Skip to main content
Log in

COMT Inhibitors in the Management of Parkinson’s Disease

  • Review Article
  • Published:
CNS Drugs Aims and scope Submit manuscript

Abstract

Levodopa treatment remains the gold standard for Parkinson’s disease, but shortcomings related to the pharmacological profile, notably, oral administration and the consequent occurrence of motor complications, have led to the development of several add-on levodopa treatments or to research to improve the method of delivery. Motor fluctuations, and to a lesser extent non-motor fluctuations, concern half of the patients with Parkinson’s disease after 5 years of disease and patients identified them as one of their most bothersome symptoms. Catechol-O-methyl transferase inhibitors (COMT-Is) are one of the recommended first-line levodopa add-on therapies for the amelioration of end-of dose motor fluctuations in patient with advanced Parkinson’s disease. Currently, two peripheral COMT-Is are considered as first-line choices - entacapone (ENT), which was approved by the US Food and Drug Administration in 1999 and the European Committee in 1998; and opicapone (OPC), which was approved by the European Committee in 2016. A second-line COMT-I that requires regular hepatic monitoring, tolcapone (TOL), was approved by the Food and Drug Administration in 1998 and the European Committee in 1997. Of note, OPC also received Food and Drug Administration approval in 2021, but it is still only marketed in a few countries, including Germany, UK, Spain, Portugal, Italy, Japan, and USA, while ENT and TOL have a wider market. Our narrative review summarizes the pharmacokinetic/pharmacodynamic properties, clinical efficacy in terms of motor fluctuations, motor/non-motor symptoms, quality of life, and safety data of these three COMT-Is, as evidenced by randomized clinical trials, as well as by real-life observational studies. Overall, a phase III non-inferiority trial showed a similar effect between ENT and OPC on off-time (−60.8 min/day and −40.3 min/day, vs placebo, respectively), with a possible additional off-time reduction of 39 min/day, obtained when there is a switch from ENT to OPC. Concomitantly, TOL can reduce off-time by an average of 98 min/day. A significant though discrete concomitant reduction on the Unified Parkinson's Disease Rating Scale motor section (2–3 points) is obtained with all three drugs vs placebo. Data on quality of life are fewer and more heterogeneous, with positive results obtained especially in open-label studies. Effects on non-motor symptoms were investigated as secondary outcome only in a few studies, frequently by means of non-specific scales and a benefit was observed in open-label studies. Dopaminergic adverse effects were the most frequent, dyskinesia being the most common for the three drugs eventually requiring levodopa dose reductions. No urine discoloration and a very low incidence of diarrhea were found with OPC compared with ENT and TOL. Regular hepatic monitoring is needed only for TOL. A combination of COMT-Is with new formulations of levodopa, including the subcutaneous, intrajejunal, or new extended-release formulation, merits further exploration to improve the management of both mild and severe motor fluctuations.

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.

Fig. 1

Similar content being viewed by others

References

  1. Dorsey ER, Bloem BR. The Parkinson pandemic: a call to action. JAMA Neurol. 2018;75(1):9–10.

    Article  PubMed  Google Scholar 

  2. LeWitt PA. Levodopa therapy for Parkinson’s disease: pharmacokinetics and pharmacodynamics. Move Disord. 2015;30(1):64–72.

    Article  CAS  Google Scholar 

  3. Ray Chaudhuri K, Poewe W, Brooks D. Motor and nonmotor complications of levodopa: phenomenology, risk factors, and imaging features. Move Disord. 2018;33(6):909–19.

    Article  CAS  Google Scholar 

  4. Rizos A, Martinez-Martin P, Odin P, Antonini A, Kessel B, Kozul TK, et al. Characterizing motor and non-motor aspects of early-morning off periods in Parkinson’s disease: an international multicenter study. Parkinsonism Relat Disord. 2014;20(11):1231–5.

    Article  CAS  PubMed  Google Scholar 

  5. Greig SL, McKeage K. Carbidopa/levodopa ER capsules (Rytary®, Numient™): a review in Parkinson’s disease. CNS Drugs. 2016;30(1):79–90.

    Article  CAS  PubMed  Google Scholar 

  6. Olanow CW, Poewe W, Rascol O, Stocchi F. On-demand therapy for OFF episodes in Parkinson’s disease. Move Disord. 2021;36(10):2243–53.

    Article  Google Scholar 

  7. Dijk JM, Espay AJ, Katzenschlager R, de Bie RMA. The choice between advanced therapies for Parkinson’s disease patients: why, what, and when? J Parkinsons Dis. 2020;10(s1):S65-73.

    Article  PubMed  PubMed Central  Google Scholar 

  8. 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.

    Article  PubMed  Google Scholar 

  9. 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. Move Disord. 2018;33(8):1248–66.

    Article  CAS  Google Scholar 

  10. Schrag A. Entacapone in the treatment of Parkinson’s disease. Lancet Neurol. 2005;4(6):366–70.

    Article  CAS  PubMed  Google Scholar 

  11. Li J, Lou Z, Liu X, Sun Y, Chen J. Efficacy and safety of adjuvant treatment with entacapone in advanced Parkinson’s disease with motor fluctuation: a systematic meta-analysis. Eur Neurol. 2017;78(3–4):143–53.

    Article  PubMed  Google Scholar 

  12. 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. Move Disord. 2018;33(10):1528–39.

    Article  Google Scholar 

  13. Artusi CA, Sarro L, Imbalzano G, Fabbri M, Lopiano L. Safety and efficacy of tolcapone in Parkinson’s disease: systematic review. Eur J Clin Pharmacol. 2021;77(6):817–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Poewe W. The role of COMT inhibition in the treatment of Parkinson’s disease. Neurology. 2004;62(1 Suppl. 1):S31–8.

    Article  CAS  PubMed  Google Scholar 

  15. Ruottinen HM, Rinne UK. COMT inhibition in the treatment of Parkinson’s disease. J Neurol. 1998;245(11 Suppl. 3):P25-34.

    Article  CAS  PubMed  Google Scholar 

  16. Dingemanse J, Jorga K, Zürcher G, Fotteler B, Sedek G, Nielsen T, et al. Multiple-dose clinical pharmacology of the catechol-O-methyl-transferase inhibitor tolcapone in elderly subjects. Eur J Clin Pharmacol. 1996;50(1–2):47–55.

    Article  CAS  PubMed  Google Scholar 

  17. Kiss LE, Ferreira HS, Torrao L, Bonifacio MJ, Palma PN, Soares-da-Silva P, et al. Discovery of a long-acting, peripherally selective inhibitor of catechol-O-methyltransferase. J Med Chem. 2010;53(8):3396–411.

    Article  CAS  PubMed  Google Scholar 

  18. Palma N, Bonifacio MJ, Loureiro AI, Soares-Da-Silva P. Computation of binding affinity of catechol-O-methyltransferase-opicapone complexes. Parkinsonism Relat Disord. 2012;18:S125.

    Article  Google Scholar 

  19. Bonifacio MJ, Torrao L, Loureiro AI, Palma PN, Wright LC, Soares-da-Silva P. Pharmacological profile of opicapone, a third-generation nitrocatechol catechol-O-methyl transferase inhibitor, in the rat. Br J Pharmacol. 2015;172(7):1739–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Bicker J, Alves G, Fortuna A, Soares-da-Silva P, Falcao A. A new PAMPA model using an in-house brain lipid extract for screening the blood-brain barrier permeability of drug candidates. Int J Pharm. 2016;501(1–2):102–11.

    Article  CAS  PubMed  Google Scholar 

  21. Truong DD. Tolcapone: review of its pharmacology and use as adjunctive therapy in patients with Parkinson’s disease. Clin Interv Aging. 2009;4:109–13.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Dingemanse J, Jorga K, Zürcher G, Schmitt M, Sedek G, Da Prada M, et al. Pharmacokinetic-pharmacodynamic interaction between the COMT inhibitor tolcapone and single-dose levodopa. Br J Clin Pharmacol. 1995;40(3):253–62.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Prada MBJ, Napolitano A. Improved therapy of Parkinson’s disease with tolcapone, a central and peripheral COMT inhibitor with an S-adenosyl-L-methionine-sparing effect. Clin Neuropharmacol. 1994;17:S26–37.

    Article  Google Scholar 

  24. Sêdek G, Jorga K, Schmitt M, Burns RS, Leese P. Effect of tolcapone on plasma levodopa concentrations after coadministration with levodopa/carbidopa to healthy volunteers. Clin Neuropharmacol. 1997;20(6):531–41.

    Article  PubMed  Google Scholar 

  25. Keränen T, Gordin A, Karlsson M, Korpela K, Pentikäinen PJ, Rita H, et al. Inhibition of soluble catechol-O-methyltransferase and single-dose pharmacokinetics after oral and intravenous administration of entacapone. Eur J Clin Pharmacol. 1994;46(2):151–7.

    Article  PubMed  Google Scholar 

  26. Ruottinen HM, Rinne UK. A double-blind pharmacokinetic and clinical dose-response study of entacapone as an adjuvant to levodopa therapy in advanced Parkinson’s disease. Clin Neuropharmacol. 1996;19(4):283–96.

    Article  CAS  PubMed  Google Scholar 

  27. Wikberg T, Vuorela A, Ottoila P, Taskinen J. Identification of major metabolites of the catechol-O-methyltransferase inhibitor entacapone in rats and humans. Drug Metab Dispos. 1993;21(1):81–92.

    CAS  PubMed  Google Scholar 

  28. Lyytinen J, Kaakkola S, Ahtila S, Tuomainen P, Teräväinen H. Simultaneous MAO-B and COMT inhibition in l-dopa-treated patients with Parkinson’s disease. Move Disord. 1997;12(4):497–505.

    Article  CAS  Google Scholar 

  29. Nutt JG, Woodward WR, Beckner RM, Stone CK, Berggren K, Carter JH, et al. Effect of peripheral catechol-O-methyltransferase inhibition on the pharmacokinetics and pharmacodynamics of levodopa in parkinsonian patients. Neurology. 1994;44(5):913–9.

    Article  CAS  PubMed  Google Scholar 

  30. Ruottinen HM, Rinne UK. Entacapone prolongs levodopa response in a one month double blind study in parkinsonian patients with levodopa related fluctuations. J Neurol Neurosurg Psychiatry. 1996;60(1):36–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Loureiro A, Fernandes-Lopes C, Wright L, et al. Sulfation of opicapone, a nitrocatechol-type COMT inhibitor, by human recombinant SULTs and human S9 fraction. Abstract, XX World Congress on Parkinson’s Disease and Related Disorders 2013, Switzerland.

  32. Almeida L, Rocha JF, Falcao A, Palma PN, Loureiro AI, Pinto R, 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.

    Article  CAS  PubMed  Google Scholar 

  33. Santos AFA, Rocha J, Soares-Da-Silva P. Influence of food on opicapone pharmacokinetics and pharmacodynamics. Eur J Neurol. 2017;24(Suppl. 1):123–444.

    Google Scholar 

  34. 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(9):1059–71.

    Article  CAS  PubMed  Google Scholar 

  35. Rocha JF, Falcao A, Lopes N, Pinto R, Santos A, Nunes T, et al. Opicapone effect on levodopa pharmacokinetics in comparison with placebo and entacapone when administered with immediate release 100/25 mg levodopa/carbidopa in healthy subjects. J Neurol. 2014;261:S119.

    Google Scholar 

  36. Falcão ASA, Ferreira JJ, Rocha J, Soares-Da-Silva P. Opicapone’s bedtime regimen and the decision-making process. Eur J Neurol. 2017;24.

  37. Zürcher G, Da Prada M, Dingemanse J. Assessment of catechol-O-methyltransferase activity and its inhibition in erythrocytes of animals and humans. BMC. 1996;10(1):32–6.

    PubMed  Google Scholar 

  38. Dingemanse J, Jorga KM, Schmitt M, Gieschke R, Fotteler B, Zürcher G, et al. Integrated pharmacokinetics and pharmacodynamics of the novel catechol-O-methyltransferase inhibitor tolcapone during first administration to humans. Clin Pharmacol Ther. 1995;57(5):508–17.

    Article  CAS  PubMed  Google Scholar 

  39. Jorga KM, Sedek G, Fotteler B, Zürcher G, Nielsen T, Aitken JW. Optimizing levodopa pharmacokinetics with multiple tolcapone doses in the elderly. Clin Pharmacol Ther. 1997;62(3):300–10.

    Article  CAS  PubMed  Google Scholar 

  40. Ruottinen HM, Rinne UK. Effect of one month’s treatment with peripherally acting catechol-O-methyltransferase inhibitor, entacapone, on pharmacokinetics and motor response to levodopa in advanced parkinsonian patients. Clin Neuropharmacol. 1996;19(3):222–33.

    Article  CAS  PubMed  Google Scholar 

  41. Rocha JF, Almeida L, Falcao A, Palma PN, Loureiro AI, Pinto R, 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Ferreira JJ, Rocha JF, Falcao A, Santos A, Pinto R, Nunes T, et al. Effects of opicapone on levodopa pharmacokinetics, catechol-O-methyltransferase activity and motor fluctuations in patients with Parkinson's disease. Eur J Neurol. 2015;22(5):815–25e56.

  43. Ferreira JJ, Lees A, Rocha JF, Poewe W, Rascol O, Soares-da-Silva P. 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.

    Article  CAS  PubMed  Google Scholar 

  44. Factor SA, Molho ES, Feustel PJ, Brown DL, Evans SM. Long-term comparative experience with tolcapone and entacapone in advanced Parkinson’s disease. Clin Neuropharmacol. 2001;24(5):295–9.

    Article  CAS  PubMed  Google Scholar 

  45. Koller W, Lees A, Doder M, Hely M. Randomized trial of tolcapone versus pergolide as add-on to levodopa therapy in Parkinson’s disease patients with motor fluctuations. Move Disord. 2001;16(5):858–66.

    Article  CAS  Google Scholar 

  46. Efficacy and tolerability of tolcapone compared with bromocriptine in levodopa-treated parkinsonian patients. Tolcapone Study Group. Move Disord. 1999;14(1):38–44.

  47. Stowe R, Ives N, Clarke CE, Deane K, Wheatley K, Gray R, et al. Evaluation of the efficacy and safety of adjuvant treatment to levodopa therapy in Parkinson's disease patients with motor complications. Cochrane Database Syst Rev. 2010;7:CD007166.

  48. Poewe WH, Deuschl G, Gordin A, Kultalahti ER, Leinonen M. Efficacy and safety of entacapone in Parkinson’s disease patients with suboptimal levodopa response: a 6-month randomized placebo-controlled double-blind study in Germany and Austria (Celomen study). Acta Neurol Scand. 2002;105(4):245–55.

    Article  CAS  PubMed  Google Scholar 

  49. Piccini P, Brooks DJ, Korpela K, Pavese N, Karlsson M, Gordin A. The catechol-O-methyltransferase (COMT) inhibitor entacapone enhances the pharmacokinetic and clinical response to Sinemet CR in Parkinson’s disease. J Neurol Neurosurg Psychiatry. 2000;68(5):589–94.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. 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(2):197–206.

    Article  PubMed  Google Scholar 

  51. Takeda A, Takahashi R, Tsuboi Y, Nomoto M, Maeda T, Nishimura A, et al. Randomized, controlled study of opicapone in Japanese Parkinson’s patients with motor fluctuations. Move Disord. 2021;36(2):415–23.

    Article  CAS  Google Scholar 

  52. 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(7):953–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  54. Takeda ATR, Tsuboi Y, Nomoto M, Maeda T, Nishimura A, Hattori N. An open-label, 1-year extension clinical study in Japan of opicapone treatment for Parkinson’s disease: comfort-PD study part 2. Move Disord. 2019;34(S2).

  55. Stocchi F, Rascol O, Kieburtz K, Poewe W, Jankovic J, Tolosa E, et al. Initiating levodopa/carbidopa therapy with and without entacapone in early Parkinson disease: the STRIDE-PD study. Ann Neurol. 2010;68(1):18–27.

    Article  CAS  PubMed  Google Scholar 

  56. Warren Olanow C, 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. Move Disord. 2013;28(8):1064–71.

    Article  CAS  Google Scholar 

  57. Hauser RA, Panisset M, Abbruzzese G, Mancione L, Dronamraju N, Kakarieka A. Double-blind trial of levodopa/carbidopa/entacapone versus levodopa/carbidopa in early Parkinson’s disease. Move Disord. 2009;24(4):541–50.

    Article  Google Scholar 

  58. Velseboer DC, Broeders M, Post B, van Geloven N, Speelman JD, Schmand B, et al. Prognostic factors of motor impairment, disability, and quality of life in newly diagnosed PD. Neurology. 2013;80(7):627–33.

    Article  PubMed  Google Scholar 

  59. Schrag A, Dodel R, Spottke A, Bornschein B, Siebert U, Quinn NP. Rate of clinical progression in Parkinson’s disease: a prospective study. Move Disord. 2007;22(7):938–45.

    Article  Google Scholar 

  60. Fabbri M, Coelho M, Abreu D, Guedes LC, Rosa MM, Godinho C, et al. Dysphagia predicts poor outcome in late-stage Parkinson’s disease. Parkinsonism Relat Disord. 2019;64:73–81.

    Article  PubMed  Google Scholar 

  61. Schrag A, Sampaio C, Counsell N, Poewe W. Minimal clinically important change on the unified Parkinson’s disease rating scale. Move Disord. 2006;21(8):1200–7.

    Article  Google Scholar 

  62. Waters CH, Kurth M, Bailey P, Shulman LM, LeWitt P, Dorflinger E, et al. Tolcapone in stable Parkinson’s disease: efficacy and safety of long-term treatment. Tolcapone Stable Study Group. Neurology. 1998;50(5 Suppl. 5):S39-45.

    Article  CAS  PubMed  Google Scholar 

  63. Onofrj M, Thomas A, Iacono D, Di Iorio A, Bonanni L. Switch-over from tolcapone to entacapone in severe Parkinson’s disease patients. Eur Neurol. 2001;46(1):11–6.

    Article  CAS  PubMed  Google Scholar 

  64. Entacapone to tolcapone switch. Multicenter double-blind, randomized, active-controlled trial in advanced Parkinson’s disease. Move Disord. 2007;22(1):14–9.

    Article  Google Scholar 

  65. Larsen JP, Worm-Petersen J, Sidén 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(2):137–46.

    Article  CAS  PubMed  Google Scholar 

  66. Hauser RRO, Poewe W, Ferreira JJ, Lees A, Klepitskaya G, Liang G, et al. Opicapone as a levodopa sparing agent: pooled analysis of BIPARK-I and II double-blind trials. Move Disord. 2019;34(Suppl. S2):S46.

  67. Adler CH, Singer C, O'Brien C, Hauser RA, Lew MF, Marek KL, et al. Randomized, placebo-controlled study of tolcapone in patients with fluctuating Parkinson disease treated with levodopa-carbidopa. Tolcapone Fluctuator Study Group III. Arch Neurol. 1998;55(8):1089–95.

  68. Welsh MD, Dorflinger E, Chernik D, Waters C. Illness impact and adjustment to Parkinson’s disease: before and after treatment with tolcapone. Move Disord. 2000;15(3):497–502.

    Article  CAS  Google Scholar 

  69. Baas H, Beiske AG, Ghika J, Jackson M, Oertel WH, Poewe W, et al. Catechol-O-methyltransferase inhibition with tolcapone reduces the “wearing off” phenomenon and levodopa requirements in fluctuating parkinsonian patients. J Neurol Neurosurg Psychiatry. 1997;63(4):421–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Reichmann H, Boas J, Macmahon D, Myllyla V, Hakala A, Reinikainen K. Efficacy of combining levodopa with entacapone on quality of life and activities of daily living in patients experiencing wearing-off type fluctuations. Acta Neurol Scand. 2005;111(1):21–8.

    Article  CAS  PubMed  Google Scholar 

  71. Fénelon G, Giménez-Roldán S, Montastruc JL, Bermejo F, Durif F, Bourdeix I, et al. Efficacy and tolerability of entacapone in patients with Parkinson’s disease treated with levodopa plus a dopamine agonist and experiencing wearing-off motor fluctuations: a randomized, double-blind, multicentre study. J Neural Transm (Vienna). 2003;110(3):239–51.

    Article  Google Scholar 

  72. Grandas F, Hernandez B. Long-term effectiveness and quality of life improvement in entacapone-treated Parkinson’s disease patients: the effects of an early therapeutic intervention. Eur J Neurol. 2007;14(3):282–9.

    Article  CAS  PubMed  Google Scholar 

  73. Gershanik O, Emre M, Bernhard G, Sauer D. Efficacy and safety of levodopa with entacapone in Parkinson’s disease patients suboptimally controlled with levodopa alone, in daily clinical practice: an international, multicentre, open-label study. Prog Neuropsychopharmacol Biol Psychiatry. 2003;27(6):963–71.

    Article  CAS  PubMed  Google Scholar 

  74. Durif F, Devaux I, Pere JJ, Delumeau JC, Bourdeix I. Efficacy and tolerability of entacapone as adjunctive therapy to levodopa in patients with Parkinson’s disease and end-of-dose deterioration in daily medical practice: an open, multicenter study. Eur Neurol. 2001;45(2):111–8.

    Article  CAS  PubMed  Google Scholar 

  75. Reichmann H, Lees A, Rocha JF, Magalhães D, Soares-da-Silva P. Effectiveness and safety of opicapone in Parkinson’s disease patients with motor fluctuations: the OPTIPARK open-label study. Transl Neurodegener. 2020;9(1):9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Schapira AHV, Chaudhuri KR, Jenner P. Non-motor features of Parkinson disease. Nat Rev Neurosci. 2017;18(7):435–50.

    Article  CAS  PubMed  Google Scholar 

  77. Müller T. Tolcapone addition improves Parkinson’s disease associated nonmotor symptoms. Ther Adv Neurol Disord. 2014;7(2):77–82.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Ebersbach G, Hahn K, Lorrain M, Storch A. Tolcapone improves sleep in patients with advanced Parkinson's disease (PD). Arch Gerontol Geriatr. 2010;51(3):e125–8.

  79. Gasparini M, Fabrizio E, Bonifati V, Meco G. Cognitive improvement during tolcapone treatment in Parkinson’s disease. J Neural Transm (Vienna). 1997;104(8–9):887–94.

    Article  CAS  Google Scholar 

  80. Oliveira C, Lees A, Ferreira J, Lopes N, Costa R, Pinto R, et al. Evaluation of non-motor symptoms in opicapone treated Parkinson’s disease patients: results from a double-blind, randomized, placebo-controlled study and open-label extension. Eur J Neurol. 2015;22:191.

    Google Scholar 

  81. Hernán MA, Checkoway H, O’Brien R, Costa-Mallen P, De Vivo I, Colditz GA, et al. MAOB intron 13 and COMT codon 158 polymorphisms, cigarette smoking, and the risk of PD. Neurology. 2002;58(9):1381–7.

    Article  PubMed  Google Scholar 

  82. Corvol JC, Bonnet C, Charbonnier-Beaupel F, Bonnet AM, Fiévet 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(1):111–8.

    Article  CAS  PubMed  Google Scholar 

  83. Olanow CW, Kieburtz K, Odin P, Espay AJ, Standaert DG, Fernandez HH, et al. Continuous intrajejunal infusion of levodopa-carbidopa intestinal gel for patients with advanced Parkinson’s disease: a randomised, controlled, double-blind, double-dummy study. Lancet Neurol. 2014;13(2):141–9.

    Article  CAS  PubMed  Google Scholar 

  84. Nyholm D, Johansson A, Lennernäs H, Askmark H. Levodopa infusion combined with entacapone or tolcapone in Parkinson disease: a pilot trial. Eur J Neurol. 2012;19(6):820–6.

    Article  CAS  PubMed  Google Scholar 

  85. Senek M, Nyholm D, Nielsen EI. Population pharmacokinetics of levodopa gel infusion in Parkinson’s disease: effects of entacapone infusion and genetic polymorphism. Sci Rep. 2020;10(1):18057.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Öthman M, Widman E, Nygren I, Nyholm D. Initial experience of the levodopa-entacapone-carbidopa intestinal gel in clinical practice. J Pers Med. 2021;11(4):254.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Schröter N, Ahrendt S, Hager A, Rijntjes M. Addition of tolcapone in intrajejunal levodopa infusion therapy requires a pronounced cost reduction. Move Disord. 2020;7(7):854–6.

    Google Scholar 

  88. Leta V, van Wamelen DJ, Sauerbier A, Jones S, Parry M, Rizos A, et al. Opicapone and levodopa-carbidopa intestinal gel infusion: the way forward towards cost savings for healthcare systems? J Parkinsons Dis. 2020;10(4):1535–9.

    Article  PubMed  Google Scholar 

  89. Giladi N, Gurevich T, Djaldetti R, Adar L, Case R, Leibman-Barak S, et al. ND0612 (levodopa/carbidopa for subcutaneous infusion) in patients with Parkinson’s disease and motor response fluctuations: a randomized, placebo-controlled phase 2 study. Parkinsonism Relat Disord. 2021;91:139–45.

    Article  PubMed  Google Scholar 

  90. Assal F, Spahr L, Hadengue A, Rubbia-Brandt L, Burkhard PR. Tolcapone and fulminant hepatitis. Lancet. 1998;352(9132):958.

    Article  CAS  PubMed  Google Scholar 

  91. Olanow CW. Tolcapone and hepatotoxic effects. Tasmar Advisory Panel Arch Neurol. 2000;57(2):263–7.

    Article  CAS  PubMed  Google Scholar 

  92. Olanow CW, Watkins PB. Tolcapone: an efficacy and safety review (2007). Clin Neuropharmacol. 2007;30(5):287–94.

    Article  CAS  PubMed  Google Scholar 

  93. Entacapone improves motor fluctuations in levodopa-treated Parkinson's disease patients. Parkinson Study Group. Ann Neurol. 1997;42(5):747–55.

  94. Rinne UK, Larsen JP, Siden A, Worm-Petersen J. Entacapone enhances the response to levodopa in parkinsonian patients with motor fluctuations. Nomecomt Study Group. Neurology. 1998;51(5):1309–14.

    Article  CAS  PubMed  Google Scholar 

  95. Brooks DJ, Sagar H, Group UK-IES. Entacapone is beneficial in both fluctuating and non-fluctuating patients with Parkinson's disease: a randomised, placebo controlled, double blind, six month study. J Neurol Neurosurg Psychiatry. 2003;74(8):1071–9.

  96. Myllyla VV, Kultalahti ER, Haapaniemi H, Leinonen M, Group FS. Twelve-month safety of entacapone in patients with Parkinson's disease. Eur J Neurol. 2001;8(1):53–60.

  97. Liao X, Wu N, Liu D, Shuai B, Li S, Li K. Levodopa/carbidopa/entacapone for the treatment of early Parkinson’s disease: a meta-analysis. Neurol Sci. 2020;41(8):2045–54.

    Article  PubMed  Google Scholar 

  98. Haapaniemi HRK, Leinonen M. Tolerability and safety of entacapone in the treatment of Parkinson’s disease. Parkinsonism Relat Disord. 2000;7(Suppl. 1):S57.

    Google Scholar 

  99. Castro Caldas A, Teodoro T, Ferreira JJ. The launch of opicapone for Parkinson’s disease: negatives versus positives. Expert Opin Drug Saf. 2018;17(3):331–7.

    Article  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  101. Lees ARH, Rocha JF, Magalhães D, Soares-da-Silva P. Onset of drug-related adverse events in Parkinson’s disease patients with motor fluctuations treated with opicapone in clinical practice: OPTIPARK post-hoc analysis. Mov Disord. 2020;35.

  102. Hauser RA, Hsu A, Kell S, Espay AJ, Sethi K, Stacy M, et al. Extended-release carbidopa-levodopa (IPX066) compared with immediate-release carbidopa-levodopa in patients with Parkinson’s disease and motor fluctuations: a phase 3 randomised, double-blind trial. Lancet Neurol. 2013;12(4):346–56.

    Article  CAS  PubMed  Google Scholar 

  103. Deuschl G, Vaitkus A, Fox GC, Roscher T, Schremmer D, Gordin A. Efficacy and tolerability of entacapone versus cabergoline in parkinsonian patients suffering from wearing-off. Move Disord. 2007;22(11):1550–5.

    Article  Google Scholar 

  104. Stocchi F, Hsu A, Khanna S, Ellenbogen A, Mahler A, Liang G, et al. Comparison of IPX066 with carbidopa-levodopa plus entacapone in advanced PD patients. Parkinsonism Relat Disord. 2014;20(12):1335–40.

    Article  PubMed  Google Scholar 

  105. Ferreira JJLA, Poewe P, Rascol O, Santos A, Magalhães D, Rocha JF, Soares-da-Silva P. Switching entacapone ‘non-responders’ to open-label opicapone: change in absolute OFF-time following the 1-year extension BIPARKI study. Move Disord. 2019;34(Suppl. S2):S1–937.

  106. Hauser RALM, Videnovic A, Poewe Q, Rascol O, Ferreira JJ, Liang GS, et al. Effects of once-daily opicapone on duration of overnight OFF and time to morning ON in patients with Parkinson’s disease and motor fluctuations. Ann Neurol. 2020;88(Suppl 25): S189.

  107. Videnovic APW, Lees A, Ferreira JJ, Klepitskaya O, Loureiro R, Magalhães D, et al. Effect of opicapone and entacapone on early morning-OFF pattern in Parkinson’s disease patients with motor fluctuations. Move Disord. 2020;34(Suppl. S2):S486.

  108. Hansen RN, Suh K, Serbin M, Yonan C, Sullivan SD. Cost-effectiveness of opicapone and entacapone in reducing OFF-time in Parkinson’s disease patients treated with levodopa/carbidopa. J Med Econ. 2021;24(1):563–9.

    Article  PubMed  Google Scholar 

  109. Jenner P, McCreary AC, Scheller DK. Continuous drug delivery in early- and late-stage Parkinson’s disease as a strategy for avoiding dyskinesia induction and expression. J Neural Transm (Vienna). 2011;118(12):1691–702.

    Article  CAS  Google Scholar 

  110. Jenner P, Rocha JF, Ferreira JJ, Rascol O, Soares-da-Silva P. Redefining the strategy for the use of COMT inhibitors in Parkinson’s disease: the role of opicapone. Expert Rev Neurother. 2021;21(9):1019–33.

    Article  CAS  PubMed  Google Scholar 

  111. Martínez-Fernández R, Schmitt E, Martinez-Martin P, Krack P. The hidden sister of motor fluctuations in Parkinson’s disease: a review on nonmotor fluctuations. Move Disord. 2016;31(8):1080–94.

    Article  Google Scholar 

  112. Ebersbach GRO, Ferreira J, Costa R, Rocha JF, Magalhães D, Soares-da-Silva P. Efficacy and safety of opicapone in Parkinson’s disease patients according to duration of motor fluctuations: post-hoc analysis of BIPARK-I and II. Move Disord. 2020;35(Suppl. 1):35.

    Google Scholar 

  113. 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(1):5–15.

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Olivier Rascol.

Ethics declarations

Funding

No specific funding was received for this work.

Conflicts of interest

Margherita Fabbri declares the following conflicts of interest: honoraria to speak: BIAL and AbbVie. Joaquim J. Ferreira declares the following conflicts of interest: consultancy for BIAL and principal investigator of the BIPARK I study; stock ownership in medically related fields: none; consultancies: Ipsen, GlaxoSmithKline, Novartis, Teva, Lundbeck, Solvay, Abbott, Merck-Serono, and Merz; advisory boards: none; partnership: none; honoraria to speak: none; grants: GlaxoSmithKline, Grunenthal, Teva, and Fundação MSD. Olivier Rascol declares the following conflicts of interest: advisory board and consultancy: BIAL; advisory boards and consultancy: AbbVie, Adamas, Acorda, Addex, AlzProtect, Apopharma, Astrazeneca, Axovant, Biogen, Britannia, Buckwang, Cerespir, Clevexel, Denali, INC Reasearch, Lundbeck, Lupin, Merck, MundiPharma, Neuratris, Neuroderm, Novartis, ONO Pharma, Osmotica, Parexel, Pfizer, Prexton Therapeutics, Quintiles, Roche, Sanofi, Servier, Sunovion, Théranexus, Takeda, Teva, UCB, Vectura, Watermark Research, XenoPort, XO, and Zambon; grant: Agence Nationale de la Recherche (ANR), CHU de Toulouse, France-Parkinson, INSERM-DHOS Recherche Clinique Translationnelle, MJFox Foundation, Programme Hospitalier de Recherche Clinique, European Commission (FP7, H2020), and Cure Parkinson IK; other: grant to participate in a symposium and contribute to the review of an IPMDS article.

Ethics approval

Not applicable.

Consent to participate

Not applicable.

Consent for publication

Not applicable.

Availability of data and material

Not applicable.

Code availability

Not applicable.

Author contributions

All co-authors have been substantially involved in the preparation of the manuscript; all the co-authors have also seen and approved the final submitted version and agreed to be accountable for the work. 1. Research project: A. Conception, B. Organization, C. Execution; 2. Statistical Analysis: A. Design, B. Execution, C. Review and Critique; 3. Manuscript Preparation: A. Writing of the first draft, B. Review and Critique. MF: 1A, 1B, 1C, 3A; JJF: 1A, 1B, 3B; OR: 1A, 2A, 3B.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fabbri, M., Ferreira, J.J. & Rascol, O. COMT Inhibitors in the Management of Parkinson’s Disease. CNS Drugs 36, 261–282 (2022). https://doi.org/10.1007/s40263-021-00888-9

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40263-021-00888-9

Navigation