A phase 3 randomized placebo-controlled trial of tadalafil for Duchenne muscular dystrophy

Ronald G Victor, H Lee Sweeney, Richard Finkel, Craig M McDonald, Barry Byrne, Michelle Eagle, Nathalie Goemans, Krista Vandenborne, Alberto L Dubrovsky, Haluk Topaloglu, M Carrie Miceli, Pat Furlong, John Landry, Robert Elashoff, David Cox, Tadalafil DMD Study Group, Hoda Abdel-Hamid, Susan Apkon, Richard Barohn, Elena Belousova, Enrico Bertini, John Brandsema, Claudio Bruno, William Burnette, Russell Butterfield, Barry Byrne, Craig Campbell, Jose Carlo, Jong-Hee Chae, Saleel Chandratre, Giacomo Comi, Anne Connolly, Imelda De Groot, Nicolas Deconinck, Joseph Dooley, Alberto Dubrovsky, Julien Durigneux, Erika Finanger, Richard Finkel, L Matthew Frank, Nathalie Goemans, Amy Harper, Ayako Hattori, Ozlem Herguner, Susan Iannaccone, Joanne Janas, Yuh-Jyh Jong, JanBerd Kirschner, Hirofumi Komaki, Nancy Kuntz, Wang-Tso Lee, Edward Leung, Jean Mah, Katherine Mathews, Craig McDonald, Eugenio Mercuri, Hugh McMillan, Wolfgang Mueller-Felber, Adolfo Lopez de Munain, Akinori Nakamura, Erik Niks, Katsuhisa Ogata, Samuel Pascual, Elena Pegoraro, Yann Pereon, Ben Renfroe, Ratna Bhavaraju Sanka, Jens Schallner, Ulrike Schara, Kathryn Selby, Isabel Illa Sendra, Laurent Servais, Edward Smith, Susan Sparks, Haluk Topaloglu, Ron Victor, Juan Jose Vilchez, Matthew Wicklund, Ekkehard Wilichoswki, Brenda Wong, Ronald G Victor, H Lee Sweeney, Richard Finkel, Craig M McDonald, Barry Byrne, Michelle Eagle, Nathalie Goemans, Krista Vandenborne, Alberto L Dubrovsky, Haluk Topaloglu, M Carrie Miceli, Pat Furlong, John Landry, Robert Elashoff, David Cox, Tadalafil DMD Study Group, Hoda Abdel-Hamid, Susan Apkon, Richard Barohn, Elena Belousova, Enrico Bertini, John Brandsema, Claudio Bruno, William Burnette, Russell Butterfield, Barry Byrne, Craig Campbell, Jose Carlo, Jong-Hee Chae, Saleel Chandratre, Giacomo Comi, Anne Connolly, Imelda De Groot, Nicolas Deconinck, Joseph Dooley, Alberto Dubrovsky, Julien Durigneux, Erika Finanger, Richard Finkel, L Matthew Frank, Nathalie Goemans, Amy Harper, Ayako Hattori, Ozlem Herguner, Susan Iannaccone, Joanne Janas, Yuh-Jyh Jong, JanBerd Kirschner, Hirofumi Komaki, Nancy Kuntz, Wang-Tso Lee, Edward Leung, Jean Mah, Katherine Mathews, Craig McDonald, Eugenio Mercuri, Hugh McMillan, Wolfgang Mueller-Felber, Adolfo Lopez de Munain, Akinori Nakamura, Erik Niks, Katsuhisa Ogata, Samuel Pascual, Elena Pegoraro, Yann Pereon, Ben Renfroe, Ratna Bhavaraju Sanka, Jens Schallner, Ulrike Schara, Kathryn Selby, Isabel Illa Sendra, Laurent Servais, Edward Smith, Susan Sparks, Haluk Topaloglu, Ron Victor, Juan Jose Vilchez, Matthew Wicklund, Ekkehard Wilichoswki, Brenda Wong

Abstract

Objective: To conduct a randomized trial to test the primary hypothesis that once-daily tadalafil, administered orally for 48 weeks, lessens the decline in ambulatory ability in boys with Duchenne muscular dystrophy (DMD).

Methods: Three hundred thirty-one participants with DMD 7 to 14 years of age taking glucocorticoids were randomized to tadalafil 0.3 mg·kg-1·d-1, tadalafil 0.6 mg·kg-1·d-1, or placebo. The primary efficacy measure was 6-minute walk distance (6MWD) after 48 weeks. Secondary efficacy measures included North Star Ambulatory Assessment and timed function tests. Performance of Upper Limb (PUL) was a prespecified exploratory outcome.

Results: Tadalafil had no effect on the primary outcome: 48-week declines in 6MWD were 51.0 ± 9.3 m with placebo, 64.7 ± 9.8 m with low-dose tadalafil (p = 0.307 vs placebo), and 59.1 ± 9.4 m with high-dose tadalafil (p = 0.538 vs placebo). Tadalafil also had no effect on secondary outcomes. In boys >10 years of age, total PUL score and shoulder subscore declined less with low-dose tadalafil than placebo. Adverse events were consistent with the known safety profile of tadalafil and the DMD disease state.

Conclusions: Tadalafil did not lessen the decline in ambulatory ability in boys with DMD. Further studies should be considered to confirm the hypothesis-generating upper limb data and to determine whether ambulatory decline can be slowed by initiation of tadalafil before 7 years of age.

Clinicaltrialsgov identifier: NCT01865084.

Classification of evidence: This study provides Class I evidence that tadalafil does not slow ambulatory decline in 7- to 14-year-old boys with Duchenne muscular dystrophy.

Copyright © 2017 The Author(s). Published by Wolters Kluwer Health, Inc. on behalf of the American Academy of Neurology.

Figures

Figure 1. Patient disposition
Figure 1. Patient disposition
Figure 2. Mean change in efficacy measures
Figure 2. Mean change in efficacy measures
(A) Mean change from baseline to week 48 in 6MWD. (B) Mean change from baseline to week 48 in NSAA (linearized score on scale of 0–100). (C) Mean 6MWD (meters), baseline to week 48. (D) Mean percent of predicted 6MWD, baseline to week 48. *Primary efficacy endpoint. Analyses for each efficacy parameter included all patients with nonmissing baseline and at least 1 nonmissing postbaseline value (for 6MWD: n = 113 in placebo, 101 in tadalafil 0.3 mg/kg, and 111 for tadalafil 0.6 mg/kg; for NSAA: n = 116 in placebo, 102 in tadalafil 0.3 mg/kg, and 112 for tadalafil 0.6 mg/kg). LS = least squares; NSAA = North Star Ambulatory Assessment; 6MWD = 6-minute walk distance.
Figure 3. Mean change in total and…
Figure 3. Mean change in total and shoulder level PUL score
(A) Total PUL score in the total population (n = 114 in placebo, 99 in tadalafil 0.3 mg/kg, and 107 in tadalafil 0.6 mg/kg). (B) Total PUL score in patients ≤10 years of age (n = 95 in placebo, 68 in tadalafil 0.3 mg/kg, and 85 in tadalafil 0.6 mg/kg). (C) Total PUL score in patients >10 years of age (n = 19 in placebo, 31 in tadalafil 0.3 mg/kg, and 22 in tadalafil 0.6 mg/kg). (D) Shoulder-level PUL subscore in the total population (n = 116 in placebo, 101 in tadalafil 0.3 mg/kg, and 111 in tadalafil 0.6 mg/kg). (E) Shoulder-level PUL subscore in patients ≤10 years of age (n = 96 in placebo, 69 in tadalafil 0.3 mg/kg, and 89 in tadalafil 0.6 mg/kg). (F) Shoulder-level PUL subscore in patients >10 years of age (n = 20 in placebo, 32 in tadalafil 0.3 mg/kg, and 22 in tadalafil 0.6 mg/kg). Changes in total PUL score are from a total score range of 0 to 72; changes in shoulder-level PUL are from a total score range of 0 to 16. Higher score represents better function. Analyses for each efficacy parameter included all patients with nonmissing baseline and at least 1 nonmissing postbaseline value. *p < 0.05. LS = least squares; PUL = Performance of Upper Limb.

References

    1. Kinnett K, Rodger S, Vroom E, Furlong P, Aartsma-Rus A, Bushby K. Imperatives for DUCHENNE MD: a simplified guide to comprehensive care for Duchenne muscular dystrophy. PLoS Curr Epub 2015 Aug 7.
    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;24:482–491.
    1. Ervasti JM, Ohlendieck K, Kahl SD, Gaver MG, Campbell KP. Deficiency of a glycoprotein component of the dystrophin complex in dystrophic muscle. Nature 1990;345:315–319.
    1. Hoffman EP, Brown RH Jr, Kunkel LM. Dystrophin: the protein product of the Duchenne muscular dystrophy locus. Cell 1987;51:919–928.
    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;82:743–752.
    1. Lai Y, Thomas GD, Yue Y, et al. . Dystrophins carrying spectrin-like repeats 16 and 17 anchor nNOS to the sarcolemma and enhance exercise performance in a mouse model of muscular dystrophy. J Clin Invest 2009;119:624–635.
    1. De Palma C, Morisi F, Pambianco S, et al. . Deficient nitric oxide signalling impairs skeletal muscle growth and performance: involvement of mitochondrial dysregulation. Skelet Muscle 2014;4:22.
    1. Froehner SC, Reed SM, Anderson KN, Huang PL, Percival JM. Loss of nNOS inhibits compensatory muscle hypertrophy and exacerbates inflammation and eccentric contraction-induced damage in mdx mice. Hum Mol Genet 2015;24:492–505.
    1. Moon Y, Balke JE, Madorma D, et al. . Nitric oxide regulates skeletal muscle fatigue, fiber type, microtubule organization, and mitochondrial ATP synthesis efficiency through cGMP-dependent mechanisms. Antioxid Redox Signal 2017;26:966–985.
    1. Rebolledo DL, Kim MJ, Whitehead NP, Adams ME, Froehner SC. Sarcolemmal targeting of nNOSµ improves contractile function of mdx muscle. Hum Mol Genet 2016;25:158–166.
    1. Hansen J, Thomas GD, Harris SA, Parsons WJ, Victor RG. Differential sympathetic neural control of oxygenation in resting and exercising human skeletal muscle. J Clin Invest 1996;98:584–596.
    1. Nelson MD, Rosenberry R, Barresi R, et al. . Sodium nitrate alleviates functional muscle ischaemia in patients with Becker muscular dystrophy. J Physiol (London) 2015;593:5183–5200.
    1. Thomas GD, Hansen J, Victor RG. Inhibition of alpha 2-adrenergic vasoconstriction during contraction of glycolytic, not oxidative, rat hind limb muscle. Am J Physiol 1994;266:H920–H929.
    1. Allen DG, Whitehead NP, Froehner SC. Absence of dystrophin disrupts skeletal muscle signaling: roles of Ca2+, reactive oxygen species, and nitric oxide in the development of muscular dystrophy. Physiol Rev 2016;96:253–305.
    1. Nelson MD, Rader F, Tang X, et al. . PDE5 inhibition alleviates functional muscle ischemia in boys with Duchenne muscular dystrophy. Neurology 2014;82:2085–2091.
    1. Sander M, Chavoshan B, Harris SA, et al. . Functional muscle ischemia in neuronal nitric oxide synthase-deficient skeletal muscle of children with Duchenne muscular dystrophy. Proc Natl Acad Sci USA 2000;97:13818–13823.
    1. Thomas GD, Sander M, Lau KS, Huang PL, Stull JT, Victor RG. Impaired metabolic modulation of alpha-adrenergic vasoconstriction in dystrophin-deficient skeletal muscle. Proc Natl Acad Sci USA 1998;95:15090–15095.
    1. Rando TA. Role of nitric oxide in the pathogenesis of muscular dystrophies: a “two hit” hypothesis of the cause of muscle necrosis. Microsc Res Tech 2001;55:223–235.
    1. Adamo CM, Dai DF, Percival JM, et al. . Sildenafil reverses cardiac dysfunction in the mdx mouse model of Duchenne muscular dystrophy. Proc Natl Acad Sci USA 2010;107:19079–19083.
    1. Asai A, Sahani N, Kaneki M, Ouchi Y, Martyn JA, Yasuhara SE. Primary role of functional ischemia, quantitative evidence for the two-hit mechanism, and phosphodiesterase-5 inhibitor therapy in mouse muscular dystrophy. PLoS One 2007;2:e806.
    1. Hammers DW, Sleeper MM, Forbes SC, Shima A, Walter GA, Sweeney HL. Tadalafil treatment delays the onset of cardiomyopathy in dystrophin-deficient hearts. J Am Heart Assoc 2016;5:e003911.
    1. Kawahara G, Karpf JA, Myers JA, Alexander MS, Guyon JR, Kunkel LM. Drug screening in a zebrafish model of Duchenne muscular dystrophy. Proc Natl Acad Sci USA 2011;108:5331–5336.
    1. Kobayashi YM, Rader EP, Crawford RW, et al. . Sarcolemma-localized nNOS is required to maintain activity after mild exercise. Nature 2008;456:511–515.
    1. Kobayashi YM, Rader EP, Crawford RW, Campbell KP. Endpoint measures in the mdx mouse relevant for muscular dystrophy pre-clinical studies. Neuromuscul Disord 2012;22:34–42.
    1. Percival JM, Whitehead NP, Adams ME, Adamo CM, Beavo JA, Froehner SC. Sildenafil reduces respiratory muscle weakness and fibrosis in the mdx mouse model of Duchenne muscular dystrophy. J Pathol 2012;228:77–87.
    1. Thomas GD, Shaul PW, Yuhanna IS, Froehner SC, Adams ME. Vasomodulation by skeletal muscle-derived nitric oxide requires alpha-syntrophin-mediated sarcolemmal localization of neuronal nitric oxide synthase. Circ Res 2003;92:554–560.
    1. Uaesoontrachoon K, Quinn JL, Tatem KS, et al. . Long-term treatment with naproxcinod significantly improves skeletal and cardiac disease phenotype in the mdx mouse model of dystrophy. Hum Mol Genet 2014;23:3239–3249.
    1. Wehling-Henricks M, Jordan MC, Roos KP, Deng B, Tidball JG. Cardiomyopathy in dystrophin-deficient hearts is prevented by expression of a neuronal nitric oxide synthase transgene in the myocardium. Hum Mol Genet 2005;14:1921–1933.
    1. Wehling M, Spencer MJ, Tidball JG. A nitric oxide synthase transgene ameliorates muscular dystrophy in mdx mice. J Cell Biol 2001;155:123–131.
    1. Zhang Y, Yue Y, Li L, et al. . Dual AAV therapy ameliorates exercise-induced muscle injury and functional ischemia in murine models of Duchenne muscular dystrophy. Hum Mol Genet 2013;22:3720–3729.
    1. Martin EA, Barresi R, Byrne BJ, et al. . Tadalafil alleviates muscle ischemia in patients with Becker muscular dystrophy. Sci Transl Med 2012;4:162ra155.
    1. Pane M, Mazzone ES, Fanelli L, et al. . Reliability of the performance of upper limb assessment in Duchenne muscular dystrophy. Neuromuscul Disord 2014;24:201–206.
    1. Henricson E, Abresch R, Han JJ, et al. . The 6-minute walk test and person-reported outcomes in boys with Duchenne muscular dystrophy and typically developing controls: longitudinal comparisons and clinically-meaningful changes over one year. PLoS Curr 2013;5:ecurrents.md.9e17658b007eb79fcd6f723089f79e06.
    1. Mayhew AG, Cano SJ, Scott E, et al. . Detecting meaningful change using the North Star Ambulatory Assessment in Duchenne muscular dystrophy. Dev Med Child Neurol 2013;55:1046–1052.
    1. Thomas GD, Ye J, De Nardi C, Monopoli A, Ongini E, Victor RG. Treatment with a nitric oxide-donating NSAID alleviates functional muscle ischemia in the mouse model of Duchenne muscular dystrophy. PLoS One 2012;7:e49350.
    1. Forgue ST, Patterson BE, Bedding AW, et al. . Tadalafil pharmacokinetics in healthy subjects. Br J Clin Pharmacol 2006;61:280–288.
    1. Wrishko RE, Dingemanse J, Yu A, et al. . Pharmacokinetic interaction between tadalafil and bosentan in healthy male subjects. J Clin Pharmacol 2008;48:610–618.
    1. Witting N, Kruuse C, Nyhuus B, et al. . Effect of sildenafil on skeletal and cardiac muscle in Becker muscular dystrophy. Ann Neurol 2014;76:550–557.
    1. Leung DG, Herzka DA, Thompson WR, et al. . Sildenafil does not improve cardiomyopathy in Duchenne/Becker muscular dystrophy. Ann Neurol 2014;76:541–549.
    1. Willcocks RJ, Arpan IA, Forbes SC, et al. . Longitudinal measurements of MRI-T2 in boys with Duchenne muscular dystrophy: effects of age and disease progression. Neuromuscul Disord 2014;24:393–401.

Source: PubMed

3
Subskrybuj