358
Views
36
CrossRef citations to date
0
Altmetric
Drug Profile

Levodopa/carbidopa/entacapone in Parkinson’s disease

&
Pages 929-940 | Published online: 09 Jan 2014

References

  • Lilienfeld DE, Perl DP. Projected neurodegenerative disease mortality in the United States, 1990–2040. Neuroepidemiology12(4), 219–228 (1993).
  • Tanner CM, Goldman SM. Epidemiology of Parkinson’s disease. Neurol. Clin.14(2), 317–335 (1996).
  • Marras C, Tanner C. Epidemiology of Parkinson’s Disease. In: Movement Disorders: Neurologic Principles and Practice (Second Edition). Watts R, Koller W (Eds). McGraw–Hill Medical Publishing, NY, USA 177–195 (2004).
  • Lang AE, Lozano AM. Parkinson’s disease. First of two parts. N. Engl. J. Med.339(15), 1044–1053 (1998).
  • Gilks WP, Abou-Sleiman PM, Gandhi S et al. A common LRRK2 mutation in idiopathic Parkinson’s disease. Lancet365(9457), 415–416 (2005).
  • Hely MA, Morris JG, Reid WG, Trafficante R. Sydney Multicenter study of Parkinson’s disease: non-L-dopa-responsive problems dominate at 15 years. Mov. Disord.20(2), 190–199 (2005).
  • Hely MA, Reid WG, Adena MA, Halliday GM, Morris JG. The Sydney multicenter study of Parkinson’s disease: the inevitability of dementia at 20 years. Mov. Disord.23(6), 837–844 (2008).
  • Pramipexole vs levodopa as initial treatment for Parkinson disease: a randomized controlled trial. Parkinson Study Group. JAMA284(15), 1931–1938 (2000).
  • Rascol O, Brooks DJ, Korczyn AD, De Deyn PP, Clarke CE, Lang AE. A five-year study of the incidence of dyskinesia in patients with early Parkinson’s disease who were treated with ropinirole or levodopa. 056 Study Group. N. Engl. J. Med.342(20), 1484–1491 (2000).
  • Molina JA, Sainz-Artiga MJ, Fraile A et al. Pathologic gambling in Parkinson’s disease: a behavioral manifestation of pharmacologic treatment? Mov. Disord.15(5), 869–872 (2000).
  • Dodd ML, Klos KJ, Bower JH, Geda YE, Josephs KA, Ahlskog JE. Pathological gambling caused by drugs used to treat Parkinson disease. Arch. Neurol.62(9), 1377–1381 (2005).
  • Biglan KM, Holloway RG Jr, McDermott MP, Richard IH. Risk factors for somnolence, edema, and hallucinations in early Parkinson disease. Neurology69(2), 187–195 (2007).
  • Kaakkola S, Gordin A, Mannisto PT. General properties and clinical possibilities of new selective inhibitors of catechol O-methyltransferase. Gen. Pharmacol.25(5), 813–824 (1994).
  • Hauser RA. Levodopa/carbidopa/entacapone (Stalevo). Neurology62(1 Suppl. 1), S64–S71 (2004).
  • Kaakkola S, Wurtman RJ. Effects of catechol-O-methyltransferase inhibitors and L-3,4-dihydroxyphenylalanine with or without carbidopa on extracellular dopamine in rat striatum. J. Neurochem.60(1), 137–144 (1993).
  • Guttman M, Leger G, Reches A et al. Administration of the new COMT inhibitor OR-611 increases striatal uptake of fluorodopa. Mov. Disord.8(3), 298–304 (1993).
  • Tornwall M, Mannisto PT. Effects of three types of catechol O-methylation inhibitors on L-3,4-dihydroxyphenylalanine-induced circling behaviour in rats. Eur. J. Pharmacol.250(1), 77–84 (1993).
  • Smith LA, Gordin A, Jenner P, Marsden CD. Entacapone enhances levodopa-induced reversal of motor disability in MPTP-treated common marmosets. Mov. Disord.12(6), 935–945 (1997).
  • Kaakkola S, Teravainen H, Ahtila S, Rita H, Gordin A. Effect of entacapone, a COMT inhibitor, on clinical disability and levodopa metabolism in parkinsonian patients. Neurology44(1), 77–80 (1994).
  • Kaakkola S, Wurtman RJ. Effects of COMT inhibitors on striatal dopamine metabolism: a microdialysis study. Brain Res.587(2), 241–249 (1992).
  • Nissinen E, Linden IB, Schultz E, Pohto P. Biochemical and pharmacological properties of a peripherally acting catechol-O-methyltransferase inhibitor entacapone. Naunyn Schmiedebergs Arch. Pharmacol.346(3), 262–266 (1992).
  • Smith LA, Jackson MJ, Al-Barghouthy G et al. Multiple small doses of levodopa plus entacapone produce continuous dopaminergic stimulation and reduce dyskinesia induction in MPTP-treated drug-naive primates. Mov. Disord.20(3), 306–314 (2005).
  • Gordin A, Brooks D. Clinical pharmacology and therapeutic use of COMT inhibition in Parkinson’s disease. J. Neurol.254(Suppl. 4), IV37–IV48 (2007).
  • Axelrod J, Tomchick R. Enzymatic O-methylation of epinephrine and other catechols. J. Biol. Chem.233, 702–705 (1958).
  • Guldberg HC, Marsden CA. Catechol-O-methyl transferase: pharmacological aspects and physiological role. Pharmacol. Rev.27(2), 135–206 (1975).
  • Park D. Monoamine methyltransferases. In: Neuromethods 5: Neurotransmitter Enzymes (3rd Edition). Boulton AA BG, Yu PH (Eds). The Humana Press, Clifton, NJ, USA (1986).
  • Boulton AA, Baker GB, Yu PH. Neurotransmitter enzymes. The Humana Press, Clifton, NJ, USA (1986).
  • Boudikova B, Szumlanski C, Maidak B, Weinshilboum R. Human liver catechol-O-methyltransferase pharmacogenetics. Clin. Pharmacol. Ther.48(4), 381–389 (1990).
  • Lee MS, Kim HS, Cho EK, Lim JH, Rinne JO. COMT genotype and effectiveness of entacapone in patients with fluctuating Parkinson’s disease. Neurology58(4), 564–567 (2002).
  • Nutt JG, Fellman JH. Pharmacokinetics of levodopa. Clin. Neuropharmacol.7(1), 35–49 (1984).
  • Cedarbaum JM. Clinical pharmacokinetics of anti-parkinsonian drugs. Clin. Pharmacokinet.13(3), 141–178 (1987).
  • Pinder RM, Brogden RN, Sawyer PR, Speight TM, Avery GS. Levodopa and decarboxylase inhibitors: a review of their clinical pharmacology and use in the treatment of parkinsonism. Drugs11(5), 329–377 (1976).
  • Gordin A, Kaakkola S, Teravainen H. Clinical advantages of COMT inhibition with entacapone – a review. J. Neural Transm.111(10–11), 1343–1363 (2004).
  • Guttman M, Leger G, Cedarbaum JM et al.3-O-methyldopa administration does not alter fluorodopa transport into the brain. Ann. Neurol.31(6), 638–643 (1992).
  • Nutt JG, Woodward WR, Gancher ST, Merrick D. 3-O-methyldopa and the response to levodopa in Parkinson’s disease. Ann. Neurol.21(6), 584–588 (1987).
  • Chang WY, Webster RA. Effects of 3-O-methyl on L-dopa-facilitated synthesis and efflux of dopamine from rat striatal slices. Br. J. Pharmacol.116(6), 2637–2640 (1995).
  • Cedarbaum JM, Leger G, Guttman M. Reduction of circulating 3-O-methyldopa by inhibition of catechol-O-methyltransferase with OR-611 and OR-462 in cynomolgus monkeys: implications for the treatment of Parkinson’s disease. Clin. Neuropharmacol.14(4), 330–342 (1991).
  • Heikkinen H, Nutt JG, LeWitt PA, Koller WC, Gordin A. The effects of different repeated doses of entacapone on the pharmacokinetics of L-dopa and on the clinical response to L-dopa in Parkinson’s disease. Clin. Neuropharmacol.24(3), 150–157 (2001).
  • Keranen T, Gordin A, Harjola VP et al. The effect of catechol-O-methyl transferase inhibition by entacapone on the pharmacokinetics and metabolism of levodopa in healthy volunteers. Clin. Neuropharmacol.16(2), 145–156 (1993).
  • Paija O, Laine K, Kultalahti ER et al. Entacapone increases levodopa exposure and reduces plasma levodopa variability when used with Sinemet CR. Clin. Neuropharmacol.28(3), 115–119 (2005).
  • Mannisto PT, Kaakkola S. Catechol-O-methyltransferase (COMT): biochemistry, molecular biology, pharmacology, and clinical efficacy of the new selective COMT inhibitors. Pharmacol. Rev.51(4), 593–628 (1999).
  • 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.46(2), 151–157 (1994).
  • 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.21(1), 81–92 (1993).
  • Heikkinen H, Saraheimo M, Antila S, Ottoila P, Pentikainen PJ. Pharmacokinetics of entacapone, a peripherally acting catechol-O-methyltransferase inhibitor, in man. A study using a stable isotope techique. Eur. J. Clin. Pharmacol.56(11), 821–826 (2001).
  • Mannisto PT, Tuomainen P, Tuominen RK. Different in vivo properties of three new inhibitors of catechol O-methyltransferase in the rat. Br. J. Pharmacol.105(3), 569–574 (1992).
  • Rouru J, Gordin A, Huupponen R et al. Pharmacokinetics of oral entacapone after frequent multiple dosing and effects on levodopa disposition. Eur. J. Clin. Pharmacol.55(6), 461–467 (1999).
  • Mannisto PT, Kaakkola S. New selective COMT inhibitors: useful adjuncts for Parkinson’s disease? Trends Pharmacol. Sci.10(2), 54–56 (1989).
  • Nutt JG, Woodward WR, Beckner RM et al. Effect of peripheral catechol-O-methyltransferase inhibition on the pharmacokinetics and pharmacodynamics of levodopa in parkinsonian patients. Neurology44(5), 913–919 (1994).
  • Heikkinen H, Varhe A, Laine T et al. Entacapone improves the availability of L-dopa in plasma by decreasing its peripheral metabolism independent of L-dopa/carbidopa dose. Br. J. Clin. Pharmacol.54(4), 363–371 (2002).
  • 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. Psychiatry68(5), 589–594 (2000).
  • Bet L, Bareggi SR, Pacei F, Bondiolotti G, Meola G, Schapira AH. Bimodal administration of entacapone in Parkinson’s disease patients improves motor control. Eur J. Neurol.15(3), 268–273 (2008).
  • Ahtila S, Kaakkola S, Gordin A et al. Effect of entacapone, a COMT inhibitor, on the pharmacokinetics and metabolism of levodopa after administration of controlled-release levodopa–carbidopa in volunteers. Clin. Neuropharmacol.18(1), 46–57 (1995).
  • Sundberg S, Scheinin M, Illi A, Akkila J, Gordin A, Keranen T. The effects of the COMT inhibitor entacapone on haemodynamics and peripheral catecholamine metabolism during exercise. Br. J. Clin. Pharmacol.36(5), 451–456 (1993).
  • Merello M, Lees AJ, Webster R, Bovingdon M, Gordin A. Effect of entacapone, a peripherally acting catechol-O-methyltransferase inhibitor, on the motor response to acute treatment with levodopa in patients with Parkinson’s disease. J. Neurol. Neurosurg. Psychiatry57(2), 186–189 (1994).
  • Myllyla VV, Sotaniemi KA, Illi A, Suominen K, Keranen T. Effect of entacapone, a COMT inhibitor, on the pharmacokinetics of levodopa and on cardiovascular responses in patients with Parkinson’s disease. Eur. J. Clin. Pharmacol.45(5), 419–423 (1993).
  • 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. Psychiatry60(1), 36–40 (1996).
  • 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.19(4), 283–296 (1996).
  • Entacapone improves motor fluctuations in levodopa-treated Parkinson’s disease patients. Parkinson Study Group. Ann. Neurol.42(5), 747–755 (1997).
  • Rinne UK, Larsen JP, Siden A, Worm-Petersen J. Entacapone enhances the response to levodopa in parkinsonian patients with motor fluctuations. Nomecomt Study Group. Neurology51(5), 1309–1314 (1998).
  • 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.105(4), 245–255 (2002).
  • Brooks DJ, Sagar H. 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. Psychiatry74(8), 1071–1079 (2003).
  • Brooks DJ, Leinonen M, Kuoppamaki M, Nissinen H. Five-year efficacy and safety of levodopa/DDCI and entacapone in patients with Parkinson’s disease. J. Neural Transm.115(6), 843–849 (2008).
  • 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. Mov. Disord.22(11), 1550–1555 (2007).
  • Jenkinson C, Fitzpatrick R, Peto V. Health-related quality-of-life measurement in patients with Parkinson’s disease. Pharmacoeconomics15(2), 157–165 (1999).
  • Jenkinson C, Fitzpatrick R. Cross-cultural evaluation of the short form 8-item Parkinson’s Disease Questionnaire (PDQ-8): results from America, Canada, Japan, Italy and Spain. Parkinsonism Relat. Disord.13(1), 22–28 (2007).
  • 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.45(2), 111–118 (2001).
  • 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.111(1), 21–28 (2005).
  • Onofrj M, Thomas A, Vingerhoets F et al. Combining entacapone with levodopa/DDCI improves clinical status and quality of life in Parkinson’s Disease (PD) patients experiencing wearing-off, regardless of the dosing frequency: results of a large multicentre open-label study. J. Neural Transm.111(8), 1053–1063 (2004).
  • 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. Psychiatry27(6), 963–971 (2003).
  • Olanow CW, Kieburtz K, Stern M et al. Double-blind, placebo-controlled study of entacapone in levodopa-treated patients with stable Parkinson disease. Arch. Neurol.61(10), 1563–1568 (2004).
  • 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.14(3), 282–289 (2007).
  • Linazasoro G, Kulisevsky J, Hernandez B. Should levodopa dose be reduced when switched to stalevo? Eur J. Neurol.15(3), 257–261 (2008).
  • Fung VS, Herawati L, Wan Y. Quality of life in early Parkinson’s disease treated with levodopa/carbidopa/entacapone. Mov. Disord.24(1), 25–31 (2009).
  • Koller W, Guarnieri M, Hubble J, Rabinowicz AL, Silver D. An open-label evaluation of the tolerability and safety of Stalevo (carbidopa, levodopa and entacapone) in Parkinson’s disease patients experiencing wearing-off. J. Neural Transm.112(2), 221–230 (2005).
  • Brooks DJ, Agid Y, Eggert K, Widner H, Ostergaard K, Holopainen A. Treatment of end-of-dose wearing-off in parkinson’s disease: stalevo (levodopa/carbidopa/entacapone) and levodopa/DDCI given in combination with Comtess/Comtan (entacapone) provide equivalent improvements in symptom control superior to that of traditional levodopa/DDCI treatment. Eur. Neurol.53(4), 197–202 (2005).
  • Myllyla V, Haapaniemi T, Kaakkola S et al. Patient satisfaction with switching to Stalevo: an open-label evaluation in PD patients experiencing wearing-off (Simcom Study). Acta Neurol. Scand.114(3), 181–186 (2006).
  • Zubair M, Jackson MJ, Tayarani-Binazir K et al. The administration of entacapone prevents L-dopa-induced dyskinesia when added to dopamine agonist therapy in MPTP-treated primates. Exp. Neurol.208(2), 177–184 (2007).
  • Hauser R. First Step: Evaluation of the Efficacy of Stalevo in de novo Parkinson’s disease. Eur J. Neurol.13(Suppl. 2), 94 (2006).
  • Hauser R, 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. Mov. Disord.24(4), 541–550 (2009).
  • 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.10(2), 137–146 (2003).
  • Myllyla VV, Kultalahti ER, Haapaniemi H, Leinonen M. Twelve-month safety of entacapone in patients with Parkinson’s disease. Eur. J. Neurol.8(1), 53–60 (2001).
  • Palmer CS, Nuijten MJ, Schmier JK, Subedi P, Snyder EH. Cost effectiveness of treatment of Parkinson’s disease with entacapone in the United States. Pharmacoeconomics20(9), 617–628 (2002).
  • Nuijten MJ, van Iperen P, Palmer C, van Hilten BJ, Snyder E. Cost-effectiveness analysis of entacapone in Parkinson’s disease: a Markov process analysis. Value Health4(4), 316–328 (2001).
  • Korchounov A, Bogomazov G. Employment, medical absenteeism, and disability perception in Parkinson’s disease: a pilot double-blind, randomized, placebo-controlled study of entacapone adjunctive therapy. Mov. Disord.21(12), 2220–2224 (2006).
  • Kushnir M, Yaar A, Reichman A, Heldman E. Transdermal delivery of a levodopa prodrug: a pilot clinical trial. Program of the 12th International Congress of Parkinson’s Disease and Movement Disorders. Chicago, IL, USA, 22–26 June 2008 (Poster 592).
  • Nyholm D. Enteral levodopa/carbidopa gel infusion for the treatment of motor fluctuations and dyskinesias in advanced Parkinson’s disease. Expert Rev. Neurother.6, 1403–1411 (2006).
  • Goole J, Deleuze P, Vanderbist F et al. New levodopa sustained-release floating minitablets coated with insoluble acrylic polymer. Eur. J. Biopharm.68, 310–318 (2008).
  • Di Stefano A, Sozio P, Cerasa L. Antiparkinson prodrugs. Molecules13, 46–68 (2008).

Website

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.