Effect of Combination l-Citrulline and Metformin Treatment on Motor Function in Patients With Duchenne Muscular Dystrophy: A Randomized Clinical Trial

Patricia Hafner, Ulrike Bonati, Andrea Klein, Daniela Rubino, Vanya Gocheva, Simone Schmidt, Jonas Schroeder, Günther Bernert, Vincent Laugel, Maja Steinlin, Andrea Capone, Monika Gloor, Oliver Bieri, Lars G Hemkens, Benjamin Speich, Thomas Zumbrunn, Nuri Gueven, Dirk Fischer, Patricia Hafner, Ulrike Bonati, Andrea Klein, Daniela Rubino, Vanya Gocheva, Simone Schmidt, Jonas Schroeder, Günther Bernert, Vincent Laugel, Maja Steinlin, Andrea Capone, Monika Gloor, Oliver Bieri, Lars G Hemkens, Benjamin Speich, Thomas Zumbrunn, Nuri Gueven, Dirk Fischer

Abstract

Importance: Nitric oxide precursors, such as the amino acid l-arginine and the biguanide antidiabetic drug metformin, have been associated with metabolism and muscle function in patients with Duchenne muscular dystrophy (DMD). The treatment of DMD remains an unmet medical need.

Objective: To evaluate the benefits and harms of a combination of l-citrulline and metformin treatment among patients with DMD.

Design, setting, and participants: A single-center randomized double-blind placebo-controlled parallel-group clinical trial was conducted between December 12, 2013, and March 30, 2016, at the University Children's Hospital Basel in Switzerland. A total of 47 ambulant male patients aged 6.5 to 10 years with genetically confirmed DMD were recruited locally and from the patient registries of Switzerland, Germany, Austria, and France. Data were analyzed from April 6, 2016, to September 5, 2019.

Interventions: Patients in the treatment group received 2500 mg of l-citrulline and 250 mg of metformin (combination therapy) 3 times a day for 26 weeks compared with patients in the control group, who received placebo.

Main outcomes and measures: The primary end point was the change in transfer and standing posture, as assessed by the first dimension of the Motor Function Measure, version 32, from baseline to week 26. Secondary end points included assessments of timed function, quantitative muscle force, biomarkers for muscle necrosis, and adverse events. The 2 prespecified subgroups comprised patients who were able to walk 350 m or more in 6 minutes (stable subgroup) and patients who were not able to walk 350 m in 6 minutes (unstable subgroup) at baseline.

Results: Among 49 ambulant male children with DMD who were screened for eligibility, 47 patients with a mean (SD) age of 8.2 (1.1) years were randomized to a treatment group receiving combination therapy (n = 23) or a control group receiving placebo (n = 24), and 45 patients completed the study. No significant differences between groups were found in the results of timed function and muscle force tests for overall, proximal and axial, and distal motor function. Among patients receiving combination therapy, the Motor Function Measure first dimension subscore decrease was 5.5% greater than that of patients receiving placebo (95% CI, -1.0% to 12.1%; P = .09). The administration of combination therapy had significantly favorable effects on the first dimension subscore decrease among the 29 patients in the stable subgroup (6.7%; 95% CI, 0.9%-12.6%; P = .03) but not among the 15 patients in the unstable subgroup (3.9%; 95% CI, -13.2% to 20.9%; P = .63). Overall, the treatment was well tolerated with only mild adverse effects.

Conclusions and relevance: Treatment with combination therapy was not associated with an overall reduction in motor function decline among ambulant patients with DMD; however, a reduction in motor function decline was observed among the stable subgroup of patients treated with combination therapy. The statistically nonsignificant difference of distal motor function in favor of combination therapy and the reduced degeneration of muscle tissue appear to support the treatment concept, but the study may have lacked sufficient statistical power. Further research exploring this treatment option with a greater number of patients is warranted.

Trial registration: ClinicalTrials.gov identifier: NCT01995032.

Conflict of interest statement

Conflict of Interest Disclosures: Dr Laugel reported receiving personal fees from Biogen, PTC Therapeutics, AveXis, Roche, and Sarepta Therapeutics outside the submitted work. Dr Gueven reported receiving grants and personal fees from Santhera Pharmaceuticals outside the submitted work. Dr Fischer reported receiving grants from the Swiss National Science Foundation and the Thomi-Hopf-Stiftung Foundation during the conduct of the study. No other disclosures were reported.

Figures

Figure 1.. CONSORT Flow Diagram
Figure 1.. CONSORT Flow Diagram
aIndicates the same patient.
Figure 2.. Effect Sizes for Primary End…
Figure 2.. Effect Sizes for Primary End Point and Selected Secondary End Points
In end points for which a decrease indicated an improvement of a test result, a sign reversal was performed to allow comparison. AA indicates amino acids; BCAA, branch chain amino acids; Cit/Met, citrulline and metformin combination therapy; D1, first dimension of the MFM, referring to transfer and standing posture; EAA, essential amino acids; FF, fat fraction; MFM, Motor Function Measure; NEAA, nonessential amino acids; qMRI, quantitative magnetic resonance imaging; 6-MWD, 6-minute walking distance; and T2, transverse weighted relaxation time.

References

    1. Mendell JR, Shilling C, Leslie ND, et al. . Evidence-based path to newborn screening for Duchenne muscular dystrophy. Ann Neurol. 2012;71(3):-. doi:10.1002/ana.23528
    1. Kuznetsov AV, Winkler K, Wiedemann FR, von Bossanyi P, Dietzmann K, Kunz WS. Impaired mitochondrial oxidative phosphorylation in skeletal muscle of the dystrophin-deficient mdx mouse. Mol Cell Biochem. 1998;183(1-2):87-96. doi:10.1023/A:1006868130002
    1. Braun U, Paju K, Eimre M, et al. . Lack of dystrophin is associated with altered integration of the mitochondria and ATPases in slow-twitch muscle cells of MDX mice. Biochim Biophys Acta. 2001;1505(2-3):258-270. doi:10.1016/S0005-2728(01)00172-4
    1. Brenman JE, Chao DS, Xia H, Aldape K, Bredt DS. Nitric oxide synthase complexed with dystrophin and absent from skeletal muscle sarcolemma in Duchenne muscular dystrophy. Cell. 1995;82(5):743-752. doi:10.1016/0092-8674(95)90471-9
    1. Millay DP, Sargent MA, Osinska H, et al. . Genetic and pharmacologic inhibition of mitochondrial-dependent necrosis attenuates muscular dystrophy. Nat Med. 2008;14(4):442-447. doi:10.1038/nm1736
    1. Ryder S, Leadley RM, Armstrong N, et al. . The burden, epidemiology, costs and treatment for Duchenne muscular dystrophy: an evidence review. Orphanet J Rare Dis. 2017;12(1):79. doi:10.1186/s13023-017-0631-3
    1. Ljubicic V, Jasmin BJ. Metformin increases peroxisome proliferator-activated receptor γ co-activator-1α and utrophin a expression in dystrophic skeletal muscle. Muscle Nerve. 2015;52(1):139-142. doi:10.1002/mus.24692
    1. Mantuano P, Sanarica F, Conte E, et al. . Effect of a long-term treatment with metformin in dystrophic mdx mice: a reconsideration of its potential clinical interest in Duchenne muscular dystrophy. Biochem Pharmacol. 2018;154:89-103. doi:10.1016/j.bcp.2018.04.022
    1. Castillo L, Chapman TE, Sanchez M, et al. . Plasma arginine and citrulline kinetics in adults given adequate and arginine-free diets. Proc Natl Acad Sci U S A. 1993;90(16):7749-7753. doi:10.1073/pnas.90.16.7749
    1. Moinard C, Nicolis I, Neveux N, Darquy S, Bénazeth S, Cynober L. Dose-ranging effects of citrulline administration on plasma amino acids and hormonal patterns in healthy subjects: the Citrudose pharmacokinetic study. Br J Nutr. 2008;99(4):855-862. doi:10.1017/S0007114507841110
    1. Osowska S, Moinard C, Neveux N, Loï C, Cynober L. Citrulline increases arginine pools and restores nitrogen balance after massive intestinal resection. Gut. 2004;53(12):1781-1786. doi:10.1136/gut.2004.042317
    1. Ventura G, Noirez P, Breuillé D, et al. . Effect of citrulline on muscle functions during moderate dietary restriction in healthy adult rats. Amino Acids. 2013;45(5):1123-1131. doi:10.1007/s00726-013-1564-3
    1. Wijnands KA, Vink H, Briedé JJ, et al. . Citrulline a more suitable substrate than arginine to restore NO production and the microcirculation during endotoxemia. PLoS One. 2012;7(5):e37439. doi:10.1371/journal.pone.0037439
    1. World Medical Association World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects. JAMA. 2013;310(20):2191-2194. doi:10.1001/jama.2013.281053
    1. International Conference on Harmonisation Working Group. Guideline for good clinical practice E6 (R1). . Published June 10, 1996. Accessed September 18, 2019.
    1. Hafner P, Bonati U, Rubino D, et al. . Treatment with L-citrulline and metformin in Duchenne muscular dystrophy: study protocol for a single-centre, randomised, placebo-controlled trial. Trials. 2016;17(1):389. doi:10.1186/s13063-016-1503-1
    1. European Medicines Agency Guideline on medicinal products for the treatment of Duchenne and Becker muscular dystrophy. . Published December 17, 2015. Accessed September 24, 2019.
    1. Hafner P, Bonati U, Erne B, et al. . 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(1):e0147634. doi:10.1371/journal.pone.0147634
    1. Motor Function Measure . Accessed September 24, 2019.
    1. Bonati U, Hafner P, Schädelin S, et al. . Quantitative muscle MRI: a powerful surrogate outcome measure in Duchenne muscular dystrophy. Neuromuscul Disord. 2015;25(9):679-685. doi:10.1016/j.nmd.2015.05.006
    1. McDonald CM, Henricson EK, Abresch RT, et al. ; PTC124-GD-007-DMD Study Group . The 6-minute walk test and other clinical endpoints in Duchenne muscular dystrophy: reliability, concurrent validity, and minimal clinically important differences from a multicenter study. Muscle Nerve. 2013;48(3):357-368. doi:10.1002/mus.23905
    1. McDonald CM, Henricson EK, Abresch RT, et al. ; PTC124-GD-007-DMD Study Group . The 6-minute walk test and other endpoints in Duchenne muscular dystrophy: longitudinal natural history observations over 48 weeks from a multicenter study. Muscle Nerve. 2013;48(3):343-356. doi:10.1002/mus.23902
    1. Fischmann A, Hafner P, Fasler S, et al. . Quantitative MRI can detect subclinical disease progression in muscular dystrophy. J Neurol. 2012;259(8):1648-1654. doi:10.1007/s00415-011-6393-2
    1. Wokke BH, Bos C, Reijnierse M, et al. . Comparison of Dixon and T1-weighted MR methods to assess the degree of fat infiltration in Duchenne muscular dystrophy patients. J Magn Reson Imaging. 2013;38(3):619-624. doi:10.1002/jmri.23998
    1. Vuillerot C, Girardot F, Payan C, et al. . Monitoring changes and predicting loss of ambulation in Duchenne muscular dystrophy with the Motor Function Measure. Dev Med Child Neurol. 2010;52(1):60-65. doi:10.1111/j.1469-8749.2009.03316.x
    1. Bérard C, Payan C, Hodgkinson I, Fermanian J; MFM Collaborative Study Group . A motor function measure for neuromuscular diseases: construction and validation study. Neuromuscul Disord. 2005;15(7):463-470. doi:10.1016/j.nmd.2005.03.004
    1. Schulz KF, Grimes DA. Unequal group sizes in randomised trials: guarding against guessing. Lancet. 2002;359(9310):966-970. doi:10.1016/S0140-6736(02)08029-7
    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. PLOS ONE Staff Correction: Long term natural history data in ambulant boys with Duchenne muscular dystrophy: 36-month changes. PLoS One. 2015;10(3):e0121882. doi:10.1371/journal.pone.0121882
    1. Mazzone ES, Coratti G, Sormani MP, et al. . Timed rise from floor as a predictor of disease progression in Duchenne muscular dystrophy: an observational study. PLoS One. 2016;11(3):e0151445. doi:10.1371/journal.pone.0151445
    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. Flanigan KM, Voit T, Rosales XQ, et al. . Pharmacokinetics and safety of single doses of drisapersen in non-ambulant subjects with Duchenne muscular dystrophy: results of a double-blind randomized clinical trial. Neuromuscul Disord. 2014;24(1):16-24. doi:10.1016/j.nmd.2013.09.004
    1. Arpan I, Willcocks RJ, Forbes SC, et al. . Examination of effects of corticosteroids on skeletal muscles of boys with DMD using MRI and MRS. Neurology. 2014;83(11):974-980. doi:10.1212/WNL.0000000000000775

Source: PubMed

3
Abonnere