Pooled Analyses of Phase III Studies of ADS-5102 (Amantadine) Extended-Release Capsules for Dyskinesia in Parkinson's Disease

Lawrence W Elmer, Jorge L Juncos, Carlos Singer, Daniel D Truong, Susan R Criswell, Sotirios Parashos, Larissa Felt, Reed Johnson, Rajiv Patni, Lawrence W Elmer, Jorge L Juncos, Carlos Singer, Daniel D Truong, Susan R Criswell, Sotirios Parashos, Larissa Felt, Reed Johnson, Rajiv Patni

Abstract

Background: Although levodopa is considered the most effective pharmacotherapy for motor symptoms of Parkinson's disease (PD), chronic use is associated with motor complications, including fluctuating response and unpredictable, involuntary movements called dyskinesia. ADS-5102 (amantadine) extended-release (ER) capsules (GOCOVRITM) is a recent US FDA-approved treatment for dyskinesia in PD patients. ADS-5102 is a high-dose, ER formulation of amantadine, administered orally once daily at bedtime, that achieves high plasma drug concentrations throughout the day.

Objective: In this study, we present pooled results from two randomized, double-blind, placebo-controlled, phase III ADS-5102 trials.

Patients and methods: The two studies in PD patients with dyskinesia shared design and eligibility criteria, differing only in treatment duration. Results from common assessment time points were pooled.

Results: At 12 weeks, the least squares (LS) mean change in total score on the Unified Dyskinesia Rating Scale among 100 patients randomized to ADS-5102 and 96 patients randomized to placebo was - 17.7 (standard error [SE] 1.3) vs. - 7.6 (1.3) points, respectively (- 10.1 points, 95% confidence interval [CI] - 13.8, - 6.5; p < 0.0001). The relative treatment difference between groups was 27.3% (p < 0.0001). At 12 weeks, the LS mean change in OFF time was - 0.59 (0.21) vs. +0.41 (0.20) h/day, a difference of - 1.00 h/day (95% CI - 1.57, - 0.44; p = 0.0006). For both efficacy measures, a significant difference from placebo was attained by two weeks, the first post-baseline assessment, and was maintained throughout 12 weeks. In the pooled ADS-5102 group, the most common adverse events were hallucination, dizziness, dry mouth, peripheral edema, constipation, falls, and orthostatic hypotension.

Conclusions: These analyses provide further evidence supporting ADS-5102 as an adjunct to levodopa for treating both dyskinesia and OFF time in PD patients with dyskinesia. Clinicaltrials.gov identifier: NCT02136914 and NCT02274766.

Conflict of interest statement

Lawrence W. Elmer has received honoraria for speaking engagements from Lundbeck, Novartis, UCB Pharma, and Teva Neuroscience; has served as a paid consultant for Teva Neuroscience and UCB Pharma; has received honoraria as a member of advisory boards for Lundbeck, Teva Neuroscience, and UCB Pharma; and has received unrestricted educational grant support from Teva Neuroscience. Jorge L. Juncos reports research grants from the National Institute of Child Health and Human Development (NICHD), the Michael J. Fox Foundation, Adamas Pharmaceuticals, US WorldMeds, Psydon, and Neurocrine. Carlos Singer has received honoraria from Neurocrine, Revance, US WorldMeds, TEVA, and Acorda, and has received grants from Allergan, Adams, Pfizer, Synovia, the Parkinson’s Foundation, National Institutes of Health (NIH), and the Huntington’s Disease Society of America. Daniel D. Truong has received research grants from Ispen, Merz, Auspex, Daiichi Sankyo Pharma, AbbVie, National Institute of Neurological Disorders and Stroke, Kyowa, and Neurocrine. Susan R. Criswell has no relevant disclosures or conflicts of interest. Sotirios Parashos has no relevant disclosures or conflicts of interest. Unrelated to the subject matter of this study, Sotirios Parashos has received consultancies from Dong-A, research support from the Park Nicollet Foundation, National Parkinson Foundation, NINDS, Patient-Centered Outcomes Research Institute, Acorda Therapeutics, Astellas, Pharma2B, and Sunovion Pharmaceuticals. Larissa Felt, Reed Johnson, and Rajiv Patni are employees of and own stock in Adamas Pharmaceuticals.

Figures

Fig. 1
Fig. 1
Pooled population used in this analysis arising from the EASE LID and EASE LID 3 trials. aThe pooled safety population included an additional two patients, both in EASE LID, who received placebo but did not provide a post-baseline UDysRS assessment. mITT modified intent-to-treat, UDysRS Unified Dyskinesia Rating Scale
Fig. 2
Fig. 2
Time course of change from baseline in a UDysRS total score, b OFF time, c ON time with troublesome dyskinesia, and d ON time without troublesome dyskinesia, by pooled treatment group. LS least squares, SE standard error, UDysRS Unified Dyskinesia Rating Scale
Fig. 3
Fig. 3
Cumulative distribution of change in UDysRS total score at 12 weeks by pooled treatment group. UDysRS Unified Dyskinesia Rating Scale
Fig. 4
Fig. 4
Subgroup analyses of 12-week change in a UDysRS total score and b OFF time (2.5 h is the median baseline OFF time). an = 100 for ADS-5102 and 96 for placebo. bn = 46 for ADS-5102 and 41 for placebo. cn = 54 for ADS-5102 and 55 for placebo. dn = 48 for ADS-5102 and 41 for placebo. en = 54 for ADS-5102 and 48 for placebo. fMedian value at baseline in the mITT population. gn = 46 for ADS-5102 and 40 for placebo. hn = 54 for ADS-5102 and 56 for placebo. in = 49 for ADS-5102 and 48 for placebo. jn = 51 for ADS-5102 and 48 for placebo. kn = 45 for ADS-5102 and 49 for placebo. CI confidence interval, LS least squares, MDSUPDRS Movement Disorder Society–Unified Parkinson’s Disease Rating Scale, UDysRS Unified Dyskinesia Rating Scale
Fig. 5
Fig. 5
Distribution of 12-week change in a OFF time, b ON time without troublesome dyskinesia, and c ON time with troublesome dyskinesia, by pooled treatment group (12-week completers). hrs/d hours/day
Fig. 6
Fig. 6
Change from baseline for CGI–C at week 12 by pooled treatment group. CGIC Clinician’s Global Impression of Change

References

    1. Lindholm D, Makela J, Di Liberto V, Mudo G, Belluardo N, Eriksson O, et al. Current disease modifying approaches to treat Parkinson’s disease. Cell Mol Life Sci. 2016;73(7):1365–1379. doi: 10.1007/s00018-015-2101-1.
    1. Hornykiewicz O. A brief history of levodopa. J Neurol. 2010;257(Suppl 2):S249–S252. doi: 10.1007/s00415-010-5741-y.
    1. LeWitt PA, Fahn S. Levodopa therapy for Parkinson disease: a look backward and forward. Neurology. 2016;86(14 Suppl 1):S3–S12. doi: 10.1212/WNL.0000000000002509.
    1. Connolly BS, Lang AE. Pharmacological treatment of Parkinson disease: a review. JAMA. 2014;311(16):1670–1683. doi: 10.1001/jama.2014.3654.
    1. Ahlskog JE, Muenter MD. Frequency of levodopa-related dyskinesias and motor fluctuations as estimated from the cumulative literature. Mov Disord. 2001;16(3):448–458. doi: 10.1002/mds.1090.
    1. Olanow CW, Stern MB, Sethi K. The scientific and clinical basis for the treatment of Parkinson disease (2009) Neurology. 2009;72(21 Suppl 4):S1–S136. doi: 10.1212/WNL.0b013e3181a1d44c.
    1. Jankovic J. Motor fluctuations and dyskinesias in Parkinson’s disease: clinical manifestations. Mov Disord. 2005;20(Suppl 11):S11–S16. doi: 10.1002/mds.20458.
    1. Lees AJ. The on-off phenomenon. J Neurol Neurosurg Psychiatry. 1989;52(Suppl):29–37. doi: 10.1136/jnnp.52.Suppl.29.
    1. Chapuis S, Ouchchane L, Metz O, Gerbaud L, Durif F. Impact of the motor complications of Parkinson’s disease on the quality of life. Mov Disord. 2005;20(2):224–230. doi: 10.1002/mds.20279.
    1. Hechtner MC, Vogt T, Zollner Y, Schroder S, Sauer JB, Binder H, et al. Quality of life in Parkinson’s disease patients with motor fluctuations and dyskinesias in five European countries. Parkinsonism Relat Disord. 2014;20(9):969–974. doi: 10.1016/j.parkreldis.2014.06.001.
    1. Rascol O, Perez-Lloret S, Damier P, Delval A, Derkinderen P, Destee A, et al. Falls in ambulatory non-demented patients with Parkinson’s disease. J Neural Transm (Vienna). 2015;122(10):1447–1455. doi: 10.1007/s00702-015-1396-2.
    1. Suh DC, Pahwa R, Mallya U. Treatment patterns and associated costs with Parkinson’s disease levodopa induced dyskinesia. J Neurol Sci. 2012;319(1–2):24–31. doi: 10.1016/j.jns.2012.05.029.
    1. Muller T, Woitalla D, Russ H, Hock K, Haeger DA. Prevalence and treatment strategies of dyskinesia in patients with Parkinson’s disease. J Neural Transm (Vienna). 2007;114(8):1023–1026. doi: 10.1007/s00702-007-0718-4.
    1. Schaeffer E, Pilotto A, Berg D. Pharmacological strategies for the management of levodopa-induced dyskinesia in patients with Parkinson’s disease. CNS Drugs. 2014;28(12):1155–1184. doi: 10.1007/s40263-014-0205-z.
    1. Chase TN, Oh JD. Striatal mechanisms and pathogenesis of parkinsonian signs and motor complications. Ann Neurol. 2000;47(4 Suppl 1):S122–9 (discussion S129–S130).
    1. Danysz W, Parsons CG, Kornhuber J, Schmidt WJ, Quack G. Aminoadamantanes as NMDA receptor antagonists and antiparkinsonian agents: preclinical studies. Neurosci Biobehav Rev. 1997;21(4):455–468. doi: 10.1016/S0149-7634(96)00037-1.
    1. Ory-Magne F, Corvol JC, Azulay JP, Bonnet AM, Brefel-Courbon C, Damier P, et al. Withdrawing amantadine in dyskinetic patients with Parkinson disease: the AMANDYSK trial. Neurology. 2014;82(4):300–307. doi: 10.1212/WNL.0000000000000050.
    1. Verhagen Metman L, Del Dotto P, van den Munckhof P, Fang J, Mouradian MM, Chase TN. Amantadine as treatment for dyskinesias and motor fluctuations in Parkinson’s disease. Neurology. 1998;50(5):1323–1326. doi: 10.1212/WNL.50.5.1323.
    1. Metman LV, Del Dotto P, LePoole K, Konitsiotis S, Fang J, Chase TN. Amantadine for levodopa-induced dyskinesias: a 1-year follow-up study. Arch Neurol. 1999;56(11):1383–1386. doi: 10.1001/archneur.56.11.1383.
    1. Wolf E, Seppi K, Katzenschlager R, Hochschorner G, Ransmayr G, Schwingenschuh P, et al. Long-term antidyskinetic efficacy of amantadine in Parkinson’s disease. Mov Disord. 2010;25(10):1357–1363. doi: 10.1002/mds.23034.
    1. Crosby NJ, Deane KH, Clarke CE. Amantadine for dyskinesia in Parkinson’s disease. Cochrane Database Syst Rev. 2003;(2):CD003467.
    1. Thomas A, Iacono D, Luciano AL, Armellino K, Di Iorio A, Onofrj M. Duration of amantadine benefit on dyskinesia of severe Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2004;75(1):141–143. doi: 10.1136/jnnp.2004.036558.
    1. GOCOVRI (amantadine) extended release capsules for oral use. .
    1. Pahwa R, Tanner CM, Hauser RA, Sethi K, Isaacson S, Truong D, et al. Amantadine extended release for levodopa-induced dyskinesia in Parkinson’s disease (EASED Study) Mov Disord. 2015;30(6):788–795. doi: 10.1002/mds.26159.
    1. Pahwa R, Tanner CM, Hauser RA, Isaacson SH, Nausieda PA, Truong DD, et al. ADS-5102 (Amantadine) extended-release capsules for levodopa-induced dyskinesia in Parkinson disease (EASE LID study): a randomized clinical trial. JAMA Neurol. 2017;74(8):941–949. doi: 10.1001/jamaneurol.2017.0943.
    1. Oertel W, Eggert K, Pahwa R, Tanner CM, Hauser RA, Trenkwalder C, et al. Randomized, placebo-controlled trial of ADS-5102 (amantadine) extended-release capsules for levodopa-induced dyskinesia in Parkinson’s disease (EASE LID 3) Mov Disord. 2017;32(12):1701–1709. doi: 10.1002/mds.27131.
    1. Hughes AJ, Daniel SE, Kilford L, Lees AJ. Accuracy of clinical diagnosis of idiopathic Parkinson’s disease: a clinico-pathological study of 100 cases. J Neurol Neurosurg Psychiatry. 1992;55(3):181–184. doi: 10.1136/jnnp.55.3.181.
    1. Hauser RA, Friedlander J, Zesiewicz TA, Adler CH, Seeberger LC, O’Brien CF, et al. A home diary to assess functional status in patients with Parkinson’s disease with motor fluctuations and dyskinesia. Clin Neuropharmacol. 2000;23(2):75–81. doi: 10.1097/00002826-200003000-00003.
    1. Goetz CG, Tilley BC, Shaftman SR, Stebbins GT, Fahn S, Martinez-Martin P, et al. Movement Disorder Society-sponsored revision of the Unified Parkinson’s Disease Rating Scale (MDS-UPDRS): scale presentation and clinimetric testing results. Mov Disord. 2008;23(15):2129–2170. doi: 10.1002/mds.22340.
    1. Goetz CG, Nutt JG, Stebbins GT. The Unified Dyskinesia Rating Scale: presentation and clinimetric profile. Mov Disord. 2008;23(16):2398–2403. doi: 10.1002/mds.22341.
    1. National Institute of Mental Health, Early clinical drug, evaluation unit (ECDEU) Clinical Global impressions. In: Guy W, editor. ECDEU assessment manual for psychopharmacology. National Institute of Mental Health: Rockville; 1976. pp. 216–222.
    1. Calon F, Rajput AH, Hornykiewicz O, Bedard PJ, Di Paolo T. Levodopa-induced motor complications are associated with alterations of glutamate receptors in Parkinson’s disease. Neurobiol Dis. 2003;14(3):404–416. doi: 10.1016/j.nbd.2003.07.003.
    1. Diederich NJ, Fenelon G, Stebbins G, Goetz CG. Hallucinations in Parkinson disease. Nat Rev Neurol. 2009;5(6):331–342. doi: 10.1038/nrneurol.2009.62.

Source: PubMed

3
订阅