Skip to main content
Log in

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

  • Original Research Article
  • Published:
Clinical Pharmacokinetics Aims and scope Submit manuscript

Abstract

Background and Objectives

Opicapone is a novel catechol-O-methyltransferase (COMT) inhibitor. The purpose of this study was to evaluate the tolerability, pharmacokinetics (including the effect of food) and pharmacodynamics (effect on COMT activity) following single oral doses of opicapone in young healthy male volunteers.

Methods

Single rising oral doses of opicapone (10, 25, 50, 100, 200, 400, 800 and 1,200 mg) were administered to eight groups of eight subjects per group (two subjects randomized to placebo and six subjects to opicapone), under a double-blind, randomized, placebo-controlled design. In an additional group of 12 subjects, a 50 mg single dose of opicapone was administered on two occasions, once having fasted overnight and once with a high-fat high-calorie meal.

Results

Opicapone was well tolerated at all doses tested. The extent of systemic exposure (area under the plasma concentration–time curve and maximum plasma concentration) to opicapone and metabolites increased in an approximately dose-proportional manner and showed a decrease following concomitant ingestion of a high-fat high-calorie meal. The apparent terminal elimination half-life of opicapone was 0.8–3.2 h. Sulphation appeared to be the main metabolic pathway for opicapone, and both opicapone and the main sulphated metabolite are likely excreted by the biliary route. Maximum COMT inhibition by opicapone was dose dependent, ranged from 36.1 % (10 mg) to 100 % (200 mg and above), and reached statistical significance at all doses tested. The long duration of COMT inhibition by opicapone, however, tended to be independent from the dose taken. The observed half-life of opicapone-induced COMT inhibition in human erythrocytes was 61.6 h (standard deviation [SD] = 37.6 h), which reflects an underlying dissociative process with a kinetic rate constant of 3.1 × 10−6 s−1 (SD = 1.9 × 10−6 s−1). Such a process compares well to the estimated dissociation rate constant (koff) of the COMT–opicapone molecular complex (koff = 1.9 × 10−6 s−1).

Conclusions

Opicapone was well-tolerated and presented dose-proportional kinetics. Opicapone demonstrated marked and sustained inhibition of erythrocyte soluble COMT activity. Based on the observation that the half-life of COMT inhibition is independent of the dose and that it reflects an underlying kinetic process that is consistent with the koff value of the COMT–opicapone complex, we propose that the sustained COMT inhibition, far beyond the observable point of clearance of circulating drug, is due to the long residence time of the reversible complex formed between COMT and opicapone. Globally, these promising results provide a basis for further clinical development of opicapone.

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
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

References

  1. Morgan JC, Sethi KD. Emerging drugs for Parkinson’s disease. Expert Opin Emerg Drugs. 2006;11(3):403–17.

    Article  PubMed  CAS  Google Scholar 

  2. Schapira AH, Emre M, Jenner P, et al. Levodopa in the treatment of Parkinson’s disease. Eur J Neurol. 2009;16(9):982–9.

    Article  PubMed  CAS  Google Scholar 

  3. Palma PN, Bonifácio MJ, Almeida L, et al. Restoring dopamine levels. In: Simth HJ, Simons C, Sewell RDE, editors. Protein misfolding in neurodegenerative diseases: mechanisms and therapeutic strategies. Boca Raton: CRC Press; 2007. p. 415–45.

    Google Scholar 

  4. Bonifácio MJ, Palma PN, Almeida L, et al. Catechol-O-methyltransferase and its inhibitors in Parkinson’s disease. CNS Drug Rev. 2007;13:352–79.

    Article  PubMed  Google Scholar 

  5. Nutt JG, Woodward WR, Gancher ST, et al. 3-O-methyldopa and the response to levodopa in Parkinson’s disease. Ann Neurol. 1987;21(6):584–8.

    Article  PubMed  CAS  Google Scholar 

  6. Benetello P, Furlanut M, Zara G, et al. Plasma levels of levodopa and its main metabolites in parkinsonian patients after conventional and controlled-release levodopa-carbidopa associations. Eur Neurol. 1993;33(1):69–73.

    Article  PubMed  CAS  Google Scholar 

  7. Wade LA, Katzman R. 3-O-methyldopa uptake and inhibition of L-dopa at the blood-brain barrier. Life Sci. 1975;17(1):131–6.

    Article  PubMed  CAS  Google Scholar 

  8. Gervas JJ, Muradas V, Bazan E, et al. Effects of 3-OM-dopa on monoamine metabolism in rat brain. Neurology. 1983;33(3):278–82.

    Article  PubMed  CAS  Google Scholar 

  9. Gomes P, Soares-da-Silva P. Interaction between L-DOPA and 3-O-methyl-L-DOPA for transport in immortalised rat capillary cerebral endothelial cells. Neuropharmacology. 1999;38(9):1371–80.

    Article  PubMed  CAS  Google Scholar 

  10. Baas H, Zehrden F, Selzer R, et al. Pharmacokinetic-pharmacodynamic relationship of levodopa with and without tolcapone in patients with Parkinson’s disease. Clin Pharmacokinet. 2001;40(5):383–93.

    Article  PubMed  CAS  Google Scholar 

  11. Muller T, Kolf K, Ander L, et al. Catechol-O-methyltransferase inhibition improves levodopa-associated strength increase in patients with Parkinson disease. Clin Neuropharmacol 2008;31(3):134–40.

    Google Scholar 

  12. Benetello P, Furlanut M, Fortunato M, et al. Levodopa and 3-O-methyldopa in cerebrospinal fluid after levodopa-carbidopa association. Pharmacol Res. 1997;35(4):313–5.

    Article  PubMed  CAS  Google Scholar 

  13. Guttman M, Leger G, Cedarbaum JM, et al. 3-O-methyldopa administration does not alter fluorodopa transport into the brain. Ann Neurol. 1992;31(6):638–43.

    Article  PubMed  CAS  Google Scholar 

  14. Himori N, Mishima K. The COMT inhibitor tolcapone potentiates the anticataleptic effect of Madopar in MPP(+)-lesioned mice. Experientia. 1994;50(10):939–42.

    Article  PubMed  CAS  Google Scholar 

  15. Keranen T, Gordin A, Karlsson M, 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  CAS  Google Scholar 

  16. Dingemanse J, Jorga KM, Schmitt M, 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  PubMed  CAS  Google Scholar 

  17. Baas H, Beiske AG, Ghika J, et al. Catechol-O-methyltransferase inhibition with tolcapone reduces the “wearing off” phenomenon and levodopa requirements in fluctuating parkinsonian patients. Neurology. 1998;50(5 Suppl 5):S46–53.

    Article  PubMed  CAS  Google Scholar 

  18. Kuoppamaki M, Korpela K, Marttila R, et al. Comparison of pharmacokinetic profile of levodopa throughout the day between levodopa/carbidopa/entacapone and levodopa/carbidopa when administered four or five times daily. Eur J Clin Pharmacol. 2009;65(5):443–55.

    Article  PubMed  Google Scholar 

  19. Brooks DJ. Safety and tolerability of COMT inhibitors. Neurology. 2004;62(1 Suppl 1):S39–46.

    Article  PubMed  CAS  Google Scholar 

  20. Dingemanse J. Issues important for rational COMT inhibition. Neurology. 2000;55 (11 Suppl 4):S24-7; discussion S8-32.

  21. Lang AE, Lees A. Management of Parkinson’s disease: an evidence-based review. Mov Disord. 2002;17(Suppl 4):S45–51.

    Google Scholar 

  22. Nissinen H, Kuoppamaki M, Leinonen M, et al. Early versus delayed initiation of entacapone in levodopa-treated patients with Parkinson’s disease: a long-term, retrospective analysis. Eur J Neurol. 2009;16(12):1305–11.

    Article  PubMed  CAS  Google Scholar 

  23. Ferreira JJ, Rascol O, Poewe W, et al. A double-blind, randomized, placebo and active-controlled study of nebicapone for the treatment of motor fluctuations in Parkinson’s disease. CNS Neurosci Ther. 2010;16:337–47.

    Article  PubMed  CAS  Google Scholar 

  24. Learmonth DA, Kiss LE, Soares-da-Silva P. The chemistry of catechol-O-methyltransferase inhibitors. Int Rev Neurobiol. 2010;95:119–62.

    Article  PubMed  CAS  Google Scholar 

  25. 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.

    Article  PubMed  CAS  Google Scholar 

  26. Palma PN, Bonifácio MJ, Loureiro AI, et al. Computation of the binding affinities of catechol-O-methyltransferase inhibitors: multisubstate relative free energy calculations. J Comput Chem. 2012;33(9):970–86.

    Article  PubMed  CAS  Google Scholar 

  27. Bonifácio MJ, Sutcliffe JS, Torrão L, et al. Brain and peripheral levodopa pharmacokinetics in the Cynomolgus monkey following administration of opicapone, a novel catechol-O-methyltransferase inhibitor. Parkinsonism Relat D. 2012;18(S2):S125.

    Google Scholar 

  28. Bonifácio MJ, Torrão L, Loureiro AI, et al. Opicapone: characterization of a novel peripheral long-acting catechol-O-methyltransferase inhibitior. Parkinsonism Relat D 2012;18(S2):S125.

    Google Scholar 

  29. Data on file, Bial, 2012.

  30. FDA/CDER. Guidance for industry: estimating the maximum safe starting dose in initial clinical trials for therapeutics in healthy adult volunteers. U.S. Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER). Rockville: FDA/CDER; 2005.

  31. Schultz E, Nissinen E, Kaakkola S. Determination of catechol-O-methyltransferase activity in erythrocytes by high performance liquid chromatography with electrochemical detection. Biomed Chromatogr. 1989;3(2):64–7.

    Article  PubMed  CAS  Google Scholar 

  32. FDA/CDER. Guidance for industry (draft). Food-effect bioavailability and fed bioequivalence studies: study design, data analysis, and labeling. U.S. Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER). Rockville: FDA/CDER; 2001.

  33. Silveira P, Vaz-da-Silva M, Almeida L, et al. Pharmacokinetic-pharmacodynamic interaction between BIA 3–202, a novel COMT inhibitor, and levodopa/benserazide. Eur J Clin Pharmacol. 2003;59(8–9):603–9.

    Article  PubMed  CAS  Google Scholar 

  34. Data on file, Bial, 2008.

  35. Ferreira JJ, Almeida L, Cunha L, et al. Effects of nebicapone on levodopa pharmacokinetics, catechol-O-methyltransferase activity, and motor fluctuations in patients with Parkinson disease. Clin Neuropharmacol. 2008;31(1):2–18.

    Google Scholar 

  36. Copeland RA, Pompliano D. Drug–target residence time and its implications for lead optimization. Nat Rev Drug Discov. 2006;1–10.

  37. Tummino PJ, Copeland RA. Residence time of receptor-ligand complexes and its effect on biological function. Biochemistry. 2008;47:5481–92.

    Article  PubMed  CAS  Google Scholar 

  38. Vauquelin G, Van Liefde I. Slow antagonist dissociation and long-lasting in vivo receptor protection. Trends Pharmacol Sci. 2006;27:356–9.

    Article  PubMed  Google Scholar 

  39. Palma PN, Bonifácio MJ, Loureiro AI, et al. Computation of binding affinity of catechol-O-methyltransferase: opicapone complex. Parkinsonism Relat D 2012;18 (S2):S125.

    Google Scholar 

  40. Vidgren J, Svensson LA, Liljas A. Crystal structure of catechol O-methyltransferase. Nature. 1994;368:354–8.

    Article  PubMed  CAS  Google Scholar 

  41. Rocha JF, Nunes T, Vaz-da-silva M, et al. Pharmacokinetics, pharmacodynamics and tolerability of opicapone, a novel COMT inhibitor, during multiple rising dose regimens in healthy male subjects [abstract]. Parkinsonism Relat D 2012;18 (S2):S126.

    Google Scholar 

Download references

Acknowledgments

This study was sponsored by BIAL - Portela & Cª, S.A.

Conflicts of interest

The authors are (Dr Rocha, Dr Palma, Dr Loureiro, Dr Pinto, Dr Bonifácio, Dr Wright, Dr Nunes and Dr Soares-da-Silva) or were (Dr Almeida) employees of Bial – Portela & Cª S.A., the sponsor of the study. A. Falcão received consultancy honoraria from BIAL – Portela & Cª, S.A.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Patrício Soares-da-Silva.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (PDF 73 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Almeida, L., Rocha, J.F., Falcão, A. et al. Pharmacokinetics, Pharmacodynamics and Tolerability of Opicapone, a Novel Catechol-O-Methyltransferase Inhibitor, in Healthy Subjects. Clin Pharmacokinet 52, 139–151 (2013). https://doi.org/10.1007/s40262-012-0024-7

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40262-012-0024-7

Keywords

Navigation