Elsevier

Maturitas

Volume 60, Issue 2, 20 June 2008, Pages 92-107
Maturitas

Review
Rationale for using raloxifene to prevent both osteoporosis and breast cancer in postmenopausal women

https://doi.org/10.1016/j.maturitas.2008.04.009Get rights and content

Abstract

Both osteoporosis with fracture and breast cancer are important health issues for postmenopausal women. It is well known that estrogen and estrogen receptors (ERs) play an important role in the pathogenesis of both diseases. In past decades, hormone therapy (HT), mainly estrogen plus progestin (EPT), has been frequently used for the purpose of preventing and treating postmenopausal osteoporosis because of its efficacy, but it also contributes to a significant increase in breast cancer. Currently, there is a dilemma regarding the use of estrogen for postmenopausal women. Fortunately, an increasing understanding of the action of estrogen has led ultimately to the design of new drugs that work by virtue of their interaction with the ER; these drugs have come to be known as selective estrogen receptor modulators (SERMs), and are not only effective in preventing osteoporosis and managing those with osteoporosis, but also in decreasing the incidence of breast cancer.

Among these SERMs, raloxifene may be the most attractive agent based on the evidence from five recent large trials (Multiple Outcomes of Raloxifene Evaluation [MORE], Continuing Outcomes Relevant to Evista [CORE], Raloxifene Use for the Heart [RUTH], Study of Tamoxifen and Raloxifene [STAR], and Evista Versus Alendronate [EVA]). The former three trials showed that raloxifene not only decreases the incidence of osteoporosis-associated fractures, but also has efficacy in breast cancer prevention. The head-to-head comparison with the anti-fracture agent alendronate (EVA trial) and the chemoprevention agent tamoxifen (STAR trial) further confirmed that raloxifene is a better choice.

We concluded that since there is an absence of a therapeutic effect on relieving climacteric symptoms and there is the presence of a potential risk of thromboembolism in the use of raloxifene, this drug can be prescribed for clear indications, such as the management of osteoporosis, the prevention of fracture, and decreasing the incidence of invasive breast cancer, with careful monitoring for thromboembolism. It is reasonable to use raloxifene as an appropriate medicine that targets climacteric symptom-free postmenopausal women because of its overall favorable risk–benefit safety profile using the global index proposed by the Women's Health Initiation (WHI).

Introduction

Menopause is a biological process that occurs as part of aging in women, and the key factor of the menopause is estrogen deficiency [1]. Estrogen and its receptors, ERα and ERβ [2], mediate the modulation of bone density [3]. Estrogen deficiency is the main cause of postmenopausal osteoporosis [4], and its occurrence after menopause leads to an increase in bone remodeling, resulting in an imbalance between bone resorption and formation. This is reflected in a decrease in bone mineral density (BMD) and an increase in fracture risk. Osteoporotic fractures lead to morbidity and mortality [5]. The incidence of osteoporosis and the associated economic burden will rise as the population ages [5]. Due to the close relationship between estrogen deficiency and osteoporosis, the use of estrogen therapy (ET) or a combination of estrogen and progestogen (EPT) after menopause, has been well accepted for decades. However, the prolonged use of either single ET or EPT (for example, in the Women's Health Initiative [WHI] trial) may be associated with a slightly, but definitely, increased risk of breast cancer [6], [7]. The weight of evidence indicates that exposure to estrogen is an important determinant of the risk of breast cancer [8]. Therefore, pure estrogens may not be a good choice in the management of postmenopausal women with osteoporosis. If agents could function like estrogen in terms of bone, but without estrogen's stimulation of the breast, they might be a better choice. Recently, selective estrogen receptor (ER) modulators (SERMs) have represented a major therapeutic advance for clinical practice [9]. Unlike estrogens, which are uniformly agonists, and anti-estrogens, which are uniformly antagonists, the SERMs exert selective agonist or antagonist effects on various estrogen target tissues [9]. The unique properties of SERMs lie in their bulky side-chain. This blocking effect in turn prevents key coregulator proteins (known as coactivators) from interacting with the receptor, and thus prevents activation [10], [11]. The mechanisms of the tissue-selective, mixed agonist–antagonist action of SERMs, although still only partly understood, are gradually becoming clearer [9]. Most of the unique pharmacology of SERMs can be explained by three interactive mechanisms: differential ER expression in a given target tissue, differential ER conformation on ligand binding, and differential expression and binding to the ER of coregulator proteins [9].

Many different types of SERMs have undergone large clinical trials. Among them, raloxifene may be the most attractive agent, because at least three large trials, including the Multiple Outcomes of Raloxifene Evaluation (MORE) [12], [13], [14], Continuing Outcomes Relevant to Evista (CORE) [15], [16], and Raloxifene Use for the Heart (RUTH) [17], have shown practical and promising results when using raloxifene for the prevention and management of postmenopausal women with osteoporosis or osteopenia. In these trials, raloxifene not only decreased the incidence of osteoporosis-associated complications, such as vertebral fractures and possible non-vertebral fractures, but also offered benefits for breast cancer prevention, with a dramatic decrease in the incidence of all breast cancers. In addition, two other trials have demonstrated raloxifene to be a potential candidate and choice for menopausal women: the Study of Tamoxifen and Raloxifene (STAR) [18], [19], and Evista Versus Alendronate (EVA) [5]. The STAR trial further confirmed the efficacy of raloxifene in preventing breast cancer, and showed it to be similar to the well-known anti-breast cancer drug tamoxifen. The EVA trial used the frequently prescribed anti-osteoporosis drug alendronate as a comparison, and the results showed equal efficacy in preventing osteoporosis-related fractures. These definitive clinical studies have highlighted the opportunities for innovation in the selective modulation of estrogen target tissues, especially raloxifene for the prevention and treatment of estrogen deficiency-related osteoporosis and possibly for estrogen-related breast cancers.

Since raloxifene has shown the above benefits, the aims of this report will be to offer data to support the rationale for using raloxifene in postmenopausal women, especially in a given population. We will review the emerging evidence of the efficacy of raloxifene in relation to both major health problems – osteoporosis and breast cancer – in postmenopausal women, summarize the results, and place in perspective their therapeutic uses for women having either a high risk of osteoporosis or a high risk of breast cancer.

Section snippets

MORE, CORE, and RUTH studies

At least three large clinical trials have studied the value, benefits, and risks of raloxifene in the management of postmenopausal women. The first one was the MORE study, a multicenter, randomized, double-blind trial, in which women taking raloxifene at 60 or 120 mg/day (5129 women) or a placebo (2576 women) were followed up [12], [13], [14]. A total of 7705 postmenopausal women (mainly in the United States and Europe), younger than 81 years (mean age, 66.5), and with osteoporosis as defined by

The anti-fracture efficacy of raloxifene: bone markers and BMD

As shown above, raloxifene was effective in preventing postmenopausal bone loss over a 3-year period in the MORE trial [12]. The BMD gains after 3 years were 2.1% in the spine and 2.6% in the femur. Concomitantly, significant decreases were noted for osteocalcin (−26.3% versus −8.6% in the placebo group) and urinary cross-linked N-telopeptides of type I collagen (NTX) (−34% versus −8.1% in the placebo group). BMD gains after 4 years were 2.6% in the spine and 2.1% in the femur. BMD increases

The anti-fracture efficacy of raloxifene: vertebral fracture and non-vertebral fracture

The anti-fracture efficacy of raloxifene has also been well established by the MORE trial [12], [14]. Raloxifene was efficacious (with a vertebral fracture reduction of 30% in women with and 55% in women without prevalent fractures over 3 years) [12], sustainable (with a 50% reduction in the fourth year versus a 55% reduction in years 0–3) [14], fast-acting (with 68% reduction, p = 0.01, in a 1-year post hoc analysis, and 90% reduction, p = 0.01, in a 6-month post hoc analysis) [28], [29], and very

Raloxifene and the prevention of breast cancer (Table 3)

An increase in mammographic density should be regarded as an unwanted side effect of HT, because increased breast density can impair the interpretation of mammograms, thus increasing the failure rate of breast cancer screening programs [35]. In fact, the WHI report showed that combination HT did indeed increase the risk of the incidence of breast cancers. Therefore, since raloxifene can reduce mammographic breast density [36] and breast cancer proliferative indices [37], it may play a

Discussion

Although the anti-osteoporosis and anti-osteoporosis-related fracture efficacy of raloxifene has been proven in large clinical trials, such as the MORE, CORE, and RUTH studies, one may question why there are so many anti-osteoporosis-related fracture agents currently available for prevention and/or treatment of postmenopausal osteoporosis. Bisphosphonates (alendronate, etidronate, pamidronate, zoledronic acid, risedronate, and ibandronate), calcitonin, calcium, vitamin D, estrogen, parathyroid

Areas of potential development

Since the publication of the WHI [6], the prescription of hormone therapy (HT) in the management of menopausal women has become controversial. Major efforts have been made to identify alternatives to hormone therapy. Compounds suggested have included SERMs, which represent a class of a growing number of compounds that act as either estrogen receptor agonists or antagonists in a tissue-specific manner. This pharmacological profile may offer the opportunity to dissociate favorable estrogenic

Conclusion

Based on these observations (the MORE, CORE, and RUTH trials), the efficacy of raloxifene in the prevention and management of osteoporosis, the decreased incidence of fracture, and the decreased incidence of invasive breast cancer has been proved. The potential risk of thromboembolism has also been emphasized. A recent meta-analysis to evaluate the effect of raloxifene on the risk of deep vein thrombosis (DVT) and pulmonary embolism (PE) showed that therapy with raloxifene was associated with a

Acknowledgements

This work was supported in part by grants from the Taipei Veterans General Hospital – National Taiwan University Hospital Joint Research Program (96VN-008 and 97VN-012), and the National Science Council (NSC-96-2629-B-010-001 and NSC-96-2314-B-010-018-MY3), Taiwan.

References (72)

  • C.J. Wu et al.

    Secretory endometrial adenocarcinoma in a tamoxifen user with breast cancer after menopause

    Taiwan J Obstet Gynecol

    (2007)
  • W.L. Lee et al.

    The role of selective estrogen receptor modulator on breast cancer—from tamoxifen to raloxifene

    Taiwan J Obstet Gynecol

    (2008)
  • P.D. Stein et al.

    Pulmonary thromboembolism in Asians/Pacific Islanders in the United States: analysis of data from the National Hospital Discharge Survey and the United States Bureau of the Census

    Am J Med

    (2004)
  • A.L. Klatsky et al.

    What protects Asians from venous thromboembolism?

    Am J Med

    (2004)
  • R.S. Blumenthal et al.

    Cardiovascular effects of raloxifene: the arterial and venous systems

    Am Heart J

    (2004)
  • A. Pines

    Postmenopausal hormone therapy: the way ahead

    Maturitas

    (2007)
  • J.F. Couse et al.

    Estrogen receptor null mice: what have we learned and where will they lead us?

    Endocr Rev

    (1999)
  • B.L. Riggs et al.

    Sex steroids and the construction and conservation of the adult skeleton

    Endocr Rev

    (2002)
  • R.R. Recker et al.

    Comparative effects of raloxifene and alendronate on fracture outcomes in postmenopausal women with low bone mass

    Bone

    (2007)
  • Risks and benefits of estrogen plus progestin in healthy postmenopausal women

    J Am Med Assoc

    (2002)
  • V. Beral

    Million Women Study Collaborators. Breast cancer and hormone-replacement therapy in the Million Women Study

    Lancet

    (2003)
  • J.D. Yager et al.

    Estrogen carcinogenesis in breast cancer

    N Engl J Med

    (2006)
  • B.L. Riggs et al.

    Selective estrogen receptor modulators—mechanism of actions and application to clinical practice

    N Engl J Med

    (2003)
  • V.C. Jordan

    SERMs: meeting the promise of multifunctional medicines

    J Natl Cancer Inst

    (2007)
  • V.C. Jordan

    Chemoprevention of breast cancer with selective estrogen-receptor modulators

    Nat Rev Cancer

    (2007)
  • B. Ettinger et al.

    Reduction of vertebral fracture risk in postmenopausal women with osteoporosis treated with raloxifene: results from a 3-year randomized clinical trial

    J Am Med Assoc

    (1999)
  • S.R. Cummings et al.

    The effect of raloxifene on risk of breast cancer in postmenopausal women: results from the MORE randomized trial. Multiple Outcomes of Raloxifene Evaluation

    J Am Med Assoc

    (1999)
  • P.D. Delmas et al.

    Efficacy of raloxifene on vertebral fracture risk reduction in postmenopausal women with osteoporosis: 4-year results from a randomized clinical trial

    J Clin Endocrinol Metab

    (2002)
  • S. Martino et al.

    Continuing Outcomes Relevant to Evista: breast cancer incidence in postmenopausal osteoporotic women in a randomized trial of raloxifene

    J Natl Cancer Inst

    (2004)
  • E.S. Siris et al.

    Skeletal effects of raloxifene after 8 years: results from the Continuing Outcomes Relevant to Evista (CORE) study

    J Bone Miner Res

    (2005)
  • E. Barrett-Connor

    Raloxifene Use for The Heart (RUTH) Trial Investigators. Effects of raloxifene on cardiovascular events and breast cancer in postmenopausal women

    N Engl J Med

    (2006)
  • V.G. Vogel et al.

    National Surgical Adjuvant Breast and Bowel Project (NSABP). Effects of tamoxifen vs. raloxifene on the risk of developing invasive breast cancer and other disease outcomes

    J Am Med Assoc

    (2006)
  • S.R. Land et al.

    Patient-reported symptoms and quality of life during treatment with tamoxifen or raloxifene for breast cancer prevention: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial

    J Am Med Assoc

    (2006)
  • J.A. Cauley et al.

    Continued breast cancer risk reduction in postmenopausal women treated with raloxifene: 4-year results from the MORE trial. Multiple outcomes of raloxifene evaluation

    Breast Cancer Res Treat

    (2001)
  • E. Barrett-Connor et al.

    Coronary heart disease in women, randomized clinical trials, HERS and RUTH

    Maturitas

    (1998)
  • E.E. Jolly et al.

    Prevention of osteoporosis and uterine effects in postmenopausal women taking raloxifene for 5 years

    Menopause

    (2003)
  • Cited by (0)

    View full text