Skip to main content
Top
Published in: Drugs 6/2000

01-06-2000 | Review Article

Clinical Pharmacology, Therapeutic Use and Potential of COMT Inhibitors in Parkinson’s Disease

Author: Dr Seppo Kaakkola

Published in: Drugs | Issue 6/2000

Login to get access

Abstract

When peripheral decarboxylation is blocked by carbidopa or benserazide, the main metabolic pathway of levodopa is O-methylation by catechol-O-methyltransferase (COMT). Entacapone and tolcapone are new potent, selective and reversible nitrocatechol-type COMT inhibitors. Animal studies have demonstrated that entacapone mainly has a peripheral effect whereas tolcapone also inhibits O-methylation in the brain. In human volunteers, both entacapone and tolcapone dose-dependently inhibit the COMT activity in erythrocytes, improve the bioavailability and decrease the elimination of levodopa, and inhibit the formation of 3-O-methyldopa (3-OMD). Entacapone is administered with every scheduled dose of levodopa whereas tolcapone is administered 3 times daily. The different administration regimens for these agents are based on their different pharmacokinetic and pharmacodynamic profiles.
Both entacapone and tolcapone enhance and extend the therapeutic effect of levodopa in patients with advanced and fluctuating Parkinson’s disease. They prolong the duration of levodopa effect. Clinical studies show that they increase the daily ON time by an average 1 to 3 hours, improve the activities of daily living and allow daily levodopa dosage to be decreased. Correspondingly, they significantly reduce the daily OFF time. No comparative studies between entacapone and tolcapone have been performed. Tolcapone also appears to have a beneficial effect in patients with nonfluctuating Parkinson’s disease.
The main adverse effects of the COMT inhibitors are related to their dopaminergic and gastrointestinal effects. Enhancement of dopaminergic activity may cause an initial worsening of levodopa-induced adverse effects, such as dyskinesia, nausea, vomiting, orthostatic hypotension, sleep disorders and hallucinations. Levodopa dose adjustment is recommended to avoid these events. Tolcapone is associated with diarrhoea in about 16 to 18% of patients and entacapone in less than 10% of patients. Diarrhoea has led to discontinuation in 5 to 6% of patients treated with tolcapone and in 2.5% of those treated with entacapone. Urine discoloration to dark yellow or orange is related to the colour of COMT inhibitors and their metabolites. Elevated liver transaminase levels are reported in 1 to 3% of patients treated with tolcapone but very rarely, if at all, in patients treated with entacapone. The descriptions of acute, fatal fulminant hepatitis and potentially fatal neurological reactions, such as neuroleptic malignant syndrome and rhabdomyolysis, in association with tolcapone led to the suspension of its marketing authorisation in the European Community and Canada. In many other countries, the use of tolcapone is restricted to patients who are not responding satisfactorily to other therapies. Regular monitoring of liver enzymes is required if tolcapone is used. No such adverse reactions have so far been described for entacapone and no laboratory monitoring has been proposed.
COMT inhibitors added to levodopa therapy are beneficial, particularly in patients with fluctuating disease. They may be combined with other antiparkinsonian drugs, such as dopamine agonists, selegiline and anticholinergics without adverse interactions. They provide a new treatment possibility in patients with Parkinson’s disease who have problems with their present levodopa therapy.
Literature
1.
go back to reference Axelrod J. O-methylation of epinephrine and other catechols in vitro and in vivo. Science 1957; 126: 400–1PubMedCrossRef Axelrod J. O-methylation of epinephrine and other catechols in vitro and in vivo. Science 1957; 126: 400–1PubMedCrossRef
2.
go back to reference Guldberg HC, Marsden CA. Catechol-O-methyl transferase: pharmacological aspects and physiological role. Pharmacol Rev 1975; 27: 135–206PubMed Guldberg HC, Marsden CA. Catechol-O-methyl transferase: pharmacological aspects and physiological role. Pharmacol Rev 1975; 27: 135–206PubMed
3.
go back to reference Ericsson AD. Potentiation of the L-dopa effect in man by the use of catechol-O-methyltransferase inhibitors. J Neurol Sci 1971; 14: 193–7PubMedCrossRef Ericsson AD. Potentiation of the L-dopa effect in man by the use of catechol-O-methyltransferase inhibitors. J Neurol Sci 1971; 14: 193–7PubMedCrossRef
4.
go back to reference Reches A, Fahn S. Catechol-O-methyltransferase and Parkinson’s disease. Adv Neurol 1984; 40: 171–9PubMed Reches A, Fahn S. Catechol-O-methyltransferase and Parkinson’s disease. Adv Neurol 1984; 40: 171–9PubMed
5.
go back to reference Männistö PT, Kaakkola S. Rationale for selective COMT inhibitors as adjuncts in the drug treatment of Parkinson’s disease. Pharmacol Toxicol 1990; 66: 317–23PubMedCrossRef Männistö PT, Kaakkola S. Rationale for selective COMT inhibitors as adjuncts in the drug treatment of Parkinson’s disease. Pharmacol Toxicol 1990; 66: 317–23PubMedCrossRef
6.
go back to reference Männistö PT, Ulmanen I, Taskinen J, et al. Catechol-O-methyltransferase (COMT) and COMT inhibitors. In: Sandier M, Smith J, editors. Design of enzyme inhibitors as drugs. Oxford: Oxford University Press, 1993; 623–46 Männistö PT, Ulmanen I, Taskinen J, et al. Catechol-O-methyltransferase (COMT) and COMT inhibitors. In: Sandier M, Smith J, editors. Design of enzyme inhibitors as drugs. Oxford: Oxford University Press, 1993; 623–46
7.
go back to reference Kaakkola S, Gordin A, Männistö PT. General properties and clinical possibilities of new selective inhibitors of catechol O-methyltransferase. Gen Pharmacol 1994; 25: 813–24PubMedCrossRef Kaakkola S, Gordin A, Männistö PT. General properties and clinical possibilities of new selective inhibitors of catechol O-methyltransferase. Gen Pharmacol 1994; 25: 813–24PubMedCrossRef
8.
go back to reference Pentikäinen PJ, Vuorela A, Järvinen M, et al. Human pharmacokinetics of OR-462, a new catechol-O-methyltransferase inhibitor. Eur J Clin Pharmacol 1989; 36 Suppl.: Al 10 Pentikäinen PJ, Vuorela A, Järvinen M, et al. Human pharmacokinetics of OR-462, a new catechol-O-methyltransferase inhibitor. Eur J Clin Pharmacol 1989; 36 Suppl.: Al 10
9.
go back to reference Kaakkola S, Gordin A, Järvinen M, et al. Effect of a novel catechol-O-methyltransferase inhibitor, nitecapone, on the metabolism of L-dopa in healthy volunteers. Clin Neuropharmacol 1990; 13: 436–47PubMedCrossRef Kaakkola S, Gordin A, Järvinen M, et al. Effect of a novel catechol-O-methyltransferase inhibitor, nitecapone, on the metabolism of L-dopa in healthy volunteers. Clin Neuropharmacol 1990; 13: 436–47PubMedCrossRef
10.
go back to reference Bieck PR, Nilsson E, Antonin KH. Effect of the new selective COMT inhibitor CGP 28014 A on the formation of 3-O-methyldopa (3OMD) in plasma of healthy subjects. J Neural Transm Suppl 1990; 32: 387–91PubMed Bieck PR, Nilsson E, Antonin KH. Effect of the new selective COMT inhibitor CGP 28014 A on the formation of 3-O-methyldopa (3OMD) in plasma of healthy subjects. J Neural Transm Suppl 1990; 32: 387–91PubMed
11.
go back to reference Bieck PR, Antonin KH, Farger G, et al. Clinical pharmacology of the new COMT inhibitor CGP 28, 014. Neurochem Res 1993; 18: 1163–7PubMedCrossRef Bieck PR, Antonin KH, Farger G, et al. Clinical pharmacology of the new COMT inhibitor CGP 28, 014. Neurochem Res 1993; 18: 1163–7PubMedCrossRef
12.
go back to reference Feuerstein C, Tanche M, Serre F, et al. Does O-methyl-dopa play a role in levodopa-induced dyskinesias? Acta Neurol Scand 1977; 56: 79–82PubMedCrossRef Feuerstein C, Tanche M, Serre F, et al. Does O-methyl-dopa play a role in levodopa-induced dyskinesias? Acta Neurol Scand 1977; 56: 79–82PubMedCrossRef
13.
go back to reference Rivera-Calimlim L, Tandon D, Anderson F, et al. The clinical picture and plasma levodopa metabolite profile of parkinsonian nonresponders. Treatment with levodopa and decarboxylase inhibitor. Arch Neurol 1977; 34: 228–32 Rivera-Calimlim L, Tandon D, Anderson F, et al. The clinical picture and plasma levodopa metabolite profile of parkinsonian nonresponders. Treatment with levodopa and decarboxylase inhibitor. Arch Neurol 1977; 34: 228–32
14.
go back to reference Tohgi H, Abe T, Kikuchi T, et al. The significance of 3-O-methyldopa concentrations in the cerebrospinal fluid in the pathogenesis of wearing-off phenomenon in Parkinson’s disease. Neurosci Lett 1991; 132: 19–22PubMedCrossRef Tohgi H, Abe T, Kikuchi T, et al. The significance of 3-O-methyldopa concentrations in the cerebrospinal fluid in the pathogenesis of wearing-off phenomenon in Parkinson’s disease. Neurosci Lett 1991; 132: 19–22PubMedCrossRef
15.
go back to reference Wade LA, Katzman R. 3-O-Methyldopa uptake and inhibition of L-dopa at the blood-brain barrier. Life Sci 1975; 17: 131–6PubMedCrossRef Wade LA, Katzman R. 3-O-Methyldopa uptake and inhibition of L-dopa at the blood-brain barrier. Life Sci 1975; 17: 131–6PubMedCrossRef
16.
go back to reference McKenzie GM, White HL. Evidence for the methylation of apomorphine by catechol-O-methyl-transferase in vivo and in vitro. Biochem Pharmacol 1973; 22: 2329–36PubMedCrossRef McKenzie GM, White HL. Evidence for the methylation of apomorphine by catechol-O-methyl-transferase in vivo and in vitro. Biochem Pharmacol 1973; 22: 2329–36PubMedCrossRef
17.
go back to reference Symes AL, Lal S, Sourkes TL. Effect of catechol-O-methyltransferase inhibitors on brain apomorphine concentrations and stereotyped behaviour in the rat. J Pharm Pharmacol 1975; 27: 947–9PubMedCrossRef Symes AL, Lal S, Sourkes TL. Effect of catechol-O-methyltransferase inhibitors on brain apomorphine concentrations and stereotyped behaviour in the rat. J Pharm Pharmacol 1975; 27: 947–9PubMedCrossRef
18.
go back to reference Coudore F, Durif F, Duroux E, et al. Effect of tolcapone on plasma and striatal apomorphine disposition in rats. Neuroreport 1997; 8: 877–80PubMedCrossRef Coudore F, Durif F, Duroux E, et al. Effect of tolcapone on plasma and striatal apomorphine disposition in rats. Neuroreport 1997; 8: 877–80PubMedCrossRef
19.
go back to reference Kohli JD, Horn PT, Glock D, et al. Dihydrexidine: a new potent peripheral dopamine D1 receptor agonist. Eur J Pharmacol 1993; 235: 31–5PubMedCrossRef Kohli JD, Horn PT, Glock D, et al. Dihydrexidine: a new potent peripheral dopamine D1 receptor agonist. Eur J Pharmacol 1993; 235: 31–5PubMedCrossRef
20.
go back to reference Keränen 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: 151–7PubMedCrossRef Keränen 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: 151–7PubMedCrossRef
21.
go back to reference 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: 508–17PubMedCrossRef 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: 508–17PubMedCrossRef
22.
go back to reference Heikkinen H, Pentikäinen PJ, Saraheimo M, et al. Pharmacokinetics of entacapone, a new COMT-inhibitor, in man: a study using stable isotope technique. New Trends Clin Neuropharm 1994; 8: 301 Heikkinen H, Pentikäinen PJ, Saraheimo M, et al. Pharmacokinetics of entacapone, a new COMT-inhibitor, in man: a study using stable isotope technique. New Trends Clin Neuropharm 1994; 8: 301
23.
go back to reference Jorga KM, Fotteler B, Heizmann P, et al. Pharmacokinetics and pharmacodynamics after oral and intravenous administration of tolcapone, a novel adjunct to Parkinson’s disease therapy. Eur J Clin Pharmacol 1998; 54: 443–7PubMedCrossRef Jorga KM, Fotteler B, Heizmann P, et al. Pharmacokinetics and pharmacodynamics after oral and intravenous administration of tolcapone, a novel adjunct to Parkinson’s disease therapy. Eur J Clin Pharmacol 1998; 54: 443–7PubMedCrossRef
24.
go back to reference Wikberg T, Vuorela A, Ottoila P, et al. Identification of major metabolites of the catechol-O-methyltransferase inhibitor entacapone in rats and humans. Drug Metab Dispos 1993; 21: 81–92PubMed Wikberg T, Vuorela A, Ottoila P, et al. Identification of major metabolites of the catechol-O-methyltransferase inhibitor entacapone in rats and humans. Drug Metab Dispos 1993; 21: 81–92PubMed
25.
go back to reference F. Hoffman-La Roche Ltd. Product monograph Tasmar. Basel: F. Hoffman-La Roche Ltd, 1997: 1–60 F. Hoffman-La Roche Ltd. Product monograph Tasmar. Basel: F. Hoffman-La Roche Ltd, 1997: 1–60
26.
go back to reference Da Prada M, Borgulya J, Napolitano A, et al. Improved therapy of Parkinson’s disease with tolcapone, a central and peripheral COMT inhibitor with an S-adenosyl-L-methionine-sparing effect. Clin Neuropharmacol 1995; 17: S26–S37CrossRef Da Prada M, Borgulya J, Napolitano A, et al. Improved therapy of Parkinson’s disease with tolcapone, a central and peripheral COMT inhibitor with an S-adenosyl-L-methionine-sparing effect. Clin Neuropharmacol 1995; 17: S26–S37CrossRef
27.
go back to reference Dingemanse J, Jorga K, Zürcher G, et al. Multiple-dose clinical pharmacology of the catechol-O-methyl-transferase inhibitor tolcapone in elderly subjects. Eur J Clin Pharmacol 1996; 50: 47–55PubMedCrossRef Dingemanse J, Jorga K, Zürcher G, et al. Multiple-dose clinical pharmacology of the catechol-O-methyl-transferase inhibitor tolcapone in elderly subjects. Eur J Clin Pharmacol 1996; 50: 47–55PubMedCrossRef
28.
go back to reference Jorga K, Fotteler B, Wiegand U. Tolcapone does not change the pharmacokinetics and pharmacodynamics of the CYP2C9 substrate tolbutamide. Mov Disord 1997; 12 Suppl. 1: 100CrossRef Jorga K, Fotteler B, Wiegand U. Tolcapone does not change the pharmacokinetics and pharmacodynamics of the CYP2C9 substrate tolbutamide. Mov Disord 1997; 12 Suppl. 1: 100CrossRef
29.
go back to reference Gordin A, Huupponen R, Rouru J, et al. Pharmacokinetics of entacapone and catechol-O-methyltransferase (COMT) inhibition after frequent multiple dosing of entacapone and effect on levodopa metabolism. Eur J Neurol 1998; 5 Suppl. 3: S165–S6 Gordin A, Huupponen R, Rouru J, et al. Pharmacokinetics of entacapone and catechol-O-methyltransferase (COMT) inhibition after frequent multiple dosing of entacapone and effect on levodopa metabolism. Eur J Neurol 1998; 5 Suppl. 3: S165–S6
30.
go back to reference Jorga KM, Sedek G, Fotteler B, et al. Optimizing levodopa pharmacokinetics with multiple tolcapone doses in the elderly. Clin Pharmacol Ther 1997; 62: 300–10PubMedCrossRef Jorga KM, Sedek G, Fotteler B, et al. Optimizing levodopa pharmacokinetics with multiple tolcapone doses in the elderly. Clin Pharmacol Ther 1997; 62: 300–10PubMedCrossRef
31.
go back to reference Dingemanse J, Jorga K, Zürcher G, et al. Pharmacokinetic-pharmacodynamic interaction between the COMT inhibitor tolcapone and single-dose levodopa. Br J Clin Pharmacol 1995; 40: 253–62PubMedCrossRef Dingemanse J, Jorga K, Zürcher G, et al. Pharmacokinetic-pharmacodynamic interaction between the COMT inhibitor tolcapone and single-dose levodopa. Br J Clin Pharmacol 1995; 40: 253–62PubMedCrossRef
32.
go back to reference Jorga K, Fotteler B, van Brummelen P. Why should tolcapone be given at a lower dose to patients with liver cirrhosis? Clin Pharmacol Ther 1997; 61: 183 Jorga K, Fotteler B, van Brummelen P. Why should tolcapone be given at a lower dose to patients with liver cirrhosis? Clin Pharmacol Ther 1997; 61: 183
33.
go back to reference Gordin A, Pentikäinen PP, Mäkimartti M, et al. Pharmacokinetics of the COMT inhibitor entacapone in liver failure and the effect of entacapone on liver function. Neurology 1998; 50 Suppl. 4: A387 Gordin A, Pentikäinen PP, Mäkimartti M, et al. Pharmacokinetics of the COMT inhibitor entacapone in liver failure and the effect of entacapone on liver function. Neurology 1998; 50 Suppl. 4: A387
34.
go back to reference Comtess Summary of Product Characteristics. Espoo, Finland: Orion Corp., 1998 Comtess Summary of Product Characteristics. Espoo, Finland: Orion Corp., 1998
35.
go back to reference Schultz E, Nissinen E. Inhibition of rat liver and duodenum soluble catechol-O-methyltransferase by a tight-binding inhibitor OR-462. Biochem Pharmacol 1989; 38: 3953–6PubMedCrossRef Schultz E, Nissinen E. Inhibition of rat liver and duodenum soluble catechol-O-methyltransferase by a tight-binding inhibitor OR-462. Biochem Pharmacol 1989; 38: 3953–6PubMedCrossRef
36.
go back to reference Lotta T, Vidgren J, Tilgmann C, et al. Kinetics of human soluble and membrane-bound catechol O-methyltransferase: a revised mechanism and description of the thermolabile variant of the enzyme. Biochemistry 1995; 34: 4202–10PubMedCrossRef Lotta T, Vidgren J, Tilgmann C, et al. Kinetics of human soluble and membrane-bound catechol O-methyltransferase: a revised mechanism and description of the thermolabile variant of the enzyme. Biochemistry 1995; 34: 4202–10PubMedCrossRef
37.
go back to reference Borges N, Vieira-Coelho MA, Parada A, et al. Studies on the tight-binding nature of tolcapone inhibition of soluble and membrane-bound rat brain catechol-O-methyltransferase. J Pharmacol Exp Ther 1997; 282: 812–7PubMed Borges N, Vieira-Coelho MA, Parada A, et al. Studies on the tight-binding nature of tolcapone inhibition of soluble and membrane-bound rat brain catechol-O-methyltransferase. J Pharmacol Exp Ther 1997; 282: 812–7PubMed
38.
go back to reference Keränen 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 1993; 16: 145–56PubMedCrossRef Keränen 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 1993; 16: 145–56PubMedCrossRef
39.
go back to reference Sêdek G, Jorga K, Schmitt M, et al. Effect of tolcapone on plasma levodopa concentrations after coadministration with levodopa/carbidopa to healthy volunteers. Clin Neuropharmacol 1997; 20: 531–41PubMedCrossRef Sêdek G, Jorga K, Schmitt M, et al. Effect of tolcapone on plasma levodopa concentrations after coadministration with levodopa/carbidopa to healthy volunteers. Clin Neuropharmacol 1997; 20: 531–41PubMedCrossRef
40.
go back to reference Jorga K, Fotteler B, Schmitt M, et al. The effect of COMT inhibition by tolcapone on tolerability and pharmacokinetics of different levodopa/benserazide formulations. Eur Neurol 1997; 38: 59–67PubMedCrossRef Jorga K, Fotteler B, Schmitt M, et al. The effect of COMT inhibition by tolcapone on tolerability and pharmacokinetics of different levodopa/benserazide formulations. Eur Neurol 1997; 38: 59–67PubMedCrossRef
41.
go back to reference Myllylä VV, Sotaniemi KA, Mäkimartti M, et al. Effect of entacapone as an adjunct to Sinemet and Madopar on the pharmacokinetics of levodopa in parkinsonian patients. Mov Disord 1997; 12 Suppl. 1: 103CrossRef Myllylä VV, Sotaniemi KA, Mäkimartti M, et al. Effect of entacapone as an adjunct to Sinemet and Madopar on the pharmacokinetics of levodopa in parkinsonian patients. Mov Disord 1997; 12 Suppl. 1: 103CrossRef
42.
go back to reference Jorga K, Fotteler B, Sedek G, et al. The effect of tolcapone on levodopa pharmacokinetics is independent of levodopa/carbidopa formulation. J Neurol 1998; 245: 223–30PubMedCrossRef Jorga K, Fotteler B, Sedek G, et al. The effect of tolcapone on levodopa pharmacokinetics is independent of levodopa/carbidopa formulation. J Neurol 1998; 245: 223–30PubMedCrossRef
43.
go back to reference 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 levodopacarbidopa in volunteers. Clin Neuropharmacol 1995; 18: 46–57PubMedCrossRef 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 levodopacarbidopa in volunteers. Clin Neuropharmacol 1995; 18: 46–57PubMedCrossRef
44.
go back to reference 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: 283–96PubMedCrossRef 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: 283–96PubMedCrossRef
45.
go back to reference 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: 222–33PubMedCrossRef 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: 222–33PubMedCrossRef
46.
go back to reference Myllylä VV, Sotaniemi KA, Illi A, et al. 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 1993; 45: 419–23PubMedCrossRef Myllylä VV, Sotaniemi KA, Illi A, et al. 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 1993; 45: 419–23PubMedCrossRef
47.
go back to reference 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. Neurology 1994; 44: 913–9PubMedCrossRef 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. Neurology 1994; 44: 913–9PubMedCrossRef
48.
go back to reference Tohgi H, Abe T, Yamazaki K, et al. Effects of the catechol-O-methyltransferase inhibitor tolcapone in Parkinson’s disease: correlations between concentrations of dopaminergic substances in the plasma and cerebrospinal fluid and clinical improvement. Neurosci Lett 1995; 192: 165–8PubMedCrossRef Tohgi H, Abe T, Yamazaki K, et al. Effects of the catechol-O-methyltransferase inhibitor tolcapone in Parkinson’s disease: correlations between concentrations of dopaminergic substances in the plasma and cerebrospinal fluid and clinical improvement. Neurosci Lett 1995; 192: 165–8PubMedCrossRef
49.
go back to reference Roberts JW, Cora-Locatelli G, Bravi D, et al. Catechol-O-methyltransferase inhibitor tolcapone prolongs levodopa/carbidopa action in parkinsonian patients. Neurology 1993; 43: 2685–8PubMedCrossRef Roberts JW, Cora-Locatelli G, Bravi D, et al. Catechol-O-methyltransferase inhibitor tolcapone prolongs levodopa/carbidopa action in parkinsonian patients. Neurology 1993; 43: 2685–8PubMedCrossRef
50.
go back to reference Limousin P, Pollak P, Pfefen JP, et al. Acute administration of levodopa-benserazide and tolcapone, a COMT inhibitor, Parkinson’s disease. Clin Neuropharmacol 1995; 18: 258–65PubMedCrossRef Limousin P, Pollak P, Pfefen JP, et al. Acute administration of levodopa-benserazide and tolcapone, a COMT inhibitor, Parkinson’s disease. Clin Neuropharmacol 1995; 18: 258–65PubMedCrossRef
51.
go back to reference Yamamoto M, Yokochi M, Kuno S, et al. Effects of tolcapone, a catechol-O-methyltransferase inhibitor, on motor symptoms and pharmacokinetics of levodopa in patients with Parkinson’s disease. J Neural Transm 1997; 104: 229–36PubMedCrossRef Yamamoto M, Yokochi M, Kuno S, et al. Effects of tolcapone, a catechol-O-methyltransferase inhibitor, on motor symptoms and pharmacokinetics of levodopa in patients with Parkinson’s disease. J Neural Transm 1997; 104: 229–36PubMedCrossRef
52.
go back to reference Kaakkola S, Teräväinen H, Ahtila S, et al. Entacapone in combination with standard or controlled-release levodopa/carbidopa: a clinical and pharmacokinetic study in patients with Parkinson’s disease. Eur J Neurol 1995; 2: 341–7CrossRef Kaakkola S, Teräväinen H, Ahtila S, et al. Entacapone in combination with standard or controlled-release levodopa/carbidopa: a clinical and pharmacokinetic study in patients with Parkinson’s disease. Eur J Neurol 1995; 2: 341–7CrossRef
53.
go back to reference Kuruma I, Bartholini G, Tissot R, et al. The metabolism of L-3-O-methyldopa, a precursor of dopa in man. Clin Pharmacol Ther 1971; 12: 678–82PubMed Kuruma I, Bartholini G, Tissot R, et al. The metabolism of L-3-O-methyldopa, a precursor of dopa in man. Clin Pharmacol Ther 1971; 12: 678–82PubMed
54.
go back to reference 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: 36–40PubMedCrossRef 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: 36–40PubMedCrossRef
55.
go back to reference Sundberg S, Scheinin M, Illi A, et al. The effects of the COMT inhibitor entacapone on haemodynamics and peripheral catecholamine metabolism during exercise. Br J Clin Pharmacol 1993; 36: 451–6PubMedCrossRef Sundberg S, Scheinin M, Illi A, et al. The effects of the COMT inhibitor entacapone on haemodynamics and peripheral catecholamine metabolism during exercise. Br J Clin Pharmacol 1993; 36: 451–6PubMedCrossRef
56.
go back to reference Illi A, Sundberg S, Koulu M, et al. COMT inhibition by highdose entacapone does not affect hemodynamics but changes catecholamine metabolism in healthy volunteers at rest and during exercise. Int J Clin Pharmacol Ther 1994; 32: 582–8PubMed Illi A, Sundberg S, Koulu M, et al. COMT inhibition by highdose entacapone does not affect hemodynamics but changes catecholamine metabolism in healthy volunteers at rest and during exercise. Int J Clin Pharmacol Ther 1994; 32: 582–8PubMed
57.
go back to reference Zürcher G, Dingemanse J, Da Prada M. Potent COMT inhibition by Ro 40-7592 in the periphery and in the brain. Preclinical and clinical findings. Adv Neurol 1993; 60: 641–7 Zürcher G, Dingemanse J, Da Prada M. Potent COMT inhibition by Ro 40-7592 in the periphery and in the brain. Preclinical and clinical findings. Adv Neurol 1993; 60: 641–7
58.
go back to reference Lyytinen J, Kaakkola S, Ahtila S, et al. Simultaneous MAO-B and COMT inhibition in L-dopa-treated patients with Parkinson’s disease. Mov Disord 1997; 12: 497–505PubMedCrossRef Lyytinen J, Kaakkola S, Ahtila S, et al. Simultaneous MAO-B and COMT inhibition in L-dopa-treated patients with Parkinson’s disease. Mov Disord 1997; 12: 497–505PubMedCrossRef
59.
go back to reference Oechsner M, Stürenburg HJ, Buhmann C, et al. Elevated serum levels of dihydroxyphenylacetic acid (DOPAC) and dopamine after catechol-O-methyltransferase (COMT) inhibition. Eur J Neurol 1998; 5 Suppl. 3: S169 Oechsner M, Stürenburg HJ, Buhmann C, et al. Elevated serum levels of dihydroxyphenylacetic acid (DOPAC) and dopamine after catechol-O-methyltransferase (COMT) inhibition. Eur J Neurol 1998; 5 Suppl. 3: S169
60.
go back to reference Davis TL, Roznoski M, Burns RS. Acute effects of COMT inhibition on L-DOPA pharmacokinetics in patients treated with carbidopa and selegiline. Clin Neuropharmacol 1995; 18: 333–7PubMedCrossRef Davis TL, Roznoski M, Burns RS. Acute effects of COMT inhibition on L-DOPA pharmacokinetics in patients treated with carbidopa and selegiline. Clin Neuropharmacol 1995; 18: 333–7PubMedCrossRef
61.
go back to reference Tasmar Summary of Product Characteristics. Basle, Switzerland: F. Hoffman-La Roche Ltd., 1998 Tasmar Summary of Product Characteristics. Basle, Switzerland: F. Hoffman-La Roche Ltd., 1998
62.
go back to reference Firnau G, Sood S, Chirakal R, et al. Metabolites of 6-[18F]fluoro-L-dopa in human blood. J Nucl Med 1988; 29: 363–9PubMed Firnau G, Sood S, Chirakal R, et al. Metabolites of 6-[18F]fluoro-L-dopa in human blood. J Nucl Med 1988; 29: 363–9PubMed
63.
go back to reference Firnau G, Sood S, Chirakal R, et al. Cerebral metabolism of 6-[18F]fluoro-L-3,4-dihydroxyphenylalanine in the primate. J Neurochem 1987; 48: 1077–82PubMedCrossRef Firnau G, Sood S, Chirakal R, et al. Cerebral metabolism of 6-[18F]fluoro-L-3,4-dihydroxyphenylalanine in the primate. J Neurochem 1987; 48: 1077–82PubMedCrossRef
64.
go back to reference Guttman M, Leger G, Cedarbaum JM. OR-611 inhibits 3-Omethyldopa formation in primates. Neurology 1991; 41: 213 Guttman M, Leger G, Cedarbaum JM. OR-611 inhibits 3-Omethyldopa formation in primates. Neurology 1991; 41: 213
65.
go back to reference Günther I, Psylla M, Reddy GN, et al. Positron emission tomography in drug evaluation: influence of three different catechol-O-methyltransferase inhibitors on metabolism of [NCA] 6-[18F]fluoro-L-dopa in rhesus monkey. Nucl Med Biol 1995; 22: 921–7PubMedCrossRef Günther I, Psylla M, Reddy GN, et al. Positron emission tomography in drug evaluation: influence of three different catechol-O-methyltransferase inhibitors on metabolism of [NCA] 6-[18F]fluoro-L-dopa in rhesus monkey. Nucl Med Biol 1995; 22: 921–7PubMedCrossRef
66.
go back to reference Doudet DJ, Chan GL, Holden JE, et al. Effects of catechol-O-methyltransferase inhibition on the rates of uptake and reversibility of 6-fluoro-L-dopa trapping in MPTP-induced parkinsonism in monkeys. Neuropharmacology 1997; 36: 363–71PubMedCrossRef Doudet DJ, Chan GL, Holden JE, et al. Effects of catechol-O-methyltransferase inhibition on the rates of uptake and reversibility of 6-fluoro-L-dopa trapping in MPTP-induced parkinsonism in monkeys. Neuropharmacology 1997; 36: 363–71PubMedCrossRef
67.
go back to reference Psylla M, Günther I, Antonini A, et al. Cerebral 6-[18F]fluoroL-DOPA uptake in rhesus monkey: pharmacological influence of aromatic amino acid decarboxylase (AAAD) and catechol-O-methyltransferase (COMT) inhibition. Brain Res 1997; 767: 45–54PubMedCrossRef Psylla M, Günther I, Antonini A, et al. Cerebral 6-[18F]fluoroL-DOPA uptake in rhesus monkey: pharmacological influence of aromatic amino acid decarboxylase (AAAD) and catechol-O-methyltransferase (COMT) inhibition. Brain Res 1997; 767: 45–54PubMedCrossRef
68.
go back to reference Holden JE, Doudet D, Endres CJ, et al. Graphical analysis of 6-fluoro-L-dopa trapping: effect of inhibition of catechol-O-methyltransferase. J Nucl Med 1997; 38: 1568–74PubMed Holden JE, Doudet D, Endres CJ, et al. Graphical analysis of 6-fluoro-L-dopa trapping: effect of inhibition of catechol-O-methyltransferase. J Nucl Med 1997; 38: 1568–74PubMed
69.
go back to reference Laihinen A, Rinne JO, Rinne UK, et al. [18F]-6-fluorodopa PET scanning in Parkinson’s disease after selective COMT inhibition with nitecapone (OR-462). Neurology 1992; 42: 199–203PubMedCrossRef Laihinen A, Rinne JO, Rinne UK, et al. [18F]-6-fluorodopa PET scanning in Parkinson’s disease after selective COMT inhibition with nitecapone (OR-462). Neurology 1992; 42: 199–203PubMedCrossRef
70.
go back to reference Sawle GV, Burn DJ, Morrish PK, et al. The effect of entacapone (OR-611) on brain [18F]-6-L-fluorodopa metabolism: implications for levodopa therapy of Parkinson’s disease. Neurology 1994; 44: 1292–7PubMedCrossRef Sawle GV, Burn DJ, Morrish PK, et al. The effect of entacapone (OR-611) on brain [18F]-6-L-fluorodopa metabolism: implications for levodopa therapy of Parkinson’s disease. Neurology 1994; 44: 1292–7PubMedCrossRef
71.
go back to reference Ishikawa T, Dhawan V, Chaly T, et al. Fluorodopa positron emission tomography with an inhibitor of catechol-O-methyltransferase: effect of the plasma 3-O-methyldopa fraction on data analysis. J Cerebral Blood Flow Metab 1996; 16: 854–63CrossRef Ishikawa T, Dhawan V, Chaly T, et al. Fluorodopa positron emission tomography with an inhibitor of catechol-O-methyltransferase: effect of the plasma 3-O-methyldopa fraction on data analysis. J Cerebral Blood Flow Metab 1996; 16: 854–63CrossRef
72.
go back to reference Ruottinen H, Rinne J, Ruotsalainen U, et al. Striatal [18F]fluorodopa utilization after COMT inhibition with entacapone studied with PET in advanced Parkinson’s disease. J Neural Transm Park Dis Dem Sect 1995; 10: 91–106CrossRef Ruottinen H, Rinne J, Ruotsalainen U, et al. Striatal [18F]fluorodopa utilization after COMT inhibition with entacapone studied with PET in advanced Parkinson’s disease. J Neural Transm Park Dis Dem Sect 1995; 10: 91–106CrossRef
73.
go back to reference Ruottinen HM, Rinne JO, Oikonen VJ, et al. Striatal 6-[18F] fluorodopa accumulation after combined inhibition of peripheral catechol-O-methyltransferase and monoamine oxidase type B: differing response in relation to presynaptic dopaminergic dysfunction. Synapse 1997; 27: 336–46PubMedCrossRef Ruottinen HM, Rinne JO, Oikonen VJ, et al. Striatal 6-[18F] fluorodopa accumulation after combined inhibition of peripheral catechol-O-methyltransferase and monoamine oxidase type B: differing response in relation to presynaptic dopaminergic dysfunction. Synapse 1997; 27: 336–46PubMedCrossRef
74.
go back to reference Merello M, Lees AJ, Webster R, et al. 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 Psychiatry 1994; 57: 186–9PubMedCrossRef Merello M, Lees AJ, Webster R, et al. 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 Psychiatry 1994; 57: 186–9PubMedCrossRef
75.
go back to reference Davis TL, Roznoski M, Burns RS. Effects of tolcapone in Parkinson’s patients taking L-dihydroxyphenylalanine/carbidopa and selegiline. Mov Disord 1995; 10: 349–51PubMedCrossRef Davis TL, Roznoski M, Burns RS. Effects of tolcapone in Parkinson’s patients taking L-dihydroxyphenylalanine/carbidopa and selegiline. Mov Disord 1995; 10: 349–51PubMedCrossRef
76.
go back to reference Parkinson Study Group. Entacapone improves motor fluctuations in levodopa-treated Parkinson’s disease patients. Ann Neurol 1997; 42: 747–55CrossRef Parkinson Study Group. Entacapone improves motor fluctuations in levodopa-treated Parkinson’s disease patients. Ann Neurol 1997; 42: 747–55CrossRef
77.
go back to reference Rinne UK, Larsen JP, Siden Å, et al. Entacapone enhances the response to levodopa in parkinsonian patients with motor fluctuations. Neurology 1998; 51: 1309–14PubMedCrossRef Rinne UK, Larsen JP, Siden Å, et al. Entacapone enhances the response to levodopa in parkinsonian patients with motor fluctuations. Neurology 1998; 51: 1309–14PubMedCrossRef
78.
go back to reference Adler CH, Singer C, O’Brien C, et al. Randomized, placebocontrolled study of tolcapone in patients with fluctuating Parkinson disease treated with levodopa-carbidopa. Arch Neurol 1998; 55: 1089–95PubMedCrossRef Adler CH, Singer C, O’Brien C, et al. Randomized, placebocontrolled study of tolcapone in patients with fluctuating Parkinson disease treated with levodopa-carbidopa. Arch Neurol 1998; 55: 1089–95PubMedCrossRef
79.
go back to reference Kurth MC, Adler CH, Hilaire MS, et al. Tolcapone improves motor function and reduces levodopa requirement in patients with Parkinson’s disease experiencing motor fluctuations: a multicenter, double-blind, randomized, placebo-controlled trial. Tolcapone Fluctuator Study Group I. Neurology 1997; 48: 81–7 Kurth MC, Adler CH, Hilaire MS, et al. Tolcapone improves motor function and reduces levodopa requirement in patients with Parkinson’s disease experiencing motor fluctuations: a multicenter, double-blind, randomized, placebo-controlled trial. Tolcapone Fluctuator Study Group I. Neurology 1997; 48: 81–7
80.
go back to reference Myllylä W, Jackson M, Larsen JP, et al. Efficacy and safety of tolcapone in levodopa-treated Parkinson’s disease patients with ‘wearing-off’ phenomenon: a multicentre, double-blind, randomized, placebo-controlled study. Eur J Neurol 1997; 4: 333–41CrossRef Myllylä W, Jackson M, Larsen JP, et al. Efficacy and safety of tolcapone in levodopa-treated Parkinson’s disease patients with ‘wearing-off’ phenomenon: a multicentre, double-blind, randomized, placebo-controlled study. Eur J Neurol 1997; 4: 333–41CrossRef
81.
go back to reference Baas H, Beiske AG, Ghika J, et al. Catechol-O-methyl-transferase inhibition with tolcapone reduces the ‘wearing off’ phenomenon and levodopa requirements in fluctuating parkinsonian patients. J Neurol Neurosurg Psychiatry 1997; 63: 421–8PubMedCrossRef Baas H, Beiske AG, Ghika J, et al. Catechol-O-methyl-transferase inhibition with tolcapone reduces the ‘wearing off’ phenomenon and levodopa requirements in fluctuating parkinsonian patients. J Neurol Neurosurg Psychiatry 1997; 63: 421–8PubMedCrossRef
82.
go back to reference Rajput AH, Martin W, Sainthilaire MH, et al. Tolcapone improves motor function in parkinsonian patients with the ‘wearing-off’ phenomenon: a double-blind, placebo-controlled, multicenter trial. Neurology 1997; 49: 1066–71PubMedCrossRef Rajput AH, Martin W, Sainthilaire MH, et al. Tolcapone improves motor function in parkinsonian patients with the ‘wearing-off’ phenomenon: a double-blind, placebo-controlled, multicenter trial. Neurology 1997; 49: 1066–71PubMedCrossRef
83.
go back to reference Limousin P, Pollak P, Gervason-Tournier CL, et al. Ro 40-7592, a COMT inhibitor, plus levodopa in Parkinson’s disease. Lancet 1993; 341: 1605PubMedCrossRef Limousin P, Pollak P, Gervason-Tournier CL, et al. Ro 40-7592, a COMT inhibitor, plus levodopa in Parkinson’s disease. Lancet 1993; 341: 1605PubMedCrossRef
84.
go back to reference Roberts JW, Cora-Locatelli G, Bravi D, et al. Catechol-O-methyltransferase (COMT) inhibitor Ro 40-7592 prolongs duration of action of levodopa/carbidopa in parkinsonian patients. Neurology 1993; 43 Suppl. 2: A332CrossRef Roberts JW, Cora-Locatelli G, Bravi D, et al. Catechol-O-methyltransferase (COMT) inhibitor Ro 40-7592 prolongs duration of action of levodopa/carbidopa in parkinsonian patients. Neurology 1993; 43 Suppl. 2: A332CrossRef
85.
go back to reference Dupont E, Burgunder JM, Findley LJ, et al. Tolcapone added to levodopa in stable parkinsonian patients: a double-blind placebo-controlled study. Mov Disord 1997; 12: 928–34PubMedCrossRef Dupont E, Burgunder JM, Findley LJ, et al. Tolcapone added to levodopa in stable parkinsonian patients: a double-blind placebo-controlled study. Mov Disord 1997; 12: 928–34PubMedCrossRef
86.
go back to reference Waters CH, Kurth M, Bailey P, et al. Tolcapone in stable Parkinson’s disease: efficacy and safety of long-term treatment. Neurology 1997; 49: 665–71PubMedCrossRef Waters CH, Kurth M, Bailey P, et al. Tolcapone in stable Parkinson’s disease: efficacy and safety of long-term treatment. Neurology 1997; 49: 665–71PubMedCrossRef
87.
go back to reference Agid Y, Destee A, Durif F, et al. Tolcapone, bromocriptine, and Parkinson’s disease. Lancet 1997; 350: 712–3PubMedCrossRef Agid Y, Destee A, Durif F, et al. Tolcapone, bromocriptine, and Parkinson’s disease. Lancet 1997; 350: 712–3PubMedCrossRef
88.
go back to reference Harper J, Vieira B. Catechol-O-methyltransferase inhibitors in Parkinson’s disease. Lancet 1998; 352: 578PubMedCrossRef Harper J, Vieira B. Catechol-O-methyltransferase inhibitors in Parkinson’s disease. Lancet 1998; 352: 578PubMedCrossRef
89.
go back to reference Henry C, Wilson JA. Catechol-O-methyltransferase inhibitors in Parkinson’s disease. Lancet 1998; 351: 1965–6PubMedCrossRef Henry C, Wilson JA. Catechol-O-methyltransferase inhibitors in Parkinson’s disease. Lancet 1998; 351: 1965–6PubMedCrossRef
90.
go back to reference Hauser RA, Molho E, Shale H, et al. A pilot evaluation of the tolerability, safety, and efficacy of tolcapone alone and in combination with oral selegiline in untreated Parkinson’s disease patients. Mov Disord 1998; 13: 643–7PubMedCrossRef Hauser RA, Molho E, Shale H, et al. A pilot evaluation of the tolerability, safety, and efficacy of tolcapone alone and in combination with oral selegiline in untreated Parkinson’s disease patients. Mov Disord 1998; 13: 643–7PubMedCrossRef
91.
92.
go back to reference EMEA. Recommendation for the suspension of the marketing authorisation for Tasmar (tolcapone) [press release]. Vol. CPMP/2457/98. London, 1998 EMEA. Recommendation for the suspension of the marketing authorisation for Tasmar (tolcapone) [press release]. Vol. CPMP/2457/98. London, 1998
93.
go back to reference Tasmar Product Label. Nutley (NJ): Roche Laboratories Inc., 1998 Tasmar Product Label. Nutley (NJ): Roche Laboratories Inc., 1998
94.
go back to reference Jorga KM, Larsen JP, Beiske A, et al. The effect of tolcapone on the pharmacokinetics of benserazide. Eur J Neurol 1999; 6: 211–19PubMedCrossRef Jorga KM, Larsen JP, Beiske A, et al. The effect of tolcapone on the pharmacokinetics of benserazide. Eur J Neurol 1999; 6: 211–19PubMedCrossRef
95.
go back to reference Tedroff J, Hartvig P, Bjurling P, et al. Central action of benserazide after COMT inhibition demonstrated in vivo by PET. J Neural Transm Gen Sect 1991; 85: 11–7PubMedCrossRef Tedroff J, Hartvig P, Bjurling P, et al. Central action of benserazide after COMT inhibition demonstrated in vivo by PET. J Neural Transm Gen Sect 1991; 85: 11–7PubMedCrossRef
96.
go back to reference Illi A, Sundberg S, Ojala-Karlsson P, et al. The effect of entacapone on the disposition and hemodynamic effects of intravenous isoproterenol and epinephrine. Clin Pharmacol Ther 1995; 58: 221–7PubMedCrossRef Illi A, Sundberg S, Ojala-Karlsson P, et al. The effect of entacapone on the disposition and hemodynamic effects of intravenous isoproterenol and epinephrine. Clin Pharmacol Ther 1995; 58: 221–7PubMedCrossRef
97.
go back to reference Lyytinen J, Kaakkola S, Teräväinen H, et al. Comparison between the effects of L-dopa + entacapone and L-dopa + placebo on exercise capacity, haemodynamics and autonomic function in patients with Parkinson’s disease. Mov Disord 1997; 12 Suppl. 1: 103CrossRef Lyytinen J, Kaakkola S, Teräväinen H, et al. Comparison between the effects of L-dopa + entacapone and L-dopa + placebo on exercise capacity, haemodynamics and autonomic function in patients with Parkinson’s disease. Mov Disord 1997; 12 Suppl. 1: 103CrossRef
98.
go back to reference Sedek G, Jorga K, Yoo K, et al. Lack of interaction between ephedrine and combination of tolcapone and sinemet. eurology 1996; 46 Suppl. 2: 374 Sedek G, Jorga K, Yoo K, et al. Lack of interaction between ephedrine and combination of tolcapone and sinemet. eurology 1996; 46 Suppl. 2: 374
99.
go back to reference Illi A, Sundberg S, Ojala-Karlsson P, et al. Simultaneous inhibition of catechol-O-methyltransferase and monoamine oxidase A: effects on hemodynamics and catecholamine metabolism in healthy volunteers. Clin Pharmacol Ther 1996; 59: 450–7PubMedCrossRef Illi A, Sundberg S, Ojala-Karlsson P, et al. Simultaneous inhibition of catechol-O-methyltransferase and monoamine oxidase A: effects on hemodynamics and catecholamine metabolism in healthy volunteers. Clin Pharmacol Ther 1996; 59: 450–7PubMedCrossRef
100.
go back to reference Illi A, Sundberg S, Ojala-Karlsson P, et al. Simultaneous inhibition of catecholamine-O-methylation by entacapone and neuronal uptake by imipramine: lack of interactions. Eur J Clin Pharmacol 1996; 51: 273–6PubMedCrossRef Illi A, Sundberg S, Ojala-Karlsson P, et al. Simultaneous inhibition of catecholamine-O-methylation by entacapone and neuronal uptake by imipramine: lack of interactions. Eur J Clin Pharmacol 1996; 51: 273–6PubMedCrossRef
101.
go back to reference Jorga K, Fotteler B, Sedek G, et al. Effect of the COMT inhibitor tolcapone on the hemodynamics effects and tolerability of the combination treatment with levodopa/carbidopa and desipramine in healthy volunteers. Neurology 1997; 48 Suppl.: A185 Jorga K, Fotteler B, Sedek G, et al. Effect of the COMT inhibitor tolcapone on the hemodynamics effects and tolerability of the combination treatment with levodopa/carbidopa and desipramine in healthy volunteers. Neurology 1997; 48 Suppl.: A185
102.
go back to reference Campbell NRC, Hasinoff BB. Iron supplements: a common cause of drug interactions. Br J Clin Pharmacol 1991; 31: 251–5PubMedCrossRef Campbell NRC, Hasinoff BB. Iron supplements: a common cause of drug interactions. Br J Clin Pharmacol 1991; 31: 251–5PubMedCrossRef
103.
go back to reference Orama M, Tilus P, Taskinen J, et al. Iron(III)-chelating properties of the novel catechol O-methyltransferase inhibitor entacapone in aqueous solution. J Pharm Sci 1997; 86: 827–31PubMedCrossRef Orama M, Tilus P, Taskinen J, et al. Iron(III)-chelating properties of the novel catechol O-methyltransferase inhibitor entacapone in aqueous solution. J Pharm Sci 1997; 86: 827–31PubMedCrossRef
104.
go back to reference Nutt JG. Catechol-O-methyltransferase inhibitors for treatment of Parkinson’s disease. Lancet 1998; 351: 1221–2PubMedCrossRef Nutt JG. Catechol-O-methyltransferase inhibitors for treatment of Parkinson’s disease. Lancet 1998; 351: 1221–2PubMedCrossRef
105.
go back to reference Khromova I, Voronina T, Kraineva VA, et al. Effects of selective catechol-O-methyltransferase inhibitors on single-trial passive avoidance retention in male rats. Behav Brain Res 1997; 86: 49–57PubMedCrossRef Khromova I, Voronina T, Kraineva VA, et al. Effects of selective catechol-O-methyltransferase inhibitors on single-trial passive avoidance retention in male rats. Behav Brain Res 1997; 86: 49–57PubMedCrossRef
106.
go back to reference Moreau JL, Borgulya J, Jenck F, et al. Tolcapone: a potential new antidepressant detected in a novel animal model of depression. Behav Pharmacol 1994; 5: 344–50PubMedCrossRef Moreau JL, Borgulya J, Jenck F, et al. Tolcapone: a potential new antidepressant detected in a novel animal model of depression. Behav Pharmacol 1994; 5: 344–50PubMedCrossRef
107.
go back to reference Da Prada M, Borgulya J, Napolitano A, et al. Improved therapy of Parkinson’s Disease with tolcapone, a central and peripheral COMT inhibitor with an S-adenosyl-L-methione-sparing effect. Clin Neuropharmacol 1994; 17 Suppl. 3: S26–37CrossRef Da Prada M, Borgulya J, Napolitano A, et al. Improved therapy of Parkinson’s Disease with tolcapone, a central and peripheral COMT inhibitor with an S-adenosyl-L-methione-sparing effect. Clin Neuropharmacol 1994; 17 Suppl. 3: S26–37CrossRef
108.
go back to reference Yassin MS, Cheng H, Ekblom J, et al. Inhibitors of catecholamine metabolizing enzymes cause changes in S-adenosylmethionine and S-adenosylhomocysteine in the rat brain. Neurochem Int 1998; 32: 53–9PubMedCrossRef Yassin MS, Cheng H, Ekblom J, et al. Inhibitors of catecholamine metabolizing enzymes cause changes in S-adenosylmethionine and S-adenosylhomocysteine in the rat brain. Neurochem Int 1998; 32: 53–9PubMedCrossRef
109.
go back to reference Gasparini M, Fabrizio E, Bonifati V, et al. Cognitive improvement during tolcapone treatment in Parkinson’s disease. J Neural Transm 1997; 104: 887–94PubMedCrossRef Gasparini M, Fabrizio E, Bonifati V, et al. Cognitive improvement during tolcapone treatment in Parkinson’s disease. J Neural Transm 1997; 104: 887–94PubMedCrossRef
Metadata
Title
Clinical Pharmacology, Therapeutic Use and Potential of COMT Inhibitors in Parkinson’s Disease
Author
Dr Seppo Kaakkola
Publication date
01-06-2000
Publisher
Springer International Publishing
Published in
Drugs / Issue 6/2000
Print ISSN: 0012-6667
Electronic ISSN: 1179-1950
DOI
https://doi.org/10.2165/00003495-200059060-00004

Other articles of this Issue 6/2000

Drugs 6/2000 Go to the issue

Adis Drug Evaluation

Exemestane

Adis Drug Evaluation

Infliximab

Disease Management

Neuroblastoma