Ian E. Smith, M.D., and Mitch Dowsett, Ph.D.


From the Royal Marsden Hospital and Institute of Cancer Research, London. Address reprint requests to Dr. Smith at the Breast Unit, Royal Marsden Hospital, Fulham Rd., London SW3 6JJ, United Kingdom, or at; E-mail: ian.smith@rmh.nthames.nhs.uk..


The third-generation aromatase inhibitors provide novel approaches to the endocrine treatment ofbreast cancer. These drugs are effec- tively challenging tamoxifen, the previous gold standard ofcare,[1-13] for use in postmenopausal patients with estrogen-receptor-positive cancers, who make up the majority ofpatients with breast cancer. These agents are also being considered for use in chemoprevention, a strategy in which tamoxifen has already been shown to reduce the incidence of breast cancer.[14,15] In this article, we review the current role of aromatase inhibitors and assess their potential for clinical use. Other reviews that may be ofinterest to specialists are also available.[16,17]



Background


Mechanisms of action

Estrogen is the main hormone involved in the development and growth ofbreast tumors; oophorectomy was first shown to cause regression ofadvanced breast cancer more than a century ago,[18] and estrogen deprivation remains a key therapeutic approach.[19] Tamoxifen inhibits the growth of breast tumors by competitive antagonism of estrogen at its receptor site (Fig. 1). Its actions are complex, however, and it also has partial estrogenagonist effects. These partial agonist effects can be beneficial, since they may help prevent bone demineralization in postmenopausal women,[20,21] but also detrimental, since they are associated with increased risks ofuterine cancer [13,22] and thromboembolism.[14] In addition, they may play a part in the development oftamoxifen resistance.[23]


f1.png



Figure 1. Mechanism ofAction ofAromatase Inhibitors and Tamoxifen.


In contrast, aromatase inhibitors markedly suppress plasma estrogen levels in postmenopausal women by inhibiting or inactivating aromatase, the enzyme responsible for the synthesis ofestrogens from androgenic substrates (specifically, the synthesis ofes- trone from the preferred substrate androstenedione and estradiol from testosterone) (Fig. 1). Unlike tamoxifen, aromatase inhibitors have no partial agonist activity.


Sources of aromatase

Aromatase, an enzyme of the cytochrome P-450 superfamily and the product of the CYP19 gene,[24] is highly expressed in the placenta and in the granulosa cells of ovarian follicles, where its expression depends on cyclical gonadotropin stimulation. Aromatase is also present, at lower levels, in several nonglandular tissues, including subcutaneous fat, liver, muscle, brain, normal breast, and breastcancer tissue.[25,26] Residual estrogen production after menopause is solely from nonglandular sources, in particular from subcutaneous fat. Thus, peripheral aromatase activity and plasma estrogen levels correlate with body-mass index in postmenopausal women.[27] At menopause, mean plasma estradiol levels fall from about 110 pg per milliliter (400 pmol per liter) to low but stable levels of about 7 pg per milliliter (25 pmol per liter). In postmenopausal women, however, the concentration of estradiol in breastcarcinoma tissue is approximately 10 times the concentration in plasma,[28] probably in part because ofthe presence ofintratumoral aromatase. Early evidence that intratumoral aromatase activity might help predict the response to aromatase inhibitors [29] remains to be confirmed in large-scale studies. Details on the control and importance ofthe sources ofaromatase have recently been published.[30,31]


Aromatase inhibition in premenopausal women

In premenopausal women, the use ofaromatase inhibitors leads to an increase in gonadotropin secretion because of the reduced feedback ofestrogen to the hypothalamus and pituitary, and in some animal models aromatase inhibition increases the weight ofthe ovaries.[32] Investigation of aromatase inhibition in breast cancer before menopause has consequently been minimal, aside from tests ofaromatase inhibition in combination with the use of a gonadotropin-releasing-hormone agonist to suppress ovarian function.[33] The short-term application of letrozole, a third-generation aromatase inhibitor, has recently been successful for the induction of ovulation in women with infertility.[34] The data in the current review, however, pertain solely to postmenopausal women.


Clinical development and pharmacology


Aminoglutethimide, the first aromatase inhibitor, was initially developed as an anticonvulsant but was withdrawn from use after reports of adrenal insufficiency. It was subsequently found to inhibit several cytochrome P-450 enzymes involved in adrenal steroidogenesis and was then redeveloped for use as “medical adrenalectomy" against advanced breast cancer.[35,36] Side effects, including drowsiness and rash, limited its use, but the discovery that its efficacy was mainly due to aromatase inhibition[37,38] stimulated the development of numerous new inhibitors during the 1980s and early 1990s. They are described as first-, second-, and third-generation inhibitors according to the chronologic order oftheir clinical development, and they are further classified as type 1 or type 2 inhibitors according to their mechanism ofaction (Table 1). Type 1 inhibitors are steroidal analogues ofandrostenedione (Fig. 2) and bind to the same site on the aromatase molecule, but unlike androstenedione they bind irreversibly, because oftheir conversion to reactive intermediates by aromatase. Therefore, they are now commonly known as enzyme inactivators. Type 2 inhibitors are nonsteroidal and bind reversibly to the heme group ofthe enzyme by way ofa basic nitrogen atom; anastrozole and letrozole, both third-generation inhibitors, bind at their triazole groups (Fig. 2).


Table 1. Classification of Aromatase Inhibitors.

t1.png



f2.png


Figure 2.Structures of the Main Aromatase Inhibitors and the Natural Substrate Androstenedione..


The second-generation aromatase inhibitors include formestane (4-hydroxyandrostenedione),[39] a type 1 compound, and fadrozole,[40] a type 2 imidazole. Each has been found to have clinical effica- cy,[11,12,41] but formestane has the disadvantage of requiring intramuscular injection, and fadrozole causes aldosterone suppression, limiting its use to doses that produce only about 90 percent inhibition.[42] Other second-generation aromatase inhibitors have been investigated clinically but have never been approved for clinical use. The third-generation inhibitors, developed in the early 1990s, include the triazoles anastrozole (Arimidex) and letrozole (Femara) and the steroidal agent exemestane (Aromasin). In contrast to aminoglutethimide and fadrozole, their specificity appears to be nearly complete at clinical doses, with little or no effect on basal levels of cortisol or aldosterone.[43-45]


Pharmacokinetics

Anastrozole, letrozole, and exemestane are administered orally. Anastrozole and letrozole have similar pharmacokinetic properties, with half-lives approximating 48 hours,[46,47] allowing a once-daily dosing schedule. The half-life ofexemestane is 27 hours.[48] Pharmacokinetic interactions between some inhibitors and tamoxifen have been described. Aminoglutethimide induces cytochrome P-450 activity, which reduces tamoxifen levels.[49] In contrast, the levels ofanastrozole and letrozole are reduced (by a mean of27 percent and 37 percent, respectively) when they are coadministered with tamoxifen, but these reductions are not associated with impaired suppression ofplasma estradiol levels.[50,51]


Comparative pharmacologic efficacy

The third-generation aromatase inhibitors have been found in preclinical studies to be more than three orders ofmagnitude more potent than aminoglutethimide.[52] All ofthem markedly suppress plasma estrogen levels, but the very low plasma estrogen levels in postmenopausal women and the limited sensitivity of immunoassays have made it difficult to estimate precisely their relative effectiveness. In contrast, isotopic measurement of whole-body aromatization has greater sensitivity and allows valid comparisons among studies. This method has demonstrated that greater inhibition is achieved with third-generation compounds than with earlier inhibitors: the mean degree of inhibition with anastrozole, exemestane, and letrozole at clinical doses is greater than 97 percent,[53,54] as compared with about 90 percent for aminoglutethimide.[55] The increased potency of the third-generation inhibitors is associated with better clinical efficacy than that offered by aminoglutethimide or the second-generation inhibitor fadrozole.[56-58]


Recently, subtle differences in potency between two of the third-generation inhibitors have been demonstrated. In a small, double-blind crossover trial, letrozole was associated with greater aromatase inhibition than anastrozole and lower plasma levels of estrone and estrone sulfate.[54]


Aromatase has intratumoral activity in the majority of breast carcinomas, and isotopic assays have shown that such activity contributes substantially to intratumoral estrogen levels; anastrozole, letrozole, and exemestane all markedly inhibit it.[59] However, the relative clinical significance of the effects of these agents on peripheral and intratumoral aromatase activity is unknown.



Current clinical role


As already noted, the data reviewed in this article pertain solely to postmenopausal women; the use of aromatase inhibitors in premenopausal women with breast cancer who have normal ovarian function is contraindicated. Their use is also, in general, contraindicated in women with estrogen-receptornegative and progesterone-receptor-negative cancer, given that such tumors are unresponsive to other forms of endocrine therapy.


Advanced disease

First-Line Therapy

One of the most important recent developments in therapy for breast cancer has been the demonstration that letrozole and probably also anastrozole are superior to tamoxifen as first-line treatment for advanced disease. Previous trials in which tamoxifen was compared with other endocrine agents, including diethylstilbestrol,[1] progestins,[2-4] androgens,[5] other antiestrogens,[6,7] and first- and second- generation aromatase inhibitors,[8-12] consistently failed to show such a difference. By current standards, these trials were underpowered, and most of them were not blinded, but nevertheless their results were interpreted as suggesting that tamoxifen, through estrogen-receptor blockade, provided the maximal possible endocrine control of breast cancer. Results with the third-generation aromatase inhibitors have refuted this hypothesis and suggest further possibilities for the development of endocrine therapy.


Three key trials of aromatase inhibitors as first- line therapy[60-62] — all ofthem multicenter, doubleblind studies involving patients whose tumors were hormone-receptor-positive (or of unknown receptor status) — have been published (Table 2). In the largest (a study involving 907 women, with a median follow-up of18 months), letrozole resulted in more tumor regressions and was associated with a longer time to disease progression than tamoxifen (9.4 vs. 6.0 months; P=0.0001).[60] This benefit was significant irrespective ofprevious adjuvant treatment with tamoxifen, the site of disease, or knowledge of the estrogen-receptor status. In the other two trials, anastrozole was compared with tamoxifen, with conflicting results. One ofthem showed that anastrozole, like letrozole, resulted in a longer time to disease progression than tamoxifen (11.1 vs. 5.6 months; P=0.005) and a trend towards more tumor regressions.[61] The other, which was similar in design, failed to confirm these findings: for each outcome variable, anastrozole was as effective as tamoxifen but not superior.[62] Several reasons for these differences have been proposed, including differences in the proportions of patients whose estrogen-receptor status was unknown or who had previously received adjuvant tamoxifen therapy, but none of these explanations are entirely adequate. Trials comparing exemestane with tamoxifen as first-line treatment are under way; promising early results [65] have led to an expanded European trial.


Table 2. Trials of Aromatase Inhibitors as Compared with Tamoxifen as First-Line Therapy.*

t2.png



In summary, in advanced disease, letrozole is clearly superior to tamoxifen as first-line therapy. For anastrozole, the data on superiority are contradictory, but the drug is convincingly at least as good as tamoxifen.


Second-Line Therapy

In the 1990s, the clinical importance of several third-generation inhibitors became clear when a series oftrials showed them to be more effective than megestrol acetate as second-line therapy after tamoxifen, despite some variation in the study re- sults [66-71] (Table 3). Trials of the second-generation inhibitors fadrozole and formestane and a trial of another third-generation agent, vorozole, now discontinued from clinical study, failed to show any such advantage.[41,72,73] The margin of additional benefit with anastrozole, letrozole, and exemestane was generally small, and the results differed slightly among the drugs,[74] but they were all associated with a very low incidence of serious side effects and with less unwanted weight gain than megestrol acetate. In practice, developments in first-line therapy rapidly diminished the clinical relevance ofthese findings.


Table 3. Trials ofAromatase Inhibitors as Compared with Megestrol Acetate as Second-Line Therapy.

t3.png



Early disease

Neoadjuvant Therapy

Trials of tamoxifen as an alternative to surgery in elderly women have consistently shown high rates ofshort-term tumor regression but poor long-term local control.[75] The option of endocrine therapy before, rather than instead of, surgery is more attractive, both as a means of down-staging primary cancers to avoid mastectomy[76] and as an in vivo measure of tumor responsiveness.[77] In small, non- randomized studies in older women (age, 59 to 88 years) with large primary tumors (diameter, >3 cm), preoperative administration of anastrozole, letrozole, or exemestane has resulted in rates of tumor regression higher than those previously reported for tamoxifen.[78,79] However, in a small, randomized trial ofpreoperative therapy, no difference was found between vorozole and tamoxifen.[80]


Evidence confirming that letrozole is superior to tamoxifen as neoadjuvant therapy has recently come from a randomized, double-blind trial in which use of the two agents for four months before surgery was assessed in older patients (median age, 67 years) with estrogen-receptor-positive or progesterone-receptor-positive large breast cancers usually requiring a mastectomy. The patients assigned to letrozole had a higher rate ofregression than those assigned to tamoxifen, and more ofthem had tumor regression sufficient to allow breast-conserving surgery [63,64] (Table 2).


There was also an unexpected and potentially important finding in a subgroup ofpatients whose tumors were available for further analysis: of17 patients whose tumors overexpressed the cell-surface growth factor receptor c-ErbB-2 (HER2), c-ErbB-1 (epidermal growth factor receptor [EGFR]) or both, 15 (88 percent) had a response to letrozole, as compared with only 4 of19 (21 percent) with a response to tamoxifen (Table 2).[64] These findings are consistent with the in vitro and in vivo observations that MCF-7 breast cancer cells and xenografts transfected with the c-erbB-2 gene do not grow without estrogen, whereas their growth continues in the presence of tamoxifen.[81] The results also support the concept of“crosstaIk” between the signal-transduction pathways for steroids and those for growth factors.


These data on the use of letrozole for neoadjuvant therapy are preliminary, however, and require verification in additionaI triaIs of aromatase inhibitors for neoadjuvant therapy, which are currentIy under way. Ifthose trials provide confirmatory data, they will support preoperative therapy with aromatase inhibitors as an effective and weII-toIerated aIternative to mastectomy for oIder patients with large, estrogen-receptor-positive cancers.


Adjuvant Therapy

Tamoxifen given for approximateIy five years after surgery to patients with earIy, estrogen-receptorpositive breast cancer is the current standard ofcare worldwide. This approach reduces the risk ofdeath by about 25 percent, a reduction that translates into an absolute improvement in 10-year survival of more than 10 percent for patients with involved nodes and 5 percent for those without.[13] This seem- ingIy Iimited increase transIates into many thousands of lives saved annually and almost certainly has contributed to the decIine in mortaIity from breast cancer seen over the past decade. It thus represents one of the main success stories in cancer medicine. However, the efficacy oftamoxifen is only partial. Furthermore, as described above, it is associated with an increased risk of uterine cancer — a risk that is small in absolute terms and far outweighed by the number of lives saved from breast cancer, but one that is very real in the public perception. Tamoxifen also increases the incidence of thromboembolism and often causes troublesome side effects, including hot flashes and vaginal discharge.[14] Thus, despite the benefits offered by tamoxifen, there is room for improvement.


The first trial ofan aromatase inhibitor given as adjuvant therapy was started more than 20 years ago with aminoglutethimide. By today’s standards, this study was very small, but it showed an early reduction in the risk of relapse or death; the reduction disappeared with longer follow-up.[82,83] In a more recent study, sequential administration of aminoglutethimide after tamoxifen therapy, as compared with tamoxifen alone,[84] was associated with a trend toward improved survival.


Trials ofadjuvant therapy with the third-generation aromatase inhibitors began roughly seven years ago. Currently, there are at least 10 ongoing studies ofthe use ofthese agents in postmenopausal women; they are scheduled to recruit almost 40,000 participants, and more such studies have been planned.


The designs ofthese trials differ, and among the key issues addressed are the use of these agents in direct comparison with tamoxifen, as combination therapy with tamoxifen, as sequential therapy with tamoxifen for a total offive years, and as maintenance therapy after five years of tamoxifen therapy. In the first and largest ofthese trials (Arimidex and Tamoxifen Alone or in Combination [ATAC] trial), which has three study groups, tamoxifen is being compared with anastrozole or with a combination oftamoxifen and anastrozole; 9366 patients have been enrolled. The first analysis, conducted at a median follow-up of 33 months, showed a small but statistically significant reduction in the rate of relapse with anastrozole as compared with tamoxifen: 89 percent ofthe patients assigned to anastrozole were relapse-free at 3 years, as compared with 87 percent of those assigned to tamoxifen (relative risk reduction, 17 percent; P=0.013).[85] The effect was seen only in patients whose tumors were known to be hormone-receptor-positive (relative risk reduction, 22 percent). So far, the ATAC trial has shown no differences in the rates ofdeath from any cause, and there have been very few breast cancer-related deaths.


Ofinterest, the combination of anastrozole and tamoxifen in the ATAC trial has not been found to be superior to tamoxifen alone. A possible explanation is that tamoxifen saturates available estrogen receptors and has partial agonist activity. The activated tamoxifen-estrogen-receptor complex cannot then be further modified by anastrozole-induced decreases in estrogen levels, and the anticancer effect remains the same as that provided by tamoxifen alone. Another finding, and one of potential relevance to breast-cancer prevention, is that the incidence ofcontralateral invasive breast cancer was significantly lower in the patients receiving anastrozole alone (0.3 percent [9 cancers]) than in those receiving tamoxifen alone (1.0 percent [30 cancers], P=0.001) or combined treatment (0.7 percent [23 cancers]).[85]


These findings are promising but preliminary. The absolute benefit in terms of freedom from relapse appears to be very small thus far, and no survival benefit has emerged. In addition, the anastrozole group has had a higher rate of fractures than the other two groups. No data on tolerability during five years oftreatment with any ofthe inhibitors are so far available. Long-term problems with tamoxifen, especially uterine cancer, emerged only after many years’ experience. It is our view that tamoxifen should remain the standard ofcare for most patients with early estrogen-receptor-positive breast cancer until further data become available. In patients with a history of thromboembolism, however, or those in whom tamoxifen is poorly tolerated, adjuvant therapy with anastrozole is now a useful alternative. This opinion is in accord with a recent American Society of Clinical Oncology evidence-based technology assessment,[86] which also appropriately advises against switching treatments in women who have already begun tamoxifen therapy. (Anastrozole has very recently been granted fast-track approval in the United States and elsewhere for adjuvant treatment of early hormone- receptor-positive breast cancer in postmenopausal women, particularly if tamoxifen is contraindicated.)



Adverse effects and long-term risks and benefits


The third-generation aromatase inhibitors appear to be very well tolerated, with a remarkably low incidence of serious short-term adverse effects, reflecting the remarkable specificity of their action. The commonest ofthese effects are hot flashes, vaginal dryness, musculoskeletal pain, and headache, but they are usually mild. Comparative trials indicate that such adverse effects are very similar in nature and frequency to those oftamoxifen.[60-63] Data from the ATAC trial, by far the largest trial of adjuvant therapy (and one that is not confounded by tumor-related symptoms), indicate that both treatments are well tolerated; however, the patients receiving anastrozole had a significantly lower incidence of hot flashes, vaginal bleeding, vaginal discharge, and venous thromboembolism and a significantly higher incidence of musculoskeletal symptoms and fractures than those receiving tamoxifen (Table 4).[85]


Table 4.Incidence ofAdverse Effects Associated with Anastrozole and Tamoxifen in the ATAC Trial.*

t4.png



Differences between the aromatase inhibitors and tamoxifen in long-term adverse effects are only starting to emerge. In contrast to findings with tamoxifen, there is no evidence to suggest an increased risk of uterine carcinoma with aromatase inhibitors (incidence, 0.1 percent, vs. 0.5 percent with tamoxifen) or venous thromboembolism (2.1 percent and 3.5 percent, respectively) (Table 4).[85]


Skeletal effects

The risk ofimportant long-term skeletal problems, including osteoporosis, may increase with the use of aromatase inhibitors. The maintenance of bone density depends in part on estrogen. Tamoxifen reduces bone demineralization through its agonist effect, at least in postmenopausal women,[20,21] whereas aromatase inhibitors may enhance this process by lowering circulating estrogen levels. Short-term use of letrozole has been shown to be associated with an increase in bone-resorption markers in plasma and urine,[87,88] and (as mentioned earlier) adjuvant therapy with anastrozole appears to be associated with a higher incidence of fractures than adjuvant therapy with tamoxifen.[85] However, it is possible that osteopenia might be prevented or modified with concurrent use of bisphosphonates.[89]


Cardiovascular effects

The cardiovascular effects of aromatase inhibitors are currently unknown. Tamoxifen appears to be estrogenic in this regard; in postmenopausal women it reduces the level of low-density lipoprotein cholesterol but causes high-density lipoprotein cholesterol to rise.[90,91] Whether such effects on lipids translate into clinical gain remains uncertain. Some trials have suggested that tamoxifen is associated with a reduction in coronary artery disease,[92-94] but so far such findings have not been confirmed, either in an overview[13] or in a large chemoprevention trial.[14] In contrast, the estrogen-lowering effects of aromatase inhibitors may prove to have an adverse effect on blood lipids: one small, short-term study in postmenopausal women with breast cancer has shown an increase in total serum cholesterol, low- density lipoprotein cholesterol, apolipoprotein B, and serum-lipid risk ratios for cardiovascular disease after 16 weeks of letrozole treatment.[95] The effect of aromatase inhibitors on lipids remains an important area for further research.


Effects on cognition

The brain is rich in estrogen receptors and contains aromatase,[26] and it has been suggested that estrogen-replacement therapy is associated with a reduced risk ofAlzheimer’s disease.[96] The results of randomized trials on the cognitive effect of estrogen in postmenopausal women are conflicting, but in one study estrogen replacement improved brain-activation patterns during working-memory tasks.[97] The long-term effects of aromatase inhibitors on cognitive function are unknown, and a great deal of careful follow-up will be required to assess this issue.


Hormone-replacement therapy and adjuvant breast-cancer therapy

Menopausal symptoms are an important source of morbidity in patients with breast cancer. Traditional wisdom has argued against the use of hormone-replacement therapy in such patients, but recently this belief has been challenged. Retrospective analyses have failed to confirm any increased risk of recurrence in women using hormone-replacement therapy after treatment for breast cancer,[98,99] and prospective trials are now addressing this issue. Theoretically, hormone-replacement therapy could be given in conjunction with adjuvant therapy with tamoxifen, on the basis ofthe efficacy oftamoxifen in premenopausal women, who have high circulating levels of estrogens. In contrast, hormone-replacement therapy would negate the action of aromatase-inhibitor therapy, and the combination would therefore be illogical.


On balance, therefore, the potential gains in efficacy with the aromatase inhibitors as compared with tamoxifen should be weighed carefully against the long-term risks and short-term quality-of-life issues associated with hormone-replacement therapy. For some women at relatively low risk ofrecurrence, a decision on the balance between efficacy and side effects may be difficult, since background information is currently inadequate.



Chemoprevention


A substantial body of evidence supports the role of estrogen in the development ofbreast cancer.[100] Such evidence includes data from prospective studies relating plasma sex-steroid levels to the risk of subsequent breast cancer.[101] Chemoprevention with aromatase inhibitors might be particularly suitable for women with relatively high plasma estrogen levels. Two chemoprevention trials have already shown that tamoxifen reduces the incidence of breast cancer,[14,15] and previous trials of adjuvant tamoxifen have likewise shown an almost 50 percent reduction in the development of cancer in the contralateral breast.[13] The results ofthe ATAC trial with regard to the development of contralateral invasive breast cancer (in 30 [1.0 percent] ofthose receiving tamoxifen vs. 9 [0.3 percent] of those receiving anastrozole after a median of 33 months of follow-up) suggest, by extrapolation, that anastrozole might reduce the early incidence ofbreast cancer to an even greater extent and thus have more potential in chemoprevention than tamoxifen.


Strategies to avoid the anticipated loss of bone density induced by aromatase inhibitors would first need to be developed. An alternative approach might be to use a much smaller dose ofaromatase inhibitor in order to lower the levels of circulating estrogens but not obliterate them. Such an approach might offer a substantial chemopreventive effect and reduce the risk of serious long-term complications.


Other issues


Is there a best third-generation aromatase inhibitor?

Letrozole resulted in greater inhibition ofaromatase than anastrozole in a crossover pharmacodynamic trial,[54] and evidence of the superiority of letrozole over tamoxifen in advanced disease is solid. Preliminary data from a comparative trial of these two inhibitors in advanced breast cancer after tamoxifen are confUsing: letrozole was associated with significantly more tumor regressions overall than anastrozole, but not in the subgroup with known estrogen-receptor-positive tumors.[102] There are no comparative data on exemestane, although occasional further responses have been reported for it and the second-generation inhibitor formestane in patients with relapses after therapy with anastrozole, letrozole, or the other nonsteroidal inhibitors.[103,104] This absence of total cross-resistance is not explained by the degree ofestrogen suppression and must involve other biochemical effects. Overall, current circumstantial evidence suggests that there are unlikely to be major clinical differences among these agents.


Aromatase inhibitors in combination with chemotherapy

No studies have compared concurrent use ofaromatase inhibitors and chemotherapy with sequential use. The concurrent use of tamoxifen and chemotherapy increases the risk ofthromboembolism,105 but this problem does not appear to occur with the aromatase inhibitors.



Conclusions


The third-generation aromatase inhibitors are a new development in the endocrine treatment of estrogen-receptor-positive breast cancer in postmenopausal women. In the treatment of advanced disease, letrozole is convincingly better than tamoxifen, and anastrozole is at least as good. In early breast cancer, adjuvant therapy with anastrozole already appears to be superior to adjuvant therapy with tamoxifen in reducing the risk of relapse, and letrozole appears to be more effective than tamoxifen as preoperative therapy. It is possible that third- generation aromatase inhibitors will have a future role in chemoprevention, but the long-term effects of profound estrogen suppression in postmenopausal women are unknown, and careful monitoring for bone demineralization and other potential problems is essential as their role evolves.


We are indebted to Alison Norton for invaluable secretarial and editorial assistance and advice and to Dr. Alistair Ring for helpful comments.




References


 Ingle JN, Ahmann DL, Green SJ, et al. Randomized clinical trial of diethylstil- bestrol versus tamoxifen in postmenopausal women with advanced breast cancer. N Engl J Med 1981;304:16-21.

 Muss HB, Wells HB, Paschold EH, et al. Megestrol acetate versus tamoxifen in advanced breast cancer: 5-year analysis — a phase III trial of the Piedmont Oncology Association. J Clin Oncol 1988;6:1098-106.

 Gill PG, Gebski V, Snyder R, et al. Randomized comparison ofthe effects oftamox- ifen, megestrol acetate, or tamoxifen plus megestrol acetate on treatment response and survival in patients with metastatic breast cancer. Ann Oncol 1993;4:741-4.

 Muss HB, Case LD, Atkins JN, et al. Tamoxifen versus high-dose oral medroxypro gesterone acetate as initial endocrine therapy for patients with metastatic breast cancer: a Piedmont Oncology Association study. J Clin Oncol 1994;12:1630-8.

 Kellokumpu-Lehtinen P, Huovinen R, Johansson R. Hormonal treatment of advanced breast cancer: a randomized trial of tamoxifen versus nandrolone decanoate. Cancer 1987;60:2376-81.

 Hayes DF, Van Zyl JA, Hacking A, et al. Randomized comparison of tamoxifen and two separate doses of toremifene in postmenopausal patients with metastatic breast cancer. J Clin Oncol 1995;13:2556-66.

 Stenbygaard LE, Herrstedt J, Thomsen JF, Svendsen KR, Engelholm SA, Domber- nowsky P. Toremifene and tamoxifen in advanced breast cancer — a double-blind cross-over trial. Breast Cancer Res Treat 1993;25:57-63.

 Smith IE, Harris AL, Morgan M, et al. Tamoxifen versus aminoglutethimide in advanced breast carcinoma: a randomised cross-over trial. Br Med J (Clin Res Ed) 1981; 283:1432-4.

 Lipton A, Harvey HA, Santen RJ, et al. A randomised trial of aminoglutethimide versus tamoxifen in metastatic breast cancer. Cancer 1982;50:2265-8.

 Gale KE, Andersen JW, Tormey DC, et al. Hormonal treatment for metastatic breast cancer: an Eastern Cooperative Oncology Group Phase III trial comparing aminoglu-tethimide to tamoxifen. Cancer 1994;73: 354-61.

 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.

 Thurlimann 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.

 Early Breast Cancer Trialists’ Collaborative Group. Tamoxifen for early breast cancer: an overview of the randomised trials. Lancet 1998;351:1451-67.

 Fisher B, Costantino JP, Wickerham DL, et al. Tamoxifen for prevention of breast cancer: report of the National Surgical Adjuvant Breast and Bowel Project P-1 Study. J Natl Cancer Inst 1998;90:1371-88.

 IBIS investigators. First results from the International Breast Cancer Intervention Study (IBIS-I): a randomised prevention trial. Lancet 2002;360:817-24.

 Goss PE, Strasser K. Aromatase inhibitors in the treatment and prevention of breast cancer. J Clin Oncol 2001;19:881-94.

 Buzdar A, Howell A. Advances in aromatase inhibition: clinical efficacy and tolerability in the treatment of breast cancer. Clin Cancer Res 2001;7:2620-35.

 Beatson GT. On the treatment ofinoper- able cases ofcarcinoma ofthe mamma: suggestions for a new method of treatment, with illustrative cases. Lancet 1896;2:104-7.

 Howell A, Dowsett M. Recent advances in endocrine therapy of breast cancer. BMJ 1997;315:863-6.

 Powles TJ, Hickish T, Kanis JA, Tidy A, Ashley S. Effect of tamoxifen on bone mineral density measured by dual-energy x-ray absorptiometry in healthy premenopausal and postmenopausal women. J Clin Oncol 1996;14:78-84.

 Love RR, Barden HS, Mazess RB, Epstein S, Chappell RJ. Effect of tamoxifen on lumbar spine bone mineral density in postmenopausal women after 5 years. Arch Intern Med 1994;154:2585-8.

 Tamoxifen and endometrial cancer. ACOG committee opinion. No. 232. Washington, D.C.: American College of Obstetricians and Gynecologists, 2000.

 DeFriend DJ, Anderson E, Bell J, et al. Effects of 4-hydroxytamoxifen and a novel pure antioestrogen (ICI 182780) on the clo- nogenic growth of human breast cancer cells in vitro. Br J Cancer 1994;70:204-11.

 Evans CT, Ledesma DB, Schulz TZ, Simpson ER, Mendelson CR. Isolation and characterization of a complementary DNA specific for human aromatase-system cytochrome P-450 mRNA. Proc Natl Acad Sci U S A 1986;83:6387-91.

 Miller WR, Hawkins RA, Forrest AP. Significance of aromatase activity in human breast cancer. Cancer Res 1982;42:Suppl: 3365s-3368s.

 Nelson LR, Bulun SE. Estrogen production and action. J Am Acad Dermatol 2001; 45:Suppl:S116-S124.

 Longcope C, Baker R, Johnston CC Jr. Androgen and estrogen metabolism: relationship to obesity. Metabolism 1986;35: 235-7.

 Thijssen JH, Blankenstein MA. Endogenous oestrogens and androgens in normal and malignant endometrial and mammary tissues. Eur J Cancer Clin Oncol 1989;25: 1953-9.

 Miller WR, O’Neill J. The importance of local synthesis ofestrogen within the breast. Steroids 1987;50:537-48.

 Simpson ER, Clyne C, Rubin G, et al. Aromatase — a brief overview. Annu Rev Physiol 2002;64:93-127.

 Simpson ER, Dowsett M. Aromatase and its inhibitors: significance for breast cancer therapy. Recent Prog Horm Res 2002;57:317-38.

 Sinha S, Kaseta J, Santner SJ, Demers LM, Bremmer WJ, Santen RJ. Effect of CGS 20267 on ovarian aromatase and gonadotropin levels in the rat. Breast Cancer Res Treat 1998;48:45-51.

 Stein RC, Dowsett M, Hedley A, Gazet JC, Ford HT, Coombes RC. The clinical and endocrine effects of 4-hydroxyandrostene- dione alone and in combination with gose- relin in premenopausal women with advanced breast cancer. Br J Cancer 1990;62: 679-83.

 Mitwally MF, Casper RF. Aromatase inhibition for ovarian stimulation: future avenues for infertility management. Curr Opin Obstet Gynecol 2002;14:255-63.

 Santen RJ, Samojlik E, Lipton A, et al. Kinetic, hormonal and clinical studies with aminoglutethimide in breast cancer. Cancer 1977;39:Suppl:2948-58.

 Smith IE, Fitzharris BM, McKinna JA, et al. Aminoglutethimide in treatment of metastatic breast carcinoma. Lancet 1978;2: 646-9.

 Santen RJ, Santner S, Davis B, Veldhuis J, Samojlik E, Ruby E. Aminoglutethimide inhibits extraglandular estrogen production in postmenopausal women with breast carcinoma. J Clin Endocrinol Metab 1978;47: 1257-65.

 Stuart-Harris R, Dowsett M, Bozek T, et al. Low-dose aminoglutethimide in treatment of advanced breast cancer. Lancet 1984;2:604-7.

 Stein RC, Dowsett M, Headley A. Treatment of advanced breast cancer in postmenopausal women with 4-hydroxyandro- stenedione. Cancer Chemother Pharmacol 1990;26:75-8.

 Steele RE, Mellor LB, Sawyer WK, Was- vary JM, Browne LJ. In vitro and in vivo studies demonstrating potent and selective oestrogen inhibition with the nonsteroidal aromatase inhibitor CGS 16949A. Steroids 1987;50:147-61.

 Buzdar AU, Smith R, Vogel C, et al. Fadrozole HCL (CGS-16949A) versus me- gestrol acetate treatment of postmenopaus

 al patients with metastatic breast carcinoma: results of two randomized double blind controlled multiinstitutional trials. Cancer 1996; 77:2503-13.

 Lonning PE, Jacobs S, Jones A, Haynes B, Powles T, Dowsett M. The influence of CGS 16949A on peripheral aromatisation in breast cancer patients. Br J Cancer 1991;63: 789-93.

 Plourde PV, DyroffM, Dowsett M, Demers L, Yates R, Webster A. ARIMIDEX: a new oral, once-a-day aromatase inhibitor. J Steroid Biochem Mol Biol 1995;53:175-9.

 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.

 Bisagni G, Cocconi G, Scaglione F, Fras- chini F, Pfister C, Trunet PF. Letrozole, a new oral non-steroidal aromatase inhibitor in treating postmenopausal patients with advanced breast cancer: a pilot study. Ann Oncol 1996;7:99-102.

 Lamb HM, Adkins JC. Letrozole: a review ofits use in postmenopausal women with advanced breast cancer. Drugs 1998;56:1125- 40.

 Wiseman LR, Adkins JC. Anastrozole: a review of its use in the management of postmenopausal women with advanced breast cancer. Drugs Aging 1998;13:321-32.

 Lonning PE. Pharmacological profiles of exemestane and formestane, steroidal aromatase inhibitors used for treatment of postmenopausal breast cancer. Breast Cancer Res Treat 1998;49:Suppl 1:S45-S52, S73-S77.

 Lien EA, Anker G, Lonning PE, Solheim E, Ueland PM. Decreased serum concentrations of tamoxifen and its metabolites induced by aminoglutethimide. Cancer Res 1990;50:5851-7.

 Pharmacokinetics of anastrozole and tamoxifen alone, and in combination, during adjuvant endocrine therapy for early breast cancer in postmenopausal women: a subprotocol of the ‘Arimidex and tamoxifen alone or in combination’ (ATAC) trial. Br J Cancer 2001;85:317-24.

 Dowsett M, Pfister C, Johnston SR, et al. Impact of tamoxifen on the pharmacokinetics and endocrine effects of the aromatase inhibitor letrozole in postmenopausal women with breast cancer. Clin Cancer Res 1999; 5:2338-43.

 Miller WR, Dixon JM. Antiaromatase agents: preclinical data and neoadjuvant therapy. Clin Breast Cancer 2000;1:Suppl 1: S9-S14.

 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.

 Geisler J, Haynes B, Anker G, Dowsett M, Lonning PE. Influence of letrozole and anastrozole on total body aromatization and plasma estrogen levels in postmenopausal breast cancer patients evaluated in a randomized, cross-over study. J Clin Oncol 2002;20: 751-7.

 MacNeill FA, Jones AL, Jacobs S, Lon- ning PE, Powles TJ, Dowsett M. The influence of aminoglutethimide and its analogue rogletimide on peripheral aromatisation in breast cancer. Br J Cancer 1992;66:692-7.

 Gershanovich M, Chaudri HA, Campos D, et al. Letrozole, a new oral aromatase inhibitor: randomised trial comparing 2.5 mg daily, 0.5 mg daily and aminoglutethimide in postmenopausal women with advanced breast cancer. Ann Oncol 1998;9:639-45.

 Tominaga T, Morimoto T, Ohashi Y. A pivotal double-blind trial in Japan of an aromatase inhibitor letrozole (third-generation) vs. its predecessor fadrozole hydrochloride (second generation). Ann Oncol 2000;11:Suppl 4:25. abstract.

 Bergh J, Bonneterre J, Illiger HJ, et al. Vorozole (Rivizor) versus aminoglutethimide (AG) in the treatment of advanced postmenopausal breast cancer relapsing after tamoxifen. Prog Proc Am Soc Clin Oncol 1997;16:155a. abstract.

 Miller WR, Dixon JM. Local endocrine effects of aromatase inhibitors within the breast. J Steroid Biochem Mol Biol 2001;79: 93-102.

 Mouridsen H, Gershanovich M, Sun Y, et al. Superior efficacy of letrozole versus tamoxifen as first-line therapy for postmenopausal women with advanced breast cancer: results of a phase III study of the International Letrozole Breast Cancer Group. J Clin Oncol 2001;19:2596-606. [Erratum, J Clin Oncol 2001;19:3302.]

 Nabholtz JM, Buzdar A, Pollak M, et al. Anastrozole is superior to tamoxifen as first-line therapy for advanced breast carcinoma in postmenopausal women: results of a North American multicenter randomized trial. J Clin Oncol 2000;18:3758-67.

 Bonneterre J, Thurlimann B, Robertson JFR, et al. Anastrozole versus tamoxifen as first-line therapy for advanced breast cancer in 668 postmenopausal women: results of the Tamoxifen or Arimidex Randomized Group Efficacy and Tolerability study. J Clin Oncol 2000;18:3748-57.

 Eiermann W, Paepke S, Appfelstaedt J, et al. Preoperative treatment of postmenopausal breast cancer patients with letrozole: a randomized double-blind multicenter study. Ann Oncol 2001;12:1527-32.

 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.

 Dirix L, Piccart MJ, Lohrisch C, et al. Efficacy ofand tolerance to exemestane (E) versus tamoxifen (T) in 1st line hormone therapy (HT) ofpostmenopausal metastatic breast cancer (MBC) patients (pts): a European Organisation for the Research and Treatment of Cancer (EORTC Breast Group) phase II trial with Pharmacia Upjohn. Prog Proc Am Soc Clin Oncol 2001;20:29a. abstract.

 Buzdar AU, Jonat W, Howell A, et al. 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. Cancer 1998;83:1142-52. [Erratum, Cancer 1999;15:1010.]

 Jonat W, Howell A, Blomqvist C, et al. A randomised trial comparing two doses of the new selective aromatase inhibitor anastrozole (Arimidex) with megestrol acetate in postmenopausal patients with advanced breast cancer. Eur J Cancer 1996;32A:404- 12.

 Buzdar A, Jonat W, Howell A, et al. Anastrozole, a potent and selective aromatase inhibitor, versus megestrol acetate in postmenopausal women with advanced breast cancer: results of overview analysis of two phase III trials. J Clin Oncol 1996; 14:2000-11.

 Dombernowsky P, Smith I, Falkson G, et al. Letrozole, a new oral aromatase inhibitor for advanced breast cancer: doubleblind randomized trial showing a dose effect and improved efficacy and tolerability compared with megestrol acetate. J Clin Oncol 1998;16:453-61.

 Buzdar A, Douma N, Davidson N, et al. Phase III, multicenter, double-blind, randomized study of letrozole, an aromatase inhibitor, for advanced breast cancer versus megestrol acetate. J Clin Oncol 2001;19: 3357-66.

 Kaufmann M, Bajetta E, Dirix LY, et al. Exemestane is superior to megestrol acetate after tamoxifen failure in postmenopausal women with advanced breast cancer: results ofa phase III randomized double-blind trial. J Clin Oncol 2000;18:1399-411.

 Goss PE, Winer EP, Tannock IF, Schwartz LH. Randomized phase III trial comparing the new potent and selective third-generation aromatase inhibitor vorozole with me- gestrol acetate in postmenopausal advanced breast cancer patients. J Clin Oncol 1999; 17:52-63.

 Thurlimann B, Castiglione M, Hsu- Schmitz SF, et al. Formestane versus meges- trol 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.

 Hamilton A, Piccart M. The third-generation non-steroidal aromatase inhibitors: a review of their clinical benefits in the second-line hormonal treatment of advanced breast cancer. Ann Oncol 1999;10:377-84.

 Cheung KL, Howell A, Robertson JF. Preoperative endocrine therapy for breast cancer. Endocr Relat Cancer 2000;7:131-41.

 Smith IE, Lipton L. Preoperative/neoad- juvant medical therapy for early breast cancer. Lancet Oncol 2001;2:561-70.

 Forrest AP, Levack PA, Chetty U, et al. A human tumour model. Lancet 1986;2: 840-2.

 Dixon JM, Neoadjuvant endocrine therapy. In: Miller WR, Santen RJ, eds. Aromatase inhibition and breast cancer. New York: Marcel Dekker, 2000:103-16.

 Miller WR, Dixon JM. Endocrine and clinical endpoints of exemestane as neoadjuvant therapy. Cancer Control 2002;9: Suppl:9-15.

 Harper-Wynne CL, Sacks NPM, Shen- ton K, et al. Comparison of the systemic and intratumoral effects of tamoxifen and the aromatase inhibitor vorozole in postmenopausal patients with primary breast cancer. J Clin Oncol 2002;20:1026-35.

 Benz CC, Scott GK, Sarup JC, et al. Estrogen-dependent, tamoxifen-resistant tu- morigenic growth of MCF-7 cells transfected with HER2/neu. Breast Cancer Res Treat 1993;24:85-95.

 Coombes RC, Powles TJ, Easton D, et al. Adjuvant aminoglutethimide therapy for postmenopausal patients with primary breast cancer. Cancer Res 1987;47:2494-7.

 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.

 Boccardo F, Rubagotti A, Amoroso D, et al. Sequential tamoxifen and aminoglute- thimide versus tamoxifen alone in the adjuvant treatment of postmenopausal breast cancer patients: results of an Italian cooperative study. J Clin Oncol 2001;19:4209-15.

 ATAC 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 2002;359:2131-9. [Erratum, Lancet 2002;360:1520.]

 Winer EP, Hudis C, Burstein HJ, et al. American Society of Clinical Oncology technology assessment on the use of aromatase inhibitors as adjuvant therapy for women with hormone receptor-positive breast cancer: status report 2002. J Clin Oncol 2002; 20:3317-27.

 Harper-Wynne C, Ross G, Sacks N, et al. Effects of the aromatase inhibitor letrozole on normal breast epithelial cell proliferation and metabolic indices in postmenopausal women: a pilot study for breast cancer prevention. Cancer Epidemiol Biomarkers Prev 2002;11:614-21.

 Heshmati HM, Khosla S, Robins SP, O’Fallon WM, Melton LJ III, Riggs BL. Role of low levels of endogenous estrogen in regulation of bone resorption in late postmenopausal women. J Bone Miner Res 2002;17: 172-8.

 Cummings SR, Black DM, Thompson DE, et al. Effect of alendronate on risk of fracture in women with low bone density but without vertebral fractures: results from the Fracture Intervention Trial. JAMA 1998; 280:2077-82.

 Bruning PF, Bonfrer JMG, Hart AAM, et al. Tamoxifen, serum lipoproteins and cardiovascular risk. Br J Cancer 1988;58:497-9.

 Saarto T, Blomqvist C, Ehnholm C, Taskinen MR, Elomaa I. Antiatherogenic effects of adjuvant antiestrogens: a randomized trial comparing the effects oftamoxifen and toremifene on plasma lipid levels in postmenopausal women with node-positive breast cancer. J Clin Oncol 1996;14:429-33.

 McDonald CC, Alexander FE, Whyte BW, Forrest AP, Stewart HJ. Cardiac and vascular morbidity in women receiving adjuvant tamoxifen for breast cancer in a randomised trial. BMJ 1995;311:977-80.

 Costantino JP, Kuller LH, Ives DG, Fisher B, Dignam J. Coronary heart disease mortality and adjuvant tamoxifen therapy. J Natl Cancer Inst 1997;89:776-82.

 Rutqvist LE, Mattsson A. Cardiac and thromboembolic morbidity among postmenopausal women with early-stage breast cancer in a randomized trial of adjuvant tamoxifen. J Natl Cancer Inst 1993;85:1398- 406.

 Elisaf MS, Bairaktari ET, Nicolaides C, et al. Effect of letrozole on the lipid profile in postmenopausal women with breast cancer. Eur J Cancer 2001;37:1510-3.

 Tang MX, Jacobs D, Stern Y, et al. Effect of oestrogen during menopause on risk and age at onset of Alzheimer’s disease. Lancet 1996;348:429-32.

 Shaywitz SE, Shaywitz BA, Pugh KR, et al. Effect of estrogen on brain activation patterns in postmenopausal women during working memory tasks. JAMA 1999;281: 1197-202.

 O’Meara ES, Rossing MA, Daling JR, Elmore JG, Barlow WE, Weiss NS. Hormone replacement therapy after a diagnosis of breast cancer in relation to recurrence and mortality. J Natl Cancer Inst 2001;93:754-62.

 Vassilopoulou-Sellin R, Asmar L, Hor- tobagyi GN, et al. Estrogen replacement therapy after localized breast cancer: clinical outcome of 319 women followed prospectively. J Clin Oncol 1999;17:1482-7.

 Henderson BE, Bernstein L. The international variation in breast cancer rates: an epidemiological assessment. Breast Cancer Res Treat 1991;18:Suppl 1:S11-S17.

 The Endogenous Hormones and Breast Cancer Collaborative Group. Endogenous sex hormones and breast cancer in postmenopausal women: reanalysis of nine prospective studies. J Natl Cancer Inst 2002; 94:606-16.

 Rose C, Vtoraya O, Pluzanska A, et al. Letrozole (Femara) vs anastrozole (Arimidex): second-line treatment in postmenopausal women with advanced breast cancer. Prog Proc Am Soc Clin Oncol 2002;21:34a. abstract.

 Lonning 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.

 Carlini P, Frassoldati A, De Marco S, et al. Formestane, a steroidal aromatase inhibitor after failure of non-steroidal aromatase inhibitors (anastrozole and letrozole): is a clinical benefit still achievable? Ann Oncol 2001;12:1539-43.

 Pritchard KI, Paterson AH, Fine S, et al. Randomized trial of cyclophosphamide, methotrexate, and fluorouracil chemotherapy added to tamoxifen as adjuvant therapy in postmenopausal women with node-positive estrogen and/or progesterone receptor-positive breast cancer: a report of the National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 1997;15: 2302-11.

 Copyright © 2003 Massachusetts Medical Society.