In vitro metabolism of tolcapone to reactive intermediates: relevance to tolcapone liver toxicity

Chem Res Toxicol. 2003 Feb;16(2):123-8. doi: 10.1021/tx025569n.

Abstract

Tolcapone is a catechol-O-methyltransferase (COMT) inhibitor used for control of motor fluctuations in Parkinson's disease (PD). Since its entry onto the market in 1998, tolcapone has been associated with numerous cases of hepatotoxicity, including three cases of fatal fulminant hepatic failure. The cause of this toxicity is not known; however, it does not occur with the use of the structurally similar drug entacapone. It is known that tolcapone is metabolized to amine (M1) and acetylamine (M2) metabolites in humans, but that the analogous metabolites were not detected in a limited human study of entacapone metabolism. We hypothesized that one or both of these tolcapone metabolites could be oxidized to reactive species and that these reactive metabolites might play a role in tolcapone-induced hepatocellular injury. To investigate this possibility, we examined the ability of M1 and M2 to undergo in vitro bioactivation by electrochemical and enzymatic methods. Electrochemical experiments revealed that M1 and M2 are more easily oxidized than the parent compound, in the order M1 > M2 > tolcapone. There was a general correlation between oxidation potential and the half-lives of the compounds in the presence of two oxidizing systems, horseradish peroxidase and myeloperoxidase. These enzymes catalyzed the oxidation of M1 and M2 to reactive species that could be trapped with glutathione (GSH) to form metabolite adducts (C1 and C2). Each metabolite was found to only form one GSH conjugate, and the structures were tentatively identified using LC-MS/MS. Following incubation of M1 and M2 with human liver microsomes in the presence of GSH, the same adducts were observed, and their structures were confirmed using LC-MS/MS and (1)H NMR. Experiments with chemical P450 inhibitors and cDNA-expressed P450 enzymes revealed that this oxidation is catalyzed by several P450s, and that P450 2E1 and 1A2 play the primary role in the formation of C1 while P450 1A2 is most important for the production of C2. Taken together, these data provide evidence that tolcapone-induced hepatotoxicity may be mediated through the oxidation of the known urinary metabolites M1 and M2 to reactive intermediates. These reactive species may form covalent adducts to hepatic proteins, resulting in damage to liver tissues, although this supposition was not investigated in this study.

MeSH terms

  • Acetylation
  • Amines / chemistry
  • Amines / metabolism
  • Benzophenones / metabolism*
  • Benzophenones / pharmacokinetics
  • Benzophenones / toxicity*
  • Catechol O-Methyltransferase Inhibitors
  • Cells, Cultured
  • Chemical and Drug Induced Liver Injury*
  • Cytochrome P-450 Enzyme Inhibitors
  • Cytochrome P-450 Enzyme System / metabolism
  • Electrochemistry / methods
  • Enzyme Inhibitors / metabolism*
  • Enzyme Inhibitors / pharmacokinetics
  • Enzyme Inhibitors / pharmacology
  • Enzyme Inhibitors / toxicity*
  • Glutathione / chemistry
  • Glutathione / metabolism
  • Half-Life
  • Hepatocytes / drug effects
  • Hepatocytes / enzymology
  • Hepatocytes / pathology
  • Horseradish Peroxidase / metabolism
  • Humans
  • Microsomes, Liver / drug effects
  • Nitrophenols
  • Nuclear Magnetic Resonance, Biomolecular
  • Oxidation-Reduction
  • Peroxidase / metabolism
  • Reactive Oxygen Species / metabolism
  • Spectrometry, Mass, Electrospray Ionization / methods
  • Tolcapone

Substances

  • Amines
  • Benzophenones
  • Catechol O-Methyltransferase Inhibitors
  • Cytochrome P-450 Enzyme Inhibitors
  • Enzyme Inhibitors
  • Nitrophenols
  • Reactive Oxygen Species
  • Cytochrome P-450 Enzyme System
  • Tolcapone
  • Horseradish Peroxidase
  • Peroxidase
  • Glutathione