You are on page 1of 12

The Pharmacogenomics Journal (2008) 8, 4–15

& 2008 Nature Publishing Group All rights reserved 1470-269X/08 $30.00
www.nature.com/tpj

REVIEW

The clinical role of genetic polymorphisms in


drug-metabolizing enzymes

D Tomalik-Scharte1, A Lazar1, For most drug-metabolizing enzymes (DMEs), the functional consequences
of genetic polymorphisms have been examined. Variants leading to reduced
U Fuhr1 and J Kirchheiner2 or increased enzymatic activity as compared to the wild-type alleles have
1
Department of Pharmacology, Clinical been identified. This review tries to define potential fields in the therapy of
Pharmacology, University of Cologne, Köln, major medical conditions where genotyping (or phenotyping) of genetically
Germany and 2Department of Pharmacology of polymorphic DMEs might be beneficial for drug safety or therapeutic
Natural Products and Clinical Pharmacology, outcome. The possible application of genotyping is discussed for depression,
University of Ulm, Ulm, Germany
cardiovascular diseases and thromboembolic disorders, gastric ulcer,
Correspondence: malignant diseases and tuberculosis. Some drugs used for relief of these
Dr D Tomalik-Scharte, Department of ailments are metabolized with participation of genetically polymorphic DMEs
Pharmacology, Clinical Pharmacology, including CYP2D6, CYP2C9, CYP2C19, thiopurine-S-methyltransferase,
University of Cologne, Gleueler Strae 24, dihydropyrimidine dehydrogenase, uridine diphosphate glucuronosyltrans-
50931 Köln, Germany.
E-mail:dorota.tomalik-scharte@uk-koeln.de ferase and N-acetyltransferase type 2. Current evidence suggests that taking
genetically determined metabolic capacities of DMEs into account has the
potential to improve individual risk/benefit relationship. However, more
prospective studies with clinical endpoints are needed before the paradigm
of ‘personalized medicine’ based on DME variants can be established.
The Pharmacogenomics Journal (2008) 8, 4–15; doi:10.1038/sj.tpj.6500462;
published online 5 June 2007

Keywords: drug-metabolizing enzymes; pharmacogenetics; genotyping; phenotyping

Introduction

Despite its profound progress, modern pharmacotherapy still faces many


challenges such as adverse drug reactions, sometimes serious or even lethal,
and nonresponse to standard therapy. The observed prominent variability in
individual response to pharmacotherapy, in part, depends on well-known factors
easily assessable, like age, sex, weight, liver and renal function, co-medication,
heterogeneity in the disease, nutritional state or smoking. Furthermore,
inherited variants in drug-metabolizing enzymes (DMEs), transporters, receptors
and molecules of signal transduction cascades may have a major impact on drug
response.
Pharmacogenetics/pharmacogenomics tries to define the influence of genetic
factors on drug efficacy and adverse drug reactions. Whereas pharmacogenetics is
focused on pharmacological consequences of a single gene mutation, pharma-
cogenomics tries to simultaneously consider numerous genes and their mutual
interaction. A major progress in pharmacogenetics/pharmacogenomics has been
possible thanks to the genetic revolution with the human genome project and
Received 16 August 2006; revised 18 April
2007; accepted 1 May 2007; published the development of modern technologies in genetic testing. It has been expected
online 5 June 2007 that personalized medicine considering patients’ individual genetic profile
Clinical role of polymorphisms in drug metabolism
D Tomalik-Scharte et al
5

would soon be introduced into clinical practice. Since conditions (Table 1) and, at the end of each chapter, a short
variation in drug concentrations resulting from respective statement is made on the potential benefit of geno-/
genetic polymorphisms in DMEs could be directly imple- phenotyping in this respective field.
mented into dose adjustments, genotyping for DMEs seems
to be the closest to incorporation into clinical practice.1 In Depression
contrast, genotyping for drug transporters and receptors
cannot be recommended as a tool for improvement of drug Pharmacotherapy of depression, one of the major psychia-
therapy in clinical practice at present, and respective gene tric disorders, is characterized by long duration of drug
tests are not ready for clinical use.2 therapy, relatively narrow therapeutic index and poor
The importance of genetic variants of DMEs to explain individual prediction of therapeutic response. According to
interindividual differences in drug concentrations and the evidence, about 30% of all depression patients do not
corresponding pharmacodynamic effects has been recog- respond sufficiently to the first antidepressant drug given.4,5
nized about 50 years ago.3 Early examples include the Failure to respond to antidepressant drug therapy as well as
metabolism of succinylcholine, isoniazid, debrisoquine or intolerable side effects not only determine personal suffer-
sparteine. Today, the functional consequences of genetic ing of individuals and their families, but also impose
polymorphisms have been examined for most DMEs. considerable costs on society. At present, there is no
Variants leading to complete deficiency of enzyme activity possibility to reliably predict the individual’s response
as well as those with reduced or increased activity compared before onset of a certain drug treatment.
to the wild-type alleles have been reported. Thus, for a
fraction of drugs, it would be desirable to consider Tricyclic antidepressants
individual activity of respective DMEs as the basis for Many tricyclic antidepressants undergo similar biotransfor-
optimizing therapy. mation in the liver with CYP2D6 catalyzing hydroxylation
The objective of this article is to critically review the data or demethylation reactions.6 CYP2D6 is characterized by a
on genetic polymorphisms of DMEs with emphasis on high interindividual variability in catalytic activity mainly
clinical relevance and the level of evidence, in order to caused by genetic polymorphisms.7 The respective pheno-
define situations where genotyping or phenotyping might type determined by CYP2D6 genotype is predicted by the
be beneficial for drug safety or therapeutic outcome. The number of functional CYP2D6 alleles, so that the presence
data on genetic polymorphisms in DMEs are presented for of two, one or no functional CYP2D6-gene copies results in
several clinically important drug therapies of major medical phenotype of, respectively, rapid or extensive metabolizer

Table 1 Summary of the reviewed medical conditions, respective medicinal treatments and involved genetically polymorphic
drug-metabolizing enzymes

Disease Treatment Drug-metabolizing enzyme/common variant alleles

Depression Tricyclic antidepressants (e.g. amitryptilline, CYP 2D6/CYP2D6*3, *4, *5 and *6 (no enzymatic
imipramine) activity)
Selective serotonin re-uptake inhibitors (e.g. CYP2C19 (for some tricyclic antidepressants)/
paroxetine) CYP2C19*2 and *3 (no enzymatic activity)
Cardiovascular diseases b-blockers (metoprolol, carvedilol) CYP2D6/CYP2D6*3, *4, *5, and *6 (no enzymatic
activity)
AT1 receptor antagonists (losartan, candesartan) CYP2C9/CYP2C9*2 and *3 (decreased enzymatic
activity)
Thromboembolic Coumarin anticoagulants (warfarin, acenocoumarol, CYP2C9/CYP2C9*2 and *3 (decreased enzymatic
disorders phenprocoumon) activity)
Ulcer disease Proton pump inhibitors (e.g. omeprazole, CYP2C19/CYP2C19*2 and *3 (no enzymatic activity)
lansoprazole, pantoprazole)
Malignant diseases Thiopurines (e.g. 6-mercaptopurine, thioguanine) Thiopurine-S-methyltransferase/*2, *3A and *3C (no
enzymatic activity)
5-Fluorouracil Dihydropyrimidine dehydrogenase/exon 14-skipping
mutation in DPYD (unclear contribution of the variant
allele to polymorphic DPD activity in vivo)
Irinotecan Uridine diphosphate glucuronosyltransferase/
UGT1A1*28, UGT1A1*6, UGT1A1*27 (decreased
enzymatic activity)
Tuberculosis Isoniazid N-acetyltransferase type 2/e.g. NAT2*5, *6 and *7
(decreased enzymatic activity)
Abbreviation: DPD, dihydropyrimidine dehydrogenase.

The Pharmacogenomics Journal


Clinical role of polymorphisms in drug metabolism
D Tomalik-Scharte et al
6

(EM), intermediate metabolizer (IM) and slow or poor Other antidepressants


metabolizer (PM).8,9 Furthermore, inheritance of three or For mirtazapine, it could be shown that CYP2D6 genotype
more functional alleles by gene duplication or gene had a significant influence on the variability in the plasma
amplification determines the ultrafast metabolizer (UM) concentration, however, when comparing UMs to EMs, the
phenotype showing higher-than-average enzymatic activ- magnitude of concentration differences was only moder-
ity.9,10 (The situation is even more complex with the ate.21
existence of functional but low-activity alleles such as CYP2D6 is responsible for the transformation of venlafax-
CYP2D6*10 and influences other than genotype on CYP2D6 ine to the equipotent O-desmethyl-venlafaxine.22–24 Thus,
activity, but as data on therapeutic consequences of these for mirtazapine and venlafaxine, CYP2D6 influence on
factors is very limited, this is not considered here.8–10) pharmacokinetics is only small and genotyping with sub-
PMs of CYP2D6 have largely (50% or more) decreased sequent dose adjustments does not seem to be of large
clearance values which was reported for amitriptyline, benefit.
clomipramine, desipramine, imipramine, nortriptyline, The existing data on effects of CYP2D6 polymorphisms on
doxepin and trimipramine.11 metabolism of the tetracyclic antidepressant maprotiline is
Intuitively, one might expect that individuals to whom contradictory.25,26
tricyclic antidepressants are prescribed could benefit from Finally, CYP2D6 polymorphism seems to have no major
CYP2D6 genotyping if the dose is adjusted for the group of influence on metabolism of nefazodone, moclobemide,
PMs and UMs of CYP2D6. reboxetine and trazodone.27–33
In addition to CYP2D6, another highly polymorphic
DME, CYP2C19, also takes part in biotransformation of Possible clinical implications
some tricyclic antidepressants like amitriptyline, imipra- The question whether the differences in pharmacokinetic
mine and clomipramine.12 However, a possible impact of parameters caused by genetic polymorphisms in DMEs really
CYP2C19 polymorphism on the pharmacokinetics of tricyc- impact the outcome of antidepressant treatment has been
lic antidepressants is by far not so well documented as that studied in some observational studies. In a German study
of CYP2D6. evaluating the effect of CYP2D6 genotype on treatment with
CYP2D6-dependent antidepressants, PMs and UMs were
significantly overrepresented as compared to the control
Selective serotonin re-uptake inhibitors (SSRIs) population, respectively, in the group of patients suffering
Some selective serotonin re-uptake inhibitors such as from adverse drug reactions (fourfold) and nonresponders
fluoxetine, fluvoxamine and paroxetine are potent inhibi- (fivefold).34 At the same time, Grasmader et al.35 did not find
tors of CYP2D6 activity. Therefore, multiple dosing causes any influence of CYP2D6 and CYP2C19 genotype on
auto-inhibition of CYP2D6 and conversion from extensive antidepressant drug response, although the incidence of
to slow metabolizer phenotype and from ultrafast to relevant side effects tended to be higher in PMs of CYP2D6.
extensive metabolism was described.13,14 In the case of A high risk for cardiotoxic events and severe arrhythmia was
fluvoxamine, differences in concentration–time curves reported in four patients treated with venlafaxine admitted
(AUCs) between CYP2D6 genotypes were described after to a cardiologic unit who all were CYP2D6 PMs.36 Further-
single doses,15,16 whereas multiple doses result in similar more, a prospective 1-year clinical study of 100 psychiatric
AUCs in PMs and in EMs indicating a strong inhibitory inpatients suggested a trend toward longer duration of
effect on CYP2D6 in EMs.17 hospital stay and higher treatment costs in UMs and PMs of
For fluoxetine undergoing enantioselective metabolism CYP2D6.37
toward the S-enantiomer, impaired demethylation of S- In conclusion, for treatment with antidepressants, there is
fluoxetine in CYP2D6 PMs was observed.18 However, since extended evidence mainly for CYP2D6 and, to a lower
the metabolite desmethylfluoxetine is also an active anti- extent, for CYP2C19 polymorphisms affecting pharmaco-
depressant, no clinical implications should be expected with kinetics of several antidepressants and possibly affecting
regard to variable metabolism of fluoxetine as the parent therapeutic outcome and adverse drug effects. The useful-
drug.19 For paroxetine, although it is also a CYP2D6 ness of genotyping procedures in depressed patients, how-
inhibitor, twofold higher AUCs of the drug were observed ever, has not been confirmed in prospective clinical trials;
in CYP2D6 PMs than in EMs after multiple doses,20 whereas therefore, currently this approach is limited to a few
and in another study, one UM carrying at least three enthusiastic hospitals, such as the Mayo Clinic, and to
functional CYP2D6 gene copies had undetectable drug some patients with adverse or no therapeutic effects.
concentrations.13 In contrast, no influences of CYP2D6 However, with the recent approval of pharmacogenetic tests
polymorphisms on pharmacokinetic parameters of sertra- for CYP2D6 and CYP2C19 by the Food and Drug Adminis-
line and citalopram have been described. tration (FDA), a more extensive application of DME-based
Thus, in conclusion, within the group of SSRIs, CYP personalized therapy in depression may be expected.
inhibition poses a problem for drug interaction, but the Tentatively, before more results of clinical studies are
CYP2D6 gene polymorphism has less effect, and CYP2D6- available, dose adjustments according to pharmacokinetic
based dose adjustments do not seem to be useful for this differences between genotypes in order to achieve a more
group of antidepressants (with the exception of paroxetine). uniform drug exposure may be used.38

The Pharmacogenomics Journal


Clinical role of polymorphisms in drug metabolism
D Tomalik-Scharte et al
7

Cardiovascular diseases polymorphic cytochrome P450 enzyme CYP2C9. CYP2C9*2


and CYP2C9*3 are the clinically best-investigated alleles
Cardiovascular diseases are the major cause of morbidity and connected with impaired intrinsic enzymatic activity, how-
mortality in developed countries. For a couple of drugs used ever, the latter, which results in changes in the substrate
in the therapy of cardiovascular diseases, like some b- binding region of CYP2C9, seems to be of primary
blockers and sartans, clinical implications resulting from importance.49
genetic polymorphisms in DMEs are discussed. Losartan, a potent antagonist of AT1, is metabolized via
CYP2C9 and CYP3A4 to its active metabolite, E-3174.50
b-Blockers McCrea et al.51 described a case of a patient showing a
For metoprolol, one of the most often prescribed b-blockers, minimal conversion of losartan to its active metabolite who
the role of CYP2D6 genetic polymorphisms in its pharma- was diagnosed as phenotypic PM for CYP2C9 and carrier of
cokinetics is well established. CYP2D6 catalyzes O-demethy- the CYP2C9*3 variant allele. Likewise, in a study of 39
lation and, even more specifically, a-hydroxylation of the healthy volunteers, the CYP2C9*3 allele was shown to be
drug.39 Metabolism by CYP2D6 showed a slight preference associated with decreased formation of E-3174.52 On the
for the (R)-enantiomer over the pharmacologically active other hand, Lee et al.53 observed no differences in the
(S)-enantiomer40 but in both enantiomers, there is a pharmacokinetics of losartan and its active metabolite
substantial difference between CYP2D6 EMs and PMs.41,42 between subjects with the genotype of CYP2C9*1/*2 and
Not only metoprolol plasma concentrations but also the *1/*3 and those expressing *1/*1. The influence of CYP2C9
effects on heart rate correlated significantly with CYP2D6 polymorphism on pharmacokinetics or the dynamics of
metabolic phenotype.39,43 Kirchheiner et al.44 described the other AT1 receptor antagonists like irbesartan or candesartan
effects of CYP2D6 genotype on pharmacokinetics of meto- has also been explored. Both drugs are metabolized by
prolol and heart rate. In UMs carrying the CYP2D6 gene CYP2C9 but in contrast to the pro-drug losartan, they are
duplication, total clearance of metoprolol was about 100% the active forms. In patients with the CYP2C9*1/*2 geno-
higher compared with EMs (367 versus 168 l/h). These type, there was a stronger reduction of the diastolic blood
pharmacokinetic differences were reflected in pharmaco- pressure (14.4%) upon therapy with irbesartan as compared
dynamics so that the reduction of exercise-induced heart to wild-type carriers (7.5%) and for systolic blood pressure
rate by metoprolol in the UM group was only about half of response, a similar trend was observed.54 Although serum
that observed in the EMs. concentrations of irbesartan were not measured in this
Another b-blocker, the racemate carvedilol, which was the study, it was suggested that the different therapeutic
first to be approved as adjunctive therapy in the treatment response in carriers of CYP2C9 variants could be explained
of heart failure, is known to be stereoselectively metabolized with a slower elimination of irbesartan and, thus, greater
by cytochrome P450 enzymes.45 Whereas b1-antagonism is concentrations of the drug. For candesartan, increased
primarily conferred by the S-enantiomer of carvedilol, the plasma concentrations and decreased clearance of the drug
R-isoform is responsible for a1-blockade.46 CYP2D6 poly- were described in a patient who was carrier of CYP2C9*1/*3
morphism has been shown to alter the disposition of genotype.55
carvedilol enantiomers. PMs demonstrated an impaired Further investigations are needed to answer the question
clearance of R-carvedilol, thus being affected by a more whether genotyping of patients with cardiovascular diseases
pronounced a1-blockade which might outweigh the bene- before treatment with AT1 receptor antagonists could be
ficial b1-blocking effects.47 Hence, CYP2D6 genotyping beneficial. Nevertheless, from our point of view, currently
might predict therapeutic outcome in patients with chronic genotyping procedures in that case are not useful.
heart failure treated with carvedilol. However, the inhibition
of CYP2D6 metabolism by administration of fluoxetine in
EMs led to significant changes in plasma pharmacokinetics Thromboembolic disorders
in favor of the R-enantiomer without any effects on blood
pressure and heart rate which casts doubt on the clinical Patients with thromboembolic disorders like pulmonary
significance of the CYP2D6 genotype for the treatment with embolism or deep vein thrombosis, or those with atrial
carvedilol in antihypertensive therapy.48 fibrillation who are at risk of thromboembolic complica-
In summary, in our opinion, CYP2D6 genotyping might tions need acute or sometimes life-long therapy with
be beneficial, if at all, for long-term treatment with anticoagulants. Coumarin anticoagulants, which are vita-
metoprolol in indications such as heart failure or in min K antagonists, belong to the most frequently prescribed
post-myocardial infarction patients where no surrogate drugs in the world and are characterized by a narrow
parameter such as blood pressure is available to predict therapeutic index with both interindividually and intra-
long-term efficacy. individually varying effectiveness. The possible complica-
tions in therapy with oral anticoagulants involve severe
AT1 receptor antagonists (sartans) bleedings or lack of efficacy resulting from, respectively,
AT1 (angiotensin II type 1) receptor antagonists, which are over- and underanticoagulation. Genetically polymorphic
also used in the treatment of hypertension and congestive CYP2C9 is one of the factors which could affect safety and
heart failure, are metabolized with involvement of the efficacy of the treatment with coumarin anticoagulants.

The Pharmacogenomics Journal


Clinical role of polymorphisms in drug metabolism
D Tomalik-Scharte et al
8

Warfarin S-phenprocoumon tended to decrease with the increasing


It is estimated that annually more than 1 million patients in number of CYP2C9*2 and *3 alleles, no influence of both
the United States are treated with warfarin.56 The drug is CYP2C9 variants on R-phenprocoumon pharmacokinetic
administered as a racemic mixture of S- and R-warfarin, with parameters was detected.66
S-warfarin being 3–5 times more potent than R-warfarin. S- Although Visser et al.64,65 could not find significant
warfarin is mainly eliminated through 6- or 7-hydroxylation differences with respect to INR or major bleeding complica-
via CYP2C9 so that the activity of this enzyme influences tions between carriers of CYP2C9 variant genotypes and
mainly the steady-state plasma concentration of S-warfarin wild-types, results of other studies support the role of
and its antithrombotic activity.57 The appropriate therapy CYP2C9 genetic polymorphism in patients treated with
with warfarin and other oral anticoagulants of the coumarin phenprocoumon. Hummers-Pradier et al.67 found an in-
type is based on evaluating international normalized ratio creased risk of hemorrhage in phenprocoumon patients
(INR), which is normalized prothrombin time. carrying the CYP2C9*3 allele, but not the CYP2C9*2 allele.
It could be shown that plasma clearance of S-warfarin, but However, in another study presenting data of 284 phenpro-
not the clearance of R-warfarin, depends significantly on coumon patients, both mutant alleles CYP2C9*3 and
CYP2C9 genotype, although nongenetic factors may also CYP2C9*2 were related to an increased risk of overanticoa-
contribute to the great interindividual variability in this gulation.68
parameter.58 Moreover, for patients with at least one variant As this review focuses on genetic polymorphisms in
allele CYP2C9*2 or CYP2C9*3, a longer induction period to DMEs, here we only want to mention briefly the importance
achieve a stable warfarin dosing and an increased risk of INR of genetic polymorphisms affecting the activity of the
values above the reference range along with a higher risk of vitamin K epoxide reductase complex subunit 1 (VKORC1),
life-threatening bleedings were observed.59 Carriers of the which is the target molecule of coumarin anticoagulants, in
CYP2C9 variant alleles were also considerably overrepre- the antithrombotic therapy. In many clinical trials which
sented in the group of patients requiring lower maintenance have been published recently, it could be shown that
doses of warfarin.60 As a result, it was suggested that CYP2C9 VKORC1 genetic polymorphisms also explain a large part
genotyping may select a population of patients who are of the variability in dose requirements of vitamin K
potentially at the risk of complications associated with antagonists and therefore, in addition to CYP2C9 poly-
warfarin therapy.59,60 morphism and demographic factors, play a dominant role in
the determination of individual dose.69–71 Moreover, Sconce
Acenocoumarol et al.69 proposed a dosing algorithm for individual adjust-
Acenocoumarol, the 40 -nitro analog of warfarin, is also ment of warfarin dose taking into account the CYP2C9 and
administered as a racemic mixture of R- and S-enantiomers VKORC1 genotype, age and height, however, such approach
and like for warfarin, the metabolism of its S-enantiomer is should be proved in prospective clinical studies before wide
predominantly mediated by CYP2C9. In contrast to warfar- clinical implementation is possible.
in, the anticoagulation potencies of the R- and S-aceno- Although genetic testing for CYP2C9 and VKORC1
coumarol are similar, although the mean oral clearance of polymorphisms could not replace the role of INR in dose
S-acenocoumarol is essentially greater and its elimination adjustment of coumarin anticoagulants, this procedure
half-life is far shorter than that of the R-enantiomer.61 certainly may help to identify a group of patients tending
Consequently, R-acenocoumarol makes the major contribu- toward a longer induction period to achieve a stable dosing
tion to the overall anticoagulation effect. and who are potentially at a higher risk of serious bleeding
Results of clinical studies dealing with the role of CYP2C9 complications.
polymorphisms in the anticoagulation effect of acenocou-
marol are partly contradictory. Whereas some studies
showed a considerable effect of CYP2C9 genotype only in Ulcer disease and proton pump inhibitors
patients with at least one CYP2C9*3 allele,62,63 recent data
showed that acenocoumarol patients carrying at least one Proton pump inhibitors (PPIs), such as omeprazole, esome-
CYP2C9*3 or CYP2C9*2 allele were at the risk of having INR prazole, lansoprazole, pantoprazole or rabeprazole, are
of six or higher and suffering from major bleeding commonly prescribed in a combination with antibiotics
complications.64,65 Moreover, a clear genotype–dose rela- for Helicobacter pylori eradication in patients with gastric and
tionship was found in this study so that significant smaller duodenal ulcer disease. PPIs undergo extensive presystemic
doses of acenocoumarol were suggested for carriers of the biotransformation in the liver with the involvement of
CYP2C9 mutant alleles as compared with the wild-type genetically polymorphic CYP2C19. CYP2C19*2 and
patients. CYP2C19*3 are the most common nonfunctional alleles
which are responsible for the majority of PM phenotypes of
Phenprocoumon CYP2C19.72
The influence of CYP2C9 polymorphisms on pharmaco- Anderson et al.73 observed that in PMs of mephenytoin (a
kinetics and dynamics of phenprocoumon, another warfarin model substrate of CYP2C19) the AUC for omeprazole,
analog which is favored in a few European countries, was lansoprazole and pantoprazole at steady state was fivefold
addressed in several clinical studies. Whereas clearance of higher compared with EMs indicating that approximately

The Pharmacogenomics Journal


Clinical role of polymorphisms in drug metabolism
D Tomalik-Scharte et al
9

80% of the dose for all three PPIs is metabolized by the standard doses of lansoprazole are administered within
CYP2C19. For different PPIs, drug exposures defined using this regimen and carriers of CYP2C19 wild-type allele might
AUC values were 3- to 13-fold higher in PMs and about 2- to benefit from a higher PPIs dosage.
4-times higher in heterozygous EMs as compared to In contrast to these results, the authors of the recently
homozygous EMs of CYP2C19.74 At the same time, the issued meta-analysis evaluating the impact of CYP2C19
elevation of intragastric pH as a pharmacodynamic response polymorphism on H. pylori eradication rates in dual and
to PPIs was related directly to the respective AUC and a triple therapies with PPIs concluded that only therapies
much higher pH was observed over 24 h following the based on omeprazole undoubtedly depend on CYP2C19
administration of PPIs in PMs than in EMs.74 genotype while those with lansoprazole and rabeprazole do
In a systematic review on pharmacogenetic studies with not.83 They suggested that genotyping for CYP2C19 could
PPIs, Chong and Ensom75 evaluated the effects of CYP2C19 be of greater importance only for the long-term PPI
genetic polymorphism on the clinical outcomes, that is, H. antisecretory therapies in which CYP2C19 impact would
pylori eradication rates upon therapy with these drugs. The be more profound.
results of the most studies supported the hypothesis that In summary, according to the results of numerous clinical
eradication rates vary with CYP2C19 genotype and in PMs, a studies, CYP2C19 polymorphisms is an important factor
significantly better efficacy of PPIs was observed. The affecting the pharmacokinetics of most PPIs, values of
authors pointed out that only in a few studies, notably intragastric pH and eradication rates of H. pylori. Therefore,
performed with rabeprazole, a similar cure rate irrespective in our opinion, genotyping for CYP2C19 polymorphisms
of CYP2C19 genotype was demonstrated. However, rabepra- could be recommended, first of all in Asian populations
zole undergoes mainly nonenzymatic metabolism and characterized by a high prevalence of defect CYP2C19
CYP3A4 in addition to CYP2C19 contribute to the enzyma- alleles. The intuitive consequence of this approach would
tically mediated fraction of its biotransformation.76 Never- be a higher dosage in the group of EMs and/or an adjusted
theless, in a clinical study determining a cure rate of H. pylori treatment regimen. The results of prospective controlled
infections upon dual rabeprazole/amoxicillin therapy in clinical trials need to be awaited to see whether patients
relation to CYP2C19 genotype status in Japanese patients, might benefit from such individually tailored therapy.
significant differences between homozygous and hetero-
zygous EMs and PMs were noticed (cure rates were 60.6, 91.7 Malignant diseases
and 93.8%, respectively).77
In another study, Furuta et al.78 investigated the impact of Most anticancer drugs are characterized by a very narrow
CYP2C19 genotype on eradication rates of H. pylori upon therapeutic index and severe consequences of over- or
triple therapy comprising PPI, clarithromycin and amox- underdosing in the form of, respectively, life-threatening
icillin. These rates were 72.7, 92.1 and 97.8% in the adverse drug reactions or increased risk of treatment
homozygous extensive, heterozygous extensive and PMs, failure. Polymorphisms in genes encoding DMEs are con-
respectively. The authors confirmed that CYP2C19 genotype sidered to be an important factor contributing to individual
seems to be one of the important factors associated with drug response in patients undergoing antineoplastic
cure rates. Moreover, the same authors could also demon- chemotherapy.
strate the significance of CYP2C19 genotype for eradication
upon dual therapy (amoxicillin as the only antibiotic and Thiopurines
high doses of omeprazole) showing cure rates of 100% in Thiopurines, such as 6-mercaptopurine and thioguanine are
PMs.79 For that reason, the authors concluded that in largely used in the treatment of acute leukemia whereas
Japanese patients homozygous for CYP2C19 defect alleles, rheumatoid arthritis is a common field of application for the
the dual therapy might be sufficient. Furuta et al.80 also immunosuppressant azathioprine. These drugs, following
suggested that CYP2C19 genotyping test could be a useful their activation to thioguanine nucleotides via the purine
tool for deciding on the optimal treatment regimen with salvage pathway, are incorporated into DNA.84 At the same
PPIs, including a dual (PPI plus antibiotic) or a triple (PPI time, thiopurine-S-methyltransferase (TPMT) inactivates
plus two antibiotics) therapy. thiopurine drugs. The individual enzymatic capacity is
The role of CYP2C19 polymorphism for eradication of inherited as an autosomal codominant trait resulting in
H. pylori was also studied in the Caucasian population. three distinct phenotypes which show a pronounced ethnic-
However, in contrast to the Asian population, the preva- specific distribution.85 Approximately 89% of Caucasians
lence of PM of CYP2C19 in Caucasians is much lower, that demonstrate a high catalytic TPMT activity while about 11
is, about 2.8 versus 21.3%, respectively, in Caucasians and and 0.3%, respectively, of that population exhibit an
Japanese.81 Schwab et al.82 could show significant differ- intermediate and low activity.86 The molecular basis of such
ences considering the efficacy of lansoprazole-based interindividual variation is provided by genetic polymorph-
quadruple therapy between Caucasian patients carrying isms and the alleles *2, *3A and *3C explain about 80–95%
wild-type alleles, one and two CYP2C19 defect alleles with of altered enzyme activity.87 Impaired TPMT activity is
cure rates of 80.2, 97.8 and 100% in the respective groups. inversely correlated with the generation of thioguanine
The authors concluded that eradication rates of H. pylori nucleotides which are responsible for toxicity.88,89 Myelo-
highly depend on CYP2C19 genotype in white patients if toxicity is the dose-limiting toxicity of thiopurines, and this

The Pharmacogenomics Journal


Clinical role of polymorphisms in drug metabolism
D Tomalik-Scharte et al
10

adverse event may necessitate discontinuation of therapy measuring individual DPD activity in peripheral mono-
and can even lead in fatal outcomes in patients with low nuclear blood cells does not provide a valid phenotyping
TPMT activity if treated with conventional doses.90,91 TPMT approach since the correlation with hepatic DPD activity
genotyping predicts the clinical phenotype with almost and 5-FU-pharmacokinetics is poor.106,107
complete concordance (at least 95%) and essentially all Polymorphisms within the human DPD gene (DPYD) have
homozygous carriers of defective alleles are affected by toxic been associated with DPD deficiency and severe 5-FU-related
side effects that require discontinuation of therapy.90,92 toxicity in cancer patients. One of the best-described
However, in analysis of 41 patients with Crohn’s disease and polymorphisms is the so-called exon 14-skipping mutation
azathioprine-related hematotoxicity, early onset of intoler- at the 50 -splice donor site of exon 14 which is present at a
ance in carriers of mutant TPMT alleles was indeed observed prevalence of only 1% in the Caucasian population but has
but genetic variants could explain only 27% of all toxic been detected in 24% of patients suffering from WHO grade
cases.93 Hepatic TPMT activity can reliably be determined by IV toxicity upon treatment with 5-FU.108 To identify
measurement of the catalytic activity of cytosolic TPMT in patients at increased risk for severe 5-FU-induced toxicity,
erythrocytes using established radiochemical or high-per- routine screening for the exon 14-skipping mutation before
formance liquid chromatography (HPLC) methods.94,86 onset of chemotherapy has been recommended.109 How-
However, several cases of shortcomings of such TPMT ever, the predictive relevance of genotyping for this
phenotyping approaches have been reported. TPMT-defi- mutation is still controversially discussed since the geno-
cient patients spuriously exhibited a normal enzyme activity type–phenotype correlation is apparently incomplete.110
after they had received erythrocyte concentrates of homo- In a study in patients who had suffered from severe
zygous wild-type donors.90,95 Moreover, the phenotyping toxicity owing to treatment with 5-FU, direct sequencing of
results can also be significantly influenced by concomitant all exons revealed possible genetic causes in 8 out of 14
therapy with furosemide, sulfasalazine, salicylic acid, intake subjects. Six of them were heterozygous carriers of the exon
of certain foodstuff and the presence of uremia.96,97 14-skipping mutation.111 Screening for known mutations in
For patients treated with thiopurines, an individualized a cohort of cancer patients undergoing 5-FU chemotherapy
therapy with regard to TPMT genotype has been suggested. explained a low DPD activity phenotype only in 17% of all
Reduction of doses to 5–10% of conventional average doses cases and the sole carrier of the exon 14-skipping mutation
in carriers of two nonfunctional TPMT alleles has been was inconspicuous with regard to enzyme activity.105 More-
shown to allow for an efficacious continuation of ther- over, the sample sizes of the previous studies are virtually
apy.87,98,99 For patients heterozygous for defective TPMT too small to dare a valid estimation of the clinical value of
alleles, a full starting dose has been recommended taking DPYD genotyping in order to prevent 5-FU-related toxicity.
into account that a dose reduction to avoid toxicity will be Currently, the predictive value of genotyping of DPYD for
probably required.99 individualized anticancer therapy needs further elucidation
In conclusion, present genotyping or phenotyping strate- in large prospective studies.
gies predict individuals being deficient with respect to TPMT
at high accuracy and should routinely precede the onset of Irinotecan
therapy with thiopurine-derived drugs in order to minimize Recently, much attention has been paid to importance of
life-threatening and cost-intensive myelotoxic adverse pharmacogenetic polymorphisms in uridine diphosphate
events. For 6-mercaptopurine, which is widely used for glucuronosyltransferase (UGT) and its role in toxicity and
therapy of acute lymphoblastic leukemia in children, the therapeutic response in cancer patients undergoing treat-
FDA has implemented respective pharmacogenetic data into ment with the potent antitumor agent irinotecan. SN-38,
the product label, considering the impact of TPMT genotype the active metabolite of irinotecan and topoisomerase I
on severe toxicity as well as the availability of genotypic and inhibitor, is glucuronidated to its inactive form by UGT1A1,
phenotypic testing. the UGT enzyme which is also responsible for glucuronida-
tion of bilirubin.112 Several variant alleles leading to reduced
5-Fluorouracil enzymatic UGT1A1 activity have been identified in the
The initial and rate-limiting enzyme in the hepatic meta- UGT1A1 gene. These polymorphisms become manifest as
bolism of the chemotherapeutic agent 5-fluorouracil (5-FU) unconjugated hyperbilirubinemia in the form of Crigler–
is dihydropyrimidine dehydrogenase (DPD), thus affecting Najjar or Gilbert’s syndromes as well as, in patients treated
its pharmacokinetics, efficacy and toxicity.100–102 Applica- with irinotecan, they result in reduced SN-38 glucuronida-
tion of 5-FU is restricted by a narrow therapeutic index and tion rates.113 For UGT1A1 genetic variations, major inter-
could be complicated by severe toxicity of WHO grade III–IV ethnic differences have been shown; whereas UGT1A1*28
with symptoms like mucositis and granulocytopenia.103 promoter polymorphism (TA repeat in the promoter) is
Moreover, severe adverse events have also been observed quite common in the Caucasian population, UGT1A1*6 and
with capecitabine, which is an orally bioavailable prodrug UGT1A1*27 situated in the coding region of UGT1A1 are
of 5-FU.104 found mainly in Asians.114
Three-to-five percent of Caucasians have been reported to Impact of UGT1A1 polymorphisms on irinotecan toxicity,
show a reduced enzymatic activity potentially leading to which is characterized by severe diarrhea and neutropenia,
severe 5-FU-related toxicity in cancer patients.105 However, has been studied in several retrospective as well as

The Pharmacogenomics Journal


Clinical role of polymorphisms in drug metabolism
D Tomalik-Scharte et al
11

prospective studies. It could be shown that the presence of metabolism of isoniazid varies interindividually, with mean
common UGT1A1 mutant alleles, even in heterozygous apparent elimination half-lives of 80 min for rapid acetyla-
carriers, significantly changed the disposition of irinotecan tors and 180 min for slow acetylators. Subsequently, these
causing severe toxicity in patients.114–116 Consecutively, earlier findings were complemented by genotyping studies
genotyping for UGT1A1 polymorphisms before therapy with showing a linear relationship of isoniazid pharmaco-
irinotecan was recommended for patients of all ethnic kinetic parameters to the number of highly active NAT2
groups. If UGT1A1 genotyping is not possible, total bilirubin genes.123–125 According to Kinzig-Schippers et al.,125 indivi-
level was proposed as an easy surrogate parameter for dual isoniazid clearance could be predicted as clearance
prediction of life-threatening toxicity due to irinotecan,115 (l/h) ¼ 10 þ 9  number of NAT2*4 alleles.
however, usefulness of this approach should definitely be At the same time, it was observed that occurrence of
confirmed in further clinical trials. Finally, given the adverse effects upon isoniazid therapy is dependent on
cumulative evidence, the FDA has approved a new labeling NAT2 activity, too. In SAs, hepatic toxicity, neurotoxic
for irinotecan in favor of UGT1A1*28 genotyping, which effects and systemic lupus erythematodes seem to appear
reflects the prevalence of the UGT1A1*28 allele in European more frequent and have more severe course than in
Americans. A reduced initial dose of irinotecan has been phenotypic RAs.126 These observations have also been
recommended for homozygous carries of this variant allele supported by later genotyping studies. In tuberculosis
as they are at increased risk for neutropenia (FDA, http:// patients treated with isoniazid and rifampicin, a significant
www.fda.gov). association between hepatotoxicity and NAT2 genotype was
In summary, importance of individually tailored medicine observed so that compared with patients with two highly
for cancer patients seems to be of critical importance to active NAT2 alleles, the relative risk was 4 for patients with
develop safe and effective chemotherapy. Even if genetically one highly active NAT2 allele and 28 for patients with no
polymorphic DMEs are not the only factor contributing to the highly active NAT2 allele. The authors suggested that NAT2
wide interindividual variation in toxicity and efficacy of genotyping before therapy could be useful.127 Likewise,
anticancer drugs, the recent implementation of the pharmaco- Huang et al.128 could show in a study in 224 tuberculosis
genetic data to the product labels of 6-mercaptopurine patients that carriers of no highly active NAT2 alleles had a
and irinotecan can be assessed as milestones in oncology higher risk for hepatotoxicity than those with at least one
and pharmacogenetics.117 However, successful implementa- highly active NAT2 allele, that is, 26 versus 11%. Further-
tion of pharmacogenetic tests in cancer patients would more, hepatic injury in SAs is more likely to have severe
depend on thorough analysis of clinical benefits and cost course than in RAs. On the other hand, efficacy of isoniazid
effectiveness of this approach. therapy seems to be better in SAs and significant differences
in the mean early bactericidal activity of isoniazid between
homozygous SAs and heterozygous or homozygous RAs
Tuberculosis and isoniazid could be demonstrated.129
Individually tailored therapy with isoniazid, which could
Tuberculosis is still a global emergency, first of all due to the warrant optimal therapeutic efficiency and limitation of
growing prevalence of drug resistant Mycobacterium tubercu- toxic effects, has not been used in clinical routine so far,
losis strains and the spread of AIDS infection.118 Isoniazid is although reliable genotyping or phenotyping approaches
a pivotal agent in the treatment of tuberculosis in combina- are available.
tion with other drugs, or alone as a prophylactic agent. It Currently, potential benefits of isoniazid dose adjustment
is metabolized to acetylisoniazid by the hepatic enzyme according to NAT2 genotype in tuberculosis patients are
N-acetyltransferase type 2 (NAT2). The enzyme is target of being assessed in two large European (‘IDANAT2’) and East
genetic polymorphisms and carriers of at least one wild-type Asian studies with participation of the authors of this review
allele (NAT2*4) or a high-activity variant allele (NAT2*12) in the former. The studies test the hypotheses that by means
demonstrate high NAT2 activity (rapid acetylator, RA), of adjustment of the isoniazid dose, early treatment failure
whereas slow acetylators (SA) have two low-activity variants. can be reduced twofold in RAs whereas in SAs hepatotoxicity
Subjects with one high-activity NAT2 allele are sometimes can be reduced threefold. IDANAT2 has a prospective,
classified as intermediate acetylators (IA). Pronounced randomized, controlled and double-blind design (Figure 1)
interethnic differences in the prevalence of the rapid- and and should provide a definite answer on dose individualiza-
slow-acetylation phenotype have been observed with the tion according to genotype of a DME in this disease of global
highest frequencies of RAs in East Asia (about 58–90%) importance.
followed by other parts of Asia and Europe (between 32 and
43%) and the lowest frequencies found in Africa.119,120
Isoniazid was the first NAT2 substrate for which acetyla- Perspectives
tion polymorphism with its well-known bimodal distribu-
tion was described following the observation that peripheral Thousands of manuscripts addressing pharmacogenetic
neuropathy, a potential side effect of the drug, is subject to questions in in vitro studies and clinical trials with healthy
interindividual variation depending on patients’ acetylation volunteers or patients have been published in the past
capacity.121 Tiittinen et al.122 could demonstrate that the decades. Even so, it seems that the way to a broader use of

The Pharmacogenomics Journal


Clinical role of polymorphisms in drug metabolism
D Tomalik-Scharte et al
12

Figure 1 Scheme of a prospective, double-blind, multicenter, parallel group randomized trial to evaluate the possible benefit of isoniazid dose
adjustment according to the genotype of N-acetyltransferase type 2 (IDANAT2).

genotyping/phenotyping approaches for DMEs at the pa- respective genetic factors are probably far more complex
tients’ bedside and, consequently, implementation of the than was initially assumed, already today there is a solid
respective results in the form of personalized pharmaco- place for genotyping for DMEs in some therapies. However,
therapy is quite laborious. There are some possible reasons more prospective studies with clinical endpoints are needed
for this slow progress in clinical pharmacogenetics/pharma- before the paradigm of ‘personalized medicine’ based on
cogenomics. Thanks to modern pharmacogenomic technol- DME variants can be established.
ogies, it was found that genetic regulation of DMEs, like
other proteins controlling biological processes in living
organisms, is actually more complex than initially expected. Duality of Interest
In reality, gene function is not a constant but it varies
depending on environmental factors as well as gene No conflict of interest exists with any of the authors.
expression could be affected by respective gene products
themselves.130 For that reason, there is no exact relationship
References
between the genotype and phenotype, that is, the actually
measured activity of DMEs, although in many cases the 1 Kirchheiner J, Sasse J, Roots I, Brockmoller J, Bauer M. The value of
genotype explains most of the interindividual variability. pharmacogenetic tests in antidepressive medication therapy. Nerve-
Another essential problem is the lack of prospective clinical narzt 2005; 76: 1340–1354.
2 de Leon J, Armstrong SC, Cozza KL. Clinical guidelines for psychiatrists
studies assessing the merits of genotyping/phenotyping
for the use of pharmacogenetic testing for CYP450 2D6 and CYP450
strategies for personalized medicine in large numbers of 2C19. Psychosomatics 2006; 47: 75–85.
patients. Similarly, no reliable data on cost effectiveness of 3 Kalow W. Familial incidence of low pseudocholinesterase level. Lancet
such screening procedures exist. A successful implementa- 1956; 2: 576–577.
4 Bauer M, Whybrow PC, Angst J, Versiani M, Moller HJ. World
tion of individually tailored medicine would depend not federation of societies of biological psychiatry (WFSBP) guidelines for
only on further improvement in multigenic testing but also biological treatment of unipolar depressive disorders part 1: acute and
needs to be preceded by numerous prospective studies with continuation treatment of major depressive disorder. World J Biol
clinical endpoints. Psychiatry 2002; 3: 5–43.
5 Nelson JC. Managing treatment-resistant major depression. J Clin
Psychiatry 2003; 64(Suppl 1): 5–12.
6 Baumann P, Jonzier Perey M, Koeb L, Küpfer A, Tinguely D, Schopf J.
In summary Amitriptyline pharmacokinetics and clinical response: II. Metabolic
polymorphism assessed by hydroxylation of debrisoquine and mephe-
nytoin. Int Clin Psychopharmacol 1986; 1: 102–112.
We reviewed the main genetic polymorphisms of DMEs and 7 Bertilsson L, Dahl ML, Dalen P, Al-Shurbaji A. Molecular genetics of
discussed their potential clinical relevance on pharmaco- CYP2D6: clinical relevance with focus on psychotropic drugs. Br J Clin
logical therapies of common human ailments, with the Pharmacol 2002; 53: 111–122.
intention to demonstrate the importance of pharmaco- 8 Sachse C, Brockmoller J, Bauer S, Roots I. Cytochrome P450 2D6
variants in a Caucasian population: allele frequencies and phenotypic
genetic factors for drug toxicity and treatment efficacy consequences. Am J Hum Genet 1997; 60: 284–295.
in patients. Although interindividual variability in drug 9 Zanger UM, Fischer J, Raimundo S, Stuven T, Evert BO, Schwab M et al.
response is also caused by many nongenetic factors and the Comprehensive analysis of the genetic factors determining expression

The Pharmacogenomics Journal


Clinical role of polymorphisms in drug metabolism
D Tomalik-Scharte et al
13

and function of hepatic CYP2D6. Pharmacogenetics 2001; 11: inhibitor. Eur Neuropsychopharmacol 1997; 7(Suppl 1): S23–S35;
573–585. discussion S71–S73.
10 Griese EU, Zanger UM, Brudermanns U, Gaedigk A, Mikus G, Morike K 30 Schoerlin MP, Blouin RA, Pfefen JP, Guentert TW. Comparison of the
et al. Assessment of the predictive power of genotypes for the in-vivo pharmacokinetics of moclobemide in poor and efficient metabolizers
catalytic function of CYP2D6 in a German population. Pharmaco- of debrisoquine. Acta Psychiatr Scand Suppl 1990; 360: 98–100.
genetics 1998; 8: 15–26. 31 Härtter S, Dingemanse J, Baier D, Ziegler G, Hiemke C. The role of
11 Kirchheiner J, Nickchen K, Bauer M, Wong ML, Licinio J, Roots I et al. cytochrome P450 2D6 in the metabolism of moclobemide. Eur
Pharmacogenetics of antidepressants and antipsychotics: the con- Neuropsychopharmacol 1996; 6: 225–230.
tribution of allelic variations to the phenotype of drug response. Mol 32 Gram LF, Guentert TW, Grange S, Vistisen K, Brøsen K. Moclobemide,
Psychiatry 2004; 9: 442–473. a substrate of CYP2C19 and an inhibitor of CYP2C19, CYP2D6, and
12 Brosen K. Some aspects of genetic polymorphism in the biotransfor- CYP1A2: a panel study. Clin Pharmacol Ther 1995; 57: 670–677.
mation of antidepressants. Therapie 2004; 59: 5–12. 33 Mihara K, Otani K, Suzuki A, Yasui N, Nakano H, Meng X et al.
13 Lam YW, Gaedigk A, Ereshefsky L, Alfaro CL, Simpson J. CYP2D6 Relationship between the CYP2D6 genotype and the steady-state
inhibition by selective serotonin reuptake inhibitors: analysis of plasma concentrations of trazodone and its active metabolite m-
achievable steady-state plasma concentrations and the effect chlorophenylpiperazine. Psychopharmacology (Berl) 1997; 133: 95–98.
of ultrarapid metabolism at CYP2D6. Pharmacotherapy 2002; 22: 34 Rau T, Wohlleben G, Wuttke H, Thuerauf N, Lunkenheimer J, Lanczik
1001–1006. M et al. CYP2D6 genotype: impact on adverse effects and nonresponse
14 Laine K, Tybring G, Hartter S, Andersson K, Svensson JO, Widen J et al. during treatment with antidepressants-a pilot study. Clin Pharmacol
Inhibition of cytochrome P4502D6 activity with paroxetine normalizes Ther 2004; 75: 386–393.
the ultrarapid metabolizer phenotype as measured by nortriptyline 35 Grasmader K, Verwohlt PL, Rietschel M, Dragicevic A, Muller M,
pharmacokinetics and the debrisoquin test. Clin Pharmacol Ther 2001; Hiemke C et al. Impact of polymorphisms of cytochrome-P450
70: 327–335. isoenzymes 2C9, 2C19 and 2D6 on plasma concentrations and clinical
15 Spigset O, Granberg K, Hagg S, Norstrom A, Dahlqvist R. Relationship effects of antidepressants in a naturalistic clinical setting. Eur J Clin
between fluvoxamine pharmacokinetics and CYP2D6/CYP2C19 phe- Pharmacol 2004; 60: 329–336.
notype polymorphisms. Eur J Clin Pharmacol 1997; 52: 129–133. 36 Lessard E, Yessine M, Hamelin B, O’Hara G, LeBlanc J, Turgeon J.
16 Carrillo JA, Dahl ML, Svensson JO, Alm C, Rodriguez I, Bertilsson L. Influence of CYP2D6 activity on the disposition and cardiovascular
Disposition of fluvoxamine in humans is determined by the poly- toxicity of the antidepressant agent venlafaxine in humans. Pharma-
morphic CYP2D6 and also by the CYP1A2 activity. Clin Pharmacol Ther cogenetics 1999; 9: 435–443.
1996; 60: 183–190. 37 Chen S, Chou WH, Blouin RA, Mao Z, Humphries LL, Meek QC et al.
17 Spigset O, Granberg K, Hagg S, Soderstrom E, Dahlqvist R. Non-linear The cytochrome P450 2D6 (CYP2D6) enzyme polymorphism: screen-
fluvoxamine disposition. Br J Clin Pharmacol 1998; 45: 257–263. ing costs and influence on clinical outcomes in psychiatry. Clin
18 Eap CB, Bondolfi G, Zullino D, Savary-Cosendai L, Powell-Golay K, Pharmacol Ther 1996; 60: 522–534.
Kosel M et al. Concentrations of the enantiomers of fluoxetine and 38 Kirchheiner J, Brosen K, Dahl ML, Gram LF, Kasper S, Roots I et al.
norfluoxetine after multiple doses of fluoxetine in cytochrome CYP2D6 and CYP2C19 genotype-based dose recommendations for
P4502D6 poor and extensive metabolizers. J Clin Psychopharmacol antidepressants: a first step towards subpopulation-specific dosages.
2001; 21: 330–334. Acta Psychiatr Scand 2001; 104: 173–192.
19 Fuller RW, Snoddy HD, Krushinski JH, Robertson DW. Comparison of 39 Lennard MS, Silas JH, Freestone S, Trevethick J. Defective metabolism
norfluoxetine enantiomers as serotonin uptake inhibitors in vivo. of metoprolol in poor hydroxylators of debrisoquine. Br J Clin
Neuropharmacology 1992; 31: 997–1000. Pharmacol 1982; 14: 301–303.
20 Sindrup SH, Brøsen K, Gram LF. Pharmacokinetics of the selective 40 Lennard MS, Tucker GT, Silas JH, Freestone S, Ramsay LE, Woods HF.
serotonin reuptake inhibitor paroxetine: nonlinearity and relation to Differential stereoselective metabolism of metoprolol in extensive
the sparteine oxidation polymorphism. Clin Pharmacol Ther 1992; 1: and poor debrisoquin metabolizers. Clin Pharmacol Ther 1983; 34:
288–295. 732–737.
21 Kirchheiner J, Henckel HB, Meineke I, Roots I, Brockmöller J. Impact of 41 Koytchev R, Alken RG, Vlahov V, Kirkov V, Kaneva R, Thyroff Friesinger
the CYP2D6 ultra-rapid metabolizer genotype on mirtazapine phar- U et al. Influence of the cytochrome P4502D6*4 allele on the
macokinetics and adverse events in healthy volunteers. J Clin pharmacokinetics of controlled-release metoprolol. Eur J Clin Pharma-
Psychopharmacol 2004; 24: 647–652. col 1998; 54: 469–474.
22 Otton SV, Ball SE, Cheung SW, Inaba T, Rudolph RL, Sellers EM. 42 McGourty JC, Silas JH, Lennard MS, Tucker GT, Woods HF. Metoprolol
Venlafaxine oxidation in vitro is catalysed by CYP2D6. Br J Clin metabolism and debrisoquine oxidation polymorphism–population
Pharmacol 1996; 41: 149–156. and family studies. Br J Clin Pharmacol 1985; 20: 555–566.
23 Fukuda T, Yamamoto I, Nishida Y, Zhou Q, Ohno M, Takada K 43 Lennard MS, Silas JH, Freestone S, Ramsay LE, Tucker GT, Woods HF.
et al. Effect of the CYP2D6*10 genotype on venlafaxine pharmaco- Oxidation phenotype – a major determinant of metoprolol metabo-
kinetics in healthy adult volunteers. Br J Clin Pharmacol 1999; 47: lism and response. N Engl J Med 1982; 307: 1558–1560.
450–453. 44 Kirchheiner J, Heesch C, Bauer S, Meisel C, Seringer A, Goldammer M
24 Veefkind AH, Haffmans PM, Hoencamp E. Venlafaxine serum levels and et al. Impact of the ultrarapid metabolizer genotype of cytochrome
CYP2D6 genotype. Ther Drug Monit 2000; 22: 202–208. P450 2D6 on metoprolol pharmacokinetics and pharmacodynamics.
25 Firkusny L, Gleiter CH. Maprotiline metabolism appears to co- Clin Pharmacol Ther 2004; 76: 302–312.
segregate with the genetically-determined CYP2D6 polymorphic 45 Neugebauer G, Akpan W, Kaufmann B, Reiff K. Stereoselective
hydroxylation of debrisoquine. Br J Clin Pharmacol 1994; 37: 383–388. disposition of carvedilol in man after intravenous and oral administra-
26 Gabris G, Baumann P, Janzier-perey MPB, Woggon B, Küpfer A. N- tion of the racemic compound. Eur J Clin Pharmacol 1990; 38:
methylation of maprotiline in debrisoquine/mephenytoin-phenotyped 108–111.
depressive patients. Biochemical Pharmacology 1985; 34: 409–410. 46 Oldham HG, Clarke SE. In vitro identification of the human
27 Faucette SR, Hawke RL, Lecluyse EL, Shord SS, Yan B, Laethem RM cytochrome P450 enzymes involved in the metabolism of R(+)- and
et al. Validation of bupropion hydroxylation as a selective marker of S()-carvedilol. Drug Metab Dispos 1997; 25: 970–977.
human cytochrome P450 2B6 catalytic activity. Drug Metab Dispos 47 Zhou HH, Wood AJJ. Stereoselective disposition of carvedilol is
2000; 28: 1222–1230. determined by CYP2D6. Clin Pharmacol Ther 1995; 57: 518–524.
28 Barbhaiya RH, Buch AB, Greene DS. Single and multiple dose 48 Graff DW, Williamson KM, Pieper JA, Carson SW, Adams Jr KF, Cascio
pharmacokinetics of nefazodone in subjects classified as extensive WE et al. Effect of fluoxetine on carvedilol pharmacokinetics, CYP2D6
and poor metabolizers of dextromethorphan. Br J Clin Pharmacol 1996; activity, and autonomic balance in heart failure patients. J Clin
42: 573–581. Pharmacol 2001; 41: 97–106.
29 Dostert P, Benedetti MS, Poggesi I. Review of the pharmacokinetics 49 Kirchheiner J, Bauer S, Meineke I, Rohde W, Prang V, Meisel C et al.
and metabolism of reboxetine, a selective noradrenaline reuptake Impact of CYP2C9 and CYP2C19 polymorphisms on tolbutamide

The Pharmacogenomics Journal


Clinical role of polymorphisms in drug metabolism
D Tomalik-Scharte et al
14

kinetics and the insulin and glucose response in healthy volunteers. 69 Sconce EA, Khan TI, Wynne HA, Avery P, Monkhouse L, King BP et al.
Pharmacogenetics 2002; 12: 101–109. The impact of CYP2C9 and VKORC1 genetic polymorphism and patient
50 Stearns RA, Chakravarty PK, Chen R, Chiu SH. Biotransformation of characteristics upon warfarin dose requirements: proposal for a new
losartan to its active carboxylic acid metabolite in human liver dosing regimen. Blood 2005; 106: 2329–2333.
microsomes. Role of cytochrome P4502C and 3A subfamily members. 70 Schalekamp T, Brasse BP, Roijers JF, van Meegen E, van der Meer FJ,
Drug Metab Dispos 1995; 23: 207–215. van Wijk EM et al. VKORC1 and CYP2C9 genotypes and phenprocou-
51 McCrea JB, Cribb A, Rushmore T, Osborne B, Gillen L, Lo MW et al. mon anticoagulation status: interaction between both genotypes
Phenotypic and genotypic investigations of a healthy volunteer affects dose requirement. Clin Pharmacol Ther 2007; 81: 185–193.
deficient in the conversion of losartan to its active metabolite E-31. 71 Montes R, Ruiz de Gaona E, Martinez-Gonzalez MA, Alberca I, Hermida
Clin Pharmacol Ther 1999; 65: 348–352. J. The c.1639G4A polymorphism of the VKORC1 gene is a major
52 Yasar U, Forslund-Bergengren C, Tybring G, Dorado P, Llerena A, determinant of the response to acenocoumarol in anticoagulated
Sjoqvist F et al. Pharmacokinetics of losartan and its metabolite patients. Br J Haematol 2006; 133: 183–187.
E-3174 in relation to the CYP2C9 genotype. Clin Pharmacol Ther 2002; 72 De Morais SM, Wilkinson GR, Blaisdell J, Meyer UA, Nakamura K,
71: 89–98. Goldstein JA. Identification of a new genetic defect responsible for the
53 Lee CR, Pieper JA, Frye RF, Hinderliter AL, Blaisdell JA, Goldstein JA. polymorphism of (S)-mephenytoin metabolism in Japanese. Mol
Tolbutamide, flurbiprofen, and losartan as probes of CYP2C9 activity Pharmacol 1994; 46: 594–598.
in humans. J Clin Pharmacol 2003; 43: 84–91. 73 Andersson T, Holmberg J, Rohss K, Walan A. Pharmacokinetics and
54 Hallberg P, Karlsson J, Kurland L, Lind L, Kahan T, Malmqvist K et al. effect on caffeine metabolism of the proton pump inhibitors,
The CYP2C9 genotype predicts the blood pressure response to omeprazole, lansoprazole, and pantoprazole. Br J Clin Pharmacol
irbesartan: results from the Swedish Irbesartan Left Ventricular 1998; 45: 369–375.
Hypertrophy Investigation vs Atenolol (S0ILVHIA) trial. J Hypertens 74 Klotz U, Schwab M, Treiber G. CYP2C19 polymorphism and proton
2002; 20: 2089–2093. pump inhibitors. Basic Clin Pharmacol Toxicol 2004; 95: 2–8.
55 Uchida S, Watanabe H, Nishio S, Hashimoto H, Yamazaki K, Hayashi H 75 Chong E, Ensom MH. Pharmacogenetics of the proton pump
et al. Altered pharmacokinetics and excessive hypotensive effect of inhibitors: a systematic review. Pharmacotherapy 2003; 23: 460–471.
candesartan in a patient with the CYP2C91/3 genotype. Clin 76 Fuhr U, Jetter A. Rabeprazole: pharmacokinetics and pharmacokinetic
Pharmacol Ther 2003; 74: 505–508. drug interactions. Pharmazie 2002; 57: 595–601.
56 Adcock DM, Koftan C, Crisan D, Kiechle FL. Effect of polymorphisms in 77 Furuta T, Shirai N, Takashima M, Xiao F, Hanai H, Nakagawa K et al.
the cytochrome P450 CYP2C9 gene on warfarin anticoagulation. Arch Effects of genotypic differences in CYP2C19 status on cure rates for
Pathol Lab Med 2004; 128: 1360–1363. Helicobacter pylori infection by dual therapy with rabeprazole plus
57 Miners JO, Birkett DJ. Cytochrome P4502C9: an enzyme of major amoxicillin. Pharmacogenetics 2001; 11: 341–348.
importance in human drug metabolism. Br J Clin Pharmacol 1998; 45: 78 Furuta T, Shirai N, Takashima M, Xiao F, Hanai H, Sugimura H et al.
525–538. Effect of genotypic differences in CYP2C19 on cure rates for
58 Scordo MG, Pengo V, Spina E, Dahl ML, Gusella M, Padrini R. Influence Helicobacter pylori infection by triple therapy with a proton pump
of CYP2C9 and CYP2C19 genetic polymorphisms on warfarin inhibitor, amoxicillin, and clarithromycin. Clin Pharmacol Ther 2001;
maintenance dose and metabolic clearance. Clin Pharmacol Ther 69: 158–168.
2002; 72: 702–710. 79 Furuta T, Ohashi K, Kamata T, Takashima M, Kosuge K, Kawasaki T
59 Higashi MK, Veenstra DL, Kondo LM, Wittkowsky AK, Srinouanpra- et al. Effect of genetic differences in omeprazole metabolism on cure
chanh SL, Farin FM et al. Association between CYP2C9 genetic variants rates for Helicobacter pylori infection and peptic ulcer. Ann Intern Med
and anticoagulation-related outcomes during warfarin therapy. JAMA 1998; 129: 1027–1030.
2002; 287: 1690–1698. 80 Furuta T, Shirai N, Sugimoto M, Ohashi K, Ishizaki T. Pharmacoge-
60 Aithal GP, Day CP, Kesteven PJ, Daly AK. Association of polymorphisms nomics of proton pump inhibitors. Pharmacogenomics 2004; 5:
in the cytochrome P450 CYP2C9 with warfarin dose requirement and 181–202.
risk of bleeding complications. Lancet 1999; 353: 717–719. 81 Wedlund PJ. The CYP2C19 enzyme polymorphism. Pharmacology
61 Takahashi H, Wilkinson GR, Padrini R, Echizen H. CYP2C9 and oral 2000; 61: 174–183.
anticoagulation therapy with acenocoumarol and warfarin:similarities 82 Schwab M, Schaeffeler E, Klotz U, Treiber G. CYP2C19 polymorphism
yet differences. Clin Pharmacol Ther 2004; 75: 376–380. is a major predictor of treatment failure in white patients by use of
62 Morin S, Bodin L, Loriot MA, Thijssen HH, Robert A, Strabach S et al. lansoprazole-based quadruple therapy for eradication of Helicobacter
Pharmacogenetics of acenocoumarol pharmacodynamics. Clin Phar- pylori. Clin Pharmacol Ther 2004; 76: 201–209.
macol Ther 2004; 75: 403–414. 83 Padol S, Yuan Y, Thabane M, Padol IT, Hunt RH. The effect of CYP2C19
63 Schalekamp T, van Geest-Daalderop JH, de Vries-Goldschmeding H, polymorphisms on H. pylori eradication rate in dual and triple
Conemans J, Bernsen MjM, de Boer A. Acenocoumarol stabilization first-line PPI therapies: a meta-analysis. Am J Gastroenterol 2006; 101:
is delayed in CYP2C93 carriers. Clin Pharmacol Ther 2004; 75: 1467–1475.
394–402. 84 Oscarson M. Pharmacogenetics of drug metabolising enzymes:
64 Visser LE, van Vliet M, van Schaik RH, Kasbergen AA, De Smet PA, Vulto importance for personalised medicine. Clin Chem Lab Med 2003; 41:
AG et al. The risk of overanticoagulation in patients with cytochrome 573–580.
P450 CYP2C9*2 or CYP2C9*3 alleles on acenocoumarol or phenpro- 85 Collie-Duguid ES, Pritchard SC, Powrie RH, Sludden J, Collier DA, Li T
coumon. Pharmacogenetics 2004; 14: 27–33. et al. The frequency and distribution of thiopurine methyltransferase
65 Visser LE, van Schaik RH, van Vliet M, Trienekens PH, De Smet PA, alleles in Caucasian and Asian populations. Pharmacogenetics 1999; 9:
Vulto AG et al. The risk of bleeding complications in patients with 37–42.
cytochrome P450 CYP2C9*2 or CYP2C9*3 alleles on acenocoumarol 86 Weinshilboum RM, Sladek SL. Mercaptopurine pharmacogenetics:
or phenprocoumon. Thromb Haemost 2004; 92: 61–66. monogenic inheritance of erythrocyte thiopurine methyltransferase
66 Kirchheiner J, Ufer M, Walter EC, Kammerer B, Kahlich R, Meisel C et al. activity. Am J Hum Genet 1980; 32: 651–662.
Effects of CYP2C9 polymorphisms on the pharmacokinetics of R- and 87 Evans WE, Hon YY, Bomgaars L, Coutre S, Holdsworth M, Janco R et al.
S-phenprocoumon in healthy volunteers. Pharmacogenetics 2004; 14: Preponderance of thiopurine S-methyltransferase deficiency and
19–26. heterozygosity among patients intolerant to mercaptopurine or
67 Hummers-Pradier E, Hess S, Adham IM, Papke T, Pieske B, Kochen azathioprine. J Clin Oncol 2001; 19: 2293–2301.
MM. Determination of bleeding risk using genetic markers in patients 88 Lennard L, Van Loon JA, Lilleyman JS, Weinshilboum RM. Thiopurine
taking phenprocoumon. Eur J Clin Pharmacol 2003; 59: 213–219. pharmacogenetics in leukemia: correlation of erythrocyte thiopurine
68 Schalekamp T, Oosterhof M, van Meegen E, van Der Meer FJ, methyltransferase activity and 6-thioguanine nucleotide concentra-
Conemans J, Hermans M et al. Effects of cytochrome P450 2C9 tions. Clin Pharmacol Ther 1987; 41: 18–25.
polymorphisms on phenprocoumon anticoagulation status. Clin 89 McLeod HL, Coulthard S, Thomas AE, Pritchard SC, King DJ, Richards
Pharmacol Ther 2004; 76: 409–417. SM et al. Analysis of thiopurine methyltransferase variant alleles in

The Pharmacogenomics Journal


Clinical role of polymorphisms in drug metabolism
D Tomalik-Scharte et al
15

childhood acute lymphoblastic leukaemia. Br J Haematol 1999; 105: 111 van Kuilenburg AB, Haasjes J, Richel DJ, Zoetekouw L, Van Lenthe H,
696–700. De Abreu RA et al. Clinical implications of dihydropyrimidine
90 Yates CR, Krynetski EY, Loennechen T, Fessing MY, Tai HL, Pui CH et al. dehydrogenase (DPD) deficiency in patients with severe 5-fluoro-
Molecular diagnosis of thiopurine S-methyltransferase deficiency: uracil-associated toxicity: identification of new mutations in the DPD
genetic basis for azathioprine and mercaptopurine intolerance. Ann gene. Clin Cancer Res 2000; 6: 4705–4712.
Intern Med 1997; 126: 608–614. 112 Sai K, Saeki M, Saito Y, Ozawa S, Katori N, Jinno H et al. UGT1A1
91 Schutz E, Gummert J, Mohr F, Oellerich M. Azathioprine-induced haplotypes associated with reduced glucuronidation and increased
myelosuppression in thiopurine methyltransferase deficient heart serum bilirubin in irinotecan-administered Japanese patients with
transplant recipient. Lancet 1993; 341: 436. cancer. Clin Pharmacol Ther 2004; 75: 501–515.
92 Nagasubramanian R, Innocenti F, Ratain MJ. Pharmacogenetics in 113 Innocenti F, Iyer L, Ratain MJ. Pharmacogenetics of anticancer agents:
cancer treatment. Annu Rev Med 2003; 54: 437–452. lessons from amonafide and irinotecan. Drug Metab Dispos 2001; 29:
93 Colombel JF, Ferrari N, Debuysere H, Marteau P, Gendre JP, Bonaz B 596–600.
et al. Genotypic analysis of thiopurine S-methyltransferase in patients 114 Araki K, Fujita K, Ando Y, Nagashima F, Yamamoto W, Endo H et al.
with Crohn’s disease and severe myelosuppression during azathioprine Pharmacogenetic impact of polymorphisms in the coding region of
therapy. Gastroenterology 2000; 118: 1025–1030. the UGT1A1 gene on SN-38 glucuronidation in Japanese patients with
94 Kroplin T, Weyer N, Gutsche S, Iven H. Thiopurine S-methyltransferase cancer. Cancer Sci 2006; 97: 1255–1259.
activity in human erythrocytes: a new HPLC method using 6- 115 Innocenti F, Undevia SD, Iyer L, Chen PX, Das S, Kocherginsky M et al.
thioguanine as substrate. Eur J Clin Pharmacol 1998; 54: 265–271. Genetic variants in the UDP-glucuronosyltransferase 1A1 gene predict
95 Schwab M, Schaeffeler E, Marx C, Zanger U, Aulitzky W, Eichelbaum the risk of severe neutropenia of irinotecan. J Clin Oncol 2004; 22:
M. Shortcoming in the diagnosis of TPMT deficiency in a patient with 1382–1388.
Crohn’s disease using phenotyping only. Gastroenterology 2001; 121: 116 Ando Y, Saka H, Ando M, Sawa T, Muro K, Ueoka H et al.
498–499. Polymorphisms of UDP-glucuronosyltransferase gene and irinotecan
96 Lennard L. Therapeutic drug monitoring of antimetabolic cytotoxic toxicity: a pharmacogenetic analysis. Cancer Res 2000; 60:
drugs. Br J Clin Pharmacol 1999; 47: 131–143. 6921–6926.
97 Pazmino PA, Sladek SL, Weinshilboum RM. Thiol S-methylation in 117 Maitland ML, Vasisht K, Ratain MJ. TPMT, UGT1A1 and DPYD:
uremia: erythrocyte enzyme activities and plasma inhibitors. Clin genotyping to ensure safer cancer therapy? Trends Pharmacol Sci 2006;
Pharmacol Ther 1980; 28: 356–367. 27: 432–437.
98 Andersen JB, Szumlanski C, Weinshilboum RM, Schmiegelow K. 118 Huang YS, Chern HD, Su WJ, Wu JC, Lai SL, Yang SY et al.
Pharmacokinetics, dose adjustments, and 6-mercaptopurine/metho- Polymorphism of the N-acetyltransferase 2 gene as a susceptibility
trexate drug interactions in two patients with thiopurine methyltrans- risk factor for antituberculosis drug-induced hepatitis. Hepatology
ferase deficiency. Acta Paediatr 1998; 87: 108–111. 2002; 35: 883–889.
99 Evans WE. Pharmacogenetics of thiopurine S-methyltransferase and 119 Brockton N, Little J, Sharp L, Cotton SC. N-acetyltransferase
thiopurine therapy. Ther Drug Monit 2004; 26: 186–191. polymorphisms and colorectal cancer: a HuGE review. Am J Epidemiol
100 Milan G, Etienne MC. 5-Fluorouracil. In: Grochow LB, Ames MM 2000; 151: 846–861.
(Hrsg) (eds.). A Clinical Guide to Chemotherapy Pharmacokinetics and 120 Garte S, Gaspari L, Alexandrie AK, Ambrosone C, Autrup H,
Pharmacodynamics. Williams & Wilkins: Baltimore, 1999 pp 289–300. Autrup JL et al. Metabolic gene polymorphism frequencies in
101 Gonzalez FJ, Fernandez-Salguero P. Diagnostic analysis, clinical control populations. Cancer Epidemiol Biomarkers Prev 2001; 10:
importance and molecular basis of dihydropyrimidine dehydrogenase 1239–1248.
deficiency. Trends Pharmacol Sci 1995; 16: 325–327. 121 Grant DM, Goodfellow GH, Sugamori KS, Durette K. Pharmacogentics
102 Wei X, McLeod HL, McMurrough J, Gonzalez FJ, Fernandez-Salguero of the human arylamine N-actyltransferases. Pharmacology 2000; 61:
P. Molecular basis of the human dihydropyrimidine dehydrogenase 204–211.
deficiency and 5-fluorouracil toxicity. J Clin Invest 1996; 98: 610–615. 122 Tiitinen H et al. Comparison of the isoniazid in activation in Finns and
103 Miller AB, Hoogstraten B, Staquet M, Winkler A. Reporting results of Lapps. Ann Med Int Finn 1968; 57: 161.
cancer treatment. Cancer 1981; 47: 207–214. 123 Parkin DP, Vandenplas S, Botha FJ, Vandenplas ML, Seifart HI, van
104 Frickhofen N, Beck FJ, Jung B, Fuhr HG, Andrasch H, Sigmund M. Helden PD et al. Trimodality of isoniazid elimination: phenotype and
Capecitabine can induce acute coronary syndrome similar to genotype in patients with tuberculosis. Am J Respir Crit Care Med 1997;
5-fluorouracil. Ann Oncol 2002; 13: 797–801. 155: 1717–1722.
105 Collie-Duguid ES, Etienne MC, Milano G, McLeod HL. Known variant 124 Kita T, Tanigawara Y, Chikazawa S, Hatanaka H, Sakaeda T, Komada F
DPYD alleles do not explain DPD deficiency in cancer patients. et al. N-Acetyltransferase 2 genotype correlated with isoniazid
Pharmacogenetics 2000; 10: 217–223. acetylation in Japanese tuberculous patients. Biol Pharm Bull 2001;
106 Fleming RA, Milano G, Thyss A, Etienne MC, Renee N, Schneider M 24: 544–549.
et al. Correlation between dihydropyrimidine dehydrogenase activity 125 Kinzig-Schippers M, Tomalik-Scharte D, Jetter A, Scheidel B, Jakob V,
in peripheral mononuclear cells and systemic clearance of fluorouracil Rodamer M et al. Should we use N-acetyltransferase type 2
in cancer patients. Cancer Res 1992; 52: 2899–2902. genotyping to personalize isoniazid doses? Antimicrob Agents Che-
107 Van Kuilenburg AB, Van Lenthe H, Tromp A, Veltman PC, Van Gennip mother 2005; 49: 1733–1738.
AH. Pitfalls in the diagnosis of patients with a partial dihydropyrimidine 126 Clark DWJ. Genetically determine variability in acetylation and
dehydrogenase deficiency. Clin Chem 2000; 46: 9–17. oxidation: therapeutic implications. Drugs 1985; 29: 342–375.
108 Raida M, Schwabe W, Hausler P, Van Kuilenburg AB, Van Gennip AH, 127 Ohno M, Yamaguchi I, Yamamoto I, Fukuda T, Yokota S, Maekura R
Behnke D et al. Prevalence of a common point mutation in the et al. Slow N-acetyltransferase 2 genotype affects the incidence of
dihydropyrimidine dehydrogenase (DPD) gene within the 50 -splice isoniazid and rifampicin-induced hepatotoxicity. Int J Tuberc Lung Dis
donor site of intron 14 in patients with severe 5-fluorouracil (5-FU)- 2000; 4: 256–261.
related toxicity compared with controls. Clin Cancer Res 2001; 7: 128 Huang YS, Chern HD, Su WJ, Wu JC, Lai SL, Yang SY et al.
2832–2839. Polymorphism of the N-acetyltransferase 2 gene as a susceptibility
109 Van Kuilenburg AB, Baars JW, Meinsma R, Van Gennip AH. Lethal 5- risk factor for antituberculosis drug-induced hepatitis. Hepatology
fluorouracil toxicity associated with a novel mutation in the 2002; 35: 883–889.
dihydropyrimidine dehydrogenase gene. Ann Oncol 2003; 14: 129 Donald PR, Sirgel FA, Venter A, Parkin DP, Seifart HI, van de Wal BW
341–342. et al. The influence of human N-acetyltransferase genotype on
110 Kollmannsberger C, Bokemeyer C, Marx C, Fischer J, Honecker F, the early bactericidal activity of isoniazid. Clin Infect Dis 2004; 39:
Schwab M et al. The association between mutations in the 1425–1430.
dihydropyrimidine dehydrogenase gene and severe toxicity of the 130 Kalow W. Pharmacogenetics and pharmacogenomics: origin, status,
treatment with s-fluorouracil: a prospective multicenter study. and the hope for personalized medicine. Pharmacogenomics J 2006; 6:
Onkologie 2001; 25(Suppl. 6): 101. 162–165.

The Pharmacogenomics Journal

You might also like