Advances in Adjuvant Hormonal Therapy for Postmenopausal Women
Kathrin Strasser-Weippl, Paul E. Goss
Journal of Clinical Oncology, Vol 23, No
8 (March 10), 2005: pp. 1751-1759
Since George Beatson
produced remissions of advanced breast cancer by performing
bilateral oophorectomy in premenopausal patients more than 100
years ago, the effect of antihormonal therapy on breast cancer
has been known. Initial endocrine therapy included oophorectomy,
hypophysectomy, and adrenalectomy. The latter two were
abandoned when tamoxifen citrate was approved by the US Food
and Drug Administration for use in advanced breast cancer in
1978. Selective estrogen receptor modulators (SERMs), such as
tamoxifen, antagonize estrogen receptor (ER) function by
binding competitively to it. Since 1978, more than 40,000
breast cancer patients, most of them postmenopausal, have been
treated with tamoxifen, either in the context of clinical
research studies or in clinical practice.The available data on the use of
tamoxifen in the adjuvant setting of ER-positive breast cancer
have been extensively reviewed by the Early Breast Cancer
Trialists' Collaborative Group (EBCTCG).In their most recent meta-analysis including 37,000 women,
adjuvant
treatment with 5 years of
tamoxifen resulted in a reduction
of 47% in disease recurrence, and a mortality reduction of 26%.The benefit of tamoxifen was restricted to the group of women
with ER-positive tumors. In this meta-analysis, 5 years of tamoxifen
therapy was better than 1 or 2 years of treatment, and the benefit
from 5 years persisted through 10 years of follow-up. In the
EBCTCG overview, the relative benefit from tamoxifen was independent
of axillary lymph node involvement, age, tamoxifen dose, menopausal
status, or use of chemotherapy. Based on these data, the 2000
US National Institutes of Health Consensus Development Conference
recommended the use of adjuvant tamoxifen therapy for women
"regardless of age, menopausal status, involvement of axillary
nodes, or tumor size."
The 2001 St Gallen Consensus Panel came to a similar
conclusion, suggesting the use of tamoxifen in most women with
ER-positive breast cancer unless the patient has an absolute
contraindication.
In both consensus statements, it was pointed out that for women
with low-risk, hormone-responsive disease, no adjuvant medical
treatment is an alternative choice to tamoxifen.
When 1,172 women who had completed 5 years of adjuvant tamoxifen
within the National Surgical Adjuvant Breast and Bowel Project
(NSABP) B-14 adjuvant study were randomly assigned to a further
5 years of tamoxifen or placebo,
no additional advantage was
obtained from extending tamoxifen beyond 5 years.
In fact, prolonged tamoxifen conferred a worse prognosis than
discontinuing therapy after 5 years. This might, in part, be
explained by the fact that tamoxifen has partial agonist
effects on the ER. In the preclinical setting, it was shown
that tamoxifen resistance is acquired if MCF-7 cells are
cultured with tamoxifen for a prolonged period.
In the clinical setting, tamoxifen's agonistic action on the ER
may even lead to growth stimulation of the tumor after 5 years.
For example, it was shown in nude mice that long-term exposure
to tamoxifen causes MCF-7 cells to grow in response to either
tamoxifen or low doses of estrogen.These demonstrable resistance/dependence mechanisms, together
with the clinical observations, have lead to the current
recommendation of restricting tamoxifen use to 5 years outside
of clinical trials. The ongoing ATLAS (Adjuvant Tamoxifen—Longer
Against Shorter) and aTTom (adjuvant Tamoxifen Treatment offer
more?) studies, in which patients are randomized to 5 versus
more than 5 years of adjuvant tamoxifen, will help to clarify
the optimal duration of tamoxifen therapy.
In contrast to SERMs, aromatase inhibitors (AIs) work by blocking
the enzyme complex responsible for the final step in estrogen
synthesis, aromatase, thus preventing the production of the
substrate of the ER. Therefore, unlike tamoxifen, the AIs have
no partial agonist activity. In postmenopausal women, all of
the third generation AIs suppress circulating estrogen levels
by approximately 98%.
In contrast to the second generation inhibitors (eg, formestane),
they are highly specific with almost no effect on cortisol or
aldosterone levels. In addition to the suppression of
circulating estrogen levels, the AIs also have the potential to
abrogate autocrine and paracrine estrogen production by
peritumoral stromal cells in both primary and metastatic sites
of the disease.Evidence suggests that this local estrogen production plays an
important role in tumor growth.
AIs can be classified by their mechanism of action into steroidal
(irreversible, type I) and nonsteroidal (reversible, type II)
inhibitors.
The inhibitors in clinical use today include the
third-generation nonsteroidal agents anastrozole and letrozole
and the third-generation steroidal aromatase inactivator exemestane.
Their pharmacokinetic profiles are similar, with anastrozole
and letrozole having longer half-lives (48 hours) than exemestane
(27 hours).All of them are administered orally once daily. Pharmacologic
interactions of tamoxifen have been demonstrated in combination
with both letrozole and anastrozole,
and while suppression of circulating estrogen is unaffected,
the therapeutic consequences of this interaction are unknown.
Efficacy of Aromatase Inhibitors
New therapeutic approaches to adjuvant therapy should address
two issues. First, is there a strategy rendering 5 years of
adjuvant treatment more effective by adding to, or substituting
tamoxifen with, AIs? And second, can adjuvant treatment be improved
by extending its duration, and administering AIs after tamoxifen?
At least 10 adjuvant trials of AIs, with a total of over 40,000
women with primary breast cancer, are currently addressing these
issues. Results of three trials are available to date and will
be discussed in the following sections.
Improving Adjuvant Therapy Within the First 5 Years
Numerous large trials are evaluating the use of AIs instead of
tamoxifen, or in combination or in sequence with tamoxifen,
during the first 5 postoperative years. Results from the first and largest of these studies,
the Arimidex, Tamoxifen, Alone or in Combination (ATAC) trial
(N = 9,366), were published in 2002 and updated in 2003.
When anastrozole was compared with tamoxifen for 5 years, the
AI led to improved disease-free survival (hazard ratio [HR],
0.86; P = .03) and time to recurrence (HR, 0.83; P = .015)
after a median follow-up of 47 months. There was also a lower incidence of contralateral breast cancer
with anastrozole (HR, 0.62; P = .062), reaching statistical
significance in the ER-positive subgroup (HR, 0.56; P =
.042). To date, there is no difference in the rates of death
from any cause or of breast cancer–related deaths. Based on the
published results from ATAC, the 2003 St Gallen consensus panel
included the option of giving anastrozole to postmenopausal
women in the adjuvant breast cancer setting "if tamoxifen is
contraindicated" in their recommendations.
A similar recommendation was published by the American Society
of Clinical Oncology Technology assessment.
In the updated position statement in 2005, the panel recommends
that adjuvant hormonal therapy for postmenopausal women with
hormone receptor–positive breast cancer should "include an AI
as initial therapy or after treatment or sequential therapy
consisting of tamoxifen (for either 2 to 3 years or 5 years)
followed by AIs for 2 to 3 years or 5 years.
Data from a trial (Intergroup Exemestane Study [IES]) employing
exemestane within the first 5 years after breast cancer diagnosis
were published recently.
After 2 to 3 years of adjuvant tamoxifen, 4,742
women were
assigned to either tamoxifen or exemestane for the remainder of
the 5 years. After a median follow-up of 30.6 months, the HR
for breast cancer recurrence in the exemestane group compared
with tamoxifen was 0.68 (P < .001), reflecting an
absolute benefit in disease-free survival of 4.7% after 3
years. Up to this point of follow-up, no difference in survival
was noted, but as 90% of patients had completed their 5 years
of therapy at the time of unblinding, assessing survival should
be possible with longer follow-up. As yet, the results from
this study have not influenced treatment recommendations in
early-stage breast cancer, because long-term toxicity data are
not yet available. However, for women finding adjuvant tamoxifen
difficult to tolerate after 2 to 3 years, continuation of adjuvant
treatment with exemestane might be an option.
A direct comparison of each of the three AIs for 5 years versus
tamoxifen for 5 years will be available in a few years. For
anastrozole, superiority to tamoxifen has already been demonstrated.
Furthermore, the sequence of an AI after 2 to 3 years of tamoxifen
has also been studied with all three agents. Data coming from
the IES look promising. The inverse sequence, tamoxifen given
after 2 to 3 years of an AI, will only be looked at in the BIGFEMTA
trial with letrozole, making this study particularly important.
To date, it can be said that AIs as monotherapy included in
a 5-year adjuvant hormonal regimen seem to improve efficacy,
and possibly, tolerability. However, as the optimal duration
of adjuvant therapy is not known, other treatment regimens including
AIs might still be superior.
Improving Adjuvant Therapy Beyond 5 Years
It is known that approximately half of all breast cancer recurrences
in women with ER-positive tumors taking 5 years of adjuvant
tamoxifen occur between 5 to 15 years after surgery, and that
the risk of recurrence appears to continue indefinitely.Based on this, there is a rationale for extending adjuvant endocrine
therapy beyond 5 years of tamoxifen with an AI.
Results from the first large study employing an AI after 5 years
of tamoxifen were published recently.
After having completed 4.5 to 6 years of prior adjuvant
tamoxifen, a total of 5,187 women were randomly assigned to a
further 5 years of letrozole 2.5 mg daily or placebo. After a
median follow-up of 2.4 years, when the first interim analysis
was conducted, the independent data and safety monitoring
committee recommended termination of the trial and prompt
communication of the results, because
women on letrozole had a
significantly superior disease-free survival (93% v 87%; P < .001), and in order to offer women taking placebo an
opportunity to take letrozole. The HR for local or metastatic
recurrence or new contralateral breast cancer in the letrozole
group was 0.57 (P = .00008) compared with placebo. There
was also a trend towards improved 4-year overall survival for
women receiving letrozole (96% v 94%), but this was not
statistically significant at the time of the first interim analysis.
The data from MA.17 show that extending adjuvant endocrine therapy
beyond 5 years with an AI offers significant benefit in disease-free
survival. The unique opportunity to re-randomize MA.17 patients
who complete 5 years of letrozole to a further 5 years or placebo
is now being undertaken. This will allow duration of efficacy
and toxicity to be further evaluated. The second trial testing
an AI after tamoxifen in the adjuvant setting was the NSABP
B-33 study. This trial compared exemestane with placebo for
5 years after the standard 5 years of tamoxifen. Based on the
results of MA.17, accrual to this trial was discontinued, the
study medication unblinded, and all participants taking placebo
were offered exemestane.
The side effect profile of tamoxifen has been examined in adjuvant
studies. In addition, important toxicity data were gained from
four large tamoxifen prevention trials in which tamoxifen was
compared with placebo, and as a result, probably most accurately
reflect its true toxicities.
The third-generation AIs are generally well tolerated. The
available data from the first-line metastatic setting indicate
at least equal short-term safety of the AIs compared with
tamoxifen, including symptoms of menopause and quality of life.
However, for use of AIs in the adjuvant setting, short-term
tolerability and long-term effects in a healthy population are
relevant. The most relevant data on the toxicity profile of AIs
are derived from adjuvant trials in which the agents are
evaluated in disease-free women over a prolonged period.
Hot Flashes and General Tolerability
Tamoxifen significantly increases bothersome hot flashes and
vaginal discharge, but this did not affect overall physical and
emotional well-being in the NSABP P-1 chemoprevention trial,
which included more than 13,000 women.
The reported effects of tamoxifen on cognition are variable and
appear dependent on the parameters that are measured. Several
ongoing studies are addressing this point.
In the two adjuvant AI studies in which tamoxifen is the standard
arm (ATAC and IES), the short-term tolerability of the AI was
at least as good as that of tamoxifen.
In ATAC, patients receiving
anastrozole suffered from
significantly fewer hot flashes, which were, however, not
confirmed in a parallel quality-of-life study.
There was a significantly decreased rate of vaginal bleeding in
patients receiving anastrozole (4.5% v 8.2%; P <
.0001). This reflects the nonstimulatory effect of the AIs on
the endometrium. In the IES trial, the frequency of hot flashes
and vaginal bleeding was similar in both arms. In a study of
letrozole in women completing tamoxifen, more rapid return to
normal endometrial thickness was noted after tamoxifen in women
receiving letrozole than those on no treatment.
Other short-term adverse effects such as nausea and vomiting,
fatigue, mood disturbance, headaches, and dizziness were noted
with equal frequency in both arms. The weakness of data coming
from the ATAC and IES studies is that there was no placebo arm
allowing for a true estimation of the toxicities of the AI.
This is now possible with data from the adjuvant MA.17 study,
comparing letrozole with placebo. In this trial, hot flashes
occurred more often in the letrozole arm (47.2% v
40.5%). Symptoms were, in general, grade 1 to 2 out of 4
according to the National Cancer Institute Common Toxicity
Criteria. Otherwise, the short-term tolerability of the study
drug in MA.17 was excellent, with symptoms such as fatigue,
sweating, constipation, headache, dizziness, and vaginal
bleeding being equally distributed between letrozole and
placebo. The percentage of women discontinuing treatment
because of adverse events was not different between the two
arms.
Lipid Metabolism
While in retrospective analyses of three randomized tamoxifen
trials a reduction in coronary heart disease was observed, no
such benefit was demonstrated in NSABP P-1.
In the EBCTCG overview of 1998, mortality rates for causes "not
attributed to breast or endometrial cancer" were nearly
identical in patients receiving tamoxifen or placebo in the
adjuvant setting.In view of the recently published data from the Women's Health
Initiative study,
it is uncertain whether the favorable influence of tamoxifen on
lipid metabolism translates into a true reduction in coronary
heart disease.
In the ATAC trial, influences of either anastrozole or tamoxifen
on the serum lipid profile have not been reported. In a small
study, no influence of anastrozole on the lipid profile was
seen when postmenopausal women were treated for less than 20
months.
Studies on the effects of letrozole on lipid metabolism have
yielded conflicting results. When letrozole was given to healthy
women for 3 months, no influence was seen on their plasma lipid
levels.
However, in another study including 20 women with breast
cancer, letrozole significantly increased total and low-density
lipoprotein (LDL) cholesterol levels, as well as the atherogenic
risk ratios total/ high-density lipoprotein (HDL) and LDL/HDL
cholesterol.
In the adjuvant MA.17 study, there was a (nonsignificant) trend
towards a higher rate of cardiovascular events in the letrozole
group compared with placebo (4.1% v 3.6%).
There were no reports of drug-related hypercholesterolemia.
Publication of the results of the lipid substudy of MA.17 is
pending and follow-up of the companion trial participants is
ongoing.
Exemestane might have effects converse to the other AIs in terms
of lipid metabolism. When exemestane was compared with tamoxifen
in breast cancer patients, it had beneficial effects on
triglycerides and a stabilizing effect on HDL and total
cholesterol levels after 24 weeks.
Similarly, in animal experiments, exemestane improved the serum
lipid profile in treated rats compared with either oophorectomy
or letrozole.
In a small European Organisation for Research and Treatment of
Cancer (EORTC) study of metastatic breast cancer patients,
exemestane had no detrimental effect on cholesterol levels and
atherogenic indices, and had a beneficial effect on
triglyceride levels compared with tamoxifen.
It is therefore possible that being an androgenic steroid makes
exemestane superior to the other inhibitors in terms of adverse
effects caused by estrogen depletion of target tissues other
than the breast. In the adjuvant IES study, the effect of exemestane
on cholesterol levels was not systematically measured.
There was no significant increase in the rates of myocardial
infarction in the exemestane arm compared with tamoxifen.
Skeletal Effects
Tamoxifen has been shown to preserve bone mineral density in
postmenopausal breast cancer patients.
NSABP P-1 is the only prospective trial that has evaluated the
effect of tamoxifen on bone fractures versus placebo and it
showed a reduction in the risk of long bone and symptomatic
vertebral fractures of borderline statistical significance
(risk ratio [RR] = 0.81; 95% CI, 0.63 to 1.05).
To date, tamoxifen has not been evaluated in a prospective
trial in women with osteoporosis.
In the ATAC trial, women taking
anastrozole were more likely
to suffer from musculoskeletal disorders (30.3% versus 23.7%,
P < .001), in particular fractures (7.1% versus 4.4%, P <
.001).
In the ATAC trial, women taking
anastrozole were more likely
to suffer from musculoskeletal disorders (30.3% v 23.7%; P <
.001), in particular, fractures (7.1% v 4.4%; P <
.001).
Similar to anastrozole, letrozole has, in several studies, been
shown to significantly increase parameters of bone resorption.
In healthy postmenopausal women, letrozole given for 12 weeks
increased bone resorption and reduced bone formation.
In the adjuvant MA.17 study, more women in the letrozole group
compared with placebo suffered from new-onset osteoporosis and
fractures (3.6% v 2.9%).
In addition, arthritis, arthralgia, and myalgia were reported
more often by women in the experimental arm (5.6% v
3.5%, 21.3% v 16.6.%, and 11.8% v 9.5%, for letrozole and
placebo, respectively).
The effect of exemestane on markers of bone metabolism was studied
in a preclinical study of ovariectomized rats. In this model,
exemestane protected against the negative effects of oophorectomy.
In healthy postmenopausal women, exemestane given for 12 weeks
appeared to cause similar increases of bone resorption as the
other inhibitors, but in addition, increased serum propeptide
of type 1 collagen, a marker of bone formation, a feature not
seen with the nonsteroidal inhibitors.
This supports the notion that
exemestane may have a superior
safety profile compared with other third-generation inhibitors
with regard to bone metabolism. In the adjuvant IES study,
there was a higher frequency of osteoporosis and arthralgia in
the exemestane group compared with tamoxifen (7.4% v
5.7% and 5.4% v 3.6% for exemestane and tamoxifen, respectively).
There was no significant increase in fractures. When studying
the effects of AIs on bone metabolism in comparison with tamoxifen,
it should be noted that tamoxifen has a beneficial
(estrogen-agonistic) effect on bone.
Therefore, the negative effects on bone caused by the AIs in
the ATAC and IES studies might, in part, be the result of a
protective effect of tamoxifen.
Thromboembolic Events and Endometrial Cancers
Participants in the tamoxifen breast cancer prevention trials
had a 2.4 times greater risk of developing invasive endometrial
cancer on tamoxifen than those receiving placebo (P = .00005),
and this effect was more pronounced in women over 50 years of
age compared with younger women (RR = 4.01 v 1.21).
When the data from the adjuvant studies included in the 1998
EBCTCG analysis are taken together,
this excess of endometrial
cancer risk is confirmed (odds ratio [OR], 3.4; P =
.00002).
Tamoxifen also leads to an excess risk of thromboembolic events
when compared with placebo. In the NSABP P-1 prevention study,
pulmonary embolism was three times as common (RR = 3.01), and
strokes were nearly twice as frequent among women > 50 years
of age receiving tamoxifen compared with younger women taking
the drug (RR = 1.75).
Venous thromboembolic events were also increased in all
prevention trials, with a relative risk of 1.9 in the tamoxifen
compared with the placebo arms (P < .0001).Overall, the increase in vascular events with tamoxifen was
comparable to that seen with hormone replacement therapy.
In all the trials comparing the third-generation AIs with tamoxifen,
the adverse events caused by tamoxifen's estrogenic properties
were significantly less common in the AI groups.
This is particularly important with respect to ischemic
cerebrovascular and venous thromboembolic events (including
deep venous thrombosis) and endometrial cancer. Compared with
placebo, letrozole did not lead to increased rates of any of
these serious adverse events in the MA.17 study and vaginal
bleeding occurred more often in the placebo arm (P = .01)
Comparison Between AIs
In a pharmacodynamic cross-over study, letrozole was associated
with a more profound suppression of aromatase than anastrozole.
In a clinical trial in advanced disease after tamoxifen, time
to progression, time to treatment failure, duration of response
and of clinical benefit were similar between letrozole and
anastrozole. In this study, letrozole was associated with a
significantly better overall response rate (19.1% v
12.3%; P = .014), which was, however, not seen in the
ER-positive subgroup.
There are no data directly comparing exemestane to any of the
other inhibitors. A large first-line adjuvant trial is now
underway, comparing anastrozole to exemestane as outlined below
(MA.27).
AIs Used Sequentially
The results of several studies suggest that breast cancer might
be sensitive to steroidal AIs even after failure of a nonsteroidal
inhibitor. Exemestane led to clinical benefit in 24.3% of 241
patients with advanced breast cancer after failure of tamoxifen
and a nonsteroidal inhibitor.A Spanish randomized cross-over study including 100 breast
cancer patients is currently evaluating the use of exemestane
after anastrozole versus the opposite in metastatic disease.
Preliminary results of a small study in the metastatic setting
comparing various sequences of AIs showed no difference between
overall response and clinical benefit rates, implying that
nonsteroidal inhibitors can be used after exemestane as well.
There are no ongoing adjuvant studies exploring the sequence of
two AIs.
Combination of Agents
Tamoxifen plus AIs The obvious
combination of tamoxifen and an AI in postmenopausal women was
evaluated in the adjuvant ATAC trial.
In the first interim analysis, the combination of the two
endocrine agents was equivalent to tamoxifen alone, but
significantly worse than anastrozole. A possible explanation of
this finding might be that the estrogen agonistic properties of
tamoxifen are more effective in an estrogen-depleted
environment. Based on these findings, the combination arm of
ATAC was suspended. It has been hypothesized, however, that use
of a less agonistic SERM with an AI may achieve the intended
total estrogen blockade. While toremifene appears equivalent to
tamoxifen as a single agent in patients in vitro in a depleted
estrogen environment, it is significantly less estrogen
agonistic. Based on this finding, toremifene is being combined
with an unregistered steroidal inhibitor, atamestane, in a
trial comparing the combination to letrozole in women with
locally advanced or metastatic breast cancer.
AIs plus COX-2 inhibitors In
recent years, several preclinical studies have implicated a
role for COX-2, an enzyme generating prostaglandins, in the
pathogenesis of breast cancer.In an analysis of 1,576 human breast cancer samples, COX-2
expression was found in 37.4% and correlated with adverse
prognostic features.
Preclinically, inhibition of COX-2 by celecoxib was effective
in the treatment and prevention of ER-positive mammary tumors.
Importantly, celecoxib also inhibits the proliferation of
ER-negative breast cancer cells.
There is a close relationship between the COX-2 and aromatase
pathways. Prostaglandin E2–dependent expression of
aromatase has been shown in several studies,
and a positive correlation between COX-2 and aromatase
expression was demonstrated in human breast cancer specimens.
Several trials are evaluating the potential of COX-2 inhibition
in treating breast cancer. In a phase II study of 13 breast
cancer patients with ER-positive or progesterone receptor–positive
metastatic disease, the combination of exemestane and celecoxib
produced a clinical benefit rate of 73%.
In the adjuvant setting, the National Cancer Institute of
Canada–Clinical Trials Group (NCIC-CTG) MA.27 trial is
evaluating the addition of celecoxib to exemestane or
anastrozole . Enrollment to MA.27 exceeds 1,000 women to date. A
neoadjuvant three-arm study (Celecoxib Anti-Aromatase
Neoadjuvant) is comparing exemestane plus celecoxib to
exemestane or to letrozole.Other studies, including the recently launched,
randomized NCIC-CTG MAP.3 study are prospectively evaluating
the chemopreventive potential of celecoxib in combination with
an AI .
Very recently, at the time of this submission, concern arose
regarding the potential cardiotoxicity of inhibitors of the
COX-2 enzyme, including adverse events noted in an adenomatous
polyposis coli trial. This has led to the suspension of the
celecoxib/placebo randomization within the MA.27 trial and to
elimination of the exemestane plus celecoxib arm from the MAP.3
trial. Review of other celecoxib-containing trials is also
being undertaken.
Strategies to Overcome Endocrine Resistance in Postmenopausal
Breast Cancer
It has been known since the early development of endocrine therapies
that some patients with ER/PR -positive breast cancer do not
benefit from endocrine therapy. In addition, even patients whose
tumors initially respond to endocrine treatment will eventually
experience disease progression. In recent years, substantial
progress has been made in understanding specific mechanisms
of endocrine resistance. One major mechanism for tumor insensitivity
to hormonal agents seems to be cross-talk between the ER and
other growth factor signaling pathways. The studies investigating
this interaction have revealed at least three different levels
of cross-talk between signal transduction pathways and steroid
hormone receptors.
In breast cancer patients, overexpression or aberrant
activation of epidermal growth factor receptor 2
(EGFR-2/ErbB2/HER-2) has been widely demonstrated in breast
tumors and linked to an adverse prognosis and endocrine resistance.
Consequently, combining endocrine therapies with signal transduction
inhibitors might be a strategy to overcome the development of
endocrine resistance.
In preclinical studies, signal transduction inhibitors such as
the tyrosine kinase inhibitor of EGFR-1, ZD-1839, seem to be
only marginally successful if used as monotherapy. However, in
a recently published study in a breast cancer xenograft mouse
model, significant synergism was shown between ZD-1839 and
tamoxifen.
In this study, mice bearing ER-positive MCF-7 breast tumors
were treated with tamoxifen, ZD-1839, or the combination of
both. ZD-1839 improved the antitumor effect of tamoxifen, and
markedly delayed the emergence of acquired resistance from 2 to
3 months to over 6 months. Importantly, in mice treated with
estrogen deprivation, there was no demonstrable benefit from
adding ZD-1839, indicating that activation of the EGFR pathway
enhances the agonist effects of tamoxifen. Apart from ZD-1839,
numerous other growth factor tyrosine kinase inhibitors are in
different stages of preclinical and clinical development,
including OSI-744, PKI-166, GW-572016, and CI-1033, which is an
irreversible pan-erbB tyrosine kinase inhibitor. These compounds
are being studied as monotherapy in numerous tumor types and
clinical studies evaluating them in combination with endocrine
agents are awaited with great interest.
Downstream of the growth factor receptor tyrosine kinases, the
Ras proteins, for which aberrant function has been demonstrated
in breast cancer, play a major role in intracellular signal
transduction. The enzyme farnesyl transferase, which is needed
for post-translational processing of Ras, can be inhibited with
the newly developed farnesyl transferase inhibitors.
One of these compounds, R115777, is active in breast cancer
models
and has already been studied in women with advanced
hormone-resistant breast cancer.
Among 76 patients, 24% of patients derived clinical benefit
from R115777. The farnesyl transferase inhibitors have, as yet,
not been studied in combination with endocrine therapy.
Other signal transduction inhibitors in clinical development
include the Raf kinase and MEK inhibitors, cell cycle inhibitors,
and the mTOR inhibitors, of which CCI-779 (rapamycin) has entered
clinical development in advanced breast cancer.For CCI-779, improved efficacy has been demonstrated in
combination with endocrine therapy, and a phase II trial
combining the compound with AIs is being planned.
Tamoxifen has played a major role in postmenopausal, hormone-sensitive,
early-stage breast cancer. De novo and acquired resistance to
tamoxifen have limited its efficacy, but identification of specific
signal transduction pathways responsible for tamoxifen resistance
promises to allow combinations of tamoxifen and inhibitors of
these pathways to improve patient outcome. AIs are proving to
be an important new class of endocrine therapy and whether to
use them instead of, or in sequence with, tamoxifen is being
determined. Understanding resistance mechanisms to AIs and finding
tumor and host prognostic and predictive markers should allow
selection of patients for treatment with the inhibitors. Long-term
adverse effects of the AIs are being determined in the follow-up
of several trials including the NCIC-CTG MA.17 and the
re-randomization phase of this trial.
Whether steroidal AIs are
superior to nonsteroidals is being determined in the NCIC-CTG
MA.27 trial now underway. The outlook of postmenopausal women
with hormone-dependent breast cancer promises to improve as a
result of these new therapeutic approaches.
|