You are on page 1of 17

Current Drug Metabolism, 2006, 7, 23-37 23

Cytochrome P450 and Anticancer Drugs

Ken-ichi Fujita*

Department of Clinical Oncology, Saitama medical school, 38 Morohongou, Moroyama-cho, Iruma-gun, 350-0495,
Japan

Abstract: Cytochrome P450 (CYP) is involved in the metabolism of a variety of anticancer drugs. CYP activities are
known to be modified by several factors including genetic polymorphisms, changes in physiological conditions such as
age, disease status or intake of certain drugs or foods or environmental factors such as smoking. These factors may cause
interindividual differences in the pharmacokinetic profiles of anticancer drugs, leading to the variations of efficacy or
toxicity of the drugs.
Genetic polymorphisms present in CYPs sometimes result in the reduced activity of the enzymes causing low metabolic
clearance of drugs or low production of active metabolites. For example, the formation of endoxifen, which is an active
metabolite of tamoxifen, was less in patients with inactive polymorphic CYP2D6 than those with the wild type enzyme.
CYP3A is the most abundant CYP expressed in the human liver and the small intestine that is involved in the metabolism
of various anticancer drugs. The catalytic activity of CYP3A shows a large interindividual variability giving rise to large
interindividual differences in the pharmacokinetic profiles of some anticancer drugs. So far, many attempts have been
made to monitor the phenotypic activity of CYP3A in order to reduce the pharmacokinetic variations of anticancer drugs.
Erythromycin, midazolam and cortisol are commonly used to monitor in vivo hepatic CYP3A activity. These methods
have been applied to reduce the pharmacokinetic variations of docetaxel.
Drug-drug interactions related to CYPs also modulate the pharmacokinetic profiles of anticancer drugs.
These factors should be considered when trying to optimize and individualize chemotherapy.
Key Words: CYP, anticancer drugs, interindividual variation, individualized medicine, chemotherapy.

INTRODUCTION since cancer patients have heterogeneous backgrounds and


complex physiological changes that may be caused by con-
In general, differences in drug absorption, distribution,
comitant disease status, organ dysfunction secondary to pre-
metabolism and excretion result in interindividual variations
vious treatments, tumor invasions, malnutrition and poly-
in the pharmacokinetics of a drug, if patients are adminis-
pharmacy. Nevertheless, recent work has clearly revealed
tered a uniform dose of the drug [1]. Among patients with
that the pharmacokinetic properties of drugs, especially me-
benign diseases treated with common drugs, relatively small
tabolism, were important factors influencing the effective-
differences in drug pharmacokinetics including metabolism
ness and the toxicity of anticancer drugs [2].
are observed. Differences in outcomes are generally negligi-
ble because of the wide therapeutic windows of common Drug metabolism is known to be catalyzed by a range of
drugs. However, in cancer chemotherapy with anticancer drug-metabolizing enzymes including the cytochromes P450
drugs, serious clinical consequences may occur from small (CYPs). CYPs are heme-containing enzymes that catalyze
changes in drug metabolism and pharmacokinetics. Dosing the oxidation of a wide variety of endogenous and exogenous
of anticancer drugs begins with Phase I studies which are compounds, including drugs, carcinogens, and other xenobi-
performed to determine a maximum-tolerated dose (MTD)1. otics [3]. The CYP superfamily is composed of families, and
Subsequently, anticancer drugs are administered clinically to four of the families have the ability to catalyze the oxidation
patients at doses of MTD level or just below the MTD level. of foreign chemicals, including a wide variety of anticancer
The therapeutic windows are narrower compared to other drugs. It is well known that CYP activities are affected by
drugs used for common diseases. Therefore, pharmacoki- several factors [1, 4, 5] including genetic polymorphisms;
netic variability is especially important in chemotherapy. changes in physiological conditions such as age and disease
Accordingly, consideration of factors affecting pharmacoki- states; intake of certain drugs or foods; or other environ-
netic profiles of anticancer drugs is needed. Recognition of mental factors such as smoking. Moreover, the large interin-
specific factors that might alter the response to chemothera- dividual differences in the constitutive expression of CYPs
peutic drugs in individual cancer patients can be difficult in hepatocytes are sometimes observed. The variation of
CYP activities due to these factors may cause interindividual
variations in pharmacokinetic profiles of anticancer drugs
*Address correspondence to this author at the Department of Clinical On- [2]. For example, genetic polymorphisms roughly segregate
cology, Saitama Medical School, 38 Morohongou, Moroyama-cho, Iruma- patients into ‘poor metabolizer (PM)’ and ‘extensive me-
gun, Saitama, 350-0495, Japan; Tel: +81-49-276-2134; Fax: +81-49-276-
2134; E-mail: fujitak@saitama-med.ac.jp tabolizer (EM)’ phenotypes. In general, PM patients possess

1389-2002/06 $50.00+.00 © 2006 Bentham Science Publishers Ltd.


24 Current Drug Metabolism, 2006, Vol. 7, No. 1 Ken-ichi Fujita

reduced or absent ability to clear anticancer drugs compared ROLES OF CYPS IN METABOLISM OF ANTI-
to EM patients, resulting in higher plasma concentrations of CANCER DRUGS
the drugs and more toxic effects. The co-administration of
CYP enzymes are known to be involved in the metabo-
certain drugs or foods that modulate the activity of CYP en-
lism of a variety of anticancer drugs (Table 1). Specific iso-
zymes may lead to decreased metabolic clearance of antican-
forms responsible for the metabolism of each drug have been
cer agents due to the inhibition of drug-metabolizing en- identified through comprehensive in vitro experiments. Since
zymes or to increased clearance due to enzyme induction. species differences in drug metabolism by CYPs have been
The decrement of metabolic clearance of anticancer agents well documented, human CYP preparations should be used.
may result in adverse drug reactions, while increased clear- The metabolism of paclitaxel, for example, may be a case
ance may cause insufficient tumor suppressive effects. Both
demonstrating the remarkable species differences in CYP-
consequences can be severe.
mediated metabolism between rats and humans [6, 7]. One of
Research related to the metabolism and pharmacokinetics major metabolites produced by human CYP2C8 (6α-
of cancer chemotherapeutic agents has been reviewed [1, 2, hydroxypaclitaxel) was not observed in rat bile. The in vitro
4, 5], and it is increasingly apparent that the individualization systems derived from human tissue developed to date in-
of the chemotherapy should take account of the factors that clude subcellular fractions (liver microsomes, cytosols and
modify the metabolism and pharmacokinetics of anticancer homogenates), precision-cut liver slices, and hepatocytes [8-
agents. 18]. Besides these systems, genetically engineered cells ex-
In this review the roles of CYP enzymes in the metabo- pressing human CYPs have been established. Various host
lism of anticancer drugs will be first described. Then, the cells are used for the heterologous expression of CYP iso-
effects of the variability of CYP activities on the interindi- forms. Among them, bacterial cells such as Escherichia coli
vidual variations of pharmacokinetic profiles of anticancer and Salmonella typhimurium have advantages with regard to
drugs will be discussed. the ease of use and the high yield of protein [16-18].

Table 1. CYP Forms Responsible for the Metabolism of Anticancer Drugs

Form Anticancer drugs Pathway Reference*

CYP2A6 Tegafur 5'-Hydroxylation 74

CYP2B6 Cyclophosphamide 4-Hydroxylation 40,41

Ifosfamide 4-Hydroxylation 40,41

N-Dechloroethylation 43

CYP2C8 Paclitaxel 6α-Hydroxylation 6,7,62

CYP2C9 Cyclophosphamide 4-Hydroxylation 42

CYP2C19 Thalidomide 5- and 5'-Hydroxylations 73

CYP2D6 Tamoxifen 4-Hydroxylation 51-53

Endoxifen formation (Secondary metabolite) 51-53

CYP3A4/5 Cyclophosphamide 4-Hydroxylation 40,41

N-Dechloroethylation 43

Docetaxel Hydroxylation 63

Etoposide O-Demethylation 21-23

Gefitinib O-Demethylation 34,35

Ifosfamide 4-Hydroxylation 40,41

N-Dechloroethylation 43

Imatinib N-Demethylation Prescription information

Paclitaxel 3'-p-Hydroxylation 6,7,62

Tamoxifen N-Demethylation 51-53

Teniposide O-Demethylation 21

Vinca alkaloids Unidentified 75-77


* The numbers refer to the numbered references in the text.
Cytochrome P450 and Anticancer Drugs Current Drug Metabolism, 2006, Vol. 7, No. 1 25

Epipodophyllotoxins ure of both platinum-based and docetaxel chemotherapy.


Gefitinib is extensively metabolized in the liver, predomi-
Etoposide and teniposide are semisynthetic derivatives of nantly by CYP3A4 [34, 35]. The known metabolic pathways
epipodophyllotoxin. These anticancer drugs are phase- include dealkylation, O-demethylation and oxidative de-
specific cytotoxic agents acting in the late S and early G2 fluorination. A recent report has shown that CYP3A5 and
phases of the cell cycle. They appear to act by causing
CYP2D6 are also involved in the metabolism of gefitinib
breaks in DNA via an interaction with DNA topoisomerase [35].
II or by the formation of free radicals.
Etoposide is a clinically important antitumor agent de- Oxazaphosphorines
rived from 4’-demethylepipodophyllotoxin which is extracted
from Podophyllum peltatum or Podophyllum emodi [19, 20]. Cyclophosphamide and ifosfamide are oxazaphosphorine
Etoposide is used during chemotherapy for a wide range of derivatives which require metabolic activation by CYP to
malignancies (e.g. small cell lung cancer, acute leukemia, exert their pharmacological activity. There are two distinct
lymphoma, testicular cancer) as a single agent or in combi- metabolic pathways of these drugs.
nation with other agents [20]. The metabolism of etoposide The cytotoxicity of the oxazaphosphorine derivatives is
was investigated by using human liver microsomes and re- produced by a 4-hydroxymetabolite, which enters cells and
combinant human CYP isoforms to identify the CYP forms spontaneously decomposes to phosphoramide mustard and
involved in the major metabolic pathway (O-demethylation) acrolein [36]. Phosphoramide mustard is a bifunctional al-
of etoposide [21-23]. According to the experiments using the kylator of DNA and is the ultimate cytotoxic metabolite of
recombinant CYP forms, the etoposide O-demethylation the oxazaphosphorine derivatives [37-39]. Liver microsomal
was mediated mainly by CYP3A4 and to a minor extent CYP2B6 and CYP3A preferentially catalyzed cyclophos-
by CYP1A2, CYP2E1 and CYP3A5, whereas CYP2A6, phamide and ifosfamide 4-hydroxylation, respectively [40,
CYP2B6, CYP2C8 and CYP2C9 did not contribute to its 41]. On the other hand, Ren et al. [42] have demonstrated
metabolism [21]. Another epipodophyllotoxin, teniposide, is that CYP2C9 and CYP3A seemed to be the major CYPs
also mainly metabolized by CYP3A4 and to a lesser degree responsible for the 4-hydroxycyclophosphamide formation at
by CYP3A5 [21]. CYP2A6, CYP2B6, CYP2C8 and CYP2C9 low and high concentrations of the parent compound, re-
do not play roles in the O-demethylation of teniposide. spectively.
Molecular Targeted Anticancer Drugs Alternatively, the oxazaphosphorine derivatives can be
metabolized to a deschloroethylmetabolite, yielding the renal
Recently, molecular targeted anticancer drugs have been and neurotoxic metabolite chloroacetaldehyde. The deschlo-
developed to target steps in signal transduction, angiogene- roethylmetabolite has no antitumor activity. The N-dechloro-
sis, or the proteasome. The molecular targeted drugs can be ethylation pathway of cyclophosphamide is catalyzed by
either small molecules or antibodies. CYPs are responsible CYP3A4, but for ifosfamide it is catalyzed by both CYP3A4
for the metabolism of small molecules but not antibodies. and CYP2B6 [43].

Imatinib Despite their structural similarities, cyclophosphamide


and ifosfamide exhibit marked differences in their metabo-
Imatinib mesylate (Gleevec, ST1571) is a potent com- lism, toxicity, and therapeutic spectrum. Approximately 45%
petitive inhibitor of tyrosine kinases (TKs) associated with of a therapeutic dose of ifosfamide is typically metabolized
BCR-ABL [24, 25], Kit [26, 27], which impedes the interac- via N-dechloroethylation, whereas only 10% of cyclophos-
tion of ATP with the Src homology 1 domain of these pro- phamide is converted via this pathway [44]. Consequently,
teins, thereby inhibiting the phosphorylation of target pro- more patients developed deschloroethylmetabolite-induced
teins acting downstream of the signal transduction. Imatinib neurotoxicity and nephrotoxicity after ifosfamide treatment
is a phenylaminopyrimidine derivative and represents the than cyclophosphamide treatment. Clinically, neurotoxicity
first of a new class of drug known as signal transduction and nephrotoxicity are dose-limiting factors for ifosfamide
inhibitors. Following initial Phase I and II dose-escalation treatment [45]. Neurotoxicity occurs in about 20% of pa-
studies of imatinib [28-31], studies have demonstrated re- tients after ifosfamide treatment, but is rare in cyclophos-
markable efficacy with minimal side effects not only in phamide treated patients. For these reasons, it is likely that
Philadelphia-positive leukemia (BCR-ABL positive) but also the safety and the therapeutic effects of oxazaphosphorine
in solid tumors such as gastrointestinal stromal tumors ex- derivatives could be improved by minimizing the N-
pressing Kit [32, 33]. CYP3A4 is the major CYP involved in dechloroethylation pathway.
the microsomal biotransformation of imatinib (Prescription
information, Novartis Pharmaceuticals Corporation, East The oxazaphosphorine derivatives contain a chiral phos-
Hanover). phorous atom. They are commonly used as racemic mixtures
of (+)-R and (-)-S enantiomers. According to an in vitro
Gefitinib study on the stereoselective metabolism of ifosfamide, the R-
enantiomer shows more favorable properties than the S-
Gefitinib (Iressa; AZ1839) is a member of a new class of enantiomer with respect to less extensive N-dechloro-
oral drugs that inhibit receptor TKs, including the epidermal ethylation and more rapid 4-hydroxylation, indicating that R-
growth factor receptor TK [34]. This anticancer drug re- ifosfamide may have a distinct clinical advantage over race-
ceived approval as monotherapy for patients with locally mic ifosfamide [46].
advanced or metastatic non-small cell lung cancer after fail-
26 Current Drug Metabolism, 2006, Vol. 7, No. 1 Ken-ichi Fujita

Tamoxifen paclitaxel, was present in both human and rat bile. On the
other hand, one of the major metabolites present in human
The selective estrogen receptor modulator, tamoxifen, bile, 6α-hydroxypaclitaxel, was not present in the rat bile
has been widely used for more than 25 years for the endo- [61], indicating species differences in the biotransformation
crine treatment of all stages of hormone receptor–positive of paclitaxel between rats and humans. The biotransforma-
breast cancer [47]. Tamoxifen is also approved by the United
tion of paclitaxel by human liver was investigated in vitro
States Food and Drug Administration for the prevention of with liver microsomes isolated from adults. Cresteil et al. [6]
breast cancer in women at high risk for developing the dis-
first concluded that CYP2C accounted for the formation of
ease [48].
6α-hydroxypaclitaxel, which was the major metabolite
Jordan et al. [49] demonstrated that hepatic metabolism formed in vivo and in vitro, while CYP3A supported the 3’-
of tamoxifen resulted in a statistically significant increase in p-hydroxypaclitaxel formation. Subsequently, Rahman et al.
its efficacy in vivo; they also showed for the first time that 4- [62] demonstrated that 6α-hydroxypaclitaxel formation from
hydroxytamoxifen, one of the human tamoxifen metabolites, paclitaxel was catalyzed by CYP2C8.
was approximately 100 times more potent than tamoxifen as In contrast to paclitaxel, the biotransformation pathway
an estrogen antagonist in vitro. The tamoxifen metabolite N- of docetaxel is similar in animals and humans. The initial
desmethyltamoxifen is 100 times less active than tamoxifen metabolite resulted from the hydroxylation of the methyl of
in vitro [50], although it is more abundant in plasma of pa- the tert-butyl group at the C 13 side chain of docetaxel. Royer
tients receiving tamoxifen treatment than tamoxifen itself. et al. [63] have documented that docetaxel hydroxylation
Recently, the activity of another tamoxifen metabolite, en- was catalyzed by human CYP3A, based on the correlation of
doxifen (4-hydroxy-N-desmethyltamoxifen), which is pre- the metabolism with CYP3A-dependent 6β-hydroxylation of
sent at notably higher concentrations than 4-hydroxytamox- testosterone and 16-hydroxylation of dehydroepiandroster-
ifen in the plasma of breast cancer patients receiving chronic one, and inhibition with CYP3A inhibitors.
tamoxifen therapy was characterized [51]. In a series of in
vitro studies [51], endoxifen exhibited the same potency and Thalidomide
efficacy as 4-hydroxytamoxifen in suppressing estrogen-
dependent breast cancer cell growth and gene expression. Thalidomide is an old compound that was originally de-
veloped as a sedative. The drug was eventually withdrawn
Tamoxifen is metabolized extensively by human liver from the general market because of its severe adverse effect
enzymes to primary and secondary metabolites [51-53]. A of teratogenesis. It has been reintroduced over the past sev-
comprehensive kinetic characterization of tamoxifen se- eral years in the treatment of leprosy. More recently, the drug
quential metabolism in vitro demonstrated that CYP3A is the has been found to be effective against various diseases in-
major CYP subfamily responsible for the formation of N- cluding multiple myeloma and prostate cancer, at least in
desmethyltamoxifen, whereas the generation of 4-hydroxy- part, through the inhibition of angiogenesis [64-66]. Results
tamoxifen and endoxifen appeared to be predominantly obtained so far suggested that thalidomide requires CYP
catalyzed by CYP2D6. catalyzed biotransformation to exert its pharmacological
Although tamoxifen therapy is associated with benefits activities including antiangiogenesis [67-69], though the
for breast cancer, it is also associated with several adverse exact mechanism of action has not been well clarified. Al-
events, including endometrial cancer [54]. Shibutani and though thalidomide is subject to spontaneous nonenzymatic
colleagues [55-58] demonstrated that a hydroxylated ta- hydrolysis, these breakdown products may not be responsi-
moxifen metabolite, α-hydroxytamoxifen, produced by hu- ble for the pharmacological activity [70]. The hydroxylated
man CYP3A was further metabolized by sulfotransferases to metabolites, 5-hydroxythalidomide, cis- and trans-5’hyd-
form an O-sulfonated metabolite that produced tamoxifen- roxythalidomide, have been found in plasma or urine of pa-
DNA adducts. The tamoxifen-DNA adducts were detected in tients [71, 72]. Recently, it has been reported that CYP2C19
the endometrium of women taking tamoxifen. These results is predominantly involved in the 5- and 5’-hydroxylation of
suggest the α-hydroxytamoxifen may be responsible for the thalidomide in humans [73].
adverse carcinogenic effects of tamoxifen.
Tegafur
Taxanes Tegafur is a prodrug of 5-fluorouracil (5-FU) synthesized
Taxanes are a family of structurally related compounds more than 30 years ago. The anticancer drug has been clini-
that share a core ring structures referred to as baccatin III. cally used for the treatment of mainly gastric and colon can-
Two members of this family, paclitaxel and docetaxel, are cers because of the lower toxicity than 5-FU. 5-FU exerts its
important anticancer drugs extensively used in the treatment cytotoxic effects via the inhibition of thymidylate synthase
of various malignancies including ovarian cancer, breast and/or its incorporation into RNA molecules. The active
cancer and lung cancer [59]. compound, 5-FU, is then further metabolized by dehydro-
pyrimidine dehydrogenase to form inactive metabolites. Te-
The metabolism of paclitaxel has been studied in both gafur is converted to 5-FU mainly in the liver through an
rats and human patients [6, 7]. The results showed that pa- unstable intermediate, 5’-hydroxytegafur. The formation of
clitaxel metabolites observed in the bile of a human patients 5-FU from tegafur is known to be mainly catalyzed by hu-
were mostly different from those observed in rat bile [6, 7]. man CYP2A6 [74] as shown in experiments with human liver
Nine metabolites were detectable in rat bile [60] and in five microsomes that revealed a significant correlation between
metabolites were in the bile of a patient with metastatic cho- 5-FU formation from tegafur and 7-hydroxylation of cou-
langiocarcinoma [61]. Only one metabolite, 3’-p-hydroxy- marin, a representative metabolic reaction for CYP2A6.
Cytochrome P450 and Anticancer Drugs Current Drug Metabolism, 2006, Vol. 7, No. 1 27

Vinca Alkaloids [74], CYP2A6 polymorphisms may affect the pharmacoki-


netics of tegafur. Patients with lower CYP2A6 activity may
Vinca alkaloids, including vinblastine, vincristine, vinde- benefit less from tegafur treatment because of insufficient
sine and vinorelbine are anticancer drugs widely used in exposure to 5-FU.
combination with other drugs in cancer chemotherapy. They
have been known to exert their anticancer effects through the In a clinical study, a newly developed anticancer drug S-
inhibition of microtubule polymerization which causes the 1, which contained tegafur and 5-chloro-2, 4-dihydroxypyri-
antimitotic effect. These anticancer drugs were proven to be dine, an inhibitor of dihydropyrimidine dehydrogenase, was
metabolized by human CYP3A [75-77]. Vinorelbine bio- orally administered to five gastric cancer patients. The total
transformation observed with human liver microsomes con- AUC in one patient was four-fold higher than in other pa-
sisted of one saturable and one nonsaturable processes with tients [81]. Alleles for the CYP2A6 genes derived from this
Km and Vmax values of 1.90 µM and 25.3 pmol/min/mg patient were completely sequenced [81]. CYP2A6*4A (iden-
protein, respectively [77]. tical to CYP2A6 *4C ) was found, which indicated the dele-
tion of an entire allele for the human CYP2A6 gene [82].
EFFECTS OF VARIABILITY OF CYPS ON INTER- Another allele, CYP2A6*11 which is a novel mutant allele in
INDIVIDUAL VARIATIONS OF PHARMACOKINE- which thymine at nucleotide 670 is changed to cytosine was
TIC PROFILES OF ANTICANCER DRUGS also found. The nucleotide change caused an amino acid
change from serine at residue 224 to proline. The Vmax
Cancer chemotherapy is characterized by a wide varia- value for tegafur metabolism by the mutant CYP2A6 was
tion in response among patients. After the intake of identical approximately one-half of the value of the intact CYP2A6,
doses of a given agent, some patients may have clinically although the Km value was nearly the same. Therefore, the
significant adverse effects, whereas others may experience higher AUC seen in the patient was attributed to the presence
no therapeutic response. The interpatient variability seen in of CYP2A6*4A and CYP2A6*11 alleles. The result supports
the response to anticancer drugs is, in part, caused by phar- the hypothesis that patients possessing valiant alleles of the
macokinetic factors. The most widely used traditional strat- CYP2A6 gene may not benefit from tegafur treatment. If
egy to decrease pharmacokinetic variability is to normalize a patients are homozygous for CYP2A6*4A genotype, the ac-
drug dose to the patient’s body surface area (BSA). How- tivity of CYP2A6 may be absent, resulting in the null activa-
ever, BSA-based dosing strategies often do not reduce inter- tion of tegafur. However, 5-FU is also known to be formed
individual variability in anticancer drug pharmacokinetics from tegafur by liver cytosolic thymidine phosphorylase
[78], though BSA-based doses did, in fact, correlate with [83]. The clinical significance of CYP2A6 activity on tegafur
paclitaxel area under the concentration-time curves (AUCs) biotransformation might also depend on the activity of the
[79]. cytosolic enzyme. Further clinical studies are needed to clar-
Some of the diversity in rates of response to anticancer ify the role of CYP2A6 in the bioactivation of tegafur. The
drugs can be ascribed to differences in the rate of drug me- CYP2A6 allele frequencies are shown in Table 2.
tabolism, particularly activity of CYP enzymes. Variability
among patients in the activity of these enzymes reflects a CYP2D6
complex interaction between both genetic and nongenetic
The CYP2D6 gene is a polymorphic gene which has more
host factors and environmental factors. Therefore, the con- than 80 distinct allelic variants with 52 reported allelic vari-
sideration of these factors may be crucial to understanding ants (http://www.imm.ki.se/CYPalleles), many of which
interpatient variability in response to anticancer drugs, and
result in the loss of CYP2D6 enzyme function [94]. Effects
ultimately to reducing it via personalized chemotherapy.
of CYP2D6 polymorphism on the metabolism of anticancer
drugs have been increasingly studied.
Genetic Variations of CYPs Related to Pharmacokinetics
of Anticancer Drugs The clinical effects of tamoxifen with respect to efficacy
and toxicity vary widely among individuals. For example,
The results of a recent meta-analysis demonstrated that among women with advanced breast cancer, roughly 35% of
adverse drug reactions were more likely for drugs metabo- those with estrogen receptor–positive tumors do not respond
lized by enzymes that had genetic polymorphisms [80]. As to tamoxifen therapy, and all tumors that do respond eventu-
described above, CYP enzymes play essential roles in the ally become resistant to tamoxifen treatment [45].
metabolism of various anticancer drugs. Therefore, the ef-
fects of genetic polymorphisms in CYP forms on the phar- Jin et al. [95] investigated the mean plasma concentra-
macokinetic properties of anticancer drugs are important to tions of tamoxifen and its metabolites after 4 months of ta-
consider. moxifen therapy in breast cancer patients with different
CYP2D6 genotype groups, based on the hypothesis that the
CYP2A6 CYP2D6 genotype might affect the pharmacokinetics of ta-
moxifen and its metabolites. CYP2D6 genotype was not sta-
Several CYP2A6 alleles which alter CYP2A6 function tistically significantly associated with mean plasma concen-
re caused by polymorphisms in the open reading frame of the trations of tamoxifen, 4-hydroxytamoxifen and N-desmethyl-
gene (see http://www.imm.ki.se/CYPalleles). Polymor- tamoxifen. However, subjects who were CYP2D6*1/*3,
phisms present in the 5’-flanking region of the CYP2A6 gene *1/*4, *1/*5, or *1/*6 heterozygotes (wild/variant genotype
which modify the transcriptional activity of CYP2A6 have group) had mean plasma endoxifen concentrations that were
also been documented (http://www.imm.ki.se/CYPalleles). 55% of those of subjects who were homozygous for the wild
Since CYP2A6 is responsible for the bioactivation of tegafur CYP2D6 genotype. Subjects who were CYP2D6*4/*4 homo-
28 Current Drug Metabolism, 2006, Vol. 7, No. 1 Ken-ichi Fujita

Table 2. CYP2A6 Allele Frequencies

Canadian
African-
Caucasian Japanese Korean Chinese Native
American
Indean

CYP2A6*1A 57.3 66.5 42.4 40.4 45.9 43.2

CYP2A6*1B 27.6 33.5 11.2 13 27.7 48.4 41.2 37.1 40.6 51.3 55

CYP2A6*1C 1.8

CYP2A6*1F 1.8 0

CYP2A6*1G 1.2 13.3

CYP2A6*1X2 0.7 0 0 0.4 0

CYP2A6*2 1.2 2 0 1.1 0 0 0 0 0 0

CYP2A6*4 1.2 1.9 20.1 24.2 11 15.1 6.7 1

(CYP2A6*4A) 3 0.5 18.4

(CYP2A6*4B) 0.24

(CYP2A6*4D) 0 0.5

CYP2A6*5 0.1 0 0 0 0.5 1 0.5 0.5

CYP2A6*6 0 0

CYP2A6*7 0.3 0 0 0 6.5 6.3 13 3.6 10 3.1 0

CYP2A6*8 0.1 0 2.2 1.6 1.4 3.6 0

CYP2A6*9 7.9 7.1 8 7.1 20.3 15.6 15.5

CYP2A6*10 0 0 1.1 1.6 0.5 0.4 0


*
CYP2A6*11

CYP2A6*12 2 0.4 0.8 0 0.5

CYP2A6*13 0 1.5

CYP2A6*14 3.6 0

CYP2A6*15 0 1.5

CYP2A6*17 0 9.4 0 0

CYP2A6*16 3.6 0

CYP2A6*18A 1.1 0 0 0.5

CYP2A6*18B 1.1 0 0 0

CYP2A6*19 0 0 0.5 1

CYP2A6*20 0 1.6 0 0
**
Reference 84 85 86 87 88 89 84 85 87 88 89 90 85 92 93 86 87 88 89 90 87 88 89 91 85 85
* Allele frequency of CYP2A6*11 has not yet been reported.
** The numbers refer to the numbered references in the text.
Details of the CYP2A6 gene polymorphisms are represented in http://www.imm.ki.se/CYPalleles/cyp2a6.htm.

zygotes (variant/variant genotype group) had mean plasma moxifen to breast cancer patients depends on the CYP2D6
endoxifen concentrations that were 26% of those of subjects genotype. The results of definitive clinical trials that include
having the wild CYP2D6 genotype. Although, Jin et al. still outcomes such as breast cancer recurrence and mortality,
did not show direct evidence indicating the relationship be- toxicity and secondary benefits are awaited. CYP2D6 allele
tween the efficacy of tamoxifen and CYP2D6 genotype, frequencies are shown in Table 3.
these results may suggest that the clinical benefit of ta-
Cytochrome P450 and Anticancer Drugs Current Drug Metabolism, 2006, Vol. 7, No. 1 29

Table 3. CYP2D6 Allele Frequencies

Saudi
Caucasian European German Russian Spaniad Japanese Chinese Zimbabwean Ethiopian
Arabian

CYP2D6*1 36.4 32.2 70.8 42.3 43 40 26.9

CYP2D6*2 32.4 32.1 28.5 9.2 12.3 13 12.5 14.3

CYP2D6*3 2.04 1.6 1 0

CYP2D6*4 20.7 17.2 19.5 21.3 18.2 11.6 0.5 0.2 0 0.8 2 1.2 3.5

CYP2D6*5 6.9 4.1 2.1 2.4 3.9 6.1 4.5 6.2 5.7 5.7 4 3.3 1

CYP2D6*6 0.93 1.2 1.2

CYP2D6*8 0 0.1 0

CYP2D6*9 2.7

CYP2D6*10 1.53 2 1.6 4.2 40.8 38.1 38.6 50.7 5.6 8.6 3

(CYP2D6*10B) 1.3 50.7

CYP2D6*11 0 0.1

CYP2D6*12 0 0.1

CYP2D6*14 0 0.7 2.2 0

CYP2D6*15 0.08

CYP2D6*17 0.1 34 9 3

CYP2D6*18 0 0.2 0 0

CYP2D6*21 1 0

CYP2D6*36 37.1

CYP2D6*1X2 1.7 0.5

CYP2D6*2X2 1 1.8 0.5 3.5 0.5 0.9 2 16 10.4


*
Reference 96 97 98 99 100 99 101 102 98 98 103 99 104 99

* The numbers refer to the numbered references in the text.


Details of the CYP2D6 gene polymorphisms are represented in http://www.imm.ki.se/CYPalleles/cyp2d6.htm.

CYP3A4/CYP3A5 CYP3A4*17, and CYP3A4*18) have been shown to result


in functional changes in CYP3A activity in vitro [118,
In adults, CYP3A4 and CYP3A5 are the most important 119]. The allele frequencies of CYP3A4 variants are show in
among the four CYP3A subfamily members for CYP3A-
Table 4.
mediated drug metabolism [105-107]. The genetic diversity
in the genes encoding these proteins has been reported [108]. CYP3A5 is known to be expressed in only about 10% of
Over 30 single nucleotide polymorphisms in CYP3A4 white subjects because of a splice variant in intron 3 of the
have been published, representing alleles CYP3A4*1 to CYP3A5 gene at nucleotide position 6986 (CYP3A5*3) [127,
CYP3A4*19. However, most are very rare and unlikely to 128]. Approximately 85 to 95% of white subjects, 70% of
impact CYP3A4 activity in vivo. The best characterized vari- Japanese, and 35 to 45% of blacks are homozygous for
ant, a promoter variant with an adenine to guanine transition CYP3A5*3 and thus deficient in CYP3A5 [127-129]. An-
at nucleotide –392 (CYP3A4*1B), was shown in vitro to ex- other splice variant (CYP3A5*6), which is observed in black
hibit increased transcriptional activity [108-110]. Two re- populations, also results in the lack of CYP3A5 expression
ports, however, indicate decreased transcriptional activity of [127, 128]. These results suggest that the metabolism of anti-
CYP3A4*1B [111, 112]. No association was observed be- cancer drugs which depend mainly for their metabolism on
tween the CYP3A4*1B variant and CYP3A activity in studies CYP3A5 may be polymorphic, although there is no antican-
with healthy subjects using the CYP3A-phenotyping probes cer drug which is specifically metabolized by CYP3A5
midazolam [113-115], erythromycin [114, 116] and nifedip- at present. The CYP3A5 allele frequencies are shown in
ine [117]. Three other CYP3A4 polymorphisms (CYP3A4*6, Table 5.
30 Current Drug Metabolism, 2006, Vol. 7, No. 1 Ken-ichi Fujita

Table 4. CYP3A4 Allele Frequencies

African- Japanese- Chinese- Hispanic- Middle Pacific


Caucasian African Asian Japanese Chinese Taiwanese Mexican
American American American American Eastern islander

CYP3A4*1

(CYP3A4*1A) 73.4

(CYP3A4*1B) 6.5 9 48 53 54.6 66.7 0 0 0 0 0 0 9.3 1 15 14

(CYP3A4*1G) 18.9

(CYP3A4*1H) 1

CYP3A4*2 0 2.7 0 0 0 0 0 0 0 0

CYP3A4*3 0 0.47 4.2 1.1 3.6 4.2 0 0 0 0 0 0 0 0

CYP3A4*4 0 0 0 0 1.47 0 0 0

CYP3A4*5 0 0 0 0 0.98 0 0 0

CYP3A4*6 0 0 0 0.1 0 0.49 0 0 0

CYP3A4*7 0 1.41 0 0 0 0 0 0

CYP3A4*8 0 0.33 0 0 0 0 0 0

CYP3A4*9 0 0.24 0 0 0 0 0 0

CYP3A4*10 2 0.24 2 0 0 5 0 0

CYP3A4*11 0 0.34 0 0 0.2 0 0 0 0

CYP3A4*12 0 0.34 0 0 0 0 0 0

CYP3A4*13 0 0.34 0 0 0 0 0 0

CYP3A4*14 0 0 0 0 0 0 0

CYP3A4*15 0 0 2 4.2 0 0 0 0 0 0

CYP3A4*16 0 0 5 1.4 0 5 0 0

CYP3A4*17 2.1 0 0

CYP3A4*18 0 0 2.1 2.8

CYP3A4*19 0 0 2.1

Reference* 120 121 118 122 123 124 125 120 125 123 124 118 118 120 126 123 120 119 124 123 125 123 120 120 120

* The numbers refer to the numbered references in the text.


Details of the CYP3A4 gene polymorphisms are represented in http://www.imm.ki.se/CYPalleles/cyp3a4.htm.

Phenotypic Variation of CYP3A4 Activity Related to Extensive attempts have been made to manipulate the
Pharmacokinetics of Anticancer Drugs large variation of CYP3A4 activity and to reduce pharma-
cokinetic variability of the relevant drugs. Since the genetic
The enzyme activity of CYP3A4, which is responsible variations seen in CYP3A4 are estimated to be small as
for the biotransformation of a wide variety of anticancer
mentioned above, phenotyping strategies to predict an indi-
drugs (Table 1), exhibits a remarkable interindividual varia- vidual’s CYP3A activity before cytotoxic chemotherapy
tion as high as 20 to 40 fold [108, 133]. In addition to the treatment could be an appropriate approach for dose indi-
large variation caused by the difference of constitutive ex- vidualization. Various noninvasive in vivo probes for evalu-
pression, CYP3A4 expression is affected by the concomitant ating CYP3A activity have been described and several have
intake of CYP3A4 inducers such as rifampicin. The co- been shown to correlate with drug clearance [134-138]. The
administration of therapeutic drugs which can modulate the most widely tested and accepted CYP3A probes are midazo-
CYP3A4 activity can alter the metabolism of anticancer
lam, erythromycin and cortisol, although selection of the
drugs. It has been recently reported that liver function to- ideal CYP3A-probe remains controversial [134-138].
gether with the concentration of the acute-phase reactant, α-1
acid glycoprotein, explained up to 18% of overall variation in Docetaxel, which is mainly metabolized by CYP3A4 in
CYP3A activity [116]. humans [63], shows large interindividual variability in its
Cytochrome P450 and Anticancer Drugs Current Drug Metabolism, 2006, Vol. 7, No. 1 31

Table 5. CYP3A5 Allele Frequencies

Southwestern
African- Pacific
Caucasian African Asian Japanese Korean Chinese Mexican American
American Islander
Indian

CYP3A5*1 15 45 31 15 25 35 60

(CYP3A5*1B) 3 0.5

(CYP3A5*1C) 3 4.3

CYP3A5*2 5.26 1.9 0 0

CYP3A5*3 91.8 88 95.1 27 35 75 71 70 72 67

CYP3A5*4 0.9

CYP3A5*5 0.9

CYP3A5*6 0 13 17 0

CYP3A5*7 0 10 8 0

CYP3A5*8 0 4 0

CYP3A5*9 0 0 2

CYP3A5*10 2 0 0

CYP3A5*11*

Reference** 127 130 131 132 128 127 128 132 132 127 128 128 131 127 128 127 127 127

* The allele frequency of CYP3A5*11 has not yet been reported.


** The numbers refer to the numbered references in the text.
Details of the CYP3A5 gene polymorphisms are represented in http://www.imm.ki.se/CYPalleles/cyp3a5.htm.

pharmacokinetic/pharmacodynamic profiles [139, 140]. The domized study, they examined whether individualized dosing
effects of inhibition and induction of CYP3A on docetaxel via CYP3A phenotyping could decrease the pharmacoki-
pharmacokinetics was investigated in mice [141]. Pretreat- netic/pharmacodynamic variability of docetaxel compared to
ment of mice with the CYP3A inhibitor, ketoconazole, in- more routine BSA-based dosing [144]. Fifty-nine patients
creased the AUC of docetaxel, whereas treatment with the with advanced non-small cell lung cancer were randomly
CYP3A inducer, dexamethasone, decreased the AUC of do- assigned to either the BSA-based arm or the individualized
cetaxel. These findings suggest that enzymatic inhibition and arm. In the BSA-based arm, 60 mg/m2 of docetaxel was ad-
induction of CYP3A might affect docetaxel pharmacokinet- ministered. In the individualized arm, the doses of docetaxel
ics and its antitumor potency in humans. were calculated from the estimated clearance and a target
AUC of 2.66 mg·h/L. The standard deviation of the AUC
Attempts have been made to quantify the phenotypic
was significantly smaller in the individualized arm than in
activity of CYP3A4 and to evaluate the correlation between
the activity and the pharmacokinetic parameters of do- the BSA-based arm. The respective percentage decreases in
cetaxel. In a study with 21 sarcoma patients who were absolute neutrophil counts were 87.1±8.7% in the BSA-
treated with 100 mg/m2 of docetaxel, the erythromycin based arm and 87.4±6.1% in the individualized arm. The
breath test (ERMBT) using [ 14C-N-methyl]erythromycin was results indicate that the interpatient variability in percent
employed to measure hepatic CYP3A activity in vivo [142]. decreases in absolute neutrophil counts was slightly smaller
The natural log of ERMBT accounted for 67% of the inter- in the individualized arm. In another study, docetaxel clear-
ance was found to be correlated with midazolam clearance
patient variability in the docetaxel clearance. In another
[145].
study, Yamamoto et al. [143] measured urinary 6β-
hydroxycortisol in 30 patients with non-small cell lung can- These studies suggest that CYP3A4 activity could be a
cer treated with 60 mg/m2 docetaxel after the administration useful indicator to predict in vivo docetaxel clearance and
of the 300 mg of hydroxycortisone. Cortisol is selectively toxicity, and that CYP3A4-guided dosing could be a useful
metabolized by CYP3A and excreted in the urine as 6β- method for docetaxel dosing.
hydroxycortisol [138]. They used 300 mg of exogenous cor- Irinotecan is a camptothecin analogue with potent antitu-
tisone, since the amount of 6β-hydroxycortisol produced
mor activity resulting from inhibition of topoisomerase I. It
from endogenous cortisol was too low to be sensitively de- is widely used for the treatment of colorectal and lung can-
tected in urine. Multivariate analysis revealed that the total cers [146-148]. Pharmacokinetic variability of irinotecan
amount of 24 h urinary 6β-hydroxycortisol was the strongest
among patients is also large. Pathways that eliminate irinote-
factor predicting docetaxel clearance. In a subsequent ran-
32 Current Drug Metabolism, 2006, Vol. 7, No. 1 Ken-ichi Fujita

can include CYP3A, carboxylesterase, UGT1A1, and drug- mg of imatinib once daily orally and on study day 8, 400 mg
transporting proteins [149]. CYP3A4 was reported to play a imatinib together with 40 mg of simvastatin was given.
role in irinotecan metabolism, though CYP3A5 did not Imatinib increased the mean maximum concentration
[150]. Mathijssen et al. [151] demonstrated a correlation (Cmax) value of simvastatin two-fold and the AUC value
between irinotecan and midazolam clearance. Because dos- 3.5-fold compared with simvastatin alone. Recently, the ef-
ing strategies that were based on BSA did not reduce the fect of ketoconazole, a potent CYP3A inhibitor, on the
interindividual variability in irinotecan pharmacokinetics, pharmacokinetics of imatinib was investigated [155]. Each
they recommended evaluating midazolam clearance com- subject received a single oral dose of imatinib 200 mg alone,
bined with the UGT1A1*28 polymorphism for optimizing and a single oral dose of imatinib 200 mg co-administered
irinotecan chemotherapy. The active irinotecan metabolite, with a single oral dose of ketoconazole 400 mg according to
SN-38, is further processed by glucuronidation. Their results a two-period crossover design. Following ketoconazole co-
supported the idea that a CYP3A-guided strategy is applica- administration, the mean imatinib Cmax and AUC increased
ble for dosing anticancer drugs that are mainly metabolized significantly by 26 and 40%, respectively. There was a sta-
by CYP3A. tistically significant decrease in apparent clearance of
imatinib with a mean reduction of 28.6%. These interactions
Drug Interactions Relevant to Anticancer Drugs should be considered when administering substrates or in-
hibitors of the CYP3A family in combination with imatinib.
Metabolic drug-drug interactions occur when one drug
Conversely, co-administration of rifampicin and St John’s
alters the activity of a drug-metabolizing enzyme, thereby
wort, which are potent inducers of CYP3A4, decreased the
affecting the pharmacokinetics of drugs metabolized by the Cmax and AUC of imatinib in healthy volunteers [156-158].
enzyme. Various interactions have been reported thus far. Therefore, the concomitant use of enzyme inducers may ne-
For example, the metabolism of terfenadine by human cessitate an increase in the imatinib dose to maintain clinical
CYP3A is known to be inhibited by azole antifungal drugs,
effectiveness.
causing a notable increase in the plasma concentration of the
parent drug causing severe adverse drug reactions associated
Paclitaxel
with prolongation of the corrected QT interval in humans
[152]. The co-administration of drug(s) with anticancer Glioma patients commonly receive anticonvulsants such
drugs has also been reported to induce drug interactions by as phenytoin, phenobarbital and carbamazepine and corti-
modifying their pharmacokinetic properties. Although, drug costeroids that may increase the hepatic metabolism of pa-
interactions are generally considered to cause adverse drug clitaxel by CYPs. A Phase I study to determine the MTD of
reactions, they sometimes might cause benefits such as an paclitaxel administration as a 3 h infusion in patients with
improvement in drug bioavailability. For example, the inhi- recurrent malignant glioma was performed [159]. The MTD
bition of CYPs in the small intestine may result in the eleva- for patients who received anticonvulsants was established to
tion of the bioavailability of drugs which are orally adminis- be 360 mg/m2, whereas the MTD for patients who did not
tered. receive anticonvulsants was 210 mg/m2. Pharmacokinetic
data demonstrated that the plasma paclitaxel levels and
Docetaxel clearance rates were similar for patients in both groups at
respective dose levels that produced dose-limiting toxicity.
A proof-of-concept study was carried out in 14 patients
These results indicate that the pharmacokinetics of paclitaxel
with solid tumors [153]. In one course, patients received oral
are altered by the co-administration of anticonvulsants, and
docetaxel of 75 mg/m2 with or without a single oral dose of
significantly larger doses of the drug can be administered to
cyclosporine A (15 mg/kg). During subsequent courses, pa-
patients with brain tumors who require anticonvulsants. Sub-
tients received 100 mg/m2 of intravenous docetaxel. The sequently, a Phase II study to confirm the effects of pacli-
AUCs seen in patients who received oral docetaxel 75 mg/m2
taxel on recurrent malignant glioma was performed using
with and without cyclosporine A were 2.71 ± 1.81 and 0.37 ± doses determined in the Phase I study. Unfortunately, no
0.33 mg·h/L, respectively. The absolute bioavailability of
objective clinical response to any dose of paclitaxel for re-
oral docetaxel observed with or without cyclosporine A, current malignant glioma was observed [160].
which was calculated from the mean AUC of intravenous
docetaxel, was 90 ± 44 or 8 ± 6%. Co-administration of oral Tamoxifen
cyclosporine A strongly enhanced the oral bioavailability of
docetaxel. These data may form the basis for the further de- Jin et al. [95] demonstrated the effects of selective sero-
velopment of a clinically useful oral formulation of do- tonin reuptake inhibitors (SSRIs) that were also inhibitors of
cetaxel. CYP2D6 on the plasma level of endoxifen. SSRIs are com-
monly prescribed to treat hot flashes in breast cancer patients
Imatinib who take tamoxifen. The concomitant administration of
SSRIs such as paroxetine substantially reduced the plasma
Pharmacokinetic interactions of imatinib with substrates concentration of endoxifen, suggesting a potential alteration
and inhibitors of CYP3A4 have been studied. O’Brien et al.
in the expression of tamoxifen activity.
[154] have evaluated the effect of the co-administration of
imatinib on the pharmacokinetics of simvastatin, a probe
Vincristine
CYP3A4 substrate. In total, 20 patients with chronic myeloid
leukemia received an oral dose of 40 mg simvastatin on Although vinca alkaloids show similarity in their chemi-
study day 1. On study days 2-7, each patient received 400 cal structures, they exhibit marked differences in their re-
Cytochrome P450 and Anticancer Drugs Current Drug Metabolism, 2006, Vol. 7, No. 1 33

spective antitumor spectra, toxicity and pharmacokinetics been developed in animal models. A CYP2B6 gene coding
[161-163]. Moreover, their clinical pharmacokinetics shows for the enzyme that is responsible for the activation of cyclo-
large intra- and/or inter-individual variations. Since these phosphamide has been transferred into tumor cells with a
agents are metabolized by CYP3A, it may be possible that retrovirus and replication-conditional adenovirus to enhance
the variability of CYP3A activity is partly responsible for the the intratumoral activation of prodrugs [174, 175].
variation in the pharmacokinetic profiles of vinca alkaloids.
The effects of the CYP3A4-inducing antiepileptic agents, PERSPECTIVES
carbamazepine and phenytoin, on the pharmacokinetics of Generally, for select drugs whose pharmacokinetic/phar-
vincristine have been studied [164]. Fifteen adult patients macodynamic relationships are known, feedback control of
with brain tumors receiving combination chemotherapy with plasma concentrations, through therapeutic drug monitoring
procarbazine, lomustine, and vincristine volunteered for this (TDM), may be invoked to minimize the variability in their
open parallel-group study. Nine of the patients used either therapeutic and toxic effects. However, for cancer che-
carbamazepine or phenytoin and six of the patients used no motherapeutic agents, TDM cannot be readily used to reduce
obvious CYP3A4-inducing medication. The results revealed pharmacokinetic/pharmacodynamic variations because of the
that drugs inducing CYP3A4 could increase the elimination following reasons:
of vincristine. Further studies are needed to determine
whether the increased clearance of vincristine by carba- § The pharmacokinetic/pharmacodynamic relationships are
not well known;
mazepine or phenytoin decreases the efficacy of vincristine.
§ The threshold of the antitumor effects is usually obscure
Local Expression of CYPs in Tumor Tissues and dependent on the patient factors, which may not be
well quantified;
Recently, a number of cancer tissues have been found to
express a variety of CYPs [165-172]. The local expression of § There is a long time lag between the treatment of patients
CYPs in tumor tissues appears to be important for tumor with anticancer drugs and the appearance of the efficacy
management, since CYPs expressed in tumors may be re- of the drugs (a large time constant);
sponsible for the localized inactivation and/or activation of
§ It is difficult to determine the target drug to be monitored
anticancer drugs. Inactivation of anticancer drugs by CYPs
because most cancer chemotherapeutics is based on in-
in tumor tissues may result in the resistance of the tumor
tentional polypharmacy; of the combination use of anti-
toward the drugs, while the activation of prodrugs by CYPs
cancer drugs;
may lead to the enhancement of effectiveness in cancer che-
motherapy. As shown in Table 1, CYP2A, CYP2B, CYP2C, § Dose adjustments may be difficult to make in chemother-
CYP2D and CYP3A are mainly responsible for the metabo- apy once dose-limiting toxicity thresholds have been
lism of anticancer drugs. These forms of CYP have been reached.
reported to be expressed in tumor tissues. CYP2A6 was
found to be expressed in breast cancer tissue obtained from a Therefore, the optimal dose of anticancer drugs should,
Japanese patient (among 34 patients) [165]. CYP2B6 was when possible, be established prior to drug administration.
demonstrated to be present in breast tumor tissues [166, Since CYP forms are responsible for the metabolism of a
167]. CYP2C enzymes were expressed in 33% of breast can- wide variety of anticancer drugs and these enzymes show a
cer tissues and 25% of prostate cancer tissues [168, 169]. vast range of variability in their activities due to genetic and
The expression of CYP2D6 in breast tumors has been re- nongenetic factors, CYP activity should be a good biomarker
ported by Iscan et al. [167]. CYP3A was found in 22% of for establishing individualized doses of anticancer drugs.
breast cancer by immunohistochemistry [170], however, the This is a feedforward concept for the determination of indi-
enzyme was not detected in 34 specimens from Japanese vidualized doses of anticancer drugs. Tests of this new con-
patients [165]. Murray et al. [171] have shown that CYP3A cept for initializing/optimizing chemotherapy are now in
was expressed in at least 60% of oesophageal carcinomas. progress.
CYP3A has been also reported to be expressed in colon car-
cinomas and prostate cancer [169, 172]. ABBREVIATIONS

It seems likely that there is a large interindividual vari- AUC = Area under the concentration-time curve
ability in CYP expression in tumor tissues. This may also BSA = Body surface area
contribute to the interindividual variability in the response to
chemotherapy. Indeed, Miyoshi et al. [173] demonstrated Cmax = Maximum concentration
that intratumoral expression of CYP3A4 mRNA was signifi- CYP = Cytochrome P450
cantly lower (about 4 fold) in responders compared with
nonresponders in docetaxel treatment. Further studies are ERMBT = Erythromycin breath test
needed to clarify the roles of CYPs expressed in tumor tis- 5-FU = 5-Fluorouracil
sues in cancer chemotherapy.
MTD = Maximum-tolerated dose
One therapeutically attractive strategy to enhance the
intratumoral activation of a prodrug is based on the direct SSRI = Selective serotonin reuptake inhibitor
transfer to the tumor of a gene which encodes an enzyme that
can activate the prodrug, and this gene therapy strategy has TK = Tyrosine kinase
34 Current Drug Metabolism, 2006, Vol. 7, No. 1 Ken-ichi Fujita

REFERENCES deville, R.; Zoellner, U.; Talpaz, M.; Druker, B.; Goldman, J.;
O'Brien, S.G.; Russell, N.; Fischer, T.; Ottmann, O.; Cony-
[1] Wilkinson, G.R. (2005) N. Engl. J. Med., 352(21), 2211-2221. Makhoul, P.; Facon, T.; Stone, R.; Miller, C.; Tallman, M.; Brown,
[2] Rochat, B. (2005) Clin. Pharmacokinet., 44(4), 349-366. R.; Schuster, M.; Loughran, T.; Gratwohl, A.; Mandelli, F.; Saglio,
[3] Nelson, D.R.; Koymans, L.; Kamataki, T.; Stegeman, J.J.; Feyere- G.; Lazzarino, M.; Russo, D.; Baccarani, M.; Morram, E. and In-
isen, R.; Waxman, D.J.; Waterman, M.R.; Gotoh, O.; Coon, M.J.; ternational STI571 CML Study Group. (2002) N. Engl. J. Med.,
Estabrook, R.W.; Gunsalus, I.C. and Nebert, D.W. (1996) Pharma- 346(9), 645-652.
cogenetics, 6(1), 1-42. [32] O'Brien, S.G.; Guilhot, F.; Larson, R.A.; Gathmann, I.; Baccarani,
[4] Evans, W.E. and Relling, M.V. (2004) Nature, 429(6990), 464- M.; Cervantes, F.; Cornelissen, J.J.; Fischer, T.; Hochhaus, A.;
468. Hughes, T.; Lechner, K.; Nielsen, J.L.; Rousselot, P.; Reiffers, J.;
[5] Evans, W.E. and McLeod, H.L. (2003) N. Engl. J. Med., 348(6), Saglio, G.; Shepherd, J.; Simonsson, B.; Gratwohl, A.; Goldman,
538-549. J.M.; Kantarjian, H.; Taylor, K.; Verhoef, G.; Bolton, A.E.;
[6] Cresteil, T.; Monsarrat, B.; Alvinerie, P.; Treluyer, J.M.; Vieira, I. Capdeville, R.; Druker, B.J. and IRIS Investigators. (2003) N. Engl.
and Wright, M. (1994) Cancer Res., 54(2), 386-392. J. Med., 348(11), 994-1004.
[7] Monsarrat, B.; Mariel, E.; Cros, S.; Gares, M.; Guenard, D.; Gue- [33] Joensuu, H.; Roberts, P.J.; Sarlomo-Rikala, M.; Andersson, L.C.;
ritte-Voegelein, F. and Wright, M. (1990) Drug Metab. Dispos., Tervahartiala, P.; Tuveson, D.; Silberman, S.; Capdeville, R.;
18(6), 895-901. Dimitrijevic, S.; Druker, B. and Demetri, G.D. (2001) N. Engl. J.
[8] Eddershaw, P.J. and Dickins, M. (1999) Pharm. Sci. Technol. To- Med., 344(14), 1052-1056.
day, 2(1), 13-19. [34] Cohen, M.H.; Williams, G.A.; Sridhara, R.; Chen, G.; McGuinn,
[9] Ekins, S.; Ring, B.J.; Grace, J.; McRobie-Belle, D.J. and Wrighton, W.D. Jr; Morse, D.; Abraham, S.; Rahman, A.; Liang, C.; Lostritto,
S.A. (2000) J. Pharmacol. Toxicol. Methods, 44(1), 313-324. R.; Baird, A. and Pazdur, R. (2004) Clin. Cancer Res., 10(4), 1212-
[10] LeCluyse, E.L. (2001) Eur. J. Pharm. Sci., 13(4), 343-368. 1218.
[11] Li, A.P.; Gorycki, P.D.; Hengstler, J.G.; Kedderis, G.L.; Koebe, [35] McKillop, D.; McCormick, A.D.; Millar, A.; Miles, G.S.; Phillips,
H.G.; Rahmani, R.; de Sousas, G.; Silva, J.M. and Skett, P. (1999) P.J. and Hutchison, M. (2005) Xenobiotica, 35(1), 39-50.
Chem. Biol. Interact., 121(1), 117-123. [36] Sladek, N.E. (1994) In Anticancer Drugs: Reactive Metabolism
[12] Rodrigues, A.D. (1994) Biochem. Pharmacol., 48(12), 2147-2156. and Drug Interactions (Powis, G. ed.), Pergamon Press, New York,
[13] Streetman, D.S.; Bertino, J.S. and Nafziger, A.N. (2000) Pharma- pp. 79-156.
cogenetics, 10(3), 187-216. [37] Colvin, M.; Padgett, C.A. and Fenselau, C. (1973) Cancer Res.,
[14] Venkatakrishnan, K.; von Moltke, L.L.; Obach, R.S. and Green- 33(4), 915-918.
blatt, D.J. (2003) Curr. Drug Metab., 4(5), 423-459. [38] Connors, T.A.; Cox, P.J.; Farmer, P.B.; Foster, A.B. and Jarman,
[15] Venkatakrishnan, K.; von Moltke, L.L. and Greenblatt, D.J. (2001) M. (1974) Biochem. Pharmacol., 23(1), 115-129.
J. Clin. Pharmacol., 41(11), 1149-1179. [39] Struck, R.F.; Kirk, M.C.; Witt, M.H. and Laster, W.R. Jr. (1975)
[16] Iwata, H.; Fujita, K.; Kushida, H.; Suzuki, A.; Konno, Y.; Naka- Biomed. Mass. Spectrom., 2(1), 46-52.
mura, K.; Fujino, A. and Kamataki, T. (1998) Biochem. Pharma- [40] Walker, D.; Flinois, J.P.; Monkman, S.C.; Beloc, C.; Boddy, A.V.;
col., 55(8), 1315-1325. Cholerton, S.; Daly, A.K.; Lind, M.J.; Pearson, A.D. and Beaune,
[17] Fujita, K.; Nakayama, K.; Yamazaki, Y.; Tsuruma, K.; Yamada, P.H. (1994) Biochem. Pharmacol., 47(7), 1157-1163.
M.; Nohmi, T. and Kamataki, T. (2001) Environ. Mol. Mutagen., [41] Chang, T.K.; Weber, G.F.; Crespi, C.L. and Waxman, D.J. (1993)
38(4), 329-338. Cancer Res., 53(23), 5629-5637.
[18] Fujita, K. and Kamataki, T. (2002) Drug Metab. Pharmacokin., [42] Ren, S.; Yang, J.S.; Kalhorn, T.F. and Slattery, J.T. (1997) Cancer
17(1), 1-22. Res., 57(19), 4229-4235.
[19] Stahelin, H.F. and von Wartburg, A. (1991) Cancer Res., 51(1), 5- [43] Huang, Z.; Roy, P. and Waxman, D.J. (2000) Biochem. Pharma-
15. col., 59(8), 961-972.
[20] Clark, P.I. and Slevin, M.L. (1987) Clin. Pharmacokinet., 12(4), [44] Kaijser, G.P.; Korst, A.; Beijnen, J.H.; Bult, A. and Underberg,
223-252. W.J. (1993) Anticancer Res., 13(5A), 1311-1324.
[21] Relling, M.V.; Nemec, J.; Schuetz, E.G.; Schuetz, J.D.; Gonzalez, [45] Fleming, R.A. (1997) Pharmacotherapy, 17(5 Pt 2), 146S-154S.
F.J. and Korzekwa, K.R. (1994) Mol. Pharmacol., 45(2), 352-358. [46] Roy, P.; Tretyakov, O.; Wright, J. and Waxman, D.J. (1999) Drug
[22] Kawashiro, T.; Yamashita, K.; Zhao, X.J.; Koyama, E.; Tani, M.; Metab. Dispos., 27(11), 1309-1318.
Chiba, K. and Ishizaki, T. (1998) J. Pharmacol. Exp. Ther., 286(3), [47] Osborne, C.K. (1998) N. Engl. J. Med., 339(22), 1609-1618.
1294-1300. [48] Fisher, B.; Costantino, J.P.; Wickerham, D.L.; Redmond, C.K.;
[23] Zhao, X.J.; Kawashiro, T. and Ishizaki, T. (1998) Drug Metab. Kavanah, M.; Cronin, W.M.; Vogel, V.; Robidoux, A.; Dimitrov,
Dispos., 26(2), 188-191. N.; Atkins, J.; Daly, M.; Wieand, S.; Tan-Chiu, E.; Ford, L. and
[24] Buchdunger, E.; Zimmermann, J.; Mett, H.; Meyer, T.; Muller, M.; Wolmark, N. (1998) J. Natl. Cancer Inst., 90(18), 1371-1388.
Druker, B.J. and Lydon, N.B. (1996) Cancer Res., 56(1), 100-104. [49] Jordan, V.C.; Collins, M.M.; Rowsby, L. and Prestwich, G. (1977)
[25] Druker, B.J.; Tamura, S.; Buchdunger, E.; Ohno, S.; Segal, G.M.; J. Endocrinol., 75(2), 305-316.
Fanning, S.; Zimmermann, J. and Lydon N.B. (1996) Nat. Med., [50] Fabian, C.; Tilzer, L. and Sternson, L. (1981) Biopharm. Drug.
2(5), 561-566. Dispos., 2(4), 381-390.
[26] Buchdunger, E.; Cioffi, C.L.; Law, N.; Stover, D.; Ohno-Jones, S.; [51] Stearns, V.; Johnson, M.D.; Rae, J.M.; Morocho, A.; Novielli, A.;
Druker, B.J. and Lydon, N.B. (2000) J. Pharmacol. Exp. Ther., Bhargava, P.; Hayes, D.F.; Desta, Z. and Flockhart, D.A. (2003) J.
295(1), 139-145. Natl. Cancer Inst., 95(23), 1758-1764.
[27] Heinrich, M.C.; Griffith, D.J.; Druker, B.J.; Wait, C.L.; Ott, K.A. [52] Crewe, H.K.; Ellis, S.W.; Lennard, M.S. and Tucker, G.T. (1997)
and Zigler, A.J. (2000) Blood, 96(3), 925-932. Biochem. Pharmacol., 53(2), 171-178.
[28] Druker, B.J.; Sawyers, C.L.; Kantarjian, H.; Resta, D.J.; Reese, [53] Desta, Z.; Ward, B.A.; Soukhova, N.V. and Flockhart, D.A. (2004)
S.F.; Ford, J.M.; Capdeville, R. and Talpaz, M. (2001) N. Engl. J. J. Pharmacol. Exp. Ther., 310(3), 1062-1075.
Med., 344(14), 1038-1042. [54] Swerdlow, A.J.; Jones, M.E. and British Tamoxifen Second Cancer
[29] Druker, B.J.; Talpaz, M.; Resta, D.J.; Peng, B.; Buchdunger, E.; Study Group. (2005) J. Natl. Cancer Inst., 97(5), 375-384.
Ford, J.M.; Lydon, N.B.; Kantarjian, H.; Capdeville, R.; Ohno- [55] Shibutani, S.; Dasaradhi, L.; Terashima, I.; Banoglu, E. and Duffel,
Jones, S. and Sawyers, C.L. (2001) N. Engl. J. Med., 344(14), M.W. (1998) Cancer Res., 58(4), 647-653.
1031-1037. [56] Shibutani, S.; Ravindernath, A.; Suzuki, N.; Terashima, I.;
[30] Talpaz, M.; Silver, R.T.; Druker, B.J.; Goldman, J.M.; Gambacorti- Sugarman, S.M.; Grollman, A.P. and Pearl, M.L. (2000) Carcino-
Passerini, C.; Guilhot, F.; Schiffer, C.A.; Fischer, T.; Deininger, genesis, 21(8), 1461-1467.
M.W.; Lennard, A.L.; Hochhaus, A.; Ottmann, O.G.; Gratwohl, A.; [57] Kim, S.Y.; Suzuki, N.; Santosh, Laxmi, Y.R.; Rieger, R. and Shi-
Baccarani, M.; Stone, R.; Tura, S.; Mahon, F.X.; Fernandes-Reese, butani, S. (2003) Chem. Res. Toxicol., 16(9), 1138-1144.
S.; Gathmann, I.; Capdeville, R.; Kantarjian, H.M. and Sawyers, [58] Kim, S.Y.; Suzuki, N.; Laxmi, Y.R.; McGarrigle, B.P.; Olson, J.R.;
C.L. (2002) Blood, 99(6), 1928-1937. Sharma, M.; Sharma, M. and Shibutani, S. (2005) Chem. Res. Toxi-
[31] Kantarjian, H.; Sawyers, C.; Hochhaus, A.; Guilhot, F.; Schiffer, col., 18(5), 889-895.
C.; Gambacorti-Passerini, C.; Niederwieser, D.; Resta, D.; Cap- [59] Rowinsky, E.K. (1997) Annu. Rev. Med., 48, 353-374.
Cytochrome P450 and Anticancer Drugs Current Drug Metabolism, 2006, Vol. 7, No. 1 35

[60] Monsarrat, B.; Alvinerie, P.; Wright, M.; Dubois, J.; Gueritte- [89] Fukami, T.; Nakajima, M.; Higashi, E.; Yamanaka, H.; McLeod,
Voegelein, F.; Guenard, D.; Donehower, R.C. and Rowinsky, E.K. H.L. and Yokoi, T. (2005) Biochem. Pharmacol., 70(5), 801-808.
(1993) J. Natl. Cancer Inst. Monogr., 15, 39-46. [90] Yoshida, R.; Nakajima, M.; Watanabe, Y.; Kwon, J.T. and Yokoi,
[61] Monsarrat, B.; Wright, M.; Dubois, J.; Gueritte-Voegelein, F.; T. (2002) Br. J. Clin. Pharmacol., 54(5), 511-517.
Guenard, D.; Rowinsky, E.K. and Donehower, R.C. (1992) Ann. [91] Oscarson, M.; McLellan, R.A.; Gullsten, H.; Agundez, J.A.;
Oncol., 3(Suppl. 1), 123. Benitez, J.; Rautio, A.; Raunio, H.; Pelkonen, O. and Ingelman-
[62] Rahman, A.; Korzekwa, K.R.; Grogan, J.; Gonzalez, F.J. and Har- Sundberg, M. (1999) FEBS Lett., 460(2), 321-327.
ris, J.W. (1994) Cancer Res., 54(21), 5543-5546. [92] Ariyoshi, N.; Takahashi, Y.; Miyamoto, M.; Umetsu, Y.; Daigo, S.;
[63] Royer, I.; Monsarrat, B.; Sonnier, M.; Wright, M. and Cresteil, T. Tateishi, T.; Kobayashi, S.; Mizorogi, Y.; Loriot, M.A.; Stucker, I.;
(1996) Cancer Res., 56(1), 58-65. Beaune, P.; Kinoshita, M. and Kamataki, T. (2000) Pharmacoge-
[64] Richardson, P.; Hideshima, T. and Anderson, K. (2002) Annu. Rev. netics, 10(8), 687-693.
Med., 53, 629-657. [93] Ariyoshi, N.; Sekine, H.; Nakayama, K.; Saito, K.; Miyamoto, A.
[65] D'Amato, R.J.; Loughnan, M.S.; Flynn, E. and Folkman, J. (1994) and Kamataki, T. (2004) Pharmacogenetics, 14(10), 701-705.
Proc. Natl. Acad. Sci. USA, 91(9), 4082-4085. [94] Caraco, Y. (2004) N. Engl. J. Med., 351(27), 2867-2869.
[66] Figg, W.D.; Dahut, W.; Duray, P.; Hamilton, M.; Tompkins, A.; [95] Jin, Y.; Desta, Z.; Stearns, V.; Ward, B.; Ho, H.; Lee, K.H.; Skaar,
Steinberg, S.M.; Jones, E.; Premkumar, A.; Linehan, W.M.; T.; Storniolo, A.M.; Li, L.; Araba, A.; Blanchard, R.; Nguyen, A.;
Floeter, M.K.; Chen, C.C.; Dixon, S.; Kohler, D.R.; Kruger, E.A.; Ullmer, L.; Hayden, J.; Lemler, S.; Weinshilboum, R.M.; Rae,
Gubish, E.; Pluda, J.M. and Reed, E. (2001) Clin. Cancer Res., J.M.; Hayes, D.F. and Flockhart, D.A. (2005) J. Natl. Cancer Inst. ,
7(7), 1888-1893. 97(1), 30-39.
[67] Singhal, S.; Mehta, J.; Desikan, R.; Ayers, D.; Roberson, P.; Ed- [96] Sachse, C.; Brockmoller, J.; Bauer, S. and Roots, I. (1997) Am. J.
dlemon, P.; Munshi, N.; Anaissie, E.; Wilson, C.; Dhodapkar, M.; Hum. Genet., 60(2), 284-295.
Zeddis, J. and Barlogie, B. (1999) N. Engl. J. Med., 341(21), 1565- [97] Marez, D.; Legrand, M.; Sabbagh, N.; Guidice, J.M.; Spire, C.;
1571. Lafitte, J.J.; Meyer, U.A. and Broly, F. (1997) Pharmacogenetics,
[68] Braun, A.G.; Harding, F.A. and Weinreb, S.L. (1986) Toxicol. 7(3), 193-202.
Appl. Pharmacol., 82(1), 175-179. [98] Kubota, T.; Yamaura, Y.; Ohkawa, N.; Hara, H. and Chiba, K.
[69] Bauer, K.S.; Dixon, S.C. and Figg, W.D. (1998) Biochem. (2000) Br. J. Clin. Pharmacol., 50(1), 31-34.
Pharmacol., 55(11), 1827-1834. [99] McLellan, R.A.; Oscarson, M.; Seidegard, J.; Evans, D.A. and
[70] Braun, A.G. and Weinreb, S.L. (1985) Teratog. Carcinog. Ingelman-Sundberg, M. (1997) Pharmacogenetics, 7(3), 187-191.
Mutagen., 5(3), 149-158. [100] Gaikovitch, E.A.; Cascorbi, I.; Mrozikiewicz, P.M.; Brockmoller,
[71] Eriksson, T.; Bjorkman, S.; Roth, B.; Bjork, H. and Hoglund, P. J.; Frotschl, R.; Kopke, K.; Gerloff, T.; Chernov, J.N. and Roots, I.
(1998) J. Pharm. Pharmacol., 50(12), 1409-1416. (2003) Eur. J. Clin. Pharmacol., 59(4), 303-312.
[72] Teo, S.K.; Sabourin, P.J.; O'Brien, K.; Kook, K.A. and Thomas, [101] Tateishi, T.; Chida, M.; Ariyoshi, N.; Mizorogi, Y.; Kamataki, T.
S.D. (2000) J. Biochem. Mol. Toxicol., 14(3), 140-147. and Kobayashi, S. (1999) Clin. Pharmacol. Ther., 65(5), 570-575.
[73] Ando, Y.; Fuse, E. and Figg, W.D. (2002) Clin. Cancer Res., 8(6), [102] Nishida, Y.; Fukuda, T.; Yamamoto, I. and Azuma, J. (2000)
1964-1973. Pharmacogenetics, 10(6), 567-570.
[74] Ikeda, K.; Yoshisue, K.; Matsushima, E.; Nagayama, S.; Kobaya- [103] Johansson, I.; Oscarson, M.; Yue, Q.Y.; Bertilsson, L.; Sjoqvist, F.
shi, K.; Tyson, C.A.; Chiba, K. and Kawaguchi, Y. (2000) Clin. and Ingelman-Sundberg, M. (1994) Mol. Pharmacol., 46(3), 452-
Cancer Res., 6(11), 4409-4415. 459.
[75] Zhou-Pan, X.R.; Seree, E.; Zhou, X.J.; Placidi, M.; Maurel, P.; [104] Aklillu, E.; Persson, I.; Bertilsson, L.; Johansson, I.; Rodrigues, F.
Barra, Y. and Rahmani, R. (1993) Cancer Res., 53(21), 5121-5126. and Ingelman-Sundberg, M. (1996) J. Pharmacol. Exp. Ther.,
[76] Zhou, X.J.; Zhou-Pan, X.R.; Gauthier, T.; Placidi, M.; Maurel, P. 278(1), 441-446.
and Rahmani, R. (1993) Biochem. Pharmacol., 45(4), 853-861. [105] de Wildt, S.N.; Kearns, G.L.; Leeder, J.S. and van den Anker, J.N.
[77] Kajita, J.; Kuwabara, T.; Kobayashi, H. and Kobayashi, S. (2000) (1999) Clin. Pharmacokinet., 37(6), 485-505.
Drug Metab. Dispos., 28(9), 1121-1127. [106] Koch, I.; Weil, R.; Wolbold, R.; Brockmoller, J.; Hustert, E.; Burk,
[78] Baker, S.D.; Verweij, J.; Rowinsky, E.K.; Donehower, R.C.; O.; Nuessler, A.; Neuhaus, P.; Eichelbaum, M.; Zanger, U. and
Schellens, J.H.; Grochow, L.B. and Sparreboom, A. (2002) J. Natl. Wojnowski, L. (2002) Drug Metab. Dispos., 30(10), 1108-1114.
Cancer Inst., 94(24), 1883-1888. [107] Williams, J.A.; Ring, B.J.; Cantrell, V.E.; Jones, D.R.; Eckstein, J.;
[79] Ando, Y.; Ohtsu, T.; Ando, M.; Ohyanagi, F.; Nagashima, F.; Na- Ruterbories, K.; Hamman, M.A.; Hall, S.D. and Wrighton, S.A.
rabayashi, M.; Saijo, N. and Sasaki, Y. (2003) J. Natl. Cancer Inst., (2002) Drug Metab. Dispos., 30(8), 883-891.
95(21), 1638-1640. [108] Lamba, J.K.; Lin, Y.S.; Schuetz, E.G. and Thummel, K.E. (2002)
[80] Phillips, K.A.; Veenstra, D.L.; Oren, E.; Lee, J.K. and Sadee, W. Adv. Drug Deliv. Rev., 54(10), 1271-1294.
(2001) JAMA, 286(18), 2270-2279. [109] Amirimani, B.; Walker, A.H.; Weber, B.L. and Rebbeck, T.R.
[81] Daigo, S.; Takahashi, Y.; Fujieda, M.; Ariyoshi, N.; Yamazaki, H.; (1999) J. Natl. Cancer Inst., 91(18), 1588-1590.
Koizumi, W.; Tanabe, S.; Saigenji, K.; Nagayama, S.; Ikeda, K.; [110] Ando, Y.; Tateishi, T.; Sekido, Y.; Yamamoto, T.; Satoh, T.; Hase-
Nishioka, Y. and Kamataki T. (2002) Pharmacogenetics, 12(4), gawa, Y.; Kobayashi, S.; Katsumata, Y.; Shimokata, K. and Saito,
299-306. H. (1999) J. Natl. Cancer Inst., 91(18), 1587-1590.
[82] Nunoya, K.; Yokoi, T.; Kimura, K.; Inoue, K.; Kodama, T.; Fu- [111] Rebbeck, T.R.; Jaffe, J.M.; Walker, A.H.; Wein, A.J. and Malk-
nayama, M.; Nagashima, K.; Funae, Y.; Green, C.; Kinoshita, M. owicz, S.B. (1998) J. Natl. Cancer Inst., 90(16), 1225-1229.
and Kamataki, T. (1998) Pharmacogenetics, 8(3), 239-249. [112] Felix, C.A.; Walker, A.H.; Lange, B.J.; Williams, T.M.; Winick,
[83] Komatsu, T.; Yamazaki, H.; Shimada, N.; Nagayama, S.; Kawa- N.J.; Cheung, N.K.; Lovett, B.D.; Nowell, P.C.; Blair, I.A. and
guchi, Y.; Nakajima, M. and Yokoi, T. (2001) Clin. Cancer Res., Rebbeck, T.R. (1998) Proc. Natl. Acad. Sci. USA, 95(22), 13176-
7(3), 675-681. 13181.
[84] Nakajima, M.; Yoshida, R.; Fukami, T.; McLeod, H.L. and Yokoi, [113] Wandel, C.; Witte, J.S.; Hall, J.M.; Stein, C.M.; Wood, A.J. and
T. (2004) FEBS Lett., 569(1-3), 75-81. Wilkinson, G.R. (2000) Clin. Pharmacol. Ther., 68(1), 82-91.
[85] Schoedel, K.A.; Hoffmann, E.B.; Rao, Y.; Sellers, E.M. and [114] Floyd, M.D.; Gervasini, G.; Masica, A.L.; Mayo, G.; George, A.L.
Tyndale, R.F. (2004) Pharmacogenetics, 14(9), 615-626. Jr.; Bhat, K.; Kim, R.B. and Wilkinson, G.R. (2003) Pharmacoge-
[86] Kiyotani, K.; Fujieda, M.; Yamazaki, H.; Shimada, T.; Guengerich, netics, 13(10), 595-606.
F.P.; Parkinson, A.; Nakagawa, K.; Ishizaki, T. and Kamataki, T. [115] Eap, C.B.; Buclin, T.; Hustert, E.; Bleiber, G.; Golay, K.P.; Aubert,
(2002) Drug Metab. Pharmacokinet., 17(5), 482-487. A.C.; Baumann, P.; Telenti, A. and Kerb, R. (2004) Eur. J. Clin.
[87] Fukami, T.; Nakajima, M.; Higashi, E.; Yamanaka, H.; Sakai, H.; Pharmacol., 60(4), 231-236.
McLeod, H.L. and Yokoi, T. (2005) Drug Metab. Dispos., 33(8), [116] Baker, S.D.; van Schaik, R.H.; Rivory, L.P.; Ten Tije, A.J.; Dinh,
1202-1210. K.; Graveland, W.J.; Schenk, P.W.; Charles, K.A.; Clarke, S.J.;
[88] Fukami, T.; Nakajima, M.; Yoshida, R.; Tsuchiya, Y.; Fujiki, Y.; Carducci, M.A.; McGuire, W.P.; Dawkins, F.; Gelderblom, H.;
Katoh, M.; McLeod, H.L. and Yokoi, T. (2004) Clin. Pharmacol. Verweij, J. and Sparreboom, A. (2004) Clin. Cancer Res., 10(24),
Ther., 76(6), 519-527. 8341-8350.
36 Current Drug Metabolism, 2006, Vol. 7, No. 1 Ken-ichi Fujita

[117] Ball, S.E.; Scatina, J.; Kao, J.; Ferron, G.M.; Fruncillo, R.; Mayer, [143] Yamamoto, N.; Tamura, T.; Kamiya, Y.; Sekine, I.; Kunitoh, H.
P.; Weinryb, I.; Guida, M.; Hopkins, P.J.; Warner, N. and Hall, J. and Saijo, N. (2000) J. Clin. Oncol., 18(11), 2301-2308.
(1999) Clin. Pharmacol. Ther., 66(3), 288-294. [144] Yamamoto, N.; Tamura, T.; Murakami, H.; Shimoyama, T.; Noki-
[118] Dai, D.; Tang, J.; Rose, R.; Hodgson, E.; Bienstock, R.J.; Mohren- hara, H.; Ueda, Y.; Sekine, I.; Kunitoh, H.; Ohe, Y.; Kodam, T.;
weiser, H.W. and Goldstein, J.A. (2001) J. Pharmacol. Exp. Ther., Shimizu, M.; Nishio, K.; Ishizuka, N. and Saijo, N. (2005) J. Clin.
299(3), 825-831. Oncol., 23(6), 1061-1069.
[119] Hsieh, K.P.; Lin, Y.Y.; Cheng, C.L.; Lai, M.L.; Lin, M.S.; Siest, [145] Goh, B.C.; Lee, S.C.; Wang, L.Z.; Fan, L.; Guo, J.Y.; Lamba, J.;
J.P. and Huang, J.D. (2001) Drug Metab. Dispos., 29(3), 268-273. Schuetz, E.; Lim, R.; Lim, H.L.; Ong, A.B. and Lee, H.S. (2002) J.
[120] Lamba, J.K.; Lin, Y.S.; Thummel, K.; Daly, A.; Watkins, P.B.; Clin. Oncol., 20(17), 3683-3690.
Strom, S.; Zhang, J. and Schuetz, E.G. (2002) Pharmacogenetics, [146] Rougier, P.; Van Cutsem, E.; Bajetta, E.; Niederle, N.; Possinger,
12(2), 121-132. K.; Labianca, R.; Navarro, M.; Morant, R.; Bleiberg, H.; Wils, J.;
[121] Eiselt, R.; Domanski, T.L.; Zibat, A.; Mueller, R.; Presecan-Siedel, Awad, L.; Herait, P. and Jacques, C. (1998) Lancet, 352(9138),
E.; Hustert, E.; Zanger, U.M.; Brockmoller, J.; Klenk, H.P.; Meyer, 1407-1412.
U.A.; Khan, K.K.; He, Y.A.; Halpert, J.R. and Wojnowski, L. [147] Kudoh, S.; Fujiwara, Y.; Takada, Y.; Yamamoto, H.; Kinoshita, A.;
(2001) Pharmacogenetics, 11(5), 447-458. Ariyoshi, Y.; Furuse, K. and Fukuoka, M. (1998) J. Clin. Oncol.,
[122] van Schaik, R.H.; de Wildt, S.N.; Brosens, R.; van Fessem, M.; van 16(3), 1068-1074.
den Anker, J.N. and Lindemans, J. (2001) Clin. Chem., 47(6), [148] Negoro, S.; Fukuoka, M.; Masuda, N.; Takada, M.; Kusunoki, Y.;
1104-1106. Matsui, K.; Takifuji, N.; Kudoh, S.; Niitani, H. and Taguchi, T.
[123] Ball, S.E.; Scatina, J.; Kao, J.; Ferron, G.M.; Fruncillo, R.; Mayer, (1991) J. Natl. Cancer Inst., 83(16), 1164-1168.
P.; Weinryb, I.; Guida, M.; Hopkins, P.J.; Warner, N. and Hall, J. [149] Mathijssen, R.H.; van Alphen, R.J.; Verweij, J.; Loos, W.J.;
(1999) Clin. Pharmacol. Ther., 66(3), 288-294. Nooter, K.; Stoter, G. and Sparreboom, A. (2001) Clin. Cancer
[124] Sata, F.; Sapone, A.; Elizondo, G.; Stocker, P.; Miller, V.P.; Zheng, Res., 7(8), 2182-2194.
W.; Raunio, H.; Crespi, C.L. and Gonzalez, F.J. (2000) Clin. [150] Santos, A.; Zanetta, S.; Cresteil, T.; Deroussent, A.; Pein, F.;
Pharmacol. Ther., 67(1), 48-56. Raymond, E.; Vernillet, L.; Risse, M.L.; Boige, V.; Gouyette, A.
[125] Walker, A.H.; Jaffe, J.M.; Gunasegaram, S.; Cummings, S.A.; and Vassal, G. (2000) Clin. Cancer Res., 6(5), 2012-2020.
Huang, C.S.; Chern, H.D.; Olopade, O.I.; Weber, B.L. and [151] Mathijssen, R.H.; de Jong, F.A.; van Schaik, R.H.; Lepper, E.R.;
Rebbeck, T.R. (1998) Hum. Mutat., 12(4), 289. Friberg, L.E.; Rietveld, T.; de Bruijn, P.; Graveland, W.J.; Figg,
[126] Fukushima-Uesaka, H.; Saito, Y.; Watanabe, H.; Shiseki, K.; W.D.; Verweij, J. and Sparreboom, A. (2004) J. Natl. Cancer Inst. ,
Saeki, M.; Nakamura, T.; Kurose, K.; Sai, K.; Komamura, K.; 96(21), 1585-1592.
Ueno, K.; Kamakura, S.; Kitakaze, M.; Hanai, S.; Nakajima, T.; [152] Honig, P.K.; Wortham, D.C.; Zamani, K.; Conner, D.P.; Mullin,
Matsumoto, K.; Saito, H.; Goto, Y.; Kimura, H.; Katoh, M.; Sugai, J.C. and Cantilena, L.R. (1993) JAMA, 269(12), 1513-1518.
K.; Minami, N.; Shirao, K.; Tamura, T.; Yamamoto, N.; Minami, [153] Malingre, M.M.; Richel, D.J.; Beijnen, J.H.; Rosing, H.; Koopman,
H.; Ohtsu, A.; Yoshida, T.; Saijo, N.; Kitamura, Y.; Kamatani, N.; F.J.; Ten Bokkel Huinink, W.W.; Schot, M.E. and Schellens, J.H.
Ozawa, S. and Sawada, J. (2004) Hum. Mutat., 23(1), 100. (2001) J. Clin. Oncol., 19(4), 1160-1166.
[127] Kuehl, P.; Zhang, J.; Lin, Y.; Lamba, J.; Assem, M.; Schuetz, J.; [154] O'Brien, S.G.; Meinhardt, P.; Bond, E.; Beck, J.; Peng, B.; Dutreix,
Watkins, P.B.; Daly, A.; Wrighton, S.A.; Hall, S.D.; Maurel, P.; C.; Mehring, G.; Milosavljev, S.; Huber, C.; Capdeville, R. and
Relling, M.; Brimer, C.; Yasuda, K.; Venkataramanan, R.; Strom, Fischer, T. (2003) Br. J. Cancer, 89(10), 1855-1859.
S.; Thummel, K.; Boguski, M.S. and Schuetz, E. (2001) Nat. [155] Dutreix, C.; Peng, B.; Mehring, G.; Hayes, M.; Capdeville, R.;
Genet., 27(4), 383-391. Pokorny, R. and Seiberling, M. (2004) Cancer Chemother. Phar-
[128] Hustert, E.; Haberl, M.; Burk, O.; Wolbold, R.; He, Y.Q.; Klein, macol., 54(4), 290-294.
K.; Nuessler, A.C.; Neuhaus, P.; Klattig, J.; Eiselt, R.; Koch, I.; Zi- [156] Frye, R.F.; Fitzgerald, S.M.; Lagattuta, T.F.; Hruska, M.W. and
bat, A.; Brockmoller, J.; Halpert, J.R.; Zanger, U.M. and Wo- Egorin, M.J. (2004) Clin. Pharmacol. Ther., 76(4), 323-329.
jnowski, L. (2001) Pharmacogenetics, 11(9), 773-779. [157] Smith, P.F.; Bullock, J.M.; Booker, B.M.; Haas, C.E.; Berenson,
[129] Lee, S.J.; Usmani, K.A.; Chanas, B.; Ghanayem, B.; Xi, T.; Hodg- C.S. and Jusko, W.J. (2004) Blood, 104(4), 1229-1230.
son, E.; Mohrenweiser, H.W. and Goldstein, J.A. (2003) Pharma- [158] Bolton, A.E.; Peng, B.; Hubert, M.; Krebs-Brown, A.; Capdeville,
cogenetics, 13(8), 461-472. R.; Keller, U. and Seiberling, M. (2004) Cancer Chemother. Phar-
[130] Jounaidi, Y.; Hyrailles, V.; Gervot, L. and Maurel, P. (1996) Bio- macol., 53(2), 102-106.
chem. Biophys. Res. Commun., 221(2), 466-470. [159] Chang, S.M.; Kuhn, J.G.; Rizzo, J.; Robins, H.I.; Schold, S.C. Jr;
[131] Chou, F.C.; Tzeng, S.J. and Huang, J.D. (2001) Drug Metab. Dis- Spence, A.M.; Berger, M.S.; Mehta, M.P.; Bozik, M.E.; Pollack, I.;
pos., 29(9), 1205-1209. Gilbert, M.; Fulton, D.; Rankin, C.; Malec, M. and Prados, M.D.
[132] Lee, S.J.; Usmani, K.A.; Chanas, B.; Ghanayem, B.; Xi, T.; (1998) J. Clin. Oncol., 16(6), 2188-2194.
Hodgson, E.; Mohrenweiser, H.W. and Goldstein, J.A. (2003) [160] Chang, S.M.; Kuhn, J.G.; Robins, H.I.; Schold, S.C. Jr.; Spence,
Pharmacogenetics, 13(8), 461-472. A.M.; Berger, M.S.; Mehta, M.; Pollack, I.F.; Rankin, C. and
[133] Shimada, T.; Yamazaki, H.; Mimura, M.; Inui, Y. and Guengerich, Prados, M.D. (2001) Cancer, 91(2), 417-422.
F.P. (1994) J. Pharmacol. Exp. Ther., 270(1), 414-423. [161] Rahmani, R.; Kleisbauer, J.P.; Cano, J.P.; Martin, M. and Barbet, J.
[134] Chiou, W.L.; Jeong, H.Y.; Wu, T.C. and Ma, C. (2001) Clin. (1985) Cancer Treat. Rep., 69(7-8), 839-844.
Pharmacol. Ther., 70(4), 305-310. [162] Rahmani, R.; Martin, M.; Favre, R.; Cano, J.P. and Barbet J. (1984)
[135] Rogers, J.F.; Rocci, M.L. Jr.; Haughey, D.B. and Bertino, J.S. Jr. Eur. J. Cancer Clin. Oncol., 20(11), 1409-1417.
(2003) Clin. Pharmacol. Ther., 73(3), 153-158. [163] Zhou, X.J. and Rahmani, R. (1992) Drugs, 44(Suppl 4), 1-16.
[136] Thummel, K.E.; Shen, D.D.; Podoll, T.D.; Kunze, K.L.; Trager, [164] Villikka, K.; Kivisto, K.T.; Maenpaa, H.; Joensuu, H. and Neu-
W.F.; Hartwell, P.S.; Raisys, V.A.; Marsh, C.L.; McVicar, J.P. and vonen, P.J. (1999) Clin. Pharmacol. Ther., 66(6), 589-593.
Barr, D.M. (1994) J. Pharmacol. Exp. Ther., 271(1), 549-556. [165] Oyama, T.; Morita, M.; Isse, T.; Kagawa, N.; Nakata, S.; So, T.;
[137] Thummel, K.E.; O'Shea, D.; Paine, M.F.; Shen, D.D.; Kunze, K.L.; Mizukami, M.; Ichiki, Y.; Ono, K.; Sugaya, M.; Uramoto, H.;
Perkins, J.D. and Wilkinson, G.R. (1996) Clin. Pharmacol. Ther., Yoshimatsu, T.; Hanagiri, T.; Sugio, K.; Kawamoto, T. and
59(5), 491-502. Yasumoto, K. (2005) Front. Biosci., 10, 1156-1161.
[138] Watkins, P.B.; Turgeon, D.K.; Saenger, P.; Lown, K.S.; Kolars, [166] El-Rayes, B.F.; Ali, S.; Heilbrun, L.K.; Lababidi, S.; Bouwman,
J.C.; Hamilton, T.; Fishman, K.; Guzelian, P.S. and Voorhees, J.J. D.; Visscher, D. and Philip, P.A. (2003) Clin. Cancer Res., 9(5),
(1992) Clin. Pharmacol. Ther., 52(3), 265-273. 1705-1709.
[139] Marre, F.; Sanderink, G.J.; de Sousa, G.; Gaillard, C.; Martinet, M. [167] Iscan, M.; Klaavuniemi, T.; Coban, T.; Kapucuoglu, N.; Pelkonen,
and Rahmani, R. (1996) Cancer Res., 56(6), 1296-1302. O. and Raunio, H. (2001) Breast Cancer Res. Treat., 70(1), 47-54.
[140] Shou, M.; Martinet, M.; Korzekwa, K.R.; Krausz, K.W.; Gonzalez, [168] Yokose, T.; Doy, M.; Taniguchi, T.; Shimada, T.; Kakiki, M.;
F.J. and Gelboin, H.V. (1998) Pharmacogenetics, 8(5), 391-401. Horie, T.; Matsuzaki, Y. and Mukai, K. (1999) Virchows. Arch.,
[141] Kamataki, T.; Yokoi, T.; Fujita, K. and Ando, Y. (1998) Cancer 434(5), 401-411.
Chemother. Pharmacol., 42(Suppl), S50-S53. [169] Murray, G.I.; Taylor, V.E.; McKay, J.A.; Weaver, R.J.; Ewen,
[142] Hirth, J.; Watkins, P.B.; Strawderman, M.; Schott, A.; Bruno, R. S.W.; Melvin, W.T. and Burke, M.D. (1995) J. Pathol., 177(2),
and Baker, L.H (2000) Clin. Cancer Res., 6(4), 1255-1258. 147-152.
Cytochrome P450 and Anticancer Drugs Current Drug Metabolism, 2006, Vol. 7, No. 1 37

[170] Murray, G.I.; Weaver, R.J.; Paterson, P.J.; Ewen, S.W.; Melvin, [173] Miyoshi, Y.; Ando, A.; Takamura, Y.; Taguchi, T.; Tamaki, Y. and
W.T. and Burke, M.D. (1993) J. Pathol., 169(3), 347-353. Noguchi, S. (2002) Int. J. Cancer, 97(1), 129-132.
[171] Murray, G.I.; Shaw, D.; Weaver, R.J.; McKay, J.A.; Ewen, S.W.; [174] Jounaidi, Y.; Hecht, J.E. and Waxman, D.J. (1998) Cancer Res.,
Melvin, W.T. and Burke, M.D. (1994) Gut, 35(5), 599-603. 58(19), 4391-401.
[172] McKay, J.A.; Murray, G.I.; Weaver, R.J.; Ewen, S.W.; Melvin, [175] Jounaidi, Y. and Waxman, D.J. (2004) Cancer Res., 64(1), 292-
W.T. and Burke, M.D. (1993) Gut, 34(9), 1234-1239. 303.

Received: June 10, 2005 Revised: August 1, 2005 Accepted: August 4, 2005

You might also like