Effectiveness and safety of opicapone in Parkinson's disease patients with motor fluctuations: the OPTIPARK open-label study

Heinz Reichmann, Andrew Lees, José-Francisco Rocha, Diogo Magalhães, Patrício Soares-da-Silva, OPTIPARK investigators, Csaba Antal Zolnai, Claudius Bartels, Andreas Barth, Kriemhild Barth, Stephan Behrens, Arnfin Bergmann, Ralf Bodenschatz, Rommy Born, Moriz Brandt, Sebastian Brock, Bernd Brockmeier, Christof Brücke, Norbert Brüggemann, Bernhard Bühler, Uwe Bungard, Lukas Cepek, Ilona Csoti, Max Deist, Carl Detlev Reimers, Ulrich Dölle, Sylke Domke, Imanuel Dzialowski, Georg Ebersbach, Heike Eggert, Karla Eggert, Reinhard Ehret, Jana Engel, Urban Fietzek, Anke Friedrich, Michael Fritzinger, Florin Gandor, Klaus Gehring, Stephan Gierer, Stephanie Gierer, Vasil Gjaurov, Doreen Gruber, Özkan Günes, Thomas Haas, Kirsten Hahn, Anna Eszter Haraszti, Rolf Hartmann, Bernhard Haslinger, Eva Heiss, Heinz P Herbst, Frank Hoffmann, Werner E Hofmann, Günter Höglinger, Wolfgang Jost, Anna-Maria Kavcic, Christoph Kellinghaus, Bertold Klemperer, Fabian Klostermann, Thomas Knoll, Natalia Koleva-Alazeh, Jiri Koschel, Diana Waltraud Kraft-Safavi, Almut Kronenberger, Andrea Kühn, Andreas Kupsch, Thomas Lehnhoff, Peter Laumen, Paul Lingor, Karla Lippmann, Michael Lorrain, Fabian Maass, Siegfried Muhlack, Thomas Müller, Michael Nagel, Stephan Neudecker, Katja Odin, Christian Oehlwein, Hakan Orbasli, Wolfram von Pannwitz, Heidi Pape, Robert Pfister, Tino Prell, Reinhard Puzich, Daniela Rau, Rene Reese, Gerd Reifschneider, Gernot Reimann, Stefani Ries, Christoph Rieth, Charlotte Rewitzer, Ali Safavi, Alexander B Schmied, Johannes Schwarz, Wolfgang Schwarz, Joachim Springub, Inga Suttrup Claus, Vera Tadic, Klaus Tiel-Wilck, Lars Tönges, Jens Tröger, Christoph Schrey, Alexander Schulze, Sven Thonke, Tobias Wächter, Achim S Wannenmacher, Tobias Warnecke, Bettina Wieder, Martin Wimmer, Christian Winkler, Otto Witte, Dirk Woitalla, Samis Zella, Uwe Ziebold, Jane Alty, Reem Amin, Michaela Boca, Stephen Butterworth, Camille Carroll, Gavin Charlesworth, K Ray Chaudhuri, Rajkumar Chinnadurai, Jemima Collins, Jeremy Stephen Cosgrove, Samantha Cravey, Dinesh Damodaran, Nikolay Dimitrov, Rory Durcan, Simon Ellis, Adbdul Elmarimi, Jonathan Evans, James Fisher, Donald Grosset, Stuart Jamieson, Christopher Kobylecki, Sze Hway Lim, Veronica Lyell, Biju Mohamed, Sophie Molloy, Nicola Pavese, Dominic Paviour, Madeleine Purchas, Khalid Rashed, Christopher Rickards, Tabish Saifee, Gillian Sare, Christine Schofield, Naveen Setty, Jagdish Sharma, Ray Sheridan, Siew Lee Shu, Monty Silverdale, Rani Sophia, Sarah Statton, Malcolm Steiger, Christopher Thomas, Richard Walker, Tai Yen Foung, Heinz Reichmann, Andrew Lees, José-Francisco Rocha, Diogo Magalhães, Patrício Soares-da-Silva, OPTIPARK investigators, Csaba Antal Zolnai, Claudius Bartels, Andreas Barth, Kriemhild Barth, Stephan Behrens, Arnfin Bergmann, Ralf Bodenschatz, Rommy Born, Moriz Brandt, Sebastian Brock, Bernd Brockmeier, Christof Brücke, Norbert Brüggemann, Bernhard Bühler, Uwe Bungard, Lukas Cepek, Ilona Csoti, Max Deist, Carl Detlev Reimers, Ulrich Dölle, Sylke Domke, Imanuel Dzialowski, Georg Ebersbach, Heike Eggert, Karla Eggert, Reinhard Ehret, Jana Engel, Urban Fietzek, Anke Friedrich, Michael Fritzinger, Florin Gandor, Klaus Gehring, Stephan Gierer, Stephanie Gierer, Vasil Gjaurov, Doreen Gruber, Özkan Günes, Thomas Haas, Kirsten Hahn, Anna Eszter Haraszti, Rolf Hartmann, Bernhard Haslinger, Eva Heiss, Heinz P Herbst, Frank Hoffmann, Werner E Hofmann, Günter Höglinger, Wolfgang Jost, Anna-Maria Kavcic, Christoph Kellinghaus, Bertold Klemperer, Fabian Klostermann, Thomas Knoll, Natalia Koleva-Alazeh, Jiri Koschel, Diana Waltraud Kraft-Safavi, Almut Kronenberger, Andrea Kühn, Andreas Kupsch, Thomas Lehnhoff, Peter Laumen, Paul Lingor, Karla Lippmann, Michael Lorrain, Fabian Maass, Siegfried Muhlack, Thomas Müller, Michael Nagel, Stephan Neudecker, Katja Odin, Christian Oehlwein, Hakan Orbasli, Wolfram von Pannwitz, Heidi Pape, Robert Pfister, Tino Prell, Reinhard Puzich, Daniela Rau, Rene Reese, Gerd Reifschneider, Gernot Reimann, Stefani Ries, Christoph Rieth, Charlotte Rewitzer, Ali Safavi, Alexander B Schmied, Johannes Schwarz, Wolfgang Schwarz, Joachim Springub, Inga Suttrup Claus, Vera Tadic, Klaus Tiel-Wilck, Lars Tönges, Jens Tröger, Christoph Schrey, Alexander Schulze, Sven Thonke, Tobias Wächter, Achim S Wannenmacher, Tobias Warnecke, Bettina Wieder, Martin Wimmer, Christian Winkler, Otto Witte, Dirk Woitalla, Samis Zella, Uwe Ziebold, Jane Alty, Reem Amin, Michaela Boca, Stephen Butterworth, Camille Carroll, Gavin Charlesworth, K Ray Chaudhuri, Rajkumar Chinnadurai, Jemima Collins, Jeremy Stephen Cosgrove, Samantha Cravey, Dinesh Damodaran, Nikolay Dimitrov, Rory Durcan, Simon Ellis, Adbdul Elmarimi, Jonathan Evans, James Fisher, Donald Grosset, Stuart Jamieson, Christopher Kobylecki, Sze Hway Lim, Veronica Lyell, Biju Mohamed, Sophie Molloy, Nicola Pavese, Dominic Paviour, Madeleine Purchas, Khalid Rashed, Christopher Rickards, Tabish Saifee, Gillian Sare, Christine Schofield, Naveen Setty, Jagdish Sharma, Ray Sheridan, Siew Lee Shu, Monty Silverdale, Rani Sophia, Sarah Statton, Malcolm Steiger, Christopher Thomas, Richard Walker, Tai Yen Foung

Abstract

Background: The efficacy and safety of opicapone, a once-daily catechol-O-methyltransferase inhibitor, have been established in two large randomized, placebo-controlled, multinational pivotal trials. Still, clinical evidence from routine practice is needed to complement the data from the pivotal trials.

Methods: OPTIPARK (NCT02847442) was a prospective, open-label, single-arm trial conducted in Germany and the UK under clinical practice conditions. Patients with Parkinson's disease and motor fluctuations were treated with opicapone 50 mg for 3 (Germany) or 6 (UK) months in addition to their current levodopa and other antiparkinsonian treatments. The primary endpoint was the Clinician's Global Impression of Change (CGI-C) after 3 months. Secondary assessments included Patient Global Impressions of Change (PGI-C), the Unified Parkinson's Disease Rating Scale (UPDRS), Parkinson's Disease Questionnaire (PDQ-8), and the Non-Motor Symptoms Scale (NMSS). Safety assessments included evaluation of treatment-emergent adverse events (TEAEs) and serious adverse events (SAEs).

Results: Of the 506 patients enrolled, 495 (97.8%) took at least one dose of opicapone. Of these, 393 (79.4%) patients completed 3 months of treatment. Overall, 71.3 and 76.9% of patients experienced any improvement on CGI-C and PGI-C after 3 months, respectively (full analysis set). At 6 months, for UK subgroup only (n = 95), 85.3% of patients were judged by investigators as improved since commencing treatment. UPDRS scores at 3 months showed statistically significant improvements in activities of daily living during OFF (mean ± SD change from baseline: - 3.0 ± 4.6, p < 0.0001) and motor scores during ON (- 4.6 ± 8.1, p < 0.0001). The mean ± SD improvements of - 3.4 ± 12.8 points for PDQ-8 and -6.8 ± 19.7 points for NMSS were statistically significant versus baseline (both p < 0.0001). Most of TEAEs (94.8% of events) were of mild or moderate intensity. TEAEs considered to be at least possibly related to opicapone were reported for 45.1% of patients, with dyskinesia (11.5%) and dry mouth (6.5%) being the most frequently reported. Serious TEAEs considered at least possibly related to opicapone were reported for 1.4% of patients.

Conclusions: Opicapone 50 mg was effective and generally well-tolerated in PD patients with motor fluctuations treated in clinical practice.

Trial registration: Registered in July 2016 at clinicaltrials.gov (NCT02847442).

Keywords: Levodopa; Motor fluctuations; Open-label; Opicapone; Parkinson’s disease.

Conflict of interest statement

HR reports acting on Advisory Boards, gave lectures and received research grants from Abbott, Abbvie, Bayer Health Care, BIAL, Boehringer/Ingelheim, Brittania, Cephalon, Desitin, GSK, Lundbeck, Medtronic, Merck-Serono, Novartis, Orion, Pfizer, TEVA, UCB Pharma, Valeant, and Zambon. AJL is funded by the Reta Lila Weston Institute of Neurological Studies, University College London, Institute of Neurology and reports consultancies from Britannia Pharmaceuticals and BIAL. He also reports grants and/or research support from the Frances and Renee Hock Fund and honoraria from Britannia Pharmaceuticals, Profile Pharma, UCB, Roche, BIAL, STADA, Nordiclnfu Care, and NeuroDerm. JFR, DM and PSS are employed by BIAL - Portela & Cª, S.A.

Figures

Fig. 1
Fig. 1
Patient disposition
Fig. 2
Fig. 2
Global Impression of Change following 3 months treatment with opicapone 50 mg (LOCF) (a) investigator rated (CGI-C, n = 477); (b) self-rated by the patient (PGI-C, n = 393)
Fig. 3
Fig. 3
Presence of PD symptoms as assessed on the WOQ-9 in patients who completed 3 months of study

References

    1. Appleman ER, Stavitsky K, Cronin-Golomb A. Relation of subjective quality of life to motor symptom profile in Parkinson’s disease. Parkinsons Dis. 2011;2011:472830.
    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;20:969–974. doi: 10.1016/j.parkreldis.2014.06.001.
    1. Cilia R, Akpalu A, Sarfo FS, Cham M, Amboni M, Cereda E, et al. The modern pre-levodopa era of Parkinson’s disease: insights into motor complications from sub-Saharan Africa. Brain. 2014;137:2731–2742. doi: 10.1093/brain/awu195.
    1. Olanow CW, Kieburtz K, Rascol O, Poewe W, Schapira AH, Emre M, et al. Factors predictive of the development of levodopa-induced dyskinesia and wearing-off in Parkinson’s disease. Mov Disord. 2013;28:1064–1071. doi: 10.1002/mds.25364.
    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;20:224–230. doi: 10.1002/mds.20279.
    1. Stocchi F, Jenner P, Obeso JA. When do levodopa motor fluctuations first appear in Parkinson’s disease? Eur Neurol. 2010;63:257–266. doi: 10.1159/000300647.
    1. Foundation MJF. Capturing and Elevating the Patient Voice. Available at . Last accessed January 2020.
    1. Fox SH, Katzenschlager R, Lim SY, Barton B, de Bie RMA, Seppi K, et al. International Parkinson and movement disorder society evidence-based medicine review: update on treatments for the motor symptoms of Parkinson’s disease. Mov Disord. 2018;33:1248–1266. doi: 10.1002/mds.27372.
    1. NICE. Parkinson’s disease in adults. Available at: . Last accessed January 2020.
    1. Pahwa R, Factor SA, Lyons KE, Ondo WG, Gronseth G, Bronte-Stewart H, et al. Practice parameter: treatment of Parkinson disease with motor fluctuations and dyskinesia (an evidence-based review). Report of the quality standards Subcommittee of the American Academy of neurology. Neurology. 2006;66:983–995. doi: 10.1212/01.wnl.0000215250.82576.87.
    1. Gordin A, Kaakkola S, Teravainen K. Position of COMT Inhibition in the Treatment of Parkinson’s Disease. In: Gordin A, editor. Parkinson’s Disease: Advances in Neurology. Philadelphia: Lippincott Williams & Wilkins; 2003. pp. 237–250.
    1. Schapira AH, Obeso JA, Olanow CW. The place of COMT inhibitors in the armamentarium of drugs for the treatment of Parkinson’s disease. Neurology. 2000;55(11 Suppl 4):S65–S68.
    1. Poewe W. The role of COMT inhibition in the treatment of Parkinson’s disease. Neurology. 2004;62(1 Suppl 1):S31–S38. doi: 10.1212/WNL.62.1_suppl_1.S31.
    1. Ferreira JJ, Katzenschlager R, Bloem BR, Bonuccelli U, Burn D, Deuschl G, et al. Summary of the recommendations of the EFNS/MDS-ES review on therapeutic management of Parkinson’s disease. Eur J Neurol. 2013;20:5–15. doi: 10.1111/j.1468-1331.2012.03866.x.
    1. Riederer P, Gerlach M, Muller T, Reichmann H. Relating mode of action to clinical practice: dopaminergic agents in Parkinson’s disease. Parkinsonism Relat Disord. 2007;13:466–479. doi: 10.1016/j.parkreldis.2007.06.015.
    1. Kiss LE, Soares-da-Silva P. Medicinal chemistry of catechol O-methyltransferase (COMT) inhibitors and their therapeutic utility. J Med Chem. 2014;57:8692–8717. doi: 10.1021/jm500572b.
    1. Rocha JF, Falcao A, Santos A, Pinto R, Lopes N, Nunes T, et al. Effect of opicapone and entacapone upon levodopa pharmacokinetics during three daily levodopa administrations. Eur J Clin Pharmacol. 2014;70:1059–1071. doi: 10.1007/s00228-014-1701-2.
    1. Ferreira JJ, Lees A, Rocha JF, Poewe W, Rascol O, Soares-da-Silva P, et al. Opicapone as an adjunct to levodopa in patients with Parkinson’s disease and end-of-dose motor fluctuations: a randomised, double-blind, controlled trial. Lancet Neurol. 2016;15:154–165. doi: 10.1016/S1474-4422(15)00336-1.
    1. Lees AJ, Ferreira J, Rascol O, Poewe W, Rocha JF, McCrory M, et al. Opicapone as adjunct to levodopa therapy in patients with Parkinson disease and motor fluctuations: a randomized clinical trial. JAMA Neurol. 2017;74:197–206. doi: 10.1001/jamaneurol.2016.4703.
    1. FDA. Submitting Documents Using Real-World Data and Real-World Evidence to FDA for Drugs and Biologics Guidance for Industry. Available at . Last accessed January 2020.
    1. Cave A, Kurz X, Arlett P. Real-world data for regulatory decision making: challenges and possible solutions for Europe. Clin Pharmacol Ther. 2019;106:36–39. doi: 10.1002/cpt.1426.
    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;55:181–184. doi: 10.1136/jnnp.55.3.181.
    1. Stacy M, Hauser R, Oertel W, Schapira A, Sethi K, Stocchi F, et al. End-of-dose wearing off in Parkinson disease: a 9-question survey assessment. Clin Neuropharmacol. 2006;29:312–321. doi: 10.1097/01.WNF.0000232277.68501.08.
    1. Fahn S, Elton RL. Unified Parkinson’s Disease Rating Scale. Recent developments in Parkinson’s disease. 2: MacMillan Healthcare Information. 1987. pp. 153–164.
    1. Jenkinson C, Fitzpatrick R, Peto V, Greenhalf R, Hyman N. The PDQ-8: development and validation of a short-form parkinson's disease questionnaire. Psychol Health. 1997;12:805–814. doi: 10.1080/08870449708406741.
    1. Chaudhuri KR, Martinez-Martin P, Brown RG, Sethi K, Stocchi F, Odin P, et al. The metric properties of a novel non-motor symptoms scale for Parkinson's disease: results from an international pilot study. Mov Disord. 2007;22:1901–1911. doi: 10.1002/mds.21596.
    1. Frieden TR. Evidence for health decision making - beyond randomized, Controlled Trials. N Engl J Med. 2017;377:465–475. doi: 10.1056/NEJMra1614394.
    1. Ferreira JJ, Lees A, Rocha JF, Poewe W, Rascol O, Soares-da-Silva P. Long-term efficacy of opicapone in fluctuating Parkinson's disease patients: a pooled analysis of data from two phase 3 clinical trials and their open-label extensions. Eur J Neurol. 2019;26:953–960.
    1. Corvol JC, Bonnet C, Charbonnier-Beaupel F, Bonnet AM, Fievet MH, Bellanger A, et al. The COMT Val158Met polymorphism affects the response to entacapone in Parkinson's disease: a randomized crossover clinical trial. Ann Neurol. 2011;69:111–118. doi: 10.1002/ana.22155.
    1. Shulman LM, Gruber-Baldini AL, Anderson KE, Fishman PS, Reich SG, Weiner WJ. The clinically important difference on the unified Parkinson’s disease rating scale. Arch Neurol. 2010;67:64–70. doi: 10.1001/archneurol.2009.295.
    1. Hauser RA, Auinger P. Determination of minimal clinically important change in early and advanced Parkinson’s disease. Mov Disord. 2011;26:813–818. doi: 10.1002/mds.23638.
    1. Hauser RA, Gordon MF, Mizuno Y, Poewe W, Barone P, Schapira AH, et al. Minimal clinically important difference in Parkinson's disease as assessed in pivotal trials of pramipexole extended release. Parkinsons Dis. 2014;2014:467131.
    1. Chaudhuri KR, Schapira AH. Non-motor symptoms of Parkinson’s disease: dopaminergic pathophysiology and treatment. Lancet Neurol. 2009;8:464–474. doi: 10.1016/S1474-4422(09)70068-7.
    1. Chaudhuri KR, Yates L, Martinez-Martin P. The non-motor symptom complex of Parkinson’s disease: a comprehensive assessment is essential. Curr Neurol Neurosci Rep. 2005;5:275–283. doi: 10.1007/s11910-005-0072-6.
    1. Franke C, Storch A. Nonmotor fluctuations in Parkinson’s disease. Int Rev Neurobiol. 2017;134:947–971. doi: 10.1016/bs.irn.2017.05.021.
    1. Storch A, Schneider CB, Wolz M, Sturwald Y, Nebe A, Odin P, et al. Nonmotor fluctuations in Parkinson disease: severity and correlation with motor complications. Neurology. 2013;80:800–809. doi: 10.1212/WNL.0b013e318285c0ed.
    1. Fabbri M, Ferreira JJ, Lees A, Stocchi F, Poewe W, Tolosa E, et al. Opicapone for the treatment of Parkinson's disease: a review of a new licensed medicine. Mov Disord. 2018;33:1528–1539. doi: 10.1002/mds.27475.
    1. Lees A, Ferreira JJ, Rocha JF, Rascol O, Poewe W, Soares-da-Silva PRI. Safety profile of Opicapone in the Management of Parkinson’s disease. J Park Dis. 2019;9:733–740.
    1. Larsen JP, Worm-Petersen J, Siden A, Gordin A, Reinikainen K, Leinonen M. The tolerability and efficacy of entacapone over 3 years in patients with Parkinson’s disease. Eur J Neurol. 2003;10:137–146. doi: 10.1046/j.1468-1331.2003.00559.x.
    1. Ahn TB, Im JH, Lee MC, Kim JW, Lee WY, Jeon BS. One-year open-label study of entacapone in patients with advanced Parkinson disease. J Clin Neurol. 2007;3:82–85. doi: 10.3988/jcn.2007.3.2.82.
    1. Papapetropoulos SS. Patient diaries as a clinical endpoint in Parkinson’s disease clinical trials. CNS Neurosci Ther. 2012;18:380–387. doi: 10.1111/j.1755-5949.2011.00253.x.

Source: PubMed

3
Sottoscrivi