You are on page 1of 22

DRUG DISPOSITION Clin Pharmacokinet 1998 Apr; 34 (4): 281-302

0312-5963/98/0004-0281/$22.00/0

© Adis International Limited. All rights reserved.

Metabolism of the
Newer Antidepressants
An Overview of the Pharmacological and
Pharmacokinetic Implications
Silvio Caccia
Istituto di Ricerche Farmacologiche ‘Mario Negri’, Milan, Italy

Contents
Summary . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 281
1. Selective Serotonin Reuptake Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 283
2. Serotonin and Noradrenaline Reuptake Inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . 286
3. The Selective Noradrenaline Reuptake Inhibitor Reboxetine . . . . . . . . . . . . . . . . . . . . . . 288
4. The Monoamine Uptake Inhibitor and 5-HT 2 Receptor Antagonist Nefazodone . . . . . . . . . . . 290
5. Tianeptine, a Serotonin Reuptake Enhancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 292
6. The Noradrenergic and Specific Serotoninergic Agent Mirtazapine . . . . . . . . . . . . . . . . . 294
7. The Reversible Inhibitor of Monoamine Oxidase Moclobemide . . . . . . . . . . . . . . . . . . . . 294
8. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 296

Summary Several chemically unrelated agents has been developed and introduced in the
past decade, to supplement the earlier antidepressants. These include inhibitors
of the reuptake of serotonin [the selective serotonin reuptake inhibitors (SSRI)]
or noradrenaline (reboxetine) or both (milnacipran and venlafaxine), as well as
drugs with distinct neurochemical profiles such as mirtazapine, nefazodone,
moclobemide and tianeptine. Like the earlier drugs, these newer antidepressants
are almost totally biotransformed before excretion, except for milnacipran whose
clearance appears to be due equally to both urinary excretion and metabolism.
Sometimes – as in the case of moclobemide – up to 20 metabolites have been
identified in body fluids. In some cases, however, only a few metabolites have
been detected, and a substantial proportion of the dose remains unaccounted for
(e.g. fluoxetine and fluvoxamine).
Metabolism generally proceeds through sequential or parallel oxidative path-
ways. These may be affected to varying degrees by physiological and pathological
factors and those mediated by cytochrome P450 (CYP) 2D6 and CYP2C19
through genetic polymorphism. Some are influenced by chirality (e.g. the dealk-
ylation of citalopram and fluoxetine), although information on this aspect of
disposition is still lacking for other drugs existing as racemates (e.g. mirtazepine
and tianeptine) and milancipran, which is probably a mixture of 4 stereoisomers.
Others again are saturable within the therapeutic range of doses (e.g. some path-
ways of metabolism of fluoxetine, fluvoxamine, nefazodone, paroxetine and
venlafaxine). This may explain the individual variability with all these drugs
282 Caccia

which, from the pharmacokinetic point of view, is the same as with tricyclic
agents.
Our knowledge of the isoenzymes involved in the various oxidation pathways
and their relevance for potential drug interactions varies from a considerable
amount for most of the SSRI and nefazodone, to minimal for reboxetine and
tianeptine. This information is useful for predicting the pharmacokinetic interac-
tions mediated through inhibition of specific isoenzymes. This would be better
appreciated if the enzymatic mechanisms involved in the biotransformation of
the metabolite(s), and their role in drug interactions, were also known. This in-
formation is still lacking for some drugs, although metabolites may exhibit in
vitro inhibitory potencies of similar to (paroxetine and its M2 metabolite as in-
hibitors of CYP2D6) or even greater than that of the parent drug (norfluoxetine
is more potent than fluoxetine as an inhibitor of CYP3A3/4, and in view of the
longer half-life (t1⁄2) of the metabolite the potential for interactions may persist
for weeks after discontinuation of the parent drug).
While we do know something about the biological activity of the metabolites
of some of these drugs, we know very little about others. With few exceptions
this knowledge refers only to the major metabolite(s) and regards the main in
vitro effects as exerted by the parent drug. However, in vitro potency and selec-
tivity may not translate directly into in vivo, and either major or minor metabolites
may have characteristic in vitro and in vivo properties. For example, unlike the
parent drug some minor ring-opened metabolites of moclobemide have mono-
amine oxidase-B inhibitory activity in the rat, and the nefazodone metabolite
m-chlorophenyl-piperazine shows activity on 5-HT 2C receptors in rats and
humans.
Data on the brain-to-blood partition of metabolites compared with their parent
drug are available only in a few cases. They are still not known for the main
metabolites of fluvoxamine, milnacipran, mirtazapine, moclobemide, nefazo-
done, paroxetine, reboxetine and venlafaxine, despite the fact that total blood
concentrations do not always reflect the metabolite : parent drug ratio in brain.
Thus, in most cases, we do not really know what part hepatic metabolism plays
in the overall effect of the administered parent drug.

Supplementing tricyclic antidepressant deriva- to the date of introduction – have been described in
tives (TCA), monoamine oxidase inhibitors (MAOI) a number of reviews, specifying the characteristics
and second generation antidepressants,[1-4] a whole governing the frequency of drug administration
new generation of chemically and neuropharmaco- and the time to reach steady state, possibility of
logically unrelated antidepressant agents has been therapeutic drug monitoring, genetic polymor-
introduced and marketed over the past decade, in- phism of metabolism and potential for drug-drug
cluding several of the drugs listed in table I. While interactions.[7-10] The importance of the formation
most of these drugs offer new approaches to the of metabolites and their possible contribution to the
treatment of depressive disorders, their therapeutic final outcome has been underlined.[11-14] This is
equivalence is not yet apparent, although they are because most of these drugs are almost entirely
considered generally safer and better tolerated than eliminated from the body after biotransformation,
the older drugs.[5,6] often yielding active metabolites. These may be-
The pharmacokinetics of the older antidepres- have either similarly to or differently from the par-
sant drugs – the term ‘old’ obviously referring only ent drug as regards pharmacokinetics and pharma-

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
Metabolism of the Newer Antidepressants 283

codynamics, sometimes resulting in a situation teristics but all are extensively biotransformed be-
which is no different in principle from polytherapy. fore excretion, initially through oxidative path-
Although exhaustive reviews have been pub- ways. However, metabolism apparently has a va-
lished on the comparative clinical pharmacokinet- riety of effects on the pharmacological activity of
ics of some of the newer antidepressants,[15-19] data the parent drug (table II). Thus, fluvoxamine is
on the in vivo biotransformation of others and the metabolised by oxidation, oxidative deamination
pharmacological significance of their metabolites and hydrolysis to many metabolites, 9 of which
are generally scarce and fragmentary. Here we re- have been identified and constitute 65% of the uri-
view the metabolism of the newer antidepressants nary excretion products. Only 2 have been tested
with emphasis on metabolites that may contribute for their effect on monoamine uptake mechanisms,
to primary and secondary pharmacological actions and showed no or only weak inhibition of serotonin
of the parent drug. uptake (i.e the carboxyl acid derivative) in compar-
ison with the parent drug.[20,28]
1. Selective Serotonin The specific isoenzymes involved in fluvoxam-
Reuptake Inhibitors ine metabolism have not been identified but in vitro
and in vivo the drug potently inhibits the metabo-
The 5 selective serotonin reuptake inhibitors
lism of substrates for CYP1A2.[18,29-32] The asso-
(SSRIs) currently available (citalopram, fluvox-
ciation between fluvoxamine metabolism and the
amine, fluoxetine, paroxetine and sertraline) differ
activity of CYP1A2 is supported by the lower
in chemical structure and pharmacokinetic charac-
fluvoxamine plasma concentrations in smokers
than nonsmokers,[33] smoking being known to in-
Table I. The newer antidepressant agents and some pharmacolog- duce the metabolism of drugs catalysed by this
ically related compounds isoenzyme. However, fluvoxamine also inhibits
Mechanism of action Drug the metabolism of typical substrates of CYP2C19,
Selective serotonin reuptake inhibition Citalopram
Fluoxetine
CYP3A4, CYP2D6 and CYP1A1.[18,29-31,34-37] Flu-
Fluvoxamine voxamine exhibits nonlinear kinetics which are not
Paroxetine due to Michaelis-Mentem saturation of a single
Sertraline
metabolic pathway, but to a complex metabolic
Serotonin and noradrenaline reuptake Duloxetine
inhibition Milnacipran picture involving multiple parallel pathways.
Venlafaxine The metabolic clearance of fluvoxamine is af-
Selective noradrenaline reuptake inhibition Reboxetine fected by gender (with higher plasma concentra-
Nisoxetine
Teniloxazine tions in women with depression than in men with
Tomoxetine depression)[38] and liver disease (cirrhosis). It does
Serotonin uptake and 5-HT2 receptor Nefazodone not appear to change in relation to age and renal
inhibition Trazodone
function after single doses.[17,18,20,28] However, all
Serotonin reuptake enhancement Tianeptine
5-HT1A receptor-partial agonism Buspirone these studies have focused on the parent drug.
Gepirone The main metabolites of paroxetine are also
Ipsapirone reported to have very weak in vitro inhibitory ac-
Selective cAMP phosphodiesterase Rolipram
inhibition
tivity on serotonin (and catecholamine) uptake
α2-Adrenergic auto- and hetero-receptor Mirtazapine mechanisms. These include an unstable catechol
blockade intermediate and its methylated and glucuronide-
Reversible and selective inhibition of Brofaromine and sulphate-conjugated derivatives.[21,29] Paroxet-
monoamine oxidase type A Cimoxatone
Moclobemide ine metabolism is partly under the pharmacoge-
Toloxatone netic control of CYP2D6[34,40-43] but another en-
Abbreviation: cAMP = cyclic adenosine monophosphate. zyme with lower affinity for the drug may also be

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
284 Caccia

Table II. Comparative in vitro potencies of selective uptake inhibitors and their main active metabolites, with mean metabolite-to-parent drug
ratios at steady state
Drug Main active Serotonin uptake Main enzyme inhibited Steady-state
metabolite inhibitorsa CYP2D6 CYP3A3/4 CYP1A2 CYP2C19 metabolic ratio
Citalopram +++ + 0.3-0.4b
Norcitalopram ++ ++
Fluoxetine +++ +++ + ++
Norfluoxetine ++/+++c +++ ++ ++ 0.6-1.7b
Fluvoxamine +++ + ++ +++ +++
Carboxy acid + NA NA NA NA NA
Paroxetine +++ +++
M2 metabolite + +++ NA
Sertraline +++ +/++
Norsertraline ++ +/++ 1.6-2.1b
a Only compared with parent drug.[20-26]
b Mean values in different populations.[18,27]
c The R-isomer is weaker than the parent drug isomers or its own S-isomer.
Abbreviation and symbols: NA = not available; +++ = substantial; ++ = moderate; + = weak.

involved.[42] This enzyme has not yet been identi- which retains weak serotonin reuptake inhibiting
fied but it may be inhibited by cimetidine, decline activity and reaches lower concentrations than the
in efficiency with age and be induced by some anti- parent drug and its main metabolite in human
convulsants.[44] plasma.[22] All the parent compounds and their nor-
At therapeutic doses paroxetine causes substa- derivatives are further biotransformed to a number
tial inhibition of CYP2D6 activity in vivo.[15-18,29,30,45] of metabolites, many of which remain to be identi-
One of its metabolites (designated M2) may con- fied.[22,46] Norcitalopram may amount to about
tribute to these actions, being a potent inhibitor of 40% of the mean parent drug steady-state plasma
this isoenzyme in vitro.[34] Paroxetine also inhibits concentration (table II), with large interindividual
its own clearance on repeated administration, al- variability, and its elimination t1⁄2 (t1⁄2β) is more than
though only in extensive, not in poor, metabolisers twice that of the parent drug.[17,18,47] However, it
of sparteine. The lack of effect on the clearance of has poor blood-brain barrier penetration.[22] In rats
desipramine in poor metabolisers confirms that this given repeated citalopram 10 mg/kg interperitone-
drug is a selective inhibitor of CYP2D6, which is ally twice daily for 14 days, the plasma concentra-
lacking from the liver of poor metabolisers.[42,43] tions were comparable with those of the parent
Clearance of the parent drug is influenced by aging, drug but concentrations in the brain were less than
severe renal impairment and hepatic disease, with 10% (within the first hours after administration),
higher plasma concentrations and slower elimina- suggesting that it was unlikely to contribute signif-
tion than in healthy volunteers,[21,39] but the phar- icantly to the pharmacological activity in this spe-
macokinetics of its main metabolites are not known cies (Caccia S., unpublished results).
for any of these populations. The formation of norcitalopram is at least partly
Citalopram, fluoxetine and sertraline are all under the genetic control of CYP2C19, being sig-
initially demethylated to their nor-derivatives nificantly lower in Caucasians who were poor me-
demethylcitalopram, norfluoxetine and norsertral- tabolisers of mephenytoin than in those who were
ine, which are also SSRI (see table II). Unlike the extensive metabolisers.[47,48] The subsequent step
other nor-derivatives, demethylcitalopram is fur- to di-demethylcitalopram appears instead to be under
ther N-demethylated to di-demethylcitalopram, the genetic control of CYP2D6. Levomepromazine,

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
Metabolism of the Newer Antidepressants 285

a potent inhibitor of this enzyme, raises the steady- metabolised faster than S-fluoxetine in vitro and in
state plasma concentrations of norcitalopram dur- vivo in humans.[54,55]
ing citalopram administration (see Baumann[18] for a Norfluoxetine retains the pharmacological
review). Norcitalopram may mediate the mild inter- properties of fluoxetine, and depending on the
action potential of citalopram with drugs metabo- species, experimental test and conditions may con-
lised by the CYP2D6 isoenzyme, being one order tribute to, or even account for, the actions seen with
of magnitude more potent as an inhibitor than the the parent compound.[25,56-59] In patients with de-
parent drug (see also table II).[17,30] pression it achieves steady-state concentrations
Using Japanese human liver microsomes and comparable with, or even higher than, those of the
recombinant human CYP isoforms, Kobayashi et parent compound, accounting for a substantial
al.[49] found that CYP2C19 was only a minor form part of the therapeutic and adverse effects of the
and CYP3A4 the major isoenzyme involved in the drug.[16-18,60] Its formation is partly under the genetic
first step of citalopram metabolism. The discrep- control of CYP2D6, being higher in extensive met-
ancy between the in vivo and in vitro findings has abolisers than in poor metabolisers of debrisoquin
been tentatively explained by ethnic differences in and dextromethorphan.[61]
the frequency of individuals heterozygous for the Fluoxetine interacts in vitro with known
extensive metabolisers genotype. Citalopram, CYP2D6 substrates.[34,40,62] In this respect the S-
however, is a racemic compound, whose disposi- isomers of fluoxetine and norfluoxetine have sim-
tion is affected by stereoselectivity, with the more ilar potency but are about 5 times more potent than
the R-isomers in vitro in human microsomes.
potent SSRI S-(+)-citalopram achieving lower
Norfluoxetine, however, is more potent than fluo-
steady-state concentrations than the R-(–)-enantio-
xetine as an inhibitor of CYP3A3/4 and, because
mer,[50] with possible age-related changes in
of its unusually long t 1⁄2β, the risk of drug-drug
stereoselective metabolism of the S-isomer.[51]
interactions persists, for a long time after fluoxet-
In vitro studies with human microsomes show
ine therapy is discontinued. The pharmacokinetics
that both CYP2C19 and CYP3A4 are involved in
and clinically relevant interactions between fluo-
the preferential N-demethylation of S-(+)-citalo-
xetine and coadministered drugs have been pre-
pram.[52] Fluvoxamine, possibly by inhibiting sented in a number of recent reviews.[16-18,29-31,63-67]
these enzymes, preferentially raises the concentra- Unlike fluoxetine, CYP2D6 does not seem to
tion of the S-isomer, changing the S/R ratio in play an important role in the demethylation of
dextromethorphan/mephenytoin phenotyped pa- sertraline,[61] which is possibly dependent on
tients given repeated doses of citalopram.[53] CYP3A isoenzyme.[18,30,44] However, sertraline
CYP2D6 is also involved in this step but with the does inhibit CYP2D6 in vitro, although less po-
opposite stereoselectivity.[52] tently than fluoxetine and paroxetine.[29,30,63-67]
Fluoxetine and norfluoxetine are also chiral Dose-dependent increases in plasma concentra-
compounds, existing as R-(–)- and S-(+)-isomers. tions of desipramine have been observed after its
While there are no major differences in pharmaco- addition to therapy (50 to 150 mg/day), although
logical potency between R- and S-fluoxetine,[23] the interaction is weaker than with fluoxetine and
the S-isomer of norfluoxetine is consistently more paroxetine at the usually effective therapeutic
effective than the R-isomer as an inhibitor of sero- doses.[17,18,29,44,67-70] Sertraline 150mg as a single
tonin uptake in vitro and ex vivo in animals, and or repeated dose also reduces the clearance of
is possibly responsible for the long-lasting seroto- imipramine.[70] Norsertraline is approximately
nin uptake inhibition in vivo.[24] Stereoselectivity equipotent to sertraline with respect to inhibition
in fluoxetine demethylation and/or subsequent of CYP2D6 in vitro[30,44] but reaches higher
pathways has been observed, the R-isomer being steady-state plasma concentrations and is elimi-

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
286 Caccia

nated more slowly than the parent compound (t1⁄2β Compared with young (between the ages of 18
3 to 5 days compared with 1 to 2 days for the parent and 65 years old) volunteers, exposure to both the
compound, with some gender and age differ- parent drug and its nor-metabolite is generally
ences).[17,27] Norsertraline possibly plays a deci- higher in elderly (between the ages of 65 and 75
sive role in the in vivo inhibitory effect of the parent years old) individuals for citalopram, fluoxetine
drug.[31,66,68,71] Sertraline and norsertraline are weaker and sertraline.[6,17-18,44] For sertraline there is also
inhibitors of CYP3A3/4 isoenzymes, and evidence an age-gender interaction, plasma concentrations
suggests that clinically significant interactions of the parent drug and its nor-derivative in young
with CYP3A substrates are unlikely.[72] men are lower than in young women or the el-
As an in vitro inhibitor of neuronal serotonin derly.[27]
uptake, norsertraline is less potent than the parent For all drugs the t1⁄2β is doubled in patients with
drug.[26] However, the in vivo activity of the nor- hepatic cirrhosis compared with healthy volun-
metabolite and its contribution to the parent drug’s teers, so these agents should be used cautiously in
effects is less clear. Thus, norsertraline was inac- patients with liver disease.[17,44,76] Citalopram
tive in the mouse Porsolt test, a model of depres- clearance is reduced and the t 1⁄2β is increased in
sion.[26] At doses of 1 to 10 mg/kg the nor-metabolite patients with renal impairment.[17] Although full
did not increase extracellular serotonin or inhibit doses of fluoxetine and sertraline have been used
the spontaneous firing rate of dorsal raphe neurons in renally impaired patients without marked phar-
in rat, 2 parameters affected dose-dependently by macokinetic changes,[26,58] caution is still advis-
sertraline (1 to 10 mg/kg administered subcutane- able because these studies have been conducted
ously), and possibly other SSRI as a consequence after single doses (and fluoxetine at least shows
of the block of serotonin reuptake.[73] However, at nonlinear behaviour with repeated dose adminis-
tration, which may be magnified by renal impair-
higher doses, norsertraline (32 mg/kg administered
ment) and the multiple-dose pharmacokinetics of
intraperitoneally) lowers brain 5-hydroxyindole-
their more polar metabolite(s) is poorly known in
acetic acid (5-HIAA) in rats, as do all SSRIs.
this population.[44]
Norsertraline dose-dependently antagonised p-
chloroamphetamine-induced serotonin depletion,
although less potently than the parent compound in 2. Serotonin and Noradrenaline
the rat [intraperitoneal the dose causing 50% antag- Reuptake Inhibitors
onism (ED50), 20 and 1 mg/kg] but almost as po- The recently introduced serotonin and nor-
tently as sertraline in mice (ED50, 6 and 4 mg/kg); adrenaline reuptake inhibitors, milnacipran and
in this species the metabolite brain concentrations venlafaxine, differ structurally from selective in-
after sertraline were comparable with those after hibitors of serotonin or noradrenaline uptake. From
norsertraline.[74] the point of view of the putative mechanism of ac-
The nor-metabolite fully retains the ability of tion, these drugs resemble the tertiary amine tri-
sertraline to cause anorexia in rodents, a property cyclic antidepressant agents (TCA), since they af-
shared by other SSRI and in general by agents that fect both serotonin and noradrenaline reuptake.
increase serotonergic transmission.[75] Milnacipran almost equipotently inhibits these
Taken together these findings suggest that mechanisms without significantly affecting dopa-
norsertraline may play a role in the pharmacologi- mine transporters.[77] Venlafaxine inhibits synapto-
cal actions of the parent drug, though this depends somal serotonin more than noradrenaline uptake
on the in vivo effect studied and experimental con- and slightly inhibits dopamine uptake.[78] A com-
ditions and animal model considered. However, it parative review of the neuropharmacological
is still not clear to what extent norsertraline con- profile of these and pharmacologically related
tributes to the therapeutic action of sertraline. compounds such as duloxetine (whose develop-

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
Metabolism of the Newer Antidepressants 287

Table III. Effects of liver and renal impairment on the disposition of milnacipran and venlafaxine. The dose of milnacipran and venlafaxine
was 50mg
Drug Main metabolic pathway Disease Outcomea
Milnacipran Glucuronide conjugation Hepatic No changes in disposition
Renalb 2- or 4-fold increase in parent drug and conjugate metabolite
levels (and t1⁄2)
Venlafaxine Oxidative O-demethylation Hepatic CL is reduced by 50%
Renalc 25 to 50% reduction in CL with increase in t1⁄2 for the drug
and its active metabolite
a Compared with age-matched healthy individuals.
b CLCR values ranged from 0.6 to 5.1 L/h.[76]
c CLCR ranged from 1.8 to 4.2 L/h for the patients with mild impairment to 0.6 to 1.8 L/h for those with moderate impairment.[81,82]
Abbreviations: CL = total body clearance; CLCR = creatinine clearance.

ment as an antidepressant has been withdrawn) has in patients with renal impairment (9 to 25 hours
been published recently.[79] compared with about 8 hours for healthy individu-
Milnacipran, a 1-aryl-2-(aminomethyl)cyclo- als) who, therefore, need some adjustment in the
propane-carboxylic acid derivative, shares some dosage and frequency of administration.[76]
structural similarities with the ‘old’ monoamine Slightly higher milnacipran plasma concentra-
oxidase (MAO) inhibitor tranylcypromine.[80] The tions (approximately 20%) and prolonged t1⁄2β
disposition of this drug, which very likely is a mix- have been observed in elderly patients given a sin-
ture of stereoisomers, has largely been studied us- gle oral dose of milnacipran 50mg compared with
ing the radiolabelled compound and seems to differ healthy individuals, this is probably a consequence
from most other antidepressants because its clear- of some loss of renal function in the elderly. Be-
ance is due equally to urinary excretion and meta- cause the differences are small the manufacturer
bolism. About 50 to 60% of the dose is recovered does not recommend dosage adjustment in these
unchanged in urine and most of the remainder as cases, except for those with impaired renal func-
its glucuronide conjugate (approximately 20%), its tion.[76] No information is available for its metabo-
N-dealkylated derivative and the corresponding lites in this population.
glucuronide conjugate after single doses in healthy Levomepromazine (a CYP2D6 inhibitor) slightly
volunteers.[76] raises milnacipran steady-state concentrations (by
In plasma, most of the radioactivity results from approximately 20%) without appreciably affecting
milnacipran and its conjugate, N-dealkylmilnaci- its metabolic profile whereas carbamazepine,
pran reaching only very low concentrations. No which is metabolised by CYP3A3/4, stimulates
data are available for concentrations of milnaci- milnacipran clearance, the plasma concentrations
pran and its norderivative in tissues, including the of N-dealkyl milnacipran rising by 50%.[76]
brain, relative to blood. However, N-dealkylmil- Venlafaxine is a phenethylamine bicyclic de-
nacipran is reported to be inactive in vivo in animal rivative, which exists as a racemic mixture of the
models predictive of antidepressant activity.[80] (R)- and (S)-enantiomers, both of which inhibit
Liver insufficiency (cirrhosis) does not change monoamine uptake in rat synaptosomes.[78] How-
the pharmacokinetics of milnacipran 50mg either ever, (–)-venlafaxine inhibits reuptake of serotonin
orally or intravenously). Renal impairment pro- and noradrenaline, whereas the (+)-isomer primar-
gressively reduces the total clearance, plasma con- ily inhibits serotonin reuptake. The pharmaco-
centrations of the parent drug and its glucuronide kinetics of the single isomers have been studied
conjugate rising 2- to 4-fold after a single oral dose after oral doses of the racemate and were stereo-
(table III). The mean t1⁄2β of milnacipran is longer selective in rats but not in dogs or humans.[83]

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
288 Caccia

O-Demethylation of (racemic) venlafaxine to This metabolite is formed more slowly, depend-


O-desmethyl-venlafaxine is the predominant route ing to some extent on renal function in renally im-
of metabolism in humans, with about 60% of the paired individuals and patients undergoing haemo-
dose appearing in the urine as such or its glucuron- dialysis. The apparent total clearance of both the
ide conjugate. Other minor routes are N-demethyl- drug and its main metabolite is reduced by 55% in
ation of the alkylamino side chain to N-desmethyl- end-stage renal failure, and the t1⁄2β is more than
venlafaxine and the loss of both the O-methyl and doubled for both compounds.[82] Similar decreases
1 of the N-methyl groups to form N,O-desmethyl- in clearance have been observed in patients with
venlafaxine.[84] Among these and other metabolites cirrhosis (table III). All these patients required a
identified in animals, the O-demethyl derivative re- reduction in dosage of 50%.[81]
tains pharmacological activity in vitro (monoamine
uptake blockade) and in vivo (reversal of reserpine 3. The Selective Noradrenaline
hypothermia, induction of pineal β-adrenergic sub- Reuptake Inhibitor Reboxetine
sensitivity) comparable with the parent drug in Reboxetine is a recently introduced drug. Chem-
rats. The N-desmethyl-derivative is also active in ically it is (RS)-2-[(RS)-α-(2-ethoxyphenoxy)]-
these in vivo tests but is a weaker inhibitor of sero- benzylmorpholine methan-sulphonate (fig. 1) and
tonin and noradrenaline uptake than venlafax- shares some chemical similarities with viloxazine,
ine.[85] and to a lesser extent fluoxetine and structurally
The O-demethylation of venlafaxine is partially related compounds. It exists as a racemic mixture
mediated by CYP2D6, whereas N-demethylation of the S,S-(+)- and R,R(–)-enantiomers[93] both of
is probably catalysed by CYP3A3/4 isoenzymes.[86,87] which inhibit noradrenaline uptake in vitro in rat
Venlafaxine, however is a less potent inhibitor of hypothalamic synaptosomes and prevent reser-
CYP2D6 enzymes in vitro than SSRIs.[88,89] Al- pine-induced blepharospasm in vivo in rodents, the
though at the usual therapeutic doses venlafaxine S,S- approximately 20 times as potently as the R,R-
should not greatly affect the biotransformation of isomer.[94,95]
drugs transformed by CYP2D6, these might affect After single oral doses of reboxetine in mice and
the O-demethylation of venlafaxine and/or one of rats the mean (R,R) : (S,S) brain concentration ra-
its isomers. tios [in terms of the peak plasma drug concentra-
The parent drug and its O- and N-desmethyl de- tion (Cmax) and area under the concentration-time
rivatives are weak inhibitors of CYP3A isoforms.[87-89] curve (AUC)] are about 2 and 4 respectively, sug-
gesting that the pharmacological effects of reboxet-
Accordingly, venlafaxine has no appreciable ef-
ine in rodents are mainly due to the more active
fects on the pharmacokinetics of diazepam[90]
(S,S)-enantiomer. The mean brain : plasma AUC
whose main routes of metabolism may partly in-
ratio for both isomers is higher in mice than in
volve CYP3A3/4.[91] Venlafaxine and metabolites
rats,[95] confirming that differences may exist in the
are also weak inhibitors of CYP1A2 activity in brain uptake of centrally acting drugs.[96,97] In hu-
vitro.[87,89,92] man plasma the (R,R) : (S,S) isomeric ratio is sim-
After single and repeated doses in young and ilar (about 2) to that in mouse plasma after single
elderly volunteers, O-demethyl venlafaxine ap- oral doses of each isomer or the racemate. In
pears rapidly in plasma and far exceeds the plasma healthy individuals the mean t1⁄2β of the 2 isomers
concentrations of the parent drug. The mean t 1⁄2β of is comparable (about 13 hours after reboxetine
the metabolite (9 to 11 hours) is about twice that of 4mg or 2mg of each isomer).[98]
the parent drug (2 to 7 hours in healthy individu- The metabolism of oral reboxetine and its re-
als,[81] and a sustained release formulation is now covery in excreta have been studied in humans and
on the market in several countries). animals using the radiolabelled drug. Reboxetine

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
Metabolism of the Newer Antidepressants 289

CI O
NH2

O= CH2-CH3
N
CH2-CH3
NH
CH3
Fluoxetine Milnacipran

CH3
N
H3 C O CH3

CI O
HO

O O

NH CH 3

Reboxetine Venlafaxine

Fig. 1. Chemical structures of some monoamine reuptake inhibitory antidepressants.

is extensively biotransformed before excretion in abolites on the activity of these drug-metabolising


all species with no, or only trace amounts of, un- enzymes are still not known.
changed drug excreted with urine. The main met- The total oral clearance of racemic reboxetine
abolic routes seem to include hydroxylation of the is low and mainly because of nonrenal mechanisms;
ethoxyphenoxy ring, oxidative O-dealkylation, it may be markedly decreased in very old people
oxidation of the morpholine ring and glucuronide (between 66 and 98 years old) compared with
conjugation of the phase I metabolites, with differ- young individuals (see also table IV).[98] Oral
ences between species.[99] reboxetine clearance is unaltered after multiple ad-
Information is limited on the enzymatic mech- ministration in both young and elderly individuals.
anisms that govern these metabolic pathways in However, exposure to reboxetine is higher and
much more variable in elderly patients with de-
humans which, however, do not seem to involve
pression than in younger volunteers, as assessed by
CYP2D6 because quinidine, a potent inhibitor of this
the steady-state AUC values. A reduction in the
enzyme, does not affect the disposition of reboxet-
total daily dosage from 8 to 6mg is recommended
ine isomers. Reboxetine 2mg twice daily given to
for elderly patients.[98]
healthy volunteers for 5.5 days does not appear to The mean plasma Cmax of reboxetine is similar
significantly alter the CYP3A3/4-mediated 6β- in healthy volunteers and in patients with alcoholic
hydroxylation of endogenous cortisol.[100] At the liver disease after a single 4mg dose. The mean
same dose it does not change the pharmacokinetics AUC is slightly higher (24 to 36%, depending on
of lorazepam,[98] a short-acting benzodiazepine the degree of impairment) and mean is t1⁄2β longer
mainly eliminated by conjugation. Reboxetine is than normal (21 to 31 hours vs 12 to 14 hours) in
reported to have negligible activity as an inhibitor those patients with alcoholic liver disease.[98,100,101]
of CYP 1A2, 2C9, 2E1 and 3A4 isoenzymes in vitro Systemic exposure to reboxetine (and renal
in human microsomes.[98] The effects of its met- clearance) is progressively affected by renal im-

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
290 Caccia

Table IV. The clearance of reboxetine in young and elderly individuals


Age (years) Gender and number CLpo (L/h) CLR (L/h)
20-39a 54m 1.18-2.42 0.126-0.336
66-98 4m, 8f 0.39-0.63 0.006-0.054
a Data are from four separate studies; reboxetine dose ranged from 1.98 to 4.15mg, as free base.[98]
Abbreviations: CLpo = total oral clearance; CLR = renal clearance; f = female; m = male.

pairment, with mean AUC and renal clearance adrenaline reuptake inhibiting properties, which
dropping with the increasing severity of the dis- together may modulate serotoninergic neurotrans-
ease. A lower daily dosage is recommended in mission through postsynaptic receptors, particu-
patients with severe renal impairment.[98] larly 5-HT1A receptors.[102]
Although no detailed pharmacokinetic studies Nefazodone too is extensively biotransformed
of the main reboxetine metabolites are available in in the liver, with the formation of 4 main met-
these populations, in the plasma of healthy volun- abolites and several minor ones.[103] The main
teers unchanged reboxetine mostly appears to re- metabolites are hydroxynefazodone, p-hydroxy-
flect the circulating radioactivity (70% in terms of nefazodone, a triazoledione derivative and m-chloro-
AUC).[101] phenyl-piperazine (mCPP). This latter results from
cleavage of the phenyl-piperazine side chain and is
4. The Monoamine Uptake Inhibitor and a metabolite common to trazodone[104] and struc-
5-HT2 Receptor Antagonist Nefazodone turally related compounds.[105] Many other cen-
This phenoxyethyl triazoline phenylpiperazine trally acting drugs containing an arylpiperazine
derivative (fig. 2) is structurally related to side chain, including the potential antidepressants
trazodone. It has potent antagonistic effects on 5- buspirone, ipsapirone and gepirone,[106,107] yield
HT2A receptors and moderate serotonin and nor- 1-arylpiperazines during biotransformation in hu-

CI C-NH-CH2-CH2-N O
N
N O

N
CH3

Mirtazapine Moclobemide

C 2H5
CH3
N
CI SO2 N
N N
OCH2CH2 (CH2)3 N N

O CH
CI
NH (CH2 )6 COONa

Nefazodone Tianeptine
Fig. 2. Chemical structures of some antidepressants with neurochemical mechanisms other than monoamine reuptake inhibition.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
Metabolism of the Newer Antidepressants 291

mans and/or animals. These metabolites concentrate etine, the persistence of the inhibitory effect pos-
in brain tissues contributing to and, or accounting sibly being related to the long half-life of fluoxet-
for, a substantial part of the central effects of the ine, and particularly norfluoxetine.[111] Likewise
parent compound.[105,107] the addition of the CYP2D6 inhibitor thioridazine
The metabolism of nefazodone is at least partly 40 mg/day to trazodone 150 to 300 mg/day raises
mediated by CYP3A4.[102,108,109] The drug and its the plasma concentrations of mCPP by about
metabolites hydroxynefazodone and p-hydroxy- 50%.[112]
nefazodone inhibit this isoenzyme in vitro[108] and Hydroxynefazodone shows affinity for 5-HT2A
impair the in vivo clearance of concomitantly ad- receptors and serotonin uptake sites comparable
ministered carbamazepine, cyclosporin and triazolo- with nefazodone in rat cortex. The triazoledione
benzodiazepines (i.e alprazolam and triazolam) derivative is much less active than the parent com-
which depend on 3A4 activity for their metabo- pound – or even inactive – in these in vitro systems,
lism.[109] Nefazodone also impairs its clearance by whereas no information is available about p-
increasing the single dose and during repeated ad- hydroxynefazodone.[102]
ministration (see table V).[109] mCPP displays potency similar to nefazodone
The pharmacokinetics of nefazodone and for serotonin reuptake inhibition and low affinity
hydroxynefadozone do not differ in extensive or for 5-HT2A receptors.[102] However, this well
poor metabolisers of dextromethorphan, and in known serotoninergic agent causes a number of
vitro nefazodone and its main metabolite are weak behavioural and endocrinological changes in ani-
inhibitors of dextromethorphan 2D6-mediated O- mals and humans. Using selective agonists, partial
demethylation. However, the steady-state plasma antagonists and antagonists for serotonin receptor
Cmax and AUC of mCPP were substantially higher subtypes and families, some pharmacological ef-
in poor metabolisers than in extensive metabolisers fects of mCPP, such as the increase in plasma
(table V), and its t1⁄2β increases from 6.1 hours in prolactin and other hormones, oral dyskinesias and
extensive metabolisers to 16.4 hours in poor meta- hyperthermia, reduction of food intake and anxiety-
bolisers. This suggests that the subsequent metabo- like behaviours, have been attributed to its action
lism of mCPP, which involves ring hydroxylation on post-synaptic 5-HT2C receptors, where it acts as
before conjugation and urinary excretion, is medi- a partial agonist.[113,114]
ated by CYP2D6.[110] Therefore, drugs that inhibit In humans mCPP serves as an interesting probe
this enzyme might reduce the clearance and raise of serotoninergic mechanisms that may modulate
the concentrations of mCPP. This was observed anxiety. Given acutely to healthy individuals and
when nefazodone was co-administered with fluox- patients with panic or obsessive-compulsive disor-

Table V. Plasma area under the concentration-time curve (AUC) of nefazodone and its active metabolites after single and repeated doses
in extensive (EM) and poor metabolisers (PM) of dextromethorphan[110]
Compound Dosage Plasma AUC (μmol/L • h)
EM PM
Nefazodone Single dose 2.60 ± 0.77 2.45 ± 0.99
Steady state 13.57 ± 3.64 15.85 ± 1.77
Hydroxy-nefazodone Single dose 0.72 ± 0.30 (0.28) 0.80 ± 0.44 (0.31)
Steady state 4.53 ± 1.04 (0.33) 5.61 ± 2.71 (0.35)
m-Chlorophenyl-piperazine Single dose 2.09 ± 1.77 (0.80) 8.34 ± 1.43 (4.00)b
Steady state 1.97 ± 1.98 (0.15) 8.66 ± 3.04 (0.55)b
a Plasma AUC after a single dose (200mg) and over the dosage interval (AUC0-12h) at steady state (200mg twice daily). The metabolite-
to-parent drug ratios are shown in parentheses.
b Statistically significant.[110]

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
292 Caccia

ders it produces manifestations of anxiety and ent in all populations. Thus in different individuals
changes in many physiological functions, although there may be a different interplay among the neuro-
with repeated administration tolerance rapidly chemical mechanisms characteristic of each com-
arises to these effects.[115] pound, with possible differences in antidepressant
In healthy volunteers the hydroxynefazodone : activity and adverse effects.
nefazodone ratio in plasma (metabolic ratio on a
g/L basis) averages 0.3 to 0.4 regardless of the dose
5. Tianeptine, a Serotonin
and schedule of treatment, supporting pharmaco-
Reuptake Enhancer
logical findings that this metabolite may contribute
to the final outcome.[102,109,116]
Unlike antidepressants which facilitate post-
Plasma concentrations of nefazodone and hydr-
synaptic effects of central neurotransmitters by
oxynefazodone, in terms of Cmax and AUC, are
blocking their reuptake (i.e. the drugs mentioned
higher in the elderly than in younger volunteers
above) or by inhibiting their catabolism (i.e. MOAIs),
after single and repeated doses, and in elderly
tianeptine increases presynaptic serotonin reuptake,
women than in elderly men after repeated doses,
thereby reducing synaptic availability of the neuro-
but the metabolic ratio remains relatively constant,
transmitter.[120,121] This original neurochemical ac-
although with wide interindividual variability.[116-118]
tion has been observed ex vivo in rat cortical and
The mean Cmax and AUC of nefazodone and hydr- hippocampal synaptosomes and in rat and human
oxynefazodone are also significantly higher and platelets after single and repeated doses of tianept-
the t1⁄2 longer in patients with liver impairment than ine. Possible explanations of how this action is
in healthy individuals after 50 to 100mg twice involved in its antidepressant activity were reviewed
daily, these patients need a lower daily nefazodone by Ansseau[122] and, more recently, by Wilde and
dosage.[117,119] Benfield.[123]
The mCPP : nefazodone ratio tends to decrease Chemically it is a modified tricylic [i.e. the so-
with higher doses and longer duration of treatment, dium salt of 3-[(3-chloro-6-methyl-5,5-dioxo-6,11-
indicating that the formation of this metabolite is dihydrodibenzo[c,f(1,2-thiazepin)]11-yl)amino]-
saturable.[102-117] At steady-state, exposure to mCPP heptanoic acid (see fig. 2)], sharing some similari-
is comparable for males and females and young and ties with amineptine whose structure also contains
elderly individuals (extensive metabolisers) with a long aminoheptanoic acid side chain. Metabo-
the metabolite : parent drug ratios averaging 0.1 : 0.2 lically too it is similar to amineptine, being rapidly
in all groups, on a molar basis.[116] Exposure to and extensively biotransformed in humans and an-
mCPP and the metabolic ratios are not altered sig- imals, mainly by β-oxidation to shorter chain de-
nificantly by renal impairment[102,117-118] though they rivatives which appear in plasma, tissues and urine.
tend to increase in patients with biopsy-confirmed With tianeptine, the pentanoic acid derivative
hepatic cirrhosis, compared with healthy individ- (MC5) is the major metabolite which is further ox-
uals.[119] As mentioned, steady-state Cmax and AUC idised to a propionic acid (MC3) and a lactam de-
of mCPP and the mean metabolic ratio are mark- rivative (LMC5). A minor route includes N-dealk-
edly higher in poor metabolisers than extensive ylation of the heptanoic acid side chain to a number
metabolisers (see table V).[110] of intermediate compounds and subsequent modi-
Although exposure to mCPP appears to be low fication of the remaining tricyclic moiety.[124]
in most patients, there is no information on the Tianeptine is a racemate and the antidepressant-
metabolite : parent drug ratio at the site of action, like properties associated with its effect on sero-
at least in preclinical use. However, considerable tonin disposition appear to depend on the (–)-
individual differences in exposure to nefazodone, isomer.[125] However, there are no studies in the
but particularly to its metabolite mCPP, are appar- literature on the pharmacokinetics and metabolism

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
Metabolism of the Newer Antidepressants 293

of the 2 isomers after administration with either the should play a decisive role in the central effects of
racemate or the single isomers. tianeptine, if its activity is at least comparable.
Tianeptine metabolism may include the forma- After single oral doses of tianeptine in healthy
tion of reactive metabolite(s) which bind covalently volunteers (12.5mg) the metabolite MC5 reaches
to liver microsomal proteins both in vitro and in mean plasma Cmax and AUC slightly lower than the
vivo, in the mouse, hamster and rat. This activation parent drug, with a mean metabolite : parent drug
also occurs in vitro in human microsomes and ap- ratio averaging 0.6 to 0.8 (on a mg/L basis because
pears to be mediated by the glucorticoid-inducible molecular weights are similar). This ratio remains
CYP3A3 but not by CYP2D6 or CYP1A1 isoen- essentially unchanged after repeated administra-
zymes. The reactive metabolite(s), however, have tion although the plasma AUC of both compounds
are slightly higher than after a single dose. After
no direct hepatotoxic potential in the hamster at
the usual therapeutic doses in adults with depres-
less than a very high, sublethal dose of tianept-
sion (12.5mg 3 times daily) steady-state plasma
ine.[126]
AUC of both compounds are again higher than af-
The main oxidised metabolite MC5 is reported
ter a single dose but the active metabolite (t 1⁄2 14
to retain antidepressant activity,[123] although de- hours) accumulates more than the parent drug (t1⁄2
tails on the test, species and the potency relative to 6.3 hours), so the steady-state metabolic ratio rises
the parent drug are not available. In rats given compared with after acute administration[123] (see
single and repeated neuropharmacologically effec- table VI).
tive doses of tianeptine, the metabolite reaches Compared with young volunteers, exposure to
lower plasma Cmax but higher AUC than the parent both tianeptine and M5 is higher in the elderly with
compound because its elimination is slower. The or without depression. The clearance of tianeptine
same is true for the brain concentrations, though is reduced (by approximately 40% after intra-
brain uptake is slightly lower for the metabolite venous administration) and the t1⁄2β prolonged
than the parent drug.[127] Thus the metabolite (from 4.7 to 9 hours). A reduction in the daily dose

Table VI. Mean plasma area under the concentration-time curve (AUC) of tianeptine and its active M5 metabolite in different populations.
The elderly individuals were aged from 73 to 86 years.[128] The renally impaired patient group comprised 12 patients with CLR <1.14 L/h, 6
patients requiring chronic haemodialysis.[130] The mean metabolite-to-parent drug ratio (on a mg/L basis) is shown in parentheses
Individual Dosage (mg) AUC (mg/L • h)
tianeptine MC5
Healthy adults 12.5 po 0.7 ± 0.3 0.5 ± 0.3 (0.8)
12.5 po 0.7 ± 0.3 0.4 ± 0.1 (0.6)
12.5 tid × 4 days 1.1 0.8 ± 0.2 (0.7)
Adults with depression 12.5 po 1.0a 0.6a (0.6)
12.5 tid × 1 month 1.3a,b 1.4a,b (1.1)
Elderly 12.5 po 1.3 ± 0.2a 1.1 ± 0.3a (0.9)
12.5 IV 1.5 ± 0.3a
1.2 ± 0.6a (0.9)
Elderly person with depression 12.5 1.1 ± 0.5 1.2 ±0.4 (1.0)
12.5 × 17 days 1.3 ± 0.5b 1.1 ± 0.8 (0.8)
Patient with renal impairment 12.5 po 0.8 ± 0.3 1.1 ± 0.6c (1.4)
Patient with cirrhosis 12.5 po 0.8 ± 0.3c 0.4 ± 0.1c (0.4)
Patient with chronic alcoholism 12.5 po 0.5 ± 0.1c 0.2 ± 0.1c (0.4)
[123,128]
a Not compared with healthy individuals.
b Steady-state AUC was significantly greater than after single dose.[129]
c Significantly different from healthy volunteers.[130,131]
Abbreviations: IV = intravenously; po = orally; tid = 3 times daily.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
294 Caccia

(to 12.5mg twice daily) has been recommended in the 8-hydroxylation of mirtazapine, while the other
very old patients.[123,128,129] 2 oxidative processes are catalysed by CYP3A4.
Chronic renal failure does not appear to greatly Although mean oral clearance and t1⁄2β of racemic
affect the pharmacokinetics of the parent drug but mirtazapine (and t1⁄2 of its demethylated metabo-
the plasma AUC (see also table VI) and t1⁄2β of the lite) are very similar in the poor and extensive me-
metabolite (14.2 vs 4.9 hours) are higher than in tabolisers of debrisoquine after a single oral dose
healthy individuals after a single 12.5mg dose of of mirtazapine (7.5 and 15mg in poor and extensive
tianeptine. Reducing the dosage to 12.5mg twice a metabolisers, respectively) the possible stereo-
day has been recommended for these patients selective dependendence of mirtazapine and nor-
too.[130] Dosage reduction is not likely to be neces- mirtazepine metabolism on this polymorphic en-
sary in patients with combined depression and al- zyme cannot be excluded.[66,137] Mirtazapine does
tered liver function since no great difference (pa- not appear to alter the pharmacokinetics of desipra-
tients with alcoholism and cirrhosis) or even a mine (CYP2D6) and diazepam (CYP2C19 and
slight reductions (alcoholism without cirrhosis) CYP3A3/4).[132,135]
have been observed in tianeptine and M5 plasma The demethylated metabolite retains pharmaco-
AUC compared with individuals with normal liver logical activity (3 to 4 times less active than mirta-
function.[131] zapine)[132] although, again, details on the test and
species are not available. Its plasma AUC is ap-
6. The Noradrenergic and Specific proximately 50% that of the parent drug. The
Serotoninergic Agent Mirtazapine plasma t1⁄2β of the parent drug (mean 22 hours,
range from 13 to 34 hours in young healthy indi-
This is the only antidepressant that increases viduals)[135] and the demethylated metabolite (about
noradrenergic and serotoninergic (5-HT1-medi- 19 hours)[137] are similar after single oral doses.
ated-) neurotransmission through a blockade of Although the Cmax and t1⁄2β of mirtazapine in el-
central α2-adrenergic auto- and hetero-recep- derly patients with cirrhosis are only slightly
tors.[132,133] Chemically, mirtazapine is 1,2,3,4, higher than in matched controls, the hepatic clear-
10,14b-hexahydro-2-methyl-pyrazino[2,1-a]-pyrido- ance is approximately 33% lower and dosage ad-
[2,3-c]benzazepin(Z)-2 butenedioate (fig. 2), the justment may be necessary in individuals with
6-aza-derivative of mianserin and is a racemate of altered liver functions.[135]
the R- and S-isomers which show quantitative and
qualitative differences in their in vivo and in vitro 7. The Reversible Inhibitors of
α2-adenoceptors antagonist properties. The com- Monoamine Oxidase Moclobemide
parative pharmacological profile of the racemate
and its enantiomers has been described by de Boer While the older monoamine oxidase inhibitor
et al.[134] and reviewed by Davis and Wilde.[135] nonselectively and irreversibly inhibit both MAO-
Information is minimal about the disposition of A and MAO-B isoenzymes, the newer generation
racemic mirtazapine. The drug undergoes an exten- preferentially (and reversibly) affects the MAO-A
sive first pass metabolism and clearance is mostly subtype, the enzyme system primarily responsible
via hepatic metabolism, urinary excretion account- for deamination of those amines believed to be im-
ing for only about 4% of the administered dose.[132] plicated in the aetiology of depression. This new
The main metabolic pathways in humans are N- class includes brofaromine, cimoxatone and mo-
demethylation, N-oxidation and 8-hydroxylation clobemide. The neuropharmacological profiles of
followed by conjugation.[136,137] A preliminary in cimoxatone[138] and moclobemide[139,140] have
vitro study using human liver microsomes and cells been reviewed. Only moclobemide is considered
expressing different enzymes suggests that CYP2D6 here. Information is minimal on the metabolism of
(and to a lesser extent CYP1A2) mainly catalyses cimoxatone which, however, is reported to be

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
Metabolism of the Newer Antidepressants 295

Table VII. Some pharmacokinetic parameters of moclobemide and its metabolites Ro 12-8095 and Ro 12-5637 in healthy individuals and
hepatically and renally impaired patients[142-145]
Participants and dose (mg) Parameter Moclobemide Ro 12-8095 Ro 12-5637
Healthy individuals (100) CLpo (L/h) 83.6 ± 41.0
Cmax (mg/L) 0.58 ± 0.23 0.49 ± 0.15 0.10 ± 0.03
Patients with cirrhosis (100) CLpo (L/h) 20.2 ± 17.8a
Cmax (mg/L) 1.61 ± 0.68a O.35 ± 0.16 0.14 ± 0.08
Patients with renal impairment (100) CLpo (L/h) 77.6 ± 45.0
Cmax (mg/L)c 0.59 (47) 0.58 (26) 0.19 (36)a
Healthy individuals (100 tid for 15 days) CLpo (L/h) 33.8 ± 7.72b
Cmin (mg/L) 0.22 ± 0.12 0.23 ± 0.09 0.07 (27)c
a Significantly different from healthy individuals.
b First day of dose administration (n = 12).
c In parentheses, the coefficient of variation (n = 13, with varying degrees of renal impairment, CLR 0 to 2.4 L/h).
Abbreviations: CLpo = oral clearance; Cmax = maximum plasma drug concentrations: Cmin = minimum steady-state concentration; tid = 3 times
daily.

extensively biotransformed to its active O-demeth- Two minor ring opened metabolites have MAO-
ylated derivative.[138] The development of brofaro- B inhibitory activity in rat liver while the primary
mine has been stopped.[141] amine Ro 16-6491 is a potent inhibitor of rat brain
Moclobemide is a benzamide derivative con- MAO-B in vitro, possibly accounting for the ex vivo
taining a morpholine ring as its main structural peripherical MAO-B inhibition by the parent drug.
characteristic (p-chloro-N-[2-morpholinoethy] The amine derivative, however, is undetectable in
benzamide) [fig. 2]. It has intermediate-to-high human plasma after a single dose of moclobemide
clearance and is subject to a substantial first pass 100mg.[147]
metabolism, resulting in low and variable oral bio- Morpholine C-oxidation to the lactam deriva-
availability. Clearance is essentially metabolic and tive Ro 12-8095 is quantitatively prominent in
decreases as the dose increases and after repeated humans and yields a number of metabolic forms
administration (table VII), possibly because of with carbamide, tertiary and secondary amine
auto-inhibition or metabolite-induced inhibition of structure.[146] This main pathway appears to in-
moclobemide metabolism. Clearance is also re- volve CYP2C19, as the oral clearance of moclo-
duced by cimetidine, an inhibitor of drug metabo- bemide is lower and t1⁄2 longer in poor metabolisers
lising systems. An extensive review of the clinical than in extensive metabolisers of S-mephenytoin,
pharmacokinetics of this drug has recently been although the difference is less marked after re-
published.[142] peated administration.[142,150]
Moclobemide undergoes a complex metabolic CYP2D6 does not appear to play a significant
fate initially involving morpholine carbon and role in the metabolism of moclobemide. The parent
nitrogen oxidation, deamination and aromatic drug and its main metabolites are weaker inhibitors
hydroxylation, with quantitative differences be- of CYP2D6 in vitro, but there is evidence that
tween species. [146,147] Of the identified metabo- moclobemide inhibits this isoenzyme in vivo[151]
lites the N-oxide derivative and a ring-opened although the clinical relevance of these findings is
metabolite retain modest MAO-A inhibitory activ- not clear.[142]
ity in vitro.[146] In the rat the N-oxide is equipotent The lactam metabolite appears rapidly in
to the parent drug as an inhibitor of brain and liver plasma, reaching concentrations comparable with
MAO-A.[140,148] A more active, but as yet uniden- those of moclobemide in healthy volunteers[143]
tified, metabolite has been suggested. [140,149] but it is unlikely to contribute to the effects of the

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
296 Caccia

parent drug since – as mentioned – it is essentially bolite whose formation has not yet been well
inactive.[140,142] The N-oxide metabolite Ro 12-5637 characterised, but appears to be carbamazepine-
is also present in blood but at lower concentrations inducible, has been identified in the body fluids of
than the parent drug and its lactam derivative.[143] volunteers and patients taking milnacipran.[76]
The single and multiple dose pharmacokinetics of The primary metabolites, often pharmacologi-
this and other metabolites in humans, and their dis- cally active, are generally substrates for further
position compared with the parent drug in animals, phase I processes. Thus most of these drugs un-
have not yet been fully characterised. dergo various parallel and/or sequential oxidation
Information is also limited on how various steps before conjugation and excretion as polar me-
physiological and pathological factors affecting tabolites. These are usually the rate-limiting steps,
drug elimination influence the pharmacokinetic as they are influenced to varying degrees by
behaviour of metabolites. The pharmacokinetics of recognised endogenous and exogenous factors af-
moclobemide are not significantly affected by age fecting drug disposition, and at least those medi-
and renal function, but the tmax and Cmax of its N- ated by CYP2D6 and CYP2C19 isoenzymes by
oxide derivative are significantly higher in patients genetic polymorphism.[17,18,66]
with moderate or severe renal impairment than in Some pathways proceed through saturable
healthy volunteers.[144,145] Chronic liver disease in- mechanisms as in the case of fluvoxamine,[152]
creases the mean Cmax of the N-oxide and reduces fluoxetine, nefazodone, paroxetine and venlafaxine.
that of the lactam derivative, besides markedly re- The clinical significance of this finding has been
ducing clearance and prolonging the t1⁄2β (3.9 vs 1.7 reviewed elsewhere.[16,17,30,44] Others may be influ-
hours) of intravenous moclobemide 90mg com- enced by chirality (e.g. citalopram and demethyl-
pared with average values in healthy individu-
citalopram and fluoxetine N-dealkylation) when
als.[139,142,145] The oral bioavailability and Cmax of
the drug is a racemic mixture of enantiomers (as
moclobemide are also significantly higher in these
are mirtazapine, reboxetine and tianeptine, al-
patients; mean Cmax of the 2 metabolites are less
though few or no data are yet available on this as-
affected while tmax was longer and far more vari-
pect of their disposition). This may explain the in-
able than in healthy individuals.[142,145]
tra- and inter-population variability with most of
these drugs which from the pharmacokinetic point
8. Conclusions of view is approximately the same as for TCAs.
Like most of the older drugs in the same ther- The main isoenzymes responsible for the meta-
apeutic class, these newer antidepressants are bolism of the older antidepressants, and lipophilic
extensively biotransformed before excretion. drugs in general (CYP 3A3/4, 2D6, 1A2, 2C), are
Sometimes, as in the case of moclobemide,[142] nu- involved to varying degrees in the oxidation of
merous metabolites have been identified in the these newer agents. They also mediate the metabo-
blood and urine of animals and humans, making it lic interactions between some of these agents and
hard to specify what role the administered drug other drugs metabolised through the same en-
plays in the overall outcome. In some cases, how- zyme(s). There are full reviews[16-19,29-31,63-65]
ever, only the main metabolites have been identi- dealing with the in vitro potency on selected en-
fied, and a substantial proportion of the dose re- zymes and the likelihood of clinically important
mains unaccounted for. drug-drug interactions. Metabolites have been less
Most of these drugs are primarily dependent on studied but where information is available it ap-
1 or more phase I oxidative pathways for their pears that these may retain in vitro inhibitory po-
metabolism, except milnacipran whose clearance tencies similar to (e.g. paroxetine and its M2 meta-
appears due equally to renal excretion and glucu- bolites as inhibitors of 2D6) or even greater than
ronide conjugation. However, a dealkylated meta- the parent drug on specific isoenzymes (for instance

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
Metabolism of the Newer Antidepressants 297

fluoxetine and norfluoxetine are equally active in Further advances in this field require addi-
inhibiting 2D6 and 2C9 but the metabolite is more tional information on the pharmacological activity
potent as an inhibitor of 3A3/4).[15,18] Hence the of the metabolites after direct administration to an-
need to identify and characterise all the pathways imals, particularly after repeated administration
in drug metabolism and the enzymes involved, and which more closely mimics the clinical situation.
any factors which by altering their activity could It would be useful to know their plasma protein
potentially modify the therapeutic response. This binding and pharmacokinetic behaviour with par-
information is lacking for some of these drugs. ticular regard to the brain-to-blood partition and
As regards metabolites’ pharmacological activ- concentrations reached at the site of action com-
ity, knowledge is clearly inadequate for many of
pared with the parent drug. This is because the
these drugs and in most cases there is no real
blood-brain distribution of the metabolites may
information about their role in the beneficial
differ from the original molecule, and as a con-
and/or adverse effects of the parent drug. With few
sequence the metabolite-to-parent drug ratio in
exceptions, only the main metabolites have been
investigated, generally with regard to their in vitro blood does not always reflect that at the site of
activity on monoaminergic mechanism(s), which action.
are of course important in mediating the anti- This picture has been described for many cen-
depressant action of the parent compound. How- trally acting drugs and their metabolites,[11,105] and
ever, in vitro potency and selectivity may translate detailed information about the relationship be-
poorly into in vivo pharmacological responses. The tween the parent drug and metabolite concentra-
MAO-A inhibitor potency of moclobemide N-oxide tions in blood and brain is essential for under-
in vitro in rat brain, for example, is several times standing the contribution of each active species to
lower than that of moclobemide but the parent drug any given pharmacological or toxic effect. This
and its metabolite have comparable potency and information needs to be extended, when appropri-
selectivity ex vivo in rats.[148] Despite the fact that ate, to phase II metabolites as some of these too
norfluoxetine and norsertraline are less potent than may enter the brain and have direct pharmacolog-
their parent drugs in inhibiting serotonin uptake in ical or toxic effects.[153]
vitro, the metabolites are at least as active in inhib- While animal studies illustrate the importance
iting food intake in animals, probably playing a of measuring drugs and metabolites at the precise
role in the hypophagic action of these compounds target site, they highlight the hazard of simply ex-
in some species,[57,75] possibly through mecha- trapolating across species because of differences in
nisms other than inhibition of serotonin uptake.
brain drug uptake.[96,97,154] Although studies in
The phenyl-piperazine derivative of nefazodone,
different species may improve predictivity, defini-
mCPP, shows 5-HT2C-like agonist properties in rat
tive evidence of metabolite activity, and how it
and humans,[114] further showing that metabolites
contributes to the overall outcome, can only come
may have certain characteristics in vitro but others
in vivo, and that their activity must, therefore, be from human studies. These are still far away, but
evaluated in a broad sense, not only for the anti- non-invasive techniques are increasingly being
depressant-like effects of the parent drug. Unlike used for evaluating the pharmacodynamic response
moclobemide, some of its minor ring-opened me- to drug therapy and to measure drug concentrations
tabolites have MAO-B inhibitory activity in the in tissues.[155-157] As these technologies become
rat,[140,146,147] illustrating the need to extend phar- more refined, the relationships between drug dose,
macological and pharmacokinetic studies to all blood and tissue concentrations and affinity for
metabolites for reliable extrapolation of pharmaco- specific neurotransmitter systems might be quanti-
logical findings to the clinical situation. fied in humans.

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
298 Caccia

References 23. Benfield P, Heel RC, Lewis SP. Fluoxetine: a review of its
1. Coccaro EF, Siever LJ. Second generation antidepressants: a pharmacodynamic and pharmacokinetic properties, and
comparative review. J Clin Pharmacol 1985; 25: 241-60 therapeutic efficacy in depressive illness. Drugs 1986; 32:
2. Blackwell B, Simon JS. Second-generation antidepressants. 481-508
Drugs Today 1986; 22: 611-33 24. Wong DT, Bymaster FP, Reid LR, et al. Norfluoxetine en-
3. Hollister LE. Current antidepressants. Annu Rev Pharmacol antiomers as inhibitors of serotonin uptake in rat brain. Neu-
Toxicol 1993; 26: 23-37 ropsychopharmacology 1993; 8: 337-44
4. Rudorfer MV, Potter WZ. Antidepressants: a comparative re- 25. Fuller, RW, Wong DT. Serotonin re-uptake blockers in vitro and
view of the clinical pharmacology and therapeutic use of the in vivo. J Clin Psychopharmacol 1987; 7: 365-435
‘newer’ versus the ‘older’ drugs. Drugs 1989; 37: 713-38 26. Murdoch D, McTavish D. Sertraline: a review of its pharmaco-
5. Möller H-J, Volz H-P. Drug treatment of depression in the dynamic and pharmacokinetic properties, and therapeutic po-
1990s: an overview of achievements and future possibilities. tential in depression and obsessive-compulsive disorder.
Drugs 1996; 52: 625-6 Drugs 1992; 44: 604-24
6. Skerritt U, Evans R, Montgomery SA. Selective serotonin 27. Ronfeld RA, Tremaine LM, Wilner KD. Pharmacokinetics of
reuptake inhibits in older patients: a tolerability perspective. sertraline and its N-demethyl metabolite in elderly and young
Drugs Aging 1997; 10: 209-18 male and female volunteers. Clin Pharmacokinet 1997; 32:
7. Amsterdam J, Brunswick D, Mendels J. The clinical application Suppl. 1: 22-30
of tricyclic antidepressant pharmacokinetics and plasma lev- 28. Benfield P, Ward A. Fluvoxamine: a review of its pharmacody-
els. Am J Psychiatry 1980; 137: 653-62 namic and pharmacokinetic properties, and therapeutic effi-
8. Guthrie S, Lane EA, Linnoila M. Monitoring of plasma drug cacy in depressive illness. Drugs 1986; 32: 313-34
concentrations in clinical psychopharmacology. In: Meltzer 29. Preskorn SH, Magnus RD. Inhibition of hepatic P-450 isoen-
HY, editor. Psychopharmacology: the third generation of zymes by serotonin selective reuptake inhibitors: in vitro and
progress. New York: Raven Press, 1987: 1323-9 in vivo findings and their implications for patient care. Psy-
9. Preskorn SH, Dorey RC, Jerkovich GS. Therapeutic drug chopharmacol Bull 1994; 30: 251-9
monitoring of tricyclic antidepressants. Clin Chem 1988; 30. Lane RM. Pharmacokinetic drug interaction potential of selec-
34: 822-8 tive serotonin reuptake inhibitors. Int Clin Psychopharmacol
10. Orsulak PJ. Therapeutic monitoring of antidepressant drugs: 1996; 11 Suppl. 5: 31-61
guidelines updated. Ther Drug Monit 1989; 11: 497-507 31. Nemeroff CB, DeVane CL, Pollock BG. Newer antidepressants
11. Caccia S, Garattini S. Formation of active metabolites of psy- and the cytochrome P450 system. Am J Psychiatry 1996; 153:
chotropic drugs: an updated review of their significance. Clin 311-20
Pharmacokinet 1990; 18: 434-59 32. Becquemont L, Ragueneau I, Le Bot MA, et al. Influence of the
12. Potter WZ, Manji HK. Antidepressants, metabolites, and appar- CYP1A2 inhibitor fluvoxamine on tacrine pharmacokinetics
ent drug resistance. Clin Neuropharmacol 1990; 13 Suppl. 1: in humans. Clin Pharmacol Ther 1997; 61: 619-27
S45-53
33. Spigset O, Carleborg L, Hedenmalm K, et al. Effect of cigarette
13. Nordin C, Bertilsson L. Active hydroxymetabolites of antide-
smoking on fluvoxamine pharmacokinetics in humans. Clin
pressants: emphasis on E-10-hydroxy-nortriptyline. Clin
Pharmacol Ther 1995; 58: 399-403
Pharmacokinet 1995; 28: 26-40
34. Crewe HK, Lennard MS, Tucker GT, et al. The effect of selec-
14. Caccia S, Garattini S. Pharmacokinetic and pharmacodynamic
tive serotonin re-uptake inhibitors on cytochrome P4502D6
significance of antidepressant drug metabolites. Pharmacol
(CYP2D6) activity in human liver microsomes. Br J Clin
Res 1992; 26: 317-29
Pharmacol 1992; 34: 262-5
15. Preskorn SH. Pharmacokinetics of antidepressants: why and
how they are relevant to treatment. J Clin Psychiatry 1993; 54 35. Skjelbo E, Brosen K. Inhibitors of imipramine metabolism by
Suppl.: 14-34 human liver microsomes. Br J Clin Pharmacol 1992; 34:
16. DeVane CL. Pharmacokinetics of the newer antidepressants: 256-61
Clinical relevance. Am J Med 1994; 97 Suppl. 6A: 13S-23S 36. von Moltke LL, Greenblatt DJ, Court MH, et al. Inhibition of
17. Goodnick PJ. Pharmacokinetic optimisation of therapy with alprazolam and desipramine hydroxylation by paroxetine and
newer antidepressants. Clin Pharmacokinet 1994; 27: 307-30 fluvoxamine: comparison with other selective serotonin
18. Baumann P. Pharmacokinetic-pharmacodynamic relationship reuptake inhibitor antidepressants. J Clin Psychopharmacol
of the selective serotonin reuptake inhibitors. Clin Phar- 1995; 15: 125-31
macokinet 1996; 31: 444-69 37. Jeppesen U, Rasmussen BB, Brosen K. Fluvoxamine inhibits
19. Rudorfer MV, Potter WZ. The role of metabolites of antidepres- the CYP2C19-catalyzed bioactivation of chloroguanide. Clin
sants in the treatment of depression. CNS Drugs 1997; 7: Pharmacol Ther 1997; 62: 279-86
273-312 38. Hartter S, Wetzel H, Hammes E, et al. Inhibition of antidepres-
20. Claassen V. Review of the animal pharmacology and pharma- sant demethylation and hydroxylation by fluvoxamine in de-
cokinetics of fluvoxamine. Br J Clin Pharmacol 1983; 15: pressed patients. Psychopharmacology 1993; 110: 302-8
349S-55S 39. Kaye CM, Haddock RE, Langley PF, et al. A review of the
21. Dechant KL, Clissold SP. Paroxetine: a review of its pharmaco- metabolism and pharmacokinetics of paroxetine in man. Acta
dynamic and pharmacokinetic properties, and therapeutic po- Psychiatr Scand 1989; 80 Suppl. 350: 60-75
tential in depressive illness. Drugs 1991; 41: 225-53 40. Crewe S, Lennard MS, Tucker GT, et al. The effect of paroxetine
22. Milne CJ, Goa KL. Citalopram: a review of its pharmacody- and other specific serotonin re-uptake inhibitors on cyto-
namic and pharmacokinetic properties, and therapeutic poten- chrome P450IID6 activity in human liver microsomes. Br J
tial in depressive illness. Drugs 1991; 41: 450-77 Clin Pharmacol 1991; 32: 658P-9P

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
Metabolism of the Newer Antidepressants 299

41. Bloomer JC, Woods FR, Haddock RE, et al. The role of cyto- 57. Anelli M, Bizzi A, Caccia S, et al. Anorectic activity of fluoxet-
chrome P4502D6 in the metabolism of paroxetine by human ine and norfluoxetine in mice, rats and guinea-pigs. J Pharm
liver microsomes. Br J Clin Pharmacol 1992; 33: 521-3 Pharmacol 1992; 44: 696-8
42. Sindrup SH, Brøsen K, Gram LF, et al. The relationship be- 58. Caccia S, Fracasso C, Garattini S, et al. Effects of short- and
tween paroxetine and the sparteine oxidation polymorphism. long-term administration of fluoxetine on the monoamine
Clin Pharmacol Ther 1992; 51: 278-87 content of rat brain. Neuropharmacology 1992; 31: 343-7
43. Brøsen K, Hansen JG, Nielsen KK, et al. Inhibition by paroxet- 59. Caccia S, Anelli M, Codegoni AM, et al. The effect of single
ine of desipramine metabolism in extensive but not in poor and repeated anorectic doses of 5-hydroxytryptamine uptake
metabolizers of sparteine. Eur J Clin Pharmacol 1993; 44: inhibitors on indole levels in rat brain. Br J Pharmacol 1993;
349-55 110: 355-9
44. Preskorn SH. Clinically relevant pharmacology of selective se- 60. Altamura AC, Moro AR, Percudani M. Clinical pharmacokinet-
rotonin reuptake inhibitors: an overview with emphasis on ics of fluoxetine. Clin Pharmacokinet 1994; 26: 201-14
pharmacokinetics and effects on oxidative drug metabolism. 61. Hamelin BA, Turgeon J, Vallée F et al. The disposition of
fluoxetine but not sertraline is altered in poor metabolizers of
Clin Pharmacokinet 1997; 32 Suppl. 1: 1-21
debrisoquin. Clin Pharmacol Ther 1996; 60: 512-21
45. Ozdemir V, Naranjo CA, Herrmann N, et al. paroxetine poten-
62. Otton SV, WU D, Joffe RT, et al. Inhibition by fluoxetine of
tiates the central nervous system side effects of perphenazine:
cytochrome P4562D6 activity. Clin Pharmacol Ther 1993;
contribution of cytocrome P4502D6 inhibition in vivo. Clin
53: 401-9
Pharmacol Ther 1997; 62: 334-47
63. Harvey A, Preskorn S. Cytochrome P450 enzymes: interpreta-
46. Tremaine LM, Welch WM, Ronfeld RA. Metabolism and dis- tion of their interactions with selective serotonin reuptake
position of the 5-hydroxytryptamine uptake blocker sertraline inhibitors. Part II. J Clin Psychopharmacol 1996; 16: 345-55
in the rat and dog. Drug Metab Dispos 1989; 17: 542-50 64. Brosen K. Are pharmacokinetic drug interactions with the
47. Sindrup SH, Brøsen K, Hansen MGJ, et al. Pharmacokinetics SSRIS and issue? Int Clin psychopharmacol 1996; 11 Suppl.
of citalopram in relation to the sparteine and the mephenytoin 1: 23-7
oxidation polymorphisms. Ther Drug Monit 1993; 15: 11-7 65. Meyer UA, Amrein R, Balant LP et al. Antidepressants and
48. Baumann P, Nil R, Souche A, et al. A double-blind placebo-con- drug-metabolizing enzymes: expert group report. Acta Psy-
trolled study of citalopram with and without lithium in the chiatr Scand 1996; 93: 71-9
treatment of therapy-resistant depressive patients: a clinical, 66. Bertilsson L, Dahl M-L, Tybring G. Pharmacogenetics of anti-
pharmacokinetic, and pharmacogenetic investigation. J Clin depressants: clinical aspects. Acta Psychiatr Scand Suppl
Psychopharmacol 1996; 16: 307-14 1997; 391: 14-21
49. Kobayashi K, Chiba K, Yagi T, et al. Identification of cyto- 67. Sproule BA, Naranjo CA, Bremner KE, et al. Selective seroto-
chrome P450 isoforms involved in citalopram N-demethyla- nin reuptake inhibitors and CNS drug interactions. Clin Phar-
tion by human liver microsomes. J Pharmacol Exp Ther 1997; macokinet 1997; 33: 454-71
280: 927-33 68. Zussman BD, Davie CC, Fowles SE, et al. Sertraline, like other
50. Rochat B, Amey M, Baumann P. Analysis of enantiomers of SSRIs, is a significant inhibitor of desipramine metabolism
citalopram and its demethylated metabolites in plasma of de- in vivo. Br J Clin Pharmacol 1995; 39: 550P
pressive patients using chiral reverse-phase liquid chroma- 69. Alderman J, Preskorn SH, Greenblatt DJ, et al. desipramine
tography. Ther Drug Monit 1995; 17: 273-9 pharmacokinetics when coadministered with paroxetine or
51. Foglia JP, Pollock BG, Kirshner MA, et al. Plasma levels of sertraline in extensive metabolizers. J Clin Psychopharmacol
citalopram enantiomers and metabolites in elderly patients. 1997; 17: 284-91
Psychopharmacol Bull 1997; 33: 109-12 70. Kurtz DL, Bergstrom RF, Goldberg MJ, Cerimele BJ. The effect
52. Rochat B, Amey M, Gillet M, et al. Identification of three cy- of sertraline on the pharmacokinetics of desipramine and
tochrome P450 isozymes involved in N-demethylation of imipramine. Clin Pharmacol Ther 1997; 62: 145-56
citalopram enantiomers in human liver microsomes. Pharma- 71. Carson SW. Pharmacokinetic and pharmacodynamic drug in-
cogenetics 1997; 7: 1-10 teractions with polypharmacotherapy of treatment-resistant
affective and obsessive-compulsive disorders. Psychopharmacol
53. Bondolfi G, Chautems C, Rochat B, et al. Non-response to
Bull 1996; 32: 555-68
citalopram in depressive patients: pharmacokinetic and clin-
72. Preskorn SH, Alderman J, Greenblatt DJ, et al. Sertraline does
ical consequences of a fluvoxamine augmentation. Psycho-
not inhibit cytochrome P4503A-mediated drug metabolism in
pharmacology 1996; 128: 421-5
vivo. Psychopharmacol Bull 1997; 33: 659
54. Torok-Both GA, Baker GB, Coutts RT, et al. Simultaneous de-
73. Sprouse J, Clarke T, Reynolds L, et al. Comparison of the ef-
termination of fluoxetine and norfluoxetine enantiomers in fects of sertraline and its metabolite desmethylsertraline on
biological samples by gas chromatography with electron-cap- blockade of central 5-HT reuptake in vivo. Neuropsycho-
ture detection. J Chromatogr B 1992; 579: 99-106 pharmacology 1996; 14: 225-31
55. Fjordside L, Jeppesen U, Eap CB et al. The stereoselective 74. Fuller RW, Hemrick-Luecke SK, Littlefield ES, et al. Compar-
metabolism of fluoxetine in poor and extensive metabo- ison of desmethylsertraline with sertraline as a monoamine
lisers of sparteine [abstract]. Eur J Clin Pharmacol 1997; uptake inhibitor in vivo. Prog Neuropsychopharmacol 1995;
Suppl. I: A127 19: 135-49
56. Fuller RW, Snoddy HD, Perry KW. Importance of duration of 75. Garattini S, Bizzi A, Caccia S, et al. Progress report on the
drug action in the antagonism of p-chloroamphetamine deple- anorectic effects of dexfenfluramine, fluoxetine and
tion of brain serotonin-comparison of fluoxetine and sertraline, Int J Obes Relat Metab Disord 1992; 16 Suppl.
chlorimipramine. Biochem Pharmacol 1978; 27: 193-8 3: S43-50

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
300 Caccia

76. Puozzo C, Leonard BE. Pharmacokinetics of milnacipran in 94. Melloni P, Carniel G, Della Torre A, et al. Potential anti-
comparison with other antidepressants. Int Clin Psy- depressant agents. α-aryloxy-benzyl derivatives of ethanol-
chopharmacol 1996; 11 Suppl. 4: 15-27 amine and morpholine. Eur J Med Chem Chim Ther 1984;
77. Briley M, Prost JF, Moret C. Preclinical pharmacology of 19: 235-42
milnacipran. Int Clin Psychopharmacol 1996; 11 Suppl. 4: 95. Strolin Benedetti M, Frigerio E, Tocchetti P, et al. Stereoselec-
10-4 tive and species-dependent kinetics of reboxetine in mouse
78. Muth EA, Haskins JT, Moyer JA, et al. Antidepressant biochem- and rat. Chirality 1995; 7: 285-9
ical profile of the novel bicyclic compound Wy-45,030, an 96. Renshaw PF, Guimaraes AR, Fava M, et al. Accumulation of
ethyl cyclohexanol derivative. Biochem Pharmacol 1986; 35: fluoxetine and norfluoxetine in human brain during therapeu-
4493-7 tic administration. Am J Psychiatry 1992; 149: 1592-4
79. Artigas F. Selective serotonin/noradrenaline reuptake inhibitors 97. Bergami A, Fracasso C, Garattini S, et al. Brain uptake and
(SNRIs): pharmacology and therapeutic potential in the treat- acute indole-depleting effect of dexfenfluramine in squirrel
ment of depressive disorders. CNS Drugs 1995; 4: 79-89 monkeys after a high-dose regimen. Pharm Sci 1995; 1: 45-8
80. Bonnaud B, Cousse H, Mouzin G, et al. 1-Aryl-2-(aminoethyl) 98. Dostert P, Benedetti MS, Poggesi I. Review of the pharmacoki-
cyclopropanecarboxylic acid derivatives: a new series of po- netics and metabolism of reboxetine, a selective noradrena-
tential antidepressants. J Med Chem 1987; 30: 318-25 line reuptake inhibitor. Eur Neuropsychopharmacol 1997; 7
81. Morton WA, Sonne SC, Verga MA. Venlafaxine: a structurally Suppl. 1: S23-35
unique and novel antidepressant. Ann Pharmacother 1995; 29: 99. Cocchiara G, Battaglia R, Pevarello P, et al. Comparison of the
387-95 disposition and of the metabolic patterns of reboxetine, a new
82. Troy SM, Schultz RW, Parker VD, et al. The effect of renal disease antidepressant, in the rat, dog, monkey and man. Eur J Drug
on the disposition of venlafaxine. Clin Pharmacol Ther 1994; Metab Pharmacokinet 1991; 16: 231-9
56: 14-21 100. Pellizzoni C, Poggesi I, Jørgensen NP, et al. Pharmacokinetics
of reboxetine in healthy volunteers: single against repeated
83. Wang CP, Howell SR, Scatina J, et al. The disposition of
oral doses and lack of enzymatic alterations. Biopharm Drug
venlafaxine enantiomers in dogs, rats, and humans receiving
Dispos 1996; 17: 623-33
venlafaxine. Chirality 1992; 4: 84-90
101. Edwards DMF, Pellizzoni C, Breuel HP, et al. Pharmacokinetics
84. Howell SR, Husbands GEM, Scatina JA, et al. Metabolic dis-
of reboxetine in healthy volunteers: single oral doses, linearity
position of 14C-venlafaxine in mouse, rat, dog, rhesus monkey
and plasma protein binding. Biopharm Drug Dispos 1995; 16:
and man. Xenobiotica 1993; 23: 349-59
443-60
85. Muth EA, Moyer JA, Haskins JT, et al. Biochemical, neuro- 102. Davis R, Whittington R, Bryson HM. Nefazodone: a review of
physiological, and behavioral effects of Wy-45,233 and other its pharmacology and clinical efficacy in the management of
identified metabolites of the antidepressant venlafaxine. Drug major depression. Drugs 1997; 53: 608-36
Dev Res 1991; 23: 191-9
103. Mayol RF, Cole CA, Luke GM, et al. Characterization of the
86. Otton SV, Ball SE, Cheung SW, et al. Venlafaxine oxidation in metabolites of the antidepressant drug nefazodone in human
vitro is catalysed by CYP2D6. Br J Clin Pharmacol 1996; 41: urine and plasma. Drug Metab Dispos 1994; 22: 304-11
149-56 104. Caccia S, Fong MH, Garattini S, et al. Plasma concentrations
87. von Moltke LL, Greenblatt DJ, Duan SX, et al. Phenacetin O- of trazodone and 1-(3-chlorophenyl)-piperazine in man after
deethylation by human liver microsomes in vitro: inhibition a single oral dose of trazodone. J Pharm Pharmacol 1982; 34:
by chemical probes, SSRI antidepressants, nefazodone and 605-6
venlafaxine. Psychopharmacology 1996; 128: 398-407 105. Caccia S, Garattini S. 1-Arylpiperazine as active metabolites of
88. Otton SV, Ball SE, Cheung SW, et al. Comparative inhibition (4-substituted aryl-1-piperazinyl)alkyl heterocyclic drugs.
of the polymorphic enzyme CYP2D6 by venlafaxine (VF) Acta Pharm Jugosl 1990; 40: 441-60
and other 5HT uptake inhibitors. Clin Pharmacol Ther 1994; 106. Caccia S, Vigano GL, Mingardi G, et al. Clinical pharmacoki-
55: 141 netics of oral buspirone in patients with impaired renal func-
89. Ball SE, Ahern D, Scatina J, et al. Venlafaxine: in vitro inhibi- tion. Clin Pharmacokinet 1988; 14: 171-7
tion of CYP2D6 dependent imipramine and desipramine me- 107. Bianchi G, Caccia S, Della Vedova F, et al. The alpha2-adreno-
tabolism. Comparative studies with selected SSRIs, and ceptor antagonist activity of ipsapirone and gepirone is medi-
effects on human hepatic CYP3A4, CYP2C9 and CYPIA2. ated by their common metabolite 1-(2-pyrimidinyl)-
Br J Clin Pharmacol 1997; 43: 619-26 piperazine (PmP). Eur J Pharmacol 1988; 151: 365
90. Troy SM, Lucki I, Peirgies AA, et al. Pharmacokinetic and phar- 108. Schmider J, Greenblatt DJ, von Moltkr LL, et al. Inhibition of
macodynamic evaluation of the potential drug interaction be- cytochrome P450 by nefazodone in vitro: studies of dextro-
tween venlafaxine and diazepam. J Clin Pharmacol 1995; 35: methorphan O- and N-demethylation. Br J Clin Pharmacol
410-9 1996; 41: 339-43
91. Ketter TA, Flockhart DA, Post RM, et al. The emerging role of 109. Greene DS, Barbhaiya RH. Clinical pharmacokinetics of
cytochrome P450 3A in psychopharmacology. J Clin Psy- nefazodone. Clin Pharmacokinet 1997; 33: 260-75
chopharmacol 1995; 15: 387-98 110. Barbhaiya RH, Buch AB, Greene DS. Single and multiple dose
92. Ereshefsky L. Drug-drug interactions involving antidepres- pharmacokinetics of nefazodone in subjects classified as ex-
sants: focus on venlafaxine. J Clin Psychopharmacol 1996; 16 tensive and poor metabolizers of dextromethorphan. Br J Clin
Suppl. 2: 37S-50S Pharmacol 1996; 42: 573-81
93. Melloni P, Della Torre A, Lazzari E, et al. Configuration studies 111. Marino MR, Langenbacher KM, Uderman HD. Interaction of
on 2[α-ethoxyphenoxy][benzyl]morpholine FCE 20124. Tet- nefazodone (N) and fluoxetine (F). Clin Pharmacol Ther
rahedron 1985; 41: 1393-9 1996; 59: 180

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
Metabolism of the Newer Antidepressants 301

112. Yasui N, Otani K, Kaneko S, et al. Inhibition of trazodone me- 130. Salvadori C, Merdjan H, Brouard R, et al. Tianeptine and its
tabolism of thioridazine in humans. Ther Drug Monit 1995; main metabolite. Disposition in chronic renal failure and
17: 333-5 haemodialysis. Fundam Clin Pharmacol 1990; 4: 663-71
113. Hamik A, Peroutka SJ. 1-(m-chlorophenyl)piperazine (mCPP) 131. Royer RJ, Royer-Morrot MJ, Paille F, et al. Tianeptine and its
interactions with neurotransmitter receptors in the human main metabolite. Pharmacokinetics in chronic alcoholism and
brain. Biol Psychiatry 1989; 25: 569-75 cirrhosis. Clin Pharmacokinet 1989; 16: 186-91
114. Murphy DL, Lesch KP, Aulakh CS, et al. Serotonin-selective 132. Sitsen JMA, Zivkov M. Mirtazapine: clinical profile. CNS
arylpiperazines with neuroendocrine, behavioral, tempera- Drugs 1995; 4 Suppl. 1: 39-48
ture, and cardiovascular effects in humans. Pharmacol Rev 133. Nutt D. Mirtazepine: pharmacology in relation to adverse ef-
1991; 43: 527-50 fects. Acta Psychiatr Scand 1997; 96 Suppl. 391: 31-7
115. Benjamin J, Greenberg BD, Murphy DL. Daily administration 134. de Boer Th, Maura G, Raiteri M, et al. Neurochemical and
of m-chlorophenylpiperazine to healthy human volunteers autonomic pharmacological profiles of the 6-aza-analogue of
rapidly attenuates many of its behavioral, hormonal, cardio- mianserin, ORG 3770 and its enantiomers. Neuropharmacol-
ogy 1988; 27: 399-408
vascular and temperature effects. Psychopharmacology 1996;
135. Davis R, Wilde MI. Mirtazapine: a review of its pharmacology
127: 140-9
and therapeutic potential in the management of major depres-
116. Barbhaiya RH, Buch AB, Greene DS. A study of the effect of
sion. CNS Drugs 1996; 389-402
age and gender on the pharmacokinetics of nefazodone after
136. Sandker GW, Vos RME, Delbressine LPC, et al. Metabolism
single and multiple doses. J Clin Psychopharmacol 1996; 16:
of three pharmacologically active drugs in isolated human
19-25 and rat hepatocytes: analysis of interspecies variability and
117. Barbhaiya RH, Shukla UA, Greene DS. Single-dose pharmaco- comparison with metabolism in vivo. Xenobiotica 1994;
kinetics of nefazodone in healthy young and elderly subjects 24: 143-55
and in subjects with renal or hepatic impairment. Eur J Clin 137. Dahl M-L, Voortman G, Alm C, et al. In vitro and in vivo studies
Pharmacol 1995; 49: 221-8 on the disposition of mirtazepine in humans. Clin Drug Invest
118. Barbhaiya RH, Brady ME, Shukla UA, et al. Steady-state phar- 1997; 13: 37-46
macokinetics of nefazodone in subjects with normal and im- 138. Poirier MF, Olié JP, Loo H, et al. Activité antidépressive,
paired renal function. Eur J Clin Pharmacol 1995; 49: 229-35 caractéristiques pharmacocinétiques et proprétés biochimi-
119. Barbhaiya RH, Shukla UA, Natarajan CS, et al. Single- and ques de la cimoxatone, un nouvel IMAO-A réversible.
multiple-dose pharmacokinetics of nefazodone in patients Encéphale 1983; IX: 331-43
with hepatic cirrhosis. Clin Pharmacol Ther 1995; 58: 390-8 139. Fitton A, Faulds D, Goa KL. Moclobemide: a review of its
120. Mocaër E, Rettori MC, Kamoun A. Pharmacological antide- pharmacological properties and therapeutic use in depressive
pressive effects and tianeptine-induced 5-HT uptake increase. illness. Drugs 1992; 43: 561-96
Clin Neuropharmacol 1988; 11 Suppl. 2: S32-42 140. Fulton B, Benfield P. Moclobemide: an update of its pharma-
121. Mennini T, Garattini S. Neurobiology of tianeptine: a new phar- cological properties and therapeutic use. Drugs 1996; 52:
maceutic agent. Presse Med 1991; 20: 1823-7 450-74
122. Ansseau M. The paradox of tianeptine. Eur Psychiatry 1993; 8 141. Volz HP, Gleiter CH, Struck M, et al. Brofaromine: insight into
Suppl. 2: 89S-93S the nature of drug development. CNS Drugs 1995; 3: 1-8
123. Wilde MI, Benfield P. Tianeptine: a review of its pharmacody- 142. Mayersohn M, Guentert TW. Clinical pharmacokinetics of the
namic and pharmacokinetic properties, and therapeutic effi- monoamine oxidase-A inhibitor moclobemide. Clin Phar-
cacy in depression and coexisting anxiety and depression. macokinet 1995; 29: 292-32
Drugs 1995; 49: 411-39 143. Schoerlin MP, Mayersohn M, Korn A, et al. Disposition kinetics
124. Grislain L, Gele P, Bertrand M, et al. The metabolic pathways of moclobemide, a monoamine oxidase-A enzyme inhibitor:
of tianeptine, a new antidepressant, in healthy volunteers. single and multiple dosing in normal subjects. Clin Phar-
Drug Metab Dispos 1990; 18: 804-8 macol Ther 1987; 42: 395-404
125. Oluyomi AO, Datla KP, Curzon G. Effects of the (+) and 144. Schoerlin MP, Horber FF, Frey FJ, et al. Disposition kinet-
ics of moclobemide, a new MAO-A inhibitor, in subjects
(–)-enantiomers of the antidepressant drug tianeptine on 5-
with impaired renal function. J Clin Pharmacol 1990; 30:
HTP-induced behaviour. Neuropharmacology 1997; 36:
272-84
383-7
145. Stoeckel K, Pfefen JP, Mayersohn M, et al. Absorption and
126. Larrey D, Tinel M, Letteron P, et al. Metabolic activation of the
disposition of moclobemide in patients with advanced age or
new tricyclic antidepressant tianeptine by human liver cyto- reduced liver or kidney function. Acta Psychiatr Scand 1990;
chrome P450. Biochem Pharmacol 1990; 40: 545-50 Suppl. 360: 94-97
127. Couet W, Girault J, Latrille F, et al. Kinetic profiles of tianeptine 146. Jauch R, Griesser E, Oesterhelt G, et al. Biotransformation of
and its MC5 metabolite in plasma, blood and brain after single moclobemide in humans. Acta Psychiatr Scand 1990; Suppl.
and chronic intraperitoneal administration in the rat. Eur J 360: 87-90
Drug Metab Pharmacokinet 1990; 15: 69-74 147. Schoerlin MP, Da Prada M. Species-specific biotransformation
128. Carlhant D, Le Garrec J, Guedes Y, et al. Pharmacokinetics and of moclobemide: a comparative study in rats and humans.
bioavailability of tianeptine in the elderly. Drug Invest 1990; Acta Psychiatr Scand 1990; Suppl. 360: 108-10
2: 167-72 148. Da Prada M, Kettler R, Keller HH, et al. Neurochemical profile
129. Demotes-Mainard F, Galley P, Manciet G, et al. Pharmacoki- of moclobemide, a short-acting and reversible inhibitor of
netics of the antidepressant tianeptine at steady state in the monoamine oxidase type A. J Pharmacol Exp Ther 1989; 248:
elderly. J Clin Pharmacol 1991; 31: 174-8 400-8

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)
302 Caccia

149. Fritze J, Laux G, Sofic E, et al. Plasma moclobemide and me- Nicolaidis S, editor. Obesity management and ReduxTM. San
tabolites: lack of correlation with clinical response and bio- Diego: Academic Press, 1997: 65-80
genic amines. Psychopharmacology 1989; 99: 252-6 155. Matzke GR, St Peter WL. Clinical pharmacokinetics 1990. Clin
150. Guentert TW, Gram LF, Grange S, et al. Reversible interaction Pharmacokinet 1990; 18: 1-19
of moclobemide with CYP2D6 and CYP2C9. Clin Pharmacol 156. Bartels, Albert K. Detection of Psychoative drugs using 19F MR
Ther 1994; 55: PI-57 spectroscopy. J Neural Transm Gen Sect 1995; 99: 1-6
151. Gram LF, Brøsen K, and the Danish University Antidepressant 157. Kennedy SH, Javanmard M, Vaccarino FJ. A review of func-
Group. Moclobemide treatment causes a substantial rise in the tional neuroimaging in mood disorders: positron emission
sparteine metabolic ratio. Br J Clin Pharmacol 1993; 35: tomography and depression. Can J Psychiatry 1997; 42:
467-75
649-52
152. Spigset O, Granberg K, Hägg S, et al. Non-linear fluvoxamine
disposition. Br J Clin Pharmacol 1998; 45: 257-63
153. Sperker B, Backman JT, Kroemer HK. The role of β-glucuro-
nidase in drug disposition and drug targeting in humans. Clin Correspondence and reprints: Dr Silvio Caccia, Istituto di
Pharmacokinet 1997; 33: 18-31 Ricerche Farmacologiche ‘Mario Negri’, via Eritrea 62,
154. Campbell B, Dard-Brunelle B, Caccia S. The use of pharmaco- 20157 Milano, Italy.
kinetics in the assessment of dexfenfluramine safety. In: E-mail: caccia@irfmn. mnegri.it

© Adis International Limited. All rights reserved. Clin Pharmacokinet 1998 Apr; 34 (4)

You might also like