Skip to main content
Log in

Selective Serotonin Reuptake Inhibitors and CNS Drug Interactions

A Critical Review of the Evidence

  • Review Article
  • Drug Interactions
  • Published:
Clinical Pharmacokinetics Aims and scope Submit manuscript

Summary

The potential for drug-drug interactions in psychiatric patients is very high as combination psychopharmacotherapy is used to treat comorbid psychiatric disorders, to treat the adverse effects of a medication, to augment a medication effect or to treat concomitant medical illnesses. Interactions can be pharmacodynamic or pharmacokinetic in nature. This paper focuses on the metabolic kinetic interactions between selective serotonin reuptake inhibitors (SSRIs) and other central nervous system (CNS) drugs. The evidence for and clinical significance of these interactions are reviewed, with special emphasis on antipsychotics, tricyclic anti-depressants and benzodiazepines.

Many psychotropic medications have an affinity for the cytochrome P450 (CYP) enzymes which promote elimination by transforming lipid soluble substances into more polar compounds. SSRIs serve both as substrates and inhibitors of these enzymes. In vitro studies provide a screening method for evaluating drug affinities for substrates, inhibitors or inducers of CYP enzymes. Although in vitro data are important as a starting point for predicting these metabolic kinetic drug interactions, case reports and controlled experimental studies in humans are required to fully evaluate their clinical significance. Several factors must be considered when evaluating the clinical significance of a potential interaction including: (a) the nature of each drugs’ activity at an enzyme site (substrate, inhibitor or inducer); (b) the potency estimations for the inhibitor/inducer; (c) the concentration of the inhibitor/inducer at the enzyme site; (d) the saturability of the enzyme; (e) the extent of metabolism of the substrate through this enzyme (versus alternative metabolic routes); (f) the presence of active metabolites of the substrate; (g) the therapeutic window of the substrate; (h) the inherent enzyme activity of the individual, phenotyping/genotyping information; (i) the level of risk of the individual experiencing adverse effects (e.g. the elderly) and (j) from an epidemiological perspective, the probability of concurrent use.

This paper systematically reviews both the in vitro and in vivo evidence for drug interactions between SSRIs and other CNS drugs. As potent inhibitors of CYP2D6, both paroxetine and fluoxetine have the potential to increase the plasma concentrations of antipsychotic medications metabolised through this enzyme, including perphenazine, haloperidol, thioridazine and risperidone in patients who are CYP2D6 extensive metabolisers. Controlled studies have demonstrated this for perphenazine with paroxetine and haloperidol with fluoxetine. Fluvoxamine, as a potent inhibitor of CYP1A2, can inhibit the metabolism of clozapine, resulting in higher plasma concentrations.

Drug interactions between the SSRIs and tricyclic antidepressants (TCAs) can occur. Fluoxetine and paroxetine, as potent inhibitors of CYP2D6, can increase the plasma concentrations of secondary and tertiary tricyclic antidepressants. Sertraline and citalopram are less likely to have this effect. Fluvoxamine can increase the plasma concentrations of tertiary TCAs.

Fluvoxamine inhibits, via CYP3A, CYP2C19 and CYP1A2, the metabolism of several benzodiazepines, including alprazolam, bromazepam and diazepam. Fluoxetine increases the plasma concentrations of alprazolam and diazepam by inhibiting CYP3A and CYP2C19, respectively. The clinical importance of the interaction with diazepam is attenuated by the presence of its active metabolite. Sertraline inhibits these enzymes only mildly to moderately at usual therapeutic doses. Therefore the potential for interactions is less; however, the in vivo evidence is minimal. Paroxetine and citalopram are unlikely to cause interactions with benzodiazepines.

The evidence is conflicting for an interaction between carbamazepine and the SSRIs fluoxetine and fluvoxamine. These combinations should be used cautiously, and be accompanied by monitoring for adverse events and carbamazepine plasma concentrations. A lack of interaction between paroxetine or sertraline and carbamazepine has been documented.

The SSRIs are not equivalent in their potential for drug interactions when combined with other CNS medications. Each combination must be assessed individually. Several factors must be considered when predicting the outcome of a potential interaction based on in vitro data (e.g. active metabolites and concentration ranges). In vivo studies are required to evaluate their clinical significance. Generally, sertraline and citalopram at the lower therapeutic dosage range appear to have less propensity for interactions. Anticipated pharmacokinetic interactions can usually be managed with careful monitoring and appropriate adjustments in dosage and titration.

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.

Institutional subscriptions

Similar content being viewed by others

References

  1. Ross HE, Glaser FB, Germanson T. The prevalence of psychiatric disorders in patients with alcohol and other drug problems. Arch Gen Psychiatry 1988; 45: 1023–31

    PubMed  CAS  Google Scholar 

  2. Rosenbaum JF. Managing selective serotonin reuptake inhibitor-drug interactions in clinical practice. Clin Pharmacokinet 1995; 29 Suppl. 1: 53–9

    PubMed  CAS  Google Scholar 

  3. Wong DT, Bymaster FT, Reid LR, et al. Fluoxetine and two other serotonin uptake inhibitors without affinity for neuronal receptors. Biochem Pharmacol 1983; 32: 1287–93

    PubMed  CAS  Google Scholar 

  4. Kasper S, Heiden A. Do SSRIs differ in their antidepressant efficacy. Hum Psychopharmacol Clin Exp Ther 1995; 10 Suppl. 3: S163–72

    CAS  Google Scholar 

  5. den Boer JA, Westenberg HGM. Serotonergic compounds in panic disorder, obsessive-compulsive disorder and anxious depression: a concise review. Hum Psychopharmacol Clin Exp Ther 1995; 10 Suppl. 3: S173–83

    Google Scholar 

  6. Nelson DR, Koymans L, Kamataki T, et al. P450 superfamily: update on new sequences, gene mapping, accession numbers and nomenclature. Pharmacogenetics 1996; 6(1): 1–42

    PubMed  CAS  Google Scholar 

  7. Gonzalez FJ, Jeffrey RI. Pharmacogenetic phenotyping and genotyping: present status and future potential. Clin Pharmacokinet 1994; 26(1): 59–70

    PubMed  CAS  Google Scholar 

  8. Bertz RJ, Granneman GR. Use of in vitro and in vivo data to estimate the likelihood of metabolic pharmacokinetic interactions. Clin Pharmacokinet 1997; 32(3): 210–58

    PubMed  CAS  Google Scholar 

  9. Shen WW. Cytochrome P450 monooxygenases and interactions of psychotropic drugs: a five-year update. Int J Psychiatry Med 1995; 25(3): 277–90

    PubMed  CAS  Google Scholar 

  10. Daly AK, Cholerton S, Gregory W, et al. Metabolic polymorphisms. Pharmacol Ther 1993; 57: 129–60

    PubMed  CAS  Google Scholar 

  11. Ketter TA, Flockhart DA, Post RM, et al. The emerging role of cytochrome P450 3A in psychopharmacology. J Clin Psychopharmacol 1995; 15(6): 387–98

    PubMed  CAS  Google Scholar 

  12. De Vane CL. Pharmacogenetics and drug metabolism of newer antidepressant agents. J Clin Psychiatry 1994; 55 Suppl. 12: 38–45

    Google Scholar 

  13. Schmider J, Greenblatt DJ, von Moltke LL, et al. Relationship of in vitro data on drug metabolism to in vivo pharmacokinetics and drug interactions: implications for diazepam disposition in humans. J Clin Psychopharmacol 1996; 16(4): 267–72

    PubMed  CAS  Google Scholar 

  14. Schmid B, Bircher J, Preisig R, et al. Polymorphic dextromethorphan metabolism: co-segregation of oxidative O-demethylation with debrisoquin hydroxylation. Clin Pharmacol Ther 1985; 38: 618–24

    PubMed  CAS  Google Scholar 

  15. Chen ZR, Somogyi AA, Bochner F. Polymorphic O-demethylation of codeine. Lancet 1988; II: 914–5

    Google Scholar 

  16. Wu D, Otton SV, Sproule BA, et al. Inhibition of human cytochrome P450 2D6 (CYP2D6) by methadone. Br J Clin Pharmacol 1993; 35: 30–4

    PubMed  CAS  Google Scholar 

  17. von Moltke LL, Greenblatt DJ, Cotreau-Bibbo MM, et al. Inhibitors of alprazolam metabolism in vitro: effect of serotonin-reuptake-inhibitor antidepressants, ketoconazole and quinidine. Br J Clin Pharmacol 1994; 38: 23–31

    CAS  Google Scholar 

  18. Shader RI, von Moltke LL, Schmider J, et al. The clinician and drug interactions: an update. J Clin Psychopharmacol 1996; 16(3): 197–201

    PubMed  CAS  Google Scholar 

  19. Preskorn SH. Clinical pharmacology of selective serotonin reuptake inhibitors. 1st ed. Caddo (OK): Professional Communications Inc., 1996

    Google Scholar 

  20. van Harten J. Clinical pharmacokinetics of selective serotonin reuptake inhibitors. Clin Pharmacokinet 1993; 24(3): 203–20

    PubMed  Google Scholar 

  21. Preskorn SH. Clinically relevant pharmacology of selective serotonin reuptake inhibitors. Clin Pharmacokinet 1997; 32 Suppl. 1: 1–21

    PubMed  CAS  Google Scholar 

  22. Jeppesen U, Gram LF, Vistisen K, et al. Dose-dependent inhibition of CYP1A2, CYP2C19 and CYP2D6 by citalopram, fluoxetine, fluvoxamine and paroxetine. Eur J Clin Pharmacol 1996; 51: 73–8

    PubMed  CAS  Google Scholar 

  23. Sindrup SH, Brosen K, Hansen MGJ, et al. Pharmacokinetics of citalopram in relation to the sparteine and the mephenytoin oxidation polymorphisms. Ther Drug Monit 1993; 15(1): 11–7

    PubMed  CAS  Google Scholar 

  24. Altamura AC, Moro AR, Percudani M. Clinical pharmacokinetics of fluoxetine. Clin Pharmacokinet 1994; 26(3): 201–14

    PubMed  CAS  Google Scholar 

  25. Benfield P, Heel RC, Lewis SP. Fluoxetine: a review of its pharmacodynamic and pharmacokinetic properties, and therapeutic efficacy in depressive illness. Drugs 1986; 32: 481–508

    PubMed  CAS  Google Scholar 

  26. Fluvoxamine in perspective: a decade of experience. Chester (UK): Adis International, 1995

  27. Dechant KL, Clissold SP. Paroxetine: a review of its pharmacodynamic and pharmacokinetic properties, and therapeutic potential in depressive illness. Drugs 1991; 41(2): 225–53

    PubMed  CAS  Google Scholar 

  28. Murdoch D, McTavish D. Sertraline: a review of its pharmacodynamic and pharmacokinetic properties, and therapeutic potential in depression and obsessive-compulsive disorder. Drugs 1992; 44(4): 604–24

    PubMed  CAS  Google Scholar 

  29. Lemberger L, Rowe H, Carmichael R, et al. Fluoxetine, a selective serotonin uptake inhibitor. Clin Pharmacol Ther 1978; 23: 421–9

    PubMed  CAS  Google Scholar 

  30. Brøsen K, Skjelbo E. Fluoxetine and norfluoxetine are potent inhibitors of P450IID6: the source of the sparteine/debrisoquine oxidation polymorphism. Br J Clin Pharmacol 1991; 32: 136–7

    PubMed  Google Scholar 

  31. 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(3): 1278–83

    Google Scholar 

  32. Crewe HK, Lennard MS, Tucker GT, et al. The effect of selective serotonin re-uptake inhibitors on cytochrome P4502D6 (CYP2D6) activity in human liver microsomes. Br J Clin Pharmacol 1992; 34: 262–5

    PubMed  CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

  34. Otton SV, Ball SE, Cheung SW, et al. Venlafaxine oxidation in vitro is catalysed by CYP2D6. Br J Clin Pharmacol 1996; 41(2): 149–56

    PubMed  CAS  Google Scholar 

  35. Brøsen K, Skjelbo E, Rasmussen JGC, et al. Fluvoxamine is a potent inhibitor of cytochrome P4501A2. Biochemical Pharmacology 1993; 45(6): 1211–4

    PubMed  Google Scholar 

  36. von Moltke LL, Greenblatt DJ, Court MH, et al. Inhibition of alprazolam and desipramine hydroxylation in vitro by paroxetine and fluvoxamine: comparison with other selective serotonin reuptake inhibitor antidepressants. J Clin Psychopharmacol 1995; 15: 125–31

    Google Scholar 

  37. Kobayashi K, Yamamoto T, Chiba K, et al. The effects of selective serotonin reuptake inhibitors and their metabolites on S-mephenytoin 4′-hydroxylase activity in human liver microsomes. Br J Clin Pharmacol 1995; 40 Suppl. 5: 481–5

    PubMed  CAS  Google Scholar 

  38. Dubovsky SL, Thomas M. Psychotic depression: advances in conceptualization and treatment. Hosp Community Psychiatry 1992; 43: 1189–98

    PubMed  CAS  Google Scholar 

  39. Plasky P. Antidepressant usage in schizophrenia. Schizophr Bull 1991; 17: 649–57

    PubMed  CAS  Google Scholar 

  40. Dahl-Puustinen M, Liden A, Alm C, et al. Disposition of perphenazine is related to polymorphic debrisoquine hydroxylation in human beings. Clin Pharmacol Ther 1989; 46: 78–81

    PubMed  CAS  Google Scholar 

  41. von Bahr C, Movin G, Nordin C, et al. Plasma levels of thioridazine and metabolites are influenced by the debrisoquine hydroxylation phenotype. Clin Pharmacol Ther 1991; 49: 234–40

    Google Scholar 

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

    PubMed  CAS  Google Scholar 

  43. Huang ML, van Peer A, Woestenborghs R, et al. Pharmacokinetics of novel antipsychotic agent risperidone and the prolactin response in healthy subjects. Clin Pharmacol Ther 1993; 54: 257–68

    PubMed  CAS  Google Scholar 

  44. Jerling M, Dahl ML, Aberg-Wistedt A, et al. The CYP2D6 genotype predicts the oral clearance of the neuroleptic agents perphenazine and zuclopenthixol. Clin Pharmacol Ther 1996; 59: 423–8

    PubMed  CAS  Google Scholar 

  45. Bertilsson L, Carrillo JA, Dahl ML, et al. Clozapine disposition covaries with CYP1A2 activity determined by a caffeine test. Br J Clin Pharmacol 1994; 38: 471–3

    PubMed  CAS  Google Scholar 

  46. Syvalahti EK, Taiminen T, Saarijarvi S, et al. Citalopram causes no significant alterations in plasma neuroleptic levels in schizophrenic patients. J Int Med Res 1997; 25(1): 24–32

    PubMed  CAS  Google Scholar 

  47. Milne RJ, Goa KL. Citalopram: a review of its pharmacodynamic and pharmacokinetic properties and therapeutic potential in depressive illness. Drugs 1991; 41: 450–77

    PubMed  CAS  Google Scholar 

  48. Gram LF, Hansen MGJ, Sindrup SH, et al. Citalopram: interaction studies with levomepromazine, imipramine and lithium. Ther Drug Monit 1993; 15: 18–24

    PubMed  CAS  Google Scholar 

  49. Øyehaug E. Effect of phenothiazines on citalopram steady-state kinetics in psychiatric patients. Nor Pharm Acta 1982; 46: 37–46

    Google Scholar 

  50. Tyndale R, Kalow W, Inaba T. Oxidation of reduced haloperidol to haloperidol: involvement of human P450TID6 (sparteine/debrisoquine monoxygenase). Br J Clin Pharmacol 1991; 31: 655–60

    PubMed  CAS  Google Scholar 

  51. Nemeroff CB, DeVane CL, Pollock BG. Newer antidepressants and the cytochrome P450 system. Am J Psychiatry 1996; 153(3): 311–20

    PubMed  CAS  Google Scholar 

  52. Llerena A, Dahl ML, Ekqvist B, et al. Haloperidol disposition is dependent on the debrisquine hydroxylation phenotype: increased plasma levels of the reduced metabolite in poor metabolizers. Ther Drug Monit 1992; 14: 261–4

    PubMed  CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

  54. Nelson MR, Dunner DL. Treatment resistance in unipolar depression and other disorders: diagnostic concerns and treatment possibilities. Psychiatr Clin North Am 1993; 16(3): 541–66

    Google Scholar 

  55. Tate JL. Extrapyramidal symptoms in a patient taking haloperidol and fluoxetine [letter]. Am J Psychiatry 1989; 146: 399–400

    PubMed  CAS  Google Scholar 

  56. Bouchard RH, Pourcher E, Vincent P. Fluoxetine and extrapyramidal side effects [letter]. Am J Psychiatry 1989; 146: 1352–3

    Google Scholar 

  57. Brod TM. Fluoxetine and extrapyramidal side effects [letter]. Am J Psychiatry 1989; 146: 1353

    Google Scholar 

  58. Centorrino F, Baldessarini RJ, Kando J, et al. Serum concentrations of clozapine and its majormetabolites: effects of cotreatment with fluoxetine or valproate. Am J Psychiatry 1994; 151(1): 123–5

    PubMed  CAS  Google Scholar 

  59. Daniel DG, Randolph C, Jaskiw G, et al. Coadministration of fluvoxamine increases serum concentrations of haloperidol. J Clin Psychopharmacol 1994; 14(5): 340–3

    PubMed  CAS  Google Scholar 

  60. Jerling M, Lindstrom L, Bondesson U, et al. Fluvoxamine inhibition and carbamazepine induction of the metabolism of clozapine: evidence from a therapeutic drug monitoring service. Ther Drug Monit 1994; 16(4): 368–74

    PubMed  CAS  Google Scholar 

  61. Szegedi A, Wiesner J, Hiemke C. Improved efficacy and fewer side effects under clozapine treatment after addition of fluvoxamine. J Clin Psychopharmacol 1995; 15(2): 141–3

    PubMed  CAS  Google Scholar 

  62. Özdemir V, Naranjo CA, Herrmann N, et al. Paroxetine potentiates CNS side effects of perphenazine: contribution of cytochrome P450 2D6 inhibition in vivo. Clin Pharmacol Ther. In press

  63. Cooper SM, Jackson D, Loudon JM, et al. The psychomotor effects of paroxetine alone and in combination with haloperidol, amylobarbitone, oxazepam or alcohol. Acta Psychiatr Scand 1989; 80 Suppl. 350: 53–5

    Google Scholar 

  64. Williams SA, Wesnes K, Oliver SD, et al. Absence of effect of sertraline on time-based sensitization of cognitive impairment with haloperidol. J Clin Psychiatry 1996; 57 Suppl. 1: 7–11

    PubMed  CAS  Google Scholar 

  65. Weilberg JB, Rosenbaum JF, Meltzer-Brody S. Tricyclic augmentation of fluoxetine. Ann Clin Psychiatry 1991; 3: 209–14

    Google Scholar 

  66. Baettig D, Bondolfi G, Montaldi S, et al. Tricyclic antidepressant plasma levels after augmentation with citalopram: a case study. Eur J Clin Pharmacol 1993; 44: 403–5

    PubMed  CAS  Google Scholar 

  67. Taylor D. Selective serotonin reuptake inhibitors and tricyclic antidepressants in combination: interactions and therapeutic uses. Br J Psychiatry 1995; 167: 575–80

    PubMed  CAS  Google Scholar 

  68. Vaughan DA. Interaction of fluoxetine with tricyclic antidepressants [letter]. Am J Psychiatry 1988; 145: 1478

    PubMed  CAS  Google Scholar 

  69. Goodnick PJ. Influence of fluoxetine on plasma levels of desipramine. Am J Psychiatry 1989; 146: 552

    PubMed  CAS  Google Scholar 

  70. Bell IR, Cole JR. Fluoxetine induces elevation of desipramine levels and exacerbation of geriatric non-psychotic depression. J Clin Psychopharmacol 1988; 8: 447–8

    PubMed  CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

  72. 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 

  73. Preskorn SH, Beber JH, Faul JC, et al. Serious adverse effects of combining fluoxetine and tricyclic antidepressants. Am J Psychiatry 1990; 147: 532

    PubMed  CAS  Google Scholar 

  74. van Harten J. Overview of the pharmacokinetics of fluvoxamine. Clin Pharmacokinet 1995; 29 Suppl. 1: 1–9

    PubMed  Google Scholar 

  75. Maskall DD, Lam RW. Increased plasma concentration of imipramine following augmentation with fluvoxamine [letter]. Am J Psychiatry 1993; 150: 1566

    PubMed  CAS  Google Scholar 

  76. Spina E, Pollicino AM, Avenoso A, et al. Fluvoxamine-induced alterations in plasma concentrations of imipramine and desipramine in depressed patients. Int J Clin Pharmacol Res 1993; 13: 167–71

    PubMed  CAS  Google Scholar 

  77. 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

    PubMed  CAS  Google Scholar 

  78. Bertschy G, Vandel S, Vandel R, et al. Fluvoxamine-tricyclic antidepressant interaction: an accidental finding. Eur J Clin Pharmacol 1991; 40: 119–20

    PubMed  CAS  Google Scholar 

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

    PubMed  Google Scholar 

  80. Preskorn SH. Pharmacokinetics of antidepressants: why and how they are relevant to treatment. J Clin Psychiatry 1993; 54 Suppl. Sep: 14–34

    PubMed  Google Scholar 

  81. Alderman J, Preskorn SH, Greenblatt DJ, et al. Desipramine pharmacokinetics when coadministered with paroxetine or sertaline in extensive metabolizers. J Clin Psychopharmacol 1997; 17(4): 284–91

    PubMed  CAS  Google Scholar 

  82. Lydiard RB, Anton RF, Cunningham T. Interactions between sertraline and tricyclic antidepressants [letter]. Am J Psychiatry 1993; 150: 1125–6

    PubMed  CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

  84. Zussmann BD, Davie CC, Fowles SE, et al. Sertraline, like other SSRIs, is a significant inhibitor of desipramine metabolism in vivo [abstract]. Br J Clin Pharmacol 1995; 39: 550–1

    Google Scholar 

  85. Kurtz DL, Bergstrom RF, Goldberg MJ, et al. The effect of sertaline on the pharmacokinetics of desipramine and imipramine. Clin Pharmacol Ther 1998; 62: 145–56

    Google Scholar 

  86. Wright CE, Lasher-Sisson TA, Steenwyk RC, et al. A pharmacokinetic evaluation of the combined administration of triazolam and fluoxetine. Pharmacotherapy 1992; 12(2): 103–6

    PubMed  CAS  Google Scholar 

  87. Lasher TA, Fleishaker JC, Steenwyk RC, et al. Pharmacokinetic pharmacodynamic evaluation of the combined administration of alprazolam and fluoxetine. Psychopharmacology 1991; 104: 323–7

    PubMed  CAS  Google Scholar 

  88. Greenblatt DJ, Preskorn SH, Cotreau MM, et al. Fluoxetine impairs clearance of alprazolam but not of clonazepam. Clin Pharmacol Ther 1992; 52: 479–86

    PubMed  CAS  Google Scholar 

  89. 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

    PubMed  CAS  Google Scholar 

  90. van Harten J, Holland RL, Wesnes K. Influence of multiple-dose administration of fluvoxamine on the pharmacokinetics of the benzodiazepine bromazepam and lorazepam: a randomized, cross-over study [abstract]. Eur Neuropsychopharmacol 1992; 2: 381

    Google Scholar 

  91. Yasumori T, Nagata K, Yang S, et al. Cytochrome P450 mediated metabolism of diazepam in human and rat: involvement of human CYP2C in N-demethylation in the substrate concentration-dependent manner. Pharmacogenetics 1993; 3: 291–301

    PubMed  CAS  Google Scholar 

  92. Andersson T, Miners JO, Birkett DJ, et al. Diazepam metabolism by human liver microsomes is mediated by both S-mephenytion hydroxylase and CYP3A isoforms [abstract]. Clin Pharmacol Ther 1994; 55: 138

    Google Scholar 

  93. Perucca E, Gatti G, Cipolla G, et al. Inhibition of diazepam metabolism by fluvoxamine: a pharmacokinetic study in normal volunteers. Clin Pharmacol Ther 1994; 56: 471–6

    PubMed  CAS  Google Scholar 

  94. Ring BJ, Binkley SN, Roskos L, et al. Effect of fluoxetine, norfluoxetine, sertraline and desmethylsertraline on human CYP3A catalyzed 1′-hydroxy midazolam formation in vitro. J Pharmacol Exp Ther 1995; 275: 1131–5

    PubMed  CAS  Google Scholar 

  95. Lemberger L, Rowe H, Bosomworth JC, et al. The effect of fluoxetine on the pharmacokinetics and psychomotor responses of diazepam. Clin Pharmacol Ther 1988; 43: 412–9

    PubMed  CAS  Google Scholar 

  96. Moskowitz H, Burns M. The effects on performance of two antidepressants, alone and in combination with diazepam. Prog Neuropsychopharmacol Biol Psychiatry 1988; 12: 783–92

    PubMed  CAS  Google Scholar 

  97. Bannister SJ, Houser VP, Hulse JD, et al. Evaluation of the potential for interactions of paroxetine with diazepam, cimetidine, warfarin, and digoxin. Acta Psychiatr Scand 1989; 80 Suppl. 350: 102–6

    Google Scholar 

  98. Gardner MJ, Baris BA, Wilner KD, et al. Effect of sertraline on the pharmacokinetics and protein binding of diazepam in healthy volunteers. Clin Pharmacokinet 1997; 32 Suppl. 1: 43–9

    PubMed  CAS  Google Scholar 

  99. Pearson HJ. Interaction of fluoxetine with carbamazepine [letter]. J Clin Psychiatry 1990; 51(3): 126

    PubMed  CAS  Google Scholar 

  100. Grimsley SR, Jann MW, Carter JG, et al. Increased carbamazepine plasma concentrations after fluoxetine coadministration. Clin Pharmacol Ther 1991; 50: 10–5

    PubMed  CAS  Google Scholar 

  101. Spina E, Avenoso A, Pollocino AM, et al. Carbamazepine coadministration with fluoxetine or fluvoxamine. Ther Drug Monit 1993; 15: 247–50

    PubMed  CAS  Google Scholar 

  102. Fritze J, Unsorg B, Lanczik M. Interaction between carbamazepine and fluvoxamine. Acta Psychiatr Scand 1991; 84: 583–4

    PubMed  CAS  Google Scholar 

  103. Wagner W, Vause EW Fluvoxamine: a review of global drug-drug interaction data. Clin Pharmacokinet 1995; 29 Suppl. 1: 26–32

    PubMed  CAS  Google Scholar 

  104. Andersen BB, Mikkelsen M, Vesterager A, et al. No influence of the antidepressant paroxetine on carbamazepine, valproate and phenytoin. Epilepsy Res 1991; 10(2-3): 201

    PubMed  CAS  Google Scholar 

  105. Rapeport WG, Williams SA, Muirhead DC, et al. Absence of a sertaline-mediated effect on pharmacokinetics and pharmacodynamics of carbamazepine. J Clin Psychiatry 1996; 57 Suppl. 1: 20–3

    PubMed  CAS  Google Scholar 

  106. Caraco Y, Sheller J, Wood AJJ. Pharmacogenetic determination of the effects of codeine and prediction of drug interactions. J Pharmacol Exp Ther 1996; 278(3): 1165–74

    PubMed  CAS  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Claudio A. Naranjo.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sproule, B.A., Naranjo, C.A., Bremner, K.E. et al. Selective Serotonin Reuptake Inhibitors and CNS Drug Interactions. Clin Pharmacokinet 33, 454–471 (1997). https://doi.org/10.2165/00003088-199733060-00004

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00003088-199733060-00004

Keywords

Navigation