Skip to main content
Log in

Opicapone: A Review in Parkinson’s Disease

  • Adis Drug Evaluation
  • Published:
CNS Drugs Aims and scope Submit manuscript

Abstract

Oral opicapone (Ongentys®), a potent third-generation, peripheral catechol-O-methyltransferase (COMT) inhibitor, is approved as adjunctive therapy to preparations of levodopa/dopa-decarboxylase inhibitor (L-dopa/DDCI) in adults with Parkinson’s disease (PD) and end-of dose (EoD) motor fluctuations. In pivotal global trials (BIPARK 1 and BIPARK 2; 14–15 weeks’ duration), open-label extensions (OLEs) of BIPARK, and in the real-world setting (OPTIPARK; 3–6 months), opicapone 50 mg once daily was an effective and generally well tolerated adjunctive therapy to L-dopa/DDCI plus other PD therapy in adults with PD and EoD motor fluctuations. Adjunctive opicapone provided better efficacy than placebo for improvements in ON- and OFF-state time and fulfilled noninferiority to adjunctive entacapone for improvements in OFF time in BIPARK 1. These beneficial effects of adjunctive opicapone on motor fluctuations were maintained during 1 year of treatment in OLE studies. Given its efficacy and safety profile, adjunctive opicapone remains an important option in the management of adults with PD and EoD motor fluctuations who cannot be stabilized on preparations of L-dopa/DDCI.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Klietz M, Greten S, Wegner F, et al. Safety and tolerability of pharmacotherapies for Parkinson’s disease in geriatric patients. Drugs Aging. 2019;36(6):511–30.

    CAS  PubMed  Google Scholar 

  2. Vijiaratnam N, Foltynie T. Therapeutic strategies to treat or prevent Off episodes in adults with Parkinson’s disease. Drugs. 2020;80(8):775–96.

    CAS  PubMed  Google Scholar 

  3. Katsaiti I, Nixon J. Are there benefits in adding catechol-O methyltransferase inhibitors in the pharmacotherapy of Parkinson’s disease patients? A systematic review. J Parkinsons Dis. 2018;8(2):217–31.

    CAS  PubMed  Google Scholar 

  4. Goetz CG, Pal G. Initial management of Parkinson’s disease. Br Med J. 2014;349:g6258.

    Google Scholar 

  5. Poewe W, Seppi K, Tanner CM, et al. Parkinson disease. Nat Rev Dis Primers. 2017;3:17013.

    PubMed  Google Scholar 

  6. Fackrell R, Carroll CB, Grosset DG, et al. Noninvasive options for “wearing-off” in Parkinson’s disease: a clinical consensus from a panel of UK Parkinson’s disease specialists. Neurodegener Dis Manag. 2018;8(5):349–60.

    PubMed  Google Scholar 

  7. Fabbri M, Rosa MM, Ferreira JJ. Adjunctive therapies in Parkinson’s disease: how to choose the best treatment strategy approach. Drugs Aging. 2018;35(12):1041–54.

    PubMed  Google Scholar 

  8. Cabreira V, Soares-da-Silva P, Massano J. Contemporary options for the management of motor complications in Parkinson’s disease: updated clinical review. Drugs. 2019;79(6):593–608.

    CAS  PubMed  Google Scholar 

  9. BIAL-Portela. Ongentys: EU summary of product characteristics. 2020. http://www.ema.europa.eu/. Accessed 9 Sep 2020.

  10. BIAL-Portela. ONGENTYS (opicapone): US prescribing information. 2020. http://www.accessdata.fda.gov/. Accessed 9 Sep 2020.

  11. Scott LJ. Opicapone: a review in Parkinson’s disease. Drugs. 2016;76(13):1293–300.

    CAS  PubMed  Google Scholar 

  12. Scott LJ. Opicapone in Parkinson’s disease: a profile of its use. Drugs Ther Perspect. 2017;33(7):303–10.

    Google Scholar 

  13. Rocha JF, Sicard É, Fauchoux N, et al. Effect of opicapone multiple-dose regimens on levodopa pharmacokinetics. Br J Clin Pharmacol. 2017;83(3):540–53.

    CAS  PubMed  Google Scholar 

  14. Loewen G, LeWitt P, Olanow CW, et al. The pharmacokinetics and pharmacodynamics of once-daily opicapone 50 mg in patients with parkinson’s disease on levodopa/carbidopa: results of a phase 1 study [abstract no. 129 plus poster]. Mov Disord Clin Pract. 2020;7(Suppl 1):S56.

    Google Scholar 

  15. Nomoto M, Takeda A, Iwai K, et al. Effect of opicapone tablets on levodopa and 3-O-methyldopa pharmacokinetics in healthy Japanese subjects: phase 1 study. Clin Pharmacol Drug Dev. 2020. https://doi.org/10.1002/cpdd.799.

    Article  PubMed  Google Scholar 

  16. Loewen G, Liang G, Jimenez R, et al. Effect of once-daily opicapone on the pharmacokinetics of repaglinide [abstract no. 1654]. Neurology. 2020;94(15 Suppl).

  17. Almeida L, Rocha JF, Falcao A, et al. Pharmacokinetics, pharmacodynamics and tolerability of opicapone, a novel catechol-O-methyltransferase inhibitor, in healthy subjects: prediction of slow enzyme-inhibitor complex dissociation of a short-living and very long-acting inhibitor. Clin Pharmacokinet. 2013;52(2):139–51.

    CAS  PubMed  Google Scholar 

  18. Bonifacio MJ, Sutcliffe JS, Torrao L, et al. Brain and peripheral pharmacokinetics of levodopa in the cynomolgus monkey following administration of opicapone, a third generation nitrocatechol COMT inhibitor. Neuropharmacology. 2014;77:334–41.

    CAS  PubMed  Google Scholar 

  19. Bonifacio MJ, Torrao L, Loureiro AI, et al. Pharmacological profile of opicapone, a third-generation nitrocatechol catechol-O-methyl transferase inhibitor, in the rat. Br J Pharmacol. 2015;172(7):1739–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Falcão A, Rocha JF, Santos A, et al. Opicapone pharmacokinetics and pharmacodynamics comparison between healthy Japanese and matched white subjects. Clin Pharmacol Drug Dev. 2016;5(2):150–61.

    PubMed  Google Scholar 

  21. Ferreira JJ, Rocha JF, Falcao A, et al. Effect of opicapone on levodopa pharmacokinetics, catechol-O-methyltransferase activity and motor fluctuations in patients with Parkinson’s disease. Eur J Neurol. 2015;22(5):815–25, e56.

    CAS  PubMed  Google Scholar 

  22. Kiss LE, Ferreira HS, Torrao L, et al. Discovery of a long-acting, peripherally selective inhibitor of catechol-O-methyltransferase. J Med Chem. 2010;53(8):3396–411.

    CAS  PubMed  Google Scholar 

  23. Pinto R, l’Hostis P, Patat A, et al. Evaluation of opicapone on cardiac repolarization in a thorough QT/QTc study. Clin Pharmacol Drug Develop. 2015;4(6):454–62.

    CAS  Google Scholar 

  24. Rocha JF, Almeida L, Falcao A, et al. Opicapone: a short lived and very long acting novel catechol-O-methyltransferase inhibitor following multiple dose administration in healthy subjects. Br J Clin Pharmacol. 2013;76(5):763–75.

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Rocha JF, Falcao A, Santos A, et al. Effect of opicapone and entacapone upon levodopa pharmacokinetics during three daily levodopa administrations. Eur J Clin Pharmacol. 2014;70:1059–71.

    CAS  PubMed  Google Scholar 

  26. Rocha JF, Santos A, Falcao A, et al. Effect of moderate liver impairment on the pharmacokinetics of opicapone. Eur J Clin Pharmacol. 2014;70(3):279–86.

    CAS  PubMed  Google Scholar 

  27. Rocha J-F, Ferreira JJ, Falcao A, et al. Effect of 3 single-dose regimens of opicapone on levodopa pharmacokinetics, catechol-O-methyltransferase activity and motor response in patients with Parkinson disease. Clin Pharmacol Drug Dev. 2015;5(3):232–40.

    PubMed  Google Scholar 

  28. Ferreira JJ, Lees A, Rocha JF, 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(2):154–65.

    CAS  PubMed  Google Scholar 

  29. Lees AJ, Ferreira J, Rascol O, et al. Opicapone as adjunct to levodopa therapy in patients with Parkinson disease and motor fluctuations: a randomized clinical trial. JAMA Neurol. 2017;74(2):197–206.

    PubMed  Google Scholar 

  30. Takeda A, Takahashi R, Tsuboi Y, et al. Randomized, controlled study of opicapone in Japanese Parkinson’s patients with motor fluctuations. Mov Disord. 2020. https://doi.org/10.1002/mds.28322.

    Article  PubMed  Google Scholar 

  31. Ferreira JJ, Lees A, Rocha JF, et al. 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(7):953–60.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Hauser R, Lew M, Kremens D, et al. Efficacy and safety of once-daily opicapone 50 mg in patients with Parkinson’s disease and motor fluctuations: pooled analysis of two randomized, double-blind, placebo-controlled studies [abstract no. 147 + poster]. Mov Disord Clin Pract. 2020;7(Suppl 1):S65.

    Google Scholar 

  33. Lew M, Hauser RA, Ferreira J, et al. Symptom improvements with once-daily opicapone in patients with Parkinson’s disease and motor fluctuations: results from a phase 3, randomized clinical trial (BIPARK-1) [abstract + poster 1]. Mov Disord. 2019;34(Suppl 1):S2.

    Google Scholar 

  34. Hauser RA, Lew M, Lees AJ, et al. BIPARK-2 results: efficacy of once-daily opicapone for the treatment of patients with Parkinson’s disease and motor fluctuations [abstract + poster 2]. Mov Disord. 2019;34(Suppl 1):S2–3.

    Google Scholar 

  35. Korlipara P, Ferreira J, Gama H, et al. Baseline off-time influence in opicapone’s efficacy outcomes in Parkinson’s disease patients with motor fluctuations: the BIPARK-I double-blind data [abstract no. EPO2169]. Eur J Neurol. 2019;26(Suppl 1):649.

    Google Scholar 

  36. Kulisevsky J, Tolosa E, Ferreira J, et al. Results from the BIPARK-I double-blind experience on the efficacy of opicapone in Parkinson’s disease patients with “early” motor fluctuations [abstract no. EPO2171]. Eur J Neurol. 2019;26(Suppl 1):650.

    Google Scholar 

  37. Lew M, Hauser R, Isaacson S, et al. Efficacy of opicapone compared to entacapone in Parkinson’s patients with motor fluctuations and ON Hoehn and Yahr ≤ 2.5: a post hoc analysis of BIPARK-1 [abstract no. 4429 plus oral presentation]. Neurology. 2020;94(15 Suppl):4429.

    Google Scholar 

  38. Rocha J, Stocchi F, Lees A, et al. Efficacy of opicapone in Parkinson’s disease patients with “early” motor fluctuations: patient and Clinical Global Impression of Change from the BIPARK-I double-blind experience [abstract no. 189]. Mov Disord. 2019;34(Suppl 2):S79.

    Google Scholar 

  39. Ferreira J, Poewe W, Rascol O, et al. Association of patient global impression of change and other endpoints of efficacy in Parkinson’s disease patients: a posthoc analysis from combined BIPARK-I and II data [abstract no. EPO1201]. Eur J Neurol. 2019;26(Suppl 1):467.

    Google Scholar 

  40. Antonini A, Poewe W, Ferreira J, et al. Super-responders to opicapone adjunct treatment to levodopa in parkinson’s disease patients with motor fluctuations: combined post-hoc analysis of BIPARK-I and II [abstract no. EPO1182]. Eur J Neurology. 2020;27(Suppl 1):631–2.

    Google Scholar 

  41. Antonini A, Ferreira J, Poewe W, et al. Impact of opicapone treatment in nonmotor symptoms scale in Parkinson’s disease patients with “early” motor fluctuations: data from the BIPARK-I double-blind experience [abstract no. EPO1171]. Eur J Neurol. 2019;26(Suppl 1):449.

    Google Scholar 

  42. Rocha J, Tolosa E, Ferreira J, et al. Effect of opicapone in Parkinson’s disease patients with “early” motor fluctuations: Parkinson’s Disease Questionnaire (PDQ-39) analysis from the BIPARK-I double-blind experience [abstract no. 188]. Mov Disord. 2019;34(Suppl 2):S78–9.

    Google Scholar 

  43. Gama H, Ferreira J, Lees A, et al. Influence of disease duration in the efficacy response of Parkinson’s disease patients with motor fluctuations: post-hoc analysis from combined BIPARK-I and II [abstract no. 93]. Mov Disord. 2019;34(Suppl 2):S41.

    Google Scholar 

  44. Santos A, Lees A, Ferreira J, et al. Influence of motor fluctuations duration, levodopa dose and duration of use in efficacy responses of Parkinson’s disease patients: post-hoc analysis from combined BIPARK-I and II [abstract no. 195]. Mov Disord. 2019;34(Suppl 2):S81.

    Google Scholar 

  45. Lees A, Ferreira J, Rascol O, et al. Efficacy of opicapone in Parkinson’s disease patients according to baseline rasagiline use: a post-hoc analysis from combined BIPARK-I and II [abstract no. 135]. Mov Disord. 2019a;34(Suppl 2):S57.

    Google Scholar 

  46. Lees A, Ferreira J, Rascol O, et al. Efficacy of opicapone in Parkinson’s disease patients according to baseline pramipexole use: a post-hoc analysis from combined BIPARK-I and II [abstract no. 134]. Mov Disord. 2019b;34(Suppl 2):S57.

    Google Scholar 

  47. Ebersbach G, Ferreira JJ, Antonini A, et al. Opicapone's added benefit as first-line adjunctive levodopa and when used promptly in the motor-fluctuation spectrum of Parkinson's disease: a post hoc analysis of BIPARK-I and II [abstract]. In: Movement Disorders Society Virtual Congress. 2020.

  48. Lees A, Ferreira J, Antonini A, et al. Efficacy of opicapone in Parkinson’s disease patients according to baseline presence of dyskinesia: a post-hoc analysis from combined BIPARK-I and II [abstract no. 133]. Mov Disord. 2019;34(Suppl 2):S56–7.

    Google Scholar 

  49. Videnovic A, Poewe W, Lees A, et al. Effect of opicapone and entacapone on early-morning-oFF pattern in Parkinson's disease patients with motor fluctuations [abstract]. In: Movement Disorders Society Virtual Congress. 2020.

  50. LeWitt P, Ferreira JJ, Ebersbach G, et al. Effect of opicapone and entacapone on levodopa short duration response [abstract]. In: Movement Disorders Society Virtual Congress. 2020.

  51. Lees A, Rascol O, Ferreira JJ, et al. Effect of opicapone and entacapone on daily pattern of motor fluctuations Parkinson's disease patients [abstract]. In: Movement Disorders Society Virtual Congress. 2020.

  52. Hauser R, Rascol O, Poewe W, et al. Opicapone as a levodopa sparing agent: pooled analysis of BIPARK-I and II double-blind trials [abstract no. 105]. Mov Disord. 2019;34(Suppl 2):S46.

    Google Scholar 

  53. Ferreira JJ, Lees AJ, Poewe W, et al. Effectiveness of opicapone and switching from entacapone in fluctuating Parkinson disease. Neurology. 2018;90(21):e1849–57.

    CAS  PubMed  Google Scholar 

  54. Takahashi R, Tsuboi Y, Nomoto M, et al. An open-label, 1-year extension clinical study in Japan of opicapone treatment for Parkinson’s disease-Comfort-PD study part 2 [abstract no. 212]. Mov Disord. 2019;34(Suppl 2):S88–9.

    Google Scholar 

  55. Tolosa E, Ferreira J, Lees A, et al. Changes in activities of daily living and motor function in patients switching from entacapone or placebo to opicapone who ended BIPARK-I extension on opicapone 50mg [abstract no. EPO3175]. Eur J Neurol. 2020;27(Suppl 1):1123.

    Google Scholar 

  56. Ferreira J, Lees A, Poewe W, et al. Switching entacapone “non-responders” to open-label opicapone: change in absolute OFF-time following the 1-year extension BIPARK-I study [abstract no. 90]. Mov Disord. 2019;34(Suppl 2):S40.

    Google Scholar 

  57. Reichmann H, Lees A, Rocha JF, et al. Effectiveness and safety of opicapone in Parkinson’s disease patients with motor fluctuations: the OPTIPARK open-label study. Transl Neurodegener. 2020;9:1–9.

    Google Scholar 

  58. Muñoz Ruiz T, Pons Pons G, Garcia Trujillo L, et al. Which factors predict the succes of opicapone? Defining the optimal patient profile [abstract no. EPO3131]. Eur J Neurol. 2020;27(Suppl 1):1095.

    Google Scholar 

  59. Lees A, Ferreira JJ, Rocha JF, et al. Safety profile of opicapone in the management of Parkinson’s disease. J Parkinsons Dis. 2019;9(4):733–40.

    CAS  PubMed  Google Scholar 

  60. Costa R, Lees A, Ferreira J, et al. Incidence of treatment-emergent adverse events in Parkinson’s disease patients according to baseline disease severity: post-hoc analysis from double-blind combined BIPARK-I and II data [abstract no. 82]. Mov Disord. 2019;34(Suppl 2):S37.

    Google Scholar 

  61. Costa R, Ferreira J, Lees A, et al. Incidence of treatment-emergent adverse events in Parkinson’s disease patients according to baseline MAO-B inhibitors use: post-hoc analysis from double-blind combined BIPARK-I and II data [abstract no. 81]. Mov Disord. 2019;34(Suppl 2):S36–7.

    Google Scholar 

  62. Ikedo F, Lees A, Ferreira J, et al. Incidence of treatment-emergent adverse events in Parkinson’s disease patients according to baseline dopamine agonist use: post-hoc analysis from double-blind combined BIPARK-I and II data [abstract no. 110]. Mov Disord. 2019;34(Suppl 2):S48.

    Google Scholar 

  63. Ikedo F, Ferreira J, Lees A, et al. Incidence of treatment-emergent adverse events in Parkinson’s disease patients according to baseline body mass index: post-hoc analysis from double-blind combined BIPARK-I and II data [abstract no. 109]. Mov Disord. 2019a;34(Suppl 2):S47.

    Google Scholar 

  64. Ikedo F, Ferreira J, Lees A, et al. Incidence of treatment-emergent adverse events in Parkinson’s disease patients according to gender: post-hoc analysis from double-blind combined BIPARK-I and II data [abstract no. 111]. Mov Disord. 2019b;34(Suppl 2):S48.

    Google Scholar 

  65. Lees A, Reichmann H, Rocha J-F, et al. Onset of drug-related adverse events in Parkinson's disease patients with motor fluctuations treated with opicapone in clinical practice: OPTIPARK post-hoc analysis [abstract]. In: Movement Disorders Society Virtual Congress. 2020.

  66. Fox SH, Katzenschlager R, Lim SY, 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(8):1248–66.

    CAS  Google Scholar 

  67. National Institute for Health and Clinical Excellence. Parkinson's disease in adults: NICE guideline NG71. 2017. http://www.nice.org.uk/guidance/ng71. Accessed 5 Oct 2020.

  68. Corvol J-C, Bonnet C, Charbonnier-Beaupel F, et al. The COMTVal158Met polymorphism effects the response to entacapone in Parkinson’s disease: a randomized crossover trial. Ann Neurol. 2011;69:111–8.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

During the peer review process, the manufacturer of the agent under review was also offered an opportunity to review this article. Changes resulting from comments received were made on the basis of scientific and editorial merit.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Lesley J. Scott.

Ethics declarations

Funding

The preparation of this review was not supported by any external funding.

Authorship and Conflict of interest

Lesley Scott is a salaried employee of Adis International Ltd/Springer Nature, and declares no relevant conflicts of interest. All authors contributed to the review and are responsible for the article content.

Ethics approval, Consent to participate, Consent to publish, Availability of material, Code availability

Not applicable.

Additional information

The manuscript was reviewed by: A. Antonini, Parkinson’s Disease & Movement Disorders Unit, IRCCS Hospital San Camillo, and 1st Neurology Clinic, Padua University Hospital, Venice, Italy; F. Brugger, Klinik für Neurologie, Kantonsspital St. Gallen, St Gallen, Switzerland; P. J. Garcia Ruiz, Department of Neurology, Fundacion Jimenez Diaz, Madrid, Spain; S. Grill, Parkinson’s Disease & Movement Disorders Center of Maryland, Elkridge, MD, USA; K. Jellinger, Institute of Clinical Neurobiology, Vienna, Austria.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Scott, L.J. Opicapone: A Review in Parkinson’s Disease. CNS Drugs 35, 121–131 (2021). https://doi.org/10.1007/s40263-020-00778-6

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40263-020-00778-6

Navigation