL) tools. Our study evaluated the relationship between 12 month changes on the Generic Core Scales (GCS), the Multidimensional Fatigue Scale and the Neuromuscular Module of the PedsQLTM with several outcome measures (6 minute walk test, North Star Ambulatory Assessment and timed items) in ambulatory DMD. Ninety-eight ambulatory DMD in a multicentric setting were included in the study. At baseline, the PedsQLTM inventories correlated with almost all the functional measures On the Child Self-Report there was a significant decrease between baseline and 12 months on the PedsQLTM GCS and its first domain, in parallel with the decrement in the functional outcome measures. Correlation between the 12 month changes on the PedsQLTM inventories and functional measures were almost all negligible. Similar results were obtained on the Parent Proxy-Report. In conclusion, PedsQLTM correlates with the level of impairment at baseline, but this does not hold true when 12 month changes are considered. Further studies comparing different tools are needed to better elucidate the complexity of the relationship between HRQOL and functional performances.
Collapse
Affiliation(s)
- Sonia Messina
- Department of Neurosciences, University of Messina, Messina, Italy; Nemo Sud Clinical Center for Neuromuscular Diseases, Messina, Italy
| | - Gian Luca Vita
- Nemo Sud Clinical Center for Neuromuscular Diseases, Messina, Italy
| | - Maria Sframeli
- Nemo Sud Clinical Center for Neuromuscular Diseases, Messina, Italy
| | | | - Elena Mazzone
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| | - Adele D'Amico
- Unit of Neuromuscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesu' Children's Research Hospital, Rome, Italy
| | - Angela Berardinelli
- IRCCS "C.Mondino" Institute, Unit of Child Neuropsychiatry, University of Pavia, Pavia, Italy
| | - Matteo La Rosa
- Department of Neurosciences, University of Messina, Messina, Italy
| | - Claudio Bruno
- Neuromuscular Disease Unit, G. Gaslini Institute, Genoa, Italy
| | | | - Giovanni Baranello
- Muscle Pathology and Neuroimmunology and Developmental Neurology Unit, Neurological Institute "Carlo Besta", Milan, Italy
| | | | - Marianna Scutifero
- Department of Experimental Medicine, Cardiomyology and Medical Genetics, Second University of Naples, Naples, Italy
| | - Sonia Marcato
- Department of Neurosciences, University of Padua, Padua, Italy
| | | | - Luisa Politano
- Department of Experimental Medicine, Cardiomyology and Medical Genetics, Second University of Naples, Naples, Italy
| | - Lucia Morandi
- Muscle Pathology and Neuroimmunology and Developmental Neurology Unit, Neurological Institute "Carlo Besta", Milan, Italy
| | - Tiziana Mongini
- Neuromuscular Center, SG. Battista Hospital, University of Turin, Turin, Italy
| | - Elena Pegoraro
- Department of Neurosciences, University of Padua, Padua, Italy
| | - Maria Grazia D'Angelo
- IRCCS E Medea Bosisio Parini Neuromuscular Unit, Department of Neurorehabilitation, Bosisio Parini, Italy
| | - Marika Pane
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| | - Carmelo Rodolico
- Department of Neurosciences, University of Messina, Messina, Italy
| | - Carlo Minetti
- Neuromuscular Disease Unit, G. Gaslini Institute, Genoa, Italy
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesu' Children's Research Hospital, Rome, Italy
| | - Giuseppe Vita
- Department of Neurosciences, University of Messina, Messina, Italy; Nemo Sud Clinical Center for Neuromuscular Diseases, Messina, Italy
| | - Eugenio Mercuri
- Department of Paediatric Neurology, Catholic University, Rome, Italy.
| |
Collapse
|
90
|
Ricotti V, Ridout DA, Pane M, Main M, Mayhew A, Mercuri E, Manzur AY, Muntoni F. The NorthStar Ambulatory Assessment in Duchenne muscular dystrophy: considerations for the design of clinical trials. J Neurol Neurosurg Psychiatry 2016; 87:149-55. [PMID: 25733532 PMCID: PMC4752678 DOI: 10.1136/jnnp-2014-309405] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 01/25/2015] [Indexed: 01/16/2023]
Abstract
OBJECTIVE With the emergence of experimental therapies for Duchenne muscular dystrophy (DMD), it is fundamental to understand the natural history of this disorder to properly design clinical trials. The aims of this study were to assess the effects produced on motor function by different DMD genotypes and early initiation of glucocorticoids. METHODS Through the NorthStar Network, standardised clinical data including the NorthStar Ambulatory Assessment score (NSAA) on 513 ambulant UK boys with DMD were analysed from 2004 to 2012. For the analysis of the genetic subpopulation, we also included data from 172 Italian boys with DMD. NSAA raw scores were converted into linear scores. RESULTS On the linearised NSAA, we observed an average decline of 8 units/year (4 units on raw NSAA analysis) after age 7. The median age at loss of ambulation (LOA) was 13 years (95% CI 12.1 to 13.5); 2 years prior to LOA, the estimated mean linearised NSAA score was 42/100 (13/34 raw scale). Starting glucocorticoids between 3 and 5 years conferred an additional gain in motor function of 3 units/year (1.3 raw units) up to age 7. When analysing the effect of genotype in the UK and Italian cumulative cohorts, individuals with deletions amenable to exons 44 and 46 skipping declined at a slower rate over 2 years (9 units (4 raw units), p<0.001), while 53 and 51 skippable deletions showed a faster decline of 14 (4.5; p<0.001) and 5 linearised units (2.4 NSAA units; p=0.02), respectively. CONCLUSIONS Our study provides a novel insight on the current natural history of DMD, which will be instrumental for the design of future clinical trials.
Collapse
Affiliation(s)
- Valeria Ricotti
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK Great Ormond Street Hospital, London, UK
| | - Deborah A Ridout
- Department of Population, Policy and Practice Programme, UCL Institute of Child Health, London, UK Great Ormond Street Hospital, London, UK
| | - Marika Pane
- Department of Paediatric Neurology, Catholic University, Rome, Italy
| | - Marion Main
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK Great Ormond Street Hospital, London, UK
| | | | - Eugenio Mercuri
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK Department of Paediatric Neurology, Catholic University, Rome, Italy
| | - Adnan Y Manzur
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK Great Ormond Street Hospital, London, UK
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, UCL Institute of Child Health, London, UK Great Ormond Street Hospital, London, UK
| | | |
Collapse
|
91
|
Hafner P, Bonati U, Erne B, Schmid M, Rubino D, Pohlman U, Peters T, Rutz E, Frank S, Neuhaus C, Deuster S, Gloor M, Bieri O, Fischmann A, Sinnreich M, Gueven N, Fischer D. Improved Muscle Function in Duchenne Muscular Dystrophy through L-Arginine and Metformin: An Investigator-Initiated, Open-Label, Single-Center, Proof-Of-Concept-Study. PLoS One 2016; 11:e0147634. [PMID: 26799743 PMCID: PMC4723144 DOI: 10.1371/journal.pone.0147634] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 01/05/2016] [Indexed: 12/31/2022] Open
Abstract
Altered neuronal nitric oxide synthase function in Duchenne muscular dystrophy leads to impaired mitochondrial function which is thought to be one cause of muscle damage in this disease. The study tested if increased intramuscular nitric oxide concentration can improve mitochondrial energy metabolism in Duchenne muscular dystrophy using a novel therapeutic approach through the combination of L-arginine with metformin. Five ambulatory, genetically confirmed Duchenne muscular dystrophy patients aged between 7–10 years were treated with L-arginine (3 x 2.5 g/d) and metformin (2 x 250 mg/d) for 16 weeks. Treatment effects were assessed using mitochondrial protein expression analysis in muscular biopsies, indirect calorimetry, Dual-Energy X-Ray Absorptiometry, quantitative thigh muscle MRI, and clinical scores of muscle performance. There were no serious side effects and no patient dropped out. Muscle biopsy results showed pre-treatment a significantly reduced mitochondrial protein expression and increased oxidative stress in Duchenne muscular dystrophy patients compared to controls. Post-treatment a significant elevation of proteins of the mitochondrial electron transport chain was observed as well as a reduction in oxidative stress. Treatment also decreased resting energy expenditure rates and energy substrate use shifted from carbohydrates to fatty acids. These changes were associated with improved clinical scores. In conclusion pharmacological stimulation of the nitric oxide pathway leads to improved mitochondria function and clinically a slowing of disease progression in Duchenne muscular dystrophy. This study shall lead to further development of this novel therapeutic approach into a real alternative for Duchenne muscular dystrophy patients.
Collapse
Affiliation(s)
- Patricia Hafner
- Division of Neuropaediatrics, University of Basel Children's Hospital, Basel, Switzerland.,Department of Neurology, University of Basel Hospital, Basel, Switzerland
| | - Ulrike Bonati
- Division of Neuropaediatrics, University of Basel Children's Hospital, Basel, Switzerland
| | - Beat Erne
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Maurice Schmid
- Division of Neuropaediatrics, University of Basel Children's Hospital, Basel, Switzerland
| | - Daniela Rubino
- Division of Neuropaediatrics, University of Basel Children's Hospital, Basel, Switzerland
| | - Urs Pohlman
- Division of Neuropaediatrics, University of Basel Children's Hospital, Basel, Switzerland
| | - Thomas Peters
- Interdisciplinary Center of Nutritional and Metabolic Diseases, St. Claraspital, Basel, Basel, Switzerland
| | - Erich Rutz
- Paediatric Orthopaedic Department, University of Basel Children's Hospital, Basel, Switzerland
| | - Stephan Frank
- Division of Neuropathology, Institute of Pathology, University of Basel Hospital, Basel, Switzerland
| | - Cornelia Neuhaus
- Therapy Department, University of Basel Children's Hospital, Basel, Switzerland
| | - Stefanie Deuster
- Hospital Pharmacy, University of Basel Hospital, Basel, Switzerland
| | - Monika Gloor
- Department of Radiology, Division of Radiological Physics, University of Basel Hospital, Basel, Switzerland
| | - Oliver Bieri
- Department of Radiology, Division of Radiological Physics, University of Basel Hospital, Basel, Switzerland
| | - Arne Fischmann
- Division of Neuroradiology, University of Basel Hospital, Basel, Switzerland
| | - Michael Sinnreich
- Department of Neurology, University of Basel Hospital, Basel, Switzerland.,Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Nuri Gueven
- Pharmacy, School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Dirk Fischer
- Division of Neuropaediatrics, University of Basel Children's Hospital, Basel, Switzerland.,Department of Neurology, University of Basel Hospital, Basel, Switzerland
| |
Collapse
|
92
|
Mendell JR, Goemans N, Lowes LP, Alfano LN, Berry K, Shao J, Kaye EM, Mercuri E. Longitudinal effect of eteplirsen versus historical control on ambulation in Duchenne muscular dystrophy. Ann Neurol 2016; 79:257-71. [PMID: 26573217 PMCID: PMC5064753 DOI: 10.1002/ana.24555] [Citation(s) in RCA: 405] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 11/03/2015] [Accepted: 11/03/2015] [Indexed: 12/13/2022]
Abstract
Objective To continue evaluation of the long‐term efficacy and safety of eteplirsen, a phosphorodiamidate morpholino oligomer designed to skip DMD exon 51 in patients with Duchenne muscular dystrophy (DMD). Three‐year progression of eteplirsen‐treated patients was compared to matched historical controls (HC). Methods Ambulatory DMD patients who were ≥7 years old and amenable to exon 51 skipping were randomized to eteplirsen (30/50mg/kg) or placebo for 24 weeks. Thereafter, all received eteplirsen on an open‐label basis. The primary functional assessment in this study was the 6‐Minute Walk Test (6MWT). Respiratory muscle function was assessed by pulmonary function testing (PFT). Longitudinal natural history data were used for comparative analysis of 6MWT performance at baseline and months 12, 24, and 36. Patients were matched to the eteplirsen group based on age, corticosteroid use, and genotype. Results At 36 months, eteplirsen‐treated patients (n = 12) demonstrated a statistically significant advantage of 151m (p < 0.01) on 6MWT and experienced a lower incidence of loss of ambulation in comparison to matched HC (n = 13) amenable to exon 51 skipping. PFT results remained relatively stable in eteplirsen‐treated patients. Eteplirsen was well tolerated. Analysis of HC confirmed the previously observed change in disease trajectory at age 7 years, and more severe progression was observed in patients with mutations amenable to exon skipping than in those not amenable. The subset of patients amenable to exon 51 skipping showed a more severe disease course than those amenable to any exon skipping. Interpretation Over 3 years of follow‐up, eteplirsen‐treated patients showed a slower rate of decline in ambulation assessed by 6MWT compared to untreated matched HC. Ann Neurol 2016;79:257–271
Collapse
Affiliation(s)
- Jerry R Mendell
- Nationwide Children's Hospital, Columbus, OH.,Department of Pediatrics and Neurology, Ohio State University, Columbus, OH.,Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, Rochester, NY
| | | | - Linda P Lowes
- Nationwide Children's Hospital, Columbus, OH.,Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, Rochester, NY
| | - Lindsay N Alfano
- Nationwide Children's Hospital, Columbus, OH.,Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, Rochester, NY
| | - Katherine Berry
- Nationwide Children's Hospital, Columbus, OH.,Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, Rochester, NY
| | | | | | | | | |
Collapse
|
93
|
Zaidman CM, Malkus EC, Connolly AM. Muscle ultrasound quantifies disease progression over time in infants and young boys with duchenne muscular dystrophy. Muscle Nerve 2015; 52:334-8. [PMID: 25704979 PMCID: PMC5931214 DOI: 10.1002/mus.24609] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Revised: 01/27/2015] [Accepted: 02/10/2015] [Indexed: 11/10/2022]
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.
Collapse
Affiliation(s)
- Craig M Zaidman
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8111, St. Louis, Missouri, 63110, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Elizabeth C Malkus
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8111, St. Louis, Missouri, 63110, USA
| | - Anne M Connolly
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, Box 8111, St. Louis, Missouri, 63110, USA
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
94
|
Quantitative muscle MRI: A powerful surrogate outcome measure in Duchenne muscular dystrophy. Neuromuscul Disord 2015; 25:679-85. [DOI: 10.1016/j.nmd.2015.05.006] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 05/11/2015] [Accepted: 05/15/2015] [Indexed: 02/04/2023]
|
95
|
Abstract
Mutations in the DMD gene result in Duchenne or Becker muscular dystrophy due to absent or altered expression of the dystrophin protein. The more severe Duchenne muscular dystrophy typically presents around ages 2 to 5 with gait disturbance, and historically has led to the loss of ambulation by age 12. It is important for the practicing pediatrician, however, to be aware of other presenting signs, such as delayed motor or cognitive milestones, or elevated serum transaminases. Becker muscular dystrophy is milder, often presenting after age 5, with ambulation frequently preserved past 20 years and sometimes into late decades.
Collapse
Affiliation(s)
- Nicolas Wein
- The Center for Gene Therapy, The Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Lindsay Alfano
- The Center for Gene Therapy, The Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA; Department of Physical Therapy, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA
| | - Kevin M Flanigan
- The Center for Gene Therapy, The Research Institute, Nationwide Children's Hospital, 700 Children's Drive, Columbus, OH 43205, USA; Department of Pediatrics, Ohio State University, 700 Children's Drive, Columbus, OH 43205, USA; Department of Neurology, Ohio State University, 700 Children's Drive, Columbus, OH 43205, USA.
| |
Collapse
|
96
|
Pillen S, Van Alfen N. Muscle ultrasound from diagnostic tool to outcome measure--Quantification is the challenge. Muscle Nerve 2015; 52:319-20. [PMID: 25703022 DOI: 10.1002/mus.24613] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 12/17/2014] [Accepted: 02/10/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Sigrid Pillen
- Department of Sleep Medicine, Kempenhaeghe Expertise Center for Epileptology, Sleep Medicine and Neurocognition, Sterkselseweg 65, 5591, VE Heeze, The Netherlands
| | - Nens Van Alfen
- Department of Neurology and Clinical Neurophysiology, Radboudumc, Nijmegen, The Netherlands
| |
Collapse
|
97
|
Davidson ZE, Ryan MM, Kornberg AJ, Walker KZ, Truby H. Strong correlation between the 6-minute walk test and accelerometry functional outcomes in boys with Duchenne muscular dystrophy. J Child Neurol 2015; 30:357-63. [PMID: 24762862 DOI: 10.1177/0883073814530502] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Accelerometry provides information on habitual physical capability that may be of value in the assessment of function in Duchenne muscular dystrophy. This preliminary investigation describes the relationship between community ambulation measured by the StepWatch activity monitor and the current standard of functional assessment, the 6-minute walk test, in ambulatory boys with Duchenne muscular dystrophy (n = 16) and healthy controls (n = 13). All participants completed a 6-minute walk test and wore the StepWatch™ monitor for 5 consecutive days. Both the 6-minute walk test and StepWatch accelerometry identified a decreased capacity for ambulation in boys with Duchenne compared to healthy controls. There were strong, significant correlations between 6-minute walk distance and all StepWatch parameters in affected boys only (r = 0.701-0.804). These data proffer intriguing observations that warrant further exploration. Specifically, accelerometry outcomes may compliment the 6-minute walk test in assessment of therapeutic interventions for Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Zoe E Davidson
- Department of Nutrition and Dietetics, Monash University, Melbourne, Victoria, Australia Murdoch Childrens Research Institute, Melbourne, Victoria, Australia
| | - Monique M Ryan
- Department of Neurology, The Royal Children's Hospital, Melbourne, Victoria, Australia Murdoch Childrens Research Institute, Melbourne, Victoria, Australia
| | - Andrew J Kornberg
- Department of Neurology, The Royal Children's Hospital, Melbourne, Victoria, Australia Murdoch Childrens Research Institute, Melbourne, Victoria, Australia
| | - Karen Z Walker
- Department of Nutrition and Dietetics, Monash University, Melbourne, Victoria, Australia
| | - Helen Truby
- Department of Nutrition and Dietetics, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
98
|
Abstract
PURPOSE Although bracing in the late ambulatory stage of Duchenne muscular dystrophy (DMD) has been described, the effects of ankle-foot orthoses (AFOs) in earlier stages have not been evaluated. The aim of this pilot study was to describe the effects of dynamic response AFO (DR-AFO) use in boys with DMD who are ambulatory. METHODS Using a crossover design, 3 boys were randomly assigned to either a 2-week DR-AFO or a placebo intervention. Phases were separated by a 1-week washout period. Primary outcomes were time to walk 10 m and a 6-Minute Walk Test. RESULTS With DR-AFO use, declines in 10-m walk time (median decline = 0.8 s) and 6-Minute Walk Distance (median = 25.0 m) occurred. Parental report suggested that the use of DR-AFOs increased falls in 2 of 3 participants. CONCLUSION This pilot study does not support the use of DR-AFOs by boys with DMD who are ambulatory.
Collapse
|
99
|
Martini J, Hukuda ME, Caromano FA, Favero FM, Fu C, Voos MC. The clinical relevance of timed motor performance in children with Duchenne muscular dystrophy. Physiother Theory Pract 2014; 31:173-81. [DOI: 10.3109/09593985.2014.989294] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
100
|
Brehm MA, Kempen JCE, van der Kooi AJ, de Groot IJM, van den Bergen JC, Verschuuren JJGM, Niks EH, Harlaar J. Age-related longitudinal changes in metabolic energy expenditure during walking in boys with Duchenne muscular dystrophy. PLoS One 2014; 9:e115200. [PMID: 25506914 PMCID: PMC4266650 DOI: 10.1371/journal.pone.0115200] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 11/19/2014] [Indexed: 12/03/2022] Open
Abstract
Objective The aim of this study was to evaluate age-related changes in metabolic walking energy expenditure in ambulant boys affected by Duchenne muscular dystrophy over a follow-up period of 12 months. Methods At baseline (T1) and 12 months later (T2), metabolic walking energy expenditure was assessed during a 6-minute walk test at comfortable speed in 14 ambulant boys with Duchenne (age range: 6.0-12.5 years, mean 8.2). Outcome measures derived from the assessment included the 6-minute comfortable walking distance (m) and net-nondimensional energy cost relative to speed-matched control cost (SMC-EC, %). Statistical comparisons were made using a two-way repeated measures ANOVA (factors: time (T1 versus T2) and age (<8 years of age (yoa) versus ≥8 yoa)). Results Over the course of the study, a significant decrease of -28m (−8.2%, p = 0.043) was noted in the walked distance at comfortable speed. Besides, SMC-EC increased with 4.4%, although this change was not significant (p = 0.452). Regarding age groups, boys below 8 yoa showed a smaller annual decrease in the walked distance (−15 m) compared to boys above 8 yoa (−37 m). SMC-EC increased with 10% in the older boys, while in the younger boys it decreased (−2.1%). The main effect of age group on walking distance and SMC-EC however was not significant (p>0.158), and also there were no interaction effects (p>0.248). Conclusions The results of our small study suggest that the natural course of walking performance in ambulant boys with Duchenne is characterized by a decrease in comfortable walking distance and an increase in walking energy cost. The rate of energy cost seems to increase with age, while walking distance decreases, which is opposite from the trend in typically developing children.
Collapse
Affiliation(s)
- Merel-Anne Brehm
- Department of Rehabilitation Medicine and MOVE Research Institute, VU University Medical Center, Amsterdam, The Netherlands
- Department of Rehabilitation, Academic Medical Center, University of Amsterdam, The Netherlands
- * E-mail:
| | - Jiska C. E. Kempen
- Department of Rehabilitation Medicine and MOVE Research Institute, VU University Medical Center, Amsterdam, The Netherlands
| | - Anneke J. van der Kooi
- Department of Neurology, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Imelda J. M. de Groot
- Department of Rehabilitation, Radboud University Medical Center Nijmegen, The Netherlands
| | | | | | - Erik H. Niks
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jaap Harlaar
- Department of Rehabilitation Medicine and MOVE Research Institute, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|