Influence of CYP2C9, 2C19 and 2D6 genetic polymorphisms on the steady-state plasma concentrations of the enantiomers of fluoxetine and norfluoxetine.

The antidepressant fluoxetine is administered as racemic mixture of two enantiomers (S- and R-fluoxetine). While S- and R-fluoxetine are equipotent in blocking serotonin reuptake, the enantiomers of the demethylated metabolite, norfluoxetine, show marked differences in pharmacological activity, S-norfluoxetine being about 20 times as potent as R- norfluoxetine as a serotonin reuptake inhibitor. In vitro and in vivo data suggest that the metabolism of fluoxetine to norfluoxetine is stereoselective and mediated, at least in part, by the polymorphic cytochrome P450 (CYP) isoenzymes CYP2D6, CYP2C9 and CYP2C19. In the present study, the influence of CYP2D6, CYP2C9 and CYP2C19 polymorphisms on the steady-state plasma concentrations of fluoxetine and norfluoxetine enantiomers was evaluated in 78 patients receiving chronic fluoxetine treatment (10-60 mg/day). The plasma concentrations of fluoxetine and norfluoxetine enantiomers were measured and CYP2D6, CYP2C9 and CYP2C19 genotypes were analyzed. No statistically significant relationship was identified between CYP2D6 or CYP2C19 genotypes and the dose normalised plasma concentrations of any of the enantiomers or the active moiety (i.e. the sum of S-fluoxetine, R-fluoxetine and S-norfluoxetine). However, the plasma concentration of S-norfluoxetine was very low in the only CYP2D6 poor metaboliser. Furthermore, the median S-norfluoxetine/S-fluoxetine ratios were higher in homozygous than in heterozygous extensive metabolisers (P<0.05). Among homozygous extensive metabolizers for CYP2D6, patients homozygous for CYP2C9*1 had lower dose-normalized R-fluoxetine concentrations and lower active moiety levels compared with those carrying detrimental CYP2C9 alleles (P<0.05). These results suggest that CYP2D6 and CYP2C9 polymorphisms contribute to the interindividual variability in fluoxetine pharmacokinetics at steady-state.

[1]  R. Berecz,et al.  Effect of CYP2D6 and CYP2C9 genotypes on fluoxetine and norfluoxetine plasma concentrations during steady-state conditions , 2004, European Journal of Clinical Pharmacology.

[2]  M. Ansseau,et al.  Polymorphisms in the CYP 2D6 Gene: Association with Plasma Concentrations of Fluoxetine and Paroxetine , 2003, Therapeutic drug monitoring.

[3]  E. Perucca,et al.  Improved enantioselective assay for the determination of fluoxetine and norfluoxetine enantiomers in human plasma by liquid chromatography. , 2003, Journal of chromatography. B, Analytical technologies in the biomedical and life sciences.

[4]  E. Spina,et al.  Influence of CYP2C9 and CYP2C19 genetic polymorphisms on warfarin maintenance dose and metabolic clearance , 2002, Clinical pharmacology and therapeutics.

[5]  P. Magni,et al.  Plasma Concentrations of the Enantiomers of Fluoxetine and Norfluoxetine: Sources of Variability and Preliminary Observations on Relations With Clinical Response , 2002, Therapeutic drug monitoring.

[6]  L. Bertilsson,et al.  Molecular genetics of CYP2D6: clinical relevance with focus on psychotropic drugs. , 2002, British journal of clinical pharmacology.

[7]  Erik Eliasson,et al.  Pharmacokinetics of losartan and its metabolite E‐3174 in relation to the CYP2C9 genotype , 2002, Clinical pharmacology and therapeutics.

[8]  Zhaoqian Liu,et al.  O-Dealkylation of fluoxetine in relation to CYP2C19 gene dose and involvement of CYP3A4 in human liver microsomes. , 2002, The Journal of pharmacology and experimental therapeutics.

[9]  M. Ingelman-Sundberg,et al.  Genetic polymorphism of cytochrome P450 2C9 in a Caucasian and a black African population. , 2001, British journal of clinical pharmacology.

[10]  J. Goldstein,et al.  Clinical relevance of genetic polymorphisms in the human CYP2C subfamily. , 2001, British journal of clinical pharmacology.

[11]  H. Zhou,et al.  Effect of the CYP2C19 oxidation polymorphism on fluoxetine metabolism in Chinese healthy subjects. , 2001, British journal of clinical pharmacology.

[12]  C. Eap,et al.  Concentrations of the Enantiomers of Fluoxetine and Norfluoxetine After Multiple Doses of Fluoxetine in Cytochrome P4502D6 Poor and Extensive Metabolizers , 2001, Journal of clinical psychopharmacology.

[13]  S. Binkley,et al.  Identification of the human cytochromes p450 responsible for in vitro formation of R- and S-norfluoxetine. , 2001, The Journal of pharmacology and experimental therapeutics.

[14]  M. Reis,et al.  Serum Concentrations of Fluoxetine in the Clinical Treatment Setting , 2001, Therapeutic drug monitoring.

[15]  H. Zhou,et al.  Effects of CYP2C19 genotype and CYP2C9 on fluoxetine N-demethylation in human liver microsomes. , 2001, Acta pharmacologica Sinica.

[16]  S. Ekins,et al.  (R)-, (S)-, and racemic fluoxetine N-demethylation by human cytochrome P450 enzymes. , 2000, Drug metabolism and disposition: the biological fate of chemicals.

[17]  C. Alm,et al.  Decreased capacity for debrisoquine metabolism among black Tanzanians: analyses of the CYP2D6 genotype and phenotype. , 1999, Pharmacogenetics.

[18]  K. Brøsen,et al.  The stereoselective metabolism of fluoxetine in poor and extensive metabolizers of sparteine. , 1999, Pharmacogenetics.

[19]  M. Ingelman-Sundberg,et al.  Validation of methods for CYP2C9 genotyping: frequencies of mutant alleles in a Swedish population. , 1999, Biochemical and biophysical research communications.

[20]  M. Ingelman-Sundberg,et al.  Genetic mechanisms for duplication and multiduplication of the human CYP2D6 gene and methods for detection of duplicated CYP2D6 genes. , 1999, Gene.

[21]  Joe C. Watson,et al.  Report from the Second European Stroke Summer School, Heidelberg, Germany , 1999, Cerebrovascular Diseases.

[22]  J. Miners,et al.  Cytochrome P4502C9: an enzyme of major importance in human drug metabolism. , 1998, British journal of clinical pharmacology.

[23]  A. Holtz,et al.  Fluoxetine tenth anniversary update: the progress continues. , 1997, Clinical therapeutics.

[24]  D. Greenblatt,et al.  Human cytochromes mediating N-demethylation of fluoxetine in vitro , 1997, Psychopharmacology.

[25]  J. Brockmöller,et al.  Cytochrome P450 2D6 variants in a Caucasian population: allele frequencies and phenotypic consequences. , 1997, American journal of human genetics.

[26]  J. Turgeon,et al.  The disposition of fluoxetine but not sertraline is altered in poor metabolizers of debrisoquin , 1996, Clinical pharmacology and therapeutics.

[27]  W. Trager,et al.  Allelic variants of human cytochrome P450 2C9: baculovirus-mediated expression, purification, structural characterization, substrate stereoselectivity, and prochiral selectivity of the wild-type and I359L mutant forms. , 1996, Archives of biochemistry and biophysics.

[28]  G R Wilkinson,et al.  Identification of a new genetic defect responsible for the polymorphism of (S)-mephenytoin metabolism in Japanese. , 1994, Molecular pharmacology.

[29]  A. Altamura,et al.  Clinical Pharmacokinetics of Fluoxetine , 1994, Clinical pharmacokinetics.

[30]  L. Wienkers,et al.  Impaired (S)-warfarin metabolism catalysed by the R144C allelic variant of CYP2C9. , 1994, Pharmacogenetics.

[31]  S. Wrighton,et al.  Interaction of the enantiomers of fluoxetine and norfluoxetine with human liver cytochromes P450. , 1993, The Journal of pharmacology and experimental therapeutics.

[32]  D. Wong,et al.  Norfluoxetine Enantiomers as Inhibitors of Serotonin Uptake in Rat Brain , 1993, Neuropsychopharmacology.

[33]  R. Fuller,et al.  Comparison of norfluoxetine enantiomers as serotonin uptake inhibitors in vivo , 1992, Neuropharmacology.

[34]  U. Meyer,et al.  Genotyping of poor metabolisers of debrisoquine by allele-specific PCR amplification , 1990, The Lancet.

[35]  D. Wong,et al.  Inhibition of serotonin uptake by optical isomers of fluoxetine , 1985 .