Adjuvant Hormonal Therapy for Early-Stage Breast Cancer




Adjuvant endocrine treatment is an essential component in therapy for hormone receptor–positive breast cancer. Among postmenopausal patients, options include tamoxifen, aromatase inhibitors, or a sequence of these agents. Tamoxifen and aromatase inhibitors have distinctive side-effect profiles. Among premenopausal women, tamoxifen remains the standard treatment. The role of ovarian suppression in addition to tamoxifen is under investigation. Questions about the duration of adjuvant endocrine therapy, the use of biomarkers for treatment selection and prognosis, and the management of side effects of adjuvant endocrine therapy remain key areas of investigation.


Adjuvant endocrine therapy is critical for reducing the risk of recurrence and promoting survival in women with hormone receptor (estrogen receptor [ER] or progesterone receptor [PgR])–positive breast cancers. Adjuvant endocrine therapy reduces the risk of tumor recurrence by approximately 40%, on average. Because of that substantial effect and because of the prevalence of hormone receptor–positive breast cancer, it is arguable that adjuvant endocrine therapy has had greater impact on reducing mortality from cancer than any other medical therapy. In addition to preventing metastatic recurrence, adjuvant endocrine therapy reduces the risk of in-breast or regional recurrences and lowers the risk of contralateral breast cancer. In light of these benefits, adjuvant endocrine therapy is recommended for nearly all patients with hormone receptor–positive breast cancer irrespective of tumor size or nodal status.


The prerequisite of benefit from treatment with adjuvant endocrine therapy is tumor expression of the ER or PgR. In the absence of tumor expression of these hormone receptors, adjuvant endocrine therapy is not of clinical benefit. This observation leads to the clinical requirement of high-quality hormone receptor testing on all newly diagnosed breast cancers. A variety of prognostic factors have emerged that are associated with recurrence risk of ER-positive breast cancer ( Box 1 ). These markers can inform the likelihood of tumor recurrence and, based on the assumption of proportional risk reduction associated with adjuvant endocrine treatment, may serve to estimate the absolute magnitude of treatment effect. To date, however, no single marker aside from expression of the hormone receptors themselves is adequate for identifying which patients might, or might not, benefit from adjuvant endocrine therapy. Similarly, no single marker can identify which endocrine treatment strategy might be optimal for a given patient.



Box 1





  • Tumor size (T)



  • Nodal status (N)



  • Tumor grade



  • Quantitative levels of hormone receptor expression



  • HER2 expression status



  • Lymphovascular invasion



  • Proliferative markers, such as Ki-67



  • Multigene prognostic signatures, such as the 21-gene recurrence score (Oncotype DX assay, Genomic Health Inc, Redwood City, CA, USA)



Prognostic factors in hormone receptor–positive breast cancer


It is now appreciated that ER-positive breast cancers have a different pattern of tumor recurrence from other forms of breast cancer. In the absence of adjuvant endocrine therapy, there is a peak of recurrences in years 2 through 5 and then a steady persistent risk of recurrence through at least 15 years after diagnosis. Introduction of adjuvant endocrine therapy dramatically reduces the incidence of early recurrences in the first 10 years after diagnosis. Endocrine therapy is also associated with a carryover effect, such that patients who have had 5 years of therapy continue to have a lower risk of recurrence through at least 15 years of follow-up after breast cancer diagnosis. With longer passage of time, second breast cancers and non–breast cancer mortality contribute a greater percentage of events in studies of adjuvant endocrine therapy. These observations underscore the need for long-term follow-up of patients receiving adjuvant endocrine therapy.


Tamoxifen


Tamoxifen, a selective ER modulator, has been the historic standard for adjuvant endocrine therapy. Multiple prospective, randomized, clinical trials of adjuvant tamoxifen therapy have been analyzed as part of the Early Breast Cancer Trialists’ Group overview analysis. Five years of tamoxifen therapy lowers the risk of breast cancer recurrence by approximately 40% and lowers the risk of breast cancer mortality by approximately 20%. The benefits of tamoxifen are seen regardless of patient age or menopausal status. The optimal duration of tamoxifen therapy seems to be a total of 5 years. Published studies that have examined continuation of tamoxifen treatment beyond 5 years have not demonstrated a significant clinical benefit for additional therapy. Preliminary studies from the large Adjuvant Tamoxifen Treatment Offers More? (ATTOM) and Adjuvant Tamoxifen—Longer Against Shorter (ATLAS) trials have raised anew questions regarding the optimal duration of tamoxifen treatment; however, at present, 5 years remains the standard for women taking tamoxifen alone.


The side effects of tamoxifen have been well characterized ( Table 1 ). Common side effects include menopausal symptoms, such as hot flashes and night sweats, and in premenopausal women, menstrual irregularities. Tamoxifen is associated with a low but increased risk of uterine cancer and deep venous thrombosis, particularly in postmenopausal women. Quality-of-life studies suggest that most women on tamoxifen have a well-preserved quality of life in all functional domains and that most women are reasonably adherent with treatment.



Table 1

Side effects of adjuvant endocrine therapy


































Tamoxifen Aromatase Inhibitors Ovarian Suppression
Gynecologic Vaginal discharge or dryness/atrophy; increased risk of vaginal bleeding and uterine cancer Vaginal dryness/atrophy Vaginal dryness/atrophy
Menstrual function Irregular menstrual cycles or amenorrhea Not applicable Amenorrhea
Menopausal symptoms Hot flashes, night sweats Hot flashes, night sweats Hot flashes, night sweats
Musculoskeletal health Mixed effects on bone density Osteopenia, osteoporotic fractures; musculoskeletal (arthralgia) syndrome Osteopenia
Cardiovascular health Increased risk of deep vein thrombosis Increased risk of hypercholesterolemia, hypertension Unknown




Adjuvant endocrine therapy for postmenopausal women with hormone receptor–positive early-stage breast cancer


The introduction of aromatase inhibitors (AIs) has defined a new era of adjuvant endocrine treatment for postmenopausal women with hormone receptor–positive breast cancer. AIs function by blocking the conversion of androgens into estrogens through the aromatase enzyme. The consequence of such therapy for postmenopausal women is profound estrogen deprivation—circulating estrogen levels are typically depleted by 90% from baseline. It is presumed that the resultant estrogen deprivation causes the antineoplastic effects of AI therapy. Because premenopausal women have residual ovarian function and the capacity to up-regulate aromatase expression in ovarian tissues in response to estrogen deprivation, AIs are contraindicated in premenopausal women.


AIs have been studied in several contexts as adjuvant endocrine treatment for postmenopausal women. Because the historical standard for adjuvant therapy has been 5 years of tamoxifen, most of the major adjuvant trials have compared AI-based treatment with 5 years of tamoxifen. The development of AIs as adjuvant treatment has included studies of primary endocrine therapy, which used AIs as initial treatment instead of tamoxifen; sequential endocrine therapy, which integrated AIs as adjuvant treatment after several (typically, 2 to 3) years of tamoxifen; and extended adjuvant therapy, which explored AI-based treatment after 5 years of adjuvant tamoxifen. Table 2 identifies the major adjuvant trials that have reported in the past decade on the role of AIs in the adjuvant setting.



Table 2

Major trials of AI therapy as adjuvant treatment of postmenopausal, early-stage, hormone receptor–positive breast cancer




































































Study Name Schema Total Duration of Therapy (y) AI References
Primary endocrine therapy ATAC TAM versus AI versus TAM + AI 5 ANA
BIG 1-98 TAM → AI versus AI → TAM versus TAM versus AI 5 LET
ABCSG 12 TAM versus AI
premenopausal at diagnosis; all patients receive ovarian suppression
5 ANA
TEAM AI versus TAM → AI 5 EXE
Sequential endocrine therapy (after 2–3 years of TAM) IES TAM versus TAM → AI 5 EXE
ARNO 95, ABSCG 8, Italian TAM versus TAM → AI 5 ANA
Extended endocrine therapy (after 5 years of TAM) MA 17 AI versus placebo 10 LET
ABCSG 6a AI versus placebo 7 ANA
NSABP B-22 AI versus placebo 10 EXE

Abbreviations: ABCSG, Austrian Breast and Colorectal Cancer Study Group; ANA, anastrozole; ARNO 95, Arimidex-Nolvadex; EXE, exemestane; IES, Intergroup Exemestane Study; LET, letrozole; NSABP, National Surgical Adjuvant Breast and Bowel Project; TAM, tamoxifen.


Collectively, these trials have demonstrated important observations. First, incorporation of an AI during the first 5 years of adjuvant endocrine treatment, or as extended therapy after 5 years of tamoxifen, is associated with a reduction in the risk of breast cancer recurrence. This reduction in risk is approximately a 15% to 20% proportionate risk reduction compared with tamoxifen alone. Because of the generally favorable prognosis for most postmenopausal women with early-stage breast cancer, the absolute difference in breast cancer events associated with AI-based therapy compared with tamoxifen is approximately 2% to 3%. The improvement in breast cancer outcomes includes reduction in the risk of distant metastatic recurrence as well as reduction in local/in-breast events and contralateral breast cancer. These modest gains are noted in comparing initial use of an AI with tamoxifen or in comparing tamoxifen alone with a sequence of tamoxifen followed by an AI. To date, studies have not reported a significant survival advantage for initial use of an AI compared with a sequential treatment program of tamoxifen and an AI. Compared with treatment with tamoxifen alone, sequential use of an AI may confer a modest survival advantage.


Second, it seems that a variety of treatment strategies yield similar outcomes, provided that an AI is incorporated at some point. Direct comparisons of results between initial use of an AI and a sequential program of a tamoxifen followed by an AI show equivalent rates of tumor recurrence in the Breast International Group (BIG) 1-98 and Tamoxifen Exemestane Adjuvant Multicentre (TEAM) trials. In the Arimidex, Tamoxifen Alone or in Combination (ATAC) study, the combination of an AI with tamoxifen was not superior to tamoxifen alone.


These findings support the recommendation for consideration of an AI at some point during adjuvant endocrine treatment in postmenopausal women. The optimal timing of AI therapy vis-à-vis tamoxifen remains unknown. It is also unknown whether or not a longer total duration of treatment, accomplished with extended therapy using tamoxifen followed by an AI, would differ from initial use of an AI. Another persistent question remains the best duration of AI treatment itself. Different studies used different durations of AI therapy. As primary treatment, data show equivalence for 2.5 or 5 years of AI therapy. Ongoing trials are examining whether or not longer-term use of an AI in excess of 5 years’ total duration is safe and effective. No studies reported to date have directly compared the cancer outcomes for one AI with another as adjuvant treatment. In broad terms, the findings with each of the commercially available AIs (anastrozole, exemestane, and letrozole) seem qualitatively similar; thus, it seems likely that the benefits seen with AI treatment represent a class effect. Ongoing trials are comparing directly one AI with another.


Clinical experience with in the adjuvant setting has identified common side effects of AI treatment (see Table 1 ). Patients taking AIs are at greater risk for musculoskeletal health problems, including accelerated osteoporosis and fractures, than are women taking tamoxifen. Bisphosphonate therapy seems to mitigate AI-associated loss of bone mineral density. AIs are associated with a unique arthralgia syndrome, characterized by muscle and joint stiffness and achiness, which is common although usually of modest intensity. AIs are also associated with a slightly greater risk of hypertension and hypercholesterolemia; the long-term cardiac consequences of these effects are not yet well characterized.


There remains considerable interest in efforts to tailor specific adjuvant endocrine treatment options for individual patients based on tumor characteristics or biomarkers or based on consideration of pharmacogenetic studies. These retrospective efforts remain inconclusive. At present, it does not seem that there are sufficient data for using CYP2D6 genotyping for deciding whether or not tamoxifen is a suitable treatment option for specific women. A variety of pathologic and other biomarker studies confirm prognostic markers for patients treated with AIs. These measures lack sufficient data, however, for predicting which treatment strategy (tamoxifen alone, AI alone, or a sequence of tamoxifen and an AI) would be best for a given woman. Currently, the recommendation for initial treatment choices should be informed by the available data on efficacy, side-effect profiles, and patient preferences in postmenopausal women.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Sep 27, 2017 | Posted by in ONCOLOGY | Comments Off on Adjuvant Hormonal Therapy for Early-Stage Breast Cancer

Full access? Get Clinical Tree

Get Clinical Tree app for offline access