Você está na página 1de 9

Clinical and Experimental Allergy, 1999, Volume 29, Supplement 3, pages 116–124

The metabolism of antihistamines and drug interactions:


the role of cytochrome P450 enzymes
A. G. RENWICK
Clinical Pharmacology Group, University of Southampton, Southampton, UK

Summary
The non-sedating antihistamines show a diversity of fates in the body and the parent drugs
and metabolites may differ in their biological properties. Clinically significant interactions
with inhibitors of cytochrome P450 have been reported primarily for terfenadine, which has
the potential for cardiac toxicity, and is metabolized to fexofenadine, an antihistamine
without cardiac effects. Astemizole shares many of these characteristics and important
safety-related interactions are likely. Loratadine undergoes extensive metabolism so that
pharmacokinetic interactions could occur, but they would be of little clinical importance
because of the lack of cardiac activity of the parent drug and its metabolites. Ebastine also
undergoes pharmacokinetic interactions, the significance of which is dependent on clar-
ification of the extent of any relevant cardiotoxicity of both ebastine and its metabolite.
Interactions would not be clinically important for cetirizine and fexofenadine which do not
show cardiac effects and are eliminated with little metabolism.
Keywords: metabolism, cytochrome P450, pharmacokinetics, cardiotoxicity

in drug metabolism with isoenzymes of CYP2B, CYP2C,


Introduction
CYP2D and CYP3A responsible for most drug oxidation
The second-generation antihistamines (H1 antagonists with reactions.
enhanced potency and reduced sedative effects) are lipid Cytochrome P450 isoenzymes (P450s) are detectable in
soluble drugs which are rapidly and well absorbed from the most tissues but the highest concentrations of most iso-
gastrointestinal tract. In common with most lipid soluble enzymes are found in the liver; the intestinal wall is also rich
xenobiotic chemicals, most antihistamines undergo meta- in one particular isoenzyme CYP3A4. This localization of
bolism, prior to elimination from the body. Lipid soluble enzyme activity means that P450 isoenzymes may be
drugs partition into cell membranes including the endo- involved in both first-pass metabolism, which determines
plasmic reticulum where they are oxidized by the cyto- the entry of drugs into the general circulation (the bioavail-
chrome P450 enzyme system(s). ability; F), and clearance (CL), which is the elimination
Cytochrome P450 is a super-family of haem-containing from the general circulation [1].
proteins present within the endoplasmic reticulum which Following a single oral dose, the maximum drug con-
show low substrate specificity and are responsible for the centration in plasma depends on the absorption rate (deter-
oxidation of a wide variety of lipophillic substrates, includ- mined by the drug lipophillicity and formulation), the
ing steroids. Cytochrome P450 reactions result in the apparent volume of distribution (V) (determined by lipo-
introduction of oxygen (derived from molecular oxygen) phillicity and protein binding), the bioavailability (F) and
into aliphatic and aromatic rings, and side chains (e.g. the terminal elimination rate [determined by distribution (V)
methyl-, ethyl-, etc.), as well as oxidation at noncarbon and clearance (CL)]. The best measure of the overall body
atoms such as nitrogen and sulphur. Cytochrome P450 exposure or internal dose is given by the area under the
families 1, 2 and 3 (CYP1, CYP2 and CYP3) are involved plasma concentration–time curve (AUC). Again, P450 iso-
enzymes are critical in determining the AUC as:
Correspondence: Dr A. G. Renwick, Clinical Pharmacology Group,
University of Southampton, Biomedical Sciences Building, Bassett Dose × F
AUC ¼
Crescent East, Southampton SO16 7PX, UK. CL
116 q 1999 Blackwell Science Ltd
Metabolism of antihistamines and drug interactions 117

Table 1. Metabolites of non-sedating antihistamines and their activities

Half-life Half-life of Activity of metabolites


Drug of drug (h) Enzyme Main metabolites metabolites (h) at therapeutic concentractions

Astemizole 48 CYP3A4 Desmethylastemizole 12 days Antihistamine; Kþ channel blockade


Azelastine 25* P450 Desmethylazelastine 48 Antihistamine
Cetirizine 9 None None (minor only) — None (most drug eliminated unchanged in urine)
Ebastine ND CYP3A4 Carebastine 14 Antihistamine
Fexofenadine 14 None None — None (drug eliminated unchanged in urine)
Loratadine 10 CYP3A4 þ Descarboethoxy loratadine 20 Antihistamine; inhibits cytokine production
CYP2D6
Terfenadine ND CYP3A4 Carboxylic acid analogue 4 Antihistamine

* non-specific assay used (measured parent drug and metabolite); ND ¼ not determined due to low or undetectable plasma concentrations;
compounds given in bold have been reported to block cardiac Kþ channels and/or increase QT interval in animal models: astemizole and
terfenadine have been reported to produce clinically significant increases in QT interval in humans associated with clinical cases of torsade
de pointes.

During chronic drug dosage the concentrations of drug in Any change in the activity of P450 isoenzymes may
the plasma and tissues build up until a steady-state is affect profoundly the values of F and CL and therefore
reached in which the daily input (dosage/dose interval) is may alter the peak concentration, the AUC and Css. The
balanced by the daily output [plasma concentration times consequence of any change in these values will depend on
plasma clearance (CL)]. The average plasma concentration the biological activity of the parent drug compared to the
at steady state (Css) is determined by: metabolite and the direction of the change in P450 activity,
Dose F ðbioavailabilityÞ i.e. an increase (enzyme induction) or a decrease (enzyme
Css ¼ × inhibition) (Table 2).
Dose interval CL ðclearanceÞ
The essential role of P450 isoenzymes in both bioavailabil-
ity and clearance for most drugs means that the activity of
The cytochrome P450 isoenzymes involved in the
these enzymes will determine the relationship between the
metabolism of antihistamines
external dosage regimen and Css, and also the extent of
accumulation. The plasma half-lives of most non-sedating Terfenadine is absorbed completely from the gut but under-
antihistamines and their active metabolites are less than 24 h goes extensive first-pass metabolism. The bioavailability is
and so they would show limited accumulation in healthy less than 0.01 and the concentrations of terfenadine in
subjects [2]; an exception is the desmethyl metabolite of the circulation are not detectable [4] or barely detectable
astemizole [3] which has a half-life of about 10 days. The (1–4 ng/mL after 120 mg oral doses [5]). The P450 iso-
half-lives and potential for accumulation of the parent drugs enzyme primarily involved in terfenadine metabolism is
would be increased by impaired hepatic oxidation due to CYP3A4 [6]; this is the principal constitutive P450 iso-
liver disease or drug interactions. enzyme in both the liver and the intestinal wall [7] and both

Table 2. Effects of inducers and inhibitors

Toxicological consequences

Change in metabolic activity Metabolic consequences Parent compound toxic Metabolite toxic

Induction ↓ [Parent] ↓
↑ [Metabolite] ↑
Inhibition ↑ [Parent] ↑
↓ [Metabolite] ↓

q 1999 Blackwell Science Ltd, Clinical and Experimental Allergy, 29, Supplement 3, 116–124
118 A. G. Renwick

sites probably contribute to the extensive first-pass metabo- Inhibition of P-glycoprotein has been reported for a
lism. The products of terfenadine metabolism are an active number of drugs, for example quinidine and verapamil
carboxylic acid and an inactive carbinol. Terfenadine is [20]. Inhibition of P-glycoprotein by CYP3A inhibitors,
oxidized initially to azacyclonol and terfenadine alcohol, such as erythromycin and ketoconazole which are discussed
with the latter further oxidized to azacyclonol and terfena- below, may have contributed to the pharmacokinetic and
dine carboxylic acid; CYP3A4 is the principal enzyme toxicological consequences detected when coadministered
involved in each of these reactions [6 8]. with antihistamines. Inhibition of P-glycoprotein, which
Loratadine is converted to descarboethoxyloratadine by also serves as an efflux transporter in the kidney [21], may
human liver microsomes, primarily by CYP3A4- and CYP explain interactions between ‘CYP3A inhibitors’ and non-
2D6-mediated oxidation [9]. Orphenadine appears to be sedating antihistamines which are absorbed and eliminated
metabolized by CYP3A isoenzymes based on in vitro without significant metabolism.
studies using rat microsomes [10].
The metabolism and pharmacokinetics of CYP3A sub-
Cardiac side-effects of antihistamines
strates, such as felodipine [11] and nifedipine [12] show
very wide interindividual differences. The variability is prob- The clinical importance of these pharmacokinetic aspects
ably due to differences in the genetic expression of the has been thrown into sharp focus by the adverse cardiac
CYP3A4 isoenzyme [13] and possibly in the expression of effects produced by some of the second generation anti-
minor forms such as CYP3A5 and the fetal form (CYP3A7). histamines. Both terfenadine and astemizole are associated
with cases of prolongation of the QT interval and torsade de
pointes. The overall therapeutic or toxic response to a drug
The role of P-glycoprotein
depends on two primary factors, the internal dose (as
Studies on the uptake of drugs by Caco-2 cells in vitro, have determined by peak concentration, or AUC or Css-see
indicated that P-glycoprotein (the multidrug resistance above) and the inherent sensitivity of the tissues to the
transporter) may be important in limiting intestinal drug drug. Low incidences of adverse reactions may arise from
absorption [14]. The transporter is on the apical side of the an abnormal relationship between external and internal dose
brush border membrane and acts to transport molecules and/or an idiosyncratically exaggerated tissue response to
actively from the cell into the intestinal lumen [15,16]. the drug. The adverse cardiac reactions to terfenadine
P-glycoprotein is an important transporter for the elim- probably arise from a combination of enhanced plasma
ination of drugs into the intestinal lumen [16]. concentrations of the parent compound, combined with an
There is probably a close interrelationship between idiosyncratic responsiveness or a predisposing sensitivity
intestinal P-glycoprotein and CYP3A4, both in relation to such as cardiac disease [22].
substrate specificity [15–17] and factors affecting their Early antihistamines, such as promethazine, are poorly
activities [15,18]. The activities of the hepatic tolerated due to sedative properties but do not show the
CYP3A4 (rather than intestinal CYP3A4) and intestinal P- cardiac effects of the second generation antihistamines.
glycoprotein both contribute significantly to interpatient Promethazine is metabolized by CYP2D6 [23] and the
variability in the pharmacokinetics of oral cyclosporin large interindividual differences in the genetic expression
[19]. Terfenadine has been shown to inhibit P-glycoprotein of this isoform would have ensured the rapid detection of a
in vitro, but no published data have been identified on the problem, such as torsade de pointes, without the need
effect of P-glycoprotein on the pharmacokinetics of non- for subtle interactions with other substrates or inhibitors of
sedating antihistamines, but analogy would suggest that an P450 oxidation.
interaction is likely. The antihistamines are lipid soluble A review of the different case reports of torsades de
drugs and therefore they would eventually be absorbed pointes associated with terfenadine [24] has suggested two
completely from the gastro-intestinal tract. However, for principal contributory factors (Table 3); firstly abnormally
antihistamines which are high affinity substrates for both high circulating levels of free terfenadine due to over-
intestinal CYP3A4 and P-glycoprotein, following uptake by dosage, drug interactions with CYP3A inhibitors or liver
enterocytes the drug would either be metabolized or trans- disease [25] and one or more factors predisposing to drug-
ferred back into the gut lumen by P-glycoprotein. The drug induced torsade de points (e.g. ischaemic heart disease or a
transferred back into the gut would then undergo passive prolonged QT interval). The importance of pharmacokinetic/
absorption into the enterocyte and present itself again metabolic interactions was shown in the first published report
for metabolism or transfer out of the cell. In consequence of terfenadine-induced torsade de pointes [26] which
P-glycoprotein would not block the absorption of the drug, occurred in a patient co-prescribed ketoconazole (plus
but greatly increase the potential for intestinal first-pass cefaclor and medroxyprogesterone), who showed elevated
metabolism. plasma concentrations of terfenadine and lower levels of its
q 1999 Blackwell Science Ltd, Clinical and Experimental Allergy, 29, Supplement 3, 116–124
Metabolism of antihistamines and drug interactions 119

Table 3. Cardiotoxicity of antihistamines: predisposing factors in was without ECG effects even after dosage for 7 days [32],
clinical cases indicating negligible risk of adverse cardiac effects.
Epinastine did not produce the ECG changes detected in
rats with terfenadine [33], but the doses of epinastine given
Pharmacokinetic Pharmacodynamic
to rats resulted in plasma concentrations of 400 ng/ml which
Overdose QT-prolongation is only 10 times therapeutic levels. An increased cardio-
CYP3A4 inhibitors Cardiac disease vascular risk due to a drug interaction plus predisposing
Cirrhosis Hypokalaemia sensitivity cannot be ruled out for this drug, based on these
animal data.
Loratadine and cetirizine do not prolong repolarization
and would not induce torsade de pointes [34].
metabolite. Terfenadine, and to a lesser extent ebastine,
prolonged the QTc interval in anaesthetized and conscious
Interactions with azole compounds
guinea-pigs given intravenous and oral doses [27]. This effect
was enhanced after oral dosage by pretreatment with keto-
Terfenadine
conazole, but ECG changes were not produced by loratadine
either alone or following ketoconazole. A subsequent study The case of torsade de pointes in a patient receiving
[28] indicated that ketoconazole alone significantly pro- coadministered terfenadine and ketoconazole [26] was one
longed the QT interval in guinea-pigs. Comparison of the of the earliest indications of the link between decreased
doses associated with a significant prolongation of QTc first-pass metabolism and adverse effects. This interaction
interval and inhibition of histamine-induced bronchocon- was studied further in six subjects given 60 mg terfenadine
striction in guinea pigs [29] showed only a 1–4-fold differ- twice daily for 7 days to establish steady-state conditions
ence in favour of H1 antagonism for terfenadine, astemizole and to assess plasma concentrations of terfenadine and ECG
and ebastine. In contrast, the corresponding oxidative changes. They were subsequently given ketoconazole
metabolites, terfenadine carboxylic acid, norastemizole and (200 mg every 12 h) while maintaining their terfenadine
carebastine showed antihistamine activity, but were essen- dosage for a period of 7 days (with close monitoring for
tially devoid of cardiac effects. Although studies using the first 3 days) followed by further kinetic and ECG
intravenous dosage of drugs which undergo essentially com- measurements. Terfenadine was detectable in only one of
plete first-pass metabolism are difficult to interpret, inter- the six subjects prior to ketoconazole but in five subjects
actions which inhibit first-pass metabolism will increase the after ketoconazole; a subject with 5 ng/ml of terfenadine in
circulating concentrations of the parent drug and may plasma prior to ketoconazole had a peak concentration of
increase the risk of cardiac complications from the 81 ng/ml after ketoconazole which was accompanied by
parent compound, whereas interactions, such as enzyme marked ECG changes. There was a clear relationship
induction, which increase the rate of metabolism would between plasma terfenadine concentration and the change
decrease the risk. The recent report that desmethylastemi- in corrected QT interval (an effect of ketoconazole per se on
zole (norastemizole) prolongs the action potential of iso- QT interval was not investigated). The plasma concen-
lated rabbit ventricular myocytes [30] suggests that the trations of unchanged terfenadine detected when terfenadine
cardiac toxicity may be due to both the parent compound was co-administered with ketoconazole were similar to
and this metabolite. those found postmortem in a woman who had suffered a
Cetirizine produced antihistamine but not cardiac effects fatal cardiac arrest (55 ng/ml) which may have resulted from
in a guinea-pig model sensitive to the cardiac actions of a terfenadine–ketoconazole interaction [35].
terfenadine and astemizole [29]. Thus cetirizine resembles Itraconazole was also one of the first drugs to be impli-
the carboxy-metabolite of terfenadine (fexofenadine) in its cated in causing symptomatic torsade de pointes associated
antihistamine activity, biological fate and safety profile. with significant increases in plasma concentrations of
Cetirizine and fexofenadine differ from the other non- terfenadine [36]. The metabolic basis for this was demon-
sedative antihistamines because they are eliminated strated in a clinical trial [37] in a small group of healthy
primarily by renal excretion of the parent drug with little volunteers. Co-administration of itraconazole with terfen-
metabolism [2]. In consequence the drug interactions adine gave detectable plasma concentrations of parent drug
critical for terfenadine and astemizole would not be in most subjects and this was associated with a prolonged
relevant, and renal disease rather than liver disease could QT interval. Itraconazole is an effective in vivo inhibitor of
result in elevated plasma concentrations, although increased the metabolism of CYP3A substrates such as felodipine.
AUC values and longer half-lives have been reported in The maximum plasma concentration and AUC of felodipine
patients with chronic liver disease [31]. However, cetirizine were increased 6–8-fold whereas the half-life only doubled
q 1999 Blackwell Science Ltd, Clinical and Experimental Allergy, 29, Supplement 3, 116–124
120 A. G. Renwick

[38] which suggests that the inhibition is primarily at the after terfenadine þ erythromycin (11, 16 and 34 ng/ml).
first-pass metabolism by the intestinal wall. Other reported Coadministration of erythromycin did not produce a sig-
interactions include a loss of consciousness when terfen- nificant change in QTc interval in the six subjects studied
adine is taken with itraconazole [39]. electrocardiographically after terfenadine þ erythromycin.
In contrast, a similar study with fluconazole did not result These data indicate that clinical doses of erythromycin
in measurable plasma concentrations of terfenadine or produce a less potent interaction than clinical doses of
changes in cardiac repolarization [40]. ketoconazole (see above). Co-administration of terfenadine
with either erythromycin or clarithromycin resulted in
significantly altered pharmacokinetics with detectable con-
Astemizole centrations of terfenadine per se in about one half of the
The plasma AUC after a single oral dose of astemizole was subjects receiving the combination [48]. Elevated concen-
increased more than 2-fold when given after a period of trations were associated with altered cardiac repolarization;
14 days’ treatment with itraconazole (200 mg twice daily) three out of six volunteers who accumulated terfenadine
[41]. There was no measurable effect of QTc intervals, but per se showed prolonged QT intervals [48]. Pretreatment
the long half-life of astemizole which increased from 2.1 to with erythromycin for 1 week [46] also increased the AUC
3.6 days, and the potential for accumulation of astemizole of the acid metabolite.
on daily dosage raises doubts about the apparently negative There was no effect of azithromycin on terfenadine
single-dose observation. concentrations [48], and the plasma concentration–time
curve of the carboxy-metabolite of terfenadine was not
altered when terfenadine was co-prescribed with azithro-
Azelastine mycin [49]: the absence of any effect on the QTc interval
indicates the absence of a clinically significant interaction
Azelastine undergoes first-pass metabolism to a desmethyl with azithromycin.
metabolite, which has a longer half-life in vivo [42], has Co-administration of loratadine with erythromycin in 24
antihistamine activity and is probably responsible for much healthy volunteers for 10 days [50] resulted in an increase in
of the activity in vivo. Blood concentrations of ‘parent þ plasma concentrations of the parent drug and its active
metabolite’ were twice as high in the elderly as in the young metabolite (Table 1) but there was no effect on QTc inter-
[43] but formal studies on drug interactions have not been vals, which is consistent with their lack of cardiac activity
identified. in vitro.

Interactions with macrolide antibiotics


Astemizole
Macrolide antibiotics are recognized inhibitors of CYP3A4
Coadministration of astemizole with clinical doses of
[44] and therefore an interaction with terfenadine, astemi-
dirithromycin in healthy volunteers resulted in a small
zole and ebastine would be predicted. Erythromycin is an
(34%) increase in oral AUC of a single oral dose of
inhibitor of a number of P450 isoenzymes, as it potentiates
astemizole (but not of its N-desmethyl-metabolite); there
the effects of carbamazepine (CYP3A) and theophylline
was no effect on QTc intervals and this interaction was
(CYP1A/2A) [45]. Macrolide antibiotics were one of the
considered not to be of clinical significance [51] despite the
drug classes which were associated with the adverse cardiac
long half-life of astemizole.
effects reported with terfenadine (see [46]). A recent review
has concluded that whereas erythromycin and clarithro-
mycin are potent inhibitors of cytochrome P450
Interactions with other CYP3A inhibitors
reactions, azithromycin and dirithromycin are not inhibitors
and would not produce clinically significant interactions An early in vivo study using cimetidine, which is a non-
[47]. specific inhibitor of cytochrome P450 isoenzymes, found no
interaction with terfenadine [52] despite the fact that it is a
good in vivo inhibitor of the metabolism of other substrates
Terfenadine
of CYP3A, such as nifedipine [53]. Similarly, cimetidine
An interaction study [46] investigated the effect of erythro- pretreatment did not increase the plasma concentration of
mycin (500 mg three times daily for 1 week) on the phar- ebastine or its active metabolite [54].
macokinetics of terfenadine (60 mg twice daily). Using an Cimetidine increased the plasma concentrations of hydro-
assay with a limit of detection of 5 ng/ml, only three of the xyzine (a first-generation antihistamine) but not of its
nine subjects had measurable terfenadine concentrations carboxylated metabolite cetirizine [55].
q 1999 Blackwell Science Ltd, Clinical and Experimental Allergy, 29, Supplement 3, 116–124
Metabolism of antihistamines and drug interactions 121

Interactions with grapefruit juice small but statistically significant increase in the QTc inter-
val (from 420 6 14 msec with water to 434 6 15 msec with
The discovery that the metabolism of felodipine was
grapefruit juice), whereas grapefruit juice given 2 h after
inhibited by grapefruit juice but not by orange juice [56]
terfenadine produced no change (408 6 18 vs. 407 6
focused attention on the possible active constituent(s)
22 msec). The difference between simultaneous and
responsible for CYP3A inhibition. Based on the known
delayed grapefruit juice is consistent with the data for
inhibitory activity of bioflavonoids on P450 oxidations
other substrates. The inhibitory effect of grapefruit juice is
in vitro [57] and differences in composition between grape-
primarily during the absorption/first pass metabolism phase;
fruit and oranges it was believed that the active agent was
grapefruit juice does not affect the clearance of systemic
naringenin and its precursor naringin. However, the in vitro
doses of other CYP3A substrates such as cyclosporin [69] or
activity of flavonoids was not reproduced by in vivo inter-
nifedipine [70]. It is probable that much of the first-pass
action studies using oral dosage of either naringin [58] or a
metabolism of CYP3A4 substrates such as terfenadine,
very high dose of quercetin [59]. In addition, the in vitro
cyclosporin and nifedipine occurs at the level of the
inhibitory activity of grapefruit juice exceeds that of an
intestinal wall.
equivalent concentration of naringin and there was not
The magnitude of the change in the pharmacokinetics of
conversion of naringin to naringenin under the in vitro
terfenadine produced by grapefruit juice was defined [5]
conditions used [60]. Recently, attention has focused on
using a sensitive radioimmunoassay method with a limit of
furanocoumarins (such as 60 ,70 -dihydroxybergamottin [61];
detection of 0.1 ng/ml. Grapefruit juice, given 30 min before
and dimers [62]) which are potent inhibitors of CYP3A4
a single oral dose of terfenadine (120 mg) produced a 3.5-
[62]; these compounds also cause a rapid loss of CYP3A4
fold increase in the maximum concentration of terfenadine
activity from a cell line in vitro [61], consistent with the loss
per se and a 2.5-fold increase in the AUC. This was
in enzyme activity reported in vivo. 60 ,70 -Dihydroxyberga-
accompanied by a small and non-significant increase in
mottin did not decrease CYP1A2 or CYP2D6 [61]; grape-
the change in QTc interval detected at the peak plasma
fruit juice produces a weak inhibition of the metabolism of
concentration.
CYP1A2 substrates in vivo [63, 64], although some studies
reported no activity in vivo [65, 66]. 60 ,70 -Dihydroxyberga-
mottin is present in grapefruit juice but not in orange juice Other in vivo interaction studies
[67], an observation consistent with the original clinical
Co-administration of clinically relevant doses of zileuton (a
observations with felodipine.
5-lipoxygenase inhibitor) with terfenadine resulted in a
Many studies on drug–grapefruit juice interactions have
small but measurable and significant increase in the maxi-
used double-strength grapefruit juice; recent studies with
mum plasma concentration and AUC of terfenadine (by
terfenadine have shown significant effects with regular-
35%) in healthy volunteers, but this was not associated with
strength grapefruit juice [68] and freshly squeezed juice [5].
significant ECG changes [71] indicating that this interaction
was not likely to be of clinical importance.
The antidepressant nefazodone increases the AUC of
Terfenadine
CYP3A substrates, such as triazolam, and coadministration
Coadministration of terfenadine (60 mg twice daily) for of nefazodone with terfenadine or astemizole is contra-
7 days and grapefruit juice gave measurable plasma con- indicated [72]. Fluvoxamine, fluoxetine and sertraline
centrations of terfenadine in 6/6 subjects when taken simul- inhibit both CYP3A4 and CYP2C [73] and these may also
taneously, but in only 2/6 subjects when the grapefruit juice show interactions with terfenadine and astemizole (but see
was taken 2 h after the terfenadine, and 0/6 subjects when below).
the same dosage of terfenadine was taken in the absence of
grapefruit juice [4]. The maximum detected plasma con-
In vitro interaction studies
centrations of terfenadine with grapefruit juice were 5–
11 ng/ml, but the sensitivity of the assay method precluded In vitro studies using primary cultures of human hepato-
characterization of the pharmacokinetic profile. Grapefruit cytes, have confirmed the metabolic basis for many of the in
juice also produced a statistically significant increase in the vivo interactions with terfenadine. The C-oxidation was
AUC of the acid metabolite when given simultaneously inhibited by ketoconazole > itraconazole > cyclosporin >
(þ55%) and when given 2 h after terfenadine (þ22%); the erythromycin > naringenin (a grapefruit constituent) [74]
interindividual variability in the data in groups with and and N-dealkylation was also inhibited but with a slightly
without grapefruit juice raises doubts about the clinical different order of potency. These in vitro rankings should be
significance of this difference. Interestingly, simultaneous interpreted with caution because in vivo activity will depend
administration of grapefruit juice and terfenadine caused a on the dosage and kinetics of the inhibitors. For example,
q 1999 Blackwell Science Ltd, Clinical and Experimental Allergy, 29, Supplement 3, 116–124
122 A. G. Renwick

selective serotonin re-uptake inhibitors (SSRIs) were weak References


in vitro inhibitors of terfenadine oxidation by human micro-
1 Thummel KE, Kunze KL, Shen DD. Enzyme-catalyzed pro-
somes [75] but in vitro–in vivo scaling indicated that this cesses of first-pass hepatic and intestinal drug extraction. Adv
would not be of clinical significance at normal clinical doses Drug Deliv Rev 1997; 27:99–127.
[76], in contrast to ketoconazole and itraconazole which 2 Estelle F, Simons R, Simons KJ. Pharmacokinetic optimisation
were 20 times more potent in vitro and have been shown to of histamine H1 receptor antagonist therapy. Clin Pharmacokin
have significant effects in vivo. Interestingly ketoconazole, 1991; 21:372–93.
fluconazole, itraconazole, erythromycin, clarithromycin and 3 Moller C, Andlin-Sobocki P, Blychert LO. Pharmacokinetics
troleandomycin were potent inhibitors of human micro- of astemizole in children. Rhinol 1992; 30 (Suppl.13):21–5.
somal terfenadine metabolism (IC50 values of < 10 nM) but 4 Benton RE, Honig PK, Zamani K, Cantilena LR, Woosley RL.
weak inhibitors of rat microsomes (IC50 values of > 80 nM) Grapefruit juice alters terfenadine pharmacokinetics, resulting
in prolongation of repolarisation on the electrocardiogram.
[77] and of oxidation of terfenadine by rat hepatocytes [78].
Clin Pharmacol Therap 1996; 59:383–8.
In vitro studies have also shown the potential for other
5 Clifford CP, Adams DA, Murray S, Taylor GW, Wilkins MR,
interactions with CYP3A substrates such as ritonavir [79] Boobis AR, Davies DS. The cardiac effects of terfenadine after
and saquinavir [80] but their in vivo significance has not inhibition of its metabolism by grapefruit juice. Eur J Clin
been defined. Pharmacol 1997; 52:311–5.
6 Yun CH, Okerholm RA, Guengerich FP. Oxidation of the
antihistaminic drug terfenadine in human liver microsomes:
Conclusions
Role of cytochrome P-450 3A (4) in N–dealkylation and
The potential for clinically important interactions with non- C–hydroxylation. Drug Metab Dispos 1993; 21:403–9.
sedating antihistamines can be subdivided on the basis of 7 Kolars JC, Lown KS, Schmiedlin-Ren P, et al. CYP3A gene
the metabolic fate and biological activity of the parent drug expression in human gut epithelium. Pharmacogenetics 1994;
and metabolite. 4:247–59.
8 Ling KHJ, Leeson GA, Burmaster SD, Hook RH, Reith MK,
Astemizole and terfenadine are both substrates for
Cheng LK. Metabolism of terfenadine associated with CYP3A
CYP3A4 and have been associated with clinical cases of
(4) activity in human hepatic microsomes. Drug Metab Dispos
torsade de pointes. The relationship between drug inter- 1995; 23:631–6.
actions and cardiovascular risk is clear in the case of 9 Yumibe N, Huie K, Chen KJ, Snow M, Clement RP, Cayen
terfenadine, because the active antihistamine metabolite MN. Identification of human liver cytochrome P450 enzymes
(fexofenadine) does not produce cardiac effects and that metabolize the non-sedating antihistamine loratadine.
coadministration of inhibitors of CYP3A4 was part of the Formation of descarboethoxyloratadine by CYP3A4 and
clinical scenario in the cases of torsade de pointes. The CYP2D6. Biochem Pharmacol 1996; 51:165–72.
situation with astemizole is probably similar but less clearly 10 Roos PH, Mahnke A. Metabolite complex formation of
defined as the antihistamine metabolite has been reported to orphenadrine with cytochrome P450. Involvement of CYP2C11
show cardiac effects; the long half-lives of the parent drug and CYP3A isozymes. Biochem Pharmacol 1996; 52:73–84.
11 Blychert E, Edgar B, Elmfeldt D, Hednor T. A population
and especially the metabolite, mean that short-term inter-
study of the pharmacokinetics of felodipine. Br J Clin
action studies are inadequate to define the potential for
Pharmacol 1991; 31:15–24.
effects on the QT interval. Ebastine is oxidized to the active 12 Ahsan CH, Renwick AG, Waller DG, et al. The influences of
antihistamine metabolite carebastine and it is probably dose and ethnic origins on the pharmacokinetics of nifedipine.
similar to terfenadine in its activities, metabolism and Clin Pharmacol Ther 1993; 54:329–38.
potential for clinically important interactions. 13 Lown KS, Kolars JC, Thummel KE, et al. Interpatient hetero-
The published data do not allow a definitive conclusion geneity in expression of CYP3A4 and CYP3A5 in small bowel.
on the potential for clinically significant interactions with Lack of prediction by erythromycin breath test. Drug Metab
azelastine. Of the non-sedating antihistamines it appears Disp 1994; 22:947–55.
that fexofenadine, cetirizine and loratadine are the least 14 Gan LSL, Thakker DR. Applications of the Caco-2 model in
likely to undergo a clinically significant drug interaction the design and development of orally active drugs: Elucidation
of biochemical and physical barriers by the intestinal epithe-
[81]. Cetirizine and fexofenadine undergo limited meta-
lium. Adv Drug Deliv Res 1997; 23:77–98.
bolism and are not associated with cardiac effects. In
15 Watkins PB. The barrier function of CYP3A4 and P-glyco-
contrast, loratadine undergoes extensive oxidation largely protein in the small bowel. Adv Drug Deliv Res 1997; 27:161–
by CYP3A, and would show pharmacokinetic interactions 70.
with azoles and macrolide antibiotics; however, neither 16 Hunter J, Hirst BH. Intestinal secretion of drugs. The role of
loratadine or the main metabolite affect cardiac Kþ P-glycoprotein and related drug efflux systems in limiting oral
channels at therapeutic concentrations, and therefore these drug absorption. Adv Drug Deliv Res 1997; 25:129–57.
interactions would not represent a safety concern. 17 Herbert MF. Contributions of hepatic and intestinal metabolism

q 1999 Blackwell Science Ltd, Clinical and Experimental Allergy, 29, Supplement 3, 116–124
Metabolism of antihistamines and drug interactions 123

and P-glycoprotein to cyclosporine and tacrolimus oral drug 34 Woosley RL. Cardiac actions of antihistamines. Ann Rev
delivery. Adv Drug Deliv Res 1997; 27:201–14. Pharmacol Toxicol 1996; 36:233–52.
18 Schuetz EG, Beck WT, Schuetz JD. Modulation and substrates 35 Honig PK, Wortham DC, Zamani K, et al. Terfenadine–
of P-glycoprotein and cytochrome P4503A coordinately up- ketoconazole interaction: Pharmacokinetic and electrocardio-
regulate these proteins in human colon carcinoma cells. Mol graphic consequences. J Am Med Assoc 1993; 269:1513–8.
Pharmacol 1996; 49:311–8. 36 Pohjola-Sintonen S, Viitasalo M, Toivonen L, Neuvonen P.
19 Lown KS, Mayo RR, Leichtman AB, et al. Role of intestinal Itraconazole prevents terfenadine metabolism and increases
P-glycoprotein (mdr 1) in interpatient variation in oral risk of torsades de pointes ventricular tachycardia. Eur J Clin
bioavailability of cyclosporine. Clin Pharmacol Ther 1997; Pharmacol 1993; 45:191–3.
62:248–60. 37 Honig PK, Wortham DC, Hull R, et al. Itraconazole affects
20 Rabbaa L, Pautrey S, Colas-Linhart N, Carbon C, Farinotti R. single-dose terfenadine pharmacokinetics and cardiac
Intestinal elimination of ofloxacin enantiomers in the rat: repolarization pharmacodynamics. J Clin Pharmacol 1993;
Evidence of a carrier-mediated process. Antimicrob Agents 33:1201–6.
Chemother 1996; 40:2126–30. 38 Jalava KM, Olkkola KT, Neuvonen PJ. Itraconazole greatly
21 Bendayan R. Renal drug transport: a review. Pharmacother increases plasma concentrations and effects of felodipine. Clin
1996; 16:971–85. Pharmacol Ther 1997; 61:410–5.
22 Koh KK, Rim MS, Yoon J, Kim SS. Torsade de pointes 39 De-Wildt SN, Van-den-Anker JN, Romkes JH, et al. Loss of
induced by terfenadine in a patient with long QT syndrome. consciousness due to simultaneous use of terfenadine and
J Electrocardiol 1994; 27:343–6. itraconazole. Ned Tijdschr Geneeskd 1997; 141:1752–4.
23 Nakamura K, Yokoi T, Inoue K, et al. CYP2D6 is the principal 40 Honig PK, Wortham DC, Zamahni K, et al. The effect of
cytochrome P450 responsible for metabolism of the histamine fluconazole on the steady-state pharmacokinetics and electro-
H1 antagonist promethazine in human liver microsomes. cardiographic pharmacodynamics of terfenadine in humans.
Pharmacogenetics 1996; 6:449–57. Clin Pharmacol Ther 1993; 53:630–6.
24 Chan TYK. Terfenadine-induced torsade de pointes: Risk 41 Lefebvre RA, Van-Peer A, Woestenborghs R. Influence of
factors and mechanisms. J Pharm Technol 1997; 13:127–32. itraconazole on the pharmacokinetics and electrocardiographic
25 Kamisako T, Adachi Y, Nakagawa H, Yamamoto T. Torsades de effects of astemizole. Br J Clin Pharmacol 1997; 43:319–22.
pointes associated with terfenadine in a case of liver cirrhosis and 42 Adusumalli VE, Wong KK, Kucharczyk N, Sofia RD.
hepatocellular carcinoma. Intern Med 1995; 34:92–5. Pharmacokinetics of azelastine and its active metabolite,
26 Monahan BP, Ferguson CL, Killeavy ES, Lloyd BK, Troy J, desmethylazelastine, in guinea pigs. Drug Metab Dispos
Cantilena LR. Torsades de pointes occurring in association 1992; 20:530–5.
with terfenadine use. J Am Med Assoc 1990; 264:2788–90. 43 Peter G, Romeis P, Borbe HO, Buker KM, Riethmuller-
27 Hey JA Del, -Prado M, Kreutner W, Egan RW. Cardiotoxic and Winzen H. Tolerability and pharmacokinetics of single and
drug interaction profile of the second generation antihistamines multiple doses of azelastine hydrochloride in elderly
ebastine and terfenadine in an experimental animal model of volunteers. Arzneim Forsch Drug Res 1995; 45:576–81.
torsade de pointes. Arzneim Forsch Drug Res 1996; 46: 159–63. 44 Gillum JG, Israel DS, Polk RE. Pharmacokinetic drug inter-
28 Gras J, Llenas J, Palacios JM, Roberts DJ. The role of actions with antimicrobial agents. Clin Pharmacokinet 1993;
ketoconazole in the QTc interval prolonging effects of H1- 25:450–82.
antihistamines in a guinea-pig model of arrhythmogenicity. Br 45 Ludden TM. Pharmacokinetic interactions of the macrolide
J Pharmacol 1996; 119:187–8. antibiotics. Clin Pharmacokinet 1985; 10:63–79.
29 Hey JA, Del-Prado M, Sherwood J, Kreutner W, Egan RW. 46 Honig PK, Woosley RL, Zamani K, Conner DP, Cantilena LR.
Comparative analysis of the cardiotoxicity proclivities of Changes in the pharmacokinetics and electrocardiographic
second generation antihistamines in an experimental model pharmacodynamics of terfenadine with concomitant
predictive of adverse clinical ECG effects. Arzneim Forsch administration of erythromycin. Clin Pharmacol Ther 1992;
Drug Res 1996; 46:153–8. 52:231–8.
30 Vorperian VR, Zhou Z, Mohammad S, et al. Torsade de pointes 47 Watkins VS, Polk RE, Stotka JL, Labrada L, Dion P. Drug
with an antihistamine metabolite: potassium channel blockade with interactions of macrolides: Emphasis on dirithromycin. Ann
desmethylastemizole. J Am College Cardiol 1996; 28:1556–61. Pharmacother 1997; 31:349–56.
31 Horsmans Y, Desager JP, Hulhoven R, Harvengt C. Single- 48 Honig PK, Wortham DC, Zamani K, Cantilena LR. Com-
dose pharmacokinetics of cetirizine in patients with chronic parison of the effect of the macrolide antibiotics erythromycin,
liver disease. J Clin Pharmacol 1993; 33:929–32. clarithromycin and azithromycin on terfenadine steady-state
32 Sale ME, Barbey JT, Woosley RL, et al. The electrocardio- pharmacokinetics and electrocardiographic parameters. Drug
graphic effects of cetirizine in normal subjects. Clin Pharmacol Invest 1994; 7:148–56.
Ther 1994; 56:295–301. 49 Harris S, Hilligoss DM, Colangelo PM, Eller M, Okerholm R.
33 Ohtani H, Kotaki H, Sawada Y, Iga T. A comparative pharma- Azithromycin and terfenadine: Lack of drug interaction. Clin
cokinetic–pharmacodynamic study of the electrocardiographic Pharmacol Ther 1995; 58:310–5.
effects of epinastine and terfenadine in rats. J Pharm Pharmacol 50 Brannan MD, Reidenberg P, Radwanski E, et al. Loratadine
1997; 49:458–62. administered concomitantly with erythromycin: pharmacokinetic

q 1999 Blackwell Science Ltd, Clinical and Experimental Allergy, 29, Supplement 3, 116–124
124 A. G. Renwick

and electrocardiographic evaluations. Clin Pharmacol Therap 66 Maish WA, Hampton EM, Whitsett TL, Shepard JD,
1995; 58:269–78. Lovallo WR. Influence of grapefruit juice on caffeine
51 Bachmann K, Sullivan TJ, Reese JH, et al. A study of the pharmacokinetics and pharmacodynamics. Pharmacotherapy
interaction between dirithromycin and astemizole in healthy 1996; 16:1046–52.
adults. Am J Ther 1997; 4:73–9. 67 Edwards DJ, Bellevue II, Woster PM. Identification of 60 ,70 -
52 Honig PK, Wortham DC, Zamani K, et al. Effect of con- dihydroxybergamottin, a cytochrome P450 inhibitor, in grape-
comitant administration of cimetidine and ranitidine on the fruit juice. Drug Metab Dispos 1996; 24: 1287–90.
pharmacokinetics and elctrocardiographic effects of 68 Rau SE, Bend JR, Arnold JMO, et al. Grapefruit juice–
terfenadine. Eur J Clin Pharmacol 1993; 45:41–6. terfenadine single-dose interaction: magnitude, mechanism
53 Renwick AG, Le Vie J, Challenor VF, et al. Factors affecting and relevance. Clin Pharmacol Ther 1997; 61:401–9.
the pharmacokinetics of nifedipine. Eur J Clin Pharmacol 69 Ducharme MP, Warbasse LH, Edwards DJ. Disposition of
1987; 32:351–5. intravenous and oral cyclosporine after administration of
54 Van-Rooij J, Schoemaker HC, Bruno R, et al. Cimetidine does grapefruit juice. Clin Pharmacol Therap 1995; 57:485–91.
not influence the metabolism of the H1-receptor antagonist 70 Rashid TJ, Martin U, Clarke H, et al. Factors affecting the
ebastine to its active metabolite carebastine. Br J Clin absolute bioavailability of nifedipine. Br J Clin Pharmacol
Pharmacol 1993; 35:661–3. 1995; 40:51–8.
55 Simons FER, Sussman GL, Simons KJ. Effect of the H2- 71 Awni WM, Cavanaugh JH, Leese P, et al. The pharmacokinetic
antagonist cimetidine on the pharmacokinetics and pharmaco- and pharmacodynamic interaction between zileuton and
dynamics of the H1-antagonists hydroxyzine and cetirizine in terfenadine. Eur J Clin Pharmacol 1997; 52:49–54.
patients with chronic urticaria. J Allergy Clin Immunol 1995; 72 Robinson DS, Roberts DL, Smith JM, et al. The safety profile
95:685–93. of nefazodone. J Clin Psychiatry 1996; 57 (Suppl.2): 31–8.
56 Bailey DG, Spence JD, Munroz C, Arnold JMO. Interaction of 73 Nemeroft CB, DeVane CL, Pollock BG. Newer antidepressants
citrus juices with felodipine and nifedipine. Lancet 1991; and the cytochrome p450 system. Am J Psychiatry 1996;
337:268–9. 153:311–20.
57 Miniscalco A, Lundahl J, Regardh CG, Edgar B, Eriksson UG. 74 Li AP, Jurima-Romet M. Applications of primary human
Inhibition of dihydropyridine metabolism in rat and human hepatocytes in the evaluation of pharmacokinetic drug —
liver microsomes by flavonoids found in grapefruit juice. drug interactions: Evaluation of model drugs terfenadine and
Pharmac Exp Ther 1992; 261:1195–9. rifampcin. Cell Biol Toxicol 1997; 13:365–74.
58 Bailey DG, Malcolm J, Arnold O, Munoz C, David-Spence J. 75 Von-Moltke LL, Greenblatt DJ, Duan SX, Harmatz JS,
Grapefruit juice–felodipine interaction. Mechanism, predict- Wright CE. Inhibition of terfenadine metabolism in vitro by
ability and effect of naringin. Clin Pharmacol Ther 1993; azole antifungal agents and by selective serotonin reuptake
53:637–42. inhibitor antidepressants: Relation to pharmacokinetic interac-
59 Rashid J, McKinstry C, Renwick AG, Dirnhuber M, Waller tions in vivo. J Clin Psychopharmacol 1996; 16:104–12.
DG, George CF. Quercetin, an in vitro inhibitor of CYP3A, 76 Von-Moltke LL, Greenblatt DJ, Schmider J, Harmatz JS,
does not contribute to the interaction between nifedipine and Shader RI. Metabolism of drugs by cytochrome P450 3A
grapefruit juice. Br J Clin Pharmacol 1993; 36:460–3. isoforms. Implications for drug interactions in psycho-
60 Edwards DJ, Bernier SM. Naringin and naringenin are not the pharmacology. Clin Pharmacokinet 1995; 29 (Suppl. 1): 33–
primary CYP3A inhibitors in grapefruit juice. Life Sci 1996; 44.
59:1025–30. 77 Jurima-Romet M, Crawford K, Cyr T, Inaba T. Terfenadine
61 Schmiedlin-Ren P, Edwards DJ, Fitzsimmons ME, et al. metabolism in human liver: In vitro inhibition by macrolide
Mechanisms of enhanced oral availability of CYP3A4 substrates antibiotics and azole antifungals. Drug Metab Dispos 1994;
by grapefruit constituents: Decreased enterocyte CYP3A4 con- 22:849–57.
centration and mechanism-based inactivation by furanocoumar- 78 Jurima-Romet M, Huang HS, Beck DJ, Li AP. Evaluation of drug
ins. Drug Metab Dispos 1997; 25: 1228–33. interactions in intact hepatocytes: Inhibitors of terfenadine meta-
62 Fukuda K, Ohta T, Oshima Y, et al. Specific CYP3A4 inhibi- bolism. Toxicol Vitro 1996; 10:655–63.
tors in grapefruit juice: Furocoumarin dimers as components of 79 Kumar GN, Rodrigues AD, Buko AM, Denissen JF, Cyto-
drug interaction. Pharmacogenetics 1997; 7:391–6. chrome P450-mediated metabolism of the HIV-1 protease
63 Fuhr W, Klittich K, Staib AH. Inhibitory effect of grapefruit inhibitor ritonavir (ABT-538) in human liver microsomes.
juice and its bitter principal, naringenin, on CYP1A2 depen- J Pharmacol Exp Ther 1996; 277:423–31.
dent metabolism of caffeine in man. Br J Clin Pharmacol 1993; 80 Fitzsimmons ME, Collins JM. Selective biotransformation of
35:431–6. the human immunodeficiency virus protease inhibitor saquina-
64 Kang JH, Roh HK, Lee YS, et al. Effect of grapefruit juice on vir by human small-intestinal cytochrome P4503A4: Potential
CYP1A2 dependent metabolism of caffeine in Koreans. contribution to high first-pass metabolism. Drug Metab Dispos
J Korean Soc Clin Pharmacol Ther 1997; 5:26–34. 1997; 25:256–66.
65 Fuhr U, Majer A, Keller A, et al. Lacking effect of grapefruit 81 Ament PW, Paterson A. Drug interactions with the nonsedating
juice on theophylline pharmacokinetics. Int J Clin Pharmacol antihistamines. Am Fam Phys 1997; 56:223–30.
Ther 1995; 33:311–4.

q 1999 Blackwell Science Ltd, Clinical and Experimental Allergy, 29, Supplement 3, 116–124

Você também pode gostar