Skip to main content
Log in

Antidepressant Drug Interactions and the Cytochrome P450 System

The Role of Cytochrome P450 2D6

  • Published:
Clinical Pharmacokinetics Aims and scope Submit manuscript

Summary

The selective serotonin reuptake inhibitors (SSRIs) and venlafaxine display the following rank order of in vitro potency against the cytochrome P450 (CYP) isoenzyme CYP2D6 as measured by their inhibition of sparteine and/or dextromethorphan metabolism: paroxetine > fluoxetine ≡ norfluoxetine ≥ sertraline ≥ fluvoxamine > venlafaxine. On this basis, paroxetine would appear to have the greatest and fluvoxamine and venlafaxine the least potential for drug interactions with CYP2D6-dependent drugs.

In vivo, inhibitory potency is affected by the plasma concentration of the free (unbound) drug, a potentially important consideration since many CYP2D6-metabolised drugs exhibit nonlinear (saturable) kinetics, and by the presence of metabolites, which might accumulate and interact with the CYP system. Under steady-state conditions, paroxetine and fluoxetine are approximately clinically equipotent inhibitors of CYP2D6 in vivo (as determined through their effects on desipramine metabolism); sertraline, in contrast, shows lower steady-state plasma concentrations than fluoxetine and, hence, a less pronounced inhibition of CYP2D6.

Of the drugs that are metabolised by CYP2D6, secondary amine tricyclic antidepressants, antipsychotics (e.g. phenothiazines and risperidone), codeine, some antiarrhythmics (e.g. flecainide) and β-blockers form the focus of clinical attention with regard to their potential interactions with the SSRIs. Coadministration of desipramine and fluoxetine (20 mg/day) at steady-state produced an ≈ 4-fold elevation in peak plasma desipramine concentrations, while the long half-life of the active metabolite norfluoxetine was responsible for a significant and long lasting (≈ 3 weeks) elevation of plasma desipramine concentrations after discontinuation of fluoxetine. Similarly, coadministration of desipramine with paroxetine produced an ≈ 3-fold increase in plasma desipramine concentrations. In contrast, coadministration of desipramine and sertraline (50 mg/day) for 4 weeks resulted in a considerably more modest (≈ 30%) elevation in plasma desipramine concentrations. Coadministration of fluoxetine (60 mg/day, as a loading dose) [equivalent to serum concentrations obtained with 20 mg/day at steady-state] with imipramine or desipramime resulted in ≈ 3- to 4-fold increases in plasma area under the curve (AUC) values for both imipramine and desipramine (illustrating a significant drug interaction potential at multiple isoenzymes). Consistent with its minimal in vitro effect on CYP2D6, fluvoxamine shows minimal in vivo pharmacokinetic interaction with desipramine, but does interact with imipramine (≈ 3- to 4-fold increase in AUC) through inhibition of CYP3A3/4, CYP1A2, and CYP2C19. Thus, the extent of the in vivo interaction between the SSRIs and tricyclic antidepressants mirrors to a large extent their in vitro inhibitory potencies against CYP2D6 and other isoenzyme systems, especially if one takes into account pharmacokinetic factors.

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

  1. Ereshefsky L, LeRoy A, Tran-Johnson T, et al. Pharmacokinetic factors affecting antidepressant drug clearance and clinical effect: evaluation of doxepin and imipramine — new data and review. Clin Chem 1988; 34: 863–80

    PubMed  CAS  Google Scholar 

  2. Toney G, Ereshefsky L. The pharmacokinetics of cyclic antidepressants and their relationship to therapeutic drug monitoring. Am Assoc Clin Chem 1990; 12(5): 7–12

    Google Scholar 

  3. Nebert DW, Nelson DR, Coon MJ, et al. The P-450 superfamily: update on new sequences, gene mapping, accession numbers, early triorial names of enzymes and nomenclature. DNA Cell Biol 1991; 10: 1–14

    Article  PubMed  CAS  Google Scholar 

  4. Preskorn SH, Alderman J, Chung M, et al. Pharmacokinetics of desipramine coadministered with sertraline and fluoxetine. J Clin Psychopharmacol 1994; 14: 90–8

    PubMed  CAS  Google Scholar 

  5. Watkins PB. Noninvasive tests of CYP3A enzymes. Pharmacogenetics 1994; 4: 171–84

    Article  PubMed  CAS  Google Scholar 

  6. von Moltke LL, Greenblatt DJ, Cotreau-Bibbo MM, et al. Inhibition of desipramine hydroxylation in vitro by serotonin reuptake inhibitor antidepressants, and by quinidine and ketoconazole: a model system to predict drug interactions in vivo. J Pharmacol Exp Ther 1994; 268: 1278–83

    Google Scholar 

  7. Geungerich FP, Muller-Enoch D, Blair IA. Oxidation of quinidine by human liver cytochrome P-450. Mol Pharmacol 1986; 30: 287–95

    Google Scholar 

  8. Brøsen K, Zeugin T, Meyer UA. Role of P450IID6, the target of the sparteine-debrisoquin oxidation polymorphism, in the metabolism of imipramine. Clin Pharmacol Ther 1991; 49: 609–17

    Article  PubMed  Google Scholar 

  9. Lemoine A, Gautier JC, Azoulay D, et al. Major pathway of imipramine metabolism is catalyzed by cytochromes P-450 1A2 and P-450 3A4 in human liver. Mol Pharmacol 1993; 43: 827–32

    PubMed  CAS  Google Scholar 

  10. Skjelbo E, Brosen K, Hallas J, et al. The mephenytoin oxidation polymorphism is partially responsible for the N-demethylation of imipramine. Clin Pharmacol Ther 1991; 49: 18–23

    Article  PubMed  CAS  Google Scholar 

  11. Ereshefsky L. Drug interactions of antidepressants [presentation]. American Psychiatric Association Annual Meeting, Miami Beach, 1995

  12. Jacqz-Aigrain E, Funck-Brentano C, Cresteil T. CYP2D6- and CYP3A-dependent metabolism of dextromethorphan in humans. Pharmacogenetics 1993; 3: 197–204

    Article  PubMed  CAS  Google Scholar 

  13. Schmid B, Bircher J, Preisig R, et al. Polymorphic dextromethorphan metabolism: cosegregation of oxidative O-demethyl-ation with debrisoquin hydroxylation. Clin Pharmacol Ther 1985; 38: 618–24

    Article  PubMed  CAS  Google Scholar 

  14. Ereshefsky L, Anderson CB, True JE, et al. Plasma concentrations of oral risperidone and active metabolite in schizophrenics [abstract]. Pharmacotherapy 1993; 13: 292

    Google Scholar 

  15. Otton SV, Wu D, Joffe R, et al. Inhibition by fluoxetine of cytochrome P450 2D6 activity. Clin Pharmacol Ther 1993; 53: 401–9

    Article  PubMed  CAS  Google Scholar 

  16. Lam YWF, Rodriguez SY. High performance liquid chromatography determination of dextromethorphan and extrorphan for oxidative phenotyping by fluorescence and ultraviolet detection. Ther Drug Monit 1993; 15: 300–4

    Article  PubMed  CAS  Google Scholar 

  17. Lam YWF, Casto DT, Dunn JF. Drug metabolizing capacity in Mexican Americans. Clin Pharmacol Ther 1991; 49: 159

    Google Scholar 

  18. Lam YWF, Rodriguez SY, Casto DT. No time-dependent change in cytochrome P450 2D6 phenotyping results during HPLC analysis. Clin Chem 1992; 38: 2350–2

    PubMed  CAS  Google Scholar 

  19. Crewe HK, Lennard MS, Tucker GT, et al. The effect of selective serotonin reuptake inhibitors on cytochrome P-450 2D6 activity in human liver microsomes. Br J Clin Pharmacol 1992; 34: 262–5

    Article  PubMed  CAS  Google Scholar 

  20. Otton SV, Ball SE, Cheung SW, et al. Comparative inhibition of the polymorphic enzyme CYP2D6 by venlafaxine and other 5-HT uptake inhibitors [abstract]. Clin Pharmacol Ther 1994; 55: 141

    Google Scholar 

  21. Stevens JC, Wrighton SA. Interaction of the enantiomers of fluoxetine and norfluoxetine with human liver cytochromes P450. J Pharmacol Exp Ther 1993; 266: 964–71

    PubMed  CAS  Google Scholar 

  22. Wong DT, Threlkeld PG, Robertson DW. Affinities of fluoxetine, its enantiomers, and other inhibitors of serotonin uptake for subtypes of serotonin receptors. Neuropsychopharmacol 1991; 5: 43–7

    CAS  Google Scholar 

  23. Preskorn SH, Magnus RD. Inhibition of hepatic P-450 isoenzymes by serotonin selective reuptake inhibitors: in vitro and in vivo findings and their implications for patient care. Psychopharmacol Bull 1994; 30: 251–9

    PubMed  CAS  Google Scholar 

  24. De Vane CL. Pharmacogenetics and drug metabolism of newer antidepressant agents. J Clin Psychiatr 1994; 55 Suppl.: 38–45

    Google Scholar 

  25. Llerena A, Alm C, Dahl M, et al. Haloperidol disposition is dependent on debrisoquine hydroxylation phenotype. Ther Drug Monit 1992; 14: 92–7

    Article  PubMed  CAS  Google Scholar 

  26. Tyndale RF, Kalow W, Inaba T. Oxidation of reduced haloperidol to haloperidolo: involvement of human P450IID6 (sparteine/debrisoquine monooxygenase). Br J Clin Pharmacol 1991; 31: 655–60

    Article  PubMed  CAS  Google Scholar 

  27. Goff DC, Midha KK, Brotman AW, et al. Elevation of plasma concentrations of haloperidol after addition of fluoxetine. Am J Psychiatr 1991; 148: 790–2

    PubMed  CAS  Google Scholar 

  28. Young D, Midha KK, Fossler MJ, et al. Effect of quinidine on the interconversion kinetics between haloperidol and reduced haloperidol in humans: implications for the involvement of cytochrome P450IID6. Eur J Clin Pharmacol 1993; 44: 433–8

    Article  PubMed  CAS  Google Scholar 

  29. Tsang MW, Shader RI, Greenblatt DJ. Metabolism of haloperidol: clinical implications and unanswered questions. J Clin Psychopharmacol 1994; 14: 159–62

    PubMed  CAS  Google Scholar 

  30. Brøsen K, Hansen JG, Nielsen KK, et al. Inhibition by paroxetine of desipramine metabolism in extensive but not in poor metabolisers of sparteine. Eur J Clin Pharmacol 1993a; 44: 349–55

    Article  PubMed  Google Scholar 

  31. Barros J, Asnis G. An interaction of sertraline and desipramine. Am J Psychiatr 1993; 150: 1751

    PubMed  CAS  Google Scholar 

  32. Lydiard RB, Anton RF, Cunningham T. Interactions between sertraline and tricyclic antidepressants. Am J Psychiatr 1993; 150: 1125–6

    PubMed  CAS  Google Scholar 

  33. Kurtz DL, Bergstrom RF, Goldberg MJ, et al. Drug interactions between sertraline and desipramine or imipramine [abstract]. J Clin Pharmacol 1994; 34: 1009–33

    Google Scholar 

  34. Bergstrom RF, Peyton AL, Lemberger L. Quantification and mechanism of the fluoxetine and tricyclic antidepressant interaction. Clin Pharmacol Ther 1992; 51: 239–48

    Article  PubMed  CAS  Google Scholar 

  35. Spina E, Pollicino AM, Avenoso A, et al. Effect of fluvoxamine on the pharmacokinetics of imipramine and desipramine in healthy subjects. Ther Drug Monit 1993; 15: 243–6

    Article  PubMed  CAS  Google Scholar 

  36. Brøsen K, Skjelbo E, Rasmussen BB, et al. Fluvoxamine is a potent inhibitor of cytochrome P-450 1A2. Biochem Pharmacol 1993b; 45: 1211–4

    Article  PubMed  Google Scholar 

  37. Fleishaker JC, Hulst LK. A pharmacokinetic and pharmacodynamic evaluation of the combined administration of alprazolam and fluvoxamine. Eur J Clin Pharmacol 1994; 46: 35–9

    Article  PubMed  CAS  Google Scholar 

  38. Serzone® prescribing information. Princeton, New Jersey, USA: Bristol Myers Squibb Co., Dec 1994

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

About this article

Cite this article

Ereshefsky, L., Riesenman, C. & Lam, Y.W.F. Antidepressant Drug Interactions and the Cytochrome P450 System. Clin. Pharmacokinet. 29 (Suppl 1), 10–19 (1995). https://doi.org/10.2165/00003088-199500291-00004

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00003088-199500291-00004

Keywords

Navigation