Eteplirsen Treatment Attenuates Respiratory Decline in Ambulatory and Non-Ambulatory Patients with Duchenne Muscular Dystrophy

Navid Khan, Helen Eliopoulos, Lixin Han, T Bernard Kinane, Linda P Lowes, Jerry R Mendell, Heather Gordish-Dressman, Erik K Henricson, Craig M McDonald, Eteplirsen Investigators and the CINRG DNHS Investigators, Navid Khan, Helen Eliopoulos, Lixin Han, T Bernard Kinane, Linda P Lowes, Jerry R Mendell, Heather Gordish-Dressman, Erik K Henricson, Craig M McDonald, Eteplirsen Investigators and the CINRG DNHS Investigators

Abstract

Background: Duchenne muscular dystrophy (DMD) patients experience skeletal muscle degeneration, including respiratory muscles. Respiratory decline in glucocorticoid-treated DMD patients, measured by percent predicted forced vital capacity (FVC% p), is typically 5% annually in patients aged 10 to 18 years.

Objective: Evaluate the effects of eteplirsen on FVC% p annual change in 3 trials versus matched Cooperative International Neuromuscular Research Group Duchenne Natural History Study (CINRG DNHS) controls.

Methods: Eteplirsen studies 201/202 evaluated eligible ambulatory DMD patients for at least 4 years, study 204 evaluated primarily non-ambulatory DMD patients for 2 years, and ongoing study 301 is evaluating ambulatory DMD patients for 2 years (interim analysis is included). Eteplirsen-treated patients (n = 74) were amenable to exon 51 skipping and were receiving glucocorticoids. Three CINRG DNHS cohorts included: glucocorticoid-treated patients amenable to exon 51 skipping (Exon 51 CINRG DNHS; n = 20), all glucocorticoid-treated CINRG patients (All CINRG DNHS; n = 172), and all glucocorticoid-treated genotyped CINRG DNHS patients (Genotyped CINRG DNHS; n = 148). FVC% p assessments between ages 10 and <18 years were included for all patients; mixed-model analyses characterized FVC% p annual change.

Results: FVC% p annual change was greater for CINRG DNHS Exon 51 controls (- 6.00) versus patients in studies 201/202, study 204, and study 301 (- 2.19, P < 0.001; - 3.66, P 0.004; and - 3.79, P 0.017, respectively). FVC% p annual change in all eteplirsen studies suggested treatment benefit compared with the Genotyped CINRG DNHS (- 5.67) and All CINRG DNHS (- 5.56) cohorts (P < 0.05, all comparisons).

Conclusions: Significant, clinically meaningful attenuation of FVC%p decline was observed in eteplirsen-treated patients versus CINRG DNHS controls.

Keywords: Dystrophin; Exondys 51; forced expiratory volume; vital capacity.

Figures

Fig.1
Fig.1
Study design for eteplirsen studies 201/202. Study 201 was a 24-week, double-blind, placebo-controlled study that randomized 12 patients to receive placebo or eteplirsen (30 or 50 mg/kg/wk). At week 25, patients originally randomized to eteplirsen treatment continued with the same dosage in study 202, an open-label extension study; patients originally randomized to receive placebo were randomized to eteplirsen 30 or 50 mg/kg. All patients were followed for 4 years. Respiratory function assessments included FVC% p as an exploratory endpoint. FVC% p, percent predicated forced vital capacity; OL, open-label; PTP, primary treatment period; R, randomized.
Fig.2
Fig.2
Study design for eteplirsen study 204. In this 2-year (96-week), open-label study, patients with advanced-stage DMD (minimally ambulatory to non-ambulatory patients; N = 24) received weekly infusions of eteplirsen 30 mg/kg. Respiratory function assessments included FVC% p as an exploratory endpoint. AEs, adverse events; ECG, electrocardiogram; FVC% p, percent predicated forced vital capacity; ECHO, echocardiogram; LVEF, left ventricular ejection fraction.
Fig.3
Fig.3
Study design for eteplirsen study 301. In this open-label study, patients with DMD (N = 71) received weekly IV infusions of eteplirsen 30 mg/kg and had both baseline and post-baseline FVC% p. An interim analysis was conducted at year 2 (96 weeks) for all patients who reached that time point. Respiratory function assessments included FVC% p as an exploratory endpoint. BL, baseline; FVC% p, percent predicted forced vital capacity.
Fig.4
Fig.4
Derivation of CINRG DNHS cohorts for comparison of eteplirsen studies. The CINRG DNHS database contains data from 440 patients with DMD across all ages. The All CINRG DNHS cohort (n = 172) comprised glucocorticoid-treated patients with FVC% p assessments obtained between the ages of 10 and 18 years, and excluded any known exon 44 skip–amenable patients. The Genotyped CINRG DNHS cohort (n = 148) comprised glucocorticoid-treated, genetically confirmed patients with FVC% p assessments between the ages of 10 and 18 years and excluded exon 44 skip-amenable patients. The Exon 51 CINRG DNHS cohort (n = 20) comprised glucocorticoid-treated patients between the ages of 10 and 18 years with mutations amenable to exon 51 skipping. CINRG DNHS, Cooperative International Neuromuscular Research Group Duchenne Natural History Study; FVC% p, percent predicted forced vital capacity; GC, glucocorticoid. Dashed box represents pre-specified comparator.

References

    1. McDonald CM, Abresch RT, Carter GT, Fowler WM Jr. V Johnson ER, Kilmer DD, Sigford BJ. Profiles of neuromus-cular diseases. Duchenne muscular dystrophy. Am J Phys Med Rehabil. 1995;74(5 Suppl):S70–92.
    1. Bushby K, Finkel R, Birnkrant DJ, Case LE, Clemens PR, Cripe L, Kaul A, Kinnett K, McDonald C, Pandya S, Poysky J, Shapiro F, Tomezsko J, Constantin C, DMD Care Considerations Working Group. Diagnosis and management of Duchenne muscular dystrophy, part 1: Diagnosis, and pharmacological and psychosocial management. Lancet Neurol. 2010;9(1):77–93.
    1. Connolly AM, Florence JM, Cradock MM, Malkus EC, Schierbecker JR, Siener CA, Wulf CO, Anand P, Golumbek PT, Zaidman CM, Philip Miller J, Lowes LP, Alfano LN, Viollet-Callendret L, Flanigan KM, Mendell JR, McDonald CM, Goude E, Johnson L, Nicorici A, Karachunski PI, Day JW, Dalton JC, Farber JM, Buser KK, Darras BT, Kang PB, Riley SO, Shriber E, Parad R, Bushby K, Eagle M, and MDA DMD Clinical Research Network. Motor and cognitive assessment of infants and young boys with Duchenne muscular dystrophy: Results from the Muscular Dystrophy Association DMD Clinical Research Network. Neuromuscul Disord. 2013;23(7):529–39.
    1. Hoffman EP, Fischbeck KH, Brown RH, Johnson M, Medori R, Loike JD, Harris JB, Waterston R, Brooke M, Specht L, et al. Characterization of dystrophin in muscle-biopsy specimens from patients with Duchenne’s or Becker’s muscular dystrophy. N Engl J Med. 1988;318(21):1363–8.
    1. Peverelli L, Testolin S, Villa L, D’Amico A, Petrini S, Favero C, Magri F, Morandi L, Mora M, Mongini T, Bertini E, Sciacco M, Comi GP, Moggio M. Histologic muscular history in steroid-treated and untreated patients with Duchenne dystrophy. Neurology. 2015;85(21):1886–93.
    1. Goldstein JA, McNally EM. Mechanisms of muscle weakness in muscular dystrophy. J Gen Physiol. 2010;136(1):29–34.
    1. Mayer O, Henricson E, McDonald C, Buyse G. Advances in pulmonary care in Duchenne muscular dystrophy. US Neurology. 2017;13(1):7.
    1. McDonald CM, Henricson EK, Abresch RT, Duong T, Joyce NC, Hu F, Clemens PR, Hoffman EP, Cnaan A, Gordish-Dressman H, 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):451–61.
    1. Bushby K, Finkel R, Birnkrant DJ, Case LE, Clemens PR, Cripe L, Kaul A, Kinnett K, McDonald C, Pandya S, Poysky J, Shapiro F, Tomezsko J, Constantin C, DMD Care Considerations Working Group. Diagnosis and management of Duchenne muscular dystrophy, part 2: Implementation of multidisciplinary care. Lancet Neurol. 2010;9(2):177–89.
    1. McDonald C, Gordish-Dressman H, Henricson E, Duong T, Joyce NC, jhawar S, Hu F, Connolly AM, Cnaan A, Abresch R. Longitudinal pulmonary function testing outcome measures in Duchenne muscular dystrophy: Long-term natural history with and without glucocorticoids. Neuromuscul Disord. 2018;28(11):897–909.
    1. Hahn A, Bach JR, Delaubier A, Renardel-Irani A, Guillou C, Rideau Y. Clinical implications of maximal respiratory pressure determinations for individuals with Duchenne muscular dystrophy. Arch Phys Med Rehabil. 1997;78(1): 1–6.
    1. Khirani S, Ramirez A, Aubertin G, Boule M, Chemouny C, Forin V, Fauroux B. Respiratory muscle decline in Duchenne muscular dystrophy. Pediatr Pulmonol. 2014;49(5):473–81.
    1. Mayer OH, Finkel RS, Rummey C, Benton MJ, Glanzman AM, Flickinger J, Lindstrom BM, Meier T. Characterization of pulmonary function in Duchenne muscular dystrophy. Pediatr Pulmonol. 2015;50(5):487–94.
    1. Miller JR, Colbert AP, Schock NC. Ventilator use in progressive neuromuscular disease: Impact on patients and their families. Dev Med Child Neurol. 1988;30(2):200–7.
    1. Birnkrant DJ, Bushby K, Bann CM, Alman BA, Apkon SD, Blackwell A, Case LE, Cripe L, Hadjiyannakis S, Olson AK, Sheehan DW, Bolen J, Weber DR, Ward LM, DMD Care Considerations Working Group. Diagnosis and management of Duchenne muscular dystrophy, part 2: Respiratory, cardiac, bone health, and orthopaedic management. Lancet Neurol. 2018;17(4):347–61.
    1. Aartsma-Rus A, Fokkema I, Verschuuren J, Ginjaar I, van Deutekom J, van Ommen GJ, den Dunnen JT. Theoretic applicability of antisense-mediated exon skipping for Duchenne muscular dystrophy mutations. Hum Mutat. 2009;30(3):293–9.
    1. Bladen CL, Salgado D, Monges S, Foncuberta ME, Kekou K, Kosma K, Dawkins H, Lamont L, Roy AJ, Chamova T, Guergueltcheva V, Chan S, Korngut L, Campbell C, Dai Y, Wang J, Barisic N, Brabec P, Lahdetie J, Walter MC, Schreiber-Katz O, Karcagi V, Garami M, Viswanathan V, Bayat F, Buccella F, Kimura E, Koeks Z, van den Bergen JC, Rodrigues M, Roxburgh R, Lusakowska A, Kostera-Pruszczyk A, Zimowski J, Santos R, Neagu E, Artemieva S, Rasic VM, Vojinovic D, Posada M, Bloetzer C, Jeannet PY, Joncourt F, Diaz-Manera J, Gallardo E, Karaduman AA, Topaloglu H, El Sherif R, Stringer A, Shatillo AV, Martin AS, Peay HL, Bellgard MI, Kirschner J, Flanigan KM, Straub V, Bushby K, Verschuuren J, Aartsma-Rus A, Beroud C, Lochmuller H. The TREAT-NMD DMD Global Database: Analysis of more than 7,000 Duchenne muscular dystrophy mutations. Hum Mutat. 2015;36(4):395–402.
    1. Bello L, Morgenroth LP, Gordish-Dressman H, Hoffman EP, McDonald CM, Cirak S, CINRG Investigators. DMD genotypes and loss of ambulation in the CINRG Duchenne Natural History Study. Neurology. 2016;87(4):401–9.
    1. Ricotti V, Ridout DA, Pane M, Main M, Mayhew A, Mercuri E, Manzur AY, Muntoni F, 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):149–55.
    1. Mendell JR, Rodino-Klapac LR, Sahenk Z, Roush K, Bird L, Lowes LP, Alfano L, Gomez AM, Lewis S, Kota J, Malik V, Shontz K, Walker CM, Flanigan KM, Corridore M, Kean JR, Allen HD, Shilling C, Melia KR, Sazani P, Saoud JB, Kaye EM, Eteplirsen Study Group. Eteplirsen for the treatment of Duchenne muscular dystrophy. Ann Neurol. 2013;74(5):637–47.
    1. Kinane TB, Mayer OH, Duda PW, Lowes LP, Moody SL, Mendell JR. Long-term pulmonary function in Duchenne muscular dystrophy: Comparison of eteplirsen-treated patients to natural history. J Neuromuscul Dis. 2018;5(1):47–58.
    1. Brooke MH, Griggs RC, Mendell JR, Fenichel GM, Shumate JB, and Pellegrino RJ. Clinical trial in Duchenne dystrophy. I. The design of the protocol. Muscle Nerve. 1981;4(3):186–97.
    1. Gauld LM, Kappers J, Carlin JB, Robertson CF. Height prediction from ulna length. Dev Med Child Neurol. 2004;46(7):475–80.
    1. McDonald CM, Henricson EK, Abresch RT, Han JJ, Escolar DM, Florence JM, Duong T, Arrieta A, Clemens PR, Hoffman EP, Cnaan A, 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.
    1. Henricson EK, Abresch RT, Cnaan A, Hu F, Duong T, Arrieta A, Han J, Escolar DM, Florence JM, Clemens PR, Hoffman EP, McDonald CM, Investigators C. 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.
    1. Duchenne Natural History. The Cooperative International Neuromuscular Research Group, 2017. Available at: . Accessed: November 6, 2018.
    1. Hankinson JL, Odencrantz JR, Fedan KB. Spirometric reference values from a sample of the general U.S. population. Am J Respir Crit Care Med. 1999;159(1):179–87.
    1. Polgar G, Varuni P Pulmonary Function Testing in Children: Techniques and Standards. Philadelphia, PA: Saunders; 1971. pp. 273.
    1. Phillips MF, Quinlivan RC, Edwards RH, Calverley PM. Changes in spirometry over time as a prognostic marker in patients with Duchenne muscular dystrophy. Am J Respir Crit Care Med. 2001;164(12):2191–4.
    1. Roberto R, Fritz A, Hagar Y, Boice B, Skalsky A, Hwang H, Beckett L, McDonald C, Gupta M. The natural history of cardiac and pulmonary function decline in patients with Duchenne muscular dystrophy. Spine. 2011;36(15):E1009–17.
    1. Birnkrant DJ, Bushby K, Bann CM, Apkon SD, Blackwell A, Brumbaugh D, Case LE, Clemens PR, Hadjiyannakis S, Pandya S, Street N, Tomezsko J, Wagner KR, Ward LM, Weber DR, Group DCCW. Diagnosis and management of Duchenne muscular dystrophy, part 1: Diagnosis, and neuromuscular, rehabilitation, endocrine, and gastrointestinal and nutritional management. Lancet Neurol. 2018;17(3):251–67.
    1. Birnkrant DJ, Bushby K, Bann CM, Apkon SD, Blackwell A, Colvin MK, Cripe L, Herron AR, Kennedy A, Kinnett K, Naprawa J, Noritz G, Poysky J, Street N, Trout CJ, Weber DR, Ward LM, DMD Care Considerations Working Group. Diagnosis and management of Duchenne muscular dystrophy, part 3: Primary care, emergency management, psychosocial care, and transitions of care across the lifespan. Lancet Neurol. 2018;17(5):445–55.

Source: PubMed

3
Abonnieren