You are on page 1of 14

Clin Pharmacokinet 2011; 50 (5): 281-294

REVIEW ARTICLE 0312-5963/11/0005-0281/$49.95/0

ª 2011 Adis Data Information BV. All rights reserved.

Duloxetine
Clinical Pharmacokinetics and Drug Interactions
Mary Pat Knadler, Evelyn Lobo, Jill Chappell and Richard Bergstrom
Eli Lilly and Company, Indianapolis, Indiana, USA

Contents

Abstract. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 281
1. Mechanism of Action and Therapeutic Use . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 282
2. Clinical Pharmacokinetics. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 282
2.1 Absorption . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 282
2.2 Distribution . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 284
2.3 Metabolism. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 285
2.4 Elimination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 286
3. Special Populations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 286
3.1 Sex and Smoking Status . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 286
3.2 Age . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 287
3.3 Ethnicity. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 287
3.4 Hepatic Impairment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 287
3.5 Renal Impairment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 287
3.6 Lactation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 287
4. Drug Interaction Evaluations. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 288
4.1 In Vitro Studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 288
4.1.1 Cytochrome P450 (CYP) Inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 288
4.1.2 CYP Induction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 288
5. Clinical Pharmacokinetic Drug Interaction Studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 288
5.1 Potential for Other Drugs to Affect Duloxetine . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 288
5.1.1 Agents Affecting Gastric pH and Activated Charcoal. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 288
5.1.2 Effect of CYP2D6 Inhibition/Genetic Polymorphism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 289
5.1.3 Effect of CYP1A2 Inhibition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 289
5.1.4 Effect of CYP1A2 Inhibition and CYP2D6 Genotype . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 289
5.2 Potential for Duloxetine to Affect Other Drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 290
5.2.1 Effects on CYP2D6 Substrates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 290
5.2.2 Effects on CYP1A2 Substrates . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 290
5.3 Pharmacodynamic Interactions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 290
5.3.1 Benzodiazepines. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 291
5.3.2 Alcohol . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 291
5.3.3 Anticoagulant Drugs . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 291
5.3.4 Serotonin Syndrome. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 291
6. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 292

Abstract Duloxetine, a potent reuptake inhibitor of serotonin (5-HT) and norepinephrine, is effective for the
treatment of major depressive disorder, diabetic neuropathic pain, stress urinary incontinence, generalized
anxiety disorder and fibromyalgia. Duloxetine achieves a maximum plasma concentration (Cmax) of ap-
proximately 47 ng/mL (40 mg twice-daily dosing) to 110 ng/mL (80 mg twice-daily dosing) approximately
282 Knadler et al.

6 hours after dosing. The elimination half-life of duloxetine is approximately 10–12 hours and the volume of
distribution is approximately 1640 L. The goal of this paper is to provide a review of the literature on
intrinsic and extrinsic factors that may impact the pharmacokinetics of duloxetine with a focus on con-
comitant medications and their clinical implications. Patient demographic characteristics found to influence
the pharmacokinetics of duloxetine include sex, smoking status, age, ethnicity, cytochrome P450 (CYP) 2D6
genotype, hepatic function and renal function. Of these, only impaired hepatic function or severely impaired
renal function warrant specific warnings or dose recommendations. Pharmacokinetic results from drug
interaction studies show that activated charcoal decreases duloxetine exposure, and that CYP1A2 inhibition
increases duloxetine exposure to a clinically significant degree. Specifically, following oral administration in
the presence of fluvoxamine, the area under the plasma concentration-time curve and Cmax of duloxetine
significantly increased by 460% (90% CI 359, 584) and 141% (90% CI 93, 200), respectively. In addition,
smoking is associated with a 30% decrease in duloxetine concentration. The exposure of duloxetine with
CYP2D6 inhibitors or in CYP2D6 poor metabolizers is increased to a lesser extent than that observed with
CYP1A2 inhibition and does not require a dose adjustment. In addition, duloxetine increases the exposure
of drugs that are metabolized by CYP2D6, but not CYP1A2. Pharmacodynamic study results indicate that
duloxetine may enhance the effects of benzodiazepines, but not alcohol or warfarin. An increase in gastric
pH produced by histamine H2-receptor antagonists or antacids did not impact the absorption of duloxetine.
While duloxetine is generally well tolerated, it is important to be knowledgeable about the potential for
pharmacokinetic interactions between duloxetine and drugs that inhibit CYP1A2 or drugs that are meta-
bolized by CYP2D6 enzymes.

1. Mechanism of Action and Therapeutic Use 2. Clinical Pharmacokinetics

Duloxetine (Cymbalta), administered as duloxetine hy- 2.1 Absorption


drochloride – or (+)-(S)-N-methyl-g-(1-naphthyloxy)-2-thio-
phenepropylamine hydrochloride – (see figure 1), is a potent Duloxetine is acid labile, requiring the development of a
inhibitor of serotonin (5-HT) and norepinephrine reuptake.[1-5] formulation that utilizes enteric-coated pellets to protect
Compared with its effects on 5-HT and norepinephrine reup- duloxetine from degradation in the acidic environment of the
take, duloxetine weakly inhibits dopamine reuptake and has stomach. The delayed-release formulation prevents dissolu-
low binding affinity for other neurotransmitter receptors, in- tion in acidic conditions (pH <5.5) such as those in the stomach
cluding adrenergic, muscarinic (nonselective) and histamine but allows for immediate release and rapid absorption at the
H1 receptors; dopamine D2 receptors; 5-HT1A, 5-HT1D and pH in the small intestine. Following oral administration, du-
5-HT2C receptors; and opioid receptors.[1,2,6] Duloxetine is used loxetine is well absorbed with a median time to maximum
clinically in many countries to treat disorders related to nor- plasma concentration (tmax) of 6 hours after dosing. The ab-
epinephrine and 5-HT, such as depression,[7-13] diabetic neu- solute oral bioavailability averaged 50%, ranging from 30% to
ropathic pain,[14,15] generalized anxiety disorder[16-19] and 80%, after a 60 mg single dose in one study[2] and averaged 43%,
fibromyalgia.[20] In Europe, duloxetine is also used to treat ranging from 19% to 71%, in another study.[24] Population
stress urinary incontinence.[21,22] The safety of duloxetine pharmacokinetic results in patients indicate that the rate of
has been extensively evaluated in a large number of patients absorption is rapid (absorption rate constant 0.168 h-1) once
across indications.[6,23] To understand the potential for drug dissolution occurs in the intestine.[25] The elimination half-life
interactions and whether dose adjustments are appropriate (t½) of duloxetine is approximately 10–12 hours and therefore
for a specific patient with any of these indications, it is im- steady state occurs within 3 days. Plasma concentrations gen-
portant to consider the intrinsic and extrinsic factors that erally range from 24.6 ng/mL to 110 ng/mL depending on the
influence the pharmacokinetics or pharmacodynamics of du- dose and dosing regimen (tables I and II and figures 2 and
loxetine. The goal of this paper is to provide a review of the 3).[24,26-29] A single dose of 60 mg of duloxetine produced a


literature on factors that may impact the pharmacokinetics of mean area under the plasma concentration-time curve (AUC)
duloxetine with a focus on the clinical implications of these from time zero to infinity (AUC1) of 591 ng h/mL (table I)
factors. and 40 or 60 mg twice-daily dosing provided a mean steady-

ª 2011 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2011; 50 (5)
Duloxetine Pharmacokinetics and Drug Interactions 283

Glucuronide
conjugate of 4-hydroxy duloxetine

Other pathways OH

N CH3
H
S
Major 4-Hydroxy duloxetine

CYP1A2
CYP2D6
O OH
OH
OH
* N CH3
H
S Major
Duloxetine
CYP1A2
CYP2D6 O O

N CH3 N CH3
H H
S S
5-Hydroxy or
Catechol duloxetine
6-hydroxy duloxetine

CH3
O

OH

N CH3
H
Sulfate and glucuronide conjugates
of 5-hydroxy-6-methoxy duloxetine S

5-Hydroxy-6-methoxy duloxetine

Fig. 1. Metabolism pathways and cytochrome P450 enzymes (CYPs) involved in the major oxidation steps of duloxetine biotransformation. The asterisk
indicates the position of the radiolabel in the material used for the distribution study. The bracketed compound is postulated, but has not been isolated.

state AUC (AUCt,ss) of 412 or 1050 ng h/mL (table II), re-


spectively.
 time and in the presence of food, especially fats. Decreased
motility may also prolong exposure of a drug to intraluminal and
Food and time of day impact duloxetine absorption, with food gut-wall degradation or metabolic processes, thus decreasing the
and bedtime administration delaying tmax by 4 hours. This finding extent of absorption. However, food had no significant effect on
is consistent with a decrease in gastrointestinal motility at bed- duloxetine maximum plasma concentration (Cmax) and reduced

ª 2011 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2011; 50 (5)
284 Knadler et al.

Table I. Summary of pharmacokinetic parameters of duloxetine and its two major metabolites following a single oral dose of 40 mg or 60 mg of duloxetine[24,26,27]
Parameter Duloxetine 40 mga Duloxetine 60 mga

duloxetine duloxetine glucuronide conjugate of sulfate conjugate of


(n = 24) (n = 50) 4-hydroxy duloxetine 5-hydroxy-6-methoxy duloxetine
(n = 26) (n = 26)
Cmax (ng/mL) 24.6 [50.8] 40.1 [47.5] 402 [47.6] 258 [49.1]
tmax (h)b 6.00 [0.99–10.01] 6.00 [2–6.03] 5.51 [3–8] 5 [3–8]
t½ (h)c 11.8 [8.04–17.5] 10.2 [6.96–14.9] 11.6 [9.06–17.1] 10.1 [6.46–13.2]


AUC1 (ng h/mL) 422 [50.5] 591 [55.6] 6830 [46.6] 3290 [42.4]
CL/F (L/h) 94.8 [50.5] 101 [55.6] NA NA
Vss/F (L) 1800 [51.2] 1620 [45.8] NA NA
Metabolite/parent drug ratiod NA NA 6.88 3.83
a Values are expressed as geometric mean [CV%] unless specified otherwise.
b Median [range].
c Geometric mean [range].
d Metabolite/parent drug ratio = AUC1,metabolite/AUC1,duloxetine.
AUC1 = area under the plasma concentration-time curve from time zero to infinity; CL/F = apparent clearance after oral administration; Cmax = maximum plasma
drug concentration; CV = coefficient of variation; NA = not applicable; t½ = elimination half-life; tmax = time to reach Cmax; Vss/F = volume of distribution at steady
state after oral administration.

AUC1 and t½ by only 11% and 18%, respectively. Bedtime ad- 2.2 Distribution
ministration had no effect on t½ and reduced duloxetine Cmax
and AUC1 by 29% and 18%, respectively.[30] Because the effects Duloxetine is extensively distributed throughout the body,
of food and bedtime administration are relatively small and as indicated by a large mean apparent steady-state volume of
clinically insignificant, dose adjustments or time-of-day restric- distribution of 1620–1800 L (table I).[31] Protein binding was
tions are not required. Patients may be instructed to take du- assessed as a factor that may impact duloxetine distribution.
loxetine with or without food and in the morning or at bedtime. Results from in vitro studies showed that more than 90% of

Table II. Summary of pharmacokinetic parameters of duloxetine and its two major metabolites following twice-daily oral dosing of 40 mg or 60 mg of duloxetine[26,28,29]
Parameter Duloxetine 40 mga Duloxetine 60 mga

duloxetine duloxetine glucuronide conjugate of sulfate conjugate of


(n = 14) (n = 36) 4-hydroxy duloxetine 5-hydroxy-6-methoxy duloxetine
(n = 22) (n = 22)
Cmin,ss (ng/mL) 22.9 [73.0] 66.3 [75.7] 353 [55.0] 185 [31.0]
Cmax,ss (ng/mL) 47.8 [58.6] 110 [66.7] 568 [43.1] 336 [28.5]
Cav,ss (ng/mL) 34.3 [58.5] 87.7 [69.4] 467 [46.2] 259 [26.6]
tmax,ss (h)b 6 [2–9] 6 [3–8] 6.00 [4–8] 6.00 [4–8]


AUCt,ss (ng h/mL) 412 [58.5] 1050 [69.4] 5600 [46.2] 3110 [26.6]
CLss/F (L/h) 97.1 [58.5] 57.0 [69.4] NA NA
Metabolite/parent drug ratioc NA NA 3.17 2.04
a Values are expressed as geometric mean [CV%] unless specified otherwise.
b Median [range].
c Metabolite/parent drug ratio = AUCt,ss,metabolite/AUCt,ss,duloxetine.
AUCs,ss = area under the plasma concentration-time curve during one dosing interval at steady state; Cav,ss = average plasma concentration at steady state;
CLss/F = apparent clearance at steady state after oral administration; Cmax,ss = maximum plasma concentration at steady state; Cmin,ss = minimum plasma
concentration at steady state; CV = coefficient of variation; NA = not applicable; tmax,ss = time to reach Cmax at steady state.

ª 2011 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2011; 50 (5)
Duloxetine Pharmacokinetics and Drug Interactions 285

Duloxetine after duloxetine 60 mg sd (n = 18) drug accounts for approximately 3% of the circulating radio-
Glucuronide conjugate of 4-hydroxy duloxetine
after duloxetine 60 mg sd (n = 14) activity and approximately 9% of the radioactivity based upon
Sulfate conjugate of 5-hydroxy-6-methoxy duloxetine
after duloxetine 60 mg sd (n = 14)
Cmax. The two major circulating metabolites of duloxetine
Duloxetine after duloxetine 40 mg sd (n = 16) are the glucuronide conjugate of 4-hydroxy duloxetine and
1000
the sulfate conjugate of 5-hydroxy-6-methoxy duloxetine
(figure 1).[36] The metabolic ratio (metabolite concentration/
Plasma concentration (ng/mL)

100 duloxetine concentration) for the glucuronide conjugate of


4-hydroxy duloxetine averaged across studies ranges from 3.2
to 6.9 and the metabolic ratio for the sulfate conjugate of
10
5-hydroxy-6-methoxy duloxetine averaged across studies ranges
from approximately 2.0 to 3.8 (tables I and II). Binding of these
1 metabolites to membranes expressing cloned human 5-HT and
norepinephrine transporters produced inhibition constant (Ki)
values >10 000 nmol/L for both receptors while the Ki for du-
0.1
loxetine was 0.8 nmol/L for 5-HT binding and 7.45 nmol/L for
0 12 24 36 48 60 72
Time (h)
norepinephrine binding.[37] The low binding of the glucuronide
conjugate of 4-hydroxy duloxetine and the sulfate conjugate of
Fig. 2. Mean plasma concentration-time profiles of duloxetine and its two major 5-hydroxy-6-methoxy duloxetine lead to a conclusion that
metabolites after a single oral dose (sd) of duloxetine 60 mg or 40 mg.[24,26]
these compounds are pharmacologically inactive metabolites.
Studies were performed to determine the cytochrome P450
duloxetine is protein bound in human plasma.[2] The binding (CYP) enzymes responsible for the biotransformation of du-
occurs primarily to albumin and a1-acid glycoprotein.[32] The loxetine to 4-, 5- and 6-hydroxy duloxetine.[38] The studies
percent bound was independent of duloxetine concentration correlated the formation of these metabolites with markers of
across the concentration range associated with therapeutic use. CYP enzymes in a human liver microsome bank, assessed
While displacement from protein binding sites could potentially which complementary DNA (cDNA)-expressed CYPs formed
impact distribution of drugs when two highly protein-bound the metabolites and utilized CYP inhibitors to determine which
drugs are co-administered, data across a wide spectrum of CYP enzymes were responsible for the initial oxidation step.[24]
drugs suggest that changes in protein binding would not result
in a clinically significant drug interaction.[33,34]
Duloxetine after duloxetine 60 mg bid (n = 36)
Duloxetine has been shown to distribute into the brains of Glucuronide conjugate of 4-hydroxy duloxetine
both rats and humans. In rats administered a 20 mg/kg dose of after duloxetine 60 mg bid (n = 22)
Sulfate conjugate of 5-hydroxy-6-methoxy duloxetine
duloxetine, the concentrations in the cerebral cortex were after duloxetine 60 mg bid (n = 22)
higher than those in plasma.[2] The plasma protein binding in Duloxetine after duloxetine 40 mg bid (n = 14)
1000
rats is similarly high as in humans. In a study of healthy male
subjects administered single doses of 5–60 mg of duloxetine or
Plasma concentration (ng/mL)

60 mg repeat doses, 5-HT transporter occupancy in the thala-


mus was studied using positron emission tomography.[35] Based 100

on these data, doses of 40 mg and higher of duloxetine resulted


in 80% occupancy, thus demonstrating that duloxetine is dis-
tributed into brain areas. 10

2.3 Metabolism
1
Duloxetine undergoes extensive metabolism into a complex 0 3 6 9 12
array of metabolites. Importantly, the major biotransforma- Time (h)
tion pathways involve oxidation in the naphthyl ring followed
Fig. 3. Mean plasma concentration-time profiles of duloxetine and its two
by further oxidation, methylation and conjugation. After a major metabolites at steady state during twice-daily (bid) oral dosing of
single oral dose of 14C-duloxetine, the relative AUC of parent duloxetine 40 mg or 60 mg.[26,28,29]

ª 2011 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2011; 50 (5)
286 Knadler et al.

The rates of formation from the in vitro studies indicated that bolite that has not been characterized account for the majority
4-hydroxy and 5-hydroxy duloxetine, but not 6-hydroxy du- of faecal radioactivity.[36] No additional work was conducted to
loxetine, define the metabolic clearance of duloxetine. These identify the unknown metabolite because it was present in very
findings for 4-hydroxy and 5-hydroxy duloxetine were corro- low levels (<5% of the dose) and faecal excretion is not a major
borated by experiments using microsomes that express specific elimination pathway.[36]
CYP enzymes. cDNA-expressed CYP2D6 and CYP1A2
formed 4-hydroxy duloxetine, and CYP2D6, CYP1A2 and
3. Special Populations
CYP2C9 (at a relatively slow rate) formed 5-hydroxy dulox-
etine. CYP2D6 was identified as a low-affinity enzyme 3.1 Sex and Smoking Status
(Michaelis-Menten constant [Km] 1 mmol/L) responsible for the
formation of both 4-hydroxy and 5-hydroxy duloxetine. Population pharmacokinetic analyses of duloxetine con-
CYP1A2 was identified as a moderate-affinity enzyme (Km centrations in 594 patients with major depressive disorder
20 mmol/L). Therefore the in vitro data indicate both enzymes (n = 223), stress urinary incontinence (n = 128), diabetic neuro-
would be likely to participate in the biotransformation of du- pathic pain (n = 112) or fibromyalgia (n = 131) show that sex
loxetine to 4-hydroxy and 5-hydroxy duloxetine, but in vivo and smoking status impact duloxetine bioavailability.[25] The
clinical studies using potent CYP2D6[29] and CYP1A2 in- effect of sex and smoking status on duloxetine exposure is
hibitors[24] (described below) reveal that CYP1A2 is the more attributable to CYP1A2 metabolism since these factors affect
important enzyme. In vitro data indicate that the role of other the expression and activity of CYP1A2.[39,40] CYP1A2 activity
CYP enzymes is small and these enzymes would not be expected in women is lower than in men and this decrement in CYP1A2
to contribute to the in vivo metabolism of duloxetine. activity has an impact on the metabolism of duloxetine, re-
sulting in higher duloxetine systemic concentration in women
2.4 Elimination compared with men. Smoking increases the expression of
CYP1A2 and this increased expression is associated with a 30%
Duloxetine oral clearance (CL/F) following a single 60 mg decrease in duloxetine concentration in smokers compared with
dose was 101 L/h, which decreased to 57 L/h following 60 mg nonsmokers. The combined effect of sex and smoking status
twice-daily dosing at steady state, but was 97.1 L/h after 40 mg typically results in duloxetine concentrations for a male smoker
twice-daily dosing (tables I and II). Another report showed that that are 57% lower than the concentrations in a female non-
mean duloxetine CL/F was 114 L/h in healthy males[31] ad- smoker. Another study revealed similar findings to the popu-
ministered multiple doses ranging from 20 mg to 40 mg. The lation pharmacokinetic study with smokers showing lower
dose dependency in CL/F was observed in patients, where the mean plasma duloxetine concentrations than nonsmokers.[41]
effect of doubling the duloxetine dose from 30 mg to 60 mg or On a population basis the magnitude of change in duloxetine
from 60 mg to 120 mg resulted in 2.3 and 2.6 times the average concentrations due to sex and smoking status is notable, but in
concentration at steady state (Cav,ss), respectively.[25] Relative individual patients these effects have a smaller impact than
to the high interindividual variability (~60%) in CL/F, the the much larger random sources of variability (approximately
magnitude of nonlinearity in duloxetine exposure with respect 60% coefficient of variation) for clearance.[25] Consequently,
to dose was considered to be minor and dose adjustments are while the distributions of the pharmacokinetic parameters
not warranted for patients on long-term therapy. and observed duloxetine concentrations broadly reflect sex
Following administration of 14C-labelled duloxetine, ap- and smoking status differences, the large ranges for these val-
proximately 72% of the radioactivity was excreted in urine ues substantially overlap. Therefore, duloxetine pharmaco-
principally as the glucuronide and/or sulfate conjugates of the kinetics are remarkably similar between smokers and non-
oxidative duloxetine metabolites. The relatively small amount smokers, and between males and females. That is, while sex
of radioactivity found in faeces as duloxetine (<5% of the dose) and smoking status have a consistent rank-order impact on
was excreted by 96 hours after dosing. These results reflect the the typical (or average) pharmacokinetic parameter estimates
substantially complete oral absorption of the dose. It is note- for each subgroup (average duloxetine concentrations in fe-
worthy that the majority of faecal radioactivity was excreted male nonsmoker > male nonsmoker > female smoker > male
beyond simply gastrointestinal transit time, suggesting a role of smoker), differential dosing recommendations for individual
biliary excretion. The profile of faecal radioactivity showed that patients based solely on sex and smoking status are not
in addition to duloxetine, 4-hydroxy duloxetine and a meta- warranted.

ª 2011 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2011; 50 (5)
Duloxetine Pharmacokinetics and Drug Interactions 287

3.2 Age subjects with moderate hepatic impairment (Child-Pugh Class


B) had a mean plasma duloxetine exposure that was 5-fold
In elderly women (age 65–77 years), the rate of elimination of higher[32] and elimination took approximately 3 times longer
duloxetine was slower than that in younger women (age 32–50 compared to age- and sex-matched healthy subjects, but Cmax
years).[42] Results based on the analysis of a large patient pop- was unaffected.[45] Use of duloxetine in patients with hepatic
ulation also showed that age had a small effect on CL/F such insufficiency is not ordinarily recommended.
that duloxetine CL/F was lower among the older patients.
Relative to the Cav,ss for a 49-year-old patient (the median age),
3.5 Renal Impairment
the Cav,ss was 13% lower for a patient who was 29 years old (at
the 5th percentile of the age distribution) and Cav,ss was 17% Duloxetine and its metabolite concentrations were elevated
higher for a patient who was 69 years old (at the 95th percentile in subjects with end-stage renal disease (ESRD) requiring dia-
of the age distribution).[25] While these findings indicate that lysis. The Cmax and AUC of duloxetine after a single 60 mg dose
age has a small effect on duloxetine pharmacokinetics, the of duloxetine were approximately twice as high in subjects with
magnitude of these pharmacokinetic differences across age is ESRD than in subjects with normal renal function.[46] Given
not great enough to warrant a specific dosing recommendation that the t½ was unaffected, the higher duloxetine exposure in
for the elderly. ESRD appears to be due to an increase in the oral bioavaila-
bility, allowing a larger portion of the drug to reach the systemic
3.3 Ethnicity circulation on first pass. More directly related to reduced renal
function, the AUCs of the major circulating metabolites,
Single-dose pharmacokinetics in Caucasian and Japanese 4-hydroxy duloxetine glucuronide and 5-hydroxy-6-methoxy
subjects revealed that the mean duloxetine Cmax and AUC were duloxetine sulfate, were approximately 7- to 9-fold higher than
approximately 20% higher in Japanese subjects. This difference the metabolite exposure in control subjects with normal renal
is likely related to the 15% lower average bodyweight in Japa- function. The magnitude of these differences may potentially
nese subjects[26] as bodyweight normalized exposures (Cmax, increase upon multiple-dose administration because the con-
AUC) were similar in Caucasians and Japanese subjects. jugated metabolites of duloxetine are principally excreted in
Conclusions could not be drawn from this study as to the effects urine.[36] Given the increase in the exposure of duloxetine and
of factors other than bodyweight, such as CYP2D6 poor me- metabolites, use of duloxetine is not ordinarily recommended
tabolizer status or smoking status, because most subjects in for patients with ESRD or for patients with severe renal impair-
both groups were CYP2D6 extensive metabolizers and non- ment (creatinine clearance [CLCR] <30 mL/min). Population
smokers. Similar small pharmacokinetic differences were also pharmacokinetic analyses show that patients with mild to moderate
observed following multiple doses of duloxetine.[26] In Chinese degrees of renal dysfunction (CLCR between 30 and 80 mL/min)
subjects, duloxetine pharmacokinetics were not statistically have no significant difference in the clearance of duloxetine,
significantly different from those reported previously in Cau- therefore a dose adjustment is unnecessary for these patients.[46]
casian and Japanese subjects.[43] Another study in Chinese
subjects found volume of distribution after oral administration
3.6 Lactation
(Vd/F), tmax, Cmax and AUC in Chinese subjects to agree with
other published studies.[44] After giving multiple doses of 40 mg of duloxetine twice daily
Population pharmacokinetic analyses found that Hispanics to lactating women, approximately 7 mg/day (ranging from
had double the Vd/F compared to non-Hispanics.[25] However, 4 to 15 mg/day) of duloxetine was secreted in their breast milk.
the effect of ethnic origin on the Vd/F did not have an impact on The estimated dose for an infant consuming breast milk is
duloxetine Cav,ss. Therefore specific dosing recommendations 2 mg/kg/day (ranging from 0.6 to 3 mg/kg/day), or approxi-
based on ethnicity are not warranted.[25] mately 0.14% of the maternal dose.[28] Although this amount of
duloxetine dosage for an infant is small, the safety of duloxetine
3.4 Hepatic Impairment in infants has not been systematically investigated and is cur-
rently unknown. If duloxetine is prescribed for mothers who
Patients with clinically evident hepatic insufficiency exhibit a may be nursing, the health care provider and the mother need to
substantial decrease in the ability to metabolize and eliminate assess the potential risks of exposing the infant to duloxetine
duloxetine. Following a single, 20 mg oral dose of duloxetine, compared with the benefits of nursing.

ª 2011 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2011; 50 (5)
288 Knadler et al.

4. Drug Interaction Evaluations Table III. In vitro cytochrome P450 (CYP) enzyme studies of the potential
inhibitory effect of duloxetine[24,38]
The evaluation of duloxetine drug interactions used a
Enzyme Ki (mmol/L) Cmax/Ki a
systematic approach that integrated information about du-
CYP2D6 2.4 0.2
loxetine physicochemical properties and data from in vitro en-
CYP1A2 17.7 <0.1
zyme studies and human clinical pharmacology studies.
CYP3A 133 <0.1
Specific drug interaction studies were designed based upon the
CYP2C9 306 <0.1
known metabolic pathways and the metabolic impact of du-
CYP2C19 7.1 <0.1
loxetine. The drug interaction studies conducted in healthy
subjects evaluated both the potential for duloxetine to affect CYP2C8 97 <0.1

other drugs and the potential for other drugs to affect dulox- a The Cmax value used was 0.48 mmol/L or 144 ng/mL after 60 mg twice-
daily dosing, the highest dose studied for efficacy.
etine. The drugs chosen for the studies were selected because of
Cmax = maximum plasma concentration; Ki = inhibition constant.
the impact on or sensitivity to specific CYP enzymes[47] rather
than the likelihood of their clinical use together with dulox-
etine. However, these specific probes provide a rigorous pharmacokinetic impact is unlikely if the Cmax/Ki ratio is <0.1.
evaluation of the potential for drug interactions and are re- For duloxetine, the Cmax/Ki ratio was 0.2 for CYP2D6 (table
presentative for many other drugs that likewise have an impact III), predicting that duloxetine would potentially affect the
on, or are affected by, specific CYP enzymes. A key aspect of pharmacokinetics of drugs metabolized by CYP2D6. In vivo
this series of studies was to coordinate results from in vitro studies were conducted with the known CYP2D6 substrates
enzyme studies with in vivo human clinical studies. desipramine, metoprolol and tolterodine (see below).
Specific interactions for duloxetine involving transporters
were not assessed in vitro or in vivo and no data on this topic 4.1.2 CYP Induction
were found in the literature. Nonetheless, it is informative to The ability of duloxetine to induce the catalytic activities
know that the equilibration of duloxetine between the blood associated with CYP1A2 and CYP3A was examined in primary
and brain of rats was rapid and reversible. In rats, duloxetine cultures of human hepatocytes. Primary cultures of human
concentrations were distinctly parallel between plasma and hepatocytes were treated for 48 hours with duloxetine at con-
brain across time.[2] These results indicate rapid equilibration of centrations ranging from 0.01 mmol/L to 100 mmol/L, and the
duloxetine into and out of the brain and that this process most effects of treatment on catalytic activities associated with
likely occurs via passive diffusion which is thought to be the CYP1A2 and CYP3A were compared to those activities in
predominant mechanism for duloxetine disposition. Therefore vehicle-treated control cultures (0.1% dimethyl sulfoxide). There
transporter-based drug-drug interactions are considered un- was no significant induction of CYP1A2[24] or CYP3A[32,38] in
likely. Since duloxetine is cleared primarily by metabolism with any of the three preparations of cultured human hepatocytes
minimal renal excretion, an interaction with renal transporters examined; therefore it is unlikely that duloxetine induces these
is also unlikely. enzymes when used clinically.

4.1 In Vitro Studies 5. Clinical Pharmacokinetic Drug Interaction Studies

4.1.1 Cytochrome P450 (CYP) Inhibition


5.1 Potential for Other Drugs to Affect Duloxetine
In vitro studies conducted using human liver microsomes
assessed the potential for duloxetine to inhibit major CYP en- 5.1.1 Agents Affecting Gastric pH and Activated Charcoal
zymes (CYP1A2, CYP2C8, CYP2C9, CYP2C19, CYP2D6 and A gelatin capsule is used to contain an enteric-coated pellet
CYP3A4).[24,38] The lowest Ki value of 2.4 mmol/L was obtained formulation of duloxetine hydrochloride.[32] The enteric-coated
for the effect of duloxetine on CYP2D6 (table III), suggesting pellets protect the acid-labile drug from normal acidic condi-
the possibility that duloxetine may inhibit CYP2D6 activity. tions of the stomach and they immediately release duloxetine
This assessment used the established criteria[47] that CYP in- hydrochloride for dissolution in the more alkaline conditions of
hibition will likely make a substantial pharmacokinetic impact the small intestine. Each capsule contains a multitude of small
if the Cmax/Ki ratio is >1, CYP inhibition will possibly make an enteric-coated pellets that are dispersed in the gastrointestinal
impact if the Cmax/Ki ratio is between 0.1 and 1, and a tract, leading to a more consistent performance than the per-

ª 2011 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2011; 50 (5)
Duloxetine Pharmacokinetics and Drug Interactions 289

formance that would be obtained with a monolithic enteric- duloxetine study who were identified as CYP2D6 intermediate
coated tablet. However, all enteric-coated formulations are metabolizers had duloxetine exposures slightly higher (16%)
responsive to changes in gastrointestinal pH. Therefore specific than the exposures in extensive metabolizers;[43] however, this
pharmacokinetic studies were conducted to assess the impact of difference is not clinically meaningful. In another study with
change in gastrointestinal pH on the duloxetine formulation. Japanese and Caucasian subjects, four subjects were genotyped
Drugs like famotidine (an H2-receptor antagonist) or alumi- as CYP2D6 poor metabolizers; two of the poor metabolizers
nium hydroxide/magnesium hydroxide/simethicone (Mylanta; had duloxetine exposures 1- to 3-fold higher than other subjects
an antacid) increase the pH in the gastrointestinal tract and are in the study who were not poor metabolizers and two poor
likely to facilitate the release and early absorption of duloxetine metabolizers had duloxetine exposures comparable to the other
after oral administration. However, these drugs did not change subjects.[26] These results indicate that exposure cannot be
the pharmacokinetics of duloxetine, suggesting that the chan- predicted by CYP2D6 metabolism status alone and factors such
ges in gastrointestinal pH due to these drugs have no impact on as CYP1A2 activity predict exposure more substantially.[26]
the rate or onset or extent of absorption. In contrast, activated Therefore in patients with a CYP2D6 poor metabolizer phe-
charcoal significantly decreased duloxetine Cmax by approxi- notype, a lower dose of duloxetine is not necessary.
mately 32% and AUC by approximately 35%, likely resulting
from the binding of duloxetine to the activated charcoal when 5.1.3 Effect of CYP1A2 Inhibition
duloxetine was released into the gastrointestinal content. These Duloxetine was administered both intravenously and orally
results indicate a potential benefit of using activated charcoal to healthy male CYP2D6 extensive metabolizers in the presence
early in the treatment of a duloxetine overdose to decrease the and absence of the potent CYP1A2 inhibitor fluvoxamine.[24]
amount of duloxetine that may be absorbed.[48] Following oral administration in the presence of fluvoxamine,
the AUC1 and Cmax of duloxetine significantly increased by
5.1.2 Effect of CYP2D6 Inhibition/Genetic Polymorphism 460% (90% CI 359, 584) and 141% (90% CI 93, 200), respect-
The co-administration of duloxetine with paroxetine, a po- ively. In the presence of fluvoxamine, the oral bioavailability of
tent CYP2D6 inhibitor, increased both the mean Cmax and duloxetine increased from 42.8% to 81.9%. The magnitude of
AUC of duloxetine by approximately 60%.[29] Although this increase in AUC and Cmax for duloxetine co-administered with
increase in exposure falls outside of the standard no-effect a potent CYP1A2 inhibitor compared with a potent CYP2D6
boundary (0.8–1.25), the clinical relevance of the increased inhibitor suggests that CYP1A2 is the predominant enzyme
concentration would depend on individual tolerance to du- responsible for duloxetine metabolism. Therefore co-adminis-
loxetine across a range of concentrations (i.e. the therapeutic tration of duloxetine with a potent CYP1A2 inhibitor (e.g.
index of the drug). Therefore the effect of paroxetine on du- fluvoxamine, some quinolone antibacterials) should be avoided
loxetine concentrations seen in this study may not necessarily and is contraindicated in some labels such as the European
be manifested by a significant change in tolerance. Further- Union summary of product characteristics (SPC). No studies
more, since CYP1A2 provides the primary pathway for du- have been reported in which duloxetine was administered at a
loxetine metabolism even though there is evidence of CYP2D6 lower dose with a potent CYP1A2 inhibitor to assess if dose
metabolism for duloxetine, the effect of CYP2D6 inhibition adjustments of duloxetine in the presence of CYP1A2 inhibitors
alone on duloxetine pharmacokinetics is considered to be less could be safely administered.
important when compared with the much larger impact of
CYP1A2 inhibition (see below). Therefore a dose adjustment 5.1.4 Effect of CYP1A2 Inhibition and CYP2D6 Genotype
does not appear to be necessary when duloxetine is co- Duloxetine was given to CYP2D6 poor metabolizer subjects
administered even with potent CYP2D6 inhibitors such as along with the potent CYP1A2 inhibitor, fluvoxamine, to
paroxetine. evaluate the effect of inhibiting both CYP1A2 and CYP2D6.[49]
A commonly recognized polymorphism in drug metabolism At steady state, there was a clinically significant increase in
is the absence of CYP2D6 activity in a small segment of the duloxetine AUC (540%) and Cmax (471%) in the presence of
population. CYP2D6 poor metabolizers represent approxi- fluvoxamine (see figure 4). The magnitude of increase in du-
mately 5–10% of the Caucasian population and they lack loxetine exposure is similar to that caused by CYP1A2 inhibi-
CYP2D6 activity due to a variety of genetic mutations of the tion alone, reflecting the predominant role of CYP1A2 in
gene responsible for the formation of this enzyme. This per- duloxetine metabolism. The effect of fluvoxamine on duloxetine
centage differs among ethnic groups. Chinese subjects in a metabolism was similar but of smaller magnitude following

ª 2011 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2011; 50 (5)
290 Knadler et al.

Duloxetine (n = 14) duloxetine with metoprolol, a CYP2D6 substrate, increased the


Glucuronide conjugate of 4-hydroxy duloxetine (n = 14)
Sulfate conjugate of 5-hydroxy-6-methoxy duloxetine (n = 14) metoprolol Cmax and t½ and decreased its clearance, leading to
800 a 180% increase in metoprolol AUC.[52] This increase in AUC
Trough plasma concentration (ng/mL)

was found to be greater than the increase in the AUC for me-
600
toprolol caused by escitalopram (89%) or sertraline (48% and
67%). However, the baseline metoprolol concentrations were
lower prior to dosing with duloxetine than with either escita-
400 lopram or sertraline, which may have resulted in the percent
change in metoprolol concentrations with duloxetine being
unusually high.[52] In general, escitalopram is considered a
200
weak inhibitor of CYP2D6, sertraline is considered a mild to
moderate inhibitor of CYP2D6 and duloxetine is considered a
0 moderate inhibitor of CYP2D6.[49]
0 2 4 6 8 10 12 14 16 18 20
Collectively, results from these drug interaction studies with
Time (d)
three CYP2D6 substrates (desipramine, tolterodine and me-
Fig. 4. Mean predose trough plasma concentrations of duloxetine and its toprolol) indicate that duloxetine is a moderately potent in-
two major metabolites in healthy cytochrome P450 2D6 poor metabolizers
hibitor of CYP2D6. Therefore caution is advised when
(n = 13–14) during twice-daily dosing of duloxetine 40 mg. Fluvoxamine was
given once daily in the evening: 50 mg on days 5 and 6, 100 mg on days 7 duloxetine is co-administered with drugs that are extensively
through 21. metabolized by CYP2D6 and have a narrow therapeutic index,
including certain tricyclic antidepressants (e.g. nortriptyline,
intravenous administration, suggesting that fluvoxamine in- amitriptyline and imipramine), phenothiazines and certain type
hibition of CYP1A2 affects both the first-pass and systemic 1C antiarrhythmics (e.g. propafenone and flecainide).
metabolism of duloxetine. In addition, the study showed that Atypical antipsychotics and antidepressant drugs such as
the steady-state Cmax and AUC values for the two major cir- duloxetine are commonly co-administered.[53-55] Some of the
culating duloxetine metabolites were decreased by about 50%. antipsychotic drugs such as risperidone and aripiprazole are
These results further emphasize that the co-administration of CYP2D6 substrates, so co-administration with duloxetine may
duloxetine with a potent CYP1A2 inhibitor should be avoided. result in higher plasma concentrations of these antipsychotics,
However, the impact of this important CYP1A2 drug inter- but not for other antipsychotics such as olanzapine and que-
action is not substantially greater for patients also taking a tiapine which are not CYP2D6 substrates.
potent CYP2D6 inhibitor or for patients having the genetic
characteristics of a CYP2D6 poor metabolizer. 5.2.2 Effects on CYP1A2 Substrates
The pharmacokinetics of theophylline, a CYP1A2 substrate,
were not significantly affected by duloxetine in either men or
5.2 Potential for Duloxetine to Affect Other Drugs
women.[24] Further, duloxetine did not have any effect on the
5.2.1 Effects on CYP2D6 Substrates urinary metabolite profiles of theophylline. As duloxetine does
The investigations of the effects of duloxetine on CYP2D6 not appear to be a clinically significant inhibitor or inducer
metabolism produced varied outcomes. When given as 60 mg of CYP1A2, dose adjustment of CYP1A2 substrates is not
twice-daily oral doses, duloxetine increased the mean AUC of necessary when such drugs are co-administered with duloxetine.
desipramine, a sensitive CYP2D6 substrate, by 192%.[29] Doses
of 30 mg twice daily increased the mean AUC of desipramine 5.3 Pharmacodynamic Interactions
122%.[50] Co-administration of 40 mg of duloxetine twice daily
with the CYP2D6 substrate tolterodine (2 mg twice daily) in- The possibility for a potentially clinically relevant pharma-
creased tolterodine steady state AUC and Cmax by 71% and codynamic interaction may be assessed by examining the
64%, respectively, and prolonged tolterodine t½ by 14%. Du- known pharmacology of each drug being considered for co-
loxetine did not affect the Cmax or AUC of the active 5-hy- administration. The adverse events reported at the highest rate
droxylmethyl tolterodine metabolite,[51] which is consistent during duloxetine use are consistent with the known pharma-
with the established fact that this clinically important tolter- cology of duloxetine rather than specific to an indication.[6]
odine metabolite is not a CYP2D6 substrate. Administration of Therefore the possibility for additive adverse effects warrants

ª 2011 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2011; 50 (5)
Duloxetine Pharmacokinetics and Drug Interactions 291

caution when other drugs are co-administered with duloxetine. Alcohol alone decreased performance on six of nine psycho-
Across indications, common adverse events reported with du- metric tests and the addition of duloxetine did not exacerbate
loxetine use include nausea, headache, dry mouth, insomnia, the cognitive and psychomotor impairment produced by alco-
constipation, dizziness, fatigue, somnolence, diarrhoea and hol. Because it is possible that duloxetine and alcohol may in-
hyperhidrosis.[6,32] teract to cause liver injury or that duloxetine may aggravate
Consistent with its adrenergic pharmacology, duloxetine pre-existing liver disease, duloxetine should ordinarily not be
produces modest increases in blood pressure and pulse rates[56] prescribed to patients with substantial alcohol use or evidence
that may be clinically important when duloxetine is used in of chronic liver disease.[32]
combination with another drug that produces similar effects.
Particularly important to consider in this regard are other 5.3.3 Anticoagulant Drugs
serotonergic drugs, which might potentiate a pharmacodyna- Recent reports have suggested an interaction between du-
mic interaction and serotonin syndrome, as described below. loxetine and anticoagulant drugs, leading to unexpected al-
Since there is a possibility for orthostatic effects with dulox- terations in the international normalized ratio (INR). In one
etine,[6,56] caution is warranted for combined use of duloxetine case, a 63-year-old woman on stable acenocoumarol therapy
with agents, such as diuretics, that might exacerbate this effect experienced a decrease in INR values from 2.58 to 1.49 after
in some patients. taking a single 60 mg dose of duloxetine.[61] The INR values
Electrocardiogram evaluations in clinical studies and a remained low for 3 weeks after discontinuation of duloxetine,
thorough QT study at supratherapeutic dosages indicate that and then returned to baseline. In another report, a 44-year old
duloxetine has no adverse effect on cardiac repolarization.[57,58] woman taking stable warfarin therapy experienced an increase
Therefore it is unlikely that duloxetine would increase the risk in INR after initiating daily therapy with duloxetine 30 mg and
for torsades de pointes when used with a concomitant QT- the INR increased even further after warfarin was dis-
prolonging drug. continued. Upon discontinuation of duloxetine, INR decreased
Because of the pharmacology of duloxetine as a central and warfarin treatment was reinstated.[62] Based on the differ-
nervous system (CNS)-acting drug, or because of concern ing metabolic pathways of duloxetine and warfarin (primarily
for a potential pharmacokinetic/pharmacodynamic interaction, via CYP2C9), a pharmacokinetic interaction does not seem a
specific studies with benzodiazepines, alcohol and warfarin likely explanation for these changes in INR values.
have been conducted to evaluate a pharmacodynamic inter- Given the known effects of serotonin inhibitors on platelet
action with duloxetine. aggregation which could lead to an increased risk of bleeding[63]
and the involvement of serotonin in the mechanism of action
5.3.1 Benzodiazepines for duloxetine, a pharmacodynamic interaction between du-
The potential for a pharmacodynamic interaction with loxetine and warfarin was considered possible prior to the
benzodiazepines was investigated using lorazepam, a drug that conduct of a drug interaction study to determine the impact of
is metabolized primarily via glucuronidation rather than via duloxetine on INR. Duloxetine 60 mg/day or 120 mg/day was
CYPs. The primary finding of this investigation was increased co-administered daily with warfarin for 14 days to healthy
sedation on subjective and objective tests in subjects who re- subjects who had a stable INR with an individualized dose of
ceived co-administration of lorazepam and duloxetine.[59] Due warfarin (2–9 mg) prior to the combined dosing period. The
to the potential for increased CNS effects when duloxetine is INR was determined daily during co-administration of duloxetine
co-administered with other CNS-acting drugs such as the benzo- and warfarin and duloxetine had no statistically or clinically
diazepines, caution should be used when co-administering significant effect on the anticoagulant effects of warfarin.[64] In
duloxetine with other CNS-acting drugs.[32] addition, duloxetine did not affect the pharmacokinetics of
either R- or S-warfarin. Since study results show neither a
5.3.2 Alcohol pharmacokinetic interaction nor a pharmacodynamic inter-
The pharmacodynamic effect of the combination of dulox- action between duloxetine and warfarin, a clear explanation of
etine and alcohol was assessed in male and female subjects the changes in INR in the two case reports cannot be made.
administered 60 mg of duloxetine alone, alcohol alone and
duloxetine with alcohol (blood alcohol level of approximately 5.3.4 Serotonin Syndrome
0.1 g/dL).[60] Pharmacodynamic assessments included computer- The combined use of serotonergic drugs with duloxetine has
based performance tests and self-administered questionnaires. the potential for additive pharmacodynamic effects that could

ª 2011 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2011; 50 (5)
292 Knadler et al.

lead to serotonin toxicities.[65] A case is reported of serotonin severe renal disease or ESRD, or in patients with hepatic im-
syndrome in a patient on duloxetine treatment when linezolid, pairment. The potential for pharmacodynamic interactions
an antibacterial that inhibits monoamine oxidase, was co- with duloxetine include co-administration with benzodiazepines
administered.[66] While the patient was on multiple other and MAOIs.
medications for treatment of sarcoma of the lower extremity, Overall, interpatient variability in duloxetine pharmaco-
the authors attributed the serotonin syndrome to linezolid due kinetics is high and a robust pharmacokinetic and pharmaco-
to its inhibition of monoamine oxidase. In another case report, dynamic relationship has not been established that provides a
a patient who had been on long-term treatment with moclo- targeted concentration range associated with efficacy or mini-
bemide (300–600 mg/day) discontinued moclobemide treat- mization of adverse events. Although some have suggested that
ment (600 mg/day) and began taking duloxetine (60 mg/day) the therapeutic drug monitoring can be conducted for dulox-
next morning. Shortly after the first duloxetine dose, the patient etine[68] in order to maximize therapeutic effects and minimize
began to experience intense restlessness, generalized tremor, side effects for an individual patient, dose adjustments should
dizziness, nausea, headache and tic-like facial movements. On principally be made on the basis of an individual patient’s
the second day of duloxetine dosing the patient experienced an clinical response and known factors that impact duloxetine
increase in blood pressure. The patient’s symptoms were gone concentrations. In cases where an individual patient is not re-
within 2 days following discontinuation of duloxetine.[67] sponding to therapy, therapeutic drug monitoring may be
Duloxetine should not be co-administered with monoamine useful to identify a noncompliant patient from a nonresponder.
oxidase inhibitors or other serotonergic drugs, including trip-
tans and drugs with secondary serotonergic properties such as
Acknowledgements
tramadol.[32] In addition, care should be taken in transitioning
patients from a selective serotonin reuptake inhibitor (SSRI) The authors acknowledge Ryan Wright’s contribution in providing
or another selective serotonin and norepinephrine reuptake writing support for this manuscript.
This review was funded by Eli Lilly and Company (Indianapolis, IN,
inhibitor (SNRI) to duloxetine. Finally, because duloxetine is
USA), the manufacturer of duloxetine. All authors have read the manu-
approved for use in certain non-psychiatric indications such as script and conflict of interest statement and approved the version sub-
the treatment of stress urinary incontinence, it is important that mitted for publication. All authors are current or former employees of Eli
prescribers consider the patient’s medication profile which may Lilly and Company; Richard Bergstrom is retired from Eli Lilly and
include monoamine oxidase inhibitor (MAOI), SSRI or SNRI Company and now works at Butler University College of Pharmacy (In-
drugs. dianapolis, IN, USA). All authors are stock owners in Eli Lilly and
Company and have participated in the development of duloxetine.

6. Conclusions
References
Duloxetine is cleared primarily by metabolism via CYP1A2 1. Bymaster FP, Dreshfield-Ahmad LJ, Threlkeld PG, et al. Comparative affinity
of duloxetine and venlafaxine for serotonin and norepinephrine transporters
and the resulting inactive metabolites are cleared after con- in vitro and in vivo, human serotonin receptor subtypes, and other neuronal
jugation by renal excretion. Duloxetine is a moderately potent receptors. Neuropsychopharmacology 2001; 25 (6): 871-80

inhibitor of CYP2D6, but does not significantly inhibit other 2. Bymaster FP, Lee TC, Knadler MP, et al. The dual transporter inhibitor du-
loxetine: a review of its preclinical pharmacology, pharmacokinetic profile,
CYP enzymes. In comparison with SSRIs, duloxetine is a and clinical results in depression. Curr Pharm Des 2005; 11 (12): 1475-93
weaker inhibitor of CYP2D6 than paroxetine and fluoxetine, 3. Turcotte JE, Debonnel G, de MC, et al. Assessment of the serotonin and
but a stronger inhibitor than escitalopram. Results from the norepinephrine reuptake blocking properties of duloxetine in healthy sub-
jects. Neuropsychopharmacology 2001; 24 (5): 511-21
drug interaction studies that have been performed with du-
4. Wong DT, Bymaster FP, Mayle DA, et al. LY248686, a new inhibitor of
loxetine and information on the pharmacokinetic properties of serotonin and norepinephrine uptake. Neuropsychopharmacology 1993;
potential concomitant drugs may be used to inform clinical 8 (1): 23-33
judgement as to the potential for drug-drug interactions with 5. Chalon SA, Granier LA, Vandenhende FR, et al. Duloxetine increases sero-
tonin and norepinephrine availability in healthy subjects: a double-blind,
duloxetine. controlled study. Neuropsychopharmacology 2003; 28 (9): 1685-93
Situations in which the pharmacokinetics of duloxetine are 6. Gahimer J, Wernicke J, Yalcin I, et al. A retrospective pooled analysis of
altered to a clinically significant extent include the co-admin- duloxetine safety in 23,983 subjects. Curr Med Res Opin 2007; 23 (1): 175-84
istration of duloxetine with potent CYP1A2 inhibitors, the 7. Berk M, du Plessis AD, Birkett M, et al. An open-label study of duloxetine
hydrochloride, a mixed serotonin and noradrenaline reuptake inhibitor, in
co-administration of CYP2D6 substrates that have a narrow patients with DSM-III-R major depressive disorder. Lilly Duloxetine De-
therapeutic index and the usage of duloxetine in patients with pression Study Group. Int Clin Psychopharmacol 1997; 12 (3): 137-40

ª 2011 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2011; 50 (5)
Duloxetine Pharmacokinetics and Drug Interactions 293

8. Detke MJ, Lu Y, Goldstein DJ, et al. Duloxetine, 60 mg once daily, for major 29. Skinner MH, Kuan HY, Pan A, et al. Duloxetine is both an inhibitor and a
depressive disorder: a randomized double-blind placebo-controlled trial. substrate of cytochrome P4502D6 in healthy volunteers. Clin Pharmacol Ther
J Clin Psychiatry 2002; 63 (4): 308-15 2003; 73 (3): 170-7
9. Detke MJ, Lu Y, Goldstein DJ, et al. Duloxetine 60 mg once daily dosing 30. Skinner MH, Skerjanec A, Seger ME, et al. The effect of food and bedtime
versus placebo in the acute treatment of major depression. J Psychiatr Res administration on duloxetine pharmacokinetics [abstract]. Clin Pharmacol
2002; 36 (6): 383-90 Ther 2000; 67 (2): 129
10. Detke MJ, Wiltse CG, Mallinckrodt CH, et al. Duloxetine in the acute and 31. Sharma A, Goldberg MJ, Cerimele BJ. Pharmacokinetics and safety of du-
long-term treatment of major depressive disorder: a placebo- and paroxetine- loxetine, a dual-serotonin and norepinephrine reuptake inhibitor. J Clin
controlled trial. Eur Neuropsychopharmacol 2004; 14 (6): 457-70 Pharmacol 2000; 40 (2): 161-7
11. Goldstein DJ, Lu Y, Detke MJ, et al. Duloxetine in the treatment of depression: 32. Cymbalta (duloxetine hydrochloride) capsules [package insert]. Indianapolis
a double-blind placebo-controlled comparison with paroxetine. J Clin Psy- (IN): Eli Lilly Pharmaceuticals, 2008
chopharmacol 2004; 24 (4): 389-99 33. Benet LZ, Hoener BA. Changes in plasma protein binding have little clinical
12. Nelson JC, Wohlreich MM, Mallinckrodt CH, et al. Duloxetine for the relevance. Clin Pharmacol Ther 2002; 71 (3): 115-21
treatment of major depressive disorder in older patients. Am J Geriatr Psy- 34. Rolan PE. Plasma protein binding displacement interactions: why are they still
chiatry 2005; 13 (3): 227-35 regarded as clinically important? Br J Clin Pharmacol 1994; 37 (2): 125-8
13. Nemeroff CB, Schatzberg AF, Goldstein DJ, et al. Duloxetine for the treat- 35. Takano A, Suzuki K, Kosaka J, et al. A dose-finding study of duloxetine based
ment of major depressive disorder. Psychopharmacol Bull 2002; 36 (4): on serotonin transporter occupancy. Psychopharmacology (Berl) 2006; 185
106-32 (3): 395-9
14. Goldstein DJ, Lu Y, Detke MJ, et al. Duloxetine vs placebo in patients with 36. Lantz RJ, Gillespie TA, Rash TJ, et al. Metabolism, excretion, and pharm-
painful diabetic neuropathy. Pain 2005; 116 (1-2): 109-18 acokinetics of duloxetine in healthy human subjects. Drug Metab Dispos
15. Raskin J, Pritchett YL, Wang F, et al. A double-blind, randomized multicenter 2003; 31 (9): 1142-50
trial comparing duloxetine with placebo in the management of diabetic per- 37. Kuo F, Gillespie TA, Kulanthaivel P, et al. Synthesis and biological activity of
ipheral neuropathic pain. Pain Med 2005; 6 (5): 346-56 some known and putative duloxetine metabolites. Bioorg Med Chem Lett
16. Allgulander C, Hartford J, Russell J, et al. Pharmacotherapy of generalized 2004; 14 (13): 3481-6
anxiety disorder: results of duloxetine treatment from a pooled analysis of 38. Ring B, Gillespe JS, Kasper SC, et al. Interaction of duloxetine with human
three clinical trials. Curr Med Res Opin 2007 Jun; 23 (6): 1245-52 cytochrome P450 [abstract]. Drug Metab Rev 2003; 35 (S2): 181
17. Hartford J, Kornstein S, Liebowitz M, et al. Duloxetine as an SNRI treatment 39. Relling MV, Lin JS, Ayers GD, et al. Racial and gender differences in N-
for generalized anxiety disorder: results from a placebo and active-controlled acetyltransferase, xanthine oxidase, and CYP1A2 activities. Clin Pharmacol
trial. Int Clin Psychopharmacol 2007; 22 (3): 167-74 Ther 1992; 52 (6): 643-58
18. Russell JM, Weisberg R, Fava M, et al. Efficacy of duloxetine in the treatment 40. Schrenk D, Brockmeier D, Morike K, et al. A distribution study of CYP1A2
of generalized anxiety disorder in patients with clinically significant pain phenotypes among smokers and non-smokers in a cohort of healthy Cauca-
symptoms. Depress Anxiety 2008; 25 (7): E1-11 sian volunteers. Eur J Clin Pharmacol 1998; 53 (5): 361-7
19. Rynn M, Russell J, Erickson J, et al. Efficacy and safety of duloxetine in the 41. Fric M, Pfuhlmann B, Laux G, et al. The influence of smoking on the serum
treatment of generalized anxiety disorder: a flexible-dose, progressive-titra- level of duloxetine. Pharmacopsychiatry 2008; 41 (4): 151-5
tion, placebo-controlled trial. Depress Anxiety 2008; 25 (3): 182-9
42. Skinner MH, Kuan HY, Skerjanec A, et al. Effect of age on the pharmaco-
20. Arnold LM, Lu Y, Crofford LJ, et al. A double-blind, multicenter trial com- kinetics of duloxetine in women. Br J Clin Pharmacol 2004; 57 (1): 54-61
paring duloxetine with placebo in the treatment of fibromyalgia patients
43. Tianmei S, Knadler MP, Lim MT, et al. Pharmacokinetics and tolerability of
with or without major depressive disorder. Arthritis Rheum 2004; 50 (9):
duloxetine following oral administration to healthy Chinese subjects. Clin
2974-84
Pharmacokinet 2007; 46 (9): 767-75
21. Guay DR. Duloxetine for management of stress urinary incontinence. Am J
44. Ma N, Zhang BK, Li HD, et al. Determination of duloxetine in human plasma
Geriatr Pharmacother 2005; 3 (1): 25-38
via LC/MS and subsequent application to a pharmacokinetic study in healthy
22. Millard RJ, Moore K, Rencken R, et al. Duloxetine vs placebo in the treatment Chinese volunteers. Clin Chim Acta 2007; 380 (1-2): 100-5
of stress urinary incontinence: a four-continent randomized clinical trial. BJU
45. Suri A, Reddy S, Gonzales C, et al. Duloxetine pharmacokinetics in cirrhotics
Int 2004; 93 (3): 311-8
compared with healthy subjects. Int J Clin Pharmacol Ther 2005; 43 (2): 78-84
23. Wernicke JF, Gahimer J, Yalcin I, et al. Safety and adverse event profile of
46. Lobo ED, Heathman M, Kuan HY, et al. Analysis of a single-dose phase I
duloxetine. Expert Opin Drug Saf 2005; 4 (6): 987-93
study and pooled steady-state data from phase II/III trials. Clin Pharmaco-
24. Lobo ED, Bergstrom RF, Reddy S, et al. In vitro and in vivo evaluations of kinet 2010; 49 (5): 311-21
cytochrome P450 1A2 interactions with duloxetine. Clin Pharmacokinet
47. Bjornsson TD, Callaghan JT, Einolf HJ, et al. The conduct of in vitro and in
2008; 47 (3): 191-202
vivo drug-drug interaction studies: a PhRMA perspective. J Clin Pharmacol
25. Lobo ED, Quinlan T, O’Brien L, et al. Population pharmacokinetics of orally 2003; 43 (5): 443-69
administered duloxetine in patients: implications for dosing recommendation.
48. Sathirakul K, Teng L, Yeo KP, et al. Impact of gastric pH and the presence of
Clin Pharmacokinet 2009; 48 (3): 189-97 activated charcoal on the absorption of duloxetine [abstract]. Clin Pharmacol
26. Chan C, Yeo KP, Pan AX, et al. Duloxetine pharmacokinetics are similar in Ther 2002; 71 (2): 18
Japanese and Caucasian subjects. Br J Clin Pharmacol 2007; 63 (3): 310-4 49. Small D, Loghin C, Lucas R, et al. Pharmacokinetic evaluation of combined
27. Kuan HY, Reddy S, Zhang L, et al. Duloxetine single- and multiple-dose duloxetine and fluvoxamine dosing in CYP2D6 poor metabolizers [abstract].
pharmacokinetics in healthy subjects [abstract]. Annual Meeting, American Clin Pharmacol Ther 2005; 77 (2): 37
Association of Pharmaceutical Scientists; 2002 Nov 10-14; Toronto (ON) 50. Patroneva A, Connolly SM, Fatato P, et al. An assessment of drug-drug in-
28. Lobo ED, Loghin C, Knadler MP, et al. Pharmacokinetics of duloxetine in teractions: the effect of desvenlafaxine and duloxetine on the pharmaco-
breast milk and plasma of healthy postpartum women. Clin Pharmacokinet kinetics of the CYP2D6 probe desipramine in healthy subjects. Drug Metab
2008; 47 (2): 103-9 Dispos 2008; 36 (12): 2484-91

ª 2011 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2011; 50 (5)
294 Knadler et al.

51. Hua TC, Pan A, Chan C, et al. Effect of duloxetine on tolterodine 61. Monastero R, Camarda R, Camarda C. Potential drug-drug interaction
pharmacokinetics in healthy volunteers. Br J Clin Pharmacol 2004; 57 (5): between duloxetine and acenocoumarol in a patient with Alzheimer’s disease.
652-6 Clin Ther 2007; 29 (12): 2706-9
52. Preskorn SH, Greenblatt DJ, Flockhart D, et al. Comparison of duloxetine, 62. Glueck CJ, Khalil Q, Winiarska M, et al. Interaction of duloxetine and war-
escitalopram, and sertraline effects on cytochrome P450 2D6 function in farin causing severe elevation of international normalized ratio. JAMA 2006;
healthy volunteers. J Clin Psychopharmacol 2007; 27 (1): 28-34 295 (13): 1517-8
53. de LJ, Armstrong SC, Cozza KL. The dosing of atypical antipsychotics. Psy- 63. de Abajo FJ, Rodriguez LA, Montero D. Association between selective sero-
chosomatics 2005; 46 (3): 262-73 tonin reuptake inhibitors and upper gastrointestinal bleeding: population
54. Englisch S, Knopf U, Scharnholz B, et al. Duloxetine for major depressive based case-control study. BMJ 1999; 319 (7217): 1106-9
episodes in the course of psychotic disorders: an observational clinical trial. 64. Chappell J, He J, Knadler MP, et al. Effects of duloxetine on the pharmaco-
J Psychopharmacol 2009; 23 (8): 875-82 dynamics and pharmacokinetics of warfarin at steady-state in healthy sub-
55. Sheffrin M, Driscoll HC, Lenze EJ, et al. Pilot study of augmentation with jects. J Clin Pharmacol 2009; 49 (12): 1456-66
aripiprazole for incomplete response in late-life depression: getting to remis- 65. Baldessarini RJ. Drugs and the treatment of psychiatric disorders: depression
sion. J Clin Psychiatry 2009; 70 (2): 208-13 and anxiety. In: Hardman JG, Limbird LE, Gilman AG, editors. Goodman
56. Derby MA, Zhang L, Chappell JC, et al. The effects of supratherapeutic doses and Gilman’s: the pharmacological basis of therapeutics. New York:
of duloxetine on blood pressure and pulse rate. J Cardiovasc Pharmacol 2007; McGraw-Hill, 2001: 447-83
49 (6): 384-93 66. Strouse TB, Kerrihard TN, Forscher CA, et al. Serotonin syndrome pre-
57. Wernicke J, Lledo A, Raskin J, et al. An evaluation of the cardiovascular safety cipitated by linezolid in a medically ill patient on duloxetine. J Clin Psycho-
profile of duloxetine: findings from 42 placebo-controlled studies. Drug Saf pharmacol 2006; 26 (6): 681-3
2007; 30 (5): 437-55 67. Jimenez-Genchi A. Immediate switching from moclobemide to duloxetine may
58. Zhang L, Chappell J, Gonzales CR, et al. QT effects of duloxetine at su- induce serotonin syndrome. J Clin Psychiatry 2006; 67 (11): 1821-2
pratherapeutic doses: a placebo and positive controlled study. J Cardiovasc 68. Waldschmitt C, Vogel F, Pfuhlmann B, et al. Duloxetine serum concentrations
Pharmacol 2007; 49 (3): 146-53 and clinical effects: data from a therapeutic drug monitoring (TDM) survey.
59. Chalon SA, Vandenhende F, Ertle S. Combined administration of duloxetine Pharmacopsychiatry 2009; 42 (5): 189-93
(DU) and lorazepam (LO): a pharmacokinetic (PK) and pharmacodynamic
(PD) study [abstract]. Clin Pharmacol Ther 2005; 77: 65
Correspondence: Mary Pat Knadler, PhD, Eli Lilly and Company, Lilly
60. Skinner MH, Weerakkody G. Duloxetine does not exacerbate the effects of
alcohol on psychometric tests [abstract]. Clin Pharmacol Ther 2002; 71 (2): Corporate Center, Indianapolis, IN 46285, USA.
P53 E-mail: Knadler_Mary_Pat@lilly.com

ª 2011 Adis Data Information BV. All rights reserved. Clin Pharmacokinet 2011; 50 (5)

You might also like