Dual-energy X-ray absorptiometry measures of lean body mass as a biomarker for progression in boys with Duchenne muscular dystrophy

Sarah P Sherlock, Jeffrey Palmer, Kathryn R Wagner, Hoda Z Abdel-Hamid, Cuixia Tian, Jean K Mah, Francesco Muntoni, Michela Guglieri, Russell J Butterfield, Lawrence Charnas, Shannon Marraffino, Sarah P Sherlock, Jeffrey Palmer, Kathryn R Wagner, Hoda Z Abdel-Hamid, Cuixia Tian, Jean K Mah, Francesco Muntoni, Michela Guglieri, Russell J Butterfield, Lawrence Charnas, Shannon Marraffino

Abstract

We evaluated whether whole-body dual-energy X-ray absorptiometry (DXA) measures of lean body mass can be used as biomarkers for disease progression and treatment effects in patients with Duchenne muscular dystrophy. This post hoc analysis utilized data from a randomized, 2-period study of domagrozumab versus placebo in 120 ambulatory boys with DMD. DXA measures of lean body mass were obtained from the whole body (excluding head), arms, legs and appendicular skeleton at baseline and every 16 weeks. Treatment effects on DXA measures for domagrozumab versus placebo were assessed at Week 49. At Week 49, domagrozumab statistically significantly increased lean body mass versus placebo in the appendicular skeleton (p = 0.050) and arms (p < 0.001). The relationship between lean body mass at Week 49 and functional endpoints at Week 97 was evaluated. Changes in lean body mass at Week 49 in all regions except arms were significantly correlated with percent change from baseline in 4-stair climb (4SC) at Week 97. DXA-derived percent lean mass at Week 49 also correlated with 4SC and North Star Ambulatory Assessment at Week 97. These data indicate that whole-body DXA measures can be used as biomarkers for treatment effects and disease progression in patients with DMD, and warrant further investigation.Trial registration: ClinicalTrials.gov, NCT02310763; registered 8 December 2014.

Conflict of interest statement

SPS, JP, LC, and SM are employees of and hold stock or stock options in Pfizer. KRW is an employee of and holds stock in F. Hoffmann-La Roche Ltd. HZA-H has received research support from and served on advisory boards for Sarepta Therapeutics, Biogen, NS Pharma, and AveXis/Novartis. CT has been site investigator for clinical trials with Pfizer, AveXis/Novartis, Catabasis, WaVe, Roche, PTC Therapeutics, FibroGen, Capricor, Santhera, Sarepta Therapeutics, and Summit. JKM has received research support for clinical trials from Pfizer, Italfarmaco, Sarepta Therapeutics, Catabasis, Roche, Biogen, Novartis, NS Pharma, PTC Therapeutics, and ReveraGen; and consultant fees from PTC Therapeutics, Roche, and Biogen. FM is a member of the Pfizer Rare Disease Scientific Advisory Board; received grant support from Biogen and Sarepta Therapeutics; and received consultant fees and/or speakers’ honoraria from Novartis, Biogen, Roche, Dyne Therapeutics, and PTC Therapeutics. MG has served as chair for a ReveraGen study; collaborated on research with ReveraGen and Sarepta Therapeutics; acted as principal investigator for clinical trials sponsored by Pfizer, Italfarmaco, Santhera Therapeutics, ReveraGen, Dynacure, Roche, PTC Therapeutics, and Summit; participated in advisory boards for Pfizer, NS Pharma, and Dyne (consultancies through Newcastle University); and performed consultancy work (speaker) for Sarepta Therapeutics. RJB has received funding via contracts for clinical trials from AveXis/Novartis, PTC Therapeutics, Sarepta Therapeutics, Pfizer, and Biogen; and has served on scientific advisory boards for Sarepta Therapeutics, Biogen, AveXis/Novartis, and Pfizer.

© 2022. The Author(s).

Figures

Figure 1
Figure 1
Representative DXA scan. A visual representation of fat mass, lean body mass and bone mass is shown for a participant (a) before treatment and (b) at the Week 49 visit. Cutlines are placed on the DXA scan to separate regions of interest for analysis (c). Primary regions for analysis included arms (left and right combined), legs (left and right combined), appendicular skeleton (arms and leg as a single region of interest) and total body excluding the head. Cutlines were placed by a trained analyst and reviewed by a radiologist prior to finalizing read results. DXA, Dual-energy X-ray absorptiometry.
Figure 2
Figure 2
Change from baseline in DXA lean body mass. Least-squares mean change from baseline in lean body mass at Weeks 17, 33 and 49 for (a) whole body, (b) appendicular skeleton, (c) total legs and (d) total arms; (e) shows percent lean mass. DXA, Dual-energy X-ray absorptiometry.

References

    1. Duan D, Goemans N, Takeda S, Mercuri E, Aartsma-Rus A. Duchenne muscular dystrophy. Nat. Rev. Dis. Primers. 2021;7:13. doi: 10.1038/s41572-021-00248-3.
    1. Birnkrant DJ, et al. Diagnosis and management of Duchenne muscular dystrophy, part 2: Respiratory, cardiac, bone health, and orthopaedic management. Lancet Neurol. 2018;17:347–361. doi: 10.1016/S1474-4422(18)30025-5.
    1. Parent Project Muscular Dystrophy. Care Guidelines by Stage: Loss of Ambulation, (2019).
    1. Birnkrant DJ, et al. Diagnosis and management of Duchenne muscular dystrophy, part 1: Diagnosis, and neuromuscular, rehabilitation, endocrine, and gastrointestinal and nutritional management. Lancet Neurol. 2018;17:251–267. doi: 10.1016/S1474-4422(18)30024-3.
    1. Finkel RS, et al. Phase 2a study of ataluren-mediated dystrophin production in patients with nonsense mutation Duchenne muscular dystrophy. PLoS ONE. 2013;8:e81302. doi: 10.1371/journal.pone.0081302.
    1. Frank DE, et al. Increased dystrophin production with golodirsen in patients with Duchenne muscular dystrophy. Neurology. 2020;94:e2270–e2282. doi: 10.1212/WNL.0000000000009233.
    1. Mendell JR, et al. Longitudinal effect of eteplirsen versus historical control on ambulation in Duchenne muscular dystrophy. Ann. Neurol. 2016;79:257–271. doi: 10.1002/ana.24555.
    1. Clemens PR, et al. Safety, tolerability, and efficacy of viltolarsen in boys with Duchenne muscular dystrophy amenable to Exon 53 skipping: A phase 2 randomized clinical trial. JAMA Neurol. 2020;77:982–991. doi: 10.1001/jamaneurol.2020.1264.
    1. Wagner KR, et al. Safety, tolerability, and pharmacokinetics of casimersen in patients with Duchenne muscular dystrophy amenable to exon 45 skipping: A randomized, double-blind, placebo-controlled, dose-titration trial. Muscle Nerve. 2021;64:285–292. doi: 10.1002/mus.27347.
    1. Goemans N, et al. Ambulatory capacity and disease progression as measured by the 6-minute-walk-distance in Duchenne muscular dystrophy subjects on daily corticosteroids. Neuromuscul. Disord. 2013;23:618–623. doi: 10.1016/j.nmd.2013.05.006.
    1. Pane M, et al. 6 minute walk test in Duchenne MD patients with different mutations: 12 month changes. PLoS ONE. 2014;9:e83400. doi: 10.1371/journal.pone.0083400.
    1. Lorente Ramos, R. M. et al. Dual energy X-ray absorptimetry: Fundamentals, methodology, and clinical applications. Radiologia. 54, 410–423 10.1016/j.rx.2011.09.023 (2012).
    1. Palmieri GM, Bertorini TE, Griffin JW, Igarashi M, Karas JG. Assessment of whole body composition with dual energy x-ray absorptiometry in Duchenne muscular dystrophy: Correlation of lean body mass with muscle function. Muscle Nerve. 1996;19:777–779. doi: 10.1002/(SICI)1097-4598(199606)19:6<777::AID-MUS15>;2-I.
    1. Skalsky AJ, Han JJ, Abresch RT, Shin CS, McDonald CM. Assessment of regional body composition with dual-energy X-ray absorptiometry in Duchenne muscular dystrophy: Correlation of regional lean mass and quantitative strength. Muscle Nerve. 2009;39:647–651. doi: 10.1002/mus.21212.
    1. Summer SS, et al. Age-related changes in appendicular lean mass in males with Duchenne muscular dystrophy: A retrospective review. Muscle Nerve. 2021;63:231–238. doi: 10.1002/mus.27107.
    1. Vuillerot C, et al. Influence of a two-year steroid treatment on body composition as measured by dual X-ray absorptiometry in boys with Duchenne muscular dystrophy. Neuromuscul. Disord. 2014;24:467–473. doi: 10.1016/j.nmd.2014.03.002.
    1. Wagner KR, et al. Randomized phase 2 trial and open-label extension of domagrozumab in Duchenne muscular dystrophy. Neuromuscul. Disord. 2020;30:492–502. doi: 10.1016/j.nmd.2020.05.002.
    1. Wagner, K. R. et al. Corrigendum to "Randomized phase 2 trial and open-label extension of domagrozumab in Duchenne muscular dystrophy" [Neuromuscul. Disord.30, 492–502 (2020)]. Neuromuscul. Disord.31, 167–168 10.1016/j.nmd.2021.01.001 (2021).
    1. Wagner KR. The elusive promise of myostatin inhibition for muscular dystrophy. Curr. Opin. Neurol. 2020;33:621–628. doi: 10.1097/WCO.0000000000000853.
    1. St Andre M, et al. A mouse anti-myostatin antibody increases muscle mass and improves muscle strength and contractility in the mdx mouse model of Duchenne muscular dystrophy and its humanized equivalent, domagrozumab (PF-06252616), increases muscle volume in cynomolgus monkeys. Skelet. Muscle. 2017;7:25. doi: 10.1186/s13395-017-0141-y.
    1. Bhattacharya I, et al. Safety, tolerability, pharmacokinetics, and pharmacodynamics of domagrozumab (PF-06252616), an antimyostatin monoclonal antibody, in healthy subjects. Clin. Pharmacol. Drug Dev. 2018;7:484–497. doi: 10.1002/cpdd.386.
    1. Sherlock SP, et al. Quantitative magnetic resonance imaging measures as biomarkers of disease progression in boys with Duchenne muscular dystrophy: A phase 2 trial of domagrozumab. J Neurol. 2022;269:4421–4435. doi: 10.1007/s00415-022-11084-0.
    1. Rybalka, E. et al. The failed clinical story of myostatin inhibitors against Duchenne muscular dystrophy: Exploring the biology behind the battle. Cells10.3390/cells9122657 (2020).
    1. Willcocks RJ, et al. Multicenter prospective longitudinal study of magnetic resonance biomarkers in a large Duchenne muscular dystrophy cohort. Ann. Neurol. 2016;79:535–547. doi: 10.1002/ana.24599.
    1. Willcocks RJ, et al. Magnetic resonance imaging of the proximal upper extremity musculature in boys with Duchenne muscular dystrophy. J. Neurol. 2017;264:64–71. doi: 10.1007/s00415-016-8311-0.
    1. Szigyarto CA, Spitali P. Biomarkers of Duchenne muscular dystrophy: Current findings. Degener. Neurol. Neuromuscul. Dis. 2018;8:1–13. doi: 10.2147/DNND.S121099.
    1. Doulgeraki AE, et al. Body composition of patients with Duchenne muscular dystrophy: The Greek experience. Acta Neurol. Belg. 2016;116:565–572. doi: 10.1007/s13760-015-0582-1.
    1. McDonald CM, et al. Body composition and water compartment measurements in boys with Duchenne muscular dystrophy. Am. J. Phys. Med. Rehabil. 2005;84:483–491. doi: 10.1097/01.phm.0000166880.91117.04.
    1. Zanardi MC, et al. Body composition and energy expenditure in Duchenne muscular dystrophy. Eur. J. Clin. Nutr. 2003;57:273–278. doi: 10.1038/sj.ejcn.1601524.
    1. Pietrobelli A, Formica C, Wang Z, Heymsfield SB. Dual-energy X-ray absorptiometry body composition model: Review of physical concepts. Am. J. Physiol. 1996;271:E941–951. doi: 10.1152/ajpendo.1996.271.6.E941.
    1. Buckinx F, et al. Pitfalls in the measurement of muscle mass: A need for a reference standard. J. Cachexia Sarcopenia Muscle. 2018;9:269–278. doi: 10.1002/jcsm.12268.
    1. Godi C, et al. Longitudinal MRI quantification of muscle degeneration in Duchenne muscular dystrophy. Ann. Clin. Transl. Neurol. 2016;3:607–622. doi: 10.1002/acn3.319.
    1. Ropars J, et al. Muscle MRI: A biomarker of disease severity in Duchenne muscular dystrophy? A systematic review. Neurology. 2020;94:117–133. doi: 10.1212/WNL.0000000000008811.
    1. Campbell C, et al. Myostatin inhibitor ACE-031 treatment of ambulatory boys with Duchenne muscular dystrophy: Results of a randomized, placebo-controlled clinical trial. Muscle Nerve. 2017;55:458–464. doi: 10.1002/mus.25268.
    1. Hanna MG, et al. Safety and efficacy of intravenous bimagrumab in inclusion body myositis (RESILIENT): A randomised, double-blind, placebo-controlled phase 2b trial. Lancet Neurol. 2019;18:834–844. doi: 10.1016/S1474-4422(19)30200-5.
    1. Pietrobelli A, Wang Z, Formica C, Heymsfield SB. Dual-energy X-ray absorptiometry: Fat estimation errors due to variation in soft tissue hydration. Am. J. Physiol. 1998;274:E808–816. doi: 10.1152/ajpendo.1998.274.5.E808.

Source: PubMed

3
Suscribir