The newer generation aromatase inhibitors (AIs) as a class show efficacy and tolerability benefits over previously established treatments inpostmenopausal women with advanced breast cancer. At clinically administered doses, the plasma half-lives of anastrozole (1 mg once daily), letrozole (2.5 mg once daily), and exemestane (25 mg once daily) are 41–48 h, 2–4 days, and 27 h, respectively. Time to steady-state plasma levels is 7 days for both anastrozole and exemestane and 60 days for letrozole. Androgenic side effects have only been reported with exemestane. Anastrozole treatment has no impact on plasma lipid levels, whereas both letrozole and exemestane have an unfavorable effect. From indirect comparisons, anastrozole shows the highest degree of selectivity compared with letrozole and exemestane, in terms of a lack of effect on adrenosteroidogenesis. To date, there are no data suggesting any major differences in clinical efficacy between the newer generation AIs anastrozole and letrozole. Based on the observed pharmacological profiles, however, it cannot be assumed that the AIs will display the same tolerability and safety profiles when given for extended periods of time in the adjuvant setting. The effects of anastrozole, letrozole, and exemestane are being investigated in the adjuvant setting, and these data will elucidate the possible long-term consequences of the pharmacological effects reported after short-term exposure.

Breast cancer is one of the leading causes of death in women in Western countries. It is anticipated that in the year 2002, 205,000 new cases of breast cancer will be diagnosed, and of those, 40,000 women will die from the disease in the United States (1). A significant proportion of patients with both early- and advanced-disease cancer have hormone-sensitive breast cancer, and this subset of patients can be identified by the presence of estrogen receptors (2, 3). Recently, a number of newer agents have become available to treat this subgroup of patients, including the AIs3 and the estrogen receptor down-regulator fulvestrant (Faslodex; Refs. 4, 5, 6, 7, 8).

In the United States, there are currently three AIs available for clinical use: (a) anastrozole; (b) letrozole; and (c) exemestane. AIs lower the estrogen level in postmenopausal women by inhibition of the P450 cytochrome enzyme aromatase, which catalyzes the conversion of androgens to estrogens. These agents are only effective in patients in whom ovarian function has been effectively suppressed either naturally or therapeutically.

Aminoglutethimide (9, 10) and testolactone (11, 12) are members of the earlier generation AIs. Testolactone was the first AI extensively investigated for treatment of advanced breast cancer. It was initially viewed as an androgen, even though virilizing effects were almost completely absent. Aminoglutethimide became established as an AI before the potential of testolactone was fully recognized. The efficacy of aminoglutethimide was evaluated in postmenopausal women and found to be similar to that of adrenolectomy and/or hypophysectomy (9, 10). However, the use of these earlier generation agents was not widespread because of substantial morbidity and lack of selectivity for the aromatase enzyme, which necessitated concomitant steroid supplementation.

The AIs are classified into two types: (a) type I, suicidal or noncompetitive inhibitors; and (b) type II, competitive inhibitors (13, 14). Type I inhibitors are steroidal compounds, and type II inhibitors are nonsteroidal drugs. Both types mimic normal substrates (androgens), competing with the substrate for access to the binding site on the enzyme. After initial binding, the next step differs for the two types: once a noncompetitive inhibitor has bound, the enzyme initiates its typical sequence of hydroxylation, but hydroxylation produces an unbreakable covalent bond between the inhibitor and the enzyme protein. Enzyme activity is thus permanently blocked; even if all unattached inhibitor is removed, enzyme activity can only be restored by new enzyme synthesis. Competitive inhibitors reversibly bind to the active enzyme site, and either no enzyme activity is triggered, or it is without effect. The inhibitor can disassociate from the binding site, allowing renewed competition between the inhibitor and the substrate for binding to the site. As a result, the effectiveness of competitive inhibitors depends on the relative concentrations and affinities of the inhibitor and the substrate. Continued activity requires constant presence of inhibitor.

To compete for binding to the active site, both competitive and noncompetitive inhibitors must necessarily share important structural features with the endogenous substrate. Noncompetitive inhibitors must also share structural features with androgens, allowing them to interact with the catalytic residual on the enzyme protein. This renders them inherently selective. By contrast, most competitive inhibitors interact with the heme iron, a common feature of all cytochrome P450 enzymes. Some may also bind to the highly conserved oxygen binding site in addition to the substrate binding site. Thus, unless the specificity of a competitive inhibitor is reinforced through other structural features, it may block the activity of a variety of cytochrome P450 enzymes, as does aminoglutethimide.

Both anastrozole and letrozole are type II nonsteroidal AIs, whereas exemestane has a steroidal structure and is classified as a type I AI, also known as an aromatase inactivator because it irreversibly binds with and permanently inactivates the enzyme. The clinical relevance of these differences in mechanism of action, if any, remains to be established.

All three drugs (anastrozole, letrozole, and exemestane) are given p.o. as once-daily doses. The recommended daily doses are as follows: for anastrozole, 1 mg; for letrozole, 2.5 mg; and for exemestane, 25 mg. The time needed to reach maximal E2 suppression is 2–4 days for anastrozole and letrozole and 7 days for exemestane (15, 16). In the exemestane study, these measurements were done every 7 days, and it is not known whether maximal estrogen suppression after exemestane may have occurred earlier. Anastrozole and exemestane achieve a steady-state drug level by 7 days (17). In contrast, letrozole has been reported to take 60 days to achieve plasma steady-state levels, which may be reflective of the accumulation that occurs with letrozole over long-term dosing (18). These data suggested a nonlinear relationship after repeated dose administration of letrozole. The half-lives of anastrozole and exemestane are 41 and 27 h (19, 20), respectively, and for letrozole, the half-life has been reported to be up to 4 days (21). All three AIs effectively reduce E2, E1, and E1S: anastrozole by 81–94% (22, 23); letrozole by 88–98% (23); and exemestane by 52–72% (17, 24). In one prospective study, a comparative evaluation was carried out to determine the extent of plasma E1, E1S, and E2 suppression after 6 weeks of treatment with either anastrozole or letrozole in postmenopausal women with metastatic disease (23). There were no significant differences in reduction of plasma E2 by either agent (84.9% and 87.8%, respectively, for anastrozole and letrozole). Anastrozole and letrozole reduced E1 levels by 81.0% and 84.3%, respectively; the extent of this suppression was significantly greater in the letrozole group (P = 0.019). Letrozole reduced plasma E1S levels significantly more than anastrozole (98% versus 93.5%, P = 0.019). The clinical significance of this additional suppression of estrogen by letrozole remains to be defined. There are no comparative studies evaluating exemestane in relation to other agents, but in individual studies, E2 levels were suppressed by 65% with exemestane (17).

Breast cancer tumor cells and also stromal cells have the potential to synthesize estrogen using local aromatase activity. Local production of estrogen by this mechanism results in a significantly higher concentration of estrogens in tumor than in the circulation in postmenopausal women. Separate studies evaluating these three agents have demonstrated that all three can reduce tumor estrogens (25, 26, 27). These studies were not carried out as comparative trials; thus, it is difficult to assess whether one agent is more effective than another in reducing intratumoral estrogen. With all three agents, reductions within the tumors have been in the range of 97–98%. The initial second-line Phase III studies evaluated different doses of AIs, and one of the questions in those studies was whether higher doses would result in further reduction in intratumoral estrogen and in higher antitumor activity. The data from these studies have not provided convincing evidence that there is a dose-dependent antitumor activity of AIs: the 1 and 10 mg doses of anastrozole had similar antitumor activity (2); and second-line studies of 0.5 and 2.5 mg of letrozole (28) have shown no consistent superiority of the higher dose. There are no comparable prospective studies in which different doses of exemestane have been studied.

All three drugs have been evaluated at concentrations of ≤500 μm for pharmacokinetic activity with the cytochrome P450 system in the human and rat microsomal preparations (29, 30, 31). Anastrozole inhibits, in decreasing order of magnitude, CYP1A2, CYP2C8/9, and CYP3A4 (29). Anastrozole has no effect on CYP2A6 or CYP2D6. Letrozole strongly inhibits CYP2A6, moderately inhibits CYP2C19, and has a low affinity for CYP3A4 (30). Exemestane is metabolized by CYP3A4 (31). There is potential for drug-drug interactions if patients are prescribed concomitant medications that interact with these cytochrome P450 enzymes. Both anastrozole and letrozole have no known drug-drug interaction with cimetidine (a marker for CYP3A4 activity) or warfarin [a marker for CYP3A4 and CYP1A2 activity (29, 30)]. No drug-drug interactions have been formally reported for exemestane, although there is potential for interactions with drugs that affect CYP3A4. There is known drug-drug interaction of tamoxifen with anastrozole and letrozole. Concomitant administration of either anastrozole or letrozole with tamoxifen decreases the plasma level of the AI. Concomitant administration of letrozole and tamoxifen decreased the level of letrozole by 38% (90% confidence interval, 32–43%; Ref. 32). Anastrozole and tamoxifen administrated concomitantly in the ATAC trial lowered the plasma anastrozole level in the combined arm by 27% (90% confidence interval, 20–30%; Ref. 33). Whereas the combined therapy arm of the ATAC trial demonstrated a decrease in the plasma concentration of anastrozole, systemic E2 suppression was similar to that observed in patients treated with anastrozole alone. Based on the observed drug-drug interaction and the lack of clinical data demonstrating the superiority of the combined arm, this arm of the study is being discontinued.

Anastrozole and letrozole have no androgenic, progestrogenic, or estrogenic effects such as weight gain, acne, or hypertrichosis. Exemestane has weak androgenic properties, and its use at higher doses has been associated with steroidal-like side effects such as weight gain and acne (24, 34). These side effects have to be evaluated if exemestane is given on a prolonged basis as in the adjuvant setting.

These three AIs have somewhat different effects on lipid profile. In one study in patients with metastatic disease (n = 952 at study entry), anastrozole showed no marked effect on lipid profile compared with baseline. Whereas these patients had advanced metastatic disease, these data do provide some evidence that anastrozole did not significantly change the lipid profile compared with the baseline studies (35). A smaller study (n = 44) that compared lipids at baseline and after 32 weeks of treatment with anastrozole (36) also did not show any significant alterations in the lipid parameters. Letrozole was evaluated in 20 women with advanced breast cancer who showed a significant increase in total cholesterol and low-density lipoprotein from baseline after 8 and 16 weeks of therapy (37). In a 9-week trial in advanced breast cancer, exemestane resulted in a significant decrease in both total cholesterol and HDL (38). Exemestane also decreased the total triglyceride levels in this study. In another European Organization of Research and Treatment of Cancer study, 24 weeks of exemestane had no impact on the lipid profile (39). Clinical implications of these changes remain to be defined.

Anastrozole data indicate that it has no impact on adrenal steroidogenesis at up to 10 times the clinically recommended dose, thus suggesting that this drug has little activity on other cytochrome P450 enzymes involved in steroid synthesis (40). In one study in healthy postmenopausal women, basal and ACTH stimulation did not differ from baseline levels after 14 days of anastrozole at a 5- or 10-mg daily dose (40). In another study, there was no change in the ACTH-stimulated response after 115 days on therapy (41). Letrozole studies showed a decrease in basal and ACTH-stimulated cortisol synthesis. In one study, patients with advanced breast cancer showed a significant decrease in ACTH-stimulated aldosterone levels after 3 months of letrozole treatment (2.5 or 0.5 mg; Ref. 42). Exemestane in one small study had no impact on cortisol or aldosterone levels with up to 7 days of treatment, and the dose-ranging studies included doses from 0.5 to 800 mg (43). Data from these limited studies demonstrate that anastrozole and exemestane have no effect on steroidogenesis. In contrast, letrozole alters cortisol and aldosterone levels even at therapeutic doses (18). However, the clinical relevance of these differences remains to be defined.

In the preclinical setting, exemestane has been evaluated and shown to prevent bone loss in that experimental system (44). The data from ongoing clinical trials will be of interest. In healthy postmenopausal women, letrozole given 2.5 mg for 6 months produced an increase in bone resorption, as assessed by increased urinary pyridinoline and deoxypyridinoline, over baseline values (45). In another study, 3 months of letrozole resulted in bone resorption as evaluated by measuring the markers of bone metabolism (46).

The preliminary data of the ATAC study do suggest that anastrozole therapy is associated with higher bone-related events, i.e., fractures, compared with the tamoxifen arm of the study (47). At the present time, there are no long-term clinical data regarding the effect of AIs on the bones, except for the ATAC trial. Bisphosphonate therapy, which can prevent or reduce the risk of bone loss, needs to be considered when offering these agents for extended periods of time to patients with early breast cancer. Currently, prospective studies are being conducted to define the benefit of bisphosphonates in this subset of patients.

In a large second-line breast cancer study (n = 713), the safety and efficacy of letrozole and anastrozole were prospectively evaluated in postmenopausal women with advanced breast cancer (48). In this multicenter, open-label, randomized trial, all patients had previously been treated with antiestrogen therapy and were allowed to have one chemotherapy for metastatic disease. Hormonal receptor status was known in 48% of the patients. The primary end point was time to progression, with secondary end points of objective response rate, duration of response, time to treatment failure, and survival. Time to progression, time to treatment failure, and survival were similar in the two arms. In the intent-to-treat analysis, a higher proportion of patients achieved an objective response with letrozole therapy (19.1% for letrozole versus 12.3% for anastrozole, P = 0.014). Similarly, clinical benefit rate was also higher in the letrozole group compared with anastrozole (27% versus 23%; odds ratio, 1.24; P = 0.218). In patients with known receptor-positive disease, the antitumor activity of the two drugs was similar. In patients with unknown receptor status, however, there were major differences in the objective response rates between the two drugs. Double-blind controlled clinical trials are needed to evaluate the safety and efficacy of these agents in hormone receptor-positive postmenopausal women. These studies will define whether there are major differences in the safety and/or efficacy of the three AIs available for clinical use.

Dr. Mitch Dowsett: I think there has to be the same sort of concern about these pharmacological comparisons as with the Phase II trials. We think of these biochemical measurements as absolute, but they’re not: the prime example of that is the levels of estrogen suppression reported in different studies using different labs. You can look at subtle differences, 85%, 88%, and 90%, but the differences represent the limits of the assays. We have assays which go down to 3 pmol/liter, which is something like 15% of the starting level. These studies just use different methods. They also have different approaches as to when they’ve made the measurements, how they have made the measurements. To get good pharmacological data, they ought to be collected within the context of randomized studies.

Dr. Aman Buzdar: You are absolutely right. I was asked to show the data, and these are the only randomized data available. I think there is no question that we need to do prospective blinded studies so that neither the investigator nor the lab has any influence.

Dr. Dowsett: I think clinicians in particular have a temptation to see biochemical data as absolutes that can be compared in a way that they would not attempt with Phase II clinical trial data.

Dr. James Ingle: There are two bits of information prior to the head-to-head study that was presented by Carson Rose at American Society of Clinical Oncology (48). One is the data from Dr. A. Brodie and Dr. B. J. Long suggesting that letrozole is more potent, at least in their model system (Q. Lu et al., Breast Cancer Res. Treat., 57: 183–192, 1999), and then we had the Geisler study from Dr. Dowsett’s laboratory that indicated that there was more suppression with letrozole than anastrozole (J. Geisler et al., J. Clin. Oncol., 20: 751–757, 2002). But is there any value to the extra little increment of estrogen suppression?

Dr. Steven Come: Are these one-time measurements or are they done serially over time in a system? What happens over time might be significant, just as it is in the models. We’re using what we see at one point in time to predict what the long-term outcome is going to be for the mouse or the patient.

Dr. Dowsett: A long time ago these measurements were done in Dick Santen’s and our studies on estrogen levels at relapse on aminoglutethimide [Santen, R. J. Overall experience with aminoglutethimide in the management of advanced breast cancer. In: R. W. Elsdon-Dew, I. M. Jackson, and G. F. B. Birdwood (eds), Aminoglutethimide: An Alternative Endocrine Therapy for Breast Carcinoma, p. 3. London: Academic Press, 1982; M. Dowsett et al., J. Clin. Endocrinol. Metab., 58: 99–104, 1984]. There was a sniff of a difference in estrogen levels, which you wouldn’t have said could explain the relapse of the patients. If we think of these now as hypersensitive cells, we can begin to interpret those findings. There is some discussion in the ATAC steering committee of collecting blood samples from patients long term to make sure these patients really have got the same degree of suppression.

Dr. Buzdar: Actually, there was a paper comparing estrogen suppression in responders and nonresponders to AIs. The degree of estrogen suppression was identical, so there must be some other mechanism.

Dr. Kent Osborne: I think we are wasting our time looking at blood estrogen levels. The action is in the tumor, where the estrogen levels are high in some of these women because of local aromatase.

Dr. Dowsett: I’m not sure I agree with that entirely. In terms of the proportional importance of intratumoral aromatase, the studies from Bill Miller and Mike Dixon, where they show the proportion of estrogen that gets into the tumor from the outside and the proportion that is synthesized within, demonstrate that there are some tumors in which there is no synthesis within the tumor (reviewed in Breast Cancer Res. Treat., 49 (Suppl. 1): S27−S32, 1998). Some of those patients respond to AIs. There are other patients in whom perhaps 80–90% of the estrogen comes from within the tumor. Overall, it’s about 50/50, but it varies much between tumors.

Dr. Brian Long: A positive correlation between aromatase expression and ER expression in the tumor has not been demonstrated.

Dr. Buzdar: If the tumor has very low aromatase activity and still you see the same degree of response, it means that perhaps the degree of suppression is not that much important.

1

Presented at the Second International Conference on Recent Advances and Future Directions in Endocrine Manipulation of Breast Cancer, June 28–29, 2002, Cambridge, MA.

3

The abbreviations used are: AI, aromatase inhibitor; E2, estradiol; E1, estrone; E1S, estrone sulfate; ATAC, Arimidex and Tamoxifen Alone or in Combination; ACTH, adrenocorticorticotropic hormone.

1
Jemal A., Thomas A., Murray T., Thun M. Cancer statistics, 2002.
CA Cancer J. Clin.
,
52
:
23
-47,  
2002
.
2
Jensen E. V., Block G. E., Smith S., Kyser K., DeSombre E. R. Estrogen receptors and breast cancer response to adrenalectomy.
Natl. Cancer Inst. Monogr.
,
34
:
55
-70,  
1971
.
3
Sedlacek S. M., Horwitz K. B. The role of progestins and progesterone receptors in the treatment of breast cancer.
Steroids
,
44
:
467
-484,  
1984
.
4
Buzdar A. U., Jonat W., Howell A., Jones S. E., Blomqvist C. P., Vogel C. L., Eiermann W., Wolter J. M., Steinberg M., Webster A., Lee D. Anastrozole versus megestrol acetate in the treatment of postmenopausal women with advanced breast carcinoma: results of a survival update based on a combined analysis of data from two mature Phase III trials.
Arimidex Study Group. Cancer (Phila.)
,
83
:
1142
-1152,  
1998
.
5
Dombernowsky P., Smith I., Falkson G., Leonard R., Panasci L., Bellmunt J., Bezwoda W., Gardin G., Gudgeon A., Morgan M., Fornasiero A., Hoffmann W., Michel J., Hatschek T., Tjabbes T., Chaudri H. A., Hornberger U., Trunet P. F. Letrozole, a new oral aromatase inhibitor for advanced breast cancer: double-blind randomized trial showing a dose effect and improved efficacy and tolerability compared with megestrol acetate.
J. Clin. Oncol.
,
16
:
453
-461,  
1998
.
6
Kaufmann M., Bajetta E., Dirix L. Y., Fein L. E., Jones S. E., Zilembo N., Dugardyn J. L., Nasurdi C., Mennel R. G., Cervek J., Fowst C., Polli A., di Salle E., Arkhipov A., Piscitelli G., Miller L. L., Massimini G. Exemestane is superior to megestrol acetate after tamoxifen failure in postmenopausal women with advanced breast cancer: results of a Phase III randomized double-blind trial. The Exemestane Study Group.
J. Clin. Oncol.
,
18
:
1399
-1411,  
2000
.
7
Osborne C. K., Pippen J., Jones S. E., Parker L. M., Ellis M., Come S., Gertler S. Z., May J. T., Burton G., Dimery I., Webster A., Morris C., Elledge R., Buzdar A. Double-blind, randomized trial comparing the efficacy and tolerability of fulvestrant versus anastrozole in postmenopausal women with advanced breast cancer progressing on prior endocrine therapy: results of a North American trial.
J. Clin. Oncol.
,
20
:
3386
-3395,  
2002
.
8
Howell A., Robertson J. F., Quaresma A. J., Aschermannova A., Mauriac L., Kleeberg U. R., Vergote I., Erikstein B., Webster A., Morris C. Fulvestrant, formerly ICI 182,780, is as effective as anastrozole in postmenopausal women with advanced breast cancer progressing after prior endocrine treatment.
J. Clin. Oncol.
,
20
:
3396
-3403,  
2002
.
9
Harvey H. A., Santen R. J., Osterman J., Samojlik E., White D. S., Lipton A. A comparative trial of transsphenoidal hypophysectomy and estrogen suppression with aminoglutethimide in advanced breast cancer.
Cancer (Phila.)
,
43
:
2207
-2214,  
1979
.
10
Santen R. J., Worgul T. J., Samojlik E., Interrante A., Boucher A. E., Lipton A., Harvey H. A., White D. S., Smart E., Cox C., Wells S. A. A randomized trial comparing surgical adrenalectomy with aminoglutethimide plus hydrocortisone in women with advanced breast cancer.
N. Engl. J. Med.
,
305
:
545
-551,  
1981
.
11
Segaloff A., Weeth J. B., Meyer K. K., Rongone E. L., Cunningham M. E. G. Hormonal therapy in cancer of the breast. XIX. Effect of oral administration of delta-1-testololactone on clinical course and hormonal excretion.
Cancer (Phila.)
,
15
:
633
-635,  
1962
.
12
Van Rymenant M., Coune A., Tagnon H. J., Somon S. Le traitement hormonal du cancer du sein en phase avancee. Methodes d’etudes: comparaison de la delta-1-testololactone avec le propionate de tetosterone.
Acta Clin. Belg.
,
18
:
469
-479,  
1963
.
13
Buzdar A., Howell A. Advances in aromatase inhibition: clinical efficacy and tolerability in the treatment of breast cancer.
Clin. Cancer Res.
,
7
:
2620
-2635,  
2001
.
14
Goss P. E., Strasser K. Aromatase inhibitors in the treatment and prevention of breast cancer.
J. Clin. Oncol.
,
19
:
881
-894,  
2001
.
15
Plourde P. V., Dyroff M., Dowsett M., Demers L., Yates R., Webster A. ARIMIDEX: a new oral, once-a-day aromatase inhibitor.
J. Steroid Biochem. Mol. Biol.
,
53
:
175
-179,  
1995
.
16
Demers L. M., Lipton A., Harvey H. A., Kambic K. B., Grossberg H., Brady C., Santen R. J. The efficacy of CGS 20267 in suppressing estrogen biosynthesis in patients with advanced stage breast cancer.
J. Steroid Biochem. Mol. Biol.
,
44
:
687
-691,  
1993
.
17
Zilembo N., Noberasco C., Bajetta E., Martinetti A., Mariani L., Orefici S. Endocrinological and clinical evaluation of exemestane, a new steroidal aromatase inhibitor.
Br. J. Cancer
,
72
:
1007
-1012,  
1995
.
18
Bajetta E., Zilembo N., Dowsett M., Guillevin L., Di Leo A., Celio L., Martinetti A., Marchiano A., Pozzi P., Stani S., Bichisao E. Double-blind, randomised, multicentre endocrine trial comparing two letrozole doses, in postmenopausal breast cancer patients.
Eur. J. Cancer
,
35
:
208
-213,  
1999
.
19
Yates R. A., Dowsett M., Fisher G. V., Selen A., Wyld P. J. Arimidex (ZD1033): a selective, potent inhibitor of aromatase in postmenopausal female volunteers.
Br. J. Cancer
,
73
:
543
-548,  
1996
.
20
Spinelli R., Jannuzzo M. G., Poggesi I., Frevola L., Broutin F., Cicioni P. Pharmacokinetics (PK) of Aromasin (Exemestane, EXE) after single and repeated doses in healthy postmenopausal volunteers (HPV).
Eur. J. Cancer
,
35 (Suppl4)
:
S295
1999
.
21
Sioufi A., Sandrenan N., Godbillon J., Trunet P., Czendlik C., Howard H. Comparative bioavailability of letrozole under fed and fasting conditions in 12 healthy subjects after 2.5 mg single oral administration.
Biopharm. Drug Dispos.
,
18
:
489
-497,  
1997
.
22
Geisler J., King N., Dowsett M., Ottestad L., Lundgren S., Walton P., Kormeset P. O., Lonning P. E. 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
,
74
:
1286
-1291,  
1996
.
23
Geisler J., Anker G., Dowsett M., Lonning P. E. Letrozole suppresses plasma estrogen levels in postmenopausal breast cancer patients more completely than anastrozole.
Proc. Am. Soc. Clin. Oncol.
,
19
:
102a
2000
.
24
Bajetta E., Zilembo N., Noberasco C., Martinetti A., Mariani L., Ferrari L., Buzzoni R., Greco M., Bartoli C., Spagnoli I., Danesini G. M., Artale S., Paolini J. The minimal effective exemestane dose for endocrine activity in advanced breast cancer.
Eur. J. Cancer
,
33
:
587
-591,  
1997
.
25
Geisler J., Bernsten L., Ottestad B., Lindtjorn B., Dowsett M., Lonning P. E. Neoadjuvant treatment with anastrozole (Arimidex) causes profound suppression of intra-tumor estrogen levels.
Proc. Am. Soc. Clin. Oncol.
,
18
:
82a
1999
.
26
Miller W. R. Biology of aromatase inhibitors: pharmacology/endocrinology within the breast.
Endocr. Relat. Cancer
,
6
:
187
-195,  
1999
.
27
Dixon J. M., Grattage L., Renshaw L., Miller W. R. Exemestane as neoadjuvant treatment for locally advanced breast cancer: endocrinologic and clinical endpoints.
Breast Cancer Res. Treat.
,
64
:
53
2000
.
28
Buzdar A., Douma J., Davidson N., Elledge R., Morgan M., Smith R., Porter L., Nabholtz J., Xiang X., Brady C. Phase III, multicenter, double-blind, randomized study of letrozole, an aromatase inhibitor, for advanced breast cancer versus megestrol acetate.
J. Clin. Oncol.
,
19
:
3357
-3366,  
2001
.
29
Grimm S. W., Dyroff M. C. Inhibition of human metabolizing cytochromes P450 by anastrozole, a potent and selective inhibitor of aromatase.
Drug Metab. Dispos.
,
25
:
598
-602,  
1997
.
30
Wirz B., Valles B., Parkinson A., Madan A., Probst A., Zimmerman A. CYP3A4 and CYP2A6 are involved in the biotransformation of letrozole (Femara).
7th North American Meeting
,
10
:
359
1996
.
31
Anonymous. Exemestane for advanced breast cancer.
Med. Lett. Drugs Ther.
,
42
:
35
-36,  
2000
.
32
Dowsett M., Pfister C., Johnson S. R. D., Miles D. W., Houston S. J., Verbeek J. A. Impact of tamoxifen on the pharmacokinetics and endocrine effects of the aromatase inhibitor letrozole in postmenopausal women with breast cancer.
Clin. Cancer Res.
,
5
:
2338
-2343,  
1999
.
33
The ATAC Trialists’ Group. Pharmacokinetics of anastrozole and tamoxifen alone, and in combination, during adjuvant endocrine therapy for early breast cancer in postmenopausal women: a sub-protocol of the “Arimidex and Tamoxifen Alone or in Combination” (ATAC) trial.
Br. J. Cancer
,
85
:
317
-324,  
2001
.
34
Michaud L. B., Buzdar A. U. Risks and benefits of aromatase inhibitors in postmenopausal breast cancer.
Drug Saf.
,
21
:
297
-309,  
1999
.
35
Dewar J., Nabholtz J-M., Bonneterre J., Buzdar A., Robertson J. F., Thurlimann B. The effect of anastrozole (Arimidex) on serum lipids: data from a randomized comparison of anastrozole (AN) versus tamoxifen (TAM) in postmenopausal (PM) women with advanced breast cancer (ABC).
Breast Cancer Res. Treat.
,
64
:
51
2000
.
36
Wojtacki J., Kruszewski W. J., Les’niewski-Kmak K., S’liwin’ska M., Czyzewska K., Kruszewska E. Short-term effects of anastrozole therapy on serum lipid profile in patients with breast cancer, previously treated with tamoxifen. Preliminary report.
Nowotwory
,
51
:
43
-47,  
2001
.
37
Elisaf M. S., Bairaktari E., Nicolaides C., Kakaidi B., Tzallas C. S., Katsaraki A. Effect of letrozole on the lipid in postmenopausal women with breast cancer.
Eur. J. Cancer
,
37
:
1510
-1513,  
2001
.
38
Engan T., Krane J., Johannessen D. C., Kvinnsland S. Plasma changes in breast cancer patients during endocrine therapy: lipid measurements and nuclear magnetic resonance (NMR) spectroscopy.
Breast Cancer Res. Treat.
,
36
:
287
-297,  
1995
.
39
Lohrisch C., Paridaens R., Dirix L. Y., Beex M., Nooij M., Cameron D. No adverse impact on serum lipids of the irreversible aromatase inactivator Aromasin [Exemestane (E)] in first-line treatment of metastatic breast cancer (MBC): companion study to a European Organization of Research and Treatment of Cancer (Breast Group) Trial with Pharmacia Upjohn.
Proc. Am. Soc. Clin. Oncol.
,
20
:
43a
2001
.
40
Plourde P. V., Dyroff M., Dukes M. Arimidex: a potent and selective fourth-generation aromatase inhibitor.
Breast Cancer Res. Treat.
,
30
:
103
-111,  
1994
.
41
Buzdar A., Esparza-Guerra L. Anastrozole “Arimidex” does not impair adrenal cortisol or aldosterone synthesis in postmenopausal women with advanced breast cancer.
Proc. Am. Soc. Clin. Oncol.
,
20
:
52b
2001
.
42
Bisagni G., Cocconi G., Scaglione F., Fraschini F., Pfister C. H., Trunet P. F. Letrozole, a new oral non-steroidal aromatase inhibitor in treating postmenopausal patients with advanced breast cancer.
Ann. Oncol.
,
7
:
99
-102,  
1996
.
43
Evans T. R., di Salle E., Ornati G., Lassus M., Benedetti M. S., Pianezzola E., Coombes R. C. Phase I and endocrine study of exemestane (FCE 24304), a new aromatase inhibitor, in postmenopausal women.
Cancer Res.
,
52
:
5933
-5939,  
1992
.
44
Goss P., Grynpas M., Qi S., Hu H. The effects of exemestane on bone and lipids in the ovariectomized rat.
Breast Cancer Res. Treat.
,
69
:
224
2001
.
45
Lai J. M., Taxel P., Raisz L. G. The effect of aromatase inhibition on bone turnover in older men.
J. Am. Geriatr. Soc.
,
46
:
S79
1998
.
46
Harper-Wynne C., Ross G., Sacks N., Gui G., Dowsett M. Effects of the aromatase inhibitor letrozole in healthy postmenopausal women rationale for prevention.
Proc. Am. Soc. Clin. Oncol.
,
20
:
335b
2001
.
47
The ATAC (Arimidex, Tamoxifen Alone or in Combination) Trialists’ Group. 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
,
359
:
2131
-2139,  
2002
.
48
Rose C., Vtoraya O., Pluzanska A., Neave F., Clemens M., Chaudri-Ross H. A., Wyld P., Lang R. Letrozole (Femara) versus anastrozole (Arimidex): second-line treatment in postmenopausal women with advanced breast cancer.
Proc. Am. Soc. Clin. Oncol.
,
21
:
34a
2002
.