Efficacy and Safety of Vamorolone in Duchenne Muscular Dystrophy: A 30-Month Nonrandomized Controlled Open-Label Extension Trial

Jean K Mah, Paula R Clemens, Michela Guglieri, Edward C Smith, Richard S Finkel, Mar Tulinius, Yoram Nevo, Monique M Ryan, Richard Webster, Diana Castro, Nancy L Kuntz, Craig M McDonald, Jesse M Damsker, Benjamin D Schwartz, Laurel J Mengle-Gaw, Stefan Jackowski, Georgia Stimpson, Deborah A Ridout, Vandana Ayyar-Gupta, Giovanni Baranello, Adnan Y Manzur, Francesco Muntoni, Heather Gordish-Dressman, Mika Leinonen, Leanne M Ward, Eric P Hoffman, Utkarsh J Dang, NorthStar UK Network and CINRG DNHS Investigators, Francesco Muntoni, Adnan Manzur, Giovanni Baranello, Stephanie Robb, Ros Quinlivan, Anna Sarkozy, Pinki Munot, Marion Main, Lianne Abbot, Volker Straub, Michela Guglieri, Chiara Bertolli, Anna Mayhew, Robert Muni-Lofra, Meredith James, Jassi Sodhi, Deepak Parasuraman, Zoya Alhaswani, Heather McMurchie, Rosanna Rabb, Anne-Marie Childs, Karen Pysden, Lindsey Pallant, Tiffany Small, Stefan Spinty, Rajesh Madhu, Alison Shillington, Sarah Gregson, Elizabeth Wraige, Heinz Jungbluth, Vasantha Gowda, Jennie Sheehan, Imelda Hughes, Sinead Warner, Emily Davies, Tracey Willis, Richa Kulshrestha, Nicholas Emery, Kate Strachan, Min Ong, Kay White, Kate Skone, Frances Gibbon, Bethan Parsons, Anirban Majumdar, Kayal Vijaykumar, Faye Mason, Claire Frimpong-Ansah, Karen Naismith, Julie Burslem, Iain Horrocks, Marina Di Marco, Sarah Brown, Sarah Williamson, Kirstie Spencer, Gabby Chow, Christian de Goede, Andrea Selley, Neil Thomas, Marjorie Illingworth, Michelle Greary, Jenni Palmer, Cathy White, Kate Greenfield, Grainne Nic Fhirleinn, Melanie Douglas, Sandya Tiraputhi, Nahin Hussain, Yvonne Julien, Gautam Ambegaonkar, Deepa Krishnakumar, Jacqui Taylor, Jane Tewnion, Elma Stephens, Saleel Chandratre, Sithara Ramdas, Hayley Ramjattan, Alex Baxter, Clare Eadie, Craig M McDonald, Erik K Henricson, R Ted Abresch, Nanette C Joyce, Venkatarman Viswanathan, Sivaprakasam Chidambaranathan, Douglas Biggar, Laura C McAdam, Jean K Mah, Mar Tulinius, Avital Cnaan, Lauren P Morgenroth, Robert Leshner, Carolina Tesi-Rocha, Mathula Thangarajh, Tina Duong, Andrew Kornberg, Monique Ryan, Yoram Nevo, Alberto Dubrovsky, Paula R Clemens, Hoda Abdel-Hamid, Anne M Connolly, Alan Pestronk, Jean Teasley, Tulio E Bertorini, Richard Webster, Hanna Kolski, Nancy Kuntz, Sherilyn W Driscoll, John B Bodensteiner, Jose Carlo, Ksenija Gorni, Timothy Lotze, John W Day, Peter Karachunski, Jean K Mah, Paula R Clemens, Michela Guglieri, Edward C Smith, Richard S Finkel, Mar Tulinius, Yoram Nevo, Monique M Ryan, Richard Webster, Diana Castro, Nancy L Kuntz, Craig M McDonald, Jesse M Damsker, Benjamin D Schwartz, Laurel J Mengle-Gaw, Stefan Jackowski, Georgia Stimpson, Deborah A Ridout, Vandana Ayyar-Gupta, Giovanni Baranello, Adnan Y Manzur, Francesco Muntoni, Heather Gordish-Dressman, Mika Leinonen, Leanne M Ward, Eric P Hoffman, Utkarsh J Dang, NorthStar UK Network and CINRG DNHS Investigators, Francesco Muntoni, Adnan Manzur, Giovanni Baranello, Stephanie Robb, Ros Quinlivan, Anna Sarkozy, Pinki Munot, Marion Main, Lianne Abbot, Volker Straub, Michela Guglieri, Chiara Bertolli, Anna Mayhew, Robert Muni-Lofra, Meredith James, Jassi Sodhi, Deepak Parasuraman, Zoya Alhaswani, Heather McMurchie, Rosanna Rabb, Anne-Marie Childs, Karen Pysden, Lindsey Pallant, Tiffany Small, Stefan Spinty, Rajesh Madhu, Alison Shillington, Sarah Gregson, Elizabeth Wraige, Heinz Jungbluth, Vasantha Gowda, Jennie Sheehan, Imelda Hughes, Sinead Warner, Emily Davies, Tracey Willis, Richa Kulshrestha, Nicholas Emery, Kate Strachan, Min Ong, Kay White, Kate Skone, Frances Gibbon, Bethan Parsons, Anirban Majumdar, Kayal Vijaykumar, Faye Mason, Claire Frimpong-Ansah, Karen Naismith, Julie Burslem, Iain Horrocks, Marina Di Marco, Sarah Brown, Sarah Williamson, Kirstie Spencer, Gabby Chow, Christian de Goede, Andrea Selley, Neil Thomas, Marjorie Illingworth, Michelle Greary, Jenni Palmer, Cathy White, Kate Greenfield, Grainne Nic Fhirleinn, Melanie Douglas, Sandya Tiraputhi, Nahin Hussain, Yvonne Julien, Gautam Ambegaonkar, Deepa Krishnakumar, Jacqui Taylor, Jane Tewnion, Elma Stephens, Saleel Chandratre, Sithara Ramdas, Hayley Ramjattan, Alex Baxter, Clare Eadie, Craig M McDonald, Erik K Henricson, R Ted Abresch, Nanette C Joyce, Venkatarman Viswanathan, Sivaprakasam Chidambaranathan, Douglas Biggar, Laura C McAdam, Jean K Mah, Mar Tulinius, Avital Cnaan, Lauren P Morgenroth, Robert Leshner, Carolina Tesi-Rocha, Mathula Thangarajh, Tina Duong, Andrew Kornberg, Monique Ryan, Yoram Nevo, Alberto Dubrovsky, Paula R Clemens, Hoda Abdel-Hamid, Anne M Connolly, Alan Pestronk, Jean Teasley, Tulio E Bertorini, Richard Webster, Hanna Kolski, Nancy Kuntz, Sherilyn W Driscoll, John B Bodensteiner, Jose Carlo, Ksenija Gorni, Timothy Lotze, John W Day, Peter Karachunski

Abstract

Importance: Vamorolone is a synthetic steroidal drug with potent anti-inflammatory properties. Initial open-label, multiple ascending dose-finding studies of vamorolone among boys with Duchenne muscular dystrophy (DMD) found significant motor function improvement after 6 months treatment in higher-dose (ie, ≥2.0 mg/kg/d) groups.

Objective: To investigate outcomes after 30 months of open-label vamorolone treatment.

Design, setting, and participants: This nonrandomized controlled trial was conducted by the Cooperative International Neuromuscular Research Group at 11 US and non-US study sites. Participants were 46 boys ages 4.5 to 7.5 years with DMD who completed the 6-month dose-finding study. Data were analyzed from July 2020 through November 2021.

Interventions: Participants were enrolled in a 24-month, long-term extension (LTE) study with vamorolone dose escalated to 2.0 or 6.0 mg/kg/d.

Main outcomes and measures: Change in time-to-stand (TTSTAND) velocity from dose-finding baseline to end of LTE study was the primary outcome. Efficacy assessments included timed function tests, 6-minute walk test, and NorthStar Ambulatory Assessment (NSAA). Participants with DMD treated with glucocorticoids from the Duchenne Natural History Study (DNHS) and NorthStar United Kingdom (NSUK) Network were matched and compared with participants in the LTE study receiving higher doses of vamorolone.

Results: Among 46 boys with DMD who completed the dose-finding study, 41 boys (mean [SD] age, 5.33 [0.96] years) completed the LTE study. Among 21 participants treated with higher-dose (ie, ≥2.0 mg/kg/d) vamorolone consistently throughout the 6-month dose-finding and 24-month LTE studies with data available at 30 months, there was a decrease in mean (SD) TTSTAND velocity from baseline to 30 months (0.206 [0.070] rises/s vs 0.189 (0.124) rises/s), which was not a statistically significant change (-0.011 rises/s; CI, -0.068 to 0.046 rises/s). There were no statistically significant differences between participants receiving higher-dose vamorolone and matched participants in the historical control groups receiving glucocorticoid treatment (75 patients in DNHS and 110 patients in NSUK) over a 2-year period in NSAA total score change (0.22 units vs NSUK; 95% CI, -4.48 to 4.04]; P = .92), body mass index z score change (0.002 vs DNHS SD/mo; 95% CI, -0.006 to 0.010; P = .58), or timed function test change. Vamorolone at doses up to 6.0 mg/kg/d was well tolerated, with 5 of 46 participants discontinuing prematurely and for reasons not associated with study drug. Participants in the DNHS treated with glucocorticoids had significant growth delay in comparison with participants treated with vamorolone who had stable height percentiles (0.37 percentile/mo; 95% CI, 0.23 to 0.52 percentile/mo) over time.

Conclusions and relevance: This study found that vamorolone treatment was not associated with a change in TTSTAND velocity from baseline to 30 months among boys with DMD aged 4 to 7 years at enrollment. Vamorolone was associated with maintenance of muscle strength and function up to 30 months, similar to standard of care glucocorticoid therapy, and improved height velocity compared with growth deceleration associated with glucocorticoid treatment, suggesting that vamorolone may be an attractive candidate for treatment of DMD.

Trial registration: ClinicalTrials.gov Identifier: NCT03038399.

Conflict of interest statement

Conflict of Interest Disclosures: Dr Mah reported receiving grants from ReveraGen BioPharma during the conduct of the study; grants from Pfizer, Italfarmaco, Sarepta, Catabasis, Roche, Biogen, Novartis, NS Pharma, PTC Therapeutics, and Alberta Children's Hospital Foundation; and personal fees from PTC Therapeutics, Biogen, and Roche outside the submitted work. Dr Clemens reported serving as a board member for Therapeutic Research in Neuromuscular Disorders Solutions (TRINDS); receiving grants from the National Institutes of Health (NIH), NS Pharma, Amicus, Sanofi, Spark, and Muscular Dystrophy Association; receiving research support through a donation from the Foundation to Eradicate Duchenne to the University of Pittsburgh; and receiving personal fees from Epirium during the conduct of the study outside the submitted work. Dr Guglieri reported receiving data management support from North Star Network during the conduct of the study; grants from the NIH, European Committee H2020, Duchenne Muscular Dystrophy UK, and Sarepta (funding through Translational Research in Europe: Assessment and Treatment of Neuromuscular Disorders); personal fees from Sarepta; and travel fees from Santhera outside the submitted work and serving as principal or chief investigator or clinical investigator for clinical trials sponsored by Pfizer, Italfarmaco, Summit, Santhera, Roche, and PTC Therapeutics. Dr Smith reported receiving grants for partial salary support from Reveragen BioPharma as principal investigator during the conduct of the study. Dr Finkel reported receiving grants from ReveraGen BioPharma during the conduct of the study and grants from Catabasis and Sarepta outside the submitted work. Dr Webster reported receiving research funding from ReveraGen BioPharma during the conduct of the study. Dr Kuntz reported receiving grants from Astellas Gene Therapies, Biogen, Genentech, Novartis, and Sarepta; personal fees from Biogen, Genentech, Novartis, and Sarepta; and data safety monitoring board services from Sarepta during the conduct of the study. Dr McDonald reported receiving grants from the NIH National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), US Department of Defense, National Institute on Disability Independent Living and Rehabilitation Research, and Parent Project Muscular Dystrophy and personal fees from Astellas, BioMarin, Epirium Bio (formerly Capricor), Catabasis, Entrada Therapeutics, Avidity Therapeutics, Edgewise Therapeutics, FibroGen, Hoffmann La Roche, Marathon, Pfizer, Santhera Pharmaceuticals, Saraepta Therapeutics, and PTC Therapeutics outside the submitted work. Dr Damsker reported being an employee and shareholder of ReveraGen BioPharma. Dr Schwartz reported receiving personal fees from ReveraGen Biopharma during the conduct of the study and personal fees from RegenXBio and TRINDS outside the submitted work. Dr Mengle-Gaw reported receiving personal fees from ReveraGen BioPharma during the conduct of the study and personal fees from RegenXBio outside the submitted work. Dr Baranello reported receiving personal fees from Roche, serving as principal investigator in clinical trials for Pfizer and Reveragen, receiving grants from Sarepta via the Dubowitz Neuromuscular Centre, and serving as an investigator in a clinical trial for NS Pharma outside the submitted work. Dr Manzur reported receiving grants from NorthStar Clinical Network during the conduct of the study and serving as clinical lead for Northstar Clinical Network UK. Dr Muntoni reported receiving grants from Muscular Dystrophy UK North Star Consortium during the conduct of the study; grants from the NIH Research Great Ormond Street Hospital-ICH Biomedical Research Centre, Genethon, and Sarepta; and personal fees from Dyne Therapeutics, Pfizer, and Sarepta outside the submitted work. Dr Gordish-Dressman reported receiving personal fees from TRINDS during the conduct of the study and co-owning TRINDS outside the submitted work. Dr Ward reported receiving grants from ReveraGen BioPharma via the Children's Hospital of Eastern Ontario (CHEO) Research Institute during the conduct of the study and grants from Amgen, Novartis, and PTC Therapeutics via the CHEO Research Institute outside the submitted work. Dr Hoffman reported receiving salary as chief executive officer and owning stock from ReveraGen BioPharma, serving as a board member for TRINDS, receiving grants from the NIH National Institute of Neurological Disorders and Stroke during the conduct of the study; serving as vice president and owning stock for Agada BioSciences outside the submitted work; and owning a patent issued to ReveraGen BioPharma. Dr Dang reported receiving grants from the Foundation to Eradicate Duchenne and personal fees from ReveraGen BioPharma during the conduct of the study and grants from the NIH NIAMS outside the submitted work. No other disclosures were reported.

Figures

Figure 1.. Flowchart of Vamorolone Long-term Extension…
Figure 1.. Flowchart of Vamorolone Long-term Extension (LTE) Study
VBP indicates vamorolone. aTreating physicians were permitted to up-titrate or down-titrate dose according to clinical judgement. There were 3 participants who ended the study at an intermediate dose of 4.0 mg/kg/d.
Figure 2.. Efficacy of Vamorolone in Motor…
Figure 2.. Efficacy of Vamorolone in Motor Function Assessments by High vs Low Starting Dose
6MWT indicates 6-minute walk test; high, a starting dose of 2.0 mg/kg/d or greater; low, a starting dose of less than 2.0 mg/kg/d; points, means; whiskers, standard error of the mean. Mean estimated differences between participants treated at higher dose and low dose, as well as P values, are provided at 6 months (ie, prior to LTE) and 30 months (ie, at conclusion of LTE).
Figure 3.. Outcome Comparisons of Vamorolone Long-term…
Figure 3.. Outcome Comparisons of Vamorolone Long-term Extension (LTE) and Duchenne Natural History (DNHS) Study Cohorts
BMI indicates body mass index. Trajectories based on model estimates, along with 95% CIs, are plotted for an individual with fixed baseline values.

References

    1. Mah JK, Korngut L, Dykeman J, Day L, Pringsheim T, Jette N. A systematic review and meta-analysis on the epidemiology of Duchenne and Becker muscular dystrophy. Neuromuscul Disord. 2014;24(6):482-491. doi:10.1016/j.nmd.2014.03.008
    1. Hoffman EP, Brown RH Jr, Kunkel LM. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell. 1987;51(6):919-928. doi:10.1016/0092-8674(87)90579-4
    1. Evans NP, Misyak SA, Robertson JL, Bassaganya-Riera J, Grange RW. Immune-mediated mechanisms potentially regulate the disease time-course of Duchenne muscular dystrophy and provide targets for therapeutic intervention. PM R. 2009;1(8):755-768. doi:10.1016/j.pmrj.2009.04.010
    1. Chen YW, Nagaraju K, Bakay M, et al. . Early onset of inflammation and later involvement of TGFbeta in Duchenne muscular dystrophy. Neurology. 2005;65(6):826-834. doi:10.1212/01.wnl.0000173836.09176.c4
    1. Rosenberg AS, Puig M, Nagaraju K, et al. . Immune-mediated pathology in Duchenne muscular dystrophy. Sci Transl Med. 2015;7(299):299rv4. doi:10.1126/scitranslmed.aaa7322
    1. Kumar A, Boriek AM. Mechanical stress activates the nuclear factor-kappaB pathway in skeletal muscle fibers: a possible role in Duchenne muscular dystrophy. FASEB J. 2003;17(3):386-396. doi:10.1096/fj.02-0542com
    1. Acharyya S, Villalta SA, Bakkar N, et al. . Interplay of IKK/NF-kappaB signaling in macrophages and myofibers promotes muscle degeneration in Duchenne muscular dystrophy. J Clin Invest. 2007;117(4):889-901. doi:10.1172/JCI30556
    1. Dogra C, Changotra H, Wergedal JE, Kumar A. Regulation of phosphatidylinositol 3-kinase (PI3K)/Akt and nuclear factor-kappa B signaling pathways in dystrophin-deficient skeletal muscle in response to mechanical stretch. J Cell Physiol. 2006;208(3):575-585. doi:10.1002/jcp.20696
    1. Pahl HL. Activators and target genes of Rel/NF-kappaB transcription factors. Oncogene. 1999;18(49):6853-6866. doi:10.1038/sj.onc.1203239
    1. Spencer MJ, Montecino-Rodriguez E, Dorshkind K, Tidball JG. Helper (CD4(+)) and cytotoxic (CD8(+)) T cells promote the pathology of dystrophin-deficient muscle. Clin Immunol. 2001;98(2):235-243. doi:10.1006/clim.2000.4966
    1. Spencer MJ, Tidball JG. Do immune cells promote the pathology of dystrophin-deficient myopathies? Neuromuscul Disord. 2001;11(6-7):556-564. doi:10.1016/S0960-8966(01)00198-5
    1. Griggs RC, Moxley RT III, Mendell JR, et al. ; Clinical Investigation of Duchenne Dystrophy Group . Prednisone in Duchenne dystrophy: a randomized, controlled trial defining the time course and dose response. Arch Neurol. 1991;48(4):383-388. doi:10.1001/archneur.1991.00530160047012
    1. Mendell JR, Moxley RT, Griggs RC, et al. . Randomized, double-blind six-month trial of prednisone in Duchenne’s muscular dystrophy. N Engl J Med. 1989;320(24):1592-1597. doi:10.1056/NEJM198906153202405
    1. Malik V, Rodino-Klapac LR, Mendell JR. Emerging drugs for Duchenne muscular dystrophy. Expert Opin Emerg Drugs. 2012;17(2):261-277. doi:10.1517/14728214.2012.691965
    1. Matthews E, Brassington R, Kuntzer T, Jichi F, Manzur AY. Corticosteroids for the treatment of Duchenne muscular dystrophy. Cochrane Database Syst Rev. 2016;2016(5):CD003725. doi:10.1002/14651858.CD003725.pub4
    1. Bello L, Gordish-Dressman H, Morgenroth LP, et al. ; CINRG Investigators . Prednisone/prednisolone and deflazacort regimens in the CINRG Duchenne Natural History Study. Neurology. 2015;85(12):1048-1055. doi:10.1212/WNL.0000000000001950
    1. Kauh E, Mixson L, Malice MP, et al. . Prednisone affects inflammation, glucose tolerance, and bone turnover within hours of treatment in healthy individuals. Eur J Endocrinol. 2012;166(3):459-467. doi:10.1530/EJE-11-0751
    1. Schakman O, Kalista S, Barbé C, Loumaye A, Thissen JP. Glucocorticoid-induced skeletal muscle atrophy. Int J Biochem Cell Biol. 2013;45(10):2163-2172. doi:10.1016/j.biocel.2013.05.036
    1. Pane M, Mazzone ES, Sivo S, et al. . Long term natural history data in ambulant boys with Duchenne muscular dystrophy: 36-month changes. PLoS One. 2014;9(10):e108205. doi:10.1371/journal.pone.0108205
    1. Kim S, Zhu Y, Romitti PA, et al. ; MD STARnet . Associations between timing of corticosteroid treatment initiation and clinical outcomes in Duchenne muscular dystrophy. Neuromuscul Disord. 2017;27(8):730-737. doi:10.1016/j.nmd.2017.05.019
    1. Connolly AM, Zaidman CM, Golumbek PT, et al. ; MDA DMD Clinical Research Network . Twice-weekly glucocorticosteroids in infants and young boys with Duchenne muscular dystrophy. Muscle Nerve. 2019;59(6):650-657. doi:10.1002/mus.26441
    1. Reeves EKM, Hoffman EP, Nagaraju K, Damsker JM, McCall JM. VBP15: preclinical characterization of a novel anti-inflammatory delta 9,11 steroid. Bioorg Med Chem. 2013;21(8):2241-2249. doi:10.1016/j.bmc.2013.02.009
    1. Liu X, Wang Y, Gutierrez JS, et al. . Disruption of a key ligand-H-bond network drives dissociative properties in vamorolone for Duchenne muscular dystrophy treatment. Proc Natl Acad Sci U S A. 2020;117(39):24285-24293. doi:10.1073/pnas.2006890117
    1. Heier CR, Yu Q, Fiorillo AA, et al. . Vamorolone targets dual nuclear receptors to treat inflammation and dystrophic cardiomyopathy. Life Sci Alliance. 2019;2(1):1-16. doi:10.26508/lsa.201800186
    1. Conklin LS, Damsker JM, Hoffman EP, et al. . Phase IIa trial in Duchenne muscular dystrophy shows vamorolone is a first-in-class dissociative steroidal anti-inflammatory drug. Pharmacol Res. 2018;136:140-150. doi:10.1016/j.phrs.2018.09.007
    1. Hoffman EP, Schwartz BD, Mengle-Gaw LJ, et al. ; Cooperative International Neuromuscular Research Group . Vamorolone trial in Duchenne muscular dystrophy shows dose-related improvement of muscle function. Neurology. 2019;93(13):e1312-e1323. doi:10.1212/WNL.0000000000008168
    1. Smith EC, Conklin LS, Hoffman EP, et al. ; CINRG VBP15 and DNHS Investigators . Efficacy and safety of vamorolone in Duchenne muscular dystrophy: an 18-month interim analysis of a non-randomized open-label extension study. PLoS Med. 2020;17(9):e1003222. doi:10.1371/journal.pmed.1003222
    1. McDonald CM, Henricson EK, Abresch RT, et al. ; CINRG Investigators . The cooperative international neuromuscular research group Duchenne natural history study—a longitudinal investigation in the era of glucocorticoid therapy: design of protocol and the methods used. Muscle Nerve. 2013;48(1):32-54. doi:10.1002/mus.23807
    1. Henricson EK, Abresch RT, Cnaan A, et al. ; CINRG Investigators . The cooperative international neuromuscular research group Duchenne natural history study: glucocorticoid treatment preserves clinically meaningful functional milestones and reduces rate of disease progression as measured by manual muscle testing and other commonly used clinical trial outcome measures. Muscle Nerve. 2013;48(1):55-67. doi:10.1002/mus.23808
    1. McDonald CM, Henricson EK, Abresch RT, et al. ; CINRG Investigators . Long-term effects of glucocorticoids on function, quality of life, and survival in patients with Duchenne muscular dystrophy: a prospective cohort study. Lancet. 2018;391(10119):451-461. doi:10.1016/S0140-6736(17)32160-8
    1. Iacus SM, King G, Porro G. Multivariate matching methods that are monotonic imbalance bounding. J Am Stat Assoc. 2011;106(493):345-361. doi:10.1198/jasa.2011.tm09599
    1. Iacus SM, King G, Porro G, Nielsen R. CEM: coarsened exact matching. The Comprehensive R Archive Network. Accessed December 8, 2021.
    1. Ricotti V, Ridout DA, Pane M, et al. ; UK NorthStar Clinical Network . The NorthStar Ambulatory Assessment in Duchenne muscular dystrophy: considerations for the design of clinical trials. J Neurol Neurosurg Psychiatry. 2016;87(2):149-155. doi:10.1136/jnnp-2014-309405
    1. Centers for Disease Control and Prevention . Percentile data files with LMS values. National Center for Health Statistics. Accessed December 8, 2021.
    1. Brooke MH, Griggs RC, Mendell JR, Fenichel GM, Shumate JB, Pellegrino RJ. Clinical trial in Duchenne dystrophy: I. the design of the protocol. Muscle Nerve. 1981;4(3):186-197. doi:10.1002/mus.880040304
    1. McDonald CM, Henricson EK, Han JJ, et al. . The 6-minute walk test as a new outcome measure in Duchenne muscular dystrophy. Muscle Nerve. 2010;41(4):500-510. doi:10.1002/mus.21544
    1. Scott E, Eagle M, Mayhew A, et al. ; North Star Clinical Network for Paediatric Neuromuscular Disease . Development of a functional assessment scale for ambulatory boys with Duchenne muscular dystrophy. Physiother Res Int. 2012;17(2):101-109. doi:10.1002/pri.520
    1. Escolar DM, Henricson EK, Mayhew J, et al. . Clinical evaluator reliability for quantitative and manual muscle testing measures of strength in children. Muscle Nerve. 2001;24(6):787-793. doi:10.1002/mus.1070
    1. Dang UJ, Ziemba M, Clemens PR, Hathout Y, Conklin LS, Hoffman EP; CINRG Vamorolone 002/003 Investigators . Serum biomarkers associated with baseline clinical severity in young steroid-naïve Duchenne muscular dystrophy boys. Hum Mol Genet. 2020;29(15):2481-2495. doi:10.1093/hmg/ddaa132
    1. McDonald CM, McDonald DA, Bagley A, et al. . Relationship between clinical outcome measures and parent proxy reports of health-related quality of life in ambulatory children with Duchenne muscular dystrophy. J Child Neurol. 2010;25(9):1130-1144. doi:10.1177/0883073810371509
    1. McDonald CM, Campbell C, Torricelli RE, et al. ; Clinical Evaluator Training Group; ACT DMD Study Group . Ataluren in patients with nonsense mutation Duchenne muscular dystrophy (ACT DMD): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2017;390(10101):1489-1498. doi:10.1016/S0140-6736(17)31611-2
    1. Fay MP, Shaw PA. Exact and asymptotic weighted logrank tests for interval censored data: the interval R package. J Stat Softw. 2010;36(2):io2. doi:10.18637/jss.v036.i02
    1. Bates D, Mächler M, Bolker BM, Walker S. Fitting linear mixed-effects models using lme4. J Stat Softw. 2015;67(1):1-48. doi:10.18637/jss.v067.i01
    1. Kuznetsova A, Brockhoff PB, Christensen RHB. lmerTest Package: Tests in Linear Mixed Effects Models. J Stat Softw. 2017;82(13). doi:10.18637/jss.v082.i13
    1. Lüdecke D. sjPlot—data visualization for statistics in social science. GitHub. Accessed December 8, 2021.
    1. Breheny P, Burchett W.. Visualization of regression models using visreg. R J. 2017;9(2):56-71. doi:10.32614/RJ-2017-046
    1. Raabe S. Vistime: pretty timelines in R. R package version 1.2.1. The Comprehensive R Archive Network. Accessed December 8, 2021.
    1. Arora H, Willcocks RJ, Lott DJ, et al. . Longitudinal timed function tests in Duchenne muscular dystrophy: ImagingDMD cohort natural history. Muscle Nerve. 2018;58(5):631-638. doi:10.1002/mus.26161
    1. Ward LM, Weber DR. Growth, pubertal development, and skeletal health in boys with Duchenne muscular dystrophy. Curr Opin Endocrinol Diabetes Obes. 2019;26(1):39-48. doi:10.1097/MED.0000000000000456
    1. Joseph S, Wang C, Bushby K, et al. ; UK NorthStar Clinical Network . Fractures and linear growth in a nationwide cohort of boys with Duchenne muscular dystrophy with and without glucocorticoid treatment: results from the UK NorthStar database. JAMA Neurol. 2019;76(6):701-709. doi:10.1001/jamaneurol.2019.0242
    1. Wood CL, Straub V, Guglieri M, Bushby K, Cheetham T. Short stature and pubertal delay in Duchenne muscular dystrophy. Arch Dis Child. 2016;101(1):101-106. doi:10.1136/archdischild-2015-308654
    1. Griggs RC, Herr BE, Reha A, et al. . Corticosteroids in Duchenne muscular dystrophy: major variations in practice. Muscle Nerve. 2013;48(1):27-31. doi:10.1002/mus.23831

Source: PubMed

3
Sottoscrivi