ChapterPDF Available

The role of pharmacogenomics in personalising the use of tamoxifen in breast cancer

Authors:

Abstract and Figures

Tamoxifen is a well-established endocrine therapy used in the treatment of hormonereceptor positive breast cancer. In recent years, its role in adjuvant and advanced breast cancer settings has been superseded by aromatase inhibitors. The prodrug tamoxifen is metabolised in the liver into several primary and secondary metabolites via cytochrome P450 enzymes. The main active forms are endoxifen and 4-hydroxytamoxifen, with CYP2D6 being a key enzyme in the production of both. There is evidence that tamoxifen-associated hot flushes are an indicator of clinical efficacy of tamoxifen. Co-prescription of CYP2D6 inhibitors, including selective serotonin re-uptake inhibitors (SSRIs) commonly used to treat hot flushes, may decrease the clinical efficacy of tamoxifen and should be used with caution. Moreover, there is evidence that patients with poor CYP2D6-mediated tamoxifen metaboliser phenotype ave worse clinical outcomes, and fewer hot flushes, than those with other phenotypes. Further research is necessary to clarify whether extensive metabolisers have the best clinical outcomes on tamoxifen. Preliminary data suggest that in individuals of certain CYP2D6 phenotype, amoxifen may have equivalent efficacy, and be considered as an alternative to aromatase inhibitors if there were concerns over the side effect profile and cost of the latter. Thus the role of tamoxifen may be redefined in a new era of personalised breast cancer treatment.
Content may be subject to copyright.
In: Tamoxifen: Properties, Biological Activity and Health Benefits ISBN
Editor: Editors Name, pp. © 2008 Nova Science Publishers, Inc.
Chapter
The role of pharmacogenomics in personalising the use of tamoxifen
in breast cancer
SJ Johnston1, LC Chen2, and KL Cheung1*
1Division of Breast Surgery and 2School of Pharmacy, University of Nottingham, UK
Abstract
Tamoxifen is a well-established endocrine therapy used in the treatment of hormone-
receptor positive breast cancer. In recent years, its role in adjuvant and advanced breast
cancer settings has been superseded by aromatase inhibitors. The prodrug tamoxifen is
metabolised in the liver into several primary and secondary metabolites via cytochrome
P450 enzymes. The main active forms are endoxifen and 4-hydroxytamoxifen, with
CYP2D6 being a key enzyme in the production of both. There is evidence that tamoxifen-
associated hot flushes are an indicator of clinical efficacy of tamoxifen. Co-prescription
of CYP2D6 inhibitors, including selective serotonin re-uptake inhibitors (SSRIs)
commonly used to treat hot flushes, may decrease the clinical efficacy of tamoxifen and
should be used with caution. Moreover, there is evidence that patients with poor
CYP2D6-mediated tamoxifen metaboliser phenotype have worse clinical outcomes, and
fewer hot flushes, than those with other phenotypes. Further research is necessary to
clarify whether extensive metabolisers have the best clinical outcomes on tamoxifen.
Preliminary data suggest that in individuals of certain CYP2D6 phenotype, tamoxifen
may have equivalent efficacy, and be considered as an alternative to aromatase inhibitors
if there were concerns over the side effect profile and cost of the latter. Thus the role of
tamoxifen may be redefined in a new era of personalised breast cancer treatment.
* E-mail address: kl.cheung@nottingham.ac.uk
SJ Johnston, LC Chen and KL Cheung
Introduction
With the introduction of tamoxifen in 19731 it became possible to personalise breast cancer
treatment according to oestrogen receptor (ER) status, as determined by an immunocytological
or histochemical technique. Selection of individual patients according to ER status for 5 years
treatment with tamoxifen following surgery for early breast cancer is made in the light of data
from worldwide randomised trials. Meta-analysis of these data indicates a reduction of around
50% in recurrence and 40% in breast cancer mortality in patients with ER positive tumours, but
no effect in patients with ER negative tumours2. For several decades, tamoxifen has been the first
line endocrine agent for adjuvant treatment and for post-menopausal women with hormone
receptor-positive advanced breast cancer. Tamoxifen is an effective and well-tolerated endocrine
treatment in these settings, but has side effects which are potentially serious, e.g. endometrial
proliferation and deep vein thrombosis, and more common side effects such as hot flushes, which
may impact adversely on quality of life and affect compliance to treatment. Moreover, between
30% and 50% of those taking adjuvant tamoxifen will eventually relapse or die of breast cancer,
which may in part be explained by tumour-dependent ER activation3. In recent years, tamoxifen
has been superseded by the third-generation aromatase inhibitors (AIs), anastrozole4,5,6,
letrozole7,8 and exemestane9, with evidence demonstrating a significant efficacy advantage for
AIs over tamoxifen. However, the magnitude of this benefit is not large: for example, long-term
follow-up of the Arimidex (anastrozole), Tamoxifen, Alone or in Combination (ATAC) trial10
demonstrated an absolute difference in disease-free survival of 4% at 100 months in favour of
anastrozole. The wide inter-individual variation in clinical effectiveness of tamoxifen may be
due to intrinsic host factors including phenotypic variation of tamoxifen metabolism into its
active components. This chapter outlines key developments over the last decade that have
increased knowledge of the metabolism of tamoxifen via cytochrome P450 enzymes and
explored links between pharmacogenetic factors and response to tamoxifen. The implications for
the role of pharmocogenetics in personalising the use of tamoxifen therapy in breast cancer are
then discussed, including the potential to enable clinicians to predict the benefits and risks of
tamoxifen therapy through testing for individual genes, and hence tailor the choice of hormone
treatment to the individual patient.
The role of pharmacogenomics in personalising the use of tamoxifen in breast cancer 3
Tamoxifen metabolism
Irrespective of selection for hormone receptor positivity, there remains wide inter-individual
variation in clinical response to tamoxifen, both in terms of efficacy and in relation to side
effects, which may be caused by variation in the metabolism of the prodrug, tamoxifen, into its
active components.
Tamoxifen is metabolised extensively in the liver into several primary and secondary metabolites
(Figure 1) via cytochrome P450 (CYP) enzymes. Studies on tamoxifen metabolites have
established that 4-hydroxytamoxifen and 4-hydroxy-N-desmethyltamoxifen (endoxifen) are the
main active therapeutic components, being of similar potency, with at least 100-fold greater
affinity to the ER, and 30- to 100-fold greater potency in the suppression of oestrogen-dependent
proliferation of breast cancer cells compared with the prodrug1.
The relative roles of endoxifen and 4-hydroxytamoxifen serum concentrations in determining the
response to tamoxifen therapy are unknown; however, the steady state concentration of
endoxifen in the plasma of patients receiving chronic tamoxifen therapy is five to ten times
higher than that of 4-hydroxytamoxifen 11 suggesting a major role for endoxifen. In-vitro
characterisation of tamoxifen sequential metabolism by CYP isoforms12 demonstrated that
CYP2D6 is the major enzyme involved in the formation of both endoxifen and 4-
hydroxytamoxifen, with a more dominant, rate limiting role in endoxifen formation via 4-
hydroxylation of the primary tamoxifen metabolite, N-desmethyltamoxifen.
Figure 1: Metabolic pathway of tamoxifen. The width of the arrow represents the relative
contribution of each pathway. [Adapted from Goetz et al, J. Clin. Oncol. 2005;23(36)9312-8]
Endoxifen
N-desmethyltamoxifen
CYP2D6
4-hydroxytamoxifen
Tamoxifen
CYP3A
CYP2C9
CYP2C9
CYP2C19
CYP2D6
CYP3A4
SJ Johnston, LC Chen and KL Cheung
CYP2D6 activity and effect on plasma endoxifen levels
Factors affecting CYP2D6 activity and thus tamoxifen metabolism in individuals on tamoxifen
may include genetic polymorphism of the enzyme, and concomitant drug interactions via
competitive inhibition (acting as substrate to the enzyme) and direct inhibition of enzyme
function.
The influence of CYP2D6 enzyme on plasma endoxifen levels during tamoxifen treatment was
demonstrated in three prospective clinical trials. In the first of these, Stearns et al13 measured the
plasma concentration of tamoxifen and its metabolites (N-desmethyl-tamoxifen, 4-
hydroxytamoxifen and endoxifen) 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. The trial found that the mean plasma concentration of endoxifen decreased by 56%
(p=0.02), whilst none of the other metabolites underwent statistically significant changes in
concentration. Analysis by CYP2D6 genotype indicated that baseline plasma endoxifen
concentration in those with variant genotype were lower, and that the reduction in endoxifen
concentration was evident primarily in women with wild-type compared to variant genotype
(64% versus 24%, p=0.03). Indeed, there was no statistical reduction in endoxifen concentration
for those with variant genotype.
Although this was a small preliminary study, the results raised the possibility that
pharmacogenetic variation in CYP2D6 activity may influence therapeutic outcomes from
tamoxifen treatment, with potential implications for CYP2D6 variant allele carriers, having
lower plasma endoxifen, and those with wild-type genotype, whose plasma endoxifen
concentrations may be reduced by concomitant enzyme inhibiting drugs.
Further evidence of the importance of CYP2D6 genotype and concomitant use of SSRIs was
subsequently presented in a larger prospective clinical trial14 of 80 patients with newly diagnosed
breast cancer beginning adjuvant tamoxifen therapy. Of these patients, 24 were taking CYP2D6
inhibiting drugs (SSRIs paroxetine (n=10), sertraline (n=4), citalopram (n=4), fluoxetine (n=2)
and the SNRI venlafaxine (n=3)). Patients were genotyped for common CYP2D6 and other
tamoxifen-metabolising enzyme (CYP2C9, CYP3A5 and sulfotransferase 1A1) alleles. The
primary outcome measure was plasma concentration of tamoxifen and its metabolites, after one
and four months of tamoxifen therapy. The study found that those with a CYP2D6 homozygous
wild-type genotype had statistically higher levels of plasma endoxifen after four months of
tamoxifen therapy compared to those with homozygous variant or heterozygous genotypes, and
that for homozygous wild-type genotype, plasma endoxifen concentration was reduced
substantially when co-administered with paroxetine. Of the other CYP2D6 inhibiting drugs,
venlafaxine was the weakest inhibitor and the effect of sertraline was intermediate between
paroxetine and venlafaxine, which is consistent with in vitro data (although data for the two
patients taking fluoxetine was not reported).
The role of pharmacogenomics in personalising the use of tamoxifen in breast cancer 5
The largest of these prospective trials was conducted by Borges et al15, in which 158 breast
cancer patients taking tamoxifen were genotyped for 33 CYP2D6 alleles and had medication
history taken. The outcome measure was plasma concentration of tamoxifen and its metabolites
at the fourth month of tamoxifen treatment. The study used a mixture model approach to define
phenotypic groups according to endoxifen/N-desmethyltamoxifen plasma concentration ratio,
identifying three distinct genotype groups in the distribution of this ratio (Table 1).
Phenotypic group
p-value
No functional CYP2D6 allele
<0.001
1 active CYP2D6 allele
2 or more active CYP2D6 allele
Table 1: Mixture model in Borges et al, (Clin. Pharmacol. Ther. 2006;80(1):61-74). “NDM” =
N-desmethyltamoxifen
The study also identified two phenotypic groups by plasma endoxifen concentration, and showed
that those with CYP2D6 extensive metaboliser phenotype taking potent CYP2D6 inhibitors had
significantly lower mean endoxifen plasma concentration than those who were not taking
CYP2D6 inhibitors (23.5 +/- 9.5 nmol/L versus 84.1 +/- 39.4 nmol/L, p<0.001).
Together, these three prospective clinical trials provided evidence that steady-state endoxifen
plasma concentrations during tamoxifen treatment were reduced in women that carry certain
CYP2D6 genetic variants, or who carry wild-type alleles and are co-prescribed CYP2D6
inhibitors, while the concentration of tamoxifen or other metabolites remained unaffected by
CYP2D6 metabolic status. Moreover, the plasma concentrations of endoxifen were on average
ten-fold higher than those of 4-hydroxytamoxifen, with a large degree of interpatient variability:
up to one hundred-fold greater endoxifen exposure relative to 4-hydroxytamoxifen in some
patients13,15,16.
Endoxifen and potential indicators of tamoxifen response
CYP2D6 phenotype
In order to determine whether changes in active metabolite concentration resulting from genetic
polymorphisms affect clinical outcomes of women receiving adjuvant tamoxifen, a prospective
adjuvant tamoxifen trial17 was used to explore the association between variation of tamoxifen
SJ Johnston, LC Chen and KL Cheung
metabolising genes and disease outcomes and toxicity in the form of hot flushes18. The study
determined the genotype of CYP2D6 (based on aforementioned preliminary study findings) and
CYP3A5 (selected because it has been shown to be the principal enzyme catalysing the
elimination of tamoxifen) from 223 patients in the adjuvant tamoxifen trial. Primary outcome
measures were relapse-free (RF) time, disease-free survival (DFS) and overall survival (OS), and
a secondary outcome measure was incidence of hot flushes. At 12 years of follow-up, for
CYP3A5 there was no difference in RF-time, DFS or OS between genotypes, whilst for
CYP2D6, unadjusted data indicated an advantage in RF-time and DFS, but not OS, for those
with CYP2D6*4*4 (homozygous variant) genotype compared to patients with one or no variant
alleles (Figure 2). However, Cox proportional hazards modelling found a significant association
between both nodal status and tumour size with RF-time, DFS and OS. Once adjustment had
been made for these associations, there was no statistically significant difference in hazard ratios
so it may have been more forthright for the article to display the Kaplan-Meier curves for the
adjusted rather than unadjusted data.
Figure 2: Kaplan-Meier estimates (approximate representation) of (I) relapse-free time; (II)
disease-free survival for unadjusted data, Wt (wild-type)/*4 or Wt/Wt vs. *4/*4 (adjusted data
hazard ratios not significant: p=0.176 and 0.089 respectively). [Adapted from Goetz et al,
2005;23(36):9312-8]
Similarly for the secondary outcome measure, the incidence of hot flushes did not vary by
CYP3A5 genotype, but there was a non-significant lower incidence of hot flushes in the
homozygous variant CYP2D6 group compared to those with wild-type alleles (Table 2).
0
10
20
30
40
50
60
70
80
90
100
0 2 4 6 8 10 12
Percentage
Years after random assignment
(I)
One or no variant alleles
Homozygous variant
0
10
20
30
40
50
60
70
80
90
100
0 2 4 6 8 10 12
Percentage
Years after random assignment
(II)
p=0.030†
p=0.020†
The role of pharmacogenomics in personalising the use of tamoxifen in breast cancer 7
CYP2D6 genotype
Total
patients
Patients with moderate or severe hot flushes
%
Homozygous variant
13
0
Heterozygous wild-type
40
23
Homozygous wild-type
137
20
Table 2: Incidence of moderate or severe hot flushes by CYP2D6 genotype
Co-prescription of SSRIs
Goetz et al19 then undertook a retrospective review of the medical records of the patients in the
North Central Cancer Treatment Group adjuvant tamoxifen trial17, from which they defined
metaboliser status based on a combination of CYP2D6 genotype and inhibition via co-prescribed
medications (classified as moderate or potent inhibitors). From this information, they classified
patients into extensive metabolisers (homozygous wild-type genotype not co-prescribed a
CYP2D6 inhibitor) or decreased metabolisers, further classified into intermediate or poor
metabolisers based on the presence of one or two CYP2D6 variant (*4) alleles or the co-
prescription of a moderate or potent CYP2D6 inhibitor. Outcome measures were breast cancer
recurrence and death.
By defining the patients in this manner, the study found that those with decreased metabolism
(n=65) had significantly shorter time to recurrence (HR=1.91, p=0.034) and worse relapse-free
survival (HR=1.74, p=0.017) relative to patients with extensive metabolism (n=115), and that
compared to extensive metabolisers, poor metabolisers had the most significant risk of breast
cancer relapse (HR 3.12, p=0.007).
Thus the study concludes that CYP2D6 metabolism as measured by genetic variation and
enzyme inhibition is an independent predictor of breast cancer outcome in post-menopausal
women receiving tamoxifen for early breast cancer although the “measurement” of CYP2D6
metabolism in this study is based on several layers of classification of retrospective data, with the
effect of moderate and potent enzyme inhibitors (co-prescribed in 13 patients) being somewhat
indeterminate.
SJ Johnston, LC Chen and KL Cheung
Hot flushes
The evidence of increased risk of disease relapse and fewer hot flushes with those with decreased
CYP2D6 mediated metabolism, along with poorer outcome with co-administration of SSRIs in
patients receiving adjuvant tamoxifen therapy, was presented as an a priori” hypothesis20 at the
Annual Scientific Meeting of the American Society of Clinical Oncology in 2007: that hot
flushes themselves are an indicator of tamoxifen efficacy, and that the absence of hot flushes
may be an indication of poor prognosis.
The evidence for this was taken from the Women’s Healthy Eating and Living (WHEL) study21,
a large randomised trial of the effect of a major increase in vegetable consumption on future
breast cancer events among breast cancer survivors. Between 1995 and 2000, the WHEL study
recruited 3088 women aged 18-70 years, diagnosed (within 2-48 months) with stage I (T1c) to
III breast cancer, and randomly assigned subjects to a WHEL healthy cancer prevention diet or
control arm (National Cancer Institute based diet). Baseline information included dietary intake,
menopause status and anti-oestrogen use, and the study recorded information about cancer
pathology and treatment, along with patient questionnaire data on symptoms.
Retrospective analysis was then performed on the control arm (n=1551) to explore links between
tamoxifen use, hot flushes and breast cancer events. Unadjusted cancer event proportions for hot
flushes were compared to those without. The average time after study entry was 7.3 years, and
there were 864 patients on tamoxifen in the analysis. At 7.3 years of follow-up, the study found
that women with hot flushes had fewer breast cancer specific events (locoregional or
contralateral recurrence, or metastatic disease) than those without hot flushes (12.9% vs. 21%,
p=0.01), and that recurrence-free survival was better for those who reported hot flushes (figure
3).
The role of pharmacogenomics in personalising the use of tamoxifen in breast cancer 9
Figure 3: Kaplan-Meier recurrence-free survival (approximate representation) as a function of
reported hot flashes in patients on tamoxifen (n=864) [Adapted from ASCO Proceedings,
Chicago, 1-5 June 2007]
The study found that hot flushes were more predictive of breast cancer specific outcomes than
age, hormone receptor status or stage I vs. II disease. Thus a relationship between hot flushes and
efficacy of adjuvant tamoxifen was suggested.
In summary, the evidence from trials looking at clinical outcomes according to variously defined
metaboliser phenotypic status has identified CYP2D6 genotype, individually and in conjunction
with SSRI co-prescription, as a potential predictive factor of tamoxifen response. The proposed
mechanism, as suggested by prior clinical trials, is CYP2D6-mediated metabolism of the prodrug
tamoxifen to endoxifen. Furthermore, there is evidence that hot flushes themselves might
indicate an efficacious, endoxifen-mediated response to tamoxifen therapy.
Association between CYP2D6 genetic variation and tamoxifen response
A further study attempted to define the predictive value of genetic variants of CYP enzymes
including CYP2D6 for tamoxifen treatment outcome22 . The study was a non-randomised,
retrospective analysis of 486 ER-positive postmenopausal breast cancer patients, with 206
patients taking adjuvant tamoxifen compared to a control population of 280 patients not taking
tamoxifen, using archival material. Patients receiving adjuvant tamoxifen and concomitant
0
10
20
30
40
50
60
70
80
90
100
0 1 2 3 4 5 6 7 8
Percentage
Years after random assignment
Recurrence-free survival
Hot flushes
No hot flushes
p=0.003
SJ Johnston, LC Chen and KL Cheung
chemotherapy or patients with an unclear ER status (n=135) were excluded from the analysis.
Information on co-prescription of SSRIs was incomplete and also excluded from analysis.
Outcome measures were relapse-free time (time from surgery to locoregional recurrence or
contralateral disease), event-free survival (time to relapse or death from any cause) and overall
survival.
At a median follow-up of 71 months, carriers of CYP2D6*4, *5, *10 and *41 alleles, with
phenotypes defined as poor and intermediate metabolisers, had significantly more breast cancer
recurrences, shorter relapse-free times, and worse event-free survival than carriers of functional
alleles (Figure 4). No such association was found for the control population.
Figure 4: Kaplan-Meier probabilities (approximate representation) of relapse-free time (RFT) of
breast cancer patients on adjuvant tamoxifen for CYP2D6-metaboliser phenotypes. [Adapted
from Scroth et al J. Clin. Oncol. 2007;25(33):5187-93]
TAM=patients on tamoxifen, NoTAM= patients not on tamoxifen, EM=extensive metabolisers,
hetEM=heterozygous EM, IM=intermediate metabolisers, PM=poor metabolisers
The authors concluded by suggesting that genetic testing for CYP2D6 status could be used to
refine the choice and / or sequencing of hormonal therapy in prospective clinical trials, and
identified a potential role for pharmacogenetics, with upfront genetic testing of CYP2D6
genotype, to predicting response to tamoxifen.
40
50
60
70
80
90
100
0 20 40 60 80 100 120
%
Relapse-free time
A. TAM (N=206)
EM
PM or IM
hetEM
40
50
60
70
80
90
100
0 20 40 60 80 100 120
%
Relapse-free time
B. NoTAM (N=280)
p=0.04
p=0.37
The role of pharmacogenomics in personalising the use of tamoxifen in breast cancer 11
Implications
Research into pharmacogenetics of tamoxifen therapy has identified CYP2D6 as a key enzyme
in the metabolism of tamoxifen, with a critical role in the formation of endoxifen. Along with 4-
hydroxytamoxifen, endoxifen is a major active metabolite of the prodrug. In women treated with
tamoxifen, plasma endoxifen levels are on average five to ten times higher than those of 4-
hydroxytamoxifen, with wide inter-individual variation, suggesting a dominant active role for
endoxifen determined via genetic variation in CYP2D6. By defining CYP2D6 phenotypes such
as extensive and poor tamoxifen metabolisers based on genotype and the co-prescription of
known CYP2D6 inhibitors such as SSRIs that are commonly used to treat hot flushes, evidence
is presented of an association between pharmacogenetic factors and clinical response to
tamoxifen. This implies that CYP2D6 genotype could be used in upfront genetic testing to help
clinicians weigh up the potential benefits versus the risks of tamoxifen therapy for the individual
patient.
Research has also asked questions about the practice of prescribing SSRIs to treat tamoxifen-
associated hot flushes, and found evidence that SSRIs, which are both competitive and direct
inhibitors of CYP2D6, contribute to decreased tamoxifen metaboliser status with an associated
diminished clinical response to tamoxifen. Alongside evidence that hot flushes are associated
with fewer breast cancer events, the implication is that hot flushes indicate an efficacious clinical
response to tamoxifen, via CYP2D6-mediated metabolism into endoxifen and SSRIs should
therefore be avoided, or at least used with caution, in patients treated with tamoxifen. Given that
hot flushes may be interpreted as a surrogate of adequate endoxifen levels in the body, patients
could be reassured that with the development of hot flushes, they are likely to be deriving
clinical benefit from tamoxifen.
Whilst aromatase inhibitors have replaced tamoxifen as the first line endocrine therapy in the
adjuvant breast cancer setting for postmenopausal women, the absolute DFS advantage is small;
thus the subgroup of patients who are extensive CYP2D6 metabolisers (e.g. homozygous wild
type alleles) may benefit more from tamoxifen therapy. Whether or not pharmacogenomics could
be used to tailor therapy to the individual would require prospective clinical trials to test whether
individual genes such as CYP2D6 can be used to predict outcomes. In the meantime, genetic
testing could be applied on an individual basis, for example if there are concerns over toxicities
of aromatase inhibitors such as osteoporotic risk, in which case tamoxifen may be preferable and
of similar efficacy for individuals with CYP2D6 homozygous wild-type genotype. With
improved knowledge of global and ethnic variation in CYP2D6 genotype, the application of
pharmacogenomics could be targeted to certain groups in order to make more informed decisions
in tailoring endocrine therapy. Thus a new role for tamoxifen may emerge in a new era of
personalised medicine.
SJ Johnston, LC Chen and KL Cheung
References
1 Cole M, Jones C, Todd I: A new antioestrogenic agent in late breast cancer. An early appraisal
of ICI 46,474. Br J Cancer 1971;25:270-5
2 Early Breast Cancer Trialists' Collaborative Group: Effects of chemotherapy and hormonal
therapy for early breast cancer on recurrence and 15-year survival: an overview of the
randomised trials. Lancet. 2005 May 14-20;365(9472):1687-717
3 Ring A and Dowsett M: Mechanisms of tamoxifen resistance. Endocr. Rel. Cancer. 11:643-58
4 Bonneterre J, Thurlimann D, Robertson J, et al. Anastrozole versus tamoxifen as first-line
therapy for advanced breast cancer in 668 postmenopausal women: results of the Tamoxifen
or Arimidex randomised group efficacy and tolerability study. J. Clin. Oncol. 2003;39:2310-7
5 Nabholtz J, Buzdar A, Pollak M, et al. Anastrozole is superior to tamoxifen as first-line
therapy for advanced breast cancer in postmenopausal women: results of a North American
multicenter randomised trial. J. Clin. Oncol. 2000;18:3758-67
6 Milla-Santos A, Milla L, Portella J, et al. Anastrozole versus tamoxifen as first-line therapy in
postmenopausal patients with hormone-dependent advanced breast cancer: a prospective,
randomised, phase III study. Am. J. Clin. Oncol. 2003;26:317-22
7 Mourisden H, Gershanovich M, Sun Y, et al. Superior efficacy of letrozole versus tamoxifen as
first-line therapy for post-menopausal women with advanced breast cancer: results of a phase
III study of the International Letrozole Breast Cancer Group. J. Clin. Oncol. 2001;19:2596-
606
8 Eirmann W, Paepke S, Appfelstaedt J, et al. Letrozole Neo-Adjuvant Breast Cancer Study
Group: Preoperative treatment of postmenopausal breast cancer patients with letrozole: a
randomised double-blind multicenter study. Ann Oncol. 2001;12:1527-32
9 Paridaens R, Dirix L, Lohrisch C, et al. European Organsiation for the Research and Treatment
of Cancer (EORTC) Investigational Drug Branch for Breast Cancer (IDBBC). Mature
results of a randomised phase II multicenter study of exemestane versus tamoxifen as first-
line hormone therapy for post-menopausal women with metastatic breast cancer. Ann. Oncol.
2003;14:1391-8
10 Forbes JF, Cuzick J, Buzdar A, et al. Effect of anastrozole and tamoxifen as adjuvant treatment
for early-stage breast cancer: 100-month analysis of the atac trial. Lancet Oncol. 2008;9:45-53
11 Lee KH, Ward BA, Desta Z, et al: Quantification of tamoxifen and three metabolites in plasma
by high-performance liquid chromatography with fluorescence detection: application to a
clinical trial. J. Chromatogr. B. Analyt. Technol. Biomed. Life Sci. 2003;791:245-53
The role of pharmacogenomics in personalising the use of tamoxifen in breast cancer 13
12 Desta Z, Ward BA, Soukhova NV, et al: Comprehensive evaluation of tamoxifen sequential
biotransformation by the human cytochrome P450 system in vitro: prominent roles for
CYP3A and CYP2D6. J. Pharmacol. Exp. Ther. 2004;310:1062-75
13 Stearns V, Johnson MD, Rae JM, et al: Active tamoxifen metabolite plasma concentrations
after coadministration of tamoxifen and the selective serotonin reuptake inhibitor paroxetine.
J. Natl. Cancer Inst. 2003;95(23):1758-64
14 Jin Y, Desta Z, Stearns V, et al: CYP2D6 genotype, antidepressant use, and tamoxifen
metabolism during adjuvant breast cancer treatment. J. Natl. Cancer Inst. 2005;97(1):30-9
15 Borges S, Desta Z, Li L, et al: Quantitative effect of CYP2D6 genotype and inhibitors on
tamoxifen metabolism: implication for optimization of breast cancer treatment. Clin.
Pharmacol. Ther. 2006;80(1):61-74
16 Desta Z, Flockhart DA: Germline pharmacogenetics of tamoxifen response: have we learned
enough? J. Clin. Oncol. 2007;25(33):5147-9 (editorial)
17 Ingle J, Suman V, Mailliard J, et al: Randomized trial of tamoxifen alone or combined with
fluoxymesterone as adjuvant therapy in postmenopausal women with resected primary breast
cancer: A North Central Cancer Treatment Group Study. Proc. Am. Soc. Clin. Oncol.
2000;19:86a (abstract)
18 Goetz MP, Rae JM, Suman VJ, et al: Pharmacogenetics of tamoxifen biotransformation is
associated with clinical outcomes of efficacy and hot flashes. J. Clin. Oncol.
2005;23(36):9312-8
19 Goetz MP, Knox SK, Suman VJ, et al: The impact of cytochrome P450 2D6 metabolism in
women receiving adjuvant tamoxifen. Breast Cancer Res. Treat. 2007;101(1):113-21. Epub
2006 Nov 18
20 Mortimer JE, Flatt SW, Parker BA, et al: Tamoxifen, hot flashes and recurrence in breast
cancer: Support for pharmacogenetics. J. Clin. Oncol. 2007;25(18S):500 (abstract)
21 Pierce JP, Natarajan L, Caan BJ, et al: Influence of a diet very high in vegetables, fruit, and
fiber and low in fat on prognosis following treatment for breast cancer: the Women's Healthy
Eating and Living (WHEL) randomized trial. JAMA. 2007;298(3):289-98.
22 Scroth W, Antoniadou L, Fritz P, et al: Breast cancer treatment outcome with adjuvant
tamoxifen relative to patient CYP2D6 and CYP2C19 genotypes. J. Clin. Oncol.
2007;25(33):5187-93
... In the future, decisions on agent of choice may be assisted by the use of pharmocogenetic profiling of individual patients. For example, patients with homozygous wild-type CYP2D6 alleles have been shown to be extensive metabolizers of tamoxifen into its active form, endoxifen, and thus may benefit more from tamoxifen than aromatase inhibitors [26]. However, this concept remains to be proven in prospective clinical trials. ...
Article
ABSTRACT A Cochrane review of randomized trials shows no difference in overall survival between surgery and primary endocrine therapy (PET) in older women with operable primary breast cancer. Most of these trials were small and unselected for estrogen receptor (ER) status. Evidence exists showing a significant correlation between the degree of ER-positivity and response and outcome in patients receiving PET. Although surgery remains the treatment of choice, patients with ER-rich tumors tend to do equally well on PET. When deciding optimal therapies, co-morbidities and frailty (which impact on the likelihood of death due to competing causes), patient choice, agent of choice (notably the third-generation aromatase inhibitors) and biology (more than just being ER-positive) should all be taken into account.
Article
Full-text available
Across the translational research continuum from biomarker discovery to public health research, “costeffectiveness” considerations are crucial, and can significantly impact the adoption of personalized medicine innovations. Cost-effectiveness is concerned with providing evidence to compare the (economic) costs and the health outcomes of competing health interventions or technologies. This also affects translational research in all stages, including clinical trials, post-market monitoring and population health outcome assessment. Indeed, economic considerations are important in determining the development and diffusion of a new technology in any scientific field. This is particularly true in healthrelated sectors, wherein governments and regulatory agencies with a mandate and commitment to efficient and rational allocation of resources require transparent and rigorous economic evidence to support or decline the adoption of a new technology. In the context of personalized medicine and theragnostics (i.e., the fusion of therapeutics and diagnostics), the use of genomics in clinical practice can be markedly facilitated by tandem evaluation of the clinical benefits/risks of customized health interventions and their cost-effectiveness. This paper provides a synthesis of the past and emerging literature on cost-effectiveness studies that evaluate pharmacogenetics tests. We conclude that despite the recent efforts, there is still a scarcity of convincing evidence on the cost-effectiveness of genomics products that creates a barrier in the uptake of pharmacogenetics in personalized medicine. Additionally, the reasons that limit a wider development of the cost-effectiveness analyses in this field are discussed, with a view to amend the above translational gaps in the literature.
Article
Full-text available
Pharmacogenomics aims to use molecular genetic markers to predict treatment outcome. Indeed, within the past decade there has been a rapid emergence of pharmacogenetic tests to aid clinicians in predicting efficacy or toxicity for some drugs. Despite this major advance in therapeutic drug management, there remain challenges to the appropriate use of pharmacogenetic tests. We discuss UGT1A1 pharmacogenetic testing to illustrate the knowledge gaps impeding widespread use of pharmacogenetic tests in the clinical setting.
Article
Full-text available
Dissecting the genetic basis of disease risk requires measuring all forms of genetic variation, including SNPs and copy number variants (CNVs), and is enabled by accurate maps of their locations, frequencies and population-genetic properties. We designed a hybrid genotyping array (Affymetrix SNP 6.0) to simultaneously measure 906,600 SNPs and copy number at 1.8 million genomic locations. By characterizing 270 HapMap samples, we developed a map of human CNV (at 2-kb breakpoint resolution) informed by integer genotypes for 1,320 copy number polymorphisms (CNPs) that segregate at an allele frequency >1%. More than 80% of the sequence in previously reported CNV regions fell outside our estimated CNV boundaries, indicating that large (>100 kb) CNVs affect much less of the genome than initially reported. Approximately 80% of observed copy number differences between pairs of individuals were due to common CNPs with an allele frequency >5%, and more than 99% derived from inheritance rather than new mutation. Most common, diallelic CNPs were in strong linkage disequilibrium with SNPs, and most low-frequency CNVs segregated on specific SNP haplotypes.
Article
Full-text available
An introductory clinical trial of the anti-oestrogenic agent IC146474 in late or recurrent carcinoma of the breast is described. Forty-six patients have been treated, of whom 10 have shown a good response. This is of the same order as that seen with oestrogens and androgens. The particular advantage of this drug is the low incidence of troublesome side effects.
Article
Full-text available
The efficacy and tolerability of anastrozole (Arimidex; AstraZeneca, Wilmington, DE, and Macclesfield, United Kingdom) and tamoxifen were compared as first-line therapy for advanced breast cancer in 353 postmenopausal women. The randomized, double-blind, multicenter study was designed to evaluate anastrozole 1 mg once daily relative to tamoxifen 20 mg once daily in patients with hormone receptor-positive tumors or tumors of unknown receptor status who were eligible for endocrine therapy. Primary end points were objective response (OR), defined as complete (CR) or partial (PR) response, time to progression (TTP), and tolerability. Anastrozole was as effective as tamoxifen in terms of OR (21% v 17% of patients, respectively), with clinical benefit (CR + PR + stabilization > or = 24 weeks) observed in 59% of patients on anastrozole and 46% on tamoxifen (two-sided P =.0098, retrospective analysis). Anastrozole had a significant advantage over tamoxifen in terms of TTP (median TTP of 11.1 and 5.6 months for anastrozole and tamoxifen, respectively; two-sided P =.005). The tamoxifen:anastrozole hazards ratio was 1.44 (lower one-sided 95% confidence limit, 1.16). Both treatments were well tolerated. However, thromboembolic events and vaginal bleeding were reported in fewer patients who received anastrozole compared with those who received tamoxifen (4.1% v 8.2% [thromboembolic events] and 1.2% v 3.8% [vaginal bleeding], respectively). Anastrozole satisfied the predefined criteria for equivalence to tamoxifen. Furthermore, we observed both a significant increase in TTP and a lower incidence of thromboembolic events and vaginal bleeding with anastrozole. These findings indicate that anastrozole should be considered as first-line therapy for postmenopausal women with advanced breast cancer.
Article
To review the labels of United States Food and Drug Administration (FDA)-approved drugs to identify those that contain pharmacogenomic biomarker information, and to collect prevalence information on the use of those drugs for which pharmacogenomic information is included in the drug labeling. Retrospective analysis. The Physicians' Desk Reference Web site, Drugs@FDA Web site, and manufacturers' Web sites were used to identify drug labels containing pharmacogenomic information, and the prescription claims database of a large pharmacy benefits manager (insuring > 55 million individuals in the United States) was used to obtain drug utilization data. Pharmacogenomic biomarkers were defined, FDA-approved drug labels containing this information were identified, and utilization of these drugs was determined. Of 1200 drug labels reviewed for the years 1945-2005, 121 drug labels contained pharmacogenomic information based on a key word search and follow-up screening. Of those, 69 labels referred to human genomic biomarkers, and 52 referred to microbial genomic biomarkers. Of the labels referring to human biomarkers, 43 (62%) pertained to polymorphisms in cytochrome P450 (CYP) enzyme metabolism, with CYP2D6 being most common. Of 36.1 million patients whose prescriptions were processed by a large pharmacy benefits manager in 2006, about 8.8 million (24.3%) received one or more drugs with human genomic biomarker information in the drug label. Nearly one fourth of all outpatients received one or more drugs that have pharmacogenomic information in the label for that drug. The incorporation and appropriate use of pharmacogenomic information in drug labels should be tested for its ability to improve drug use and safety in the United States.
Article
The basic concepts of pharmacogenetics, pharmacogenetic study approaches, factors to consider when applying pharmacogenetic discoveries to patient care, and potential roles for pharmacists in pharmacogenetics are discussed. The Food and Drug Administration (FDA) has recognized pharmacogenomics as an opportunity to identify new biomarkers that may expedite the drug development process. Currently, there are over 50 drugs with pharmacogenetic discoveries on their labeling. Sequence variations in drug disposition genes can alter the pharmacokinetics of a drug, while sequence variations in drug target genes can change the pharmacodynamics of the drug. The two most common strategies to test a pharmacogenetic question are the candidate-gene approach and genomewide association study. Given the complex interplay among the many factors that influence a drug dose, determination of an appropriate dose of a particular drug for a given patient will eventually require knowledge about both genetic and nongenetic factors that affect drug disposition and pharmacodynamics. Many factors can influence the application of pharmacogenetic discoveries to patient care. Before these discoveries find widespread application in clinical practice, additional work is needed, including randomized clinical trials to evaluate the clinical utility of a pharmacogenetic test, the development of guidelines for the clinical use of various pharmacogenetic tests, and provider education on pharmacogenetics. Pharmacogenetics has made significant progress in the past decade, and many pharmacogenetic discoveries have now been included on FDA-approved drug labeling. Pharmacogenetic discoveries may further promote safe and effective use of medications by more accurately predicting an individual's drug response.
Article
Before pharmacogenetic (PGx) testing has a major impact on clinical practice, two levels of evidence must be generated. First, studies demonstrating the links between genetic variation and response to medications in defined populations are needed, along with development of valid tests to measure these specific variants. Second, studies should be conducted to evaluate whether PGx testing improves health outcomes for patients and whether the decision to test is cost-effective relative to usual care. This latter set of questions is typically of greatest relevance to clinicians and payers, the ultimate gatekeepers for the clinical integration of pharmacogenetics. To date, nearly all of the research efforts and funding for PGx have been focused on the first set of issues-getting the science right. However, now is the time to increase our research efforts on the second set of issues-to improve the PGx evidence base for both clinical and economic decision making.