Você está na página 1de 20

Clin Pharmacokinet 2005; 44 (6): 571-590

REVIEW ARTICLE 0312-5963/05/0006-0571/$34.95/0

© 2005 Adis Data Information BV. All rights reserved.

Clinical Pharmacokinetics
of Atomoxetine
John-Michael Sauer,1 Barbara J. Ring2 and Jennifer W. Witcher2
1 Elan Pharmaceuticals, Inc., South San Francisco, California, USA
2 Department of Drug Disposition, Lilly Research Laboratories, Eli Lilly and Company,
Indianapolis, Indiana, USA

Contents
Abstract . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 571
1. Overview of Pharmacological Agents Used for the Treatment of Attention-Deficit Hyperactivity
Disorder . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 572
2. Physicochemical Properties . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 574
3. Pharmacokinetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 574
3.1 Overview . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 574
3.2 Absorption and Bioavailability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 576
3.3 Distribution and Protein Binding . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 577
3.4 Metabolism and Excretion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 577
4. Overview of Pharmacodynamic Properties . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 581
4.1 Mechanism of Action . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 581
4.2 In Vitro and In Vivo Activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 581
4.3 Clinical Pharmacodynamics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 582
5. Pharmacokinetic Interactions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 583
5.1 Interactions with Food . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 583
5.2 Interactions with Paroxetine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 583
5.3 Interactions with Desipramine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 584
5.4 Interactions with Midazolam . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 584
6. Implications of Pharmacokinetic Properties for Therapeutic Use . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 585
6.1 Dosages and Therapeutic Range . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 585
6.2 Sex and Racial Differences in Atomoxetine Pharmacokinetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . 585
6.3 Influence of Age and Bodyweight on Atomoxetine Pharmacokinetics . . . . . . . . . . . . . . . . . . . . 585
6.4 Diseases and the Pharmacokinetics of Atomoxetine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 586
6.4.1 Chronic Impairment of Renal Function . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 586
6.4.2 Hepatic Insufficiency . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 586
7. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 587

Abstract Atomoxetine (Strattera®), a potent and selective inhibitor of the presynaptic


norepinephrine transporter, is used clinically for the treatment of attention-deficit
hyperactivity disorder (ADHD) in children, adolescents and adults. Atomoxetine
has high aqueous solubility and biological membrane permeability that facilitates
its rapid and complete absorption after oral administration. Absolute oral bioavai-
lability ranges from 63 to 94%, which is governed by the extent of its first-pass
metabolism. Three oxidative metabolic pathways are involved in the systemic
clearance of atomoxetine: aromatic ring-hydroxylation, benzylic hydroxylation
572 Sauer et al.

and N-demethylation. Aromatic ring-hydroxylation results in the formation of the


primary oxidative metabolite of atomoxetine, 4-hydroxyatomoxetine, which is
subsequently glucuronidated and excreted in urine. The formation of 4-hydroxy-
atomoxetine is primarily mediated by the polymorphically expressed enzyme
cytochrome P450 (CYP) 2D6. This results in two distinct populations of individu-
als: those exhibiting active metabolic capabilities (CYP2D6 extensive metabolis-
ers) and those exhibiting poor metabolic capabilities (CYP2D6 poor metabolisers)
for atomoxetine.
The oral bioavailability and clearance of atomoxetine are influenced by the
activity of CYP2D6; nonetheless, plasma pharmacokinetic parameters are predict-
able in extensive and poor metaboliser patients. After single oral dose, atomoxe-
tine reaches maximum plasma concentration within about 1–2 hours of
administration. In extensive metabolisers, atomoxetine has a plasma half-life of
5.2 hours, while in poor metabolisers, atomoxetine has a plasma half-life of 21.6
hours. The systemic plasma clearance of atomoxetine is 0.35 and 0.03 L/h/kg in
extensive and poor metabolisers, respectively. Correspondingly, the average
steady-state plasma concentrations are approximately 10-fold higher in poor
metabolisers compared with extensive metabolisers. Upon multiple dosing there
is plasma accumulation of atomoxetine in poor metabolisers, but very little
accumulation in extensive metabolisers. The volume of distribution is 0.85 L/kg,
indicating that atomoxetine is distributed in total body water in both extensive and
poor metabolisers. Atomoxetine is highly bound to plasma albumin (approximate-
ly 99% bound in plasma). Although steady-state concentrations of atomoxetine in
poor metabolisers are higher than those in extensive metabolisers following
administration of the same mg/kg/day dosage, the frequency and severity of
adverse events are similar regardless of CYP2D6 phenotype.
Atomoxetine administration does not inhibit or induce the clearance of other
drugs metabolised by CYP enzymes. In extensive metabolisers, potent and
selective CYP2D6 inhibitors reduce atomoxetine clearance; however, administra-
tion of CYP inhibitors to poor metabolisers has no effect on the steady-state
plasma concentrations of atomoxetine.

1. Overview of Pharmacological Agents ing oppositional defiant disorder, conduct disorder,


Used for the Treatment of depression, anxiety disorders and tic disorders.[7,8]
Attention-Deficit Hyperactivity Disorder The exact aetiology of ADHD is unknown, al-
Attention-deficit hyperactivity disorder (ADHD) though neurotransmitter deficits, genetic traits and
is the most common neurobehavioural disorder of prenatal complications have been implicated.[9]
childhood. The incidence of ADHD is 5–10% in Symptoms of ADHD respond to treatment with psy-
children and the symptoms are known to persist into chostimulants. These drugs have been demonstrated
adulthood in 10–60% of cases.[1-5] Behavioural fea- to increase the availability of extracellular dopamine
tures of ADHD include inattention, hyperactivity by blocking dopamine transporters[10] and in some
and impulsivity, which may lead to academic under- cases enhancing dopamine release.[11] Psychostimu-
achievement, poor interpersonal relationships and lants are the most common pharmacological inter-
low self esteem.[6] Additionally, comorbid medical vention used in the treatment of ADHD.[12] A detri-
conditions are often associated with ADHD, includ- mental attribute associated with the drugs in this

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)
Clinical Pharmacokinetics of Atomoxetine 573

class (pemoline, methylphenidate and amphet- clearance for the amphetamines.[18] Definitive
amines) is their potential for abuse.[13] These effects clinical drug interaction studies with CYP2D6 in-
place the psychostimulant drugs as Schedule II or IV hibitors or substrates have not been reported for
(pemoline), putting them under close monitoring by amfetamine. Likewise, the effect of the polymorphic
regulators. As these drugs were developed many expression of CYP2D6 on the metabolism of
years ago, contemporary clinical metabolism and amfetamine has not been reported.
drug interaction studies are lacking or have only Pemoline (Cylert®, Abbott Laboratories, Abbott
recently received description. Markowitz and Pat- Park, IL, USA), unlike the other psychostimulants,
rick[14] provide an authoritative review on the is not a phenethylamine derivative. The risk of
pharmacokinetic and pharmacodynamic interactions
hepatotoxicity has significantly limited the clinical
of psychostimulants with other drugs.
usage of pemoline.[19] The half-life of pemoline is
Among the psychostimulants, methylphenidate approximately 12 hours and it is typically adminis-
in a variety of formulations (Ritalin® 1, Ritalin LA®, tered once daily. The isoforms of CYP responsible
Novartis Pharmaceuticals Corporation, East Hano- for oxidative metabolism of pemoline are unknown.
ver, NJ, USA; Concerta®, Alza Corporation, Moun- Additionally, there are no published reports on the
tain View, CA, USA) is the most widely pre- interaction of pemoline with other drugs.
scribed medication for the treatment of ADHD.
Several other pharmacological interventions in-
Methylphenidate, a dopamine transporter inhibitor,
cluding tricyclic antidepressants and selective sero-
does not appear to be a substrate for transport into
the neuron and it elicits little presynaptic dopamine tonin reuptake inhibitors have been used to treat
release. Methylphenidate is a racemic mixture of ADHD.[20] However, until the recent introduction of
threo-(R,R)-(+)- and threo-(S,S)-(–)-isomers, but atomoxetine (Strattera®; Eli Lilly and Company,
some marketed forms only include a single enanti- Indianapolis, IN, USA), a potent and selective
omer [threo-(R,R)-(+)-isomer]. Methylphenidate is norepinephrine reuptake inhibitor,[21] no new medi-
rapidly cleared from the systemic circulation and cations have been developed for the treatment of this
has a half-life of 2.5 hours. Recently, several modi- disease over the last 3 decades. Atomoxetine in-
fied-release products (Concerta®, Ritalin LA®) creases both dopamine and norepinephrine in the
have been introduced into the market to extend the prefrontal cortex.[22] This is markedly different from
apparent plasma half-life of methylphenidate by al- the actions of psychostimulants like methylpheni-
tering its absorption kinetics.[15,16] date, which increases dopamine in the prefrontal
cortex, as well as the striatum and nucleus accum-
Dexamfetamine, the (S)-(+)-isomer of amfe-
bens.[22] Neurotransmitter changes in the striatum
tamine, as well as mixed isomers (81% [–] and 19%
and nucleus accumbens are postulated to be respon-
[+]) of amfetamine salts have been demonstrated to
sible for the abuse potential associated with psy-
be an effective treatment of ADHD. Amfetamine is
cleared from the systemic circulation by oxidative chostimulants.[13]
metabolism and has a half-life of approximately 12 With a favourable safety profile and novel mech-
hours. Recently, a longer acting formulation, Adder- anism of action, atomoxetine represents an advance
all XR® (Shire Laboratories, Inc., Wayne, PA, for the treatment of ADHD in children, adolescents
USA), was introduced into the market. Adderall and adults.[23-27] In 2003, atomoxetine was launched
XR® alters the pharmacokinetics of the mixed am- in the US and is currently marketed under the brand
phetamine salts by prolonging the absorption kinet- name Strattera®. Atomoxetine is the first nonstimu-
ics.[17] Aromatic oxidation catalysed by cytochrome lant treatment approved by the US FDA for ADHD
P450 (CYP) 2D6 is the primary means of systemic and has the advantage of not being a scheduled drug.

1 The use of trade names is for product identification purposes only and does not imply endorsement.

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)
574 Sauer et al.

2. Physicochemical Properties enzyme (table I and figure 2).[31] The majority of


people (>90%), who metabolise atomoxetine and
Atomoxetine hydrochloride is known chemically other CYP2D6 substrates relatively rapidly, are des-
as (–)-N-methyl-3-phenyl-3-(o-tolyloxy)-propy-
ignated as CYP2D6 extensive metabolisers. These
lamine hydrochloride, or (–)-N-methyl-γ-(2-
individuals possess a range of activities considered
methylphenoxy) benzenepropanamine hydrochlo-
to be normal CYP2D6 activity. Mutations or dele-
ride (figure 1). Atomoxetine is marketed as the R(–)
tion of the CYP2D6 gene results in a minority of
isomer (apparent inhibition constant [Ki] = 1.9
people (up to 7%) who are known as poor
nmol/L), which is approximately 9-fold more potent
metabolisers of CYP2D6 substrates and metabolise
as an inhibitor of the norepinephrine transporter than
atomoxetine relatively slowly. As the clearance of
the S(+) isomer (Ki = 16.8 nmol/L).[21] Compared to
atomoxetine is influenced by its metabolism, it is not
its effect on the norepinephrine transporter, atomox-
surprising that its clearance across patients results in
etine (R(–) isomer) has very little affinity for the
a bimodal distribution indicative of a genetic poly-
dopamine (Ki = 1800 nmol/L) or serotonin (Ki = 750
morphism (figure 3).
nmol/L) transporters.[21] Atomoxetine hydrochloride
(C17H21NO • HCl) has a molecular weight of 291.8 The extensive metaboliser population can be sub-
and a free base molecular weight of 255.4. Atomox- divided into three groups based on the number of
etine hydrochloride is a white to practically white available wild-type alleles, homozygous, heterozy-
solid. Its solubility is 27.8 mg/mL in water and its gous and ultra-rapid metabolisers. Ultra-rapid
dissociation constant (pKa) is 10.13. metabolisers of CYP2D6 substrates result from gene
duplication of functional CYP2D6 alleles.[32] The
3. Pharmacokinetics ultra-rapid metabolisers genotype accounts for ap-
proximately 3–7% of the population.[33] The ultra-
3.1 Overview rapid metaboliser population appears to be compara-
ble to the upper end of the extensive metaboliser
Following oral administration, atomoxetine is range, with a great deal of overlap of apparent
well absorbed. It is primarily cleared from the body plasma clearance (CL/F) values between ultra-rapid
via oxidative metabolism and is subsequently elimi- metaboliser and extensive metaboliser patients (fig-
nated into the urine as conjugated metabolites.[28] ure 3). The extensive metaboliser subpopulations
Similar to many compounds, the biotransformation are not distinguishable from each other on an indi-
of atomoxetine governs its overall disposition in vidual patient basis. Therefore, genotype is more
humans. As a result of the central role of CYP2D6 in useful for differentiating extensive and poor
the metabolism of atomoxetine, the activity of this metabolisers, rather than predicting an individual’s
enzyme plays a significant role in its pharmacoki- precise clearance within the extensive metaboliser
netics.[28-30] range of clearances.
The enzymatic activity of CYP2D6 is determined The plasma pharmacokinetic parameters of ato-
by a genetic polymorphism resulting in two primary
moxetine in extensive and poor metaboliser subjects
populations of individuals with either extensive or
have been reported in a number of publications
poor metabolic capabilities for substrates of this
(table II).[28-30,34-37] In extensive metaboliser individ-
H uals, the absorption is rapid and the maximum plas-
H3C N ma concentration (Cmax) of atomoxetine is 533
ng/mL (coefficient of variance [CV] 32%) after a
O 1 mg/kg dose and occurs at a median time of 1–2
CH3 hours following oral administration.[36] The mean
Fig. 1. Chemical structure of atomoxetine, a selective nore- half-life is 5.2 hours (range 3.7–7.5 hours) with a
pinephrine transporter inhibitor. mean CL/F of 0.35 L/h/kg (intersubject CV

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)
Clinical Pharmacokinetics of Atomoxetine 575

Table I. Noncompartmental pharmacokinetic parameters for atomoxetine and its metabolites in cytochrome P450 (CYP) 2D6 extensive and
poor metabolisers following oral administration of atomoxetine 20mg twice daily (≈0.5 mg/kg/day)[28]
Parameter Arithmetic mean (CV %)
atomoxetine 4-hydroxyatomoxetine N-desmethylatomoxetine 4-hydroxyatomoxetine-
O-glucuronide
CYP2D6 extensive metabolisers
Cmax,ss (ng/mL) 159.70 (51.9) 2.03 (17.5) 7.02 (71.5) 413.88 (35.5)
Cmin,ss (ng/mL) 36.05 (115.8) 0.52 (115.6) 3.12 (113.6) 104.36 (19.3)
Cav,ss (ng/mL) 89.64 (64.3) 5.15 (86.4) 228.70 (13.6)
tmaxa (h) 2.00 (1.00–3.00) 2.50 (2.00–4.00) 3.50 (2.00–6.00) 2.00 (2.00–4.00)
t1/2b (h) 5.34 (3.67–9.09) 8.97 (2.11–21.9) 6.74 (5.90–8.30)
AUCτ (μg • h/mL) 1.08 (64.3) 0.0618 (86.4) 2.74 (13.6)
CLss/F (L/h/kg) 0.373 (75.1)
Vz/F (L/kg) 2.33 (51.0)

CYP2D6 poor metabolisers


Cmax,ss (ng/mL) 914.72 (30.5) 259.22 (39.6) 88.00 (16.9)
Cmin,ss (ng/mL) 502.84 (29.2) 193.09 (40.6) 69.27 (16.4)
Cav,ss (ng/mL) 703.63 (26.9) 234.89 (41.2) 77.88 (17.0)
tmaxa (h) 2.00 (2.00–3.00) 6.00 (3.00–6.00) 4.00 (2.00–6.00)
t1/2b (h) 20.0 (16.8–25.2) 33.3 (27.7–42.7) 19.0 (15.2–22.8)
AUCτ (μg • h/mL) 8.44 (26.9) 2.82 (41.2) 0.935 (17.0)
CLss/F (L/h/kg) 0.0357 (26.2)
Vz/F (L/kg) 1.06 (42.9)
a Median (range).
b Mean (range).
AUCτ = area under the plasma concentration-time curve during a dosage interval (τ) at steady state; CLss/F = apparent plasma clearance at
steady-state; Cav,ss = average steady-state drug concentration in plasma; Cmax,ss = maximum (peak) steady-state drug concentration in the
plasma; Cmin,ss = minimum (trough) steady-state drug concentration in the plasma; CV = coefficient of variance; t1/2 = half-life; tmax = time to
maximum plasma concentration; Vz/F = apparent volume of distribution during terminal phase.

56%).[37] The volume of distribution is 0.85 L/kg morphism associated with CYP2D6, the phenotypic
(CV 16%) after an intravenous dose, indicating that manifestation of this condition can result from expo-
atomoxetine is distributed in total body water in sure to potent inhibitors of this metabolic pathway.
both extensive and poor metabolisers.[37] With a In individuals lacking CYP2D6 activity, oxidative
short half-life and rapid clearance, accumulation metabolism is still the primary route of atomoxetine
after twice-daily administration is minimal (mean clearance, albeit at a much slower rate than that
9%, range 5–15%) with a mean plasma fluctuation observed in extensive metabolisers. In poor
of 273%.[36] The mean maximum steady-state plas- metabolisers, the mean Cmax after a single 1 mg/kg
ma concentration (Cmax,ss) is 584 ng/mL (CV 46%) dose is approximately 2-fold higher than Cmax for
after 1 mg/kg twice-daily administration and, when extensive metabolisers. The mean half-life in poor
compared with Cmax following a single dose, indi- metabolisers is considerably longer than in exten-
cates minimal accumulation.[36] sive metabolisers (21.6 hours, range 14.1–26.8
The poor metaboliser trait, inherited as an autoso- hours) and the CL/F (0.03 L/h/kg, intersubject CV
mal recessive characteristic, is an important source 19%) is about one-tenth of the extensive metabolis-
of intersubject variability in metabolism for a num- ers’ value.[37] The mean apparent volume of distribu-
ber of drugs metabolised through CYP2D6.[38,39] tion (1.06 L/kg, CV 43%) is about half the extensive
Although the inability to metabolise CYP2D6 sub- metabolisers’ value, likely due to differences in first
strates is principally mediated by the genetic poly- pass metabolism.[28] The average steady-state plas-

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)
576 Sauer et al.

Atomoxetine
N-desmethylatomoxetine
4-Hydroxyatomoxetine
4-Hydroxyatomoxetine-O-glucuronide

a b

1000

800

600

400
Plasma concentration (μg/L)

200

1000

100

10

0.1
−12 0 12 24 36 48 60 72 0 24 48 72 96 120 144 168 192 216
Time (h)
Fig. 2. Mean plasma concentration-time profiles (cartesian [upper panel] and semi-log [lower panel]) for cytochrome P450 2D6 extensive
metabolisers (a) and poor metabolisers (b). Multiple 20mg doses of atomoxetine were administered twice daily for 6 days.

ma concentration (Cav,ss) is about 10-fold higher tween extensive and poor metabolisers are princi-
than observed in extensive metabolisers and is readi- pally mediated by differences in first-pass metabo-
ly predictable from half-life and dosing interval. lism and clearance rather than being driven by dif-
Although Cav,ss is approximately 10-fold higher in ferences in absorption of atomoxetine.
poor metabolisers compared with extensive
metabolisers, at the Cmax,ss the difference is only 3.2 Absorption and Bioavailability
about 5-fold.[37] For example, following 6 days of
As a result of its high water solubility, as well as
20mg twice-daily administration of atomoxetine its favourable dissolution and intestinal permeability
(≈0.5 mg/kg/day) the mean Cav,ss was 90 ng/mL characteristics, atomoxetine is rapidly absorbed fol-
(CV 64%) in extensive metabolisers and 704 ng/mL lowing oral administration.[28] The absolute oral
(CV 27%) in poor metabolisers.[28] The mean bioavailability of atomoxetine is 94% (90% CI 0.88,
Cmax,ss was 160 ng/mL (CV 52%) in extensive 0.99) in poor metabolisers and 63% (90% CI 0.59,
metabolisers and 915 ng/mL (CV 31%) in poor 0.67) in extensive metabolisers, indicating nearly
metabolisers.[28] The dissimilarities observed be- complete absorption with higher first-pass metabo-

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)
Clinical Pharmacokinetics of Atomoxetine 577

lism in extensive metabolisers.[40] Food does not 3.4 Metabolism and Excretion
affect the extent of atomoxetine absorption but does
decrease Cmax (37% with a standard high-fat break- As with many compounds, the rate of metabolism
fast and 9% with a more typical meal) and delays of atomoxetine governs its overall pharmacokinetic
time to reach maximum plasma concentration (tmax) profile in humans.[28] Atomoxetine is primarily
by 3 hours.[37,40] cleared from the body by oxidative metabolism and
nearly all of its metabolites are eliminated by excre-
tion into urine. Three primary biotransformation
3.3 Distribution and Protein Binding reactions direct the overall metabolism of atomoxe-
tine: aromatic ring-hydroxylation, benzylic hydrox-
The tissue and organ distribution of atomoxetine ylation and N-demethylation. For most of the prima-
was evaluated in rats following an oral dose (50 ry metabolites, secondary aromatic oxidation is also
mg/kg) of radiolabelled atomoxetine.[41] Peak con- observed. Conjugation of hydroxylated metabolites
centrations of radiocarbon occurred at 1 hour after by uridine diphosphate glucuronosyltransferase is
administration in nearly all tissues. By 8 hours after the only conjugation (phase II) metabolic pathway
administration, radiocarbon associated with most to participate in the biotransformation of atomoxe-
tissues, including the brain, had declined to either tine. Figure 4 illustrates the overall biotransforma-
low or background levels. tion of atomoxetine in humans.
In humans, regardless of CYP2D6 status, atom- Aromatic ring-hydroxylation forms the primary
oxetine is well distributed and its volume of distri- oxidative metabolite of atomoxetine, 4-hydroxyato-
bution is equivalent to total body water at 0.85 moxetine. This metabolite is subsequently glu-
L/kg.[37] Atomoxetine and its metabolites undergo curonidated and excreted in urine.[28] Biotrans-
only limited partitioning into human red blood formation of atomoxetine to 4-hydroxyatomoxetine
cells.[28] In plasma, atomoxetine is highly protein- is primarily mediated by CYP2D6.[43] Therefore, the
bound (98.7% bound in plasma) primarily to albu- overall pharmacokinetic parameters of atomoxetine
min.[28] The plasma protein binding of N-desmethy- are influenced by polymorphic expression of this
latomoxetine (99.1% bound) is similar to atomoxe- enzyme. In individuals lacking CYP2D6 activity, 4-
tine, while the binding of 4-hydroxyatomoxetine to hydroxyatomoxetine is still the major oxidative me-
plasma protein (66.6% bound) is substantially less tabolite. This fact demonstrates that the metabolite
than atomoxetine.[28] In vitro drug-displacement can also be formed by several isoforms of CYP,
studies demonstrated that atomoxetine did not affect albeit at a much lower efficiency than CYP2D6. The
the binding of warfarin, aspirin (acetylsalicylic ac-
80 PM
id), phenytoin and diazepam to human albumin.[37] EM
Similarly, these compounds did not affect the bind- UM
60
ing of atomoxetine to human albumin.
No. of patients

The ability of atomoxetine and its metabolites to


40
cross the placenta was examined in gravid rats fol-
lowing an oral dose (50 mg/kg) of radiolabelled
20
atomoxetine on gestational day 18.[42] Atomoxetine
and/or its metabolites crossed the placenta. None-
0
theless, fetal tissue exposure was substantially less
than that observed in maternal tissues. The distribu- 0 1 2 3 4 5
Ln (CL/F) [L/h/kg]
tion of radioactivity in milk was investigated in
Fig. 3. Frequency distribution of individual atomoxetine clearance
lactating rats. Only a small amount of atomoxetine values based on cytochrome P450 2D6 genotype. EM = extensive
and/or its metabolites were present in rat milk (<1% metaboliser; Ln (CL/F) = log transformed apparent plasma clear-
of the dose). ance; PM = poor metaboliser; UM = ultra-rapid metaboliser.

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)
Table II. Summary of atomoxetine pharmacokinetic parametersa

Dosage No. of subjects Age group tmax Cmax or Cmax,ss t1/2 CL/F Vz/F Reference
578
(h) (ng/mL) (h) (L/h/kg) (L/kg)
Healthy CYP2D6 extensive metabolisers (single dose)

10mg PO 7 Paediatric 2 144 3.1 0.46 2.0 36

20mg PO 10 Adult 1 142 4.3 0.51 2.7 34

Healthy CYP2D6 extensive metabolisers (multiple dose)

20mg PO, bid 4 Adult 2 160 5.3 0.37 2.3 35

20mg PO, bid 21 Adult 1 184 4.0 0.40 2.2 30

20–45mg PO, bid 16 Paediatric 1.7 537 3.3 0.48 2.3 36

© 2005 Adis Data Information BV. All rights reserved.


40mg PO, bid 6 Adult 1 552 4.0 0.33 1.5 35

60mg PO, bid 15 Adult 1 591 3.4 0.40 1.8 35

Healthy CYP2D6 poor metabolisers (multiple dose)

20mg PO, bid 3 Adult 2 915 20 0.04 1.1 28

60mg PO, bid 6 Adult 4 2610 0.04 35

Healthy CYP2D6 extensive metabolisers (multiple dose) with CYP2D6 inhibitor

20mg PO, bid 14 Adult 1.5 690 11 0.06 0.8 30

CYP2D6 extensive metabolisers with hepatic impairment (single dose)

20mg POb 6 Adult 3 116 11 0.21 3.3 34

20mg POc 4 Adult 6 126 16 0.16 2.7 34

a The study by Farid et al.[29] contains compartmental analysis for atomoxetine in extensive and poor metabolisers, which is not shown in this table as different parameters
were calculated.

b Child-Pugh B.

c Child-Pugh C.

bid = twice daily; CL/F = apparent plasma clearance; Cmax = maximum observed plasma concentration; Cmax,ss = maximum (peak) steady-state drug concentration in the plasma;
CYP = cytochrome P450; PO = oral; t1/2 = half-life; tmax = time to reach Cmax; Vz/F = apparent volume of distribution during terminal phase.

Clin Pharmacokinet 2005; 44 (6)


Sauer et al.
Clinical Pharmacokinetics of Atomoxetine 579

average Michaelis-Menten constant (Km) for 4- and poor metabolisers.[28] Although the overall me-
hydroxyatomoxetine formation in human liver mi- tabolism of atomoxetine is similar regardless of
crosomes containing a full complement of CYP was CYP2D6 status, quantitative amounts of metabolites
2.3 μmol/L, and in CYP2D6-deficient microsomes formed and the rate of their formation are different
was 149 μmol/L. In addition, the estimated intrinsic between extensive and poor metabolisers. Radio-
clearance (CLint) for 4-hydroxyatomoxetine forma- labelled atomoxetine has been given to extensive
tion was 200-fold higher for the microsomes with a and poor metabolisers. The mean Cmax of radioac-
full complement of CYPs than that calculated for the tivity is essentially the same in both extensive and
CYP2D6-deficient microsomes (103 versus 0.2 μL/ poor metabolisers, but the area under the plasma
min/mg, respectively).[43] concentration-time curve from zero to infinity
Atomoxetine undergoes the same biotransforma- (AUC∞) was larger in poor metabolisers because of
tion regardless of CYP2D6 activity, with no pheno- the slower metabolism and elimination. The mean
type-specific metabolites being formed in extensive half-life of atomoxetine-derived radioequivalents is
HO2C
H2N H2N O
CYP2C19 (CYP2D6) OH UGT H2N OH
O OH
HO
O O
O
CH3 CH3
CH3
N-desmethylatomoxetine N-desmethyl-4-hydroxyatomoxetine N-desmethyl-4-hydroxyatomoxetine-O-glucuronide

HO2C
H H
H3C N H3 C N H O
CYP2D6 UGT H3C N OH
OH
O OH
HO
O O
O
CH3 CH3
CH3
Atomoxetine 4-Hydroxyatomoxetine 4-Hydroxyatomoxetine-O-glucuronide

H
H H3C N
H3 C N
UGT HO2C
O O
O OH
C O OH
CH2OH H2 HO
2-Hydroxymethylatomoxetine 2-Hydroxymethylatomoxetine-O-glucuronide

H H H
H3C N H3C N H3C N HO2C
OH OH
UGT O
OH
OH
O O O HO
COOH COOH COOH
2-Carboxyatomoxetine Hydroxy-2-carboxyatomoxetine Hydroxy-2-carboxyatomoxetine-O-glucuronide

H H HO2C
H3C N H3C N
OH OH O
UGT OH
OH
O O HO
CH2OH CH2OH
Dihydroxyatomoxetine Dihydroxyatomoxetine-O-glucuronide

Fig. 4. Metabolic biotransformation of atomoxetine. For hydroxy-2-carboxyatomoxetine, dihydroxyatomoxetine and their respective glucuro-
nide conjugates, multiple sites of aromatic hydroxylation were observed. Although only cytochrome P450 (CYP) 2D6 and CYP2C19 are
shown for the formation of 4-hydroxyatomoxetine and N-desmethylatomoxetine, respectively, several other isoforms of CYP (with reduced
affinity) can form these metabolites. UGT = uridine diphosphate glucuronosyltransferase.

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)
580 Sauer et al.

18 hours in extensive metabolisers and 62 hours in microsomal CYP2D6 content.[43] These studies indi-
poor metabolisers. cated that CYP2C19 is the primary enzyme respon-
In extensive metabolisers, the majority of the sible for N-desmethylatomoxetine formation. How-
radioactive dose is excreted within the first 24 hours ever, because of its minor role in atomoxetine clear-
with >96% of the dose excreted in the urine.[28] ance it was predicted that perturbations in this route
Excretion rate is slower in poor metabolisers and the of metabolism would not influence the overall clear-
majority of the radioactive dose is excreted within ance of atomoxetine. Thus, the higher plasma con-
72 hours. Only 80% of total radioactivity is excreted centrations of N-desmethylatomoxetine in poor
in the urine of poor metabolisers and the remaining metabolisers are not due to enhanced formation of
20% is excreted in faeces. The difference between N-desmethylatomoxetine, but rather due to a parallel
the amount of metabolites formed in the extensive decrease in the systemic clearance of this metabo-
and poor metaboliser patient populations is reflected lite. Elimination of N-desmethylatomoxetine re-
by the relative formation of 4-hydroxyatomoxetine. quires secondary oxidation (aromatic hydroxyl-
The fraction of the dose excreted in the urine and ation) that appears to be primarily mediated by
faeces as 4-hydroxyatomoxetine and 4-hydroxy- CYP2D6. Because this enzyme is functionally ab-
atomoxetine-O-glucuronide is greater in the exten- sent in poor metabolisers, similar to atomoxetine,
sive metabolisers (approximately 86%) than the there is an accumulation of N-desmethylatomoxe-
poor metabolisers (approximately 40%). Although tine to higher steady-state concentrations.
only 40% of the excreted dose is 4-hydroxyatomox- Thus, differences in atomoxetine concentrations
etine-O-glucuronide in poor metabolisers, this prod- between extensive and poor metabolisers are attrib-
uct represents the most abundant metabolite. In poor uted to a decrease in the rate of formation of 4-
metabolisers, less prominent routes of biotrans- hydroxyatomoxetine, and subsequent 4-hydroxy-
formation such as N-desmethylatomoxetine- and 2- atomoxetine-O-glucuronide. These differences re-
hydroxymethylatomoxetine-derived metabolites are sult in a reduction in the overall rate of elimination
increased. Regardless of CYP2D6 metabolic status, of atomoxetine in poor metabolisers.
very little atomoxetine (<3%) was excreted into the
In vitro enzyme studies of the potential for
urine unchanged, indicating a relatively minor role
atomoxetine and its two primary oxidative metabo-
for direct renal elimination.
lites, N-desmethylatomoxetine and 4-hydroxy-
Atomoxetine and 4-hydroxyatomoxetine-O- atomoxetine, to inhibit CYP1A2, CYP2C9,
glucuronide are the principle circulating species in CYP2D6 and CYP3A were conducted in human
the plasma of extensive metabolisers, while atomox- hepatic microsomes.[35] These compounds exhibit
etine and N-desmethylatomoxetine are the principle very little inhibition of CYP2C9 (diclofenac 4′-
circulating species in poor metabolisers (figure hydroxylation) or CYP1A2 (phenacetin O-deethyla-
2).[28] Although N-desmethylatomoxetine is a major tion) enzymatic activity. Although apparent Ki val-
circulating metabolite in poor metabolisers, the con- ues could not be determined for atomoxetine or 4-
tribution of the metabolic pathway to the overall hydroxyatomoxetine, the formation of 4′-hydroxy
metabolism of atomoxetine is relatively minor (ap- diclofenac was inhibited by approximately 37% by
proximately 6% of the total dose). Extensive atomoxetine at concentrations of 800 μmol/L, and
metabolisers have relatively low plasma concentra- 34% by 4-hydroxyatomoxetine at concentrations of
tions of N-desmethylatomoxetine; nonetheless, the 500 μmol/L. The formation of paracetamol (ac-
formation amounts of this metabolite are only slight- etaminophen) was inhibited by approximately 30%
ly smaller than the amount formed in poor by atomoxetine at concentrations of 800 μmol/L and
metabolisers. Human microsomal studies provide an 10% by 4-hydroxyatomoxetine at concentrations of
average apparent Km value of 83 μmol/L for the 500 μmol/L. For reference, the estimated maximum
formation of N-desmethylatomoxetine regardless of plasma concentrations of atomoxetine and its me-

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)
Clinical Pharmacokinetics of Atomoxetine 581

tabolites following 1.0 mg/kg twice-daily doses The ability of atomoxetine to induce catalytic
(2.0 mg/kg/day) of atomoxetine hydrochloride are activities associated with CYP1A2 and CYP3A was
12.9 μmol/L for atomoxetine, 6.3 μmol/L for N- studied in primary cultures of human hepatocytes.
desmethylatomoxetine and 0.03 μmol/L for 4-hy- Atomoxetine had no effect on the catalytic activities
droxyatomoxetine. N-desmethylatomoxetine inhib- of 7-ethoxyresorufin O-deethylation (CYP1A2) or
its CYP2C9 and CYP1A2 enzymatic activity at high midazolam 1′-hydroxylation (CYP3A).[35]
concentrations with Ki values of 53 μmol/L and
4. Overview of
271 μmol/L, respectively. Thus, the likelihood of in
Pharmacodynamic Properties
vivo inhibition of CYP2C9 and CYP1A2 is very
low.
4.1 Mechanism of Action
Both atomoxetine and N-desmethylatomoxetine
inhibit CYP3A (midazolam 1′-hydroxylation) en- Atomoxetine is a potent inhibitor of the presy-
zyme activity and yield Ki values of 34 μmol/L and naptic norepinephrine transporter with minimal af-
16 μmol/L, respectively. In addition, atomoxetine, finity for other monoamine transporters or recep-
N-desmethylatomoxetine and 4-hydroxyatomoxe- tors.[21,45-47] Furthermore, as a result of its novel
tine significantly inhibit CYP2D6 (bufuralol 1′-hy- mechanism of action, atomoxetine does not share
droxylation) enzyme activity and yield Ki values of the abuse potential associated with psychostimulant
3.6 μmol/L, 5.3 μmol/L and 17 μmol/L, respective- drugs.[48]
ly.
4.2 In Vitro and In Vivo Activity
These in vitro data predict the possible effect of
atomoxetine on the clearance of coadministered In vitro studies demonstrate that atomoxetine has
drugs. The relationship (I/[I + Ki]) • 100, where I is potent nanomolar to subnanomolar affinity for the
inhibitor concentration, can be used to calculate an rat and human norepinephrine transporters and min-
estimate of expected percent inhibition.[44] Differ- imal affinity for serotonin and dopamine transport-
ences in accumulation and overall exposure profiles ers, as well as other receptors and ion channels.[21]
The human transporter affinities of the primary oxi-
between extensive and poor metaboliser populations
dative metabolites of atomoxetine, 4-hydroxy-
were considered in the interpretation of the in vitro
atomoxetine and N-desmethylatomoxetine have
findings. A conservative approach was used to esti-
been assessed. 4-Hydroxyatomoxetine has similar
mate inhibitor concentration in these calculations,
affinity for the norepinephrine transporter (Ki = 12.5
which assumes that all of the drug measured in the
μmol/L) to that of atomoxetine (Ki = 8.5 μmol/L),
plasma is potentially an inhibitor. As such, the cal- and little affinity for the dopamine transporter (Ki
culations did not take into account the high plasma >1000 μmol/L). Unlike atomoxetine (Ki >1000
protein binding of either atomoxetine or N- μmol/L), 4-hydroxyatomoxetine (Ki = 11.3 μmol/L)
desmethylatomoxetine. Significant enzyme inhibi- has relatively high affinity for the human serotonin
tion was predicted for CYP3A (56% predicted inhi- transporter. N-desmethylatomoxetine, on the other
bition in poor metaboliser patients) and CYP2D6 hand, had less affinity for the monoamine transport-
(60% predicted inhibition in extensive metaboliser er system compared with atomoxetine and 4-hy-
patients) for typical therapeutic plasma concentra- droxyatomoxetine. As with atomoxetine, these me-
tions. On the basis of these in vitro findings, drug tabolites do not have affinity for other neuronal
interaction studies in healthy subjects were conduct- receptors or ion channels.[49]
ed using probe substrates for CYP2D6 (de- A number of preclinical in vivo studies defined
sipramine) in CYP2D6 extensive metaboliser sub- the central and peripheral potency of atomoxetine as
jects and CYP3A (midazolam) in CYP2D6 poor an inhibitor of norepinephrine reuptake sites.[45,46,50]
metaboliser subjects (see sections 5.3 and 5.4). In rats, orally administered atomoxetine inhibits

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)
582 Sauer et al.

radioligand binding to the norepinephrine transport- tine is an effective and well tolerated treatment for
er with a dose that produces 50% response (ED50) of paediatric, adolescent and adult patients with
2.4 mg/kg, while a small amount (30%) of inhibition ADHD. In children and adolescents, atomoxetine in
of the serotonin transporter is observed at the higher a single or divided daily dose of 1.2 mg/kg has been
doses of atomoxetine (60 mg/kg). Norepinephrine shown to be superior to placebo in randomised,
transporter inhibition was maximal 1 hour after ad- placebo-controlled studies.[25,26] Efficacy was as-
ministration and yet still significant for up to 6 hours sessed by ADHD Rating Scale-IV-Parent Version:
after administration. Interestingly, the half-life of Investigator Administered and Scored (ADHDRS-
atomoxetine in rats is approximately 2 hours,[51] IV-Parent:Inv). Effectiveness of atomoxetine in im-
suggesting a difference between the pharmacody- proving ADHD symptoms is comparable to treat-
namic and pharmacokinetic half-lives of atomoxe- ment with methylphenidate.[27]
tine.
In adult patients with ADHD, atomoxetine effi-
Microdialysis measurement of brain extracellular cacy was initially evaluated in a small, double-blind,
monoamines was utilised to evaluate the central placebo-controlled, crossover study.[23] Atomoxe-
effects of atomoxetine.[22] In these studies, atomoxe- tine, at an average dose of 76 mg/day, was well
tine increased extracellular norepinephrine in the
tolerated. Improvement in ADHD symptomatology
prefrontal cortex 3-fold, but did not alter serotonin.
was significant overall and sufficiently robust to be
Atomoxetine also increased dopamine concentra-
detectable in a parallel-group comparison. Eleven of
tions in the prefrontal cortex 3-fold, but did not alter
21 patients showed improvement after receiving
dopamine in striatum or nucleus accumbens. In con-
atomoxetine, while only 2 of 21 patients improved
trast, methylphenidate increased norepinephrine and
after receiving placebo. These results demonstrated
dopamine equally in prefrontal cortex, and also in-
that atomoxetine was effective in treating adult
creased dopamine in the striatum and nucleus ac-
ADHD, resulting in clinically and statistically sig-
cumbens. The expression of the neuronal activity
nificant improvement in ADHD symptoms, and was
marker Fos was increased 3.7-fold in prefrontal
well tolerated. A number of additional studies have
cortex by atomoxetine administration, but was not
been conducted demonstrating the effectiveness of
increased in the striatum or nucleus accumbens,
atomoxetine treatment in adults with ADHD.[24]
consistent with the regional increases of dopamine.
Bymaster et al.[22] concluded that due to the promis- The safety, tolerability and efficacy of atomoxe-
cuous transport of dopamine via the norepinephrine tine in children and adolescents with ADHD have
transporter in the prefrontal cortex, atomoxetine se- been rigorously characterised in comparison to the
lectively increases both dopamine and nore- other drugs used to treat this disease.[25,53] The most
pinephrine neurotransmission in a region of the common drug-related adverse event reported was
brain involved in attention and memory. In contrast decreased appetite and an initial period of weight
to methylphenidate, atomoxetine does not increase loss followed by an apparently normal rate of weight
dopamine in striatum or nucleus accumbens, sug- gain. There are no effects on the corrected QT
gesting it would have less potential for motoric or interval, and CYP2D6 phenotype of patients does
drug abuse liabilities. This conclusion is supported not affect the overall safety and tolerability of the
by the finding that atomoxetine did not substitute drug. However, tachycardia, as well as increases in
appreciably for metamfetamine in drug discrimina- diastolic blood pressure (2–5mm Hg) and systolic
tion studies in monkeys.[52] blood pressure (3mm Hg) have been observed fol-
lowing treatment with atomoxetine.[54-56] No serious
4.3 Clinical Pharmacodynamics adverse events have been associated with atomoxe-
tine administration, and there have been few discon-
The results of prospective, placebo-controlled tinuations resulting from adverse events. The safety
and dose-finding studies demonstrate that atomoxe- and tolerability associated with atomoxetine admin-

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)
Clinical Pharmacokinetics of Atomoxetine 583

istration in children are similar to those previously pears that an increase in gastric pH had no substan-
reported for adults.[23,57,58] tial effect on the absorption of atomoxetine.[40]
Since individuals lacking CYP2D6 activity have
higher atomoxetine plasma concentrations after 5.2 Interactions with Paroxetine
multiple doses, it was initially believed that poor
metabolisers would not tolerate higher concentra- The role of CYP2D6 in the biotransformation of
tions. Moreover, there was concern that poor atomoxetine[28,29,43] and the potential for interaction
metaboliser patients would need to be phenotypical- with other drugs[30,35] make it important to assess the
ly or genotypically identified before the initiation of impact of concomitant drug treatment. Although the
treatment with atomoxetine. In early clinical trials, systemic clearance of atomoxetine is dependent
patients were given doses in accordance with their upon the polymorphic expression of CYP2D6,
genotype and poor metabolisers received lower chemical inhibitors of this enzymatic pathway also
doses than extensive metabolisers.[25,55] After dem- alter the pharmacokinetics of atomoxetine. In
onstrating safety in initial safety and tolerability humans, coadministration of paroxetine, a potent
studies in poor metabolisers, atomoxetine was ad- inhibitor of CYP2D6-catalysed reactions,[60,61] with
ministered without regard for genotype in later known CYP2D6 substrates (such as metoprolol,
clinical trials.[59] A comparison of 1290 extensive desipramine or perphenazine) result in signifi-
metabolisers with 67 poor metabolisers taking at cant pharmacokinetic interactions in extensive
least 1.2 mg/kg/day of atomoxetine, which is the metabolisers.[62-65] Although paroxetine substantial-
initial target dose for efficacy, showed little differ- ly inhibits CYP2D6, it does not seem to affect
ence in discontinuations or reporting rates of ad- CYP1A2, CYP2C9, CYP2C19 and CYP3A4 en-
verse events.[53,59] However, poor metabolisers did zyme activities in vivo at therapeutic dose.[66-71]
develop a slightly higher increase in heart rate (10.6 Administration of paroxetine (20mg once daily)
bpm versus 6.9 bpm) and lost slightly more weight for 17 days results in steady-state plasma concentra-
(–1.2kg versus +0.8kg) than extensive metabolisers. tions that are in the same range as its Ki for CYP2D6
Thus, with the apparently large therapeutic index, (0.15 μmol/L). Consequently, concomitant adminis-
differential dosing based on genotype is not re- tration of paroxetine and atomoxetine (20mg twice
quired. daily) led to an increase in the plasma concentra-
tions of atomoxetine and its N-demethylated metab-
5. Pharmacokinetic Interactions olite.[30] Paroxetine increased mean Cmax,ss and
AUC during a dosage interval (τ) at steady state
(AUCτ) values of atomoxetine by about 3.5- and
5.1 Interactions with Food 6.5-fold, respectively, which is in the same order of
magnitude as the reported effects of paroxetine on
Food does not affect the extent of atomoxetine other CYP2D6 substrates.[62-64] Correspondingly,
absorption. However, ingestion of the atomoxetine the concentration of 4-hydroxyatomoxetine, the ma-
dose with food decreases Cmax by 37% with a stan- jor CYP2D6 oxidative metabolite, was lower.
dard high-fat breakfast or by 9% with a more typical After concomitant treatment with paroxetine,
meal. Food also delays tmax by 3 hours.[37,40] The steady-state pharmacokinetic parameters of atom-
effect of food on atomoxetine pharmacokinetics is oxetine in extensive metabolisers are similar to
regarded as not clinically important given the small the pharmacokinetic values obtained for poor
decrease in Cmax observed in clinical efficacy stud- metabolisers who were administered atomoxetine
ies. The relative oral bioavailability of atomoxetine alone.[28,29] For example, the CL/F of atomoxetine
administered in the presence of either Maalox® averaged 0.0599 L/h/kg after coadministration with
(Novartis Consumer Health, Parsippany, NJ, USA) paroxetine, which is comparable to that observed in
or omeprazole was approximately 100% and it ap- poor metabolisers (CL/F = 0.03 L/h/kg).[37] In-

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)
584 Sauer et al.

creases in N-desmethylatomoxetine concentrations Effects of atomoxetine on desipramine metabo-


after concomitant treatment with paroxetine are also lism were not studied in poor metabolisers because
consistent with the pharmacokinetics of atomoxe- of the lack of CYP2D6 activity in this population.
tine in poor metabolisers. Thus, coadministration of
paroxetine and atomoxetine results in an inhibition 5.4 Interactions with Midazolam
of CYP2D6-mediated conversion of atomoxetine to
4-hydroxyatomoxetine, and produces an atomoxe- In vitro enzyme inhibition studies suggest that
tine pharmacokinetic profile similar to that found in atomoxetine may impact the pharmacokinetics of
poor metabolisers. agents metabolised by CYP3A, but only when
atomoxetine and N-desmethylatomoxetine plasma
concentrations are relatively high (see section
5.3 Interactions with Desipramine 3.4).[35] These are potential conditions for poor
metaboliser patients. Therapeutic doses of atomoxe-
In vitro inhibition studies designed to evaluate tine in poor metabolisers may achieve concentra-
the 1′-hydroxylation of bufuralol suggest that atom- tions that approach the Ki values for inhibition of
oxetine has the potential to inhibit CYP2D6-mediat- CYP3A. The in vitro predictions indicate that both
ed metabolism (see section 3.4).[35] The in vitro atomoxetine (28% predicted inhibition) and N-
predictions indicate that atomoxetine (54% predict- desmethylatomoxetine (28% predicted inhibition)
ed inhibition) but not its metabolites (5% predicted could participate in CYP3A inhibition. These find-
inhibition) would be primarily responsible for inhi- ings led to a clinical study evaluating the coadminis-
bition. These in vitro findings led to an in vivo study tration of atomoxetine and midazolam in healthy
in healthy CYP2D6 extensive metabolisers evaluat- CYP2D6 poor metabolisers.[35,74] Midazolam is a
ing the coadministration of atomoxetine and de- sensitive and selective probe drug for CYP3A-
sipramine.[35,72] Desipramine is a sensitive and se- dependent metabolism.[75] Twice-daily 60mg doses
lective probe drug for CYP2D6-dependent metabo- of atomoxetine ranged from 1.5 to 2.7 mg/kg/day,
lism; its biotransformation to 2-hydroxydesipramine with a median dose of 1.7 mg/kg/day. At these
doses, Cmax,ss of atomoxetine ranged from 1456 to
is almost exclusively mediated by CYP2D6 and is
3319 ng/mL (5.6–12.9 μmol/L) and N-desmethy-
not affected by inhibitors of other isoforms of
latomoxetine ranged from 755 to 2485 ng/mL
CYP.[73] Twice-daily 60mg doses of atomoxetine
(3.2–10.4 μmol/L), well below their respective pre-
ranged from 1.2 to 2.3 mg/kg/day, with a median
dicted Ki values for CYP3A (34 μmol/L [8757 ng/
dose of 1.6 mg/kg/day. At these doses, Cmax,ss of
mL] and 16 μmol/L [3837 ng/mL], respectively).[35]
atomoxetine ranged from 241 to 1046 ng/mL Following a single 5mg oral dose of midazolam in
(0.9–4.1 μmol/L) and these values are in the range healthy CYP2D6 poor metabolisers, the midazolam
of and exceed the estimated Ki values of atomoxe- Cmax and AUC∞ were about 16% larger when
tine for CYP2D6 (Ki = 3.6 μmol/L [919 ng/ midazolam was concomitantly administered with
mL]).[35] However, because of the rapid elimination atomoxetine; however, the increase was not statisti-
of atomoxetine, the Cav,ss are below the Ki value cally significant or regarded as a change that would
(Cav,ss range 105–569 ng/mL [0.4–2.2 μmol/L]). reflect a clinically important inhibition of CYP3A
Upon the concomitant administration of a single activity.
50mg oral dose of desipramine with atomoxetine, no Coadministration of the potent CYP3A inhibitor
change in desipramine pharmacokinetics was ob- ketoconazole with midazolam results in a 15-fold
served. Thus, in clinical practice, atomoxetine at increase in midazolam AUC and a 4-fold increase in
maximum steady-state conditions is not likely to Cmax.[76] Furthermore, midazolam half-life increases
inhibit the metabolic clearance of drugs metabolised from 2.9 hours to 7.0 hours. Unlike the interaction
by CYP2D6. observed with ketoconazole, atomoxetine did not

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)
Clinical Pharmacokinetics of Atomoxetine 585

alter the half-life of midazolam or increase midazo- improvements in efficacy are similar to those ob-
lam exposure statistically.[35] Yuan et al.,[77] demon- served between atomoxetine dose and efficacy.
strated that moderate inhibitors of CYP3A, such as
grapefruit juice, cause pharmacokinetic changes in 6.2 Sex and Racial Differences in
midazolam (increased Cmax and AUC) that are sub- Atomoxetine Pharmacokinetics
stantially greater than those observed for atomoxe-
In general, the plasma pharmacokinetic parame-
tine. Because there are only minimal changes in
ters and biotransformation of atomoxetine are not
midazolam pharmacokinetics observed, it is predict-
affected by sex or race.[37] However, as described
ed that atomoxetine at maximum recommended
earlier, the disposition of atomoxetine is dependent
clinical doses will not inhibit the metabolic clear-
upon CYP2D6 activity, a trait that differs among
ance of drugs metabolised by CYP3A.
races. For example, the poor metaboliser trait, inher-
ited as an autosomal recessive characteristic, is most
6. Implications of Pharmacokinetic
prevalent in Caucasians; up to 7% of the Caucasian
Properties for Therapeutic Use
population can be genetically classified as poor
metabolisers,[31] while this genetic trait is observed
6.1 Dosages and Therapeutic Range in <1% of the Asian population,[79,80] <2% in Arabi-
an populations[81] and approximately 3% in African
The relationship between exposure and efficacy
populations.[82] Likewise, mutations (allelic varia-
of atomoxetine has been evaluated in a randomised,
tions) observed in the extensive metaboliser popula-
double-blind, placebo-controlled study conducted in
tion that have relatively minimal functional conse-
children and adolescents with ADHD.[78] Patients
quences for CYP2D6 are also distributed differently
were randomised to a target dose of atomoxetine
among Caucasian, Asian, Arabian and African
(placebo, 0.5 mg/kg/day, 1.2 mg/kg/day, 1.8 mg/kg/
populations.[83]
day). Using a population pharmacokinetic model,
the modelled clearance values for each patient pro- 6.3 Influence of Age and Bodyweight on
vided estimated AUCτ values as a measure of Atomoxetine Pharmacokinetics
atomoxetine exposure. A nonlinear model (maxi-
mum effect [Emax] model) was fit to the observed Witcher et al.,[36] evaluated single-dose and
AUC and change from baseline in ADHDRS-IV- steady-state pharmacokinetics of atomoxetine in
Parent:Inv total scores data to explore the relation- children and adolescent patients across a wide range
ship between AUC and efficacy. The modelling was of doses. Identical to the pharmacokinetics in adults,
restricted to extensive metaboliser data, as there was the paediatric pharmacokinetic parameters of
only a sparse amount of poor metaboliser data avail- atomoxetine demonstrated a linear and predictable
able. The relationship between AUCτ and change pharmacokinetic behaviour. As atomoxetine doses
from baseline in ADHDRS-IV-Parent:Inv total are increased, proportional increases in plasma ex-
score suggests that the expected maximum improve- posure (AUC and Cmax) are observed. Furthermore,
ment from baseline is –17.4 (compared with –6.2 for repeated administration of atomoxetine results in a
8 weeks of placebo administration) in ADHDRS- predictable pharmacokinetic profile based on the
IV-Parent:Inv total score. The overall maximum single-dose data. Because of the rapid absorption
benefit of atomoxetine treatment was –11.2 over and elimination of the drug, steady-state profiles are
that for placebo. At the median AUC values for remarkably similar to single-dose profiles in paedia-
atomoxetine doses of 0.5 mg/kg/day, 1.2 mg/kg/day tric extensive metaboliser patients. The ability of
or 1.8 mg/kg/day, there was a 62%, 78% and 85% children and adolescents to metabolise atomoxetine
maximum improvement over baseline, respectively. rapidly and similarly to adults suggests CYP2D6
Therefore, a relationship between systemic exposure metabolic activity is mature and has reached adult
to atomoxetine and efficacy was apparent and these competency by 7–14 years of age. Phenotype distri-

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)
586 Sauer et al.

a b
26.3kg
400
29.6kg
35.3kg
Atomoxetine plasma
concentration (μg/L)

300 26.3kg
43.6kg 29.6kg
35.3kg
200
70.0kg 43.6kg
70.0kg
100

0
0 3 6 9 12 15 18 21 24 0 3 6 9 12 15 18 21 24
Time from dose (h)
Fig. 5. Predicted effect of bodyweight on atomoxetine plasma concentrations for (a) a 40mg dose and (b) a 1 mg/kg dose.

bution studies for CYP2D6 in children (mean age of systemic clearance of atomoxetine is influenced by
10.4 ± 3.7 years) and adolescents are a mirror of the renal disease. However, the metabolites of atomoxe-
distributions observed in adults.[84,85] tine are primarily excreted in urine. Therefore, the
The paediatric pharmacokinetic data reported by pharmacokinetics of atomoxetine and its primary
Witcher et al.[36] are similar to data described in metabolites were evaluated in six adults with end-
adults after adjustment for bodyweight. This study stage renal disease and compared with age-matched
provides evidence that it is appropriate to ad- healthy subjects.[37] Atomoxetine mean plasma con-
minister atomoxetine on a bodyweight-adjusted ba- centrations with end-stage renal disease patients
sis (mg/kg) in paediatric patients. Bodyweight has a were higher than the mean for healthy subjects
significant impact on atomoxetine pharmacokinet- (about a 65% increase). However, after adjustment
ics, which was demonstrated in a separate analysis for bodyweight, the differences between these
using population pharmacokinetic modelling. As groups were minimal. As expected, the plasma con-
bodyweight increases, both the clearance and vol- centrations of 4-hydroxyatomoxetine-O-glucuro-
ume of distribution increase in an essentially propor- nide, a metabolite excreted in urine with no known
tional manner.[37] Figure 5 shows the predicted ef- pharmacological actions, were substantially higher
fect of bodyweight on atomoxetine plasma concen- in individuals with end-stage renal disease. Pharma-
trations for a fixed 40mg dosing regimen in cokinetics of atomoxetine and its metabolites in
comparison to a weight-adjusted dose. The similari- individuals with end-stage renal disease suggest that
ty between the profiles for the weight-based dose no dosage adjustments are necessary.[37]
administration suggests this is a useful means to
provide comparable exposures between patients of 6.4.2 Hepatic Insufficiency
different bodyweights. This dosing method also
Since atomoxetine is primarily cleared from the
maintains constant exposure to drug as a patient
systemic circulation through oxidative metabolism,
matures and bodyweight increases.
it is reasonable to assume that the disposition of
atomoxetine would be affected by impaired hepatic
6.4 Diseases and the Pharmacokinetics function. Recently, Chalon et al.[34] compared the
of Atomoxetine pharmacokinetic parameters of atomoxetine in ten
adults with hepatic impairment (six moderate, four
6.4.1 Chronic Impairment of Renal Function severe) and ten age- and sex-matched healthy sub-
Because atomoxetine is primarily cleared jects, all being genotyped as CYP2D6 extensive
through oxidative metabolism, it is unlikely that the metabolisers.

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)
Clinical Pharmacokinetics of Atomoxetine 587

Systemic clearance of atomoxetine was signifi- Atomoxetine is rapidly and completely absorbed
cantly reduced in patients with hepatic impairment after oral administration with a median tmax of 1–2
compared with healthy subjects. There was an in- hours. The presence of food in the gastrointestinal
crease in overall atomoxetine exposure (AUC∞ 1.58 tract does not affect the extent of atomoxetine ab-
versus 0.85 μg • h/mL) but no change in Cmax or sorption but does decrease Cmax (by 37% with a
peak exposure. Mean 4-hydroxyatomoxetine AUC standard high-fat breakfast and by 9% with a more
from zero to time t (AUCt) and Cmax were increased modest meal) and delays tmax by 3 hours. Pharma-
approximately 7- and 2-fold, respectively, indicat- cokinetics of atomoxetine are linear over the range
ing a reduced rate of glucuronidation in individuals of doses studied in both extensive and poor
with hepatic impairment. For the glucuronide conju- metabolisers. The pharmacokinetics following
gate of 4-hydroxyatomoxetine, the mean half-life once-daily and twice-daily dosage regimens are
was longer and mean AUC∞ and Cmax values were readily predictable from single-dose data. Despite
also lower, similar to the type of differences noted greater systemic exposures experienced by poor
for individuals lacking CYP2D6 activity.[28] De-
metabolisers versus extensive metabolisers, there
creased atomoxetine clearance in hepatically im-
has been no clinical significance related to these
paired patients was correlated with decreased
pharmacokinetic differences. The same mean doses
CYP2D6 activity (debrisoquine clearance) and de-
were found to be well tolerated and efficacious for
creased hepatic blood flow (sorbitol clearance).
atomoxetine, and atomoxetine dosage recommenda-
Chalon et al.,[34] concluded that for ADHD patients
tions are the same regardless of CYP2D6 genotype
who have hepatic impairment, a dosage reduction is
recommended. Initial doses of atomoxetine should or phenotype.
be reduced to 25% or 50% of the normal dose for In both extensive and poor metabolisers, the pri-
patients with moderate or severe hepatic impair- mary oxidative metabolite of atomoxetine is 4-hy-
ment, respectively. droxyatomoxetine. This metabolite undergoes fur-
ther metabolism, resulting in the formation of 4-
hydroxyatomoxetine-O-glucuronide, which is pri-
7. Conclusions
marily excreted in the urine. As a result of either
genetic polymorphism or pharmacologial inhibition,
Atomoxetine is a well-tolerated inhibitor of the individuals may exhibit a poor metaboliser pheno-
presynaptic norepinephrine transporter, and is a type that is characterised by slower metabolic elimi-
drug that has shown notable efficacy for the treat- nation and higher steady-state plasma concentra-
ment of ADHD in children, adolescents and adults. tions of atomoxetine and N-demethylated metab-
The metabolism, pharmacokinetics and absolute olite compared with those observed in extensive
oral bioavailability of atomoxetine are influenced by metabolisers.
the CYP2D6 polymorphism. The mean half-life of
atomoxetine is approximately 5-fold longer (5.2 ver- Atomoxetine administration does not result in
sus 21.6 hours) and its mean CL/F is about 10-fold clinically significant inhibition or induction of
slower (0.35 versus 0.03 L/h/kg) in poor metabolis- the clearance of other drugs metabolised by
ers compared with extensive metabolisers. After CYP. In extensive metabolisers, selective inhibitors
single oral dose, the mean atomoxetine Cmax is 2- of CYP2D6 (paroxetine) increased atomoxetine
fold higher in poor metabolisers compared with steady-state plasma concentrations to exposures less
extensive metabolisers. At steady state after twice- than or similar to those observed in poor metabolis-
daily dosing, the mean atomoxetine Cmax is approxi- ers. In vitro studies indicate that the coadministra-
mately 5-fold higher and the mean plasma concen- tion of CYP inhibitors to individuals lacking
tration is approximately 10-fold higher in poor CYP2D6 activity is not expected to increase the
metabolisers compared with extensive metabolisers. plasma concentrations of atomoxetine.

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)
588 Sauer et al.

The results of the conventional pharmacokinetic 6. Barkley FA, Fischer M, Edelbrock CS, et al. The adolescent
outcome of hyperactive children diagnosed by research crite-
analyses in adults, as well as conventional and popu- ria: I. An 8-year prospective follow-up study. J Am Acad Child
lation pharmacokinetic analyses in children, demon- Adolesc Psychiatry 1990; 29: 546-57
7. Munir K, Biederman J, Knee D. Psychiatric comorbidity in
strate that the pharmacokinetics of atomoxetine are patients with attention deficit disorder: a controlled study. J
similar between these age groups after adjustment Am Acad Child Adolesc Psychiatry 1987; 26: 844-8
for bodyweight. Weight-based dosage regimens 8. Biederman J, Newcorn J, Sprich S. Comorbidity of attention
deficit hyperactivity disorder with conduct, depressive, anxie-
(mg/kg) are recommended for paediatric patients. ty, and other disorders. Am J Psychiatry 1991; 148: 564-77
The pharmacokinetic parameters of atomoxetine 9. Spencer TJ, Biederman J, Wilens TE, et al. Overview and
neurobiology of attention-deficit/hyperactivity disorder. J Clin
were affected by moderate and severe hepatic insuf- Psychiatry 2002; 63 Suppl. 12: 3-9
ficiency, and a dosage reduction is recommended in 10. Volkow ND, Fowler JS, Wang G-J, et al. Role of dopamine in
patients who have hepatic insufficiency. Pharma- the therapeutic and reinforcing effects of methylphenidate in
humans: results from imaging studies. Eur Neuropsychophar-
cokinetics of atomoxetine and its metabolites in macol 2002; 12: 557-66
individuals with end-stage renal disease suggest that 11. Giros B, Caron MG. Molecular characterization of the
no dosage adjustment is necessary. dopamine transporter. Trends Pharmacol Sci 1993; 14: 43-9
12. Patrick KS, Markowitz JS. Pharmacology of methylphenidate,
Overall, the pharmacokinetics and metabolism of amphetamine enantiomers and pemoline in attention-deficit
atomoxetine are predictable based on an individu- hyperactivity disorder: a review. Hum Psychopharmacol 1997;
12: 527-46
al’s CYP2D6 phenotype. Both the efficacy and safe-
13. Dackis CA, Gold MS. Addictiveness of central stimulants. Adv
ty of atomoxetine have been demonstrated for the Alcohol Subst Abuse 1990; 9: 9-26
treatment of ADHD in children, adolescents and 14. Markowitz JS, Patrick KS. Pharmacokinetic and pharmacody-
namic drug interactions in the treatment of attention-deficit
adults. As the first FDA-approved, nonstimulant hyperactivity disorder. Clin Pharmacokinet 2001; 40: 753-72
pharmaceutical treatment for ADHD, atomoxetine 15. Pelham WE, Gnagy EM, Burrows-Maclean L, et al. Once-a-
represents a significant advance in the treatment of day Concerta methylphenidate versus three-times-daily
methylphenidate in laboratory and natural settings [abstract].
this disease. Furthermore, due to its novel mecha- Pediatrics 2001; 107: E105
nism of action, atomoxetine does not share the abuse 16. Lyseng-Williamson KA, Keating GM. Extended-release
potential associated with psychostimulant drugs. methylphenidate (Ritalin LA). Drugs 2002; 62: 2251-9
17. Tulloch SJ, Zhang Y, McLean A, et al. SLI381 (Adderall XR), a
two-component, extended-release formulation of mixed am-
Acknowledgements phetamine salts: bioavailability of three test formulations and
comparison of fasted, fed, and sprinkled administration.
The authors wish to thank Dr Richard F. Bergstrom from Pharmacotherapy 2002; 22: 1405-15
Department of Drug Disposition, Lilly Research Laborato- 18. Wu D, Otton SV, Inaba T, et al. Interactions of amphetamine
ries, Eli Lilly and Company, Indianapolis, IN, USA, for his analogs with human liver CYP2D6. Biochem Pharmacol 1997;
53: 1605-12
helpful guidance and insightful scientific comments. The
19. Nehra A, Mullick R, Ishak KG, et al. Pemoline-associated
authors also thank Ms Amanda J. Long for her contributions hepatic injury. Gastroenterology 1990; 99: 1517-9
to the work presented herein. Finally, the authors thank Mr 20. Popper CW. Antidepressants in the treatment of attention-defi-
Nathan P. Sanburn for his editorial comments. At the time of cit/hyperactivity disorder. J Clin Psychiatry 1997; 58 Suppl.
preparation of this review all authors were employees of Eli 14: 14-29
Lilly and Company, Indianapolis, IN, USA. 21. Wong DT, Threlkeld PG, Best KL, et al. A new inhibitor of
norepinephrine uptake devoid of affinity for receptors in rat
brain. J Pharmacol Exp Ther 1982; 222: 61-5
References 22. Bymaster FP, Katner JS, Nelson DL, et al. Atomoxetine in-
1. Spencer T, Biederman J, Wilens T, et al. Pharmacotherapy of creases extracellular levels of norepinephrine and dopamine in
attention-deficit hyperactivity disorder across the life cycle. prefrontal cortex of rat: a potential mechanism for efficacy in
Child Adol Psychiatry 1996; 35: 409-32 attention deficit/hyperactivity disorder. Neuropsychopharma-
2. Swanson JM, Sergeant JA, Sonuga-Barke EJS, et al. Attention- cology 2002; 27: 699-711
deficit hyperactivity disorder and hyperkinetic disorder. Lan- 23. Spencer T, Biederman J, Wilens T, et al. Effectiveness and
cet 1998; 351: 429-33 tolerability of tomoxetine in adults with attention deficit hyper-
3. Weiss G. Follow-up studies on outcome of hyperactive children. activity disorder. Am J Psychiatry 1998; 155: 693-5
Psychopharmacol Bull 1985; 21: 169-77 24. Michelson D, Adler L, Spencer T, et al. Atomoxetine in adults
4. Pary R, Lewis S, Matuschka PR, et al. Attention deficit disorder with ADHD: two randomized, placebo-controlled studies. Biol
in adults. Ann Clin Psychiatry 2002; 14: 105-11 Psychiatry 2003; 53: 112-20
5. Adler LA, Chua HC. Management of ADHD in adults. J Clin 25. Michelson D, Faries DE, Wernicke J, et al. Atomoxetine in the
Psychiatry 2002; 63 Suppl. 12: 29-35 treatment of children and adolescents with ADHD: a random-

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)
Clinical Pharmacokinetics of Atomoxetine 589

ized, placebo-controlled dose-response study [abstract]. Pedi- 43. Ring BJ, Gillespie JS, Eckstein JA, et al. Identification of the
atrics 2001; 108: E83 human cytochromes P450 responsible for atomoxetine metab-
26. Michelson D, Allen AJ, Busner J, et al. Once-daily atomoxetine olism. Drug Metab Dispos 2002; 30: 319-23
treatment for children and adolescents with attention deficit 44. Segel IH. Enzyme kinetics. New York: John Wiley and Sons,
hyperactivity disorder: a randomized, placebo-controlled Inc., 1975
study. Am J Psychiatry 2002; 159: 1896-901 45. Oberlender R, Nichols DE, Ramachandran PV, et al. Tomoxe-
27. Kratochvil CJ, Heiligenstein JH, Dittmann R, et al. Atomoxetine tine and the stereoselectivity of drug action. J Pharm Pharma-
and methylphenidate treatment in children with ADHD: a col 1987; 39: 1055-6
prospective, randomized, open-label trial. Am Acad Child 46. Gehlert DR, Gackenheimer SL, Robertson DW. Localization of
Adolesc Psychiatry 2002; 41: 776-84 rat brain binding sites for [3H]tomoxetine, an enantiomerically
28. Sauer JM, Ponsler GD, Mattiuz EL, et al. Disposition and pure ligand for norepinephrine reuptake sites. Neurosci Lett
metabolic fate of atomoxetine hydrochloride: the role of 1993; 157: 203-6
CYP2D6 in human disposition and metabolism. Drug Metab 47. Gehlert DR, Schober DA, Gackenheimer SL. Comparison of
Dispos 2003; 31: 98-107 (R)-[3H]tomoxetine and (R/S)-[3H]nisoxetine binding in rat
29. Farid NA, Bergstrom RF, Ziege EA, et al. Single-dose and brain. J Neurochem 1995; 64: 2792-800
steady-state pharmacokinetics of tomoxetine in normal sub-
48. Heil SH, Holmes HW, Bickel WK, et al. Comparison of the
jects. J Clin Pharmacol 1985; 25: 296-301
subjective, physiological, and psychomotor effects of atomox-
30. Belle DJ, Ernest S, Sauer JM, et al. Effect of potent CYP2D6 etine and methylphenidate in light drug users. Drug Alcohol
inhibition by paroxetine on atomoxetine pharmacokinetics. J Depend 2002; 67: 149-56
Clin Pharmacol 2002; 42: 1-9
49. Wheeler WJ, Bymaster FP, Calligaro DO, et al. Strattera®
31. Guengerich FP. Human cytochrome P450 enzymes. In: Ortiz de (atomoxetine HCl), an inhibitor of the norepinephrine trans-
Montellano PR, editor. Cytochrome P450: structure, mecha- porter. I: the preparation of C-14 labeled atomoxetine, and two
nism, and biochemistry. New York: Plenum Press, 1995: 473- of its metabolites; II: The preparation and biological evaluation
535 of some additional putative metabolites of atomoxetine. In:
32. Sachse C, Brockmoller J, Bauer S, et al. Cytochrome P450 2D6 Dean DC, Filer CN, McCarthy KE, editors. Synthesis and
variants in a Caucasian population: allele frequencies and applications of isotopically labelled compounds. Vol 8. New
phenotypic consequences. Am J Hum Genet 1997; 60: 284-95 York: John Wiley and Sons, Inc., 2004: 357-60
33. Agundez JA, Ledesma MC, Ladero JM, et al. Prevalence of 50. Fuller RW, Hemrick-Luecke SK. Antagonism by tomoxetine of
CYP2D6 gene duplication and its repercussion on the oxida- the depletion of norepinephrine and epinephrine in rat brain by
tive phenotype in a white population. Clin Pharmacol Ther alpha-methyl-m-tyrosine. Res Commun Chem Path Pharmacol
1995; 57: 265-9 1983; 41: 169-72
34. Chalon S, Desager JP, DeSante K, et al. Effect of liver impair-
51. Mattiuz EL, Ponsler GD, Barbuch RJ, et al. Disposition and
ment on the pharmacokinetics of atomoxetine and its metabo-
metabolic fate of atomoxetine hydrochloride: pharmacokinet-
lites. Clin Pharmacol Ther 2003; 73: 178-91
ics, metabolism, and excretion in the fischer 344 rat and beagle
35. Sauer JM, Long AJ, Ring B, et al. Disposition and metabolic dog. Drug Metab Dispos 2003; 31: 88-97
fate of atomoxetine hydrochloride: clinical drug-drug interac-
52. Tidey JW, Bergman J. Drug discrimination in metham-
tion prediction and outcome. J Pharmacol and Exp Ther 2004;
phetamine-trained monkeys: agonist and antagonist effects of
308: 410-8
dopaminergic drugs. J Pharmacol Exp Ther 1998; 285: 1163-
36. Witcher JW, Long AJ, Sauer JM, et al. Atomoxetine 74
pharmacokinetics in children with attention deficit hyperactiv-
ity disorder. J Child Adolesc Psychopharmacol 2003; 13: 53- 53. Wernicke JF, Kratochvil CJ. Safety profile of atomoxetine in the
64 treatment of children and adolescents with ADHD. J Clin
Psychiatry 2002; 63 Suppl. 12: 50-5
37. Strattera™ (atomoxetine) package insert (NDA 21-411). Indian-
apolis (IN); Eli Lilly and Co., 2003 54. Wernicke JF, Allen AJ, Faries D, et al. Safety of tomoxetine in
clinical trials [abstract]. Biol Psychiatry 2001; 49 (8 Suppl.):
38. Evans DAP, Maghoub A, Sloan TP, et al. A family and popula-
159S
tion study of the genetic polymorphism of the debrisoquine
oxidation in a white British population. J Med Genet 1980; 17: 55. Spencer T, Heiligenstein JH, Biederman J, et al. Results from 2
102-5 proof-of-concept, placebo-controlled studies of atomoxetine in
39. Steiner E, Bertilsson L, Sawe J, et al. Polymorphic debrisoquine children with attention-deficit/hyperactivity disorder. J Clin
hydroxylation in 757 Swedish subjects. Clin Pharmacol Ther Psychiatry 2002; 63: 1140-7
1988; 44: 431-5 56. Wernicke JF, Faries D, Girod D, et al. Cardiovascular effects of
40. DeSante K, Long A, Smith B, et al. Atomoxetine absolute atomoxetine in children, adolescents, and adults. Drug Saf
bioavailability and effects of food, Maalox or omeprazole on 2003; 26: 729-40
atomoxetine bioavailability. AAPS Annual Meeting and Expo- 57. Chouinard G, Annable L, Bradwejn J. An early phase II clinical
sition; 2001 Oct 21-25; Denver trial of tomoxetine (LY139603) in the treatment of newly
41. Herman JL, Kou F, Sauer JM, et al. Tissue disposition of 14C- admitted depressed patients. Psychopharmacologia 1984; 83:
tomoxetine in male Fischer 344 rats following a single oral 126-8
dose administration. Society for Whole-Body Autoradiogra- 58. Zerbe RL, Rowe H, Enas GG, et al. Clinical pharmacology of
phy Meeting; 1999 Apr 18-20; St Louis tomoxetine, a potential antidepressant. J Pharmacol Exp Ther
42. Hamilton MM, Herman JL, Kou F, et al. Placental transfer and 1985; 232: 139-43
milk excretion in rats after a single oral 50 mg/kg dose of 59. Allen AJ, Wernicke JF, Dunn D, et al. Safety and efficacy of
[14C]atomoxetine administered as the hydrochloride salt. Eu- atomoxetine in pediatric CYP2D6 extensive and poor
ropean Society for Whole Body Autoradiography; 2000, Paris metabolizers. Biol Psychiatry 2001; 49 (8 Suppl.): 37S

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)
590 Sauer et al.

60. Caccia S. Metabolism of the newer antidepressants: an overview 74. Sanburn N, Long A, Witcher J, et al. Co-administration of
of the pharmacological and pharmacokinetic implications. atomoxetine hydrochloride and midazolam results in no clini-
Clin Pharmacokinet 1998; 34: 281-302 cally significant drug-drug interaction. AAPS Annual Meeting
61. Preskorn SH. Clinically relevant pharmacology of selective and Exposition; 2001 Oct 21-25, Denver
serotonin reuptake inhibitors: an overview with emphasis on 75. Kronbach T, Mathys D, Umeno M, et al. Oxidation of midazo-
pharmacokinetics and effects on oxidative drug metabolism.
lam and triazolam by human liver cytochrome P450IIIA4. Mol
Clin Pharmacokinet 1997; 32S: 1-21
Pharmacol 1989; 36: 89-96
62. Brøsen K, Hansen JG, Nielsen KK, et al. Inhibition by paroxe-
76. Olkkola KT, Backman JT, Neuvonen PJ. Midazolam should be
tine of desipramine metabolism in extensive but not in poor
metabolizers of sparteine. Eur J Clin Pharmacol 1993; 44: 349- avoided in patients receiving the systemic antimycotics
55 ketoconazole or itraconazole. Clin Pharmacol Ther 1994; 55:
481-5
63. Özdemir V, Naranjo CA, Herrmann N, et al. Paroxetine poten-
tiates the central nervous system side effects of perphenazine: 77. Yuan R, Flockhart D, Balian J. Pharmacokinetic and pharmaco-
contribution of cytochrome P4502D6 inhibition in vivo. Clin dynamic consequences of metabolism-based drug interactions
Pharmacol Ther 1997; 62: 334-47 with alprazolam, midazolam, and triazolam. J Clin Pharmacol
64. Alderman J, Preskorn SH, Greenblatt DJ, et al. Desipramine 1999; 39: 1109-25
pharmacokinetics when coadministered with paroxetine or ser- 78. Witcher JW, Kurtz DL, Sauer JM, et al. Pharmacokinetic/
traline in extensive metabolizers. J Clin Psychopharmacol
pharmacodynamic relationship of atomoxetine exposure and
1997; 17: 284-91
efficacy in child and adolescent ADHD patients. Philadelphia
65. Hemeryck A, Lefebvre RA, De Vriendt C, et al, editor. Paroxe- (PA): American Psychiatric Association, 2002
tine affects metoprolol pharmacokinetics and pharmacody-
namics in healthy volunteers. Drug Metab Dispos 2001; 29: 79. Horai Y, Nakano M, Ishizaki T, et al. Metoprolol and
656-63 mephenytoin oxidation polymorphisms in Far Eastern Oriental
subjects: Japanese versus mainland Chinese. Clin Pharmacol
66. Kobayashi K, Yamamoto T, Chiba K, et al. The effects of
selective serotonin reuptake inhibitors and their metablites on Ther 1989; 46: 198-207
S-mephenytoin 4′-hydroxylase activity in human liver micro- 80. Zanger UM, Eichelbaum M. CYP2D6. In: Levy RH, Thummel
somes. Br J Clin Pharmacol 1995; 40: 481-5 KE, Trager WF, et al, editors. Metabolic drug interactions.
67. von Moltke LL, Greenblatt DJ, Court MH, et al. Inhibition of New York: Lippincott Williams & Wilkins, 2000: 87-94
alprazolam and desipramine hydroxylation in vitro by paroxe- 81. McLellan RA, Oscarson M, Seidegard J, et al. Frequent occur-
tine and fluvoxamine: comparison with other selective seroto- rence of CYP2D6 gene duplication in Saudi Arabians.
nin reuptake inhibitor antidepressants. J Clin Psychopharma-
Pharmacogenetics 1997; 7: 187-91
col 1995; 15: 125-31
82. Griese EU, Asante-Poku S, Ofori-Adjei D, et al. Analysis of the
68. Jeppesen U, Gram LF, Vistisen K, et al. Dose-dependent inhibi-
CYP2D6 gene mutations and their consequences for enzyme
tion of CYP1A2, CYP2C19 and CYP2D6 by citalopram,
fluoxetine, fluvoxamine and paroxetine. Eur J Clin Pharmacol function in a West African population. Pharmacogenetics
1996; 51: 73-8 1999; 9: 715-23
69. Martin DE, Zussman BD, Everitt DE, et al. Paroxetine does not 83. Bradford LD. CYP2D6 allele frequency in European Cauca-
affect the cardiac safety and pharmacokinetics of terfenadine sians, Asians, Africans and their descendants. Pharmacoge-
in healthy adult men. J Clin Psychopharmacol 1997; 17: 451-9 nomics 2002; 3: 229-43
70. Schmider J, Greenblatt DJ, von Moltke LL, et al. Inhibition of 84. Evans W, Relling M, Petros W, et al. Dextromethorphan and
CYP2C9 by selective serotonin reuptake inhibitors in vitro: caffeine as probes for simultaneous determination of debriso-
studies of phenytoin p-hydroxylation. Br J Clin Pharmacol quin-oxidation and N-acetylation phenotypes in children. Clin
1997; 44: 495-8 Pharmacol Ther 1989; 45: 568-73
71. Hemeryck A, De Vriendt C, Belpaire FM. Inhibition of 85. Relling M, Cherrie J, Schell M, et al. Lower prevalence of the
CYP2C9 by selective serotonin reuptake inhibitors: in vitro debrisoquin oxidative poor metabolizer phenotype in Ameri-
studies with tolbutamide and (S)-warfarin using human liver
microsomes. Eur J Clin Pharmacol 1999; 54: 947-51 can black versus white subjects. Clin Pharmacol Ther 1991;
50: 308-13
72. Long A, Witcher J, Smith B, et al. Atomoxetine does not alter
the plasma pharmacokinetics of desipramine in healthy sub-
jects. AAPS Annual Meeting and Exposition; 2001 Oct 21-25,
Correspondence and offprints: Dr Jennifer W. Witcher, De-
Denver
partment of Drug Disposition, Lilly Research Laboratories,
73. Spina E, Avenoso A, Campo GM, et al. Effect of ketoconazole
on the pharmacokinetics of imipramine and desipramine in Eli Lilly and Company, Lilly Corporate Center, Indianapo-
healthy subjects. Br J Clin Pharmacol 1997; 43: 315-8 lis, IN 46285, USA.

© 2005 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2005; 44 (6)

Você também pode gostar