Skip to main content
Log in

Formation of pyridinium species of haloperidol in human liver and brain

  • Original Investigation
  • Published:
Psychopharmacology Aims and scope Submit manuscript

Abstract

Recent interest in the neurotoxicity of haloperidol is based on its oxidation in rodents to the pyridinium derivative, HPP+, a structural analog of the neurotoxin, 1-methyl-4-phenylpyridinium (MPP+). Recently, we reported that HPP+ and a newly identified reduced pyridinium, RHPP+, were present in blood and urine of haloperidol-treated schizophrenics and that the concentrations of RHPP+ exceeded those of HPP+. In this study, we examined pathways for formation of RHPP+ in subcellular fractions of human liver (n=5) and brain (basal ganglia;n=5). The major pathway was reduction of HPP+ (20 µM) to RHPP+ in cytosol (0.17–0.39 and 0.03–0.07 µM RHPP+/g cytosolic protein per h in liver and brain, respectively). The reactions were inhibited significantly by menadione and in brain also by daunorubicin. The inhibition profile, cytosolic location and strict NADPH dependence suggest that the enzymes involved are ketone reductases. A second pathway was oxidation of reduced haloperidol (50 µM), a major metabolite of haloperidol in blood and brain, to RHPP+. In liver microsomes, 0.17–0.63 µmol RHPP+ was formed /g microsomal protein per h. A potent inhibitor of the pathway was ketoconazole (IC50, 0.8 µM), which suggests that P-450 3A isozymes could be involved. In brain mitochondria but not microsomes, reduced haloperidol (120 µM) was oxidised to RHPP+ at a small but significant rate (0.005–0.020 µmol RHPP+/g mitochondrial protein per h) which was not attenuated by SKF 525A, quinidine, ketoconazole, or monoamine oxidase inhibitors. Further studies are warranted to establish the biological importance of these metabolites in vivo.

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.

Similar content being viewed by others

References

  • Bachur NR (1976) Cytoplasmic aldo-keto reductases: a class of drug metabolising enzymes. Science 193:595–597

    PubMed  Google Scholar 

  • Bloomquist J, King E, Wright A, Mytilineou C, Kimura K, Castagnoli K, Castagnoli Jr N (1994) 1-Methyl-4-phenylpyridinium-like neurotoxicity of a pyridinium metabolite derived from haloperidol: cell culture and neurotransmitter uptake studies. J Pharmacol Exp Ther 270:822–830

    PubMed  Google Scholar 

  • Bocker RH, Guengerich FP (1986) Oxidation of 4-aryl- and 4-alkyl-substituted 2,6-dimethly-3,5-bis(alkoxycarbonyl)-1,4-dihydropyridines by human liver microsomes and immunochemical evidence for the involvement of a form of cytochrome P-450. J Med Chem 29:1596–1603

    Article  PubMed  Google Scholar 

  • Eyles DW, Pond SM (1992) Stereospecific reduction of haloperidol in human tissues. Biochem Pharmacol 44:867–871

    Article  PubMed  Google Scholar 

  • Eyles DW, McLennan HR, Jones A, McGrath JJ, Stedman TJ, Pond SM (1994a) Quantitative analysis of two pyridinium metabolites of haloperidol in patients with schizophrenia. Clin Pharmacol Ther 56:512–520

    PubMed  Google Scholar 

  • Eyles DW, Stedman TJ, Pond SM (1994b) Nonlinear relationship between circulating concentrations of reduced haloperidol and haloperidol: evaluation of possible mechanisms. Psychopharmacology 116:161–166

    Article  PubMed  Google Scholar 

  • Fang J, Gorrod JW (1993) High performance liquid chromatographic method for the detection and quantitation of haloperidol and seven of its metabolites in microsomal preparations. J Chromatogr 614:267–273

    PubMed  Google Scholar 

  • Forsman A, Folsch G, Larsson M, Ohman R (1977) On the metabolism of haloperidol in man. Curr Ther Res 21:606–617

    Google Scholar 

  • Ghersi-egea J-F, Perrin R, Leininger-Muller B, Grassiot M-C, Jeandel C, Floquet J, Cuny G, Siest G, Minn A (1993) Subcellular location of cytochrome P450, and activities of several enzymes responsible for drug metabolism in the human brain. Biochem Pharmacol 45:647–658

    Article  PubMed  Google Scholar 

  • Gorrod JW, Fang J (1993) On the metabolism of haloperidol. Xenobiotica 23:495–508

    PubMed  Google Scholar 

  • Guengerich FP (1990) Mechanism-based inactivation of liver microsomal cytochrome P-450 IIIA4 by gestodene. Chem Res Toxicol 3:363–371

    Article  PubMed  Google Scholar 

  • Halpert JR, Guengerich FP, Bend JR, Correia MA (1994) Selective inhibitors of cytochromes P450. Toxicol Appl Pharmacol 125:163–175

    Article  PubMed  Google Scholar 

  • Igarashi K, Castagnoli Jr N (1992) Determination of the pyridinium metabolite derived from haloperidol in brain tissue, plasma and urine by high-performance liquid chromatography with fluorescence detection. J Chromatogr 579:277–283

    PubMed  Google Scholar 

  • Korpi ER, Kleinman JE, Costakos DT, Linnoila M, Wyatt RJ (1984) Reduced haloperidol in the post-mortem brains of haloperidol-treated patients. Psychiatry Res 11:259–269

    Article  PubMed  Google Scholar 

  • Langston JW, Ballard P, Tetrud JW, Irwin I (1983) Chronic parkinsonism in humans due to a product of meperidine-analog synthesis. Science 219:979–980

    Google Scholar 

  • Lerena AL, Alm C, Dahl M-L, Ekqvist B, Bertilsson L (1992) Haloperidol disposition is dependent on debrisoquine hydroxylation phenotype. Ther Drug Monit 14:92–97

    PubMed  Google Scholar 

  • Lowry OH, Rosebrough NJ, Farr AL, Randall RJ (1951) Protein measurement with the folin phenol reagent. J Biol Chem 193:265–275

    PubMed  Google Scholar 

  • Oida T, Terauchi Y, Yoshida K, Kagemoto A, Sekine Y (1989) Use of antisera in the isolation of human specific conjugates of haloperidol. Xenobiotica 19:781–793

    PubMed  Google Scholar 

  • Rollema H, Skolnik M, D'Engelbronner J, Igarashi K, Usuki E, Castagnoli Jr N (1994) MPP+-like neurotoxicity of a pyridinium metabolite derived from haloperidol: in vivo microdialysis and in vitro mitochondrial studies. J Pharmacol Exp Ther 268:380–387

    PubMed  Google Scholar 

  • Someya T, Shibasaki M, Noguchi T, Takahashi S, Inaba T (1992) Haloperidol metabolism in psychiatric patients: importance of glucuronidation and carbonyl reduction. J Clin Psychopharmacol 12:169–174

    PubMed  Google Scholar 

  • Spina E, Ancione M, Di Rosa AE, Meduri M, Caputi AP (1992) Polymorphic debrisoquine oxidation and acute neuroleptic-induced adverse effects. Eur J Clin Pharmacol 42:347–348

    Article  PubMed  Google Scholar 

  • Stedman TJ, Whiteford HA Eyles DW, Welham JL, Pond SM (1991) Effects of nifedipine on psychosis and tardive dyskinesia in schizophrenic patients. J Clin Psychopharmacl 11:43–47

    Google Scholar 

  • Subramanyam B, Rollema H, Woolf T, Castagnoli Jr N (1990) Identification of a potentially neurotoxic pyridinium metabolite of haloperidol in rats. Biochem Biophys Res Commun 166:238–244

    Article  PubMed  Google Scholar 

  • Subramanyam B, Woolf T, Castagnoli Jr N (1991) Studies on the in vitro conversion of haloperidol to a potentially neurotoxic pyridinium metabolite. Chem Res Toxicol 4:123–128

    Article  PubMed  Google Scholar 

  • Tipton KF, Singer TP (1993) Advances in our understanding of the mechanisms of the neurotoxicity of MPTP and related compounds. J Neurochem 61:1191–1206

    PubMed  Google Scholar 

  • Van der Schyf CJ, Castagnoli K, Usuki E, Fouda HG, Rimoldi JM, Castagnoli Jr N (1994) Metabolic studies of haloperidol and its tetrahydropyridine analog in C57BL/6 mice. Chem Res Toxicol 7:281–285

    Article  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

About this article

Cite this article

Eyles, D.W., McGrath, J.J. & Pond, S.M. Formation of pyridinium species of haloperidol in human liver and brain. Psychopharmacology 125, 214–219 (1996). https://doi.org/10.1007/BF02247331

Download citation

  • Received:

  • Revised:

  • Issue Date:

  • DOI: https://doi.org/10.1007/BF02247331

Key words

Navigation