Evidence Summary

Breast Cancer: Medication Use to Reduce Risk

September 03, 2019

Recommendations made by the USPSTF are independent of the U.S. government. They should not be construed as an official position of the Agency for Healthcare Research and Quality or the U.S. Department of Health and Human Services.

By Heidi D. Nelson, MD, MPH, MACP, FRCP; Rongwei Fu, PhD; Bernadette Zakher, MBBS, MPH; Miranda Pappas, MA; Marian McDonagh, PharmD

The information in this article is intended to help clinicians, employers, policymakers, and others make informed decisions about the provision of health care services. This article is intended as a reference and not as a substitute for clinical judgment.

This article may be used, in whole or in part, as the basis for the development of clinical practice guidelines and other quality enhancement tools, or as a basis for reimbursement and coverage policies. AHRQ or U.S. Department of Health and Human Services endorsement of such derivative products may not be stated or implied.

This article was published in JAMA on September 3, 2019 (JAMA. 2019;322(9):868-886. doi:10.1001/jama.2019.5780)

Return to Table of Contents

Importance: Medications to reduce risk of breast cancer are effective for women at increased risk but also cause adverse effects.

Objective: To update the 2013 US Preventive Services Task Force systematic review on medications to reduce risk of primary (first diagnosis) invasive breast cancer in women.

Data Sources: Cochrane Central Register of Controlled Trials and Database of Systematic Reviews, EMBASE, and MEDLINE (January 1, 2013, to February 1, 2019); manual review of reference lists.

Study Selection: Discriminatory accuracy studies of breast cancer risk assessment methods; randomized clinical trials of tamoxifen, raloxifene, and aromatase inhibitors for primary breast cancer prevention; studies of medication adverse effects.

Data Extraction and Synthesis: Investigators abstracted data on methods, participant characteristics, eligibility criteria, outcome ascertainment, and follow-up. Results of individual trials were combined by using a profile likelihood random-effects model.

Main Outcomes and Measures: Probability of breast cancer in individuals (area under the receiver operating characteristic curve [AUC]); incidence of breast cancer, fractures, thromboembolic events, coronary heart disease events, stroke, endometrial cancer, and cataracts; and mortality.

Results: A total of 46 studies (82 articles [>5 million participants]) were included. Eighteen risk assessment methods in 25 studies reported low accuracy in predicting the probability of breast cancer in individuals (AUC, 0.55-0.65). In placebo-controlled trials, tamoxifen (risk ratio [RR], 0.69 [95% CI, 0.59-0.84]; 4 trials [n = 28,421]), raloxifene (RR, 0.44 [95% CI, 0.24-0.80]; 2 trials [n = 17,806]), and the aromatase inhibitors exemestane and anastrozole (RR, 0.45 [95% CI, 0.26-0.70]; 2 trials [n = 8424]) were associated with a lower incidence of invasive breast cancer. Risk for invasive breast cancer was higher for raloxifene than tamoxifen in 1 trial after long-term follow-up (RR, 1.24 [95% CI, 1.05-1.47]; n = 19,747). Raloxifene was associated with lower risk for vertebral fractures (RR, 0.61 [95% CI, 0.53-0.73]; 2 trials [n = 16 929]) and tamoxifen was associated with lower risk for nonvertebral fractures (RR, 0.66 [95% CI, 0.45-0.98]; 1 trial [n = 13,388]) compared with placebo. Tamoxifen and raloxifene were associated with increased thromboembolic events compared with placebo; tamoxifen was associated with more events than raloxifene. Tamoxifen was associated with higher risk of endometrial cancer and cataracts compared with placebo. Symptomatic effects (eg, vasomotor, musculoskeletal) varied by medication.

Conclusions and Relevance: Tamoxifen, raloxifene, and aromatase inhibitors were associated with lower risk of primary invasive breast cancer in women but also were associated with adverse effects that differed between medications. Risk stratification methods to identify patients with increased breast cancer risk demonstrated low accuracy.

Return to Table of Contents

Although periodic mammography screening is currently the main approach to early detection of primary breast cancer,1 medications to reduce risk of breast cancer provide an additional prevention option for women at increased risk.2 Clinical trials indicate that the selective estrogen receptor modulators raloxifene and tamoxifen3 and the aromatase inhibitors anastrozole4 and exemestene5,6 are associated with reduced incidence of primary invasive breast cancer. However, these medications are also associated with adverse effects,3 and candidates for risk-reducing medications need to be accurately identified to optimize potential benefits and minimize harms.

In 2013, the US Preventive Services Task Force (USPSTF) issued a B recommendation for clinicians to offer to prescribe risk-reducing medications for women at increased risk for breast cancer and low risk for adverse medication effects.2 However, use of medications for breast cancer risk reduction is low in clinical practice7-9 because of women’s concerns about adverse effects and beliefs that benefits are not worth the harms,3 difficulty in identifying candidates for therapy, and primary care physicians’ unfamiliarity with tamoxifen and aromatase inhibitors because they are predominantly used for breast cancer treatment.10 This review was conducted to update evidence on the efficacy and harms of risk-reducing medications and the accuracy of clinical risk assessment methods to select candidates for therapy to inform new USPSTF recommendations.

Return to Table of Contents

Scope of Review

Detailed methods are available in the full evidence report at https://www.uspreventiveservicestaskforce.org/uspstf/recommendation/breast-cancer-medications-for-risk-reduction.11 Figure 1 shows the analytic framework, key questions (KQs), and contextual questions that guided the review. Contextual questions provide additional information for the USPSTF but are not systematically reviewed or represented in the analytic framework.

Data Sources and Searches

Cochrane Central Register of Controlled Trials and Database of Systematic Reviews, Ovid EMBASE, and MEDLINE (January 1, 2013, to February 1, 2019) were searched for relevant English-language articles, and reference lists were manually reviewed. Studies published before 2013 were identified from prior systematic reviews for the USPSTF.3,13

Study Selection

Investigators reviewed abstracts and full-text articles using prespecified eligibility criteria.11 A second reviewer independently confirmed results of the initial review; discrepancies were resolved by consensus.

Studies reporting discriminatory accuracy (ie, area under the receiver operating characteristic curve [AUC]) of risk assessment methods that could be used in primary care settings to identify women at higher than average risk for breast cancer were included for KQ1. For KQ2 and KQ4 (efficacy), only double-blind, placebo-controlled or head-to-head randomized clinical trials (RCTs) tamoxifen, raloxifene, or aromatase inhibitors for primary prevention of breast cancer that enrolled women without preexisting breast cancer and reported breast cancer incidence as a primary or secondary outcome were included. For KQ3 and KQ4 (harms), RCTs and observational studies of these medications that had nonuser comparison groups or direct comparisons between the medications were included. All adverse outcomes at all reported follow-up times were considered in an effort to capture potential short- and long-term adverse effects. Studies reporting only intermediate outcomes rather than health outcomes, such as bone density rather than fractures, were not included.

Data Extraction and Quality Assessment

Investigators abstracted data from studies of risk assessment methods (study design; population characteristics; eligibility criteria; reference standards; risk factors included in the models; and performance measures) and trials of medications (study design; setting; population characteristics; eligibility criteria; interventions [dose and duration]; numbers enrolled and lost to follow-up; method of outcome ascertainment; and results for each outcome). A second investigator reviewed accuracy of abstracted data.

Two investigators independently applied criteria developed by the USPSTF12 to rate the quality of each study as good, fair, or poor. Discrepancies were resolved through a consensus process. 

Data Synthesis and Analysis

For all KQs, the overall quality of evidence was rated good, fair, or poor, based on study quality, consistency of results, precision of estimates, study limitations, risk of reporting bias, and applicability and was summarized in a table.12

Statistical Meta-Analysis

Results of placebo-controlled trials were combined for each medication separately using meta-analysis that considered clinical and methodological differences. Estimates of risk ratios (RRs [rate ratio, hazard ratio, or relative risk]) and their standard errors were abstracted or calculated from each study and used as effect measures. Statistical heterogeneity was assessed using Cochrane χ2 tests and the magnitude of heterogeneity using the I2 statistic.14 The RRs were combined by using a profile likelihood random-effects model to account for variation among studies.15 All analyses were performed using Stata version 13.1 (StataCorp).

To determine outcomes for subgroups, analysis was performed by age (≤50 years; >50 years), family history of breast cancer (yes; no), use of menopausal hormone therapy (yes; no), menopausal status (pre; post), and body mass index (≤25; >25 [calculated as weight in kilograms divided by height in meters squared]), when at least 2 studies reported results. For outcomes of major adverse events, analyses were stratified by active vs posttreatment periods, although this was possible for tamoxifen only.

Estimating Number of Events Reduced or Increased

To interpret the clinical effect of medications, the numbers of events reduced for benefits or increased for harms compared with placebo per 1000 women, assuming 5 years of medication use, were estimated when the meta-analysis indicated a significant difference between treatment and placebo groups. Estimates used combined RRs from the meta-analyses and combined event rates from placebo groups of included trials. Combined event rates were determined from a meta-analysis of placebo event rates from each trial by using a random effects Poisson model and raw data of the number of events and women-years of follow-up. This analysis was performed using PROC NLMIXED in SAS version 9.4 (SAS Institute Inc). The 95% CIs were estimated using a simulation method that assumed that the logs of both the RRs and the event rates have normal distributions and then drew 10,000 random samples from the distribution. The numbers of events reduced or increased were then estimated from each sample; 95% CIs were obtained by computing the 2.5% and 97.5% quantiles of the full sample.

Return to Table of Contents

A total of 46 studies4,16-60 (82 articles [>5 million participants]) met inclusion criteria for KQs (Figure 2), including 25 studies of discriminatory accuracy and 21 studies of the efficacy and harms of medications (20 RCTs; 1 observational study). In addition, 14 studies addressed the contextual question regarding clinician and patient attitudes and practices.7,61-73

Accuracy of Breast Cancer Risk Assessment Methods

Key Question 1. In adult women without preexisting breast cancer, what is the accuracy of risk assessment methods to identify women who could benefit from medications to reduce risk for primary breast cancer?

A total of 25 studies reporting results of evaluations of 18 risk assessment methods based on data from more than 5 million women met inclusion criteria (Table 1).16-40 Although most methods shared common risk factors, they differed by including additional variables and using dissimilar reference populations. Three new studies expanded existing methods with new data by adding breast density to the Gail and Tyrer-Cuzick models;21 modifying the Gail model for Asian Americans;31 and adding benign breast disease to the Breast Cancer Surveillance Consortium (BCSC) model.37 A fourth new study developed models to predict estrogen receptor–positive and estrogen receptor–negative breast cancer.30 No studies evaluated the optimal age or frequency of risk assessment (KQ1a and KQ1b).

Studies reported AUC values from 0.55 to 0.65, indicating low accuracy in predicting incidence of breast cancer in individual women.16-39 Only 1 study reported AUC values above 0.70 for both the Gail-2 model (AUC, 0.74 [95% CI, 0.67-0.80]) and the Tyrer-Cuzick model (AUC, 0.76 [95% CI, 0.70-0.82]).17 However, this study was small and did not include a primary care population, limiting its clinical applicability. One study determined how well the BCSC-Tice model stratified women into low vs high-risk groups based on the risk threshold used in the National Surgical Adjuvant Breast and Bowel Project (NSABP P-1) and the Study of Tamoxifen and Raloxifene (STAR) trials (≥1.66% 5-year breast cancer risk).36 Results indicated AUC values from 0.61 to 0.64.36

Benefits of Risk-Reducing Medications

Key Question 2. In adult women without preexisting breast cancer, what is the effectiveness and comparative effectiveness of medications to reduce risk for primary breast cancer on improvement in short- and long-term health outcomes, including invasive breast cancer, noninvasive breast cancer (including ductal carcinoma in situ), breast cancer mortality, all-cause mortality, and other beneficial outcomes (such as reduced fractures caused by certain medications and improved quality of life)?

Ten RCTs (40 articles) provided results for KQ2, including 7 new publications. These included updated long-term results of the International Breast Cancer Intervention Study (IBIS-I) trial of tamoxifen,74 a placebo-controlled trial of low-dose tamoxifen,41 and placebo-controlled trials of anastrozole4,75,76 and exemestane.44,77.

Trials include the STAR head-to-head trial of tamoxifen and raloxifene;49,78,79 5 placebo-controlled trials of tamoxifen, including IBIS-I,42,74,80 NSABP P-1,46,81,82 Royal Marsden Hospital Trial,47,83 Italian Tamoxifen Prevention Study,43,84-87 and the Hormone Replacement Therapy Opposed by Low-dose Tamoxifen (HOT) study;41 2 placebo-controlled trials of raloxifene, the Multiple Outcomes of Raloxifene Evaluation (MORE) with long-term follow-up in the Continuing Outcomes Relevant to Evista (CORE) study45,88-102 and the Raloxifene Use for the Heart (RUTH) trial;48,103 and 2 placebo-controlled trials of aromatase inhibitors, the International Breast Cancer Intervention Study II (IBIS-II) of anastrozole4,75,76 and the Mammary Prevention.3 trial (MAP.3) of exemestane.44,77 The most recent placebo-controlled tamoxifen trial, HOT, using a lower dose than the other trials (5 mg/d vs 20 mg/d), did not indicate reduction in invasive breast cancer risk (RR, 0.83 [95% CI, 0.42-1.62]; n = 1884) and was not included in the meta-analyses of tamoxifen trials.41 Details of individual trials are provided in Table 2 and the full report.11 Trials met criteria for fair or good quality.

Trials included large numbers of women, ranging in size from 188441 to 19,747,49 primarily from North America, Europe, and the United Kingdom. Most participants were white, and none of the trials provided outcomes specific to racial or ethnic groups. Participants ranged in age from 30s to 80s at baseline, and only the placebo-controlled tamoxifen trials enrolled premenopausal women. The Italian trial of tamoxifen exclusively enrolled women who had undergone prior hysterectomy, including some with oophorectomy.43 Participants used exogenous estrogen in the Italian (14% of women), Royal Marsden (15%-27%), IBIS-I (40%), and HOT (100%) tamoxifen trials. The raloxifene trials enrolled older women with osteoporosis45,88-102 or increased cardiovascular risk.45,88-102

Results of the meta-analysis for KQ2 are summarized in Table 3. In placebo-controlled trials, tamoxifen (RR, 0.69 [95% CI, 0.59-0.84]; 7 fewer cases per 1000 women over 5 years of use [95% CI, 4-12]; 4 trials [n = 28,421]), raloxifene (RR, 0.44 [95% CI, 0.24-0.80]; 9 fewer cases [95% CI, 3-15]; 2 trials [n = 17,806]), and the aromatase inhibitors exemestane and anastrozole (RR, 0.45 [95% CI, 0.26-0.70]; 16 fewer cases [95% CI, 8-24]; 2 trials [n = 8424]) (Figure 3) were associated with reduced invasive breast cancer. Risk for invasive breast cancer was higher for raloxifene than tamoxifen in the STAR head-to-head trial (RR, 1.24 [95% CI, 1.05-1.47]; n = 19,747) after long-term follow-up. All medications were associated with reduced estrogen receptor–positive but not estrogen receptor–negative invasive breast cancer. Tamoxifen was associated with reduced noninvasive cancer in the NSABP P-182 and IBIS-I74 trials but not in the meta-analysis of all 4 trials (RR, 0.72 [95% CI, 0.56-1.41]; 4 trials [n = 28,421]). Medications were not associated with reductions in breast cancer–specific and all-cause mortality.

In placebo-controlled trials, raloxifene (RR, 0.61 [95% CI, 0.53-0.73]; 2 trials [n = 16,929]) was associated with reduced vertebral fractures; tamoxifen was associated with reduced nonvertebral fractures in the NSABP P-1 trial (RR, 0.66 [95% CI, 0.45-0.98]; n = 13,388) and the aromatase inhibitors had no effect on fractures. Tamoxifen and raloxifene had similar effects on fracture incidence at multiple vertebral and nonvertebral sites in the STAR head-to-head trial.

Key Question 2a. Does the effectiveness of risk-reducing medications vary by timing of initiation or duration of use?

Eight trials reported similar breast cancer outcomes regardless of age, although age categories varied by trial. No studies specifically compared shorter vs longer regimens of medication use or initiation based on time since menopause. While most trials intended 5 years of medication use, mean exposure times varied across the trials from 3 to 5 years. However, trials of similar medications indicated general consistency in their associations with breast cancer risk reduction, despite exposure time.

Key Question 2b. Does the effectiveness of risk-reducing medications persist beyond discontinuation of use?

The IBIS-I80 and Royal Marsden83 trials provided results for invasive and estrogen receptor–positive breast cancer for both the active treatment (mean duration, 5 years) and the posttreatment (median follow-up, 13 and 16 years, respectively) periods. These results indicate continued associations with reduced risk after discontinuation of tamoxifen, providing point estimates of even larger reductions in invasive and estrogen receptor–positive breast cancer during the posttreatment period. For IBIS-I, the RR for invasive breast cancer was 0.74 (95% CI, 0.60-0.93) for the 0- to 10-year follow-up period and 0.70 (95% CI, 0.52-0.95) for the greater than 10 years follow-up period,4 although the difference between periods was not statistically significant.

Harms of Risk-Reducing Medications

Key Question 3. What are the harms of using medications to reduce risk for primary breast cancer?

A total of 22 studies (54 articles) met inclusion criteria, including updated long-term results of the IBIS-I trial of tamoxifen,74 a placebo-controlled trial of low-dose tamoxifen,41 and placebo-controlled trials of anastrozole4,75,76 and exemestane.44,77

For tamoxifen, information on adverse effects was confined to the 5 large placebo-controlled primary prevention trials41-43,46,47,74,80-84,86,87,105-108 and the STAR head-to-head trial.49,78,79,109 The HOT trial of low-dose tamoxifen indicated no statistically significant differences in outcomes compared with placebo and was not included in the meta-analyses of tamoxifen trials.41 For raloxifene, adverse effects were reported from the 2 large placebo-controlled trials, MORE/CORE and RUTH;88-94,104 the STAR head-to-head trial;49,78,79 8 smaller trials (in 11 publications) evaluating either bone density, biochemical profiles, or fractures;51-60,110 and 1 observational study.50 These additional studies contribute little to the evaluation of harms because they involve few women relative to the large primary prevention trials, although these results are generally consistent with those of the larger trials. Consequently, they were not included in the meta-analyses of raloxifene trials. For anastrozole and exemestane, information on adverse effects was based on the 2 large placebo-controlled primary prevention trials.4,44 Similar to tamoxifen, no other RCTs or observational studies evaluated adverse effects of aromatase inhibitors in women without breast cancer.

Results of the meta-analysis for KQ3 are summarized in Table 3. In placebo-controlled trials, tamoxifen (RR, 1.93 [95% CI, 1.33-2.68]; 4 trials [n = 28,421]) and raloxifene (RR, 1.56 [95% CI, 1.11-2.60]; 2 trials [n = 17,806]) were associated with increased thromboembolic events. Raloxifene was associated with fewer thromboembolic events than tamoxifen in the STAR head-to-head trial (RR, 0.75 [95% CI, 0.60-0.93]; n = 19,490). Tamoxifen, raloxifene, and aromatase inhibitors were not associated with increased coronary heart disease events or strokes.

In placebo-controlled trials, tamoxifen was associated with increased incidence of endometrial cancer (RR, 2.25 [95% CI, 1.17-4.41]; 3 trials [n = 15,421]). In the STAR head-to-head trial, raloxifene was associated with fewer cases of endometrial cancer (RR, 0.55 [95% CI, 0.36-0.83]; n = 19,490) and endometrial hyperplasia (RR,0.19 [95% CI,0.12-0.29]; n = 19,490) and with fewer hysterectomies (RR, 0.45 [95% CI, 0.37-0.54]; n = 19,490) than tamoxifen. Tamoxifen was associated with increased incidence of cataracts (RR, 1.22 [95% CI, 1.08-1.48]; 3 trials [n = 22,832]) and cataract surgery compared with placebo. Risks for thromboembolic events and endometrial cancer with tamoxifen were higher for older compared with younger women and returned to normal after discontinuation. All medications were associated with adverse effects, such as vasomotor or musculoskeletal symptoms,that varied by medication.

Key Question 3a. Do the harms of risk-reducing medications vary by timing of initiation or duration of use?

The NSABP P-1 placebo-controlled trial of tamoxifen reported point estimates consistent with higher risks for deep vein thrombosis, pulmonary embolus, and stroke for women 50 years and older than for women younger than 50 years, although results were not statistically significant.46 Results of the NSABP P-1 trial also indicated that the risk of thromboembolic events was elevated only during the first 3 years of tamoxifen use.111 Age older than 60 years was also an important risk factor for venous thrombosis in the Italian trial.87 The NSABP P-1 trial found that endometrial cancer was more common among women 50 years and older than among women younger than 50 years (RR, 4.01 [95% CI, 1.70-10.90]; n = 7998 for those ≥50 years vs RR, 1.21 [95% CI, 0.41-3.60]; n = 5177 for those <50 years).46 Initiation based on time since menopause was not reported.

Key Question 3b. Do the harms of risk-reducing medications persist beyond discontinuation of use?

Although tamoxifen was associated with increased thromboembolic events compared with placebo during the trials, risk returned to normal after discontinuation of tamoxifen in the 2 trials (IBIS-I80 and Royal Marsden83) that reported post-treatment data (RR,0.98 [95% CI, 0.48-1.80]; 2 trials; n = 10,130).74 In the IBIS-I trial, risk for endometrial cancer was higher for tamoxifen compared with placebo during the first 5 years of follow-up (RR, 3.76 [95% CI, 1.20-15.56]) but declined after discontinuation (RR for 5- to 10-year followup, 0.64 [95% CI, 0.21-1.80]; RR for ≥10-year follow-up, 1.40 [95% CI, 0.38-5.61]).74

Outcomes in Subgroups

Key Question 4. Do the outcomes of using medications to reduce risk for primary breast cancer vary by population subgroups?

Studies included for KQ2 and KQ3 also provided results for KQ4. Medications were associated with lower risks for invasive breast cancer in all population subgroups evaluated based on menopausal status; family history of breast cancer; body mass index categories; modified Gail model risk categories; and age at menarche, parity, or age at first live birth. Tamoxifen and anastrozole were associated with reduced risk, regardless of history of previous breast lesions (lobular carcinoma in situ, atypical ductal hyperplasia, or atypical lobular hyperplasia) but demonstrated larger estimates of effect in women with previous lesions.

Clinician and Patient Attitudes and Practices

Factors associated with adherence and nonadherence in patient use of risk-reducing medications were examined in systematic reviews7,69,72 and additional observational studies.63,64,66-68,70,71,73 Factors associated with adherence included higher breast cancer risk; clinician recommendation; peers with good experiences using medications; belief that medications are effective; anxiety or worry about breast cancer; and history of an abnormal breast biopsy result. Factors associated with nonadherence included concern for adverse effects; estrogen contraindication; peers with poor experiences using medications; belief that medications are for treatment, not risk reduction; medication is a daily reminder of illness; preference for other risk-reducing approaches such as mastectomy; and knowledge of the benefits and harms of medication.

Prescribing risk-reducing medications is an uncommon practice among primary care physicians surveyed in 3 US studies.61,62,65 Factors associated with prescribing included more breast cancer diagnoses in clinical practice; belief that benefits outweigh harms; patients asking about medications; personal experience with breast cancer in self or a relative; and belief that eligibility for medications is easy to determine. Barriers to prescribing included lack of training, experience, or comfort with medications; belief that benefit may not be worth harms; belief that patients lack interest in medications; preference that specialists prescribe medications; lack of comfort or certainty with identifying women eligible for medications; and time constraints.

Return to Table of Contents

This evidence report reviewed trials of the efficacy and harms of medications to reduce the risk of primary invasive breast cancer and studies of the accuracy of clinical risk assessment methods to select patients for therapy. Table 4 summarizes the evidence included in this review. Although most results are consistent with the 2013 USPSTF review,3 this update provides additional evidence of the inaccuracy of risk assessment methods;21,30,31,37 long-term follow-up of the IBIS-1 tamoxifen trial demonstrating persistent breast cancer risk reduction and normalization of endometrial cancer risk after discontinuation of tamoxifen;4 and new trials of aromatase inhibitors.4,44,75-77 In addition, a placebo-controlled trial of low-dose tamoxifen indicated no reduction in risk of invasive breast cancer.41

Results of 4 recently published studies of breast cancer risk assessment methods indicated low discriminatory accuracy in predicting the probability of breast cancer in individual women,21,30,31,37 similar to previous studies. Most methods performed only slightly better than age alone as a risk predictor. Based on these studies, current practices of selecting women for risk-reducing medications according to a modified 5-year Gail score of 1.66% or higher, as used for inclusion criteria in primary prevention trials and US Food and Drug Administration approval of tamoxifen and raloxifene for risk reduction, are likely inaccurate. Most women 60 years and older without other risk factors would meet this threshold by age alone. Studies also provide no clinical guidance on optimal ages or frequencies for risk assessment because these components have not yet been evaluated.

Primary prevention trials of anastrozole4,75,76 and exemestane44,77 provide new evidence of the efficacy and harms of aromatase inhibitors for breast cancer risk reduction. However, no long-term follow-up data are available to determine whether harms demonstrated in treatment trials of women with noninvasive and early stage breast cancer, such as fractures and cardiovascular events, apply to risk reduction. An RCT of 2980 women with locally excised estrogen receptor–positive ductal carcinoma in situ compared anastrozole (1 mg/d) with tamoxifen (20 mg/d) for 5 years, with median follow-up of 7.2 years.112 Results indicated increased risk of fractures (odds ratio, 1.36 [95% CI, 1.03-1.80]) and stroke (odds ratio, 3.36 [95% CI, 1.04-14.18]) with anastrozole and increased venous thromboembolic events with tamoxifen.112 A meta-analysis of individual-level data from 31,920 postmenopausal women with estrogen receptor–positive early breast cancer in treatment RCTs of aromatase inhibitors vs tamoxifen also indicated associations with increased fractures for aromatase inhibitors but no differences for venous thromboembolic events or stroke.113 Also, 7 RCTs that compared extended aromatase inhibitor treatment with treatment followed by placebo or no treatment showed associations with increased fractures and stroke for extended aromatase inhibitors and suggested increased cardiovascular events.114 Although these trials imply associations of aromatase inhibitors with increased risk for fractures and stroke, it is unclear how well the results of treatment trials translate to women without cancer, particularly in the absence of true placebo comparison groups. For example, it is not known whether the increase in fractures reflects the direct harm of aromatase inhibitors or the protective effect of tamoxifen.

Future research to determine optimal candidates for risk-reducing medications should focus on the women mostly likely to benefit. Applying research findings to clinical selection criteria would improve identification of candidates in practice settings and clinical decision making. For example, no new studies and no studies in the 2013 review evaluated risk-reducing medications specifically in carriers of pathogenic BRCA1/2 mutations. Mutation testing was not a common practice when most of the trials were conducted, and it is not known how many BRCA1/2 carriers were enrolled. The NSABP P-1 trial of tamoxifen described results for 288 mutation carriers who developed breast cancer during the trial.115 Of the 8 women with breast cancer who had BRCA1 mutations, 5 received tamoxifen and 3 placebo (RR, 1.67 [95% CI, 0.32-10.70]). Of 11 women with breast cancer and BRCA2 mutations, 3 received tamoxifen and 8 placebo (RR, 0.38 [95% CI, 0.06-1.56]). Also, 6 of 7 women (86%) with BRCA1 mutations had estrogen receptor–negative breast cancer and 6 of 9 (67%) with BRCA2 mutations had estrogen receptor–positive cancer. Tamoxifen is only effective in reducing risk for estrogen receptor–positive breast cancer.

Limitations

This review had several limitations. First, there was potential publication bias as well as biases of the literature review process, such as including only English-language articles. Second, studies of risk assessment methods varied by size, study populations, reference groups, and methods. Third, RCTs were limited by clinical heterogeneity related to different eligibility criteria, exposure durations and follow-up, adherence, and ascertainment of outcomes. The trials were not designed for subgroup analysis and may have been underpowered to demonstrate treatment effects. Furthermore, no trials directly compared the effects of timing and duration of medication use. Fourth, research is lacking for optimal doses, duration of use, persistence of effects after treatment for most medications, and outcomes in women who are nonwhite, premenopausal, have comorbidities, or are taking additional medications for other indications

Return to Table of Contents

Tamoxifen, raloxifene, and aromatase inhibitors were associated with lower risk of primary invasive breast cancer in women but also were associated with adverse effects that differed between medications. Risk stratification methods to identify patients with increased breast cancer risk demonstrated low accuracy.

Return to Table of Contents

Source: This article was first published in the Journal of the American Medical Association on September 3, 2019 (JAMA. 2019;322(9):868-886. doi:10.1001/jama.2019.5780)

Conflict of Interest Disclosures: None reported.

Funding/Support: This research was funded under contract HHSA290201500009I, Task Order 7, from the Agency for Healthcare Research and Quality (AHRQ), US Department of Health and Human Services, under a contract to support the US Preventive Services Task Force (USPSTF).

Role of the Funder/Sponsor: Investigators worked with USPSTF members and AHRQ staff to develop the scope, analytic framework, and key questions for this review. AHRQ had no role in study selection, quality assessment, or synthesis. AHRQ staff provided project oversight, reviewed the report to ensure that the analysis met methodological standards, and distributed the draft for peer review. Otherwise, AHRQ had no role in the conduct of the study; collection, management, analysis, and interpretation of the data; and preparation, review, or approval of the manuscript findings. The opinions expressed in this document are those of the authors and do not reflect the official position of AHRQ or the US Department of Health and Human Services.

Additional Contributions: We gratefully acknowledge the following individuals for their contributions to this project: AHRQ medical officer Tina Fan, MD, MPH; and Pacific Northwest Evidence-based Practice Center expert consultant Rachel Yung, MD, research librarian Andrew Hamilton, MLS, MS, and research assistant Lucy Stillman, BS. We also acknowledge past and current USPSTF members who contributed to topic deliberations. USPSTF members, external reviewers, and federal partner reviewers did not receive financial compensation for their contribution

Additional Information: A draft version of this evidence report underwent external peer review from 4 content experts (Therese Bartholomew Bevers, MD, University of Texas MD Anderson Cancer Center, Houston; Jack Cuzick, PhD, FRS, CBE, Wolfson Institute of Preventive Medicine, Queen Mary University, London; Sam G. Smith, MSc, PhD, Leeds Institute of Health Sciences, London; and Diana Petitti, MD, MPH, University of Arizona, Tucson) and from 4 federal partners at the Centers for Disease Control and Prevention and National Cancer Institute. Comments from reviewers were presented to the USPSTF during its deliberation of the evidence and were considered in preparing the final evidence review.

Return to Table of Contents

1. US Preventive Services Task Force (USPSTF). Final recommendation statement: breast cancer screening. USPSTF website. https://www.uspreventiveservicestaskforce.org/Page/Document/RecommendationStatementFinal/breast-cancer-screening1. 2016. Accessed April 25, 2019.
2. Moyer VA; U.S. Preventive Services Task Force. Medications to decrease the risk for breast cancer in women: recommendations from the U.S. Preventive Services Task Force. Ann Intern Med. 2013;159(10):698-708.
3. Nelson HD, Smith ME, Griffin JC, Fu R. Use of medications to reduce risk for primary breast cancer: a systematic review for the U.S. Preventive Services Task Force. Ann Intern Med. 2013;158(8):604-614. doi:10.7326/0003-4819-158-8-201304160-00005
4. Cuzick J, Sestak I, Forbes JF, et al; IBIS-II Investigators. Anastrozole for prevention of breast cancer in high-risk postmenopausal women (IBIS-II): an international, double-blind, randomised placebo-controlled trial. [published correction appears in Lancet. 2014;383(9922):1040]. Lancet. 2014;383(9922):1041-1048. doi:10.1016/S0140-6736(13)62292-8
5. Richardson H, Johnston D, Pater J, Goss P. The National Cancer Institute of Canada Clinical Trials Group MAP.3 trial: an international breast cancer prevention trial. Curr Oncol. 2007;14(3):89-96. doi:10.3747/co.2007.117
6. van de Velde CJ, Rea D, Seynaeve C, et al. Adjuvant tamoxifen and exemestane in early breast cancer (TEAM): a randomised phase 3 trial. Lancet. 2011;377(9762):321-331. doi:10.1016/S0140-6736(10)62312-4
7. Smith SG, Sestak I, Forster A, et al. Factors affecting uptake and adherence to breast cancer chemoprevention: a systematic review and meta-analysis. Ann Oncol. 2016;27(4):575-590. doi:10.1093/annonc/mdv590
8. Pinsky PF, Miller E, Heckman-Stoddard B, Minasian L. Use of raloxifene and tamoxifen by breast cancer risk level in a Medicare-eligible cohort. Am J Obstet Gynecol. 2018;218(6):606.e1-606.e9. doi:10.1016/j.ajog.2018.03.031
9. Waters EA, Cronin KA, Graubard BI, Han PK, Freedman AN. Prevalence of tamoxifen use for breast cancer chemoprevention among U.S. women. Cancer Epidemiol Biomarkers Prev. 2010;19(2):443-446. doi:10.1158/1055-9965.EPI-09-0930
10. Smith SG, Foy R, McGowan JA, et al. Prescribing tamoxifen in primary care for the prevention of breast cancer: a national online survey of GPs’ attitudes. Br J Gen Pract. 2017;67(659):e414-e427. doi:10.3399/bjgp17X689377
11. Nelson HD, Fu R, Zakher B, et al. Medication Use for the Risk Reduction of Primary Breast Cancer in Women: A Systematic Review for the U.S. Preventive Services Task Force. Evidence Synthesis No. 180. AHRQ Publication No. 19-05249-EF-1. Rockville, MD: Agency for Healthcare Research and Quality; 2019.
12. US Preventive Services Task Force (USPSTF). Methods and processes. USPSTF website. https://www.uspreventiveservicestaskforce.org/Page/Name/methods-and-processes. 2018. Accessed March 27, 2019.
13. Nelson HD, Fu R, Griffin JC, Nygren P, Smith ME, Humphrey L. Systematic review: comparative effectiveness of medications to reduce risk for primary breast cancer. Ann Intern Med. 2009;151(10):703-715. doi:10.7326/0000605-200911170-00147
14. Higgins JP, Thompson SG. Quantifying heterogeneity in a meta-analysis. Stat Med. 2002;21(11):1539-1558. doi:10.1002/sim.1186
15. Hardy RJ, Thompson SG. A likelihood approach to meta-analysis with random effects. Stat Med. 1996;15(6):619-629. doi:10.1002/(SICI)1097-0258(19960330)15:6<619::AID-SIM188>3.0.CO;2-A
16. Adams-Campbell LL, Makambi KH, Palmer JR, Rosenberg L. Diagnostic accuracy of the Gail model in the Black Women’s Health Study. Breast J. 2007;13(4):332-336. doi:10.1111/j.1524-4741.2007.00439.x
17. Amir E, Evans DG, Shenton A, et al. Evaluation of breast cancer risk assessment packages in the family history evaluation and screening programme. J Med Genet. 2003;40(11):807-814. doi:10.1136/jmg.40.11.807
18. Barlow WE, White E, Ballard-Barbash R, et al. Prospective breast cancer risk prediction model for women undergoing screening mammography. J Natl Cancer Inst. 2006;98(17):1204-1214. doi:10.1093/jnci/djj331
19. Boughey JC, Hartmann LC, Anderson SS, et al. Evaluation of the Tyrer-Cuzick (International Breast Cancer Intervention Study) model for breast cancer risk prediction in women with atypical hyperplasia. J Clin Oncol. 2010;28(22):3591-3596. doi:10.1200/JCO.2010.28.0784
20. Boyle P, Mezzetti M, La Vecchia C, Franceschi S, Decarli A, Robertson C. Contribution of three components to individual cancer risk predicting breast cancer risk in Italy. Eur J Cancer Prev. 2004;13(3):183-191. doi:10.1097/01.cej.0000130014.83901.53
21. Brentnall AR, Harkness EF, Astley SM, et al. Mammographic density adds accuracy to both the Tyrer-Cuzick and Gail breast cancer risk models in a prospective UK screening cohort. Breast Cancer Res. 2015;17(1):147. doi:10.1186/s13058-015-0653-5
22. Chen J, Pee D, Ayyagari R, et al. Projecting absolute invasive breast cancer risk in white women with a model that includes mammographic density. J Natl Cancer Inst. 2006;98(17):1215-1226. doi:10.1093/jnci/djj332
23. Chlebowski RT, Anderson GL, Lane DS, et al; Women’s Health Initiative Investigators. Predicting risk of breast cancer in postmenopausal women by hormone receptor status. J Natl Cancer Inst. 2007;99(22):1695-1705. doi:10.1093/jnci/djm224
24. Colditz GA, Rosner B. Cumulative risk of breast cancer to age 70 years according to risk factor status: data from the Nurses’ Health Study. Am J Epidemiol. 2000;152(10):950-964. doi:10.1093/aje/152.10.950
25. Colditz GA, Rosner BA, Chen WY, Holmes MD, Hankinson SE. Risk factors for breast cancer according to estrogen and progesterone receptor status. J Natl Cancer Inst. 2004;96(3):218-228. doi:10.1093/jnci/djh02
26. Costantino JP, Gail MH, Pee D, et al. Validation studies for models projecting the risk of invasive and total breast cancer incidence. J Natl Cancer Inst. 1999;91(18):1541-1548. doi:10.1093/jnci/91.18.1541
27. Decarli A, Calza S, Masala G, Specchia C, Palli D, Gail MH. Gail model for prediction of absolute risk of invasive breast cancer: independent evaluation in the Florence-European Prospective Investigation Into Cancer and Nutrition Cohort. J Natl Cancer Inst. 2006;98(23):1686-1693. doi:10.1093/jnci/djj463
28. Gail MH, Anderson WF, Garcia-Closas M, Sherman ME. Absolute risk models for subtypes of breast cancer. J Natl Cancer Inst. 2007;99(22):1657-1659. doi:10.1093/jnci/djm228
29. Gail MH, Brinton LA, Byar DP, et al. Projecting individualized probabilities of developing breast cancer for white females who are being examined annually. J Natl Cancer Inst. 1989;81(24):1879-1886. doi:10.1093/jnci/81.24.1879
30. Li K, Anderson G, Viallon V, et al. Risk prediction for estrogen receptor–specific breast cancers in two large prospective cohorts. Breast Cancer Res. 2018;20(1):147. doi:10.1186/s13058-018-1073-0
31. Matsuno RK, Costantino JP, Ziegler RG, et al. Projecting individualized absolute invasive breast cancer risk in Asian and Pacific Islander American women. J Natl Cancer Inst. 2011;103(12):951-961. doi:10.1093/jnci/djr154
32. Petracci E, Decarli A, Schairer C, et al. Risk factor modification and projections of absolute breast cancer risk. J Natl Cancer Inst. 2011;103(13):1037-1048. doi:10.1093/jnci/djr172
33. Rockhill B, Byrne C, Rosner B, Louie MM, Colditz G. Breast cancer risk prediction with a log-incidence model: evaluation of accuracy. J Clin Epidemiol. 2003;56(9):856-861. doi:10.1016/S0895-4356(03)00124-0
34. Rockhill B, Spiegelman D, Byrne C, Hunter DJ, Colditz GA. Validation of the Gail et al. model of breast cancer risk prediction and implications for chemoprevention. J Natl Cancer Inst. 2001;93(5):358-366. doi:10.1093/jnci/93.5.358
35. Tamimi RM, Rosner B, Colditz GA. Evaluation of a breast cancer risk prediction model expanded to include category of prior benign breast disease lesion. Cancer. 2010;116(21):4944-4953.doi:10.1002/cncr.25386
36. Tice JA, Cummings SR, Smith-Bindman R, Ichikawa L, Barlow WE, Kerlikowske K. Using clinical factors and mammographic breast density to estimate breast cancer risk: development and validation of a new predictive model. Ann Intern Med. 2008;148(5):337-347. doi:10.7326/0003-4819-148-5-200803040-00004
37. Tice JA, Miglioretti DL, Li CS, Vachon CM, Gard CC, Kerlikowske K. Breast density and benign breast disease: risk assessment to identify women at high risk of breast cancer. J Clin Oncol. 2015;33(28):3137-3143. doi:10.1200/JCO.2015.60.8869
38. Tyrer J, Duffy SW, Cuzick J. A breast cancer prediction model incorporating familial and personal risk factors. Stat Med. 2004;23(7):1111-1130. doi:10.1002/sim.1668
39. Vacek PM, Skelly JM, Geller BM. Breast cancer risk assessment in women aged 70 and older. Breast Cancer Res Treat. 2011;130(1):291-299. doi:10.1007/s10549-011-1576-1
40. Warwick J, Birke H, Stone J, et al. Mammographic breast density refines Tyrer-Cuzick estimates of breast cancer risk in high-risk women: findings from the placebo arm of the International Breast Cancer Intervention Study I. Breast Cancer Res. 2014;16(5):451. doi:10.1186/s13058-014-0451-5
41. DeCensi A, Bonanni B, Maisonneuve P, et al; Italian HOT Study Group. A phase-III prevention trial of low-dose tamoxifen in postmenopausal hormone replacement therapy users: the HOT study. Ann Oncol. 2013;24(11):2753-2760. doi:10.1093/annonc/mdt244
42. Cuzick J, Forbes J, Edwards R, et al; IBIS Investigators. First results from the International Breast Cancer Intervention Study (IBIS-I): a randomised prevention trial. Lancet. 2002;360(9336):817-824. doi:10.1016/S0140-6736(02)09962-2
43. Veronesi U, Maisonneuve P, Costa A, et al; Italian Tamoxifen Prevention Study. Prevention of breast cancer with tamoxifen: preliminary findings from the Italian randomised trial among hysterectomised women. Lancet. 1998;352(9122):93-97. doi:10.1016/S0140-6736(98)85011-3
44. Goss PE, Ingle JN, Alés-Martínez JE, et al; NCIC CTG MAP.3 Study Investigators. Exemestane for breast-cancer prevention in postmenopausal women [published correction appears in N Engl J Med. 2011;365(14):1361].. N Engl J Med. 2011;364(25):2381-2391. doi:10.1056/NEJMoa1103507
45. Martino S, Cauley JA, Barrett-Connor E, et al; CORE Investigators. Continuing outcomes relevant to Evista: breast cancer incidence in postmenopausal osteoporotic women in a randomized trial of raloxifene. J Natl Cancer Inst. 2004;96(23):1751-1761. doi:10.1093/jnci/djh319
46. 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(18):1371-1388. doi:10.1093/jnci/90.18.1371
47. Powles T, Eeles R, Ashley S, et al. Interim analysis of the incidence of breast cancer in the Royal Marsden Hospital tamoxifen randomised chemoprevention trial. Lancet. 1998;352(9122):98-101. doi:10.1016/S0140-6736(98)85012-5
48. Barrett-Connor E, Mosca L, Collins P, et al; 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;355(2):125-137. doi:10.1056/NEJMoa062462
49. Vogel VG, Costantino JP, Wickerham DL, 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: the NSABP Study of Tamoxifen and Raloxifene (STAR) P-2 trial. JAMA. 2006;295(23):2727-2741. doi:10.1001/jama.295.23.joc60074
50. Christodoulakos GE, Botsis DS, Lambrinoudaki IV, et al. A 5-year study on the effect of hormone therapy, tibolone and raloxifene on vaginal bleeding and endometrial thickness. Maturitas. 2006;53(4):413-423. doi:10.1016/j.maturitas.2005.07.003
51. Cohen FJ, Watts S, Shah A, Akers R, Plouffe L Jr. Uterine effects of 3-year raloxifene therapy in postmenopausal women younger than age 60. Obstet Gynecol. 2000;95(1):104-110.
52. Delmas PD, Bjarnason NH, Mitlak BH, et al. Effects of raloxifene on bone mineral density, serum cholesterol concentrations, and uterine endometrium in postmenopausal women. N Engl J Med. 1997;337(23):1641-1647. doi:10.1056/NEJM199712043372301
53. Johnston CC Jr, Bjarnason NH, Cohen FJ, et al. Long-term effects of raloxifene on bone mineral density, bone turnover, and serum lipid levels in early postmenopausal women: three-year data from 2 double-blind, randomized, placebo-controlled trials. Arch Intern Med. 2000;160(22):3444-3450. doi:10.1001/archinte.160.22.3444
54. Jolly EE, Bjarnason NH, Neven P, et al. Prevention of osteoporosis and uterine effects in postmenopausal women taking raloxifene for 5 years. Menopause. 2003;10(4):337-344. doi:10.1097/01.GME.0000058772.59606.2A
55. Lufkin EG, Whitaker MD, Nickelsen T, et al. Treatment of established postmenopausal osteoporosis with raloxifene: a randomized trial. J Bone Miner Res. 1998;13(11):1747-1754. doi:10.1359/jbmr.1998.13.11.1747
56. McClung MR, Siris E, Cummings S, et al. Prevention of bone loss in postmenopausal women treated with lasofoxifene compared with raloxifene. Menopause. 2006;13(3):377-386. doi:10.1097/01.gme.0000188736.69617.4f
57. Meunier PJ, Vignot E, Garnero P, et al; Raloxifene Study Group. Treatment of postmenopausal women with osteoporosis or low bone density with raloxifene. Osteoporos Int. 1999;10(4):330-336. doi:10.1007/s001980050236
58. Morii H, Ohashi Y, Taketani Y, et al. Effect of raloxifene on bone mineral density and biochemical markers of bone turnover in Japanese postmenopausal women with osteoporosis: results from a randomized placebo-controlled trial. Osteoporos Int. 2003;14(10):793-800. doi:10.1007/s00198-003-1424-1
59. Nickelsen T, Lufkin EG, Riggs BL, Cox DA, Crook TH. Raloxifene hydrochloride, a selective estrogen receptor modulator: safety assessment of effects on cognitive function and mood in postmenopausal women. Psychoneuroendocrinology. 1999;24(1):115-128. doi:10.1016/S0306-4530(98)00041-9
60. Palacios S, Farias ML, Luebbert H, et al. Raloxifene is not associated with biologically relevant changes in hot flushes in postmenopausal women for whom therapy is appropriate. Am J Obstet Gynecol. 2004;191(1):121-131. doi:10.1016/j.ajog.2003.10.701
61. Armstrong K, Quistberg DA, Micco E, Domchek S, Guerra C. Prescription of tamoxifen for breast cancer prevention by primary care physicians. Arch Intern Med. 2006;166(20):2260-2265. doi:10.1001/archinte.166.20.2260
62. Corbelli J, Borrero S, Bonnema R, et al. Use of the Gail model and breast cancer preventive therapy among three primary care specialties. J Womens Health (Larchmt). 2014;23(9):746-752. doi:10.1089/jwh.2014.4742
63. Hackett J, Thorneloe R, Side L, et al. Uptake of breast cancer preventive therapy in the UK: results from a multicentre prospective survey and qualitative interviews. Breast Cancer Res Treat. 2018;170(3):633-640. doi:10.1007/s10549-018-4775-1
64. Holmberg C, Bandos H, Fagerlin A, et al. NRG Oncology/National Surgical Adjuvant Breast and Bowel Project Decision-Making Project-1 results: decision making in breast cancer risk reduction. Cancer Prev Res (Phila). 2017;10(11):625-634. doi:10.1158/1940-6207.CAPR-17-0076
65. Kaplan CP, Haas JS, Pérez-Stable EJ, Des Jarlais G, Gregorich SE. Factors affecting breast cancer risk reduction practices among California physicians. Prev Med. 2005;41(1):7-15. doi:10.1016/j.ypmed.2004.09.04
66. Kaplan CP, Kim SE, Wong ST, Sawaya GF, Walsh JM, Pérez-Stable EJ. Willingness to use tamoxifen to prevent breast cancer among diverse women. Breast Cancer Res Treat. 2012;133(1):357-366. doi:10.1007/s10549-012-1960-5
67. Liede A, Mansfield CA, Metcalfe KA, et al; Kathleen Cuningham Foundation Consortium for Research into Familial Breast Cancer. Preferences for breast cancer risk reduction among BRCA1/BRCA2 mutation carriers: a discrete-choice experiment. Breast Cancer Res Treat. 2017;165(2):433-444. doi:10.1007/s10549-017-4332-3
68. Martinez KA, Fagerlin A, Witteman HO, Holmberg C, Hawley ST. What matters to women when making decisions about breast cancer chemoprevention? Patient. 2016;9(2):149-159. doi:10.1007/s40271-015-0134-z
69. Meiser B, Wong WKT, Peate M, Julian-Reynier C, Kirk J, Mitchell G. Motivators and barriers of tamoxifen use as risk-reducing medication amongst women at increased breast cancer risk: a systematic literature review. Hered Cancer Clin Pract. 2017;15:14. doi:10.1186/s13053-017-0075-8
70. Reimers LL, Sivasubramanian PS, Hershman D, et al. Breast cancer chemoprevention among high-risk women and those with ductal carcinoma in situ. Breast J. 2015;21(4):377-386. doi:10.1111/tbj.12418
71. Roetzheim RG, Lee JH, Fulp W, et al. Acceptance and adherence to chemoprevention among women at increased risk of breast cancer. Breast. 2015;24(1):51-56. doi:10.1016/j.breast.2014.11.0
72. Ropka ME, Keim J, Philbrick JT. Patient decisions about breast cancer chemoprevention: a systematic review and meta-analysis. J Clin Oncol. 2010;28(18):3090-3095. doi:10.1200/JCO.2009.27.807
73. Skandarajah AR, Thomas S, Shackleton K, Chin-Lenn L, Lindeman GJ, Mann GB. Patient and medical barriers preclude uptake of tamoxifen preventative therapy in women with a strong family history. Breast. 2017;32:93-97. doi:10.1016/j.breast.2017.01.002
74. Cuzick J, Sestak I, Cawthorn S, et al; IBIS-I Investigators. Tamoxifen for prevention of breast cancer: extended long-term follow-up of the IBIS-I breast cancer prevention trial. Lancet Oncol. 2015;16(1):67-75. doi:10.1016/S1470-2045(14)71171-4
75. Sestak I, Singh S, Cuzick J, et al. Changes in bone mineral density at 3 years in postmenopausal women receiving anastrozole and risedronate in the IBIS-II bone substudy: an international, double-blind, randomised, placebo-controlled trial. [published correction appears in Lancet Oncol. 2014;15(13):e587]. Lancet Oncol. 2014;15(13):1460-1468. doi:10.1016/S1470-2045(14)71035-6
76. Spagnolo F, Sestak I, Howell A, Forbes JF, Cuzick J. Anastrozole-induced carpal tunnel syndrome: results from the International Breast Cancer Intervention Study II Prevention Trial. J Clin Oncol. 2016;34(2):139-143. doi:10.1200/JCO. 2015.63.4972
77. Maunsell E, Goss PE, Chlebowski RT, et al. Quality of life in MAP.3 (Mammary Prevention 3): a randomized, placebo-controlled trial evaluating exemestane for prevention of breast cancer. J Clin Oncol. 2014;32(14):1427-1436. doi:10.1200/JCO.2013.51.2483
78. Land SR, Wickerham DL, Costantino JP, 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. JAMA. 2006;295(23):2742-2751. doi:10.1001/jama.295.23.joc6007
79. Vogel VG, Costantino JP, Wickerham DL, et al; National Surgical Adjuvant Breast and Bowel Project. Update of the National Surgical Adjuvant Breast and Bowel Project Study of Tamoxifen and Raloxifene (STAR) P-2 Trial: preventing breast cancer. Cancer Prev Res (Phila). 2010;3(6):696-706. doi:10.1158/1940-6207.CAPR-10-0076
80. Cuzick J, Forbes JF, Sestak I, et al; International Breast Cancer Intervention Study I Investigators. Long-term results of tamoxifen prophylaxis for breast cancer—96-month follow-up of the randomized IBIS-I trial. J Natl Cancer Inst. 2007;99(4):272-282. doi:10.1093/jnci/djk049
81. Day R, Ganz PA, Costantino JP. Tamoxifen and depression: more evidence from the National Surgical Adjuvant Breast and Bowel Project’s Breast Cancer Prevention (P-1) randomized study. J Natl Cancer Inst. 2001;93(21):1615-1623. doi:10.1093/jnci/93.21.1615
82. Fisher B, Costantino JP, Wickerham DL, et al. Tamoxifen for the prevention of breast cancer: current status of the National Surgical Adjuvant Breast and Bowel Project P-1 study. J Natl Cancer Inst. 2005;97(22):1652-1662. doi:10.1093/jnci/dji372
83. Powles TJ, Ashley S, Tidy A, Smith IE, Dowsett M. Twenty-year follow-up of the Royal Marsden randomized, double-blinded tamoxifen breast cancer prevention trial. J Natl Cancer Inst. 2007;99(4):283-290. doi:10.1093/jnci/djk050
84. Veronesi U, Maisonneuve P, Rotmensz N, et al; Italian Tamoxifen Study Group. Tamoxifen for the prevention of breast cancer: late results of the Italian Randomized Tamoxifen Prevention Trial among women with hysterectomy. J Natl Cancer Inst. 2007;99(9):727-737. doi:10.1093/jnci/djk154
85. Veronesi U, Maisonneuve P, Sacchini V, Rotmensz N, Boyle P; Italian Tamoxifen Study Group. Tamoxifen for breast cancer among hysterectomised women. Lancet. 2002;359(9312):1122-1124. doi:10.1016/S0140-6736(02)08159-X
86. Veronesi U, Maisonneuve P, Rotmensz N, et al; Italian Tamoxifen Study Group. Italian randomized trial among women with hysterectomy: tamoxifen and hormone-dependent breast cancer in high-risk women. J Natl Cancer Inst. 2003;95(2):160-165.
87. Decensi A, Maisonneuve P, Rotmensz N, et al; Italian Tamoxifen Study Group. Effect of tamoxifen on venous thromboembolic events in a breast cancer prevention trial. Circulation. 2005;111(5):650-656. doi:10.1161/01.CIR.0000154545.84124.AC
88. Ettinger B, Black DM, Mitlak BH, et al. Reduction of vertebral fracture risk in postmenopausal women with osteoporosis treated with raloxifene: results from a 3-year randomized clinical trial. Multiple Outcomes of Raloxifene Evaluation (MORE) Investigators. JAMA. 1999;282(7):637-645. doi:10.1001/jama.282.7.637
89. Cauley JA, Norton L, Lippman ME, 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;65(2):125-134. doi:10.1023/A:1006478317173
90. Cummings SR, Eckert S, Krueger KA, et al; Multiple Outcomes of Raloxifene Evaluation. The effect of raloxifene on risk of breast cancer in postmenopausal women: results from the MORE randomized trial. JAMA. 1999;281(23):2189-2197. doi:10.1001/jama.281.23.2189
91. Barrett-Connor E, Cauley JA, Kulkarni PM, Sashegyi A, Cox DA, Geiger MJ. Risk-benefit profile for raloxifene: 4-year data from the Multiple Outcomes of Raloxifene Evaluation (MORE) randomized trial. J Bone Miner Res. 2004;19(8):1270-1275. doi:10.1359/JBMR.040406
92. Barrett-Connor E, Grady D, Sashegyi A, et al; MORE Investigators (Multiple Outcomes of Raloxifene Evaluation). Raloxifene and cardiovascular events in osteoporotic postmenopausal women: four-year results from the MORE (Multiple Outcomes of Raloxifene Evaluation) randomized trial. JAMA. 2002;287(7):847-857.
93. Delmas PD, Ensrud KE, Adachi JD, et al; Multiple Outcomes of Raloxifene Evaluation Investigators. Efficacy of raloxifene on vertebral fracture risk reduction in postmenopausal women with osteoporosis: four-year results from a randomized clinical trial. J Clin Endocrinol Metab. 2002;87(8):3609-3617. doi:10.1210/jcem.87.8.8750
94. Delmas PD, Genant HK, Crans GG, et al. Severity of prevalent vertebral fractures and the risk of subsequent vertebral and nonvertebral fractures: results from the MORE trial. Bone. 2003;33(4):522-532. doi:10.1016/S8756-3282(03)00241-2
95. Duvernoy CS, Kulkarni PM, Dowsett SA, Keech CA. Vascular events in the Multiple Outcomes of Raloxifene Evaluation (MORE) trial: incidence, patient characteristics, and effect of raloxifene. Menopause. 2005;12(4):444-452. doi:10.1097/01.GME.0000151653.02620.89
96. Grady D, Ettinger B, Moscarelli E, et al; Multiple Outcomes of Raloxifene Evaluation Investigators. Safety and adverse effects associated with raloxifene: multiple outcomes of raloxifene evaluation. Obstet Gynecol. 2004;104(4):837-844. doi:10.1097/01.AOG.0000137349.79204.b8
97. Keech CA, Sashegyi A, Barrett-Connor E. Year-by-year analysis of cardiovascular events in the Multiple Outcomes of Raloxifene Evaluation (MORE) trial. Curr Med Res Opin. 2005;21(1):135-140. doi:10.1185/030079904X18045
98. Lippman ME, Cummings SR, Disch DP, et al. Effect of raloxifene on the incidence of invasive breast cancer in postmenopausal women with osteoporosis categorized by breast cancer risk. Clin Cancer Res. 2006;12(17):5242-5247. doi:10.1158/1078-0432.CCR-06-0688
99. Martino S, Disch D, Dowsett SA, Keech CA, Mershon JL. Safety assessment of raloxifene over eight years in a clinical trial setting. Curr Med Res Opin. 2005;21(9):1441-1452. doi:10.1185/030079905X61839
100. Silverman SL, Delmas PD, Kulkarni PM, Stock JL, Wong M, Plouffe L Jr. Comparison of fracture, cardiovascular event, and breast cancer rates at 3 years in postmenopausal women with osteoporosis. J Am Geriatr Soc. 2004;52(9):1543-1548. doi:10.1111/j.1532-5415.2004.52420.x
101. Siris ES, Harris ST, Eastell R, et al; Continuing Outcomes Relevant to Evista (CORE) Investigators. Skeletal effects of raloxifene after 8 years: results from the continuing outcomes relevant to Evista (CORE) study. J Bone Miner Res. 2005;20(9):1514-1524. doi:10.1359/JBMR.050509
102. Johnell O, Cauley JA, Kulkarni PM, Wong M, Stock JL. Raloxifene reduces risk of vertebral fractures [corrected] in postmenopausal women regardless of prior hormone therapy. J Fam Pract. 2004;53(10):789-796.
103. Grady D, Cauley JA, Geiger MJ, et al; Raloxifene Use for The Heart Trial Investigators. Reduced incidence of invasive breast cancer with raloxifene among women at increased coronary risk. J Natl Cancer Inst. 2008;100(12):854-861. doi:10.1093/jnci/djn153
104. Ensrud KE, Stock JL, Barrett-Connor E, et al. Effects of raloxifene on fracture risk in postmenopausal women: the Raloxifene Use for the Heart Trial. J Bone Miner Res. 2008;23(1):112-120. doi:10.1359/jbmr.070904
105. Brisson J, Brisson B, Coté G, Maunsell E, Bérubé S, Robert J. Tamoxifen and mammographic breast densities. Cancer Epidemiol Biomarkers Prev. 2000;9(9):911-915.
106. Bruno S, Maisonneuve P, Castellana P, et al. Incidence and risk factors for non-alcoholic steatohepatitis: prospective study of 5408 women enrolled in Italian tamoxifen chemoprevention trial. BMJ. 2005;330(7497):932. doi:10.1136/bmj.38391.663287.E0
107. Chalas E, Costantino JP, Wickerham DL, et al. Benign gynecologic conditions among participants in the Breast Cancer Prevention Trial. Am J Obstet Gynecol. 2005;192(4):1230-1237. doi:10.1016/j.ajog.2004.12.083
108. Reis SE, Costantino JP, Wickerham DL, Tan-Chiu E, Wang J, Kavanah M; National Surgical Adjuvant Breast and Bowel Project Breast Cancer Prevention Trial Investigators. Cardiovascular effects of tamoxifen in women with and without heart disease: Breast Cancer Prevention Trial. J Natl Cancer Inst. 2001;93(1):16-21. doi:10.1093/jnci/93.1.16
109. Runowicz CD, Costantino JP, Wickerham DL, et al. Gynecologic conditions in participants in the NSABP breast cancer prevention Study of Tamoxifen and Raloxifene (STAR). Am J Obstet Gynecol. 2011;205(6):535.e1-535. doi:10.1016/j.ajog.2011.06.067
110. Goldstein SR, Johnson S, Watts NB, Ciaccia AV, Elmerick D, Muram D. Incidence of urinary incontinence in postmenopausal women treated with raloxifene or estrogen. Menopause. 2005;12(2):160-164. doi:10.1097/00042192-200512020-00010
111. Abramson N, Costantino JP, Garber JE, Berliner N, Wickerham DL, Wolmark N. Effect of Factor V Leiden and prothrombin G20210-->A mutations on thromboembolic risk in the National Surgical Adjuvant Breast and Bowel Project Breast Cancer Prevention Trial. J Natl Cancer Inst. 2006;98(13):904-910. doi:10.1093/jnci/djj262
112. Forbes JF, Sestak I, Howell A, et al; IBIS-II Investigators. Anastrozole versus tamoxifen for the prevention of locoregional and contralateral breast cancer in postmenopausal women with locally excised ductal carcinoma in situ (IBIS-II DCIS): a double-blind, randomized controlled trial. Lancet. 2016;387(10021):866-873. doi:10.1016/S0140-6736(15)01129-0
113. Early Breast Cancer Trialists' Collaborative Group (EBCTCG). Aromatase inhibitors versus tamoxifen in early breast cancer: patient-level meta-analysis of the randomised trials. Lancet. 2015;386(10001):1341-1352. doi:10.1016/S0140-6736(15)61074-1
114. Goldvaser H, Barnes TA, Šeruga B, et al. Toxicity of extended adjuvant therapy with aromatase inhibitors in early breast cancer: a systematic review and meta-analysis. J Natl Cancer Inst. 2018;110(1). doi:10.1093/jnci/djx141
115. King MC, Wieand S, Hale K, et al; National Surgical Adjuvant Breast and Bowel Project. Tamoxifen and breast cancer incidence among women with inherited mutations in BRCA1 and BRCA2: National Surgical Adjuvant Breast and Bowel Project (NSABP-P1) Breast Cancer Prevention Trial. JAMA. 2001;286(18):2251-2256. doi:10.1001/jama.286.18.2251

Return to Table of Contents

Figure 1 is a flow diagram of the selection of candidates for medications to reduce the risk for primary breast cancer. Women without preexisting breast cancer are screened and sorted into either appropriate or inappropriate candidates for medication. Women who are appropriate candidates for medication receive medications to reduce the risk for primary breast cancer. Women may experience adverse effects due to these medications. Women who are treated with these medications may have reduced incidence of invasive breast cancer and reduced incidence of noninvasive breast cancer, which may lead to improved breast cancer related mortality and all-cause mortality.

Evidence reviews for the US Preventive Services Task Force (USPSTF) use an analytic framework to visually display key questions addressed by the review to allow the USPSTF to evaluate the effectiveness and safety of a preventive service. The questions are depicted by linkages that relate interventions to outcomes. A dashed line indicates a health outcome that precedes subsequent outcomes. A circle that is half blue and half orange indicates a key question that pertains to both benefits and harms, such as key question 4. Refer to the USPSTF procedure manual for further details.12

Return to Table of Contents

Figure 2 is a literature flow diagram depicting the search and selection of articles for the review. The diagram shows that 1786 abstracts of potentially relevant articles were identified through MEDLINE, Cochrane databases (including the Cochrane Central Register of Controlled Trials and the Cochrane Database of Systematic Reviews), EMBASE and other sources (reference lists of relevant articles, studies, and systematic reviews, suggestions from reviewers, etc.). Of these 1269 citations were excluded for not being relevant to the key questions, and 50 papers were pulled for relevance to the contextual questions. The remaining 467 articles were retrieved and reviewed at the full-text level. Of these 454 were excluded for the following reasons: background information only (68), used for contextual questions only (14), population not applicable (34), intervention not applicable (38), wrong outcomes (74), companion paper, data not used (15), non-systematic or outdated review (78), wrong study design for key question (9), wrong publication type (122), or non-English language paper (2). An additional 101 full-text articles from the prior report were reviewed for inclusion in this report. Of these 32 were excluded for the following reasons:  background information only (2), used for contextual questions only (6), intervention not appropriate (3), wrong outcomes (13), companion paper, data not used (7), or non-systematic review (1). After exclusion of these studies, 46 studies in 82 publications were included that provide evidence for the key questions, as follows: 6 new studies and 19 studies from the prior report for Key Question 1,  4 new trials in 7 publications and 7 trials in 33 publications from the prior report for Key Question 2, 5 new trials in 8 publications and 18 studies in 46 publications from the prior report for Key Question 3, and 2 new trials in 2 publications and 7 trials in 10 publications from the prior report for Key Question 4.

KQ indicates key question.
a Forty-six studies in 82 publications provided data; some addressed more than 1 KQ.

Return to Table of Contents

Figure 3 is a forest plot of studies reporting the risk reduction of invasive breast cancer stratified by type of medication.

The area of the square representing the risk ratio is proportional to the number of events in each subgroup. CORE indicates Continuing Outcomes Relevant to Evista; HOT, Hormone replaced therapy Opposed by low-dose Tamoxifen; IBIS, International Breast Cancer Intervention Study; MAP.3, Mammary Prevention.3; MORE, Multiple Outcomes of Raloxifene; NSABP-1, National Surgical Adjuvant Breast and Bowel Project P-1; RUTH, Raloxifene Use for the Heart.
a Veronesi (2007) and Barrett-Connor (2006) reported mean or median duration of the treatment period.
b Includes data from both MORE (4-year treatment) and CORE (4-year additional treatment); total follow-up time is averaged over both studies for 7705 participants.
c Intended treatment duration is 5 years or until a breast, neoplastic, cardiovascular, or toxicity event.

Return to Table of Contents

Model Age, y Age at Menarche, y Age at Birth of First Child, y No. First-Degree Relatives With
Breast Cancer
No. Previous Breast Biopsies Other Factors Summary of Accuracy, AUC (95% CI)a
Gail 2 (5-y risk) <50; ≥50 <12; 12-13; ≥14 <20; 20-24; 25-29; ≥30; no children 0; 1; ≥2 0; 1; ≥2
Atypical hyperplasia: 0; ≥1
Not included 0.55 (0.51-0.60)16
0.6026
0.58 (0.56-0.60)34
0.5820
0.59 (0.54-0.63)27
0.6022
0.61 (0.60-0.62)36
Gail 2 (10-y risk) <50; ≥50 <12; 12-13; ≥14 <20; 20-24; 25-29; ≥30; no children 0; 1; ≥2 0; 1; ≥2
Atypical hyperplasia: 0; ≥1
Not included 0.74 (0.67-0.80)17
0.54 (0.52-0.56)21
African American Gail (5-y risk) <50; ≥50 ≤13; >13 Not included 0; 1; ≥2 0; 1; ≥2 African American race 0.56 (0.54-0.58)28
0.56 (0.51-0.60)16
Asian American Gail (5-y risk) <50; ≥50 ≤13; >13 Not included 0; 1; ≥2 0; 1; ≥2 Asian American race 0.61 (0.59 to 0.64)31
Gail + breast density (10-y risk) <50; ≥50 <12; 12-13; ≥14 <20; 20-24; 25-29; ≥30; no children 0; 1; ≥2 Yes; no Breast density (%); BMI 0.59 (0.57-0.61)21
Gail + breast density (5-y risk) <50; ≥50 <12; 12-13; ≥14 <20; 20-24; 25-29; ≥30; no children 0; 1; ≥2 0; 1; ≥2 Breast density (%); BMI 0.6422
BCSC (premenopausal; 1-y risk) 45-84 by 5-y groups Not included Not included 0; 1; ≥2; unknown Yes; no; unknown Breast density (BI-RADS)b 0.63 (0.60-0.66)18
BCSC (postmenopausal; 1-y risk) 45-84 by 5-y groups Not included First- and second-degree 0; 1; ≥2; unknown 0; ≥1; unknown Breast density (BI-RADS), prior false-positive mammogram, BMI, menopause type, hormone therapy, race or ethnicity 0.62 (0.62-0.63)18
BCSC (5-y risk) 45-84 by 5-y groups Not included Not included Yes; no Yes; no Breast density (BI-RADS), race or ethnicity 0.66 (0.65-0.66)36
0.66437
BCSC + benign breast disease (5-y risk)c 45-84 by 5-y groups Not included Not included Yes; no Yes; no Breast density (BI-RADS), race or ethnicity, benign breast disease 0.66537
Rosner-Colditzd <50; ≥50 <12; 12-13; ≥14 <20; 20-24; 25-29; ≥30; no children Yes; no Not included BMI, benign breast disease, menopause type, menopause age, hormone therapy use and duration, height, alcohol use, parity 0.57 (0.55-0.59)33
0.64 (0.63-0.66) (ER+/PR+)25
0.61 (0.58-0.64) (ER−/PR−)25
Rosner-Colditz 2d <50; ≥50 <12; 12-13; ≥14 <20; 20-24; 25-29; ≥30; no children Yes; no Atypical hyperplasia: 0; ≥1 Benign breast disease presence or type 0.63 (0.61-0.65)33
0.64 (type)33
Tyrer-Cuzick (10-y risk) <50; ≥50 ≤12; >12 ≤30; >30; no children 1; 2; ≥3 0; 1; ≥2
Lobular carcinoma in situ: 0; ≥1
BMI, height, menopause age, family history of ovarian or other cancer, age of cancer onset, bilateral or male breast cancer 0.76 (0.70-0.82)17
0.54 (0.42-0.65)19
0.57 (0.55-0.59)21
Tyrer-Cuzick + breast density (10-y risk) <50; ≥50 ≤12; >12 ≤30; >30; no children 1; 2; ≥3 Yes; no BMI, height, menopause age, family history of ovarian or other cancer, age of cancer onset, bilateral or male breast cancer; breast density (%) 0.61 (0.58-0.63)21
0.6240
Italian 1 (5-y risk) <50; ≥50 <12; 12-13; ≥14 <20; 20-24; 25-29; ≥30; no children 0; 1; ≥2 Not included Age of relative at breast cancer diagnosis, diet score, alcohol use, BMI, hormone therapy, physical activity 0.59 (vitamin)20
0.60 (diet)20
Italian 2 (20-y risk)e <50; ≥50 <12; 12-13; ≥14 <20; 20-24; 25-29; ≥30; no children 0; 1; ≥2 0; 1; ≥2 Occupational and leisure physical activity, education, alcohol use, BMI 0.62 (0.56-0.69) (age <50 y)32
0.57 (0.52-0.61) (age ≥50 y)32
Chlebowski (5-y risk) 50-59; 60-69; 70-79 <12; 12-13; ≥14 <20; 20-24; 25-29; ≥30; no children 0; ≥1 0; 1; ≥2 BMI, menopause age, hormone therapy use and duration, race, alcohol use, parity, breastfeeding, smoking status, physical activity 0.61 (0.59-0.63)23
0.62 (0.60-0.64) (ER+)23
0.53 (0.47-0.58) (ER−)23
Chlebowski-simplified (5-y risk) <50; ≥50 Not included Not included 0; ≥1 0; 1; ≥2 Not included 0.58 (0.56-0.60) (ER+)23
ModelER+ Not included <12; 12-13; ≥14 ≤20; 20.1-25; 25.1-30; 30.1-35; >35 Not included Not included BMI, menopause status and age, alcohol use, hormone therapy use, breast-feeding duration, parity 0.59 (0.58 to 0.60)30

Abbreviations: AUC, area under the receiver operating characteristic curve; BCSC, Breast Cancer Surveillance Consortium; BI-RADS, Breast Imaging–Reporting and Data System; BMI, body mass index; ER−, estrogen receptor–negative; ER+, estrogen receptor–positive.
a From studies of discriminatory accuracy for invasive breast cancer unless otherwise indicated.
b BI-RADS categories include 0 (unknown), 1 (entirely fat), 2 (scattered fibroglandular densities), 3 (heterogeneously dense), 4 (extremely dense).
c Includes nonproliferative, proliferative without atypia, proliferative with atypia, and lobular carcinoma in situ.
d Invasive and noninvasive breast cancer.
e Included an Italian population and used incidence rates from the Italian multicenter case-control study of diet and breast cancer and from Italian cancer registries.

Return to Table of Contents

Source Group, No.a Breast Cancer Risk
Criteria
Participants,
Setting
Age, Median, y No. (%)a Primary
Outcomes
Median, y Quality Rating
1 2 White Post-hyster-ectomy Used Estrogen During Trial Follow-Up Exposure
Tamoxifen (20 mg/d) vs Raloxifene (60 mg/d)
STAR
Vogel et al,49 2006
Land et al,78 2006
Vogel et al,79 2010
9872 9875 5-y predicted breast cancer risk ≥1.66% based on the modified Gail modelb Postmenopausal, aged ≥35 y, US-based with sites in North America 58.5c 18,204
(93.5)
10,027 (51.5) 0 Invasive breast cancer 3.9 initial; 6.8 long-termc 3.6-3.9c Good
Tamoxifen (20 mg/d) vs Placebo
IBIS-I
Cuzick et al,42 2002
Cuzick et al,80 2007
Cuzick et al,74 2015
3573 3566 2-fold relative risk for breast cancer for ages 45-70 y, 4-fold for ages 40-44 y, 10-fold for ages 35-39 y based on family history criteriad 35-70 y, United Kingdom, Australia, New Zealand, Europe 50.8c NR 2515 (35) 2844 (40) Invasive and noninvasive breast cancer 4.2 initial; 8.0 long-term; 16 longer-term 5 Good
NSABP-P1
Fisher et al,46 1998
Fisher et al,82 2005
Day et al, 81 2001
6576 6599 Age ≥60 y or 35-59 y with a 5-y predicted breast cancer risk ≥1.66% based on the modified Gail model or history of LCISb ≥35 y, US-based with sites in North America Median not reported; 5177 (39.3%) <50 12,706 (96.4) 4884 (37) NR (<10) Invasive and noninvasive breast cancer 4.6 initial; 7.0 long-term 4.0 when unblinded Good
Royal Marsden Hospital Trial
Powles et al,47 1998
Powles et al,83 2007
1238 1233 Family history of breast cancere 30-70 y, United Kingdom 47 NR NR 389 (15.6) Invasive breast cancer 5.8 initial; 13.2 long-term NR Good
Italian Tamoxifen Prevention Study
Veronesi et al,43 1998
Veronesi et al,86 2003
Veronesi et al,84 2007
Decensi et al,87 2005
2700 2708 None 35-70 y, Italy-based with sites in Europe and South America 51 NR 100 (100) 751 (14) Breast cancer incidence and mortality 3.8 initial; 11.2 long-term 4 Fair; dropout rate 26.3%
Tamoxifen (5 mg/d) vs Placebo
HOT
DeCensi et al,41 2013
938 946 None Postmenopausal, Italy-based 53c NR NR 100 (100) Invasive breast cancer 6.2c 5c Good
Raloxifene (60 or 120 mg/d) vs Placebo
MORE and CORE
Cauley et al,89 2001
Cummings et al,90 1999
Ettinger et al,88 1999
Barrett-Connor et al,92 2002
Delmas et al,93 2002
Delmas et al,94 2003
Grady et al,96 2004
Barrett-Connor et al,91 2002
Silverman et al,100 2004
Johnell et al,102 2004;
Martino et al,45 2004;
Martino et al,99 2005
Duvernoy et al,95 2005
Keech et al,97 2005
Siris et al,101 2005
Lippman et al,98 2006
MORE: 5129l
CORE: 2725
MORE: 2576;
CORE: 1286
None Postmenopausal, aged 31-80 y, with osteoporosis, US-based with sites in 25 countries; CORE includes a subset of MORE participantsf 66.9 NR (96) NR (23) 0 MORE: Incident radiographic vertebral fractures and clinical nonvertebral fractures;
CORE: Breast cancer
MORE: 3, 4
CORE: 4,8 (combines data)
NR Good
RUTH
Barrett-Connor et al,48 2006
Grady et al,103 2008
Ensrud et al,104 2008
5044 5057 None Postmenopausal, aged ≥55 y, CHD or risk factors, US-based with sites in 26 countriesg 67.5 8481 (84) 2319 (23) 0 Coronary events, invasive breast cancer 5.6 5.1 Good
Anastrozole (1 mg/d) vs Placebo
IBIS-II
Cuzick et al,4 2014
Sestak et al,75 2014
Spagnolo et al,76 2016
1920 1944 Increased risk for breast cancer: ages 45-60 y ≥2 times higher than the general population; ages 60-70 y 1.5 times higher; ages 40-44 y 4 times higher Postmenopausal, aged 40-70 y, United Kingdom-based with sites in 18 countries 59.5 NR 1287 (33.3) 0 Invasive and noninvasive breast cancer 5 5 Good
Exemestane (25 mg/d) vs Placebo
MAP.3
Goss et al,44 2011
Maunsell et al,77 2014
2285 2275 Risk factors for breast cancer: age ≥60 y; Gail risk score >1.66%; prior ADH, ALH, LCIS, or DCIS Postmenopausal, age ≥35 y, US-based with sites in 4 countries 62.5 4261 (93.4) NR 0 Invasive breast cancer 2.9 3 Good

Abbreviations: ADH, atypical ductal hyperplasia; ALH, atypical lobular hyperplasia; CHD, coronary heart disease; CORE, Continuing Outcomes Relevant to Evista; DCIS, ductal carcinoma in situ; HOT, Hormone replacement therapy Opposed by low-dose Tamoxifen; IBIS, International Breast Cancer Intervention Study; KQ, key question; LCIS, lobular carcinoma in situ; MAP.3, Mammary Prevention.3 trial; MORE, Multiple Outcomes of Raloxifene Evaluation; NR, not reported; NSABP-P1, National Surgical Adjuvant Breast and Bowel Project P-1; RUTH, Raloxifene Use for the Heart; STAR, Study of Tamoxifen and Raloxifene.
a At time of randomization.
b STAR and NSABP-1: The Gail model includes age, number of first-degree relatives with breast cancer, nulliparity or age at first live birth, number of benign breast biopsy results, pathologic diagnosis of atypical hyperplasia, and age at menarche. The original model was further modified to predict expected rates of invasive breast cancer only (not invasive and noninvasive as originally designed) and to allow for race-specific determinations of risk.
c Values are means.
d IBIS: All criteria permit entry to trial at age 45 years: first-degree relative with breast cancer at 50 years or younger; first-degree relative with bilateral breast cancer (permits entry from age 40 years; if relative age ≤40 years, permits entry at age 35 years); 2 or more first-degree or second-degree relatives with breast cancer (permits entry from age 40 years if both developed breast cancer before age 50 years; permits entry at age 35 years if both relatives are first-degree and both relatives developed breast cancer before age 50 years); benign breast biopsy and first-degree relative with breast cancer; lobular carcinoma in situ (permits entry from age 35 years); atypical hyperplasia (permits entry from age 40 years); nulliparous and a first-degree relative who developed breast cancer; risk equivalent (strong family history, not fitting specific categories, but judged to be at higher risk than eligibility category by the study chairman).
e Family history criteria, Royal Marsden Hospital Trial: 1 first-degree relative younger than 50 years with breast cancer, or 1 first-degree relative with bilateral breast cancer, or 1 affected first-degree of any age plus another affected first-degree or second-degree relative; benign breast biopsy result and a first-degree relative with breast cancer.
f MORE: study group 1, femoral neck or lumbar spine bone mineral density T-score less than −2.5; study group 2, low bone mineral density and 1 or more moderate or severe vertebral fractures or 2 or more milder vertebral fractures (20%-25%reduction in height); or 2 or more moderate fractures (25%-40% reduction from expected vertebral height), regardless of bone mineral density.
g Cardiovascular risk score of 4 or greater: established coronary heart disease (4 points), arterial disease of the leg (4 points), 70 years or older (2 points), diabetes mellitus (3 points), cigarette smoking (1 point), hypertension (1 point), and hyperlipidemia (1 point).

Return to Table of Contents

Outcome Tamoxifen vs Raloxifene (Single Trial) Tamoxifen vs Placebo Raloxifene vs Placebo Aromatase Inhibitors vs Placebo
RR (95% CI) No. of Events Reduced or Increased (95% CI)a RR (95% CI) No. of Trialsb Placebo
Rate (SE)c
No. of Events Reduced or Increased (95% CI)a RR (95% CI) No. of Trialsb Placebo
Rate (SE)c
No. of Events Reduced or Increased (95% CI)a RR (95% CI) No. of Trialsb Placebo
Rate (SE)c
No. of Events Reduced or Increased (95% CI)a
Benefits
Invasive breast cancer 1.24 (1.05-1.47)d 5 (1-9) fewer with tamoxifen 0.69 (0.59-0.84) 4 4.58 (0.96) 7 (4-12) fewer with tamoxifen 0.44 (0.24-0.80) 2 3.19 (0.59) 9 (3-15) fewer with raloxifen 0.45 (0.26-0.70) 2 5.90 (0.6) 16 (8-24) fewer with AIs
ER+ breast cancer 0.93 (0.72-1.24)e NA 0.58 (0.42-0.81 4 3.62 (0.76) 8 (4-13) fewer with tamoxifen 0.33 (0.15-0.70) 2 2.45 (0.42) 8 (4-13) fewer with raloxifene 0.37 (0.19-0.63) 2 4.55 (0.53) 15 (8-20) fewer with AIs
ER− breast cancer 1.15 (0.75-1.77)e NA 1.18 (0.93-1.53) 4 NA NA 1.25 (0.60-2.58) 2 NA NA 0.79 (0.35-1.79) 2 NA NA
Noninvasive breast cancer 1.22 (0.95-1.59)d NA 0.72 (0.56-1.41)f 4 NA NA 1.47 (0.61-3.85) 2 NA NA 0.46 (0.16-1.42) 2 NA NA
Breast cancer mortality 0.36 (0.08-1.21)d NA 1.20 (0.79-1.79) 4 NA NA Not reportedg NA NA NA Not reported NA NA NA
All-cause mortality 0.84 (0.70-1.02)d NA 1.07 (0.91-1.23) 4 NA NA 0.90 (0.63-1.05) 2 NA NA 1.02 (0.58-1.82) 2 NA NA
Vertebral fracture 0.98 (0.65-1.46)e NA 0.75 (0.48-1.15)h 1 NA NA 0.61 (0.53-0.73) 2 3.45 (0.35)i 7 (5-9) fewer with raloxifene 1.28 (0.59-2.75) 2 NA NA
Nonvertebral fracture Not reported NA 0.66 (0.45-0.98)h 1 1.55 (0.20) 3 (0.2-5) fewer with tamoxifen 0.97 (0.86-1.12) 2 NA NA 1.05 (0.87-1.28) 2 NA NA
Harms
Venous thromboembolismj 0.75 (0.60-0.93)d 4 (1-7) more with tamoxifen 1.93 (1.33-2.68) 4 0.91 (0.19) 5 (2-9) more with tamoxifen 1.56 (1.11-2.60) 2 2.34 (0.25) 7 (0.3-17) more with raloxifene 1.24 (0.65-2.16) 2 NA NA
DVT 0.72 (0.54-0.95)d 3 (1-5) more with tamoxifen 1.45 (0.73-2.59) 2 NA NA 1.66 (0.79-5.14) 2 NA NA Not reported NA NA NA
PE 0.80 (0.57-1.11)d NA 2.69 (0.54-8.13) 2 NA NA 2.11 (0.82-6.12) 2 NA NA Not reported NA NA NA
CHD events 1.10 (0.85-1.43)e NA 1.00 (0.75-1.30) 4 NA NA 0.95 (0.80-1.10) 2 NA NA 0.76 (0.41-1.49) 2 NA NA
Stroke 0.96 (0.64-1.43)e NA 1.36 (0.78-2.20) 4 NA NA 1.04 (0.64-1.36) 2 NA NA 0.98 (0.27-2.56) 2 NA NA
Endometrial cancer 0.55 (0.36-0.83)d 5 (2-9) more with tamoxifen 2.25 (1.17-4.41) 3 0.62 (0.10) 4 (1-8) more with tamoxifen 1.14 (0.54-2.17) 2 NA NA 0.60 (0.09-3.07) 1 NA NA
Cataracts 0.80 (0.72-0.95)d 15 (8-22) more with tamoxifen 1.22 (1.08-1.48) 3 22.85 (0.75)k 26 (5-50) more with tamoxifen 0.93 (0.82-1.06) 2 NA NA 0.94 (0.70-1.27) 1 NA NA

Abbreviations: AI, aromatase inhibitor; CHD, coronary heart disease; DVT, deep vein thrombosis; ER−, estrogen receptor–negative; ER+, estrogen receptor–positive; KQ, key question; NA, not applicable; NSABP, National Surgical Adjuvant Breast and Bowel Project; PE, pulmonary embolism; RR, risk ratio; RUTH, Raloxifene Use for the Heart; SE, standard error; VTE, venous thromboembolism.
a Numbers of events reduced for benefits or increased for harms vs comparator per 1000 women assuming 5 years of use.
b Number of trials included in meta-analysis.
c Per 1000 women, estimated from ameta-analysis of rates from the placebo groups from the same trials included in the risk ratio estimate.
d Updated results from STAR (2010).52
e Initial results from STAR (2006).50
f Reduced in NSABP P-1 (2005) (60 vs 93 events; RR, 0.63 [95% CI, 0.45-0.89]).57
g Two breast cancer deaths in 7601 women for raloxifene vs 0 in 7633 women for placebo.75,83
h NSABP P-1 (2007).57
i Estimated from the placebo group of the RUTH trial (2006).82
j Includes DVT and PE.
k Placebo rate was from NSABP P-1 (2005).57

Return to Table of Contents

Intervention No. of Studies (No. of
Participants)
Summary of Findings Consistency and Precision Other Limitaitons Strength of Evidence Applicability
KQ1: Diagnostic Accuracy of Risk Assessment Methods
Breast cancer risk assessment 25 discriminatory accuracy studies of 18 risk stratification methods (>5,000,000) Methods have low discriminatory accuracy in predicting the probability of breast cancer in individuals (AUC, 0.55-0.65) Consistent; precise While some studies used inappropriate reference groups, enrolled small numbers, or inadequately described methods, most studies met criteria for good quality High High
KQ1a: Optimal Age at Which to Begin Risk Assessment
Breast cancer risk assessment No studies NA NA NA Insufficient NA
KQ1b: Optimal Frequency of Risk Assessment
Breast cancer risk assessment No studies NA NA NA Insufficient NA
KQ2: Benefits of Risk-Reducing Medications
Tamoxifen vs raloxifene 1 RCT (19,747) Risk for invasive breast cancer was higher for raloxifene compared with tamoxifen (RR, 1.24 [95% CI, 1.05-1.47]; 5 more cases [95% CI, 1-9]a)

No differences for ER+, ER−, or noninvasive breast cancer; all-cause or breast cancer-specific mortality; or fractures

NA None High; 1 large
definitive trial
High
Tamoxifen vs placebo 4 RCTs (28,193) Tamoxifen was associated with reduced invasive breast cancer (RR, 0.69 [95% CI, 0.59-0.84]; 7 fewer cases [95% CI, 4-12]a), ER+ breast cancer (RR, 0.58 [95% CI, 0.42-0.81]; 8 fewer cases [95% CI, 4-13]a), and nonvertebral fractures (RR, 0.66 [95% CI, 0.45-0.98]; 3 fewer cases [95% CI, 0.2-5]a) compared with placebo

No differences for ER− or noninvasive breast cancer, all-cause or breast cancer-specific mortality, or vertebral fractures

Consistent; precise Clinical heterogeneity across trials from varying eligibility criteria, adherence, and ascertainment of certain outcomes High for all outcomes except fractures (based on 1 trial) High
Raloxifene vs placebo 2 RCTs (17,806) Raloxifene was associated with reduced invasive breast cancer (RR, 0.44 [95% CI, 0.24-0.80]; 9 fewer cases [95% CI, 3-15]a), ER+ breast cancer (RR, 0.33 [95% CI, 0.15-0.70]; 8 fewer cases [95% CI, 4-13]a), and vertebral fractures (RR, 0.61 [95% CI, 0.53-0.73]; 7 fewer cases [95% CI, 5-9]a) compared with placebo

No differences for ER− or noninvasive breast cancer, all-cause or breast cancer-specific mortality, or nonvertebral fractures

Consistent; precise Trials were primarily designed for osteoporosis and cardiovascular outcomes; participants were not selected based on breast cancer risk High for all outcomes High
Aromatase inhibitors (anastrozole; exemestane) vs placebo 2 RCTs (8424) Aromatase inhibitors were associated with reduced invasive breast cancer (RR, 0.45 [95% CI, 0.26-0.70]; 16 fewer cases [95% CI, 8-24]a) and ER+ breast cancer (RR, 0.37 [95% CI, 0.19-0.63]; 15 fewer cases [95% CI, 8-20]a) compared with placebo

No differences for ER− or noninvasive breast cancer, all-cause or breast cancer-specific mortality, or fractures

Consistent; precise Trials used different medications and exposure durations High for all outcomes High
KQ2a: Benefits of Risk-Reducing Medications—Timing and Duration
Tamoxifen, raloxifene, aromatase inhibitors (anastrozole; exemestane) 9 RCTs (74,170) No differences in breast cancer outcomes by age

Despite variations in exposure times from 3-5 y, comparisons across similar medications indicated consistency in risk reduction for invasive breast cancer

Consistent; precise No trials compared timing and duration directly

Age categories and durations varied across trials

Moderate for tamoxifen; insufficient for other medications High
KQ2a: Benefits of Risk-Reducing Medications—Persistence of Effects
Tamoxifen, raloxifene, aromatase inhibitors (anastrozole; exemestane) 2 RCTs of tamoxifen (9610); no trials of other medications Tamoxifen reduced invasive and ER+ breast cancer 8 y after discontinuation Consistent; precise Long term follow-up data are lacking from most trials Moderate for tamoxifen; insufficient for other medications High
KQ3: Harms of Risk-Reducing Medications
Tamoxifen vs raloxifene 1 RCT (19,747) Tamoxifen was associated with increased thromboembolic events (RR, 0.75 [95% CI, 0.60-0.93]; 4 more cases [95% CI, 1-7]a), DVT (RR, 0.72 [95% CI, 0.54-0.95]; 3 more cases [95% CI, 1-5]a), endometrial cancer (RR, 0.55 [95% CI, 0.36-0.83]; 5 more cases [95% CI, 2-9]a), and cataracts (RR, 0.80 [95% CI, 0.72-0.95]; 15 more cases [95% CI, 8-22]a) compared with raloxifene

No differences for PE, CHD events, or stroke

NA None High; 1 large definitive trial High
Tamoxifen vs placebo 4 RCTs (28,193) Tamoxifen was associated with increased thromboembolic events (RR 1.93 [95% CI, 1.33-2.68]; 5 more cases [95% CI, 2-9]a), endometrial cancer (RR, 2.25 [95% CI, 1.17-4.41]; 4 more cases [95% CI, 1-8]a), and cataracts (RR, 1.22 [95% CI, 1.08-1.48]; 26 more cases [95% CI, 5-50]a) compared with placebo

No differences for DVT, PE, CHD events, or stroke

Consistent; precise Clinical heterogeneity across trials from varying eligibility criteria, adherence, and ascertainment of certain outcomes High for all outcomes except DVT, PE (based on 2 trials) High
Raloxifene vs placebo 2 RCTs (17,806) Raloxifene was associated with increased thromboembolic events (RR, 1.56 [95% CI, 1.11-2.60]; 7 more cases [95% CI, 0.3-17]a), endometrial cancer (RR, 2.25 [95% CI, 1.17-4.41]; 4 more cases [95% CI, 1-8]a), and cataracts (RR, 1.22 [95% CI, 1.08-1.48]; 26 more cases [95% CI, 5-50]a) compared with placebo

No differences for DVT, PE, CHD events, stroke, endometrial cancer, or cataracts

Consistent; precise Trials primarily designed for osteoporosis and cardiovascular outcomes; participants not selected based on breast cancer risk High for all outcomes High
Aromatase inhibitors (anastrozole; exemestane) vs placebo 2 RCTs (8424) No differences between aromatase inhibitors and placebo for thromboembolic events, DVT, PE, CHD events, stroke, endometrial cancer, or cataract Consistent; precise Trials used different medications and exposure durations; no long-term follow-up data Low to moderate; follow-up inadequate for several outcomes High
KQ3a: Harms of Risk-Reducing Medication—Timing and Duration
Tamoxifen, raloxifene, aromatase inhibitors (anastrozole; exemestane) 2 RCTs of tamoxifen for thromboembolic events (18, 583); 1 RCT of tamoxifen (13,175) for endometrial cancer; no trials of other medications Risks for thromboembolic events and endometrial cancer with tamoxifen were higher for older compared with younger women Consistent; precise No trials compared timing and duration directly

Age categories and durations varied across trials

Moderate for tamoxifen; insufficient for other medications High
KQ3a: Harms of Risk-Reducing Medication—Persistence of Effects
Tamoxifen, raloxifene, aromatase inhibitors (anastrozole; exemestane) 2 RCTs of tamoxifen for thromboembolic events (9610); 1 RCT of tamoxifen (7139) for endometrial cancer; no trials of other medications Risks for thromboembolic events and endometrial cancer with tamoxifen declined to normal after discontinuation Consistent; precise Long-term follow-up data are lacking from most trials Moderate for tamoxifen; insufficient for other medications High
KQ4: Variability by Subpopulations
Tamoxifen, raloxifene, aromatase inhibitors (anastrozole; exemestane) 2 RCTs with menopausal status (12,547); 5 RCTs with family history (56,136); 4 RCTs with BMI (26,230); 4 RCTs with breast lesions (41,346); 4 RCTs with risk categories (13,965); 1 RCT with reproductive factors (10,101) Reduced risk for invasive cancer for tamoxifen for both premenopausal and postmenopausal women; tamoxifen and raloxifene for women with or without family history of breast cancer; raloxifene, anastrozole, and exemestane for all BMI categories

Tamoxifen and anastrozole had more effects for women with previous breast lesions (LCIS, ADH, ALH)

Risks were reduced for tamoxifen, raloxifene, and anastrozole in all modified Gail model risk categories and for raloxifene regardless of age at menarche, parity, or age at first live birth

Inconsistent; imprecise Trials not designed for subgroup comparisons, and analysis of differences between groups may be underpowered Low and insufficient High

Abbreviations: ADH, atypical ductal hyperplasia; ALH, atypical lobular hyperplasia; AUC, area under the receiver operating characteristic curve; BMI, body mass index; CHD, coronary heart disease; DVT, deep vein thrombosis; ER+, estrogen receptor–positive; ER−, estrogen receptor–negative; KQ, key question; LCIS, lobular carcinoma in situ; NA, not applicable; PE, pulmonary embolism; RCT, randomized clinical trial; RR, risk ratio.
a Per 1000 women over 5 years of use.

Return to Table of Contents