ArticlePDF AvailableLiterature Review

Pharmacogenomics of Tamoxifen Therapy

Authors:

Abstract and Figures

Tamoxifen is a standard endocrine therapy for the prevention and treatment of steroid hormone receptor-positive breast cancer. Tamoxifen requires enzymatic activation by cytochrome P450 (CYP) enzymes for the formation of active metabolites 4-hydroxytamoxifen and endoxifen. As compared with the parent drug, both metabolites have an approximately 100-fold greater affinity for the estrogen receptor and the ability to inhibit cell proliferation. The polymorphic CYP2D6 is the key enzyme in this biotransformation, and recent mechanistic, pharmacologic, and clinical evidence suggests that genetic variants and drug interaction by CYP2D6 inhibitors influence the plasma concentrations of active tamoxifen metabolites and the outcomes of tamoxifen-treated patients. In particular, nonfunctional (poor metabolizer) and severely impaired (intermediate metabolizer) CYP2D6 alleles are associated with higher recurrence rates. Accordingly, CYP2D6 (cytochrome P450, family 2, subfamily D, polypeptide 6) genotyping before treatment to predict metabolizer status may open new avenues for individualizing endocrine treatment, with the maximum benefit being expected for extensive metabolizers. Moreover, strong CYP2D6 inhibitors such as the selective serotonin reuptake inhibitors paroxetine and fluoxetine, which are used to treat hot flashes, should be avoided because they severely impair formation of the active metabolites.
Content may be subject to copyright.
Pharmacogenomics of Tamoxifen Therapy
Hiltrud Brauch,
1,2*
Thomas E. Mu¨ rdter,
1,2
Michel Eichelbaum,
1,2
and Matthias Schwab
1,2,3
BACKGROUND:Tamoxifen is a standard endocrine ther-
apy for the prevention and treatment of steroid hor-
mone receptor–positive breast cancer.
CONTENT:Tamoxifen requires enzymatic activation by
cytochrome P450 (CYP) enzymes for the formation of
active metabolites 4-hydroxytamoxifen and endoxifen.
As compared with the parent drug, both metabolites
have an approximately 100-fold greater affinity for the
estrogen receptor and the ability to inhibit cell prolif-
eration. The polymorphic CYP2D6 is the key enzyme in
this biotransformation, and recent mechanistic, pharma-
cologic, and clinical evidence suggests that genetic vari-
ants and drug interaction by CYP2D6 inhibitors influence
the plasma concentrations of active tamoxifen metabo-
lites and the outcomes of tamoxifen-treated patients. In
particular, nonfunctional (poor metabolizer) and se-
verely impaired (intermediate metabolizer) CYP2D6 al-
leles are associated with higher recurrence rates.
SUMMARY:Accordingly, CYP2D6 (cytochrome P450,
family 2, subfamily D, polypeptide 6) genotyping be-
fore treatment to predict metabolizer status may open
new avenues for individualizing endocrine treatment,
with the maximum benefit being expected for exten-
sive metabolizers. Moreover, strong CYP2D6 inhibi-
tors such as the selective serotonin reuptake inhibitors
paroxetine and fluoxetine, which are used to treat hot
flashes, should be avoided because they severely impair
formation of the active metabolites.
© 2009 American Association for Clinical Chemistry
The pharmacogenomics of drug-metabolizing en-
zymes involved in the biotransformation of tamoxifen
has become a major area of interest, owing to its poten-
tial to predict a breast cancer patient’s treatment out-
come before the initiation of treatment. If the tamox-
ifen pharmacogenomic paradigm were to be borne out
in proof of principle, patients eligible for endocrine
treatment would be able to exploit it by opting for their
personally most powerful medication. Most breast
cancers, particularly those of postmenopausal women,
are hormone receptor positive; therefore, hundreds of
thousands of women worldwide initiate endocrine
treatment each year. On the basis of results of the Early
Breast Cancer Trialist Collaborative Group, the stan-
dard recommendation has been 5 years of therapy with
the selective estrogen receptor (ER)
4
modulator ta-
moxifen (1 ). Tamoxifen is currently prescribed in
120 countries worldwide as a component of standard
adjuvant therapy in early breast cancer and in the met-
astatic setting for patients with steroid hormone
receptor–positive breast tumors. In primary breast
cancer, adjuvant tamoxifen significantly decreases re-
lapse rates and mortality in pre- and postmenopausal
patients, and the therapy benefit from 5 years of adju-
vant tamoxifen is maintained, even 10 years after pri-
mary diagnosis (1 ). In postmenopausal women with
endocrine-responsive disease, tamoxifen is a valid
therapy option, along with aromatase inhibitors (AIs)
(2 ), and is considered the standard care for the preven-
tion of invasive breast cancer in premenopausal
women at high risk, including those who have had duc-
tal carcinoma in situ (3 ), and for the treatment of male
breast cancer (4 ). Tamoxifen is generally well toler-
ated, and menopausal symptoms, including hot
flashes, are the most common side effects. Severe side
effects, such as thromboembolic events or endometrial
carcinoma, are rare (1 ). The clinical benefit of tamox-
ifen has been evident for more than 3 decades; how-
ever, up to 50% of patients who receive adjuvant
tamoxifen relapse or die from tumor-specific resis-
tance or host genome–associated factors.
The field of tamoxifen pharmacogenomics gained
substantial impetus from the elucidation of tamoxifen
metabolism and metabolite pharmacology through
studies that identified major active metabolites formed
by cytochrome P450 (CYP) enzymes, particularly
CYP2D6, which exhibit substantial genetic and pheno-
typic polymorphism. Several clinical studies have re-
ported on the relationship of genotype and/or pheno-
type variants with the clinical outcome of tamoxifen
1
Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Ger-
many;
2
University Tu¨ bingen, Tu¨bingen, Germany;
3
Department of Clinical
Pharmacology, University Hospital Tu¨ bingen, Tu¨bingen, Germany.
* Address correspondence to this author at: Dr. Margarete Fischer-Bosch Institute
of Clinical Pharmacology, Auerbachstrasse 112, 70376 Stuttgart, Germany. Fax
49-(0)711-859295; e-mail hiltrud.brauch@ikp-stuttgart.de.
Received November 28, 2008; accepted June 8, 2009.
Previously published online at DOI: 10.1373/clinchem.2008.121756
4
Nonstandard abbreviations: ER, estrogen receptor; CYP, cytochrome P450; UGT,
UDP-glucuronosyltransferase; SULT, sulfotransferase; EM, extensive metabo-
lizer; PM, poor metabolizer; IM, intermediate metabolizer; UM, ultrarapid
metabolizer; SSRI, serotonin reuptake inhibitor; AI, aromatase inhibitor.
Clinical Chemistry 55:10
1770–1782 (2009) Review
1770
therapy, and international efforts are currently under
way to clarify this relationship.
In light of the potential for a future translation of
tamoxifen pharmacogenomics into clinical practice,
this review seeks to impart the underlying pharmaco-
logic, genetic, and phenotypic principles for a mecha-
nistic explanation of tamoxifen efficacy. It highlights
the biotransformation of tamoxifen into primary and
secondary metabolites with an emphasis on the clini-
cally active metabolites 4-hydroxytamoxifen and
4-hydroxy-N-desmethyltamoxifen (endoxifen). Ow-
ing to the key role of CYP2D6, this review focuses on
the relationships between the CYP2D6
5
(cytochrome
P450, family 2, subfamily D, polypeptide 6) genotype
and phenotype. This discussion also includes the phe-
nocopying effect of CYP2D6 inhibitors, which are fre-
quently coadministered to alleviate hot flashes in post-
menopausal women treated with tamoxifen. These
basic research findings provide the scientific back-
ground for a thorough discussion of the currently
available literature on tamoxifen pharmacogenetic
studies. Finally, there is a possibility that other drug-
metabolizing enzymes and even nonmetabolic factors
can influence tamoxifen efficacy. In considering these
topics, this review provides an overview of the princi-
ples of the emerging practice of personalized medicine
for the improvement of the outcomes of endocrine
drug treatment in breast cancer.
Tamoxifen Metabolism and Active Metabolites
trans-Tamoxifen {(Z)-2-[4-(1,2-diphenylbut-1-enyl)-
phenoxy]-N,N-dimethyl-ethanamine} undergoes ex-
tensive phase I and phase II metabolism in the hu-
man liver (Fig. 1). The bioconversion of tamoxifen
involves N-oxidation, N-demethylation, and hy-
droxylation. Formation of the major metabolite
N-desmethyltamoxifen is primarily catalyzed by
CYP3A4 and 3A5, with minor contributions by
CYP2D6, 1A1, 1A2, 2C19, and 2B6 (5–7 ). The steady-
state plasma concentration of N-desmethyltamoxifen
after 20 mg tamoxifen is administered daily for at least
3 months is approximately twice as high as that of the
parent drug (100 –290
g/L vs 72–160
g/L) (8 –14 ).
This fact is of utmost clinical importance because
N-desmethyltamoxifen is subject to hydroxylation,
predominantly at the para position, to produce the ma-
jor clinically active metabolite endoxifen. Importantly,
the conversion of N-desmethyltamoxifen into endox-
ifen is catalyzed almost exclusively by CYP2D6
(15, 16). Plasma concentrations of endoxifen have
been observed to range from a mean of 8.1
g/L (n 51)
for patients with 2 variant CYP2D6 alleles to 20.7
g/L
(n 55) for patients with 2 wild-type alleles (17 ). In ad-
dition, N-desmethyltamoxifen can also be desmethylated
by CYP3A4 to form N,N-didesmethyltamoxifen.
Another clinically active metabolite is 4-
hydroxytamoxifen, which is formed by 4-hydroxylation,
also at the para position of the phenyl ring of the parent
drug. This conversion is catalyzed by a number of CYPs,
including CYP2D6, 3A4, 2C9, 2B6, and 2C19 (7, 18–
21 ). Compared with endoxifen, the steady-state con-
centrations of 4-hydroxytamoxifen are lower, ranging
from 1.15
g/L to 6.4
g/L (11, 14, 22 ). With the ex-
ception of endoxifen and 4-hydroxytamoxifen, no
other highly active metabolites have been described
thus far.
Further hydroxylation also occurs at the 4posi-
tion of the other phenyl ring system, leading to 4-
hydroxytamoxifen, which is mainly mediated by
CYP2B6 and 2D6 (7 ), and to 4-hydroxy-N-
desmethyltamoxifen. Another hydroxylated metabo-
lite,
-hydroxytamoxifen, is produced mainly by
CYP3A4 (5, 6, 23, 24 ).
4-Hydroxylated metabolites undergo in vitro
chemical isomerization into the respective E or cis iso-
mers (25 ), which are weak ER antagonists. In addition,
isomerization of 4-hydroxytamoxifen is catalyzed by
CYP1B1, 2B6, and 2C19 (7 ). Of note, an accumulation
of cis-4-hydroxytamoxifen was observed in tumor tis-
sues of patients whose tumors showed resistance to ta-
moxifen treatment (26 ); however, because data on the
plasma concentrations of cis isomers are sparse, this
observation may be regarded as preliminary. Addi-
tional hydroxylation of 4-hydroxytamoxifen by
CYP3A4 and 2D6 at the phenyl moiety leads to 3,4-
dihydroxytamoxifen (27 ), a compound that is capable
of binding covalently to protein and to DNA, thereby
contributing to the reported toxic and carcinogenic ef-
fects associated with tamoxifen treatment (28, 29).
Another route of tamoxifen metabolism is the for-
mation of tamoxifen-N-oxide by flavin-containing
monooxygenases 1 and 3, with a chance for tamoxifen-
N-oxide to be reduced back to tamoxifen by a number
of different CYPs, including CYP2A6, 1A1, 3A4, and
others (30, 31). From an analytical point of view, how-
ever, this metabolite cannot be ignored because of the
likelihood of chemical reduction of the N-oxide during
5
Human genes:
CYP2D6
, cytochrome P450, family 2, subfamily D, polypeptide 6;
CYP2D7P1
, cytochrome P450, family 2, subfamily D, polypeptide 7 pseudogene
1;
CYP2D7P2
, cytochrome P450, family 2, subfamily D, polypeptide 7 pseudo-
gene 2;
CYP2D8P1
, cytochrome P450, family 2, subfamily D, polypeptide 8
pseudogene 1;
CYP2D8P2
, cytochrome P450, family 2, subfamily D, polypeptide
8 pseudogene 2;
CYP2C9
, cytochrome P450, family 2, subfamily C, polypeptide
9;
CYP2C19
, cytochrome P450, family 2, subfamily C, polypeptide 19;
CYP2B6
,
cytochrome P450, family 2, subfamily B, polypeptide 6;
CYP3A4
, cytochrome
P450, family 2, subfamily A, polypeptide 4;
CYP3A5
, cytochrome P450, family
2, subfamily A, polypeptide 5;
SULT1A1
, sulfotransferase family, cytosolic, 1A,
phenol-preferring, member 1;
BRCA1
, breast cancer 1, early onset;
BRCA2
,
breast cancer 2, early onset.
Tamoxifen Pharmacogenomics Review
Clinical Chemistry 55:10 (2009) 1771
Fig. 1. Metabolic transformation of tamoxifen in humans.
Major metabolic pathways are highlighted with bold arrows. Enzymes preferentially catalyzing a distinct metabolic step are
indicated in bold. Hb, hemoglobin; FMO1, flavin-containing monooxygenase 1.
Review
1772 Clinical Chemistry 55:10 (2009)
sample preparation, a reason why the quantification of
tamoxifen-N-oxide may be regarded as a problem in
the analysis of tamoxifen metabolites. Thus far, few
data on this issue are available, suggesting that the
N-oxide in a patient’s plasma accounts for 15% of
tamoxifen (32 ).
At the level of phase II tamoxifen metabolism, sul-
fation and glucuronidation are major mechanisms.
O-glucuronidation of 4-hydroxytamoxifen is mainly
mediated by UDP-glucuronosyltransferases (UGTs)
UGT1A4, 2B15, 2B7, 1A8, and various others to produce
4-hydroxytamoxifen-O-glucuronide (33–35 ). Endox-
ifen is predominantly glucuronidated by UGT1A10 and
1A8 to the corresponding O-glucuronide. Of note is
that in addition to hydroxylated metabolites that un-
dergo phase II metabolism at the hydroxyl moiety,
tamoxifen itself is conjugated by UGT1A4 to the cor-
responding N
-glucuronide (36, 37). In contrast to
endoxifen, which does not form any N
-glucuronide,
4-hydroxytamoxifen is glucuronidated by UGT1A4 at
the amino group to produce 4-hydroxytamoxifen-
N
-glucuronide (34, 37). The formation of sulfates
of 4-hydroxytamoxifen and endoxifen is catalyzed
by sulfotransferases (SULTs) SULT1E1, 1A1, and 2A1
(32, 38). E isomers of both 4-hydroxytamoxifen and
endoxifen are also substrates for these conjugation
reactions but seem to have different affinities for differ-
ent isoenzymes (33 ).
-Hydroxytamoxifen is sulfatized
by SULT2A1 (39 ); the resulting
-hydroxytamoxifen
sulfate is suspected to exert carcinogenic effects after
covalently binding to DNA (40, 41).
Although the number of tamoxifen metabolites
that have been identified in vitro is large (Fig. 1), in
vivo analytical measurements of plasma samples
from tamoxifen-treated patients have quantified few
metabolites, including N-desmethyltamoxifen, endox-
ifen, 4-hydroxytamoxifen, N,N-didesmethyltamoxifen,
-hydroxytamoxifen, and tamoxifen-N-oxide (Table 1).
Therefore, there may be other, yet-unidentified tamox-
ifen metabolites present at relevant concentrations in pa-
tients’ plasma.
CYP2D6 Biochemistry and Genetics
CYP2D6 is a member of CYP enzyme family 2, which in
humans constitutes one third of all CYPs and is one of the
largest and best studied of isoenzyme families. Human
CYPs are heme-containing monooxygenases, and the hu-
man genome contains 57 CYP genes and about the same
number of pseudogenes grouped into 18 families and
44 subfamilies according to sequence similarities (http://
drnelson.utmem.edu/CytochromeP450.html). CYP2D6
is involved in the metabolism of many clinically im-
portant drugs, including
-blockers, antiarrhythmics,
antihypertensives, antipsychotics, antidepressants,
opioids, and others. A recent analysis of the routes of
Table 1. Tamoxifen and metabolites.
Compounds
Mean plasma
concentrations,
nmol/LaEffect on ER/affinity for ER
(estradiol 100%)
Involvement of
CYP2D6
Tamoxifen 190–420 Weak antagonist/2%
b
N
-Desmethyltamoxifen 280–800 Weak antagonist/1%
b
Minor
N
,
N
-Didesmethyltamoxifen 90–120 Weak antagonist No
Endoxifen 14–130 Strong antagonist/equal to 4-hydroxytamoxifen Almost exclusively
4-Hydroxytamoxifen 3–17
c
Strong antagonist/188%
b
Among others
-Hydroxytamoxifen 1 None No
3,4-Dihydroxytamoxifen No data available Weak antagonist/high affinity Together with CYP3A4
Tamoxifen-
N
-oxide 15–24 Weak antagonist
d
No
4-Hydroxytamoxifen-
O
-glucuronide No data available No antagonist
e
See 4-hydroxy-tamoxifen
4-Hydroxytamoxifen-
N
-glucuronide No data available No antagonist
e
See 4-hydroxy-tamoxifen
Endoxifen-
O
-glucuronide No data available No antagonist
e
See endoxifen
-Hydroxytamoxifen sulfate No data available No data available No
a
Range of mean plasma concentrations according to different investigators [Dowsett et al.
(9)
, Hutson et al.
(10)
, Jin et al.
(11)
, Lee et al.
(12)
, Sheth et al.
(14)
,
Lim et al.
(17)
, Stearns et al.
(22)
, Gjerde et al.
(32)
, Langan-Fahey et al.
(108)
].
b
According to Wakeling and Slater
(109)
.
c
MacCallum et al.
(13)
reported much higher concentrations (67 nmol/L).
d
Might be due to reduction to tamoxifen.
e
According to Lazarus et al.
(110)
.
Tamoxifen Pharmacogenomics Review
Clinical Chemistry 55:10 (2009) 1773
elimination for the “top 200 drugs” in the US (http://
www.rxlist.com; most frequently prescribed 200 drugs,
April 2008) showed that 15% were drugs that are
CYP2D6 substrates, compared with subfamilies
CYP3A (37%) and CYP2C (33%) (42 ).
The human CYP2D6 locus on chromosome 22 in-
cludes the CYP2D6 gene and pseudogenes CYP2D7P1
(cytochrome P450, family 2, subfamily D, polypep-
tide 7 pseudogene 1), CYP2D7P2 (cytochrome P450,
family 2, subfamily D, polypeptide 7 pseudogene 2),
CYP2D8P1 (cytochrome P450, family 2, subfamily D,
polypeptide 8 pseudogene 1), and CYP2D8P2 (cyto-
chrome P450, family 2, subfamily D, polypeptide 8
pseudogene 2) originally described as pseudogenes
CYP2D7 and CYP2D8 (43 ). The CYP2D6 gene consists
of 9 exons and 8 introns, and the sequence is highly
polymorphic. By way of clinical observation (i.e., ad-
ministration of the antiarrhythmic and oxocytic drug
sparteine (44 ) and the antihypertensive agent debriso-
quine (45 )), the first CYP2D6 phenotypic variant
(sparteine/debrisoquine polymorphism) distinct from
an extensive metabolizer (EM) phenotype was identi-
fied more than 30 years ago and was termed a “poor
metabolizer” (PM) phenotype. Currently, 4 CYP2D6
phenotypes are commonly observed in Caucasian pop-
ulations on the basis of their drug-oxidation capacities:
EM, intermediate metabolizer (IM), PM, and ultra-
rapid metabolizer (UM) (46–48). Among Caucasians,
about 7%–10% of individuals are PMs, 10%–15% are
IMs, and, at the opposite end of the activity spectrum,
up to 10%–15% are UMs.
The PM status can be deduced with 99% cer-
tainty from the presence of 2 nonfunctional alleles,
with 20 null alleles having been identified (43 ).
Therefore, it is possible to exactly predict the CYP2D6
PM phenotype (i.e., lack of catalytic function of the
enzyme) by genotyping the patient’s DNA without the
need to phenotype (42, 46, 48, 49 ). The EM phenotype
is due to the presence of 1 or 2 allelic variants with
wild-type function, such as *1 or *2. This phenotype
can be separated by genotype into homozygous or
heterozygous EMs, depending on whether they carry
1 or 2 functional alleles. Because heterozygous EMs
who carry one *1 or *2 allele in combination with an
IM or PM allele have somewhat impaired enzyme
production and function, they have been classified as
IMs, assuming a gene-dosage effect such that het-
erozygous EMs would have only 50% of the enzyme
amount and catalytic activity of homozygous EMs.
This assumption is not correct, however, and there is
substantial overlap between homozygous and het-
erozygous EMs in both enzyme content and activity.
Consequently, the genotype has a rather poor pre-
dictive value. Of note is that the IM has a phenotype
and genotype distinct from the heterozygous EM
(47, 50 –52 ) that involves impaired gene expression
and enzyme function (these variants include *9, *10,
and *41) and/or nonfunctional variants (47, 52 ).
Within the German population, 2%–3% are carriers
of a duplicated/multiplied CYP2D6 gene and there-
fore have very high enzyme activity (UM). These dif-
ferences in enzyme activity can have profound con-
sequences on the plasma concentrations of drug
metabolites, as has been observed for the tricyclic
antidepressant nortriptyline. A 30-fold difference
between PMs and UMs in steady-state plasma con-
centrations of nortriptyline was observed when nor-
triptyline was prescribed as a standard daily dose of
100–150 mg (53, 54). With respect to UM pheno-
type, however, only 20%–30% of UM phenotypes
observed in the Caucasian population are identifi-
able through genotyping (46, 48, 55 ).
Thus far, systematic genetic analyses of large
numbers of individuals have led to the discovery of
100 different alleles [http://www.cypalleles.ki.se,
(56 )]. At least 15 of these alleles encode nonfunc-
tional gene products caused by aberrant splicing,
nonsense codons, mutations of single base pairs,
small insertions/deletions, larger chromosomal de-
letions of the entire CYP2D6 gene, CYP2D6/CYP2D7
hybrid genes, or mutations that cause lack of heme
incorporation or otherwise produce nonfunctional
full-length proteins.
There are significant ethnic differences with re-
spect to PM, IM, and UM frequencies, heralding the
possibility that different ethnic groups vary with re-
spect to the clinical outcomes of drug therapy with
CYP2D6 substrates. Within this context it is important
to appreciate that the frequency of gene duplication is
much higher in northeastern African populations
[e.g., 29% in Ethiopia (57 )] and in Saudi Arabia
[21% (58 )] compared with populations of European
descent (59, 60 ). In Asian populations, however, the
CYP2D6*10-associated IM is prevalent (61 ), with
the frequency in Han Chinese being 57% and the PM
playing a minor role (59 ).
Overall, an awareness of the CYP2D6 genotype–
phenotype relationship may influence treatment deci-
sions, particularly in cases for which an effective alter-
native drug is available. As in the case of orally
administered codeine, which in the 10% of Caucasians
who are PMs is not metabolized efficiently to mor-
phine and therefore provides little analgesic effect,
there is a chance that women with a CYP2D6 PM or IM
genotype/phenotype also will not benefit from the an-
tiestrogenic effects of tamoxifen, owing to insufficient
production of active metabolites. With respect to UMs,
who in cases of codeine treatment develop severe opi-
oid side effects due to rapid morphine formation
(62, 63), it is important to note that such women pa-
Review
1774 Clinical Chemistry 55:10 (2009)
tients may be more susceptible to hot flashes during
tamoxifen therapy.
CYP2C9,2C19,2B6,3A4, and 3A5 Genetics
Other important CYP isoenzymes of subfamily 2 that
are involved in the bioactivation of tamoxifen are
CYP2C9, 2C19, and 2B6 (15, 18); these enzymes are
also polymorphic. Of the 30 variant alleles of
CYP2C9 (cytochrome P450, family 2, subfamily C,
polypeptide 9), the *2 and *3 alleles have been thor-
oughly investigated and found to be associated with
significant but highly variable reductions in intrinsic
clearance, depending on the substrate (64 ). The *3 al-
lele is more strongly affected than *2, with a reduction
in enzyme activity of up to 90% for some specific drugs
(65 ). Both alleles are present in approximately 35% of
Caucasians but are less prevalent in black and Asian
populations (42, 66). About 2% and 24% of individu-
als in the Caucasian population are homozygous and
heterozygous for the variants, respectively (67 ). Nu-
merous clinical studies have demonstrated the clinical
significance of CYP2C9 genetics with respect to an as-
sociation with higher incidences of adverse drug reac-
tions. The most prominent example is warfarin, an an-
ticoagulant, and several retrospective and prospective
studies have confirmed that CYP2C9 genetics is clini-
cally useful for adjusting warfarin dosage to reduce se-
rious warfarin-related bleeding events (68, 69). The
anticoagulant response also depends on the genetics of
vitamin K epoxide reductase (68 ). Moreover, gastroin-
testinal bleeding from nonsteroidal antiinflammatory
drugs (70 ) and such side effects as hypoglycemia
caused by sulfonylureas (71 ) have also been attributed
to CYP2C9 polymorphisms.
For the CYP2C19 gene (cytochrome P450, family
2, subfamily C, polypeptide 19), the known null alleles
(CYP2C19*2,*3,*4,*5,*6,*7, and *8) have no
CYP2C19 enzyme activity (PM); the *2 allele is preva-
lent in Caucasians. These null alleles are due to a splice
defect (*2), a premature stop codon (*3), or an alter-
ation in CYP2C19 structure and/or stability (72 )
(http://www.cypalleles.ki.se/). Recently, several new
CYP2C19 alleles have been identified (*9*25) in indi-
viduals from different racial groups; however, whether
these mutations produce significant alterations in en-
zyme activity in vivo is not clear. CYP2C19*2 and *3
are the most frequent variants. According to geno-
typing and phenotyping results and in analogy to
CYP2D6, the distribution of PMs shows wide inter-
ethnic differences. In Caucasian Europeans, the mean
frequency of PM individuals is 3%, whereas PM fre-
quencies as high as 23% have been identified in Asian/
Oceanian populations (72, 73). Carriers of heterozy-
gous variants constitute 32% of Caucasians, however
(74 ). A promoter variant of CYP2C19*17 has recently
been identified and shown to be associated with in-
creased CYP2C19 activity in vivo (UM) with the
CYP2C19 substrate omeprazole [a proton pump in-
hibitor (75 )] and the antidepressant escitalopram
(76 ). Differences in CYP2C19*17 allele frequency have
been reported: 18% in both a Swedish and an Ethiopian
population (75 ), 25% in a German population (77 ),
and 27% in a Polish population (78 ). A lower fre-
quency (4%) has been reported for Chinese individuals
(75 ). Given these genotype/phenotype relationships,
there is a possibility that the CYP2C19 UM may play a
role in tamoxifen metabolism and clinical outcome, as
we have reported for our breast cancer tamoxifen phar-
macogenetic study (79 ).
With respect to CYP2B6 (cytochrome P450, family
2, subfamily B, polypeptide 6), the most common vari-
ant allele, *6, occurs at frequencies of 15%– 60% across
different populations (80 ). Genotyping of CYP2B6*6
predicted increased plasma concentrations of efavirenz
and nevirapine and efavirenz-related neurotoxicity in
HIV-infected individuals (81, 82), and the results sug-
gested reducing the dose by 35% in African patients
who were homozygous for CYP2B6*6 (83 ). These find-
ings are in agreement with the lower activities of
CYP2B6*6 isoenzyme, which may be substrate depen-
dent, however. At present, any contribution of CYP2B6
variants to tamoxifen outcome is unknown.
The most important CYP isoenzyme subfamilies
involved in human drug metabolism are CYP3A4 and
3A5, which participate in the metabolism of 40% of the
drugs that are most frequently prescribed (42 ). There is
little evidence for a relevant contribution of CYP3A4
(cytochrome P450, family 2, subfamily A, polypeptide
4) gene expression and enzyme function, although de-
fective CYP3A4 mutants may account for toxicity in
very rare cases (84 ). In contrast, genetic polymor-
phisms define much of the variation in CYP3A5 (cyto-
chrome P450, family 2, subfamily A, polypeptide 5)
expression. The higher incidence of the inactive
CYP3A5*3 variant in Caucasians (85%–95%) vs Afri-
can Americans (30%–50%) causes the lower CYP3A5
protein level seen in Caucasians compared with African
Americans (30% vs 50%). CYP3A5*6 and *7 lack any
functional activity and occur solely in individuals of
African origin. Apart from a clear effect on the immu-
nosuppressant tacrolimus (85 ), the contribution of the
polymorphic CYP3A5 enzyme to CYP3A-mediated
metabolism remains controversial. It is difficult to de-
lineate the relative contributions of CYP3A4 and
CYP3A5 because their protein structures, functions,
and substrates are so similar. In fact, one of these en-
zymes may functionally compensate for the lack of the
other. Whether CYP3A4 and/or CYP3A5 variants con-
tribute to tamoxifen outcome is unknown.
Tamoxifen Pharmacogenomics Review
Clinical Chemistry 55:10 (2009) 1775
Tamoxifen Pharmacogenomics
The rationale underlying the tamoxifen pharmaco-
genomic principle is that variant DNA sequences of
drug-metabolizing enzymes that encode proteins
with reduced or absent enzyme function may be as-
sociated with lower plasma concentrations of active
tamoxifen metabolites, which could have an impact
on the efficacy of tamoxifen treatment. About 30
years ago, Jordan et al. characterized the first potent
antiestrogen metabolite, 4-hydroxytamoxifen, and
reported a 100-fold greater affinity for the ER than
the parent drug (86 ). This metabolite was later
shown to be 30- to 100-fold more potent than ta-
moxifen in suppressing estrogen-dependent cell
proliferation (86 – 89 ). Despite its potency as an an-
tiestrogen, the contribution of this metabolite to the
overall clinical effect of tamoxifen has remained un-
clear, because its plasma concentrations are rela-
tively low compared with those of tamoxifen and
other metabolites (86 ). Our knowledge of the link
between tamoxifen metabolism and treatment re-
sponse rapidly expanded after the characterization
of endoxifen (16, 22 ), which, although it had been
identified in the late 1980s, initially remained ob-
scure with respect to its biological activity. Finally, a
series of laboratory studies for the characterization
of its pharmacology established that endoxifen has a
potency equivalent to 4-hydroxytamoxifen in terms
of its binding affinity for ERs (16 ), suppression of
estrogen-dependent proliferation of breast cancer
cells (16, 89, 90 ), and modulation of estrogen-
mediated global gene expression (91 ). A detailed in
vitro analysis showed that endoxifen is formed
mainly by 4-hydroxylation of the primary metabo-
lite N-desmethyltamoxifen, with the CYP2D6 en-
zyme catalyzing this rate-limiting step (15 ). Owing to
the dominant role of CYP2D6 in the formation of en-
doxifen, variation in the CYP2D6 genotype and pheno-
type is at the heart of tamoxifen pharmacogenetics. The
currently available evidence for this notion is based on
findings obtained at 2 levels of clinical investigations,
which addressed (a) the association between the con-
centrations of active tamoxifen metabolites either with
CYP2D6 genotype or by clinical outcome, and (b) the
association between CYP2D6 genotype and clinical
outcome. The latter approach has shown that patients
with 2 functional CYP2D6 alleles benefited the most
from tamoxifen treatment. Further elucidation of the
relationship between plasma concentrations of en-
doxifen in vivo and clinical outcomes will require
additional detailed investigations with large patient
cohorts.
Effects of Tamoxifen Metabolite Concentrations
Prospective cohort studies of adjuvant tamoxifen treat-
ment have shown wide interindividual variation in the
formation of tamoxifen metabolites and substantial re-
ductions in the steady-state plasma concentrations of
endoxifen during tamoxifen treatment in women car-
rying CYP2D6 gene variants (8, 11, 22 ). Moreover,
convincing evidence have shown that selective seroto-
nin reuptake inhibitors (SSRIs) such as paroxetine and
fluoxetine, which are known to be strong CYP2D6 in-
hibitors, reduce plasma endoxifen concentrations. In
particular, the phenocopy of a significant reduction in
endoxifen plasma concentrations induced by SSRIs
was observed in breast cancer patients homozygous for
the wild-type CYP2D6 genotype, whereas the concen-
trations of other metabolites remained unaffected by
the CYP2D6 genotype/phenotype. Although the rela-
tionship between CYP2D6 variants and plasma endox-
ifen concentrations was first shown for patients with
the PM CYP2D6*4 genotype (11 ), a quantitative ap-
proach that included PM, IM, and UM genotypes sub-
stantiated this relationship (8 ); however, endoxifen
concentrations overlap across genotypes. It follows
that other factors may modify plasma endoxifen
concentrations.
A relationship between CYP2D6 variants and
higher concentrations of N-desmethyltamoxifen (i.e.,
the endoxifen precursor) has been reported at the level
of chemoprevention. Significantly higher plasma con-
centrations of N-desmethyltamoxifen were reported
for mutation carriers after 1 year of tamoxifen therapy,
indicating that the conversion to clinically active en-
doxifen may be impaired (92 ).
A more recent study addressed the relationship be-
tween CYP2D6 and SULT1A1 (sulfotransferase family,
cytosolic, 1A, phenol-preferring, member 1) geno-
types, including the effect of SULT1A1 copy number
on the pharmacokinetics of tamoxifen during steady-
state treatment (32 ). Whereas both CYP2D6 and
SULT1A1 genotypes influenced the pharmacokinet-
ics of tamoxifen metabolites, SULT1A1 copy num-
ber did not. Lower metabolic ratios with respect to
the formation of endoxifen and 4-hydroxytamoxifen
but higher metabolic ratios for the formation of
N-desmethyltamoxifen (endoxifen precursor) were
observed in carriers of CYP2D6 variant genotypes, a
result consistent with a gene-dosage effect. In contrast,
patients carrying CYP2D6 alleles with high predicted
enzymatic activity showed higher metabolic ratios for
both active metabolites. Whether such metabolic ratios
are of clinical relevance remains to be determined.
Similarly, a study of a prospective cohort of Ko-
rean patients with early or metastatic breast cancer
found an association between the IM CYP2D6*10 ho-
Review
1776 Clinical Chemistry 55:10 (2009)
mozygous variant and lower steady-state plasma con-
centrations of 4-hydroxytamoxifen and endoxifen
(17 ), and a Chinese study found that patients homozy-
gous for CYP2D6*10 had lower serum concentrations
of 4-hydroxytamoxifen (93 ). The high prevalence of
the CYP2D6*10 allele in East Asia, together with the IM
association of impaired formation of an active metabolite,
confirms the CYP2D6 PM findings in Caucasians.
Clinical Outcome of Tamoxifen Therapy and
Prediction
The first evidence linking CYP2D6 variants with treat-
ment response was obtained from a prospective ran-
domized phase III trial of postmenopausal women
with ER-positive breast cancer (North Central Cancer
Treatment Group adjuvant breast cancer trial) for the
investigation of the effect of adding the androgen flu-
oxymesterone for 1 year to the standard regimen of 5
years of adjuvant tamoxifen. The pharmacogenetic in-
vestigation of patients from the tamoxifen-only arm
showed that after a median follow-up of 11.4 years, the
CYP2D6*4 variant allele was an independent predictor
of a higher risk of relapse and a lower incidence of hot
flashes (94 ). A follow-up study found that in addition
to CYP2D6 genetics, the phenocopying due to the
coprescription of CYP2D6 inhibitors (SSRIs) was an
independent predictor of breast cancer outcome in
postmenopausal women taking tamoxifen (95 ). Re-
cently, a robust association between CYP2D6 genotype
and treatment outcome was obtained from a nonran-
domized retrospective cohort of ER-positive post-
menopausal breast cancer patients undergoing adju-
vant tamoxifen therapy (79 ). At a median follow-up of
71 months, carriers of PM and IM genotypes (i.e., car-
riers of CYP2D6*4,*5,*10, and *41 alleles) had signif-
icantly more breast cancer recurrences, shorter relapse-
free times, and worse event-free survival than carriers
of functional alleles (Fig. 2). This association was not
observed in postmenopausal ER-positive patients not
treated with tamoxifen. Interestingly, the UM
CYP2C19*17 variant also had a favorable effect on ta-
moxifen treatment outcome. Patients with the ho-
mozygous *17 genotype had significantly fewer breast
cancer recurrences, longer relapse-free times, and bet-
ter event-free survival than non-*17 carriers. Overall,
this study suggested that genotyping for CYP2D6*4,*5,
*10, and *41 could identify patients who would derive
little benefit from adjuvant tamoxifen therapy. Al-
though the CYP2D6 EM phenotype will identify the
patients likely to benefit from tamoxifen, accounting
for about 50% of all patients, the benefit will be maxi-
mal for individuals with the combination of fully func-
tional CYP2D6 alleles and the CYP2C19 UM. The lat-
ter will apply to one third of all patients, indicating that
the tamoxifen pharmacogenetics issue will be relevant
Fig. 2. Kaplan–Meier probabilities of relapse-free time (RFT) of breast cancer patients for CYP2D6-metabolizer
phenotypes predicted from genotypes.
(A), Patients treated with adjuvant tamoxifen (TAM). EMs had a significantly more favorable RFT than patients with impaired
phenotypes (PMs or IMs). (B), Patients without TAM showed no differences with respect to a relationship between the
CYP2D6
-
predicted phenotype and RFT [Schroth et al.
(79)
]. hetEM, heterozygous EM. Originally published in Schroth, W et al.: J Clin Oncol 25
(33), 2007: 5187–93. Reprinted with permission. © 2008 American Society of Clinical Oncology. All rights reserved.
Tamoxifen Pharmacogenomics Review
Clinical Chemistry 55:10 (2009) 1777
for a substantial fraction of breast cancer patients re-
ceiving endocrine treatment.
Clinical studies from Korea, China, and Japan also
have linked poor clinical outcome with CYP2D6 genet-
ics. As expected for populations with a high prevalence
of the IM CYP2D6*10 allele, the *10 homozygote geno-
type was associated with a poor clinical outcome in a
Korean cohort of metastatic breast cancer patients,
whereas the *10 heterozygote and wild-type homozy-
gote genotypes were not (17 ). Likewise, patients from
China who were homozygous for the CYP2D6*10 allele
(93 ) showed an association with unfavorable disease-
free survival. The latter result was substantiated
through comparison with a control patient group
without tamoxifen treatment, in which no association
between clinical outcome and the CYP2D6*10 variant
was observed. Moreover, patients homozygous for
CYP2D6*10 from a Japanese breast cancer cohort that
underwent adjuvant tamoxifen monotherapy showed
a significantly higher incidence of recurrence within 10
years of follow-up, compared with patients with wild-
type CYP2D6 (96 ). Although some of the sample sizes
were low in the Asian studies demonstrating the geno-
type– efficacy correlation, the findings of the clinical
implications of CYP2D6 genotypes predictive for ta-
moxifen efficacy are in line with the findings of others.
On the other hand, a study from the US reported
no association between CYP2D6 genetics and tamox-
ifen outcome (97 ), and contradictory results for this
relationship were reported in a study from Sweden,
which found the CYP2D6*4 variant to be associated
with a better clinical outcome in tamoxifen-treated pa-
tients (98 ). An extended study showed favorable
disease-free survival in CYP2D6*4 carriers compared
with patients homozygous or heterozygous for the
functional CYP2D6 allele (99 ).
The issue of the role of CYP2D6 in tamoxifen ther-
apy for breast cancer has also been addressed within the
context of breast cancer prevention. For example, data
from the Italian Tamoxifen Trial suggest that women
with a CYP2D6*4/*4 genotype may be less likely to ben-
efit from tamoxifen as a chemopreventive agent. This
finding supports the notion of CYP2D6 playing an im-
portant role in tamoxifen’s metabolic activation and
efficacy (100 ). Moreover, the “a priori” hypothesis that
hot flashes may be an independent predictor of tamox-
ifen efficacy has been addressed in the Women’s
Healthy Eating and Living randomized trial (101 ).Of
the 864 patients taking tamoxifen, 674 (78%) reported
hot flashes, and 12.9% of these patients had experi-
enced recurrent breast cancer after 7.3 years of follow-
up, whereas 21% of the patients who did not have hot
flashes had recurrent breast cancer during this period.
Because hot flashes were a stronger predictor of a breast
cancer–specific outcome than age, hormone receptor
status, or tumor stage at diagnosis, the authors sug-
gested an association between side effects, tamoxifen
metabolism, and efficacy. Finally, a small study of fa-
milial breast cancer patients who were carriers of either
BRCA1 (breast cancer 1, early onset) or BRCA2 (breast
cancer 2, early onset) mutations and treated with ta-
moxifen suggested a relationship between CYP2D6 PM
status and a worse survival in familial breast cancer
(102 ); however, because of the small numbers of pa-
tients as well as the inclusion of ER-positive and ER-
negative patients in this investigation, clarification
provided by further studies will be needed to distin-
guish a pharmacogenetic effect from a poor prognostic
effect in carriers of these BRCA mutations.
Given the current treatment practice of long-term
estrogen deprivation in ER-positive postmenopausal
breast cancer patients with the use of AIs as a valid
option, the question of the impact of pharmacogenetic
variation on the optimal choice for adjuvant endocrine
therapy has been addressed in a modeling analysis
(103 ). A Markov model was created to examine
whether the optimal treatment strategy for patients
with the wild-type CYP2D6 gene differs from that for
carriers of the CYP2D6*4 mutation. The study used
patients from the BIG1–98 trial, information from this
trial on relapse risk, and the corresponding genotype
data of Goetz et al. (94 ). Under the assumption that AI
metabolism is independent from CYP2D6, the model
suggests that the 5-year benefit of adjuvant tamoxifen
therapy may exceed even that of up-front AI treatment
in postmenopausal CYP2D6 EM patients.
Conclusion: Clinical Relevance of CYP2D6 in Breast
Cancer
Strong mechanistic, pharmacologic, and clinical evi-
dence, as well as modeling data, now indicate that ta-
moxifen efficacy and clinical outcome depend on
CYP2D6 metabolism controlled by CYP2D6 enzyme
polymorphisms and on pharmacologic interactions.
Data from international studies have consistently dem-
onstrated that plasma concentrations of active tamox-
ifen metabolites are linked with genetically determined
CYP2D6 metabolizer status, phenocopying by strong
CYP2D6 inhibitors, and clinical outcome. The few
conflicting data may be explained by variation in the
studies with respect to patient-inclusion criteria, ta-
moxifen doses, length of treatment, additional chemo-
therapy regimens, or a lack of consistent ER testing.
Importantly, most authors agree that CYP2D6 gene
variants, as well as inhibition of CYP2D6 by prescribed
comedications such as SSRIs, may decrease tamoxifen
metabolism and thus negatively affect tamoxifen effi-
cacy and treatment outcome.
Review
1778 Clinical Chemistry 55:10 (2009)
There are a number of potential clinical conse-
quences from these emerging data on CYP2D6 and the
outcomes of tamoxifen treatment. First, potent SSRIs
such as paroxetine or fluoxetine should not be used to
relieve hot flashes in breast cancer patients receiving ta-
moxifen. Although SSRIs are one of the few evidence-
based therapy options for menopausal vasomotor symp-
toms (104 ), convincing data now indicate that these
drugs may compromise tamoxifen efficacy via a pheno-
copying effect due to interference with CYP2D6-
dependent tamoxifen metabolism. Yet, differences in the
plasma concentrations of tamoxifen metabolites have
been observed, depending on the strength of the CYP2D6
inhibitor (11, 105 ). If treatment of hot flashes is indicated,
an SSRI such as citalopram or escitalopram or a selective
norepinephrine reuptake inhibitor such as venlafaxine
should be used, because these compounds have shown no
appreciable inhibition of CYP2D6.
Second, the relationship between CYP2D6 geno-
type, phenotype, and treatment outcome points to a
possible benefit of up-front CYP2D6 genotyping prior
to a decision on an adjuvant endocrine treatment. A
comprehensive robust, standardized, and quality-
controlled CYP2D6-genotyping assay will have to test
for genetic variants that could affect tamoxifen metab-
olism. According to the data of Goetz et al. (94 ) and
Schroth et al. (79 ), such assays should include testing
for common PM alleles (CYP2D6*3,*4, and *5) and for
IM alleles, depending on the individual’s ethnic origin.
Of note, *41 is the most frequent IM allele in Europe-
ans, *17 is the principal IM allele in Africans, and *10
dominates in Asians (*9 should also be considered)
(59 ). Other areas of interest with respect to clinical
application are the measurement of plasma endoxifen
concentrations as a surrogate of CYP2D6 phenotype.
Given alternative treatment options (i.e., tamox-
ifen vs AI), and considering the available scientific and
clinical evidence, an individualized approach for en-
docrine treatment of postmenopausal breast cancer
patients is desirable. One may speculate that tamoxi-
fen alone is adequate for CYP2D6 EMs and EM carri-
ers, whereas postmenopausal patients with variant
CYP2D6 alleles may fare better with up-front AI ther-
apy. Although this approach may be regarded as
straightforward for PM patients, the best treatment
may be less clear for IM patients. IM is a common phe-
notype among many ethnic groups, including Cauca-
sians, African Americans, and Asians, so data on link-
ing IM genotypes with therapeutic threshold and
efficacy are in demand to adequately address the clini-
cally important question of tamoxifen dose adjust-
ment. Similarly, any impact of UM phenotypes on
metabolite concentrations, treatment efficacy, and tox-
icity that have potential implications for dosing re-
quires further investigations. Formal recommenda-
tions on the integration of CYP2D6 genotypes into
treatment decisions still must await validation of these
genotypes in larger retrospective studies, as is being
attempted by the International Tamoxifen Pharmaco-
genetics Consortium (http://www.pharmgkb.org/do/
serve?objId63&objClsProject), or prospective
clinical trials. Thus far, no study has addressed the
question of whether genetically predisposed differ-
ences in 4-hydroxytamoxifen and endoxifen concen-
trations are associated with treatment response or dis-
ease progression and with side effects such as hot
flashes, including phenocopying effects; therefore,
therapeutic drug monitoring as a useful surrogate is
currently not available in the case of tamoxifen.
Whether determination of the CYP2D6 genotype will
become a diagnostic tool for selecting the appropriate
adjuvant endocrine therapy for ER-positive postmeno-
pausal breast cancer patients awaits validation in pro-
spective clinical trials that randomize tamoxifen vs AI
treatment according to CYP2D6 genotypes. Such pro-
spective clinical trials are currently being planned.
Other open questions may address the clinical rel-
evance of other drug-metabolizing enzymes and muta-
tions, as well as ethnic variation, in the prevalence of
their treatment outcome–relevant genotypes. Finally,
there is the possibility that pharmacokinetic genes will
only partly explain the pharmacogenomics of tamox-
ifen. It will therefore be important to also explore the
contribution of pharmacodynamic genes in evaluating
antiestrogen resistance as a feature of the tumor cell
and in addressing the role of genes associated with
estrogen-mediated cell proliferation. Within this con-
text, it will be interesting to learn whether genes encod-
ing the ER, its coactivators, or its corepressors (106 ),as
well as antiestrogen resistance genes (107 ) and their
variants, will affect the response to tamoxifen. Such
results may increase the overall potential of tamoxifen
pharmacogenomics.
To this end, it is important to appreciate that most
cancer therapies in current use have been established
empirically. The recent progress in our understanding
of the pharmacology and pharmacogenetics of tamox-
ifen, however, holds promise for the improvement of
treatments through personalized medicine. Because
the genome-based approach uses CYP2D6 genotyping
to predict a patient’s metabolizer phenotype, ethical
issues need to be sufficiently addressed. In the light of
acceptable alternatives, an informed choice about ad-
juvant endocrine treatment and, most importantly,
avoiding a therapy that may lack efficacy must be of
prime interest. It will therefore be important to make
patients and their caregivers aware of these issues and
to initiate discussions with regulatory authorities.
Tamoxifen Pharmacogenomics Review
Clinical Chemistry 55:10 (2009) 1779
Author Contributions: All authors confirmed they have contributed to
the intellectual content of this paper and have met the following 3 re-
quirements: (a) significant contributions to the conception and design,
acquisition of data, or analysis and interpretation of data; (b) drafting
or revising the article for intellectual content; and (c) final approval of
the published article.
Authors’ Disclosures of Potential Conflicts of Interest: Upon
manuscript submission, all authors completed the Disclosures of Poten-
tial Conflict of Interest form. Potential conflicts of interest:
Employment or Leadership: None declared.
Consultant or Advisory Role: None declared.
Stock Ownership: None declared.
Honoraria: None declared.
Research Funding: Robert Bosch Foundation, Stuttgart, Germany,
and Bundesministerium fu¨ r Bildung und Forschung Grant No.
01ZP0502.
Expert Testimony: None declared.
Role of Sponsor: The funding organizations played no role in the
design of study, choice of enrolled patients, review and interpretation
of data, or preparation or approval of manuscript.
References
1. Early Breast Cancer Trialists’ Collaborative
Group (EBCTCG). Effects of chemotherapy and
hormonal therapy for early breast cancer on
recurrence and 15-year survival: an overview of
the randomised trials. Lancet 2005;365:1687–
717.
2. Goldhirsch A, Wood WC, Gelber RD, Coates AS,
Thu¨ rlimann B, Senn HJ. Progress and promise:
highlights of the international expert consensus
on the primary therapy of early breast cancer
2007. Ann Oncol 2007;18:1133–44.
3. Fisher B, Costantino JP, Wickerham DL, Cecchini
RS, Cronin WM, Robidoux A, et al. Tamoxifen
for the prevention of breast cancer: current sta-
tus of the National Surgical Adjuvant Breast and
Bowel Project P-1 study. J Natl Cancer Inst
2005;97:1652–62.
4. Fentiman IS, Fourquet A, Hortobagyi GN. Male
breast cancer. Lancet 2006;367:595–604.
5. Boocock DJ, Brown K, Gibbs AH, Sanchez E,
Turteltaub KW, White IN. Identification of hu-
man CYP forms involved in the activation of
tamoxifen and irreversible binding to DNA. Car-
cinogenesis 2002;23:1897–901.
6. Coller JK, Krebsfaenger N, Klein K, Wolbold R,
Nussler A, Neuhaus P, et al. Large interindi-
vidual variability in the in vitro formation of
tamoxifen metabolites related to the develop-
ment of genotoxicity. Br J Clin Pharmacol 2004;
57:105–11.
7. Crewe HK, Notley LM, Wunsch RM, Lennard MS,
Gillam EM. Metabolism of tamoxifen by recom-
binant human cytochrome P450 enzymes:
formation of the 4-hydroxy, 4-hydroxy and
N-desmethyl metabolites and isomerization of
trans-4-hydroxytamoxifen. Drug Metab Dispos
2002;30:869–74.
8. Borges S, Desta Z, Li L, Skaar TC, Ward BA,
Nguyen A, et al. Quantitative effect of CYP2D6
genotype and inhibitors on tamoxifen
metabolism: implication for optimization of
breast cancer treatment. Clin Pharmacol Ther
2006;80:61–74.
9. Dowsett M, Cuzick J, Howell A, Jackson I, and
the ATAC Trialists’ Group. Pharmacokinetics of
anastrozole and tamoxifen alone, and in com-
bination, during adjuvant endocrine therapy for
early breast cancer in postmenopausal women:
a sub-protocol of the ‘Arimidex™ and Tamox-
ifen Alone or in Combination’ (ATAC) trial. Br J
Cancer 2001;85:317–24.
10. Hutson PR, Love RR, Havighurst TC, Rogers E,
Cleary JF. Effect of exemestane on tamoxifen
pharmacokinetics in postmenopausal women
treated for breast cancer. Clin Cancer Res 2005;
11:8722–7.
11. Jin Y, Desta Z, Stearns V, Ward B, Ho H, Lee KH,
et al. CYP2D6 genotype, antidepressant use,
and tamoxifen metabolism during adjuvant
breast cancer treatment. J Natl Cancer Inst
2005;97:30–9.
12. Lee KH, Ward BA, Desta Z, Flockhart DA, Jones
DR. Quantification of tamoxifen and three me-
tabolites in plasma by high-performance liquid
chromatography with fluorescence detection: ap-
plication to a clinical trial. J Chromatogr B Analyt
Technol Biomed Life Sci 2003;791:245–53.
13. MacCallum J, Cummings J, Dixon JM, Miller WR.
Concentrations of tamoxifen and its major me-
tabolites in hormone responsive and resistant
breast tumours. Br J Cancer 2000;82:1629–35.
14. Sheth HR, Lord G, Tkaczuk K, Danton M, Lewis
LM, Langenberg P, et al. Aging may be associ-
ated with concentrations of tamoxifen and its
metabolites in breast cancer patients. J Womens
Health (Larchmt) 2003;12:799–808.
15. Desta Z, Ward BA, Soukhova NV, Flockhart DA.
Comprehensive evaluation of tamoxifen sequen-
tial biotransformation by the human cytochrome
P450 system in vitro: prominent roles for CYP3A
and CYP2D6. J Pharmacol Exp Ther 2004;310:
1062–75.
16. Johnson MD, Zuo H, Lee KH, Trebley JP, Rae JM,
Weatherman RV, et al. Pharmacological charac-
terization of 4-hydroxy-
N
-desmethyl tamoxifen,
a novel active metabolite of tamoxifen. Breast
Cancer Res Treat 2004;85:151–9.
17. Lim HS, Ju LH, Seok LK, Sook LE, Jang IJ, Ro
J. Clinical implications of CYP2D6 genotypes
predictive of tamoxifen pharmacokinetics in
metastatic breast cancer. J Clin Oncol 2007;25:
3837–45.
18. Coller JK, Krebsfaenger N, Klein K, Endrizzi K,
Wolbold R, Lang T, et al. The influence of
CYP2B6, CYP2C9 and CYP2D6 genotypes on the
formation of the potent antioestrogen
Z
-4-
hydroxy-tamoxifen in human liver. Br J Clin
Pharmacol 2002;54:157–67.
19. Crewe HK, Ellis SW, Lennard MS, Tucker GT.
Variable contribution of cytochromes P4502D6,
2C9 and 3A4 to the 4-hydroxylation of tamox-
ifen by human liver microsomes. Biochem Phar-
macol 1997;53:171–8.
20. Dehal SS, Kupfer D. CYP2D6 catalyzes tamoxifen
4-hydroxylation in human liver. Cancer Res
1997;57:3402–6.
21. Mani C, Gelboin HV, Park SS, Pearce R, Parkin-
son A, Kupfer D. Metabolism of the antimam-
mary cancer antiestrogenic agent tamoxifen. I.
Cytochrome P-450-catalyzed N-demethylation
and 4-hydroxylation. Drug Metab Dispos 1993;
21:645–56.
22. Stearns V, Johnson MD, Rae JM, Morocho A,
Novielli A, Bhargava P, et al. Active tamoxifen
metabolite plasma concentrations after coad-
ministration of tamoxifen and the selective se-
rotonin reuptake inhibitor paroxetine. J Natl
Cancer Inst 2003;95:1758–64.
23. Kim SY, Suzuki N, Santosh Laxmi YR, Rieger R,
Shibutani S. Alpha-hydroxylation of tamoxifen
and toremifene by human and rat cytochrome
P450 3A subfamily enzymes. Chem Res Toxicol
2003;16:1138–44.
24. Notley LM, Crewe KH, Taylor PJ, Lennard MS,
Gillam EM. Characterization of the human cy-
tochrome P450 forms involved in metabolism of
tamoxifen to its
-hydroxy and
,4-dihydroxy
derivatives. Chem Res Toxicol 2005;18:1611–8.
25. Katzenellenbogen JA, Carlson KE, Katzenellenbo-
gen BS. Facile geometric isomerization of phenolic
non-steroidal estrogens and antiestrogens: limita-
tions to the interpretation of experiments charac-
terizing the activity of individual isomers. J Steroid
Biochem 1985;22:589–96.
26. Osborne CK, Wiebe VJ, McGuire WL, Ciocca DR,
DeGregorio MW. Tamoxifen and the isomers of
4-hydroxytamoxifen in tamoxifen-resistant tu-
mors from breast cancer patients. J Clin Oncol
1992;10:304–10.
27. Dehal SS, Brodie AMH, Kupfer D. The aromatase
inactivator 4-hydroxyandrostenedione (4-OH-A)
inhibits tamoxifen metabolism by rat hepatic
cytochrome P-450 3A: potential for drug-drug
interaction of tamoxifen and 4-OH-A in com-
bined anti-breast cancer therapy. Drug Metab
Dispos 1999;27:389–94.
28. Dehal SS, Kupfer D. Evidence that the catechol
3,4-dihydroxytamoxifen is a proximate interme-
diate to the reactive species binding covalently
to proteins. Cancer Res 1995;56:1283–90.
29. Liu X, Pisha E, Tonetti DA, Yao D, Li Y, Yao J, et
al. Antiestrogenic and DNA damaging effects
induced by tamoxifen and toremifene metabo-
lites. Chem Res Toxicol 2003;16:832–7.
30. Hodgson E, Rose RL, Cao Y, Dehal SS, Kupfer D.
Flavin-containing monooxygenase isoform spec-
ificity for the N-oxidation of tamoxifen deter-
mined by product measurement and NADPH
oxidation. J Biochem Mol Toxicol 2000;14:118
20.
31. Parte P, Kupfer D. Oxidation of tamoxifen by
human flavin-containing monooxygenase (FMO)
Review
1780 Clinical Chemistry 55:10 (2009)
1 and FMO3 to tamoxifen-
N
-oxide and its novel
reduction back to tamoxifen by human cyto-
chromes P450 and hemoglobin. Drug Metab
Dispos 2005;33:1446–52.
32. Gjerde J, Hauglid M, Breilid H, Lundgren S,
Varhaug JE, Kisanga ER, et al. Effects of CYP2D6
and SULT1A1 genotypes including SULT1A1
gene copy number on tamoxifen metabolism.
Ann Oncol 2008;19:56–61.
33. Nishiyama T, Ogura K, Nakano H, Ohnuma T,
Kaku T, Hiratsuka A, et al. Reverse geometrical
selectivity in glucuronidation and sulfation of
cis
- and
trans
-4-hydroxytamoxifens by human
liver UDP-glucuronosyltransferases and sulfo-
transferases. Biochem Pharmacol 2002;63:
1817–30.
34. Ogura K, Ishikawa Y, Kaku T, Nishiyama T,
Ohnuma T, Muro K, Hiratsuka A. Quaternary
ammonium-linked glucuronidation of
trans
-4-
hydroxytamoxifen, an active metabolite of ta-
moxifen, by human liver microsomes and UDP-
glucuronosyltransferase 1A4. Biochem Pharmacol
2006;71:1358–69.
35. Sun D, Sharma AK, Dellinger RW, Blevins-
Primeau AS, Balliet RM, Chen G, et al. Glucu-
ronidation of active tamoxifen metabolites by
the human UDP glucuronosyltransferases. Drug
Metab Dispos 2007;35:2006–14.
36. Kaku T, Ogura K, Nishiyama T, Ohnuma T, Muro
K, Hiratsuka A. Quaternary ammonium-linked
glucuronidation of tamoxifen by human liver
microsomes and UDP-glucuronosyltransferase
1A4. Biochem Pharmacol 2004;67:2093–102.
37. Sun D, Chen G, Dellinger RW, Duncan K, Fang
JL, Lazarus P. Characterization of tamoxifen and
4-hydroxytamoxifen glucuronidation by human
UGT1A4 variants. Breast Cancer Res 2006;8:
R50.
38. Falany JL, Pilloff DE, Leyh TS, Falany CN. Sulfa-
tion of raloxifene and 4-hydroxytamoxifen by
human cytosolic sulfotransferases. Drug Metab
Dispos 2006;34:361–8.
39. Apak TI, Duffel MW. Interactions of the stereo-
isomers of alpha-hydroxytamoxifen with human
hydroxysteroid sulfotransferase SULT2A1 and
rat hydroxysteroid sulfotransferase STa. Drug
Metab Dispos 2004;32:1501–8.
40. Kim SY, Laxmi YR, Suzuki N, Ogura K, Watabe T,
Duffel MW, Shibutani S. Formation of
tamoxifen-DNA adducts via O-sulfonation, not
O-acetylation, of alpha-hydroxytamoxifen in rat
and human livers. Drug Metab Dispos 2005;33:
1673–8.
41. Osborne MR, Hewer A, Phillips DH. Resolution
of alpha-hydroxytamoxifen; R-isomer forms
more DNA adducts in rat liver cells. Chem Res
Toxicol 2001;14:888–93.
42. Zanger UM, Turpeinen M, Klein K, Schwab M.
Functional pharmacogenetics/genomics of hu-
man cytochromes P450 involved in drug bio-
transformation. Anal Bioanal Chem 2008;392:
1093–108.
43. Zanger UM. The CYP2D subfamily. In: Ioannides
C, ed. Cytochromes P450: role in the metabo-
lism and toxicity of drugs and other xenobiotics.
London: Royal Chemical Society; 2008. p 241–
75.
44. Eichelbaum M, Spannbrucker N, Dengler HJ.
Proceedings: N-oxidation of sparteine in man
and its interindividual differences. Naunyn
Schmiedebergs Arch Pharmacol 1975;287
(Suppl):R94.
45. Mahgoub A, Idle JR, Dring LG, Lancaster R,
Smith RL. Polymorphic hydroxylation of debriso-
quine in man. Lancet 1977;2:584–6.
46. Griese EU, Zanger UM, Brudermanns U, Gaedigk
A, Mikus G, Morike K, et al. Assessment of the
predictive power of genotypes for the in-vivo
catalytic function of CYP2D6 in a German pop-
ulation. Pharmacogenetics 1998;8:15–26.
47. Raimundo S, Toscano C, Klein K, Fischer J,
Griese EU, Eichelbaum M, et al. A novel intronic
mutation, 2988GA, with high predictivity for
impaired function of cytochrome P450 2D6 in
white subjects. Clin Pharmacol Ther 2004;76:
128–38.
48. Sachse C, Brockmoller J, Bauer S, Roots I. Cyto-
chrome P450 2D6 variants in a Caucasian
population: allele frequencies and phenotypic
consequences. Am J Hum Genet 1997;60:284
95.
49. Marez D, Legrand M, Sabbagh N, Guidice JM,
Spire C, Lafitte JJ, et al. Polymorphism of the
cytochrome P450 CYP2D6 gene in a European
population: characterization of 48 mutations
and 53 alleles, their frequencies and evolution.
Pharmacogenetics 1997;7:193–202.
50. Raimundo S, Fischer J, Eichelbaum M, Griese
EU, Schwab M, Zanger UM. Elucidation of the
genetic basis of the common ‘intermediate me-
tabolizer’ phenotype for drug oxidation by
CYP2D6. Pharmacogenetics 2000;10:577–81.
51. Toscano C, Klein K, Blievernicht J, Schaeffeler E,
Saussele T, Raimundo S, et al. Impaired expres-
sion of CYP2D6 in intermediate metabolizers
carrying the *41 allele caused by the intronic
SNP 2988GA: evidence for modulation of
splicing events. Pharmacogenet Genomics 2006;
16:755–66.
52. Zanger UM, Fischer J, Raimundo S, Stuven T,
Evert BO, Schwab M, Eichelbaum M. Compre-
hensive analysis of the genetic factors determin-
ing expression and function of hepatic CYP2D6.
Pharmacogenetics 2001;11:573–85.
53. Bertilsson L, Dahl ML, Sjo¨ qvist F, Aberg-Wistedt
A, Humble M, Johansson I, et al. Molecular basis
for rational megaprescribing in ultrarapid hy-
droxylators of debrisoquine. Lancet 1993;341:
63.
54. Dalen P, Dahl ML, Bernal Ruiz ML, Nordin J,
Bertilsson L. 10-Hydroxylation of nortriptyline in
white persons with 0, 1, 2, 3, and 13 functional
CYP2D6 genes. Clin Pharmacol Ther 1998;63:
444–52.
55. Lovlie R, Daly AK, Matre GE, Molven A, Steen
VM. Polymorphisms in CYP2D6 duplication-
negative individuals with the ultrarapid metabo-
lizer phenotype: a role for the CYP2D6*35 allele
in ultrarapid metabolism? Pharmacogenetics
2001;11:45–55.
56. Algeciras-Schimnich A, O’Kane DJ, Snozek CL.
Pharmacogenomics of tamoxifen and irinotecan
therapies. Clin Lab Med 2008;28:553–67.
57. Aklillu E, Persson I, Bertilsson L, Johansson I,
Rodrigues F, Ingelman-Sundberg M. Frequent
distribution of ultrarapid metabolizers of de-
brisoquine in an Ethiopian population carrying
duplicated and multiduplicated functional
CYP2D6 alleles. J Pharmacol Exp Ther 1996;278:
441–6.
58. McLellan RA, Oscarson M, Seidegard J, Evans
DA, Ingelman-Sundberg M. Frequent occurrence
of CYP2D6 gene duplication in Saudi Arabians.
Pharmacogenetics 1997;7:187–91.
59. Sistonen J, Sajantila A, Lao O, Corander J, Bar-
bujani G, Fuselli S. CYP2D6 worldwide genetic
variation shows high frequency of altered activ-
ity variants and no continental structure. Phar-
macogenet Genomics 2007;17:93–101.
60. Ingelman-Sundberg M. Genetic polymorphisms
of cytochrome P450 2D6 (CYP2D6): clinical con-
sequences, evolutionary aspects and functional
diversity. Pharmacogenomics J 2005;5:6–13.
61. Bradford LD. CYP2D6 allele frequency in Euro-
pean Caucasians, Asians, Africans and their de-
scendants. Pharmacogenomics 2002;3:229–43.
62. Gasche Y, Daali Y, Fathi M, Chiappe A, Cottini S,
Dayer P, Desmeules J. Codeine intoxication as-
sociated with ultrarapid CYP2D6 metabolism.
N Engl J Med 2004;351:2827–31.
63. Koren G, Cairns J, Chitayat D, Gaedigk A, Leeder
SJ. Pharmacogenetics of morphine poisoning in
a breastfed neonate of a codeine-prescribed
mother. Lancet 2006;368:704.
64. Lee CR, Goldstein JA, Pieper JA. Cytochrome
P450 2C9 polymorphisms: a comprehensive re-
view of the in-vitro and human data. Pharma-
cogenetics 2002;12:251–63.
65. King BP, Khan TI, Aithal GP, Kamali F, Daly
AK. Upstream and coding region CYP2C9
polymorphisms: correlation with warfarin dose
and metabolism. Pharmacogenetics 2004;14:
813–22.
66. Garcia-Martin E, Martinez C, Ladero JM, Agun-
dez JA. Interethnic and intraethnic variability of
CYP2C8 and CYP2C9 polymorphisms in healthy
individuals. Mol Diagn Ther 2006;10:29–40.
67. Xie HG, Kim RB, Wood AJ, Stein CM. Molecular
basis of ethnic differences in drug disposition
and response. Annu Rev Pharmacol Toxicol
2001;41:815–50.
68. Flockhart DA, O’Kane D, Williams MS, Watson
MS, Flockhart DA, Gage B, et al. Pharmacoge-
netic testing of CYP2C9 and VKORC1 alleles for
warfarin. Genet Med 2008;10:139–50.
69. Limdi NA, Veenstra DL. Warfarin pharmacoge-
netics. Pharmacotherapy 2008;28:1084–97.
70. Pilotto A, Seripa D, Franceschi M, Scarcelli C, Co-
laizzo D, Grandone E, et al. Genetic susceptibility
to nonsteroidal anti-inflammatory drug-related
gastroduodenal bleeding: role of cytochrome P450
2C9 polymorphisms. Gastroenterology 2007;133:
465–71.
71. Holstein A, Plaschke A, Ptak M, Egberts EH,
El-Din J, Brockmoller J, Kirchheiner J. Associa-
tion between CYP2C9 slow metabolizer geno-
types and severe hypoglycaemia on medication
with sulphonylurea hypoglycaemic agents. Br J
Clin Pharmacol 2005;60:103–6.
72. Desta Z, Zhao X, Shin JG, Flockhart DA. Clinical
significance of the cytochrome P450 2C19 ge-
netic polymorphism. Clin Pharmacokinet 2002;
41:913–58.
73. Xie HG, Stein CM, Kim RB, Wilkinson GR, Flock-
hart DA, Wood AJ. Allelic, genotypic and phe-
notypic distributions of
S
-mephenytoin 4-
hydroxylase (CYP2C19) in healthy Caucasian
populations of European descent throughout
the world. Pharmacogenetics 1999;9:539–49.
74. Wedlund PJ. The CYP2C19 enzyme polymor-
Tamoxifen Pharmacogenomics Review
Clinical Chemistry 55:10 (2009) 1781
phism. Pharmacology 2000;61:174–83.
75. Sim SC, Risinger C, Dahl ML, Aklillu E, Chris-
tensen M, Bertilsson L, Ingelman-Sundberg M. A
common novel CYP2C19 gene variant causes
ultrarapid drug metabolism relevant for the drug
response to proton pump inhibitors and antide-
pressants. Clin Pharmacol Ther 2006;79:103–
13.
76. Rudberg I, Mohebi B, Hermann M, Refsum H,
Molden E. Impact of the ultrarapid CYP2C19*17
allele on serum concentration of escitalopram in
psychiatric patients. Clin Pharmacol Ther 2008;
83:322–7.
77. Justenhoven C, Hamann U, Pierl CB, Baisch C,
Harth V, Rabstein S, et al. CYP2C19*17 is as-
sociated with decreased breast cancer risk.
Breast Cancer Res Treat 2008;115:391–6.
78. Kurzawski M, Gawronska-Szklarz B, Wrzes-
niewska J, Siuda A, Starzynska T, Drozdzik M.
Effect of CYP2C19*17 gene variant on
Helico-
bacter pylori
eradication in peptic ulcer patients.
Eur J Clin Pharmacol 2006;62:877–80.
79. Schroth W, Antoniadou L, Fritz P, Schwab M,
Muerdter T, Zanger UM, et al. Breast cancer
treatment outcome with adjuvant tamoxifen rel-
ative to patient CYP2D6 and CYP2C19 geno-
types. J Clin Oncol 2007;25:5187–93.
80. Zanger UM, Klein K, Saussele T, Blievernicht J,
Hofmann MH, Schwab M. Polymorphic CYP2B6:
molecular mechanisms and emerging clinical
significance. Pharmacogenomics 2007;8:743–
59.
81. Rotger M, Colombo S, Furrer H, Bleiber G, Buclin
T, Lee BL, et al. Influence of CYP2B6 polymor-
phism on plasma and intracellular concentra-
tions and toxicity of efavirenz and nevirapine in
HIV-infected patients. Pharmacogenet Genom-
ics 2005;15:1–5.
82. Rotger M, Tegude H, Colombo S, Cavassini M,
Furrer H, Decosterd L, et al. Predictive value of
known and novel alleles of CYP2B6 for efavirenz
plasma concentrations in HIV-infected individu-
als. Clin Pharmacol Ther 2007;81:557–66.
83. Nyakutira C, Ro¨ shammar D, Chigutsa E, Chonzi
P, Ashton M, Nhachi C, Masimirembwa C. High
prevalence of the CYP2B6 516G3T(*6) variant
and effect on the population pharmacokinetics
of efavirenz in HIV/AIDS outpatients in Zimba-
bwe. Eur J Clin Pharmacol 2008;64:357–65.
84. Westlind-Johnsson A, Hermann R, Huennemeyer
A, Hauns B, Lahu G, Nassr N, et al. Identification
and characterization of CYP3A4*20, a novel
rare CYP3A4 allele without functional activity.
Clin Pharmacol Ther 2006;79:339–49.
85. Anglicheau D, Legendre C, Beaune P, Thervet E.
Cytochrome P450 3A polymorphisms and immu-
nosuppressive drugs: an update. Pharmacog-
enomics 2007;8:835–49.
86. Jordan VC. Metabolites of tamoxifen in animals
and man: identification, pharmacology, and sig-
nificance. Breast Cancer Res Treat 1982;2:123–
38.
87. Coezy E, Borgna JL, Rochefort H. Tamoxifen and
metabolites in MCF7 cells: correlation between
binding to estrogen receptor and inhibition of
cell growth. Cancer Res 1982;42:317–23.
88. Robertson DW, Katzenellenbogen JA, Long DJ,
Rorke EA, Katzenellenbogen BS. Tamoxifen an-
tiestrogens. A comparison of the activity, phar-
macokinetics, and metabolic activation of the
cis and trans isomers of tamoxifen. J Steroid
Biochem 1982;16:1–13.
89. Buck MB, Coller JK, Mu¨ rdter TE, Eichelbaum M,
Knabbe C. TGF
2 and T
RII are valid molecular
biomarkers for the antiproliferative effects of
tamoxifen and tamoxifen metabolites in breast
cancer cells. Breast Cancer Res Treat 2008;107:
15–24.
90. Lim YC, Desta Z, Flockhart DA, Skaar TC. En-
doxifen (4-hydroxy-
N
-desmethyl-tamoxifen) has
anti-estrogenic effects in breast cancer cells
with potency similar to 4-hydroxy-tamoxifen.
Cancer Chemother Pharmacol 2005;55:471–8.
91. Lim YC, Li L, Desta Z, Zhao Q, Rae JM, Flockhart
DA, Skaar TC. Endoxifen, a secondary metabo-
lite of tamoxifen, and 4-OH-tamoxifen induce
similar changes in global gene expression pat-
terns in MCF-7 breast cancer cells. J Pharmacol
Exp Ther 2006;318:503–12.
92. Decensi A, Gandini S, Serrano D, Cazzaniga M,
Pizzamiglio M, Maffini F, et al. Randomized
dose-ranging trial of tamoxifen at low doses in
hormone replacement therapy users. J Clin On-
col 2007;25:4201–9.
93. Xu Y, Sun Y, Yao L, Shi L, Wu Y, Ouyang T, et al.
Association between CYP2D6 *10 genotype and
survival of breast cancer patients receiving ta-
moxifen treatment. Ann Oncol 2008;19:1423–9.
94. Goetz MP, Rae JM, Suman VJ, Safgren SL, Ames
MM, Visscher DW, et al. Pharmacogenetics of
tamoxifen biotransformation is associated with
clinical outcomes of efficacy and hot flashes.
J Clin Oncol 2005;23:9312–8.
95. Goetz MP, Kamal A, Ames MM. Tamoxifen
pharmacogenomics: the role of CYP2D6 as a
predictor of drug response. Clin Pharmacol Ther
2008;83:160–6.
96. Kiyotani K, Mushiroda T, Sasa M, Bando Y,
Sumitomo I, Hosono N, et al. Impact of
CYP2D6*10 on recurrence-free survival in breast
cancer patients receiving adjuvant tamoxifen
therapy. Cancer Sci 2008;99:995–9.
97. Nowell SA, Ahn J, Rae JM, Scheys JO, Trovato A,
Sweeney C, et al. Association of genetic varia-
tion in tamoxifen-metabolizing enzymes with
overall survival and recurrence of disease in
breast cancer patients. Breast Cancer Res Treat
2005;91:249–58.
98. Wegman P, Vainikka L, Stal O, Nordenskjold B,
Skoog L, Rutqvist LE, Wingren S. Genotype of
metabolic enzymes and the benefit of tamoxifen
in postmenopausal breast cancer patients.
Breast Cancer Res 2005;7:R284–90.
99. Wegman P, Elingarami S, Carstensen J, Stal O,
Nordenskjold B, Wingren S. Genetic variants of
CYP3A5, CYP2D6, SULT1A1, UGT2B15 and ta-
moxifen response in postmenopausal patients
with breast cancer. Breast Cancer Res 2007;9:
R7.
100. Bonanni B, Macis D, Maisonneuve P, Johansson
HA, Gucciardo G, Oliviero P, et al. Polymor-
phism in the CYP2D6 tamoxifen-metabolizing
gene influences clinical effect but not hot
flashes: data from the Italian Tamoxifen Trial.
J Clin Oncol 2006;24:3708–9.
101. Mortimer JE, Flatt SW, Parker BA, Gold EB,
Wasserman L, Natarajan L, Pierce JP. Tamox-
ifen, hot flashes and recurrence in breast cancer.
Breast Cancer Res Treat 2008;108:421–6.
102. Newman WG, Hadfield KD, Latif A, Roberts SA,
Shenton A, McHague C, et al. Impaired tamox-
ifen metabolism reduces survival in familial
breast cancer patients. Clin Cancer Res 2008;
14:5913–8.
103. Punglia RS, Burstein HJ, Winer EP, Weeks JC.
Pharmacogenomic variation of CYP2D6 and the
choice of optimal adjuvant endocrine therapy
for postmenopausal breast cancer: a modeling
analysis. J Natl Cancer Inst 2008;100:642–8.
104. Carlson RW, Hudis CA, Pritchard KI. Adjuvant
endocrine therapy in hormone receptor-positive
postmenopausal breast cancer: evolution of
NCCN, ASCO, and St Gallen recommendations.
J Natl Compr Canc Netw 2006;4:971–9.
105. Lien EA, Solheim E, Kvinnsland S, Ueland PM.
Identification of 4-hydroxy-
N
-desmethyltamoxifen
as a metabolite of tamoxifen in human bile. Can-
cer Res 1988;48:2304–8.
106. Jordan VC, O’Malley BW. Selective estrogen-
receptor modulators and antihormonal resis-
tance in breast cancer. J Clin Oncol 2007;25:
5815–24.
107. van Agthoven T, Sieuwerts AM, Meijer-van
Gelder ME, Look MP, Smid M, Veldscholte J, et
al. Relevance of breast cancer antiestrogen re-
sistance genes in human breast cancer progres-
sion and tamoxifen resistance. J Clin Oncol
2009;27:542–9.
108. Langan-Fahey SM, Tormey DC, Jordan VC. Ta-
moxifen metabolites in patients on long-term
adjuvant therapy for breast cancer. Eur J Cancer
1990;26:883–8.
109. Wakeling AE, Slater SR. Estrogen-receptor bind-
ing and biologic activity of tamoxifen and its
metabolites. Cancer Treat Rep 1980;64:741–4.
110. Lazarus P, Blevins-Primeau AS, Zheng Y, Sun D.
Potential role of UGT pharmacogenetics in can-
cer treatment and prevention: focus on tamox-
ifen. Ann N Y Acad Sci 2009;1155:99–111.
Review
1782 Clinical Chemistry 55:10 (2009)
... Tamoxifen has a relatively low anti-estrogenic activity compared with these three active metabolites. 3,4 Both 4-hydroxy-tamoxifen and endoxifen have comparable anti-estrogenic activities, which is nearly 30-to 100-fold more potent than tamoxifen. 5 Yet, usually endoxifen is considered the most important active metabolite of tamoxifen due to the 5-10 times higher concentrations than 4-hydroxy-tamoxifen. 6 CYP2D6 enzyme is considered the rate-limiting step in tamoxifen metabolism, 3,4,7,8 and it is encoded by the highly polymorphic gene CYP2D6. ...
... 3,4 Both 4-hydroxy-tamoxifen and endoxifen have comparable anti-estrogenic activities, which is nearly 30-to 100-fold more potent than tamoxifen. 5 Yet, usually endoxifen is considered the most important active metabolite of tamoxifen due to the 5-10 times higher concentrations than 4-hydroxy-tamoxifen. 6 CYP2D6 enzyme is considered the rate-limiting step in tamoxifen metabolism, 3,4,7,8 and it is encoded by the highly polymorphic gene CYP2D6. 8 For many years, CYP2D6 genotyping has been investigated as a potential tool that could be used to predict tamoxifen efficacy. ...
... A possible explanation for this controversy might be the fact that genetic polymorphisms in CYP2D6 genotypes only partially contribute to explaining the inter-variability of endoxifen concentrations. 4,7,[10][11][12] Also the effect on survival of tamoxifen in the adjuvant setting is generally low in absolute percentages, and therefore, a difference in systemic exposure may have little effect on survival. Therefore, CYP2D6 genotyping has not been commonly implemented in the clinical practice as a manner to anticipate tamoxifen effectiveness. ...
Article
Tamoxifen is part of the standard of care of endocrine therapy for adjuvant treatment of breast cancer. However, survival outcomes with tamoxifen are highly variable. The concentration of endoxifen, the 30–100 times more potent metabolite of tamoxifen and bioactivated by the CYP2D6 enzyme, has been described as the most relevant metabolite of tamoxifen metabolism. A genome‐wide association study (GWAS) was performed with the objective to identify genetic polymorphisms associated with endoxifen serum concentration levels and clinical outcome in early‐stage breast cancer patients receiving tamoxifen. A GWAS was conducted in 608 women of the CYPTAM study (NTR1509/PMID: 30120701). Germline DNA and clinical and survival characteristics were readily available. Genotyping was performed on Infinium Global Screening Array (686,082 markers) and single nucleotide polymorphism (SNP) imputation by using 1000 Genomes. Relapse‐free survival during tamoxifen (RFSt) was defined the primary clinical outcome. Endoxifen serum concentration was analyzed as a continuous variable. Several genetic variants reached genome‐wide significance ( P value: ≤5 × 10 ⁻⁸ ). Endoxifen concentrations analysis identified 430 variants, located in TCF20 and WBP2NL genes (chromosome 22), which are in strong linkage disequilibrium with CYP2D6 variants. In the RFSt analysis, several SNP were identified ( LPP gene: rs77693286, HR 18.3, 95% CI: 15.2–21.1; rs6790761, OR 18.2, 95% CI: 15.5–21.1). Endoxifen concentrations have a strong association with the chromosome 22, which contains the CYP2D6 gene.
... The cells were then permanently maintained in media containing only 1 µM of tamoxifen as seen in (Supplementary Scheme 1). The continued treatment with 1 µM was done as this concentration is within range of the serum concentrations of the sum of tamoxifen and its metabolites in breast cancer patients treated with tamoxifen [24]. Cells were labeled 50, 35x6, and 4+1 (each group contains 9 samples) to represent the concentration and number of treatments the cells received throughout the metabolic and molecular studies. ...
Article
Full-text available
Background: Tamoxifen is commonly used in the treatment of hormonal-positive breast cancer. However, 30%-40% of tumors treated with tamoxifen develop resistance; therefore, an important step to overcome this resistance is to understand the underlying molecular and metabolic mechanisms. In the present work, we used metabolic profiling to determine potential biomarkers of tamoxifen resistance, and gene expression levels of enzymes important to these metabolites and then correlated the expression to the survival of patients receiving tamoxifen. Methods: Tamoxifen-resistant cell lines previously developed and characterized in our laboratory were metabolically profiled with nuclear magnetic resonance spectroscopy (NMR) using cryogenic probe, and the findings were correlated with the expression of genes that encode the key enzymes of the significant metabolites. Moreover, the effect of significantly altered genes on the overall survival of patients was assessed using the Kaplan-Meier plotter web tool. Results: We observed a significant increase in the levels of glutamine, taurine, glutathione, and xanthine, and a significant decrease in the branched-chain amino acids, valine, and isoleucine, as well as glutamate and cysteine in the tamoxifen-resistant cells compared to tamoxifen sensitive cells. Moreover, xanthine dehydrogenase and glutathione synthase gene expression were downregulated, whereas glucose-6-phosphate dehydrogenase was upregulated compared to control. Additionally, increased expression of xanthine dehydrogenase was associated with a better outcome for breast cancer patients. Conclusion: Overall, this study sheds light on metabolic pathways that are dysregulated in tamoxifen-resistant cell lines and the potential role of each of these pathways in the development of resistance.
... [14][15][16] NDM-tamoxifen is further metabolized to endoxifen; hence, CYP2D6 is considered a critical enzyme in tamoxifen metabolism to this active metabolite. 17 CYP2D6 gene is a highly polymorphic gene, which carries over 100 genetic variants, some of which may result in a CYP2D6 enzyme, which is non-functional or has reduced function. 9 These variants may result in variability in endoxifen plasma concentrations and may impact the success of tamoxifen therapy. ...
Article
Full-text available
Tamoxifen is the most used hormonal therapy for oestrogen receptor‐positive breast cancer. CYP2D6 is the main enzyme in the metabolic pathway of tamoxifen to endoxifen. Variations in endoxifen plasma concentrations are associated with CYP2D6 polymorphisms. This study aimed to determine the association between the CYP2D6 polymorphisms and endoxifen plasma concentrations in a cohort of Zimbabwean breast cancer patients (n = 40). TaqMan genotyping and copy number assays were done to determine CYP2D6 genotypes. Tamoxifen and metabolites were quantitated using LC‐MS/MS. The population had high frequencies of the CYP2D6 reduced function alleles, *17 (15%) and *29 (18%). The median endoxifen concentration was 4.78 ng/mL, and in 55% of the patients, mostly intermediate metabolizers were below the endoxifen therapeutic threshold of 5.97 ng/mL. The CYP2D6 phenotypes and activity scores were significantly associated with endoxifen plasma concentrations (P = 0.0151) and with endoxifen to N‐desmethyl‐tamoxifen ratios (P = 0.0006).
... The cells were then permanently maintained in media containing only 1 µM of tamoxifen as seen in (Supplementary Scheme 1). The continued treatment with 1 µM was done as this concentration is within range of the serum concentrations of the sum of tamoxifen and its metabolites in breast cancer patients treated with tamoxifen [24]. Cells were labeled 50, 35x6, and 4+1 (each group contains 9 samples) to represent the concentration and number of treatments the cells received throughout the metabolic and molecular studies. ...
Article
Full-text available
Glutathione (GSH) is an antioxidant with an important protective intracellular role against reactive oxygen species. Increased glutathione level is associated with higher rates of metastases and a more aggressive behavior in breast cancer. This study aims to study the molecular and metabolic changes in glutathione synthesis in tamoxifen resistant (TAM-R) MCF-7 breast cancer cell lines.Methods: Three tamoxifen resistant MCF-7 cell lines were produced by using two methods. The first was achieved by treating the cells with tamoxifen and gradually increasing the amount until reaching a predetermined concentration of tamoxifen. The second was achieved by giving multiple fixed concentrations of tamoxifen, the third model was produced from the second by treating the cells with continuous 1µM of tamoxifen. mRNA and metabolites were extracted from the cell lines. cDNA was synthesized from the extracted mRNA, and gene expression of glutathione synthetase (GSS) was measured using real-time PCR. Nuclear Magnetic Resonance technique with PABO probe was used for metabolic profiling of extracted metabolites from the resistant and sensitive MCF-7cells and Chenomix software for quantification. Additionally, Kaplan Meier plotter (https://kmplot.com/) was used to predict the significance of GSS gene expression to overall survival of breast cancer patients. Results: A significant increase in glutathione accompanied with significant decrease in cysteine levels were found in TAM-R cells compared to control TAM sensitive cells. However, there was a 2 times reduction in the expression of GSS in TAM-R cells. Correlation of GSS gene expression among ER +ve, PR +ve patients who received tamoxifen in their therapy showed that its down regulation is significantly linked to poor overall survival (p-value 0.022 and HR of 0.7). Conclusion: The significant increase of glutathione in TAM-R cells may contribute to their increased resistance to oxidative stress and exhibition of a more aggressive behavior. Moreover, the decrease in GSS expression is a strong indicator of decreased glutathione synthesis in TAM-R cells as a result of cysteine depletion in the cells that was correlated to poor overall survival among BC patients. However, high glutathione levels in TAM-R cells could be due to increased glutathione regeneration in TAM-R cell lines compared to control. Citation Format: Yazan I. Hamadneh, Mohammad AlWahsh, Jawad Alrawabdeh, Roland Hergenröder, Lina A. Dahabiyeh, Lama Hamadneh. Glutathione as a potential marker of tamoxifen resistance in breast cancer. [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3902.
Article
Nowadays, drug-induced liver toxicity (DILT) is one of the main contributing factors to severe liver disease. In the United States (US) alone, DILT is the cause of more than 50% of instances of acute liver failure. Prescription or over-the-counter drugs, xenobiotics, and herbal and nutritional supplements can cause DILT and could produce anomalies in hepatic function tests. Some drugs induce hepatotoxicity directly, and others induce it indirectly (i. e. through their toxic or reactive metabolites). Currently, the United States Food and Drug Administration (US FDA) has issued black box warnings for about 1279 drugs due to their hepatotoxicity. When we analyzed their mechanism in inducing hepatotoxicity, we found nearly 18 drugs causing hepatotoxicity by their toxic metabolites. In this review, we attempted to highlight the well-known drugs that induce hepatotoxicity indirectly through their toxic metabolites including the enzymes involved in the formation of these metabolites. The Cytochrome P-450 (CYP), Hypoxanthine phosphoribosyltransferase 1, Alcohol oxidase, Uridine diphosphate (UDP)-glucuronosyltransferases, Xanthine dehydrogenase, Purine-nucleoside phosphorylase, Xanthine oxidase, Thiopurine S-methyltransferase, Inosine-5'-monophosphate dehydrogenase, and aldehyde dehydrogenase are involving in the formation of toxic metabolites. The metabolic reactions and enzymes discussed in this review help toxicologists, pharmacologists, and chemists to design and develop hepatotoxicity-free pharmaceutical products containing the inhibitors of these enzymes to reduce hepatotoxicity and improve human health.
Article
Pharmacogenomics (PGx) enables personalized treatment for the prediction of drug response and to avoid adverse drug reactions. Currently, PGx mainly relies on the genetic information of absorption, distribution, metabolism, and excretion (ADME) targets such as drug-metabolizing enzymes or transporters to predict differences in the patient's phenotype. However, there is evidence that the phenotype-genotype concordance is limited. Thus, we discuss different phenotyping strategies using exogenous xenobiotics (e.g., drug cocktails) or endogenous compounds for phenotype prediction. In particular, minimally invasive approaches focusing on liquid biopsies offer great potential to preemptively determine metabolic and transport capacities. Early studies indicate that ADME phenotyping using exosomes released from the liver is reliable. In addition, pharmacometric modeling and artificial intelligence improve phenotype prediction. However, further prospective studies are needed to demonstrate the clinical utility of individualized treatment based on phenotyping strategies, not only relying on genetics. The present review summarizes current knowledge and limitations. Expected final online publication date for the Annual Review of Pharmacology and Toxicology, Volume 64 is January 2024. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Article
Objectives: The present study investigated the analysis of adverse drug reactions (ADRs) to Tamoxifen (TAM) in breast cancer patients in relation to the carriage of genetic polymorphisms of genes encoding enzymes of CYP system and transporters of P-glycoprotein (Pg) and predictive models based on it. Methods: 120 women with breast cancer taking adjuvant TAM were examined for the gene polymorphisms such as CYP2D6*4, CYP3A5*3, CYP2C9*2, CYP2C9*3, CYP2C19*2, CYP2C19*3 and ABCB1 (C3435T). Allelic variants were determined using the real-time polymerase chain reaction method. The research material was double sampling of buccal epithelium. Medical history data and extracts from case histories were used as sources of medical information, on the basis of which questionnaires specially created by us were filled out. Results: An associative analysis showed association with the development of ADRs to tamoxifen indicating their clinical significance from different genetic polymorphisms of CYP2D6, CYP3A5, CYP2C9 and ABCB1. The complex associative analysis performed using mathematical modeling made it possible to build predictive risk models for the development of ADRs such as hot flashes, dyspepsia, bone pain, and asthenia. Conclusions: Models that include both genetic and non-genetic determinants of ADRs of TAM may further improve the prediction of individual response to tamoxifen.
Article
Oxysterols play significant roles in many physiological and pathological processes including cancer. They modulate some of the cancer hallmarks pathways, influence the efficacy of anti-cancer drugs, and associate with patient survival. In this study, we aimed to analyze the role of 7-ketocholesterol (7-KC) in breast carcinoma cells and its potential modulation of the tamoxifen effect. 7-KC effects were studied in two estrogen receptor (ER)-positive (MCF-7 and T47D) and one ER-negative (BT-20) breast cancer cell lines. First, we tested the viability of cells in the presence of 7-KC. Next, we co-incubated cells with tamoxifen and sublethal concentrations of 7-KC. We also tested changes in caspase 3/7 activity, deregulation of the cell cycle, and changes in expression of selected genes/proteins in the presence of tamoxifen, 7-KC, or their combination. Finally, we analyzed the effect of 7-KC on cellular migration and invasion. We found that the presence of 7-KC slightly decreases the efficacy of tamoxifen in MCF-7 cells, while an increased effect of tamoxifen and higher caspase 3/7 activity was observed in the BT-20 cell line. In the T47D cell line, we did not find any modulation of tamoxifen efficacy by the presence of 7-KC. Expression analysis showed the deregulation in CYP1A1 and CYP1B1 with the opposite trend in MCF-7 and BT-20 cells. Moreover, 7-KC increased cellular migration and invasion potential regardless of the ER status. This study shows that 7-KC can modulate tamoxifen efficacy as well as cellular migration and invasion, making 7-KC a promising candidate for future studies.
Article
Tamoxifen, the gold standard drug for endocrine therapy for breast cancer, modulates the phosphorylation status of the TAU protein in Alzheimer's disease by inhibiting CDK5 kinase activity. Its binding to p25 prevents CDK5/p25 complexation and hence a decrease of CDK5 activity. In breast tumors, this complex is involved in the proliferation and survival of cancer cells, as well as in the disease's prognosis. Still, the molecular stability of the CDK5/p25 complex following tamoxifen exposure in this cancer type has not yet been clearly deciphered. Here, we report the functional characterization of CDK5 and its p25 regulatory subunit in the absence and presence of tamoxifen. In addition, two novel inhibitors of the kinase activity of the CDK5/p25 complex are identified, both of which would reduce the risk of recurrence of estrogen receptor-positive (ER+) breast cancers and prevent drawbacks induced by tamoxifen exposure. Accordingly, 6His-CDK5 and 6His-p25 have been expressed and purified. Fluorescence anisotropy measurements have been used to assess that the two proteins do form an active complex, and thermodynamic parameters of their interaction were measured. It was also confirmed that tamoxifen directly binds to p25 and inhibits CDK5 kinase activity. Similar observations were obtained using 4-hydroxytamoxifen, an active metabolized form of tamoxifen. Two novel compounds have been identified here that harbor a benzofuran moiety and were shown to target directly p25, and their bindings resulted in decreased CDK5 kinase activity. This encouraging alternative opens the way to the ensuing chemical optimization of this scaffold. It also promises a more specific therapeutic approach that may both tackle the pathological signaling in breast cancer and provide a potential new drug for Alzheimer's disease.
Article
Full-text available
Objective To investigate the disposition and effects of nortriptyline and its major metabolite 10-hydroxynortriptyline in panels of white subjects with different CYP2D6 genotypes, including those with duplicated and multiduplicated CYP2D6*2 genes and to evaluate the contribution of the number of functional CYP2D6 alleles to the metabolism of nortriptyline, used here as a model drug for CYP2D6 substrates.Methods Oral single doses of 25 to 50 mg nortriptyline were given to five poor metabolizers of debrisoquin (INN, debrisoquine) with no functional CYP2D6 gene, five extensive metabolizers with one functional CYP2D6 gene, five extensive metabolizers with two functional CYP2D6 genes, five ultrarapid metabolizers with duplicated CYP2D6*2 genes, and one ultrarapid metabolizer with 13 copies of the CYP2D6*2 gene. Plasma kinetics of nortriptyline and 10-hydroxynortriptyline were analyzed. Anticholinergic effects (inhibition of salivation and accommodation disturbances), sedation, blood pressure, and effect on supine and erect pulse rate were measured.ResultsThere was a clear relation between the CYP2D6 genotype and the plasma kinetics of nortriptyline and 10-hydroxynortriptyline. The proportion between the apparent oral clearances of nortriptyline in the groups with 0, 1, 2, 3, and 13 functional genes was 1:1:4:5:17. The proportions between AUC(nortriptyline) to AUC(10-hydroxynortriptyline) ratios in the groups with 0, 1, 2, 3, and 13 functional genes were 36:25:10:4:1. Oral plasma clearance of nortriptyline and AUC(nortriptyline) to AUC(10-hydroxynortriptyline) ratio both correlated significantly with the debrisoquin metabolic ratio (rS = −0.89, p = 0.0001; rS = 0.92, p = 0.0001). Although ultrarapid metabolizer subjects were given double the nortriptyline dose (50 mg), inhibition of salivation was not more pronounced compared with the other genotype groups given 25 mg nortriptyline.Conclusion The results of this study show the quantitative importance of the CYP2D6 genotype, especially the presence of multiple functional CYP2D6 genes for the pharmacokinetics of nortriptyline and 10-hydroxynortriptyline. Genotyping of subjects with multiple copies of functional genes may be of great value for differentiating ultrarapid metabolizers from patients who do not comply with the prescription and for assuring adequate drug choice and dosage for these patients.Clinical Pharmacology & Therapeutics (1998) 63, 444-452; doi:
Article
4-Hydroxytamoxifen is 100 times more potent as an oestrogen receptor antagonist than the parent drug. The aim of this study was to identify the cytochrome P450 enzymes involved in the 4-hydroxylation of tamoxifen by human liver microsomes. Microsomes from each of 10 human livers tested catalysed the reaction [ranges 0.6 to 2.9 pmol/mg protein/min (1 μM) and 6 to 25 pmol/mg protein/min (18 μM)]. Three of the livers with the lowest tamoxifen 4-hydroxylase activity were from poor CYP2D6 metabolisers. Inhibition of activity by quinidine (1 μM), sulphaphenazole (20 μM) and ketoconazole (2 μM) ranged from 0-80%, 0-80% and 12-57%, respectively. The proportion of activity inhibited by quinidine correlated positively with total microsomal tamoxifen 4-hydroxylase activity (rs = 0.89, p < 0.01). Recombinant human CYPs 2D6, 2C9 and 3A4 but not CYPs 1A1, 1A2, 2C19 and 2E1 displayed significant 4-hydroxylase activity. These findings indicate that the 4-hydroxylation of tamoxifen is catalysed almost exclusively by CYPs 2D6, 2C9 and 3A4 in human liver microsomes. However, there was marked between-subject variation in the contribution of these isoforms.
Article
Background: Tamoxifen, a selective estrogen receptor modulator (SERM), is converted to 4-hydroxy-tamoxifen and other active metabolites by cytochrome P450 (CYP) enzymes. Selective serotonin reuptake inhibitors (SSRIs), which are often prescribed to alleviate tamoxifen-associated hot flashes, can inhibit CYPs. In a prospective clinical trial, we tested the effects of coadministration of tamoxifen and the SSRI paroxetine, an inhibitor of CYP2D6, on tamoxifen metabolism. Methods: Tamoxifen and its metabolites were measured in the plasma of 12 women of known CYP2D6 genotype with breast cancer who were taking adjuvant tamoxifen before and after 4 weeks of coadministered paroxetine. We assessed the inhibitory activity of pure tamoxifen metabolites in an estradiol-stimulated MCF7 cell proliferation assay. To determine which CYP isoforms were involved in the metabolism of tamoxifen to specific metabolites, we used CYP isoform-specific inhibitors. All statistical tests were two-sided. Results: We separated, purified, and identified the metabolite 4-hydroxy-N-desmethyl-tamoxifen, which we named endoxifen. Plasma concentrations of endoxifen statistically significantly decreased from a mean of 12.4 ng/mL before paroxetine coadministration to 5.5 ng/mL afterward (difference = 6.9 ng/mL, 95% confidence interval [CI] = 2.7 to 11.2 ng/mL) (P = .004). Endoxifen concentrations decreased by 64% (95% CI = 39% to 89%) in women with a wild-type CYP2D6 genotype but by only 24% (95% CI = 23% to 71%) in women with a variant CYP2D6 genotype (P = .03). Endoxifen and 4-hydroxy-tamoxifen inhibited estradiol-stimulated MCF7 cell proliferation with equal potency. In vitro, troleandomycin, an inhibitor of CYP3A4, inhibited the demethylation of tamoxifen to N-desmethyl-tamoxifen by 78% (95% CI = 65% to 91%), and quinidine, an inhibitor of CYP2D6, reduced the subsequent hydroxylation of N-desmethyl-tamoxifen to endoxifen by 79% (95% CI = 50% to 108%). Conclusions: Endoxifen is an active tamoxifen metabolite that is generated via CYP3A4-mediated N-demethylation and CYP2D6-mediated hydroxylation. Coadministration of paroxetine decreased the plasma concentration of endoxifen. Our data suggest that CYP2D6 genotype and drug interactions should be considered in women treated with tamoxifen.
Article
The cytochrome P450 (P450)-mediated biotransformation of tamoxifen is important in determining both the clearance of the drug and its conversion to the active metabolite,trans-4-hydroxytamoxifen. Biotransformation by P450 forms expressed extrahepatically, such as in the breast and endometrium, may be particularly important in determining tissue-specific effects of tamoxifen. Moreover, tamoxifen may serve as a useful probe drug to examine the regioselectivity of different forms. Tamoxifen metabolism was investigated in vitro using recombinant human P450s. Forms CYP1A1, 1A2, 1B1, 2A6, 2B6, 2C9, 2C19, 2D6, 2E1, 3A4, 3A5, and 3A7 were coexpressed in Escherichia coli with recombinant human NADPH-cytochrome P450 reductase. Bacterial membranes were harvested and incubated with tamoxifen ortrans-4-hydroxytamoxifen under conditions supporting P450-mediated catalysis. CYP2D6 was the major catalyst of 4-hydroxylation at low tamoxifen concentrations (170 ± 20 pmol/40 min/0.2 nmol P450 using 18 μM tamoxifen), but CYP2B6 showed significant activity at high substrate concentrations (28.1 ± 0.8 and 3.1 ± 0.5 nmol/120 min/0.2 nmol P450 for CYP2D6 and CYP2B6, respectively, using 250 μM tamoxifen). These two forms also catalyzed 4′-hydroxylation (13.0 ± 1.9 and 1.4 ± 0.1 nmol/120 min/0.2 nmol P450, respectively, for CYP2B6 and CYP2D6 at 250 μM tamoxifen; 0.51 ± 0.08 pmol/40 min/0.2 nmol P450 for CYP2B6 at 18 μM tamoxifen). Tamoxifen N-demethylation was mediated by CYP2D6, 1A1, 1A2, and 3A4, at low substrate concentrations, with contributions by CYP1B1, 2C9, 2C19 and 3A5 at high concentrations. CYP1B1 was the principal catalyst of 4-hydroxytamoxifentrans-cis isomerization but CYP2B6 and CYP2C19 also contributed.
Article
The genotoxic tamoxifen metabolite alpha -hydroxytamoxifen has been resolved into R- and S-enantiomers. This was achieved by preparing its ester with S-camphanic acid, chromatographic separation into two diastereoisomers, and hydrolysis to give (+)- and (-)-alpha -hydroxytamoxifen. The configuration of the (-)-isomer was shown to be S- by degradation of an ester to a derivative of (-)-2-hydroxy-1-phenyl-1-propanone, which has already been shown to have S-configuration. Metabolism of tamoxifen by rat liver microsomes gave equal amounts of the two enantiomers. They have the same chemical properties but, on treatment of rat hepatocytes in culture, R-(+)-alpha -hydroxytamoxifen gave at least eight times as many DNA adducts as the S-(-)-isomer.
Article
Objectives: To assess whether CYP2C9 alleles other than CYP2C9*2 and *3 are associated with a low-warfarin dose requirement and the relevance of upstream CYP2C9 polymorphisms to dose requirement and metabolism. Methods: CYP2C9 exons, intron-exon boundaries and 3 kb of upstream sequence in 20 patients requiring ≤ 1.5 mg warfarin per day and with apparently homozygous wild-type or heterozygous CYP2C9*2 genotypes were screened for novel polymorphisms by single-strand conformational polymorphism analysis. PCR-based genotyping assays for novel upstream and other known polymorphisms were used to screen a larger patient population of known CYP2C9*2 and *3 genotype requiring a range of warfarin doses. Results: Polymorphisms at eight different upstream sites were found, five of which were already described. We found that the majority of the upstream polymorphisms were in complete linkage disequilibrium with previously described coding region polymorphisms. However, two polymorphisms, T-1188C and the novel ΔG-2664ΔT-2665, occurred both in individuals who were otherwise wild-type and in individuals positive for coding region polymorphisms. Evidence for 11 haplotypes, including 8 with frequencies ≥ 0.01, was obtained. In individuals negative for coding region polymorphisms, neither individual genotypes for T-1188C or ΔG- 2664ΔT-2665 or particular combinations of haplotype pairs were predictive of dose requirement or S-warfarin total clearance, suggesting neither upstream polymorphism was functionally significant. Dose requirements in CYP2C9*11 heterozygotes were not statistically significantly different from homozygous wild-type individuals. Conclusions: The coding region non-synonymous polymorphisms associated with the CYP2C9*2 and CYP2C9*3 alleles are the major CYP2C9-related factor affecting warfarin dose in UK Caucasians. Upstream CYP2C9 polymorphisms do not appear to be important independent determinants of dose requirement.
Article
In many estrogen responsive systems the isomers of tamoxifen are known to have different biological character-the trans isomer is generally an antagonist and the cis isomer an agonist. Attempts to similarly characterize the isomers of hydroxytamoxifen (which differ greatly in their affinity for the estrogen receptor) are shown to be complicated by their facile isomerization. This isomerization was studied in cultures of estrogen receptor positive MCF-7 human breast cancer cells and monitored by HPLC under reversed phase conditions. Hydroxytamoxifen isomers that are initially 99% pure, undergo a time and temperature dependent isomerization, so that after 2 days in tissue culture medium at 37°C they have isomerized to the extent of 20%. This isomerization occurs in the cell-free medium alone and cannot be attributed to a metabolic conversion by the cells. The isomerization occurs much more slowly at 4 than at 37° C and can be reduced considerably by various antioxidants (butylated hydroxytoluene, ascorbate, α-tocopherol, retinoic acid and retinal); however, at concentrations that block isomerization, these antioxidants are toxic to the cells. Although the medium contains both the cis and trans isomers of hydroxytamoxifen, the MCF-7 cells preferentially accumulate the trans isomer and the material associated with the nuclear estrogen receptor is, in all cases, mainly the higher affinity trans isomer. A similar preference of the estrogen receptor for the trans isomer is seen with diethylstilbestrol, resulting again in almost exclusive accumulation of the trans isomer in the receptor binding site.