The development of the novel third-generation aromatase inhibitors and inactivators for breast cancer treatment is one of the most successful contemporary achievements in cancer therapy. Parallel to studies evaluating toxicity and clinical efficacy in metastatic disease, the endocrine effects of multiple compounds were evaluated, leading to the identification of the highly potent third-generation aromatase inhibitors based on estrogen deprivation and aromatase inhibition in vivo. Thus, translational studies have been of vital importance identifying the unique characteristics of these compounds. Whereas first- and second-generation aromatase inhibitors inhibit estrogen synthesis in vivo by up to 90%, the third-generation compounds anastrozole, exemestane, and letrozole were found to cause ≥98% aromatase inhibition. This article summarizes and discusses the “translational research” that provided the background for the implementation of the third-generation aromatase inhibitors and inactivators into large clinical trials. The need for future translational research exploiting the mechanisms of resistance to these compounds for future improvement of endocrine therapy is emphasized.

The development of contemporary endocrine treatment for breast cancer is one of the most remarkable stories in cancer therapy. Initiated more than a century ago by Beatson's oophorectomy (1), followed by other pioneering discoveries like the therapeutic potential of ablative strategies (adrenalectomy or hypophysectomy) in postmenopausals (2, 3) and the beneficial effects of different additive therapies (refs. 47; Table 1), endocrine treatment of breast cancer was established a long time before implementation of cytotoxic therapy (814). Currently, we are facing a major novel achievement: the successful development of the third-generation aromatase inhibitors and inactivators in early breast cancer.

Table 1.

Development of endocrine therapy

First publishedAuthorType of therapyReferences
1896 Beatson Oophorectomy (1) 
1922 DeCourmelles Ovarian irradiation (9) 
1939 Ulrich Androgens (10) 
1944 Haddow et al. Synthetic estrogens (5) 
1952 Douglas Pituitary irradiation (11) 
1953 Huggins et al. Adrenalectomy (12) 
1953 Luft et al. Hypophysectomy (3) 
1956 Taylor et al. Cortical steroids (6) 
1971 Cole Antiestrogen (14) 
1973 Schwarzel et al. Aromatase inhibition (22) 
First publishedAuthorType of therapyReferences
1896 Beatson Oophorectomy (1) 
1922 DeCourmelles Ovarian irradiation (9) 
1939 Ulrich Androgens (10) 
1944 Haddow et al. Synthetic estrogens (5) 
1952 Douglas Pituitary irradiation (11) 
1953 Huggins et al. Adrenalectomy (12) 
1953 Luft et al. Hypophysectomy (3) 
1956 Taylor et al. Cortical steroids (6) 
1971 Cole Antiestrogen (14) 
1973 Schwarzel et al. Aromatase inhibition (22) 

Traditionally, endocrine therapy has been developed through interplay between clinical observations, endocrine evaluations in vivo, and laboratory experiments. The initial clinical discoveries with respect to oophorectomy, additive therapy with estrogens, and adrenalectomy were empirical observations at a time when knowledge about endocrinology in general and with respect to breast cancer in particular was poor. At this stage, the clinical observation of the effects of ablative as well as additive treatment added key information to our understanding of the endocrine biology of breast cancer. As such, the development of endocrine therapy remains a pioneering example of successful translational research, transferring knowledge between basic research and the clinic in an interactive two-way process (15, 16). As may be seen, this applies to the successful development of third-generation aromatase inhibitors and inactivators in particular.

The discovery of the estrogen receptor and, subsequently, the finding three decades ago that its expression was a requisite for response to endocrine therapy (17, 18) gave us a predictive factor, something yet to be identified for chemotherapy. Although far from perfect (we still lack knowledge why so many tumors harboring the estrogen receptor-α do not respond to endocrine therapy), the lack of response among tumors with negative receptor status means that a substantial number of patients may avoid an ineffective therapy. The functional role of the ligand-receptor mechanism is further underlined by the additional predictive value of the progesterone receptor, known to be a marker of a functional estradiol (E2)-receptor ligand interaction (19). Whether its predictive value differs with respect to treatment with tamoxifen and aromatase inhibitors (20) remains to be confirmed in additional studies.

The list of unexpected clinical observations leading to a “conceptual breakthrough,” thus, “translating the clinic back to the laboratory,” is extensive. The introduction of the first aromatase inhibitor in the clinic was based on a conceptual misunderstanding. The therapeutic effect of surgical adrenalectomy initiated attempts to develop medical adrenalectomies by use of corticosteroids and, later, an adrenotoxic antiepileptic compound named aminoglutethimide (13, 2123). Whereas treatment with corticosteroids was only partly successful (24, 25), the clinical efficacy of aminoglutethimide triggered endocrine studies revealing the compound to be a potent inhibitor of aromatization, albeit with additional effects on adrenal steroid synthesis (26, 27).

The observation that the response to hormone ablation was (at least) as good in postmenopausal patients undergoing adrenalectomy as for premenopausals treated with ovarian ablation and, later, the finding that aminoglutethimide may work even in women expressing supra-low plasma estrogen levels due to a previous adrenalectomy or hypophysectomy (28) revealed the (still puzzling) observation that endogenous estrogens may stimulate the growth of hormone-sensitive tumors more or less independent of plasma hormone levels, suggesting the antitumor effects of hormone suppression may be due to relative suppression and not absolute hormone levels (29).

Another clinical observation translating into an improved conceptual understanding relates to the clinical experiences made during treatment of breast cancer patients with androgens. Whereas androgens have antitumor effects but were abandoned from breast cancer therapy due to their side effects (7), the discovery of their antitumor effects may add important information to our understanding of the puzzling lack of cross-resistance between aromatase inhibitors and inactivators (see later). Finally, recent clinical observations with use of tamoxifen in combination with luteinizing hormone–releasing hormone analogues in premenopausals (30) and tamoxifen in combination with aromatase inhibitors in postmenopausals (31) have added to our understanding of the estrogen agonistic/antagonistic effects of tamoxifen in relation to estrogen suppression.

An important issue with respect to most forms of therapy is the search for biochemical surrogate markers predicting in vivo efficacy. In contrast to other forms of endocrine treatment, like use of antiestrogens, for which biochemical surrogate variables remain an issue of debate (32, 33), or chemotherapy, for which dose-finding is based on a combined assessment of preclinical efficacy and in vivo toxicity, the effect of an aromatase inhibitor on its therapeutic target, the peripheral aromatase enzyme, can be determined in vivo (34). Whereas aromatase inhibitors may be characterized based on in vitro experiments, their potency in vivo will depend on additional factors, including plasma half-life and tissue penetration. Thus, whereas fadrozole was found superior compared with letrozole in vitro (35, 36), letrozole was found to be much more potent in vivo (37, 38).

This review will focus on the translational work leading to the successful development of novel aromatase inhibitors. The issues of endocrine assessment will be discussed in detail, putting emphasis on methodologic limitations, as these are critical to the interpretation of our findings. The fact that aromatase inhibitors may improve relapse-free and, most likely, overall survival compared with tamoxifen (31, 39, 40), something few had believed a decade ago, underlines the potential for improvement in this therapeutic area.

The first aromatase inhibitor for clinical use, an adrenotoxic antiepileptic named aminoglutethimide, was introduced in an attempt to induce a medical adrenalectomy (13, 41). However, over the next decade, Santen et al. (27, 42), in a series of elegant experiments, revealed plasma dehydroepiandrosterone sulfate to be suppressed, but plasma androstenedione sustained or elevated depending on glucocorticoid replacement. Despite this, they found plasma estrogens to be profoundly suppressed. Inspired by the work of Thompson and Siiteri (23), who found aminoglutethimide to inhibit in vitro aromatization in 1974, they evaluated and found aminoglutethimide to inhibit in vivo aromatization in postmenopausal women (26; see ref. 43 for a detailed discussion of the early clinical development and endocrine effects of aminoglutethimide). The clinical development of aminoglutethimide as an aromatase inhibitor was then pursued. However, due to the side effects of the nonselective compound aminoglutethimide, many attempts were made to find more selective drugs that were better tolerated.

At the same time, Brodie and colleagues identified a panel of steroidal compounds inhibiting the aromatase enzyme in preclinical models (44, 45). The first approach was developing substrate analogues, derivatives of androstenedione (the major substrate of aromatase) or testosterone.

Following the results of Santen and colleagues, two independent events, one endocrine study and one pilot study of the antitumor effects of a compound, confirmed the principle of aromatase inhibition, independent of adrenal enzyme inhibition, as a therapeutic strategy in breast cancer. The endocrine study related to the compound named testololactone. This weak androgen agonist had been evaluated for breast cancer therapy in the 1970s and found to cause antitumor effects (46) albeit with a significant toxicity. Subsequently, Barone et al. (47) in 1979 found testololactone to inhibit peripheral aromatization by about 90%, confirming the compound to be a “steroidal” aromatase inhibitor, albeit with intrinsic androgenic effects. The “clinical event” was the pilot study reporting 4-hydroxyandrostenedione (today known as formestane), one of the compounds developed by the Brodie team, to cause antitumor effects in patients suffering from metastatic breast cancer (48). As this compound has no effects on adrenal hormone synthesis nor does it express any androgenic activity following parenteral administration (49, 50), this small study provided the “proof of principle” revealing a selective aromatase inhibitor to be an active antitumor compound.

There are two classes of aromatase inhibitors; the nonsteroidal, or type II compounds, and the steroidal derivatives (type I), currently coined aromatase inactivators (see explanation in this section). Considering the nonsteroidal compounds, these drugs are either phenobarbitone derivatives (like aminoglutethimide) or belong to the imidazole/triazole class (Fig. 1). The steroidal compounds are derivatives of the aromatase substrate, androstenedione (Fig. 2).

Fig. 1.

Chemical structures of some nonsteroidal aromatase inhibitors.

Fig. 1.

Chemical structures of some nonsteroidal aromatase inhibitors.

Close modal
Fig. 2.

Chemical structures of androstenedione and some steroidal aromatase inactivators.

Fig. 2.

Chemical structures of androstenedione and some steroidal aromatase inactivators.

Close modal

Based on in vitro experiments, the nonsteroidal compounds are characterized by reversible binding to the P450 part of the aromatase enzyme. In contrast, the two steroidal compounds used for breast cancer therapy (4-OH-androstenedione and exemestane) both bind irreversibly to or near by the substrate-binding pocket due to covalent bonds causing aromatase inactivation (5154). Through this mechanism the enzyme is irreversibly inactivated (also called mechanism based or suicide inhibition; ref. 55). The potential importance of these differences (56), in particular with respect to the well-confirmed lack of complete cross-resistance between aromatase inhibitors and inactivators, has been an issue of debate. So has the observation that the steroidal compounds express slight androgenic effects as detected by a dose-dependent suppression of plasma sex hormone binding globuline following oral but not parenteral administration (57, 58). Whereas the important issue of lack of cross-resistance is addressed later in this article, it should be mentioned that this observation, similar to the discovery of aminoglutethimide as an aromatase inhibitor, was made for the first time in a clinical trial (59) for which the hypothesis (that formestane should be a more potent inhibitor compared with aminoglutethimide) was incorrect, but the therapeutic result turned out to be positive, triggering the postulation of an alternative hypothesis.

There are three types of methods by which the biochemical efficacy of an aromatase inhibitor may be assessed: in vitro measurements, evaluation in animal models, and in vivo assessment of endocrine efficacy in humans. Whereas the two first issues will be covered here briefly and the reader referred to other contemporary reviews, this article will review the subject of endocrine assessment in humans in detail.

In general, in vitro assessment of aromatase inhibition is conducted using placental or ovarian aromatase as test substrate (60). Notably, whereas aromatase expression may be regulated by different promoters in different tissues (refs. 61, 62; see later), only one aromatase gene, coding a single protein, is detected in human tissues (63). The subject of in vitro assessment of aromatase inhibitors has been reviewed by others (64, 65); only a few issues will be highlighted here.

Whereas in vitro data may outline the potency of individual compounds suggesting which may be chosen for clinical development, the importance of in vivo assessment of endocrine effects is illustrated by the comparison between fadrozole and letrozole, revealing fadrozole to be more potent in vitro (35, 36), whereas letrozole was superior in vivo (37, 38). Whether the discrepancy between in vitro and in vivo findings may be related to differences in pharmacokinetic disposition (6668) alone or other factors may contribute is not known.

The animal model most frequently used to assess endocrine effects of aromatase inhibitors is the ovariectomized nude mouse inoculated with MCF-7 cells transfected with the human aromatase gene (MCF-7CA) and treated with androgens (postmenopausal model; ref. 64, 69). Whereas results achieved with this model have been interpreted in favor of intratumor estrogen synthesis versus systemic delivery (69, 70), there is evidence that estrogens synthesized by the MCF-7 xenografts in this model may contribute to circulating hormone levels (71). However, the relevance of this source to circulating estrogens in breast cancer patients remains uncertain.

Recently, insights into the effects of estrogens on different target tissues have been generated by animal models using targeted disruption of key genes (knockout models). This group comprises the aromatase-knockout mouse (ref. 72; ArKO mouse), the ERKO mouse (disrupted estrogen receptor α), the βERKO mouse (disrupted estrogen receptor β), as well as the α/βERKO mouse (disrupted estrogen receptors α and β; ref. 73). These model systems are currently used by several research groups and allow scientists to study the role of aromatase and the individual estrogen receptors in vivo.

Plasma estrogens of interest are E2, estrone (E1), and estrone sulfate (E1S). Whereas only E2 is biologically active, plasma levels of E1 (the main product of aromatization in postmenopausal women) and E1S are about 4- and 20-fold higher than the concentration of E2, respectively, in postmenopausal women (74, 75). As a consequence, E1S is a useful surrogate variable due to a ratio of >100 between the plasma levels of E1S in postmenopausals and the sensitivity limit for E1S analysis (29).

The importance of plasma estrogens as an intratumor source as well as the validity of using plasma estrogen suppression as a variable for aromatase inhibitor efficacy remains controversial. There are several reasons for this. First, the mean tissue level of E2 is an order of magnitude higher than plasma level in postmenopausal women, with particular high levels in many breast tumors (76, 77). Because plasma levels of E1S are 10- to 20-fold higher than plasma E2 (78), circulating E1S has been suggested a major estrogen source to the tumor tissue (79). This hypothesis is partly challenged by contemporary findings that breast intratumor levels of E1S are less than half the level of E2 (77). On the other hand, the identification of an active transport mechanism for E1S across the cell membrane (80) and high expression of sulfatase (81) as well as 17β-dehydrogenase type 1 (82, 83) in tumor tissue may be consistent with a rapid uptake of E1S followed by conversion to E2. Whereas one study suggested an active uptake of E2 from plasma into tumor tissue in rats (84) and studies evaluating the contribution of circulating estrogens to intratumor levels in breast cancer patients have reported a substantial interindividual variation (85, 86), the general opinion is that the high concentration of E2 in tumor tissue is due to high expression of the aromatase enzyme leading to local estrogen synthesis from androgen precursors Fig. 3; ref. 56).

Fig. 3.

Origin of breast cancer tissue estrogens and sites of action of aromatase inhibitors and inactivators.

Fig. 3.

Origin of breast cancer tissue estrogens and sites of action of aromatase inhibitors and inactivators.

Close modal

The finding of a high tissue to plasma gradient, in particular for E2, has questioned the validity of measuring plasma estrogen levels as well as total body aromatization as surrogate markers for tissue estrogen levels. Postmenopausal estrogen synthesis occurs in most nonglandular tissues including fat, muscle, liver, and skin, and in normal breast as well as malignant tissue (87101). All circulating estrogens in postmenopausals are synthesized in nonglandular tissue compartments, whereafter they leak into the circulation by a passive gradient for subsequent removal by the liver and kidneys (102). Whereas interindividual variation in estrogen clearance rates (103) would influence plasma hormone levels, the drop in plasma levels recorded in relation to drug therapy reflects an effect on total body estrogen production and, thereby, tissue production in general. Whereas the issue has sometimes been a subject to discussion, circulating estrogens thus reflect tissue synthesis and may be considered as surrogate markers for normal tissue levels.

Intratumor estrogen levels remain a different topic. Aromatase expression may be particularly high in tumors (86), and the observation made by different groups, including our own (77), of a lack of correlation between plasma and intratumor estrogen levels may reflect a substantial interindividual variation with respect to local (tumor) estrogen synthesis. Whereas the aromatase enzyme is encoded by a single copy of the CYP19 gene localized at human chromosome 15q21.2 (104), a total of nine promoters have been identified thus far; these are differentially expressed in different tissues (105, 106). The promoters I.3, II, and I.7, which dominate in breast cancer tissue (107), may be stimulated by interleukins (interleukins 6 and 11) as well as tumor necrosis factor α and prostaglandin 2 (108, 109), all commonly expressed in tumor tissue. Whereas a limited number of studies have evaluated intratumor estrogen suppression during treatment with aromatase inhibitors (110), data thus far suggest a similar degree of estrogen suppression in tumors expressing high hormone levels and in plasma (77), arguing against a local escape phenomenon.

Determination of plasma and tissue estrogen levels. Due to successful development of sensitive assays for plasma estrogens, such methods in general have replaced traditional methods of urinary estrogen measurements (111). Notably, there is no international consensus regarding normal values for plasma estrogen levels in postmenopausal women. Whereas studies by different groups have reported high plasma estrogen levels to be a significant risk factor for breast cancer development in postmenopausal women (112120), the fact that the mean values of E2 recorded by different investigators may vary by a factor of up to 10 (refs. 38, 114, 115, 121125; Fig. 4) questions the validity of doing a meta-analysis on these data (126, 127). Theoretical calculations based on androgen production, androgen and estrogen plasma levels, aromatase activity, and hormone clearance rates suggest plasma levels of E2 to be in the range of 10 to 20 pmol/L (128), consistent with values reported by groups using highly sensitive RIAs for estrogen measurements (38).

Fig. 4.

Plasma E2 levels (pmol/L) in postmenopausal breast cancer patients as published in the literature (in chronological order). The first (black) column presents the theoretical plasma E2 value calculated from androgen production rates, plasma hormone levels, and mean level of aromatization [see Lønning et al. (128) for details].

Fig. 4.

Plasma E2 levels (pmol/L) in postmenopausal breast cancer patients as published in the literature (in chronological order). The first (black) column presents the theoretical plasma E2 value calculated from androgen production rates, plasma hormone levels, and mean level of aromatization [see Lønning et al. (128) for details].

Close modal

Considering RIA methods in general, the major problems are lack of sensitivity in general and lack of specificity against potential cross-reactive compounds in particular; each factor may result in false high values. Using highly sensitive RIAs, we and others recorded mean values for E2 in the range of 15 to 25 pmol/L, with mean levels of E1 and E1S of about 100 and 4 to 500 pmol/L, respectively (50, 77). However, taking into account sensitivity limits in the range of 2 to 3 pmol/L (somewhat higher for E1), this means that for the majority of patients we are able to record plasma hormone suppression below 1% of pretreatment values with respect to E1S, but not for E2 or E1 (29). Thus, the measurement of E1S in plasma during treatment with aromatase inhibitors is a useful surrogate variable for plasma estrogen suppression in general. This is illustrated by a recent study comparing the two third-generation aromatase inhibitors anastrozole and letrozole in a crossover design (38). Here, we found a significant difference with respect to plasma levels of E1 and E1S during treatment with the two compounds but no difference in plasma levels of E2 (Fig. 5). This was due to the fact that 9 of 12 and 12 of 12 patients had their plasma E2 levels suppressed below the sensitivity of the method during treatment with anastrozole and letrozole, respectively. We did not evaluate the clinical responses comparing anastrozole and letrozole due to the study design with small numbers of patients and short-term exposure to the compounds. However, a direct comparison of the clinical effects of anastrozole versus letrozole has been recently published by Rose et al. (129).

Fig. 5.

Plasma E1, E1S, and E2 levels (pmol/L) in postmenopausal breast cancer patients treated in a crossover trial [see Geisler et al. (38) for details] comparing anastrozole (A) and letrozole (L). C, baseline. Arrows indicate detection limits of the RIAs.

Fig. 5.

Plasma E1, E1S, and E2 levels (pmol/L) in postmenopausal breast cancer patients treated in a crossover trial [see Geisler et al. (38) for details] comparing anastrozole (A) and letrozole (L). C, baseline. Arrows indicate detection limits of the RIAs.

Close modal

Due to their steroidal structure, aromatase inactivators and their metabolites are particularly prone to cross-reactivity in the RIAs. Whereas interactions involving the mother compound and major metabolites can be tested for, it is difficult to exclude potential minor, probably unidentified, cross-reacting metabolites. The daily dose of exemestane for clinical treatment is 25 mg, and total estrogen synthesis in postmenopausal women about 50 μg daily (111). Assuming aromatase inhibition of about 98% (130), a metabolite of exemestane accounting for 1% of its metabolism with a plasma clearance rate resembling E2 and a cross-reactivity against the antibodies of 10% may in theory increase the value of plasma E2 measured during therapy by 100% (128). The problem of metabolite interaction is illustrated by the fact that plasma estrogen levels in patients treated with exemestane can be measured only after high-performance liquid chromatography purification of the plasma samples (57).

The problems related to sensitivity of the assays are of relevance to tissue estrogen measurements as well. However, due to different levels of the individual estrogens in plasma and tissue and the sensitivity of the individual assays, E2 seems to be the most reliable indicator for assessment of alterations in tissue estrogen levels (29, 77, 131). Notably, tissue estrogen measurements are time and resource consuming (131), for which reason such studies may include a limited number of patients only.

Measurement of in vivo aromatization by use of isotope techniques. This may in principle be done by two methods: assessment of aromatase inhibition by measuring the isotope ratio in plasma E1 during steady-state infusion (26) or by measuring the isotope ratio in the main estrogen metabolites extracted from the urine following an i.v. bolus injection.

In all studies done thus far, the tracers used have been 3H-labeled androstenedione and 14C-labeled E1. With the plasma method, the aim is to achieve a steady-state level through concomitant infusion of the two tracers (26, 132, 133). With the urine method, a bolus injection of 3H-labeled androstenedione and 14C-labeled E1 is followed by total urine collection over a period of 4 days with isolation and measurement of the isotope ratio in the major estrogen metabolites (34). For practical reasons, this may be limited to E1 and estriol (E3), leaving out the major catechol (2-hydroxylated) metabolites. The reason is that these metabolites need to be protected from oxidation and epoxide formation following glucurolysis (134).

Whereas different research groups have determined the in vivo aromatase inhibition capacity of different compounds (26, 47, 135, 136), these groups have used different methods, for which reason the results are not directly comparable.

In a collaborative program between the Royal Marsden Hospital and our group, we determined the in vivo aromatase inhibition with a panel of first-, second-, and third-generation inhibitors (37, 38, 130, 137142). The results are summarized in Table 2. Notably, whereas first- and second-generation compounds in general caused aromatase inhibition <90%, the third-generation compounds caused a mean aromatase inhibition ≥98%. It is important to underline that the differences between first-, second-, and third-generation compounds concerning aromatase inhibition are not due to improvements in methodology but mirror a significant difference in efficacy between these compounds. The tracer method used in collaboration by our groups in Bergen and in London (head: Prof. M. Dowsett) has been used without any major changes from the time first published more than a decade ago (34). However, whereas a dose-response effect has been evaluated for several compounds (Table 2), a direct intrapatient comparison of two different drugs has been done for anastrozole and letrozole only. Notably, the method was able to discriminate the biochemical efficacy of these highly potent compounds, providing indirect evidence for its sensitivity (38).

Table 2.

Effects of different aromatase inhibitors and inactivators on whole-body aromatization

CompoundDrug dose in mg% Aromatase inhibitionRef.
Aminoglutethimide 250 qid 90.6% (138) 
Formestane (p.o.) 125 od/125 bid/250 od 72.3%/70%/57.3% (140) 
Formestane (i.m.) 250 2w/500 2w/500w 84.8%/91.9%/92.5% (137) 
Rogletimide 200 bid/400 bid/800 bid 50.6%/63.5%/73.8% (138) 
Fadrozole 1 bid/2 bid 82.4%/92.6% (37) 
Anastrozole 1 od/10 od 96.7%/98.1% (142) 
 1 od 97.3% (38) 
Letrozole 0.5 od/2.5 od 98.4%/98.9% (141) 
 2.5 od >99.1% (38) 
Exemestane 25 od 97.9% (130) 
CompoundDrug dose in mg% Aromatase inhibitionRef.
Aminoglutethimide 250 qid 90.6% (138) 
Formestane (p.o.) 125 od/125 bid/250 od 72.3%/70%/57.3% (140) 
Formestane (i.m.) 250 2w/500 2w/500w 84.8%/91.9%/92.5% (137) 
Rogletimide 200 bid/400 bid/800 bid 50.6%/63.5%/73.8% (138) 
Fadrozole 1 bid/2 bid 82.4%/92.6% (37) 
Anastrozole 1 od/10 od 96.7%/98.1% (142) 
 1 od 97.3% (38) 
Letrozole 0.5 od/2.5 od 98.4%/98.9% (141) 
 2.5 od >99.1% (38) 
Exemestane 25 od 97.9% (130) 

NOTE: All values are determined by the same assay at the Academic Department of Biochemistry, Royal Marsden Hospital, London, United Kingdom (Head: Prof. M. Dowsett) and the Breast Cancer Research Group at the Haukeland University Hospital in Bergen, Norway (Head: Prof. P.E. Lønning).

Abbreviations: od, once daily; bid, twice daily; qid, four times daily; w, weekly; 2w, every 2 weeks.

This important question may be addressed in two different ways. (a) Are we able to differentiate potential differences between compounds and, if so, are those differences related to clinical efficacy? (b) Do we have evidence suggesting any correlation between the degree of estrogen suppression and clinical outcome among individual patients treated with the same compound?

Based on the tracer results reported above, aromatase inhibitors may be divided into contemporary third-generation compounds (anastrozole, exemestane, and letrozole), causing ≥98% inhibition, and the first- and second-generation compounds, causing 85% to 90% inhibition. Whereas the trials comparing second-generation compounds to progestins and tamoxifen included a limited number of patients by today's standards (143147), there is no indication from these data suggesting improved clinical efficacy of any of these aromatase inhibitors compared with conventional treatment. In contrast, despite some diverging observations between individual trials, in general the superiority of the third-generation compounds has been established (see ref. 148 for a detailed review of these trials). Notably, whereas we found letrozole to enhance aromatase inhibition and plasma E1 as well as E1S suppression when compared with anastrozole in a crossover-designed study (Fig. 5; ref. 38), it is not clear whether this difference translates into an improved clinical efficacy (129).

Two articles have addressed the issue of correlation between degree of plasma estrogen suppression and clinical outcome (149, 150). The first study (149) was conducted more than 20 years ago during the pioneering period of development of aminoglutethimide; the second was reported more recently, and compared the degree of estrogen suppression to clinical outcome in patients treated with formestane (150). Notably, in that study (150) plasma estrogen levels were measured without doing high-performance liquid chromatography purification (57) of the samples before RIA, and the contrast between their finding (mean estrogen suppression of 40-60%) and the degree of aromatase inhibition (85-90%) recorded with tracer methods (151) suggests estrogen measurement in the study done by Bajetta et al. (150) to be disturbed by cross-reacting metabolites.

At this stage, the issue whether there may be any correlation between degree of estrogen suppression and the likelihood of achieving a response among individuals treated with the same aromatase inhibitor remains unsettled. Taking into account the high number of potential mechanisms of resistance to endocrine therapy in general (172), such a study, apart from requiring suitable assays combining the requirement of analytic simplicity and high sensitivity, would need a substantial number of patients to address the issue properly. Even if a correlation could be found, it remains unlikely that such a test would be routinely implemented to select patients for therapy or to define patient drug dosing at an individual level.

However, another interesting question remains to be addressed. Realizing that premenopausal as well as postmenopausal women with breast cancer respond to estrogen suppression, and that the third-generation inhibitors are more effective compared with the less potent first- and second-generation ones, these observations indirectly suggest a dose-response relationship between degree of estrogen suppression and clinical efficacy. Notably, long-term estrogen deprivation of hormone-sensitive MCF-7 cells in vitro may sensitize these cells to E2 concentrations 10−4 to 10−4−5 times the concentration normally required for maximal growth stimulation (152). Assuming third-generation aromatase inhibitors and inactivators to reduce estrogen production by 98% to 99%, we may ask the question whether a more potent inhibition (e.g., 99.9%) will cause a better clinical outcome. And if so, should such a therapy be implemented following relapse on a third-generation inhibitor, or given as first-line treatment? Would more aggravated estrogen suppression further sensitize the tumor cells to subsequent additive therapy with estrogens in pharmacologic doses (see later)? Whereas these questions are of clinical as well as biological relevance, our main problem is the technical incapacity to assess the endocrine effects of aggravated hormone suppression in vivo with the methods available today.

Lack of cross-resistance between aromatase inhibitors and inactivators. An interesting clinical observation is the rather surprising finding of a lack of complete cross-resistance between nonsteroidal aromatase inhibitors and the steroidal aromatase inactivators. The issue has been reviewed in detail elsewhere (153); only the key points will be summarized here. Whereas the response rate in general is low, there is no doubt that a significant number of patients may achieve durable stable disease if treated with a steroidal inactivator following a nonsteroidal compound; this effect is not explained through a more extensive suppression of plasma estrogens or total body aromatization. The most obvious reason for non-cross-resistance between steroidal and nonsteroidal drugs is the major difference in their biochemical structure. Another mechanism could be the slight androgenic effects caused by the steroidal compounds or some of their metabolites, illustrated by the dose-dependent drop in sex hormone binding globulin seen with exemestane due to its 17-hydrometabolite (57, 154). Thus, an important aim for future studies is to explore the mechanisms behind this interesting observation.

From what has been discussed above, it seems unlikely that we should be able to detect differences in plasma or tissue estrogen levels between groups of patients treated with third-generation aromatase inhibitors and inactivators. However, any difference with respect to clinical effects should be reflected in different biological effects at the tumor level. Thus, an issue for future studies could be to assess alterations in gene transcription in relation to treatment with compounds of the different classes using techniques like cDNA microarrays (155, 156).

Estrogen suppression and selective estrogen receptor modulators: combination versus sequential therapy. Whereas attempts to combine different types of endocrine therapy have in general been unsuccessful (157163), lessons from some recent clinical studies provide important information to our understanding of the mechanisms by which estrogen suppression provides antitumor effects in vivo. The combination of tamoxifen and a luteinizing hormone–releasing hormone analogue was found superior to tamoxifen monotherapy in treatment of premenopausal patients suffering from metastatic breast cancer (30, 164). Whereas the issue of mechanisms of action and resistance to selective estrogen receptor modulators is beyond the scope of this article, certain biological aspects are of interest to our understanding of the antitumor effects of estrogen deprivation and will therefore be discussed here. Whereas tamoxifen is an active compound in premenopausal women (165), due to disturbance of ovarian function, it increases plasma E2 levels by a factor of 2 to 4 (166169). Thus, it has been questioned whether this effect could be detrimental to the clinical outcome, consistent with the finding that addition of a luteinizing hormone–releasing hormone analogue improved tamoxifen efficacy. In contrast, tamoxifen does not influence plasma levels of E2 in postmenopausals (170). In the recent Arimidex, Tamoxifen, Alone or in Combination study, anastrozole was found superior to tamoxifen monotherapy preventing relapses (31). Here, tamoxifen and anastrozole in combination did not improve outcome compared with tamoxifen monotherapy, suggesting a detrimental effect of tamoxifen in postmenopausal patients treated with anastrozole. Taken together, these results are consistent with the hypothesis that plasma E2 in premenopausal concentrations may be detrimental by competing with tamoxifen for the estrogen receptor. Contrary, in postmenopausal women experiencing supralow estrogen levels during treatment with an aromatase inhibitor, the estrogen agonistic effect of tamoxifen (171) may be detrimental. Thus, combining these data provides important information to our understanding of the “antiestrogenic” effects of selective estrogen receptor modulators in vivo.

A major obstacle to the clinical use of endocrine therapy is the occurrence of primary or acquired endocrine resistance. Whereas the issue in general has been discussed in several recent publications (172178) and would be beyond the scope of this article, we will briefly summarize certain topics of relevance to resistance to estrogen deprivation in general and treatment with aromatase inhibitors in particular.

Contemporary clinical research is focusing on optimizing adjuvant treatment with aromatase inhibitors (the design of the major studies is summarized in Fig. 6), taking into consideration important issues like potential differences regarding compound efficacy, drug administration (sequential treatment versus monotherapy), duration of therapy, and side effects. Although several trials involving aromatase inhibitors in the adjuvant setting are still ongoing, three major adjuvant studies involving anastrozole, letrozole, and exemestane have been published recently (31, 39, 40). In conclusion, all three trials reported superiority of aromatase inhibitors either given alone (Arimidex, Tamoxifen, Alone or in Combination) or in sequence with tamoxifen (OEXE-31 and MA-17) compared with tamoxifen alone.

Fig. 6.

Adjuvant trials evaluating the effects of aromatase inhibitors or inactivators in early breast cancer.

Fig. 6.

Adjuvant trials evaluating the effects of aromatase inhibitors or inactivators in early breast cancer.

Close modal

Thus, future research exploring the mechanisms of action and resistance to aromatase inhibitors may potentially lead to even more effective strategies. Here, the potential differences between steroidal aromatase inactivators (like exemestane) and the nonsteroidal aromatase inhibitors, manifested by the lack of total cross-resistance between compounds of the two classes (179), merit special consideration. The fact that exemestane, in addition to being a potent aromatase inactivator, expresses weak androgen-agonistic effects through its main metabolite in vivo (57) indicates that exemestane may provide additional mechanisms acting in concert with estrogen deprivation (180). In addition, recent findings by Dr. Brodie's group, suggesting combined treatment with exemestane and tamoxifen to be superior to either tamoxifen or exemestane given as monotherapy in a mouse tumor model (181), underline the importance of exploring the consequences of structural differences between aromatase inhibitors and inactivators.

The extension of basic biological knowledge, together with methodologic improvements, offers the possibility to explore the mechanisms of therapy resistance in vivo in a manner we could just dream about 10 to 20 years ago (155, 182, 183). Importantly, whereas experimental data may generate hypotheses considering the mechanisms of therapy resistance, the proof is to verify the concepts in vivo—by translational research. Here, we will focus on selected issues including the mechanisms of estrogen sensitization, cross-talks between hormonal regulation and other growth factor pathways, and the potential of aggravated aromatase inhibition.

Several authors have shown that MCF-7 cells grown in culture may adapt to a low estrogen environment by developing hormonal hypersensitivity (152, 184, 185), achieving maximal growth stimulation by an E2 concentration 10−4 to 10−5 times the concentration needed for the mother cell line. Whereas the mechanism is not fully understood, several signaling pathways, including mitogen-activated protein kinase, seem to be involved (186). Estrogen stimulation in vitro is following a bell-shaped curve inasmuch as estrogens in concentrations above the level required for maximal growth stimulation inhibit cellular growth. This bell-shaped profile was also recorded for the hypersensitized cells, inasmuch as these cells could be growth inhibited by adding E2 in concentrations that would have stimulated the growth of the wild-type cells. The concept is substantiated through clinical observations: before the era of contemporary endocrine therapy, estrogens in high doses were used for breast cancer therapy (187189). Based on the hypothesis that estrogen deprivation could sensitize tumors to treatment with estrogens in pharmacologic doses, we exposed patients whose tumors had developed resistance to aromatase inhibitors to estrogens in high doses (diethylstilbestrol, 5 mg thrice daily). During this regimen, we recorded an objective response in 10 of 29 patients, some of them lasting for >2 years (190). Our challenge is to explore molecular changes in tumors exposed to such therapy elucidating the mechanism of action. Exact knowledge about these mechanisms, based on translational research, may open for new therapeutic strategies.

There is a growing body of literature suggesting that cross-talk between growth factors and estrogen receptors may contribute to endocrine resistance (176, 186, 191, 192). Of particular interest are some recent findings with respect to human epidermal growth factor receptor 2 overexpression, suggesting this may reduce responsiveness to tamoxifen (193) but not to aromatase inhibition (194, 195). Of particular interest is an article by Zhu et al. (195), revealing treatment with aromatase inhibitors to eradicate human epidermal growth factor receptor 2–positive cells in vivo. In addition, recent findings by Modlich et al. (196) suggest that the time points for any gene expression analyses are very critical as immediate changes in gene expression might occur after the initiation of therapy. All in all, a better understanding of the multidirectional cross-talk between growth factor pathways, the estrogen receptors (both at the genomic and nongenomic level), and estrogen receptor cofactors is currently one of the most promising scientific attempts to overcome endocrine resistance (191).

A final issue relates to aggravated hormone suppression. Prostaglandin E2, the product of cyclooxygenase 2 (COX-2), has been shown to be one of the most important factors controlling aromatase expression in vitro via promoter II (109), and aromatase expression is strongly correlated with COX-2 expression in series of breast cancer patients (61). Whereas COX-2–derived signals influence cell cycle progression, invasiveness, and angiogenesis in preclinical trials (197), the potential influence on estrogen synthesis provided the main background for implementing celecoxib into adjuvant and breast cancer prevention trials in concert with aromatase inhibitors. This approach, however, has several pitfalls. Whereas COX-2 overexpression has been associated with a significant inferior disease-free survival in subsets of breast cancer patients (see ref. 197 for references), any association between expression of a certain biological variable and prognosis could be misleading regarding biological interpretation. Often, a prognostic factor may be just a covariate to other biological variables (182), illustrated by the finding that expression of the estrogen receptor—the target for one of the most potent mitogens in breast cancer—has been associated with an improved prognosis also in breast cancer patients not exposed to adjuvant endocrine therapy (198, 199). Thus far, there has been no study reporting intratumor estrogen levels in patients treated with a COX-2 inhibitor. Even if the concept—combining a COX-2 inhibitor with an aromatase inhibitor—may improve therapeutic outcome, there is a need to explore the mechanisms determining intratumor hormone disposition (as well as the effects on other potential targets).

As shown in this review, translational research has played a key role during the successful development of aromatase inhibitors for breast cancer treatment. Following the implementation of aromatase inhibitors and inactivators of the third-generation as a first-line therapy in metastatic breast cancer, recent findings of large studies evolving the role for aromatase inhibitors in the adjuvant setting have caused optimism. However, whereas the adjuvant studies provided further evidence for the superiority of aromatase inhibitors compared with tamoxifen, the clinical improvements are in fact marginal, although statistically significant. Moreover, these studies are not designed to elucidate fundamental biological mechanisms leading to the major problem in the clinical use of these drugs: endocrine resistance. However, with the merging knowledge from basic research, together with method developments in molecular biology, we are now facing the opportunity of exploring the mechanisms of resistance to estrogen deprivation. Again, carefully designed translational research protocols are the most promising tools of physicians and basic scientists to change the situation for breast cancer patients fundamentally.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1
Beatson GT. On the treatment of inoperable cases of carcinoma of the mamma. Suggestions for a new method of treatment with illustrative cases.
Lancet
1896
;
2
:
104
–7.
2
Dao TL, Huggins C. Bilateral adrenalectomy in the treatment of cancer of the breast.
Arch Surg
1955
;
71
:
645
–57.
3
Luft R, Olivecrona H, Sjögren B. Hypophysektomy in man.
Nord Med
1952
;
14
:
351
–4.
4
Binnie GG. Regression of tumors following treatment by stilboestrol and X-ray therapy, with notes on case of breast tumour which regressed with stilboestrol alone.
Br J Radiol
1944
;
17
:
42
–5.
5
Haddow A, Watkinson JM, Paterson E. Influence of synthetic oestrogens upon advanced malignant disease.
Br Med J
1944
;
2
:
393
–8.
6
Taylor SG, Ayer JP, Morris RS. Cortical steroids in treatment of cancer.
JAMA
1956
;
144
:
1058
–64.
7
Nosaquo ND. Androgens and estrogens in the treatment of disseminated mammary carcinoma.
JAMA
1960
;
172
:
135
–47.
8
Greenspan EM. Regression of metastatic hepatomegaly from mammary carcinoma. Cytotoxic combination chemotherapy.
NY State J Med
1964
;
64
:
2442
–9.
9
DeCourmelles FV. La radiotherapie indirecte ou dirigée par les correlation organique.
Arch Elect Med
1922
;
32
:
264
.
10
Ulrich P. Testosterone (hormone male) et son role possible dans le traitment de certains cancers du sein.
Acta Union Int Centre Cancer
1939
;
4
:
377
–80.
11
Douglas M. The treatment of advanced breast cancer by hormone therapy.
Br J Cancer
1952
;
6
:
32
–6.
12
Huggins C, Dao TL-Y. Adrenalectomy and oophorectomy in treatment of advanced carcinoma of the breast.
JAMA
1953
;
151
:
1388
–94.
13
Cash R, Brough AJ, Cohen MNP, Satoh PS. Aminoglutethimide (Elipten-Ciba) is an inhibitor of adrenal steroidogenesis: mechanism of action and therapeutic trial.
J Clin Endocrinol Metab
1967
;
27
:
1239
–48.
14
Cole MP, Jones CTA, Todd IDH. A new anti-oestrogenic agent in late breast cancer. An early clinical appraisal of ICI46474.
Br J Cancer
1971
;
25
:
270
–5.
15
Rustgi AK. Translational research: What is it?
Gastroenterology
1999
;
116
:
1285
.
16
Friedman HS, Bigner SH, Bigner DD. Cyclosphosphamide therapy of medulloblastoma—from the laboratory to the clinic and back again (and again and again).
J Neurooncol
1995
;
24
:
103
–8.
17
Jensen EV, DeSombre ER, Jungblut PP. Estrogen receptors in hormone-responsive tissues and tumors. In: Wissler RW, Dao TL, Wood S Jr, editors. Endogenous factors influencing host-tumor balance. Chicago: University of Chicago Press; 1967. p. 15–30.
18
McGuire WL. Steroid receptors in human breast cancer.
Cancer Res
1978
;
38
:
4289
–91.
19
Horwitz KB, McGuire WL. Predicting response to endocrine therapy in human breast cancer: a hypothesis.
Science
1975
;
189
:
726
–7.
20
Dowsett M. Analysis of time to recurrence in the ATAC (arimidex, tamoxifen, alone or in combination) trial according to estrogen receptor and progesterone receptor status.
Breast Cancer Res Treat
2003
;
82
:
S7
.
21
Camacho AM, Brough AJ, Cash R, Wilroy RS. Adrenal toxicity associated with the administration of an anticonvulsant drug.
J Pediatr
1966
;
68
:
852
–3.
22
Schwarzel WC, Kruggel WG, Brodie HJ. Studies on the mechanism of estrogen biosynthesis: 8. The development of inhibitors of the enzyme system in human placenta.
Endocrinology
1973
;
92
:
866
–80.
23
Thompson EA Jr, Siiteri PK. The involvement of human placental microsomal cytochrome P-450 in aromatization.
J Biol Chem
1974
;
249
:
5373
–8.
24
Kofman S, Nagamani D, Buenger RF, Taylor SG. The use of prednisolone in the treatment of disseminated breast carcinoma.
Cancer
1958
;
11
:
226
–32.
25
Lemon HM. Prednisone therapy of advanced mammary cancer.
Cancer
1959
;
12
:
93
–107.
26
Santen RJ, Santner S, Davis B, et al. Aminoglutethimide inhibits extraglandular estrogen production in postmenopausal women with breast carcinoma.
J Clin Endocrinol Metab
1978
;
47
:
1257
–65.
27
Samojlik E, Veldhuis JD, Wells SA, Santen RJ. Preservation of androgen secretion during estrogen suppression with aminoglutethimide in the treatment of metastatic breast carcinoma.
J Clin Invest
1980
;
65
:
602
–12.
28
Samojlik E, Santen RJ, Worgul TJ. Suppression of residual oestrogen production with aminoglutethimide in women following surgical hypophysectomy or adrenalectomy.
Clin Endocrinol (Oxf)
1984
;
20
:
43
–51.
29
Lønning PE. Stepwise estrogen suppression manipulating the estrostat.
J Steroid Biochem Mol Biol
2001
;
79
:
127
–32.
30
Klijn JGM, Blamey RW, Boccardo F, et al. Combined tamoxifen and luteinizing hormone-releasing hormone (LHRH) agonist versus LHRH agonist alone in premenopausal advanced breast cancer: a meta-analysis of four randomized trials.
J Clin Oncol
2001
;
19
:
343
–53.
31
Baum M, Buzdar AU, Cuzick J, et al. Anastrozole alone or in combination with tamoxifen versus tamoxifen alone for adjuvant treatment of postmenopausal women with early breast cancer: first results of the ATAC randomised trial.
Lancet
2002
;
359
:
2131
–9.
32
Decensi A, Gandini S, GuerrieriGonzaga A, et al. Effect of blood tamoxifen concentrations on surrogate biomarkers in a trial of dose reduction in healthy women.
J Clin Oncol
1999
;
17
:
2633
–8.
33
Jordan VC. Tamoxifen: Too much of a good thing?
J Clin Oncol
1999
;
17
:
2629
–30.
34
Jacobs S, Lønning PE, Haynes B, Griggs L, Dowsett M. Measurement of aromatisation by a urine technique suitable for the evaluation of aromatase inhibitors in vivo.
J Enzyme Inhib
1991
;
4
:
315
–25.
35
Batzl C, Hausler A, Schieweck K, Lang M, Trunet P. Pharmacology of nonsteroidal aromatase inhibitors. In: Pasqualini J, Katzenellenbogen B, editors. Hormone-dependent cancer. New York: Marcel Dekker, Inc.; 1996. p. 155–68.
36
Batzl-Hartmann C, Evans DB, Bhatnagar A. Comparative aromatase enzyme kinetic studies on fadrozole, formestane, letrozole, anastrozole and exemestane, San Antonio Breast Cancer Conference, San Antonio, TX, USA, 2003.
37
Lønning PE, Jacobs S, Jones A, et al. The influence of CGS 16949A on peripheral aromatisation in breast cancer patients.
Br J Cancer
1991
;
63
:
789
–93.
38
Geisler J, Haynes B, Anker G, Dowsett M, Lønning PE. Influence of letrozole (Femara) and anastrozole (Arimidex) on total body aromatization and plasma estrogen levels in postmenopausal breast cancer patients evaluated in a randomized, cross-over-designed study.
J Clin Oncol
2002
;
20
:
751
–7.
39
Goss PE, Ingle JN, Martino S, et al. A randomized trial of letrozole in postmenopausal women after five years of tamoxifen for early-stage breast cancer.
N Engl J Med
2003
;
349
:
1793
–802.
40
Coombes RC, Hall E, Gibson LJ, et al. A randomized trial of exemestane after two to three years of tamoxifen therapy in postmenopausal women with primary breast cancer.
N Engl J Med
2004
;
350
:
1081
–92.
41
Santen RJ, Lipton A, Kendall J. Successful medical adrenalectomy with aminoglutethimide.
JAMA
1974
;
230
:
1661
–5.
42
Samojlik E, Santen RJ, Wells SA. Adrenal suppression with aminoglutethimide. II. Differential effects of aminoglutethimide on plasma androstenedione and estrogen levels.
J Clin Endocrinol Metab
1977
;
45
:
480
–7.
43
Lønning PE, Kvinnsland S. Mechanisms of action of aminoglutethimide as endocrine therapy of breast cancer.
Drugs
1988
;
35
:
685
–710.
44
Brodie AMH, Schwarzel WC, Shaikh AA, Brodie HJ. The effect of an aromatase inhbitor, 4-hydroxy-androstene-3,17-dione, on estrogen-dependent processes in reproduction and breast cancer.
Endocrinology
1977
;
100
:
1684
–95.
45
Brodie AMH, Longcope C. Inhibition of peripheral aromatization by aromatase inhibitors, 4-hydroxy- and 4-acetoxy-androstene-3,17-dione.
Endocrinology
1980
;
106
:
19
–21.
46
Volk H, Deupree RH, Goldenberg IS, et al. A dose response evaluation of δ-1-testololactone in advanced breast cancer.
Cancer
1974
;
33
:
9
–13.
47
Barone RM, Shamonki IM, Siiteri PK, Judd HL. Inhibition of peripheral aromatization of androstenedione to estrone in postmenopausal women with breast cancer using Δ1-testoloclactone.
J Clin Endocrinol Metab
1979
;
49
:
672
–6.
48
Coombes RC, Goss P, Dowsett M, Gazet J-C, Brodie A. 4-Hydroxyandrostenedione in treatment of postmenopausal patients with advanced breast cancer.
Lancet
1984
;
ii
:
1237
–9.
49
Dowsett M, Goss PE, Powles TJ, et al. Use of the aromatase inhibitor 4-hydroxyandrostenedione in postmenopausal breast cancer: Optimization of therapeutic dose and route.
Cancer Res
1987
;
47
:
1957
–61.
50
Dowsett M, Cunningham DC, Stein RC, et al. Dose-related endocrine effects and pharmacokinetics of oral and intramuscular 4-hydroxyandrostenedione in postmenopausal breast cancer patients.
Cancer Res
1989
;
49
:
1306
–12.
51
Brodie AM, Garrett WM, Hendrickson JR, et al. Inactivation of aromatase in vitro by 4-hydroxy-4-androstene-3,17-dione and 4-acetoxy-4-androstene-3,17-dione and sustained effects in vivo.
Steroids
1981
;
38
:
693
–702.
52
Covey DF, Hood WF. Enzyme-generated intermediates derived from 4-androstene-3,6,17-trione and 1,4,6-androstatriene-3,17-dione cause a time-dependent decrease in human placental aromatase activity.
Endocrinology
1981
;
108
:
1597
–9.
53
di Salle E, Ornati G, Giudici D, et al. Exemestane (FCE 24304), a new steroidal aromatase inhibitor.
J Steroid Biochem Mol Biol
1992
;
43
:
137
–43.
54
di Salle E, Giudici D, Briatico G, Ornati G. Novel irreversible aromatase inhibitors.
Ann N Y Acad Sci
1990
;
595
:
357
–67.
55
Brodie AM, Njar VC. Aromatase inhibitors in advanced breast cancer: mechanism of action and clinical implications.
J Steroid Biochem Mol Biol
1998
;
66
:
1
–10.
56
Miller WR, Dixon JM. Local endocrine effects of aromatase inhibitors within the breast.
J Steroid Biochem Mol Biol
2001
;
79
:
93
–102.
57
Johannessen DC, Engan T, Di Salle E, et al. Endocrine and clinical effects of exemestane (PNU 155971), a novel steroidal aromatase inhibitor, in postmenopausal breast cancer patients: a phase I study.
Clin Cancer Res
1997
;
3
:
1101
–8.
58
Dowsett M, Mehta A, King N, et al. An endocrine and pharmacokinetic study of four oral doses of formestane in postmenopausal breast cancer patients.
Eur J Cancer
1992
;
28
:
415
–20.
59
Murray R, Pitt P. Aromatase inhibition with 4-OH androstenedione after prior aromatase inhibition with aminoglutethimide in women with advanced breast cancer.
Breast Cancer Res Treat
1995
;
35
:
249
–53.
60
Purba HS, Bhatnagar A. A comparison of methods measuring aromatase activity in human placenta and rat ovary.
J Enzyme Inhib
1990
;
4
:
169
–78.
61
Brodie AM, Lu Q, Long BJ, et al. Aromatase and COX-2 expression in human breast cancers.
J Steroid Biochem Mol Biol
2001
;
79
:
41
–7.
62
Chen S, Zhou D, Okubo T, Kao Y-C, Yang C. Breast tumor aromatase: functional role and transcriptional regulation.
Endocrine-Rel Cancer
1999
;
6
:
149
–56.
63
Chen SU, Zhou DJ, Yang C, et al. Modulation of aromatase expression in human breast tissue.
Journal of Steroid Biochemistry and Molecular Biology
2001
;
79
:
35
–40.
64
Brodie A, Lu Q, Liu Y, Long B. Aromatase inhibitors and their antitumor effects in model systems.
End Rel Cancer
1999
;
6
:
205
–10.
65
Miller WR, Dixon JM. Antiaromatase agents: preclinical data and neoadjuvant therapy.
Clin Breast Cancer
2000
;
1
(Suppl 1):
S9
–14.
66
Kochak GM, Mangat S, Mulagha MT, et al. The pharmacodynamic inhibition of estrogen synthesis by fadrozole, an aromatase inhibitor, and its pharmacokinetic disposition.
J Clin Endocrinol Metab
1990
;
71
:
1349
–55.
67
Sioufi A, Gauducheau N, Pineau V, et al. Absolute bioavailability of letrozole in healthy postmenopausal women.
Biopharm Drug Dispos
1997
;
18
:
779
–89.
68
Pfister C, Dowsett M, Iveson T, et al. Pharmacokinetics of new aromatase inhibitor CGS 20267.
Eur J Drug Metab Pharmacokinet
1993
;
18
:
117
.
69
Yue W, Zhou D, Chen S, Brodie A. A new nude mouse model for postmenopausal breast cancer using MCF-7 cells transfected with the human aromatase gene.
Cancer Res
1994
;
54
:
5092
–5.
70
Yue W, Brodie A. MCF-7 human breast carcinomas in nude mice as a model for evaluating aromatase inhibitors.
J Steroid Biochem Mol Biol
1993
;
44
:
671
–3.
71
Lu Q, Yue W, Wang J, Liu Y, Brodie A. The effects of aromatase inhibitors and antiestrogens in the nude mouse model.
Breast Cancer Res Treat
1998
;
50
:
63
–71.
72
Fisher CR, Gracves KH, Parlow AF, Simpson ER. Characterization of mice deficient in aromatase (ArKO) because of targeted disruption of the CYP19 gene.
Proc Natl Acad Sci U S A
1998
;
95
:
6965
–70.
73
Couse JF, Korach KS. Estrogen receptor null mice: what have we learned and where will they lead us?
Endocr Rev
1999
;
20
:
358
–417.
74
Lønning PE, Helle SI, Johannessen DC, et al. Relations between sex hormones, sex hormone binding globulin, insulin-like growth factor-I and insulin-like growth factor binding protein-1 in post-menopausal breast cancer patients.
Clin Endocrinol
1995
;
42
:
23
–30.
75
Geisler J, Lien EA, Ekse D, Lønning PE. Influence of aminoglutethimide on plasma levels of estrone sulphate and dehydroepiandrosterone sulphate in postmenopausal breast cancer patients.
J Steroid Biochem Mol Biol
1997
;
63
:
53
–8.
76
vanLandeghem AAJ, Poortman J, Nabuurs M, Thijssen JHH. Endogenous concentration and subcellular distribution of estrogens in normal and malignant breast tissue.
Cancer Res
1985
;
45
:
2900
–4.
77
Geisler J, Detre S, Berntsen H, et al. Influence of neoadjuvant anastrozole (Arimidex) on intratumoral estrogen levels and proliferation markers in patients with locally advanced breast cancer.
Clin Cancer Res
2001
;
7
:
1230
–6.
78
Lønning PE, Ekse D. A sensitive assay for measurement of plasma estrone sulphate in patients on treatment with aromatase inhibitors.
J Steroid Biochem Mol Biol
1995
;
55
:
409
–12.
79
Santner SJ, Leszcynski D, Wright C, et al. Estrone sulfate: A potential source of estradiol in human breast cancer tissue.
Breast Cancer Res Treat
1986
;
7
:
35
–44.
80
Pizzagalli F, Varga Z, Huber RD, et al. Identification of Steroid Sulfate Transport Processes in the Human Mammary Gland.
J Clin Endocrinol Metab
2003
;
88
:
3902
–12.
81
Chetrite GS, Cortes-Prieto J, Philippe JC, Wright F, Pasqualini J. Comparison of estrogen concentrations, estrone sulfatase and aromatase activities in normal, and in cancerous, human breast tissues.
J Steroid Biochem Mol Biol
2000
;
72
:
23
–7.
82
Poutanen M, Isomaa V, Peltoketo H, Vihko R. Role of 17β-hydroxysteroid dehydrogenase type 1 in endocrine and intracrine estradiol biosynthesis.
J Steroid Biochem Mol Biol
1995
;
55
:
525
–32.
83
Sasano H, Frost AR, Saitoh R, et al. Aromatase and 17β-hydroxysteroid dehydrogenase type 1 in human breast carcinoma.
J Clin Endocrinol Metab
1996
;
81
:
4042
–6.
84
Masamura S, Santner SJ, Gimotty P, George J, Santen RJ. Mechanism for maintenance of high breast tumor estradiol concentrations in the absence of ovarian function: Role of very high affinity tissue uptake.
Breast Cancer Res Treat
1997
;
42
:
215
–26.
85
James VHT, Reed MJ, Adams EF, et al. Oestrogen uptake and metabolism in vivo. In: Beck JS, editors. Oestrogens and the Human Breast.
Proc Roy Soc Edinb
1989
;
95B
:
185
–93.
86
Miller WR. Importance of intratumour aromatase, and its susceptibility to inhibitors. In: Dowsett M, editors. Aromatase inhibition—then, now and tomorrow. London: Partenon Publishing Group; 1994. p. 43–53.
87
Frisch RE, Canick JA, Tulchinsky D. Human fatty marrow aromatizes androgen to estrogen.
J Clin endocrinol Metab
1980
;
51
:
394
–6.
88
Matsumine H, Hirato K, Yanaihara T, Tamada T, Yoshida M. Aromatization by skeletal muscle.
J Clin Endocrinol Metab
1986
;
63
:
717
–20.
89
Nimrod A, Ryan KJ. Aromatization of androgens by human abdominal and breast fat tissue.
J Clin Endocrinol Metab
1975
;
40
:
367
–72.
90
Osawa Y, Tochigi B, Higashiyama T, et al. Multiple forms of aromatase and response of breast cancer aromatase to antiplacental aromatase II antibodies.
Cancer Res
1982
;
42
(Suppl):
3299
–306s.
91
Perel E, Killinger DW. The interconversion and aromatization of androgens by human adipose tissue.
J Steroid Biochem
1979
;
10
:
623
–7.
92
Schweikert HU, Milewich L, Wilson JD. Aromatization of androstenedione by isolated human hairs.
J Clin Endocrinol Metab
1975
;
40
:
413
–7.
93
Schweikert HU, Milewich L, Wilson JD. Aromatization of androstenedione by cultured human fibroblasts.
J Clin Endocrinol Metab
1976
;
43
:
785
–95.
94
Smuk M, Schwers J. Aromatization of androstenedione by human adult liver in vitro.
J Clin Endocrinol Metab
1977
;
45
:
1009
–12.
95
Tilson-Malett N, Santner SJ, PD F, Santen RJ. Biological significance of aromatase activity in human breast tumors.
J Clin Endocrinol Metab
1983
;
57
:
1125
–8.
96
Brodie A, Long B, Lu Q. Aromatase expression in the human breast.
Breast Cancer Res Treat
1998
;
49
:
S85
–91.
97
Santen RJ, Martel J, Hoagland M, et al. Demonstration of aromatase activity and its regulation in breast tumor and benign breast fibroblasts.
Breast Cancer Res Treat
1998
;
49
:
S93
–9.
98
Sasano H, Murakami H. Immunolocalization of aromatase in human breast disorders using different antibodies.
Breast Cancer Res Treat
1998
;
49
:
S79
–84.
99
Shenton KC, Dowsett M, Lu Q, et al. Comparison of biochemical aromatase activity with aromatase immunohistochemistry in human breast carcinomas.
Breast Cancer Res Treat
1998
;
49
:
S101
–7.
100
Silva MC, Rowlands MG, Dowsett M, et al. Intratumoral aromatase as a prognostic factor in human breast carcinoma.
Cancer Res
1989
;
49
:
2588
–91.
101
Miller WR, Anderson TJ, Jack WJL. Relationship between tumour aromatase activity, tumour characteristics and response to therapy.
J Steroid Biochem Mol Biol
1990
;
37
:
1055
–9.
102
Bolt HM. Metabolism of estrogens—natural and synthetic.
J Clin Pharma Ther
1979
;
4
:
155
–81.
103
Lønning PE, Kvinnsland S, Thorsen T, Ueland PM. Alterations in the metabolism of oestrogens during treatment with aminoglutethimide in breast cancer patients. Preliminary findings.
Clin Pharmacokinet
1987
;
13
:
393
–406.
104
Sebastian S, Bulun SE. A highly complex organization of the regulatory region of the human CYP19 (aromatase) gene revealed by the Human Genome Project.
J Clin Endocrinol Metab
2001
;
86
:
4600
–2.
105
Simpson ER, Michael MD, Agarwal VR, et al. Cytochromes P450 11: expression of the CYP19 (aromatase) gene: an unusual case of alternative promoter usage.
Faseb J
1997
;
11
:
29
–36.
106
Simpson ER, Clyne C, Rubin G, et al. Aromatase—a brief overview.
Annu Rev Physiol
2002
;
64
:
93
–127.
107
Sebastian S, Takayama K, Shozu M, Bulun SE. Cloning and characterization of a novel endothelial promoter of the human CYP19 (aromatase P450) gene that is up-regulated in breast cancer tissues.
Mol Endocrinol
2002
;
16
:
2243
–54.
108
Harada N, Utsumi T, Takagi Y. Molecular and epidemiological analysis of abnormal expression of aromatase in breast cancer.
Pharmacogenetics
1995
;
5
:
S59
–64.
109
Zhao Y, Agarwal VR, Mendelson CR, Simpson ER. Estrogen biosynthesis proximal to a breast tumor is stimulated by PGE2 via cyclic AMP, reading to activation of promoter II of the CYP19 (aromatase) gene.
Endocrinology
1996
;
137
:
5739
–42.
110
Geisler J. Breast cancer tissue estrogens and their manipulation with aromatase inhibitors and inactivators.
J Steroid Biochem Mol Biol
2003
;
86
:
245
–53.
111
Lønning PE, Dowsett M, Powles TJ. Postmenopausal estrogen synthesis and metabolism: Alterations caused by aromatase inhibitors used for the treatment of breast cancer.
J Steroid Biochem Mol Biol
1990
;
35
:
355
–66.
112
Garland CF, Friedllander NJ, Khaw KT. Sex hormones and postmenopausal breast cancer: a prospective study in an adult community.
Am J Epidemiol
1992
;
135
:
1220
–30.
113
Helzlsouer KJ, Alberg AJ, Bush TL, Longcope C, Gordon GB. A prospective study of endogenous hormones and breast cancer.
Cancer Detect Prev
1994
;
18
:
79
–85.
114
Toniolo PG, Levitz M, Zeleniuch-Jacquotte A, et al. A prospective study of endogenous estrogens and breast cancer in postmenopausal women.
J Natl Cancer Inst
1995
;
87
:
190
–7.
115
Berrino F, Muti P, Micheli A, et al. Serum sex hormone levels after menopause and subsequent breast cancer.
J Natl Cancer Inst
1996
;
88
:
291
–6.
116
Thomas HV, Key TJ, Allen DS, et al. A prospective study of endogenous serum hormone concentrations and breast cancer risk in postmenopausal women on the island of Guernsey.
Br J Cancer
1997
;
76
:
401
–5.
117
Hankinson SE, Willett WC, Manson JE, et al. Plasma sex steroid hormone levels and risk of breast cancer in postmenopausal women.
J Natl Cancer Inst
1998
;
90
:
1292
–9.
118
Cauley JA, Lucas FL, Kuller LH, et al. Elevated serum estradiol and testosterone concentrations are associated with a high risk for breast cancer.
Ann Intern Med
1999
;
130
:
270
–7.
119
Dorgan JF, Longcope C, Stanczyk FZ, Stephenson HE, Hoover RN. Plasma sex steroid hormone levels and risk of breast cancer in postmenopausal women.
J Natl Cancer Inst
1999
;
91
:
380
–1.
120
Kabuto M, Akiba S, Stevens RG, Neriishi K, Land CE. A prospective study of estradiol and breast cancer in Japanese women.
Cancer Epidem Biomarkers Prev
2000
;
9
:
575
–9.
121
Bruning PF, Bonfrer JM, Hart AA. Non-protein bound oestradiol, sex hormone binding globulin, breast cancer and breast cancer risk.
Br J Cancer
1985
;
51
:
479
–84.
122
Reed MJ, Beranek PA, Cheng RW, Ghilchik MW, James VH. The distribution of oestradiol in plasma from postmenopausal women with or without breast cancer: relationships with metabolic clearance rates of oestradiol.
Int J Cancer
1985
;
35
:
457
–60.
123
Secreto G, Recchione C, Cavalleri A, Miraglia M, Dati V. Circulating levels of testosterone, 17β-oestradiol, luteinising hormone and prolactin in postmenopausal breast cancer patients.
Br J Cancer
1983
;
47
:
269
–75.
124
Siiteri PK, Simberg N, Murai J. Estrogens and breast cancer.
Ann N Y Acad Sci
1986
;
464
:
100
–5.
125
Bernstein L, Ross RK, Pike MC, Brown JB, Henderson BE. Hormone levels in older women: a study of post-menopausal breast cancer patients and healthy population controls.
Br J Cancer
1990
;
61
:
298
–302.
126
Dorgan JF, Longcope C, Franz C, et al. Endogenous sex hormones and breast cancer in postmenopausal women: Reanalysis of nine prospective studies.
J Natl Can Inst
2002
;
94
:
606
–16.
127
Key TJ, Appleby PN, Reeves GK, et al. Body mass index, serum sex hormones, and breast cancer risk in postmenopausal women.
J Natl Can Inst
2003
;
95
:
1218
–26.
128
Lønning PE, Geisler J, Johannessen DC, Ekse D. Plasma Estrogen Suppression with Aromatase Inhibitors Evaluated by a Novel, Sensitive Assay for Estrone Sulphate.
J Steroid Biochem Mol Biol
1997
;
61
:
255
–60.
129
Rose C, Vtoraya O, Pluzanska A, et al. An open randomised trial of second-line endocrine therapy in advanced breast cancer. comparison of the aromatase inhibitors letrozole and anastrozole.
Eur J Cancer
2003
;
39
:
2318
–27.
130
Geisler J, King N, Anker G, et al. In vivo inhibition of aromatization by exemestane, a novel irreversible aromatase inhibitor, in postmenopausal breast cancer patients.
Clin Cancer Res
1998
;
4
:
2089
–93.
131
Geisler J, Berntsen H, Lønning PE. A novel HPLC-RIA method for the simultaneous detection of estrone, estradiol and estrone sulphate levels in breast cancer tissue.
J Steroid Biochem Mol Biol
2000
;
72
:
259
–64.
132
Gurpide E, Mann J, Lieberman S. Analysis of open systems of multiple pools by administration of tracers at a constant rate or as a single dose as illustrated by problems involving steroid hormones.
J Clin Endocrinol Metab
1963
;
23
:
1155
–76.
133
Gurpide E, MacDonald PC, Wiele RLV, Lieberman S. Measurement of the rates of secretion and of peripheral metabolism of two interconvertible compounds: dehydroisoandrosterone-dehydroisoandrosterone sulfate.
J Clin Endocrinol Metab
1963
;
23
:
346
–54.
134
Lønning PE, Skulstad P, Sunde A, Thorsen T. Separation of urinary metabolites of radiolabelled estrogens in man by HPLC.
J Steroid Biochem Mol
Biol1989
;
32
:
91
–7.
135
Reed MJ, Lai LC, Owen AM, et al. Effect of treatment with 4-hydroxyandrostenedione on the peripheral conversion of androstenedione to estrone and in vitro tumor aromatase activity in postmenopausal women with breast cancer.
Cancer Res
1990
;
50
:
193
–6.
136
Wall Evd, Donker TH, Frankrijker Ed, et al. Inhibition of the in vivo conversion of androstenedione to estrone by the aromatase inhibitor vorozole in healthy postmenopausal women.
Cancer Res
1993
;
53
:
4563
–6.
137
Jones AL, MacNeill F, Jacobs S, et al. The influence of intramuscular 4-hydroxyandrostenedione on peripheral aromatisation in breast cancer patients.
Eur J Cancer
1992
;
28A
:
1712
–6.
138
MacNeill FA, Jones AL, Jacobs S, et al. The influence of aminoglutethimide and its analogue rogletimide on peripheral aromatisation in breast cancer.
Br J Cancer
1992
;
66
:
692
–7.
139
MacNeill FA, Jacobs S, Lønning PE, Powles TJ, Dowsett M. Combined treatment with 4-hydroxyandrostenedione and aminoglutethimide: Effects on aromatase inhibition and oestrogen suppression.
Br J Cancer
1994
;
69
:
1171
–5.
140
MacNeill FA, Jacobs S, Dowsett M, Lønning PE, Powles TJ. The effects of oral 4-hydroxyandrostenedione on peripheral aromatisation in post-menopausal breast cancer patients.
Cancer Chemother Pharmacol
1995
;
36
:
249
–54.
141
Dowsett M, Jones A, Johnston SR, et al. In vivo measurement of aromatase inhibition by letrozole (CGS 20267) in postmenopausal patients with breast cancer.
Clin Cancer Res
1995
;
1
:
1511
–5.
142
Geisler J, King N, Dowsett M, et al. Influence of anastrozole (Arimidex®), a selective, non-steroidal aromatase inhibitor, on in vivo aromatisation and plasma oestrogen levels in postmenopausal women with breast cancer.
Br J Cancer
1996
;
74
:
1286
–91.
143
Thürlimann B, Castiglione M, HsuSchmitz SF, et al. Formestane versus megestrol acetate in postmenopausal breast cancer patients after failure of tamoxifen: A phase III prospective randomised cross over trial of second-line hormonal treatment (SAKK 20/90).
Eur J Cancer
1997
;
33
:
1017
–24.
144
Buzdar AU, Smith R, Vogel C, et al. Fadrozole HCL (CGS-16949A) versus megestrol acetate treatment of postmenopausal patients with metastatic breast carcinoma. Results of two randomized double blind controlled multiinstitutional trials.
Cancer
1996
;
77
:
2503
–13.
145
Falkson CI, Falkson HC. A randomised study of CGS 16949A (fadrozole) versus tamoxifen in previously untreated postmenopausal patients with metastatic breast cancer.
Ann Oncol
1996
;
7
:
465
–9.
146
Thürlimann B, Beretta K, Bacchi M, et al. First-line fadrozole HCI (CGS 16949A) versus tamoxifen in postmenopausal women with advanced breast cancer—prospective randomised trial of the Swiss Group for Clinical Cancer Research SAKK 20/88.
Ann Oncol
1996
;
7
:
471
–9.
147
Pérez-Carrión R, Candel VA, Calabresi F, et al. Comparison of the selective aromatase inhibitor formestane with tamoxifen as first-line hormonal therapy in postmenopausal women with advanced breast cancer.
Ann Oncol
1994
;
5
:
S19
–24.
148
Lønning PE. Aromatase inhibitors and inactivators for breast cancer therapy.
Drugs Aging
2002
;
19
:
277
–98.
149
Santen RJ, Worgul TJ, Lipton A, Harvey H, Boucher A. Aminoglutethimide as treatment of postmenopausal women with advanced breast carcinoma.
Ann Intern Med
1982
;
96
:
94
–101.
150
Bajetta E, Zilembo N, Bichisao E, et al. Tumor response and estrogen suppression in breast cancer patients treated with aromatase inhibitors.
Ann Oncol
2000
;
11
:
1017
–22.
151
Jones AL, Powles TJ, Law M, et al. Adjuvant aminoglutethimide for postmenopausal patients with primary breast cancer: analysis at 8 years.
J Clin Oncol
1992
;
10
:
1547
–52.
152
Masamura S, Santner SJ, Heitjan DF, Santen RJ. Estrogen deprivation causes estradiol hypersensitivity in human breast cancer cells.
J Clin Endocrinol Metab
1995
;
80
:
2918
–25.
153
Lønning PE. Comparison between aromatase inhibitors and sequential use.
J Steroid Biochem Mol Biol
2003
;
86
:
275
–82.
154
Lønning PE, Paridaens R, Thürlimann B, Piscitelli G, Di Salle E. Exemestane experience in breast cancer treatment.
J Steroid Biochem Mol Biol
1997
;
61
:
151
–5.
155
Sorlie T, Perou CM, Tibshirani R, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications.
Proc Nat Acad Sci U S A
2001
;
98
:
10869
–74.
156
Sorlie T, Tibshirani R, Parker J, et al. Repeated observation of breast tumor subtypes in independent gene expression data sets.
Proc Natl Acad Sci U S A
2003
;
100
:
8418
–23.
157
Corkery J, Leonard RCF, Henderson IC, et al. Tamoxifen and aminoglutethimide in advanced breast cancer.
Cancer Res
1982
;
42
:
3409
–14s.
158
Smith IE, Harris AL, Morgan M, Gazet J-C, McKinna JA. Tamoxifen versus aminoglutethimide versus combined tamoxifen and aminoglutethimide in the treatment of advanced breast carcinoma.
Cancer Res
1982
;
42
:
3430
–3s.
159
Milsted R, Habeshaw T, Kaye S, et al. A randomized trial of tamoxifen versus tamoxifen with aminoglutethimide in postmenopausal women with advanced breast cancer.
Cancer Chemother Pharmacol
1985
;
14
:
272
–3.
160
Ingle JN, Green SJ, Ahmann DL, et al. Progress report on two clinical trials in women with advanced breast cancer. Trial I: tamoxifen versus tamoxifen plus aminoglutethimide; trial II: aminoglutethimide in patients with prior tamoxifen exposure.
Cancer Res
1982
;
42
(Suppl):
3461
–7s.
161
Alonso-Munoz MC, González MBO, Beltran-Fabregat M, et al. Randomized trial of tamoxifen versus aminoglutethimide and versus combined tamoxifen and aminglutethimide in advanced postmenopausal breast cancer.
Oncology
1988
;
45
:
350
–3.
162
Powles TJ, Gordon C, Coombes RC. Clinical trial of multiple endocrine therapy for metastatic and locally advanced breast cancer with tamoxifen-aminoglutethimide-danazol compared to tamosifen used alone.
Cancer Res
1982
;
42
(Suppl):
3458
–60s.
163
Hardy JR, Powles TJ, Judson IR, et al. Combination of tamoxifen, aminoglutethimide, danazol and medroxyprogesterone acetate in advanced breast cancer.
Eur J Cancer
1990
;
26
:
824
–7.
164
Klijn JGM, Beex LVAM, Mauriac L, et al. Combined treatment with buserelin and tamoxifen in premenopausal metastatic breast cancer: a randomized study.
J Natl Cancer Inst
2000
;
92
:
903
–11.
165
Jordan VC. Biochemical pharmacology of antiestrogen action.
Pharmacol Rev
1984
;
36
:
245
–76.
166
Sherman BM, Chapler FK, Crickard K, Wycoff D. Endocrine consequences of continuous antiestrogen therapy with tamoxifen in premenopausal women.
J Clin Invest
1979
;
64
:
398
–404.
167
Cohen I, Tepper R, Figer A, et al. Successful co-treatment with LHRH-agonist for ovarian over-stimulation and cystic formation in premenopausal tamoxifen exposure.
Breast Cancer Res Treat
1999
;
55
:
119
–25.
168
Groom GV, Griffiths K. Effect of the anti-oestrogen tamoxifen on plasma levels of luteinizing hormone, follicle-stimulating hormone, prolactin, oestradiol and progesterone in normal pre-menopausal women.
J Endocrinol
1976
;
70
:
421
–8.
169
Senior BE, Cawood ML, Oakey RE, McKiddie JM, Siddle DR. A comparison of the effects of clomiphene and tamoxifen treatment on the concentrations of oestradiol and progesterone in the peripheral plasma of infertile women.
Clin Endocrinol (oxf)
1978
;
8
:
381
–9.
170
Lønning PE, Johannessen DC, Lien EA, et al. Influence of tamoxifen on sex hormones, gonadotrophins and sex hormone binding globulin in postmenopausal breast cancer patients.
J Steroid Biochem Mol Biol
1995
;
52
:
491
–6.
171
Lønning PE, Lien E. Mechanisms of action of endocrine treatment in breast cancer.
Crit Rev Oncol Hematol
1995
;
21
:
158
–93.
172
Geisler J, Lønning P. Resistance to endocrine therapy of breast cancer: recent advances and tomorrows challenges.
Clin Breast Cancer
2001
;
1
:
297
–308.
173
Osborne CK, Schiff R. Growth factor receptor cross-talk with estrogen receptor as a mechanism for tamoxifen resistance in breast cancer.
Breast
2003
;
12
:
362
–7.
174
Osborne CK, Schiff R, Fuqua SA, Shou J. Estrogen receptor: current understanding of its activation and modulation.
Clin Cancer Res
2001
;
7
:
4338
–42s; discussion 4411–2s.
175
Osipo C, Liu H, Meeke K, Jordan VC. The consequences of exhaustive antiestrogen therapy in breast cancer: Estrogen-induced tumor cell death.
Exp Biol Med (Maywood)
2004
;
229
:
722
–31.
176
Schiff R, Massarweh SA, Shou J, et al. Cross-talk between estrogen receptor and growth factor pathways as a molecular target for overcoming endocrine resistance.
Clin Cancer Res
2004
;
10
:
331
–6S.
177
Jensen EV, Jordan VC. The estrogen receptor: A model for molecular medicine.
Clin Cancer Res
2003
;
9
:
1980
–9.
178
Lewis JS, Cheng D, Jordan VC. Targeting oestrogen to kill the cancer but not the patient.
Br J Cancer
2004
;
90
:
944
–9.
179
Lønning PE, Bajetta E, Murray R, et al. Activity of exemestane in metastatic breast cancer after failure of nonsteroidal aromatase inhibitors: A phase II trial.
J Clin Oncol
2000
;
18
:
2234
–44.
180
Lønning PE, Geisler J, Krag L, et al. Effect of exemestane on bone: a randomized placebo controlled study in postmenopausal women with early breast cancer at low risk.
Am Soc Clin Oncol
2004
;
23
:
6
.
181
Brodie AH, Jelovac D, Long B. The intratumoral aromatase model: studies with aromatase inhibitors and antiestrogens.
J Steroid Biochem Mol Biol
2003
;
86
:
283
–8.
182
Lønning PE. Study of suboptimum treatment response: lessons from breast cancer.
Lancet Oncol
2003
;
4
:
177
–85.
183
Lønning PE. Genes causing inherited cancer as beacons identifying the mechanisms of chemoresistance.
Trends Mol Med
2004
;
10
:
113
–8.
184
Lippman M, Bolan G, Huff K. The effects of estrogens and antiestrogens on hormone-responsive human breast cancer in long-term tissue culture.
Cancer Res
1976
;
36
:
4595
–601.
185
Martin LA, Farmer I, Johnston SR, et al. Enhanced estrogen receptor (ER) α, ERBB2, and MAPK signal transduction pathways operate during the adaptation of MCF-7 cells to long term estrogen deprivation.
J Biol Chem
2003
;
278
:
30458
–68.
186
Yue W, Wang J-P, Conaway MR, Li Y, Santen R. Adaptive hypersensitivity following long-term estrogen deprivation: involvement of multiple signal pathways.
J Steroid Biochem Mol Biol
2003
;
86
:
265
–74.
187
Kennedy BJ. Massive estrogen administration in premenopausal women with metastatic breast cancer.
Cancer
1962
;
15
:
641
–8.
188
Kennedy BJ. Hormonal therapies in breast cancer.
Semin Oncol
1974
;
1
:
119
–29.
189
Ingle JN, Ahmann DL, Green SJ, et al. Randomized clinical trial of diethylstilstilbestrol versus tamoxifen in postmenopausal women with advanced breast cancer.
N Engl J Med
1981
;
304
:
16
–21.
190
Lønning PE, Taylor PD, Anker G, et al. High-dose estrogen treatment in postmenopausal breast cancer patients heavily exposed to endocrine therapy.
Breast Cancer Res Treat
2001
;
67
:
111
–6.
191
Schiff R, Massarweh S, Shou J, Osborne CK. Breast cancer endocrine resistance: how growth factor signaling and estrogen receptor coregulators modulate response.
Clin Cancer Res
2003
;
9
:
447
–54S.
192
Thiantanawat A, Long BJ, Brodie AM. Signaling pathways of apoptosis activated by aromatase inhibitors and antiestrogens.
Cancer Res
2003
;
63
:
8037
–50.
193
Osborne CK, Bardou V, Hopp TA, et al. Role of the estrogen receptor coactivator AIB1 (SRC-3) and HER-2/neu in tamoxifen resistance in breast cancer.
J Natl Cancer Inst
2003
;
95
:
353
–61.
194
Ellis MJ, Coop A, Singh B, et al. Letrozole is more effective neoadjuvant endocrine therapy than tamoxifen for ErbB-1- and/or ErbB-2-positive, estrogen receptor-positive primary breast cancer: evidence from a phase III randomized trial.
J Clin Oncol
2001
;
19
:
3808
–16.
195
Zhu L, Chow LW, Loo WT, Guan XY, Toi M. Her2/neu expression predicts the response to antiaromatase neoadjuvant therapy in primary breast cancer: subgroup analysis from celecoxib antiaromatase neoadjuvant trial.
Clin Cancer Res
2004
;
10
:
4639
–44.
196
Modlich O, Prisack HB, Munnes M, Audretsch W, Bojar H. Immediate gene expression changes after the first course of neoadjuvant chemotherapy in patients with primary breast cancer disease.
Clin Cancer Res
2004
;
10
:
6418
–31.
197
Osborne CK, Arteaga CL. Role of molecular and genetic markers in breast cancer treatment decisions.
J Clin Oncol
2003
;
21
:
285
–7s.
198
Hähnel R, Woodings T, Vivian AB. Prognostic value of estrogen receptors in primary breast cancer.
Cancer
1979
;
44
:
671
–5.
199
Vollenweider-Zerargui L, Barrelet L, Wong Y, Lemarchand-Béraud T, Gómez F. The predictive value of estrogen and progesterone receptors' concentrations on the clinical behavior of breast cancer in women. Clinical correlation on 547 patients.
Cancer
1986
;
57
:
1171
–80.