Abstract
Purpose
Treatment-emergent symptoms with adjuvant tamoxifen and aromatase inhibitors (AIs) have been associated with superior recurrence-free survival (RFS). We hypothesized that MA.27 anastrozole- or exemestane-treated patients with new or worsening vasomotor and/or joint symptoms would have improved RFS.
Patients and Methods
MA.27 randomly assigned 7,576 postmenopausal women with breast cancer to 5 years of anastrozole or exemestane. Patient-reported symptoms were collected using the Common Terminology Criteria for Adverse Events version 3.0 at protocol-specified baseline and 6- and 12-month clinical visits. Symptoms were considered present with either vasomotor and/or joint complaints. Associations between symptoms and baseline patient characteristics were examined with χ2 and Fisher's exact tests. Subsequent effects of new or worsening symptoms on RFS were examined with landmark analyses and stratified univariable and multivariable Cox models. We examined the effects of 3-month symptoms arising from unplanned clinic visits as a result of severe toxicity.
Results
Patients were assessable if eligible for the MA.27 trial, received some trial therapy, and had no disease recurrence at the end of a symptom assessment period; 96% of patients (n = 7,306 patients) were included at 6 months, and 96% (n = 7,246) were included at 12 months. Thirty-four percent of patients had baseline symptoms. For patients without baseline symptoms, 25% and 52% had new symptoms by 6 and 12 months, respectively. Neither treatment-emergent nor baseline symptoms significantly impacted RFS (P > .10) in patients with or without baseline symptoms.
Conclusion
In MA.27, anastrozole or exemestane treatment-emergent symptoms were not associated with improved RFS. Women should be supported through treatment and encouraged to remain on their AI regardless of their symptoms.
INTRODUCTION
An aromatase inhibitor (AI) as monotherapy for 5 years or for 3 years after 2 years of tamoxifen is standard adjuvant therapy for postmenopausal women with hormone receptor–positive invasive breast cancer.1 Approximately one in two women starting an AI will complain of early-onset new vasomotor or joint symptoms, often leading to treatment discontinuation.2–4
Several investigators have assessed the relationship between endocrine therapy–related symptoms and survival outcomes; however, results have been mixed. The Arimidex, Tamoxifen, Alone or in Combination (ATAC) trial investigators reported that new vasomotor or joint symptoms within 3 months of initiating tamoxifen or anastrozole were associated with superior recurrence-free survival (RFS).5 In the Tamoxifen Exemestane Adjuvant Multinational (TEAM) trial, women were randomly assigned to exemestane for 5 years or tamoxifen for 2.5 to 3 years followed by exemestane to a total of 5 years. Patients with vasomotor and joint symptoms in the first year of TEAM participation had improved survival outcomes.6 Likewise, Breast International Group (BIG) 1-98 study investigators reported that patients assigned to monotherapy tamoxifen or letrozole arms with 3-month or 12-month emergent vasomotor or musculoskeletal symptoms had improved outcomes.7
The NCIC Clinical Trials Group (CTG) MA.27 trial was the first adjuvant comparison of two third-generation AIs, nonsteroidal anastrozole versus steroidal exemestane. During a median of 4.1 years of follow-up in MA.27, 31.6% of patients discontinued treatment as a result of adverse effects, concomitant disease, or study refusal, with joint pain being the most frequent reason.8–10 Neither of the AIs was superior in efficacy, and there were no significant differences in treatment adherence observed.8 We hypothesized that new or worsening vasomotor or joint symptoms would be associated with improved RFS. If treatment-emergent symptoms are indeed associated with improved breast cancer outcomes, clinicians and patients can use the information to make important decisions regarding persistence of chronic AI therapy.
PATIENTS AND METHODS
Study Design
NCIC CTG MA.27 (ClinicalTrials.gov identifier: NCT00066573) was an open-label, randomized phase III multinational trial,8 approved by health regulatory authorities and the institutional review boards of participating centers; patients provided informed consent. Between June 2003 and July 2008, 7,576 patients were randomly assigned 1:1 to exemestane 25 mg (n = 3,789) or anastrozole 1 mg (n = 3,787). A factorial design of celecoxib or placebo with concomitant prophylactic aspirin use (< 81 mg/d [yes, no]) was discontinued because of concerns of celecoxib cardiac toxicity after 1,622 patients were randomly assigned.11 Trial stratification factors were use of celecoxib, aspirin, and trastuzumab; lymph node status (negative, positive, or unknown); and prior adjuvant chemotherapy (yes or no). Analyses reported here used the final analysis database, with a median of 4.1 years of follow-up.
MA.27 was funded by the Canadian Cancer Society Research Institute, US National Cancer Institute, and Pfizer. This is a retrospective analysis of treatment-associated symptoms and outcomes. Data were collected, managed, and analyzed by the NCIC CTG. The authors vouch for the integrity of the data and are the sole writers of the article.
Study Population
MA.27 enrolled postmenopausal women with estrogen receptor– and/or progesterone receptor–positive primary invasive breast cancer within 3 to 12 weeks of completing their initial treatment.8 Prior hormones or steroids had to be discontinued ≥ 3 weeks before random assignment. Prior treatment with an AI or tamoxifen was not permitted, although raloxifene was allowed for bone health if discontinued ≥ 3 weeks before random assignment. Patients with normal bone mineral density T-scores better than −2.0 enrolled onto the MA.27B (Bone) substudy were not permitted bisphosphonate use.
We included here all eligible MA.27 patients who received at least one dose of protocol therapy, classified by treatment received. Patients randomly assigned to MA.27 included women with and without pre-existing vasomotor and/or joint symptom(s). We investigated symptoms in the following three groups: women without baseline vasomotor or grade 3 or 4 joint symptoms who developed symptoms of any grade of vasomotor or grade 3 or 4 joint symptoms (group 1); women without baseline vasomotor or joint symptoms who developed symptoms of any grade of new vasomotor or joint symptoms (group 2, a subgroup of group 1); and all women, regardless of baseline symptoms, who developed new or worsening vasomotor and/or joint symptoms (group 3, which includes groups 1 and 2).
Adverse Events
Patient-reported symptoms were collected using the Common Terminology Criteria for Adverse Events (CTCAE) version 3.0. Vasomotor symptoms included hot flushes or sweating. Joint symptoms included joint pain, muscle pain, bone pain, arthritis, diminished joint function, or other musculoskeletal problems.9
Symptoms were recorded at baseline, at protocol-mandated 6- and 12-month treatment follow-up, annually thereafter, and on treatment case report forms any time after random assignment, when a clinical visit was deemed necessary. Three-month follow-up, 91 days from random assignment, was not mandated, and adverse events (AEs) if noted, were reported on a follow-up form or on an off-treatment report. The 3-month time point corresponded to that of the ATAC investigation and would represent in MA.27 (unacceptable) early toxicity reported before the first scheduled follow-up; early toxicity reports were included in the MA.27 genome-wide association studies of the genetic basis for musculoskeletal events.9
Study End Points
The primary end point in MA.27 was event-free survival (EFS), defined as time from random assignment to time of locoregional or distant recurrence, new primary breast cancer, or death. Other studies that looked at the relationship between new or worsening symptoms with outcomes used recurrence or RFS as the end point. Therefore, we used RFS as the end point for this current investigation, defined as time from random assignment to time of locoregional or distant disease recurrence or contralateral breast cancer. Five breast cancer deaths without prior recurrence were excluded. Patients were censored at death or longest follow-up.
Statistical Analyses
MA.27 baseline patient characteristics are listed in Table 1 and are described for each of the three subgroups of women in the analysis. Differences among the three subgroups were examined by χ2 or Fisher's exact tests.
Table 1.
Baseline Characteristics of Women by Vasomotor and/or Joint Symptoms at Baseline and 12 Months
| Variable | No Baseline Symptoms* |
With or Without Baseline Symptoms† |
||||||||
|---|---|---|---|---|---|---|---|---|---|---|
| No Vasomotor or Joint Symptom at 12 Months‡ (n = 3,458) |
Vasomotor or Joint Symptom at 12 Months‡ (n = 2,115) |
P§ | No New or Worsening Vasomotor or Joint Symptom at 12 Months‖ (n = 3,412) |
New or Worsening Vasomotor or Joint Symptom at 12 Months‖ (n = 3,834) |
P§ | |||||
| No. of Patients | % | No. of Patients | % | No. of Patients | % | No. of Patients | % | |||
| Age, years | .012 | .060 | ||||||||
| ≥ 50 or missing | 3,399 | 98.3 | 2,058 | 97.3 | 3,334 | 97.7 | 3,719 | 97.0 | ||
| < 50 | 59 | 1.7 | 57 | 2.7 | 78 | 2.3 | 115 | 3.0 | ||
| Race | < .001 | .008 | ||||||||
| Nonwhite | 238 | 6.9 | 95 | 4.5 | 217 | 6.4 | 189 | 4.9 | ||
| White | 3,220 | 93.1 | 2,020 | 95.5 | 3,195 | 93.6 | 3,645 | 95.1 | ||
| Performance status | .006 | .021 | ||||||||
| Other | 642 | 18.6 | 332 | 15.7 | 630 | 18.5 | 629 | 16.4 | ||
| Fully | 2,816 | 81.4 | 1,783 | 84.3 | 2,782 | 81.5 | 3,205 | 83.6 | ||
| Postmenopausal status | < .001 | < .001 | ||||||||
| Other | 3,314 | 95.8 | 1,964 | 92.9 | 3,228 | 94.6 | 3,516 | 91.7 | ||
| ≤ 55 years old | 54 | 1.6 | 80 | 3.8 | 79 | 2.3 | 168 | 4.4 | ||
| 45-59 years old (< 12 months; HRT) | 25 | 0.7 | 23 | 1.1 | 23 | 0.7 | 51 | 1.3 | ||
| 45-59 years old (< 12 months; hysterectomy) | 65 | 1.9 | 48 | 2.3 | 82 | 2.4 | 99 | 2.6 | ||
| Most extensive surgery | .230 | |||||||||
| Partial mastectomy or missing | 2,327 | 67.3 | 1,456 | 68.8 | 2,312 | 67.8 | 2,634 | 68.7 | .391 | |
| Mastectomy NOS | 1,131 | 32.7 | 659 | 31.2 | 1,100 | 32.2 | 1,200 | 31.3 | ||
| Axillary dissection | .682 | .251 | ||||||||
| Yes or missing | 1,751 | 50.6 | 1,059 | 50.1 | 1,732 | 50.8 | 1,998 | 52.1 | ||
| No | 1,707 | 49.4 | 1,056 | 49.9 | 1,680 | 49.2 | 1,836 | 47.9 | ||
| Tumor laterality | .889 | |||||||||
| Right | 1,679 | 48.6 | 1,031 | 48.7 | 1,670 | 48.9 | 1,835 | 47.9 | .357 | |
| Bilateral, left, and missing | 1,779 | 51.4 | 1,084 | 51.3 | 1,742 | 51.1 | 1,999 | 52.1 | ||
| T stage | .005 | .138 | ||||||||
| T1 | 2,425 | 70.1 | 1,558 | 73.7 | 2,434 | 71.3 | 2,795 | 72.9 | ||
| Other | 1,033 | 29.9 | 557 | 26.3 | 978 | 28.7 | 1,039 | 27.1 | ||
| N stage | < .001 | .995 | ||||||||
| Other | 1,041 | 30.1 | 542 | 25.6 | 960 | 28.1 | 1,079 | 28.1 | ||
| N0 | 2,417 | 69.9 | 1,573 | 74.4 | 2,452 | 71.9 | 2,755 | 71.9 | ||
| Prior adjuvant chemotherapy | .282 | .004 | ||||||||
| Yes | 1,031 | 29.8 | 602 | 28.5 | 984 | 28.8 | 1,226 | 32.0 | ||
| No or missing | 2,427 | 70.2 | 1,513 | 71.5 | 2,428 | 71.2 | 2,608 | 68.0 | ||
| Prior raloxifene | .555 | .273 | ||||||||
| Yes | 56 | 1.6 | 30 | 1.4 | 56 | 1.6 | 51 | 1.3 | ||
| No, missing, unknown | 3,402 | 98.4 | 2,085 | 98.6 | 3,356 | 98.4 | 3,783 | 98.7 | ||
| Prior or concurrent radiotherapy | .415 | .152 | ||||||||
| Yes | 2,412 | 69.8 | 1,497 | 70.8 | 2,396 | 70.2 | 2,751 | 71.8 | ||
| No, missing, unknown | 1,046 | 30.2 | 618 | 29.2 | 1,016 | 29.8 | 1,083 | 28.2 | ||
| Fractures within 10 years before allocation | .008 | .295 | ||||||||
| Yes | 355 | 10.3 | 172 | 8.1 | 349 | 10.2 | 364 | 9.5 | ||
| No, missing, unknown | 3,103 | 89.7 | 1,943 | 91.9 | 3,063 | 89.8 | 3,470 | 90.5 | ||
| Cardiovascular history | .014 | .006 | ||||||||
| Yes | 1,977 | 57.2 | 1,138 | 53.8 | 1,893 | 55.5 | 2,003 | 52.2 | ||
| No, missing, unknown | 1,481 | 42.8 | 977 | 46.2 | 1,519 | 44.5 | 1,831 | 47.8 | ||
Abbreviations: HRT, hormone replacement therapy; NOS, not otherwise specified.
Patients had no baseline vasomotor symptoms or grade 3 or 4 joint symptoms (group 1).
Patients did or did not have any baseline vasomotor and/or joint symptoms (group 3).
Patients did or did not have any grade of vasomotor symptoms or grade 3 or 4 joint symptoms (group 1).
P values from a χ2 or Fisher's exact test.
Patients did or did not have any grade of new or worsening vasomotor and/or joint symptoms (group 3).
To examine the effects of new or worsening symptoms on subsequent RFS, we used landmark analyses, which excluded patients who had already experienced a recurrence, and looked at the effect of having or not having symptoms in the initial 3, 6, or 12 months after random assignment on subsequent RFS. We used stratified Cox models for univariable and multivariable assessments, where stratification was by the trial stratification factors. Kaplan-Meier curves were used to illustrate the univariable RFS experience for women who did, or did not, have symptoms. Exploratory multivariable models had forced inclusion of trial therapy and presence of vasomotor/joint symptoms with stepwise addition of multivariable significant (P ≤ .05) baseline patient characteristics. Hazard ratios (HR) and 95% CIs were reported.
RESULTS
MA.27 randomly assigned 7,576 patients; the median follow-up time was 4.1 years for both the overall study population and the patients included in this analysis. Of the 7,576 patients, 7,344 (97%) were eligible for MA.27, received at least one dose of trial therapy, and were alive without relapse at 3 months, with 2,467 patients (34%) reporting baseline vasomotor and/or joint symptoms. At 6 months, 7,306 patients (96%) were evaluable, with 2,452 (34%) having baseline symptoms. At 12 months, 7,246 patients (96%) were evaluable, with 2,434 (34%) having baseline symptoms (Fig 1).
Fig 1.
CONSORT diagram for NCIC Clinical Trials Group MA.27. Group 1 patients are a subgroup of group 3; group 2 patients are a subgroup of group 1; and group 3 patients are all patients assessed for musculoskeletal (MSK) and vasomotor (VM) symptoms.
We evaluated the presence of vasomotor and/or joint symptoms in each the three groups of patients, as described earlier, at landmark time points. The occurrence or worsening of symptoms was similar in each group (reported as group 1, group 2, and group 3) at 6 months (19.2%, 25.4%, and 26.4%, respectively) and 12 months (38.0%, 52.9%, and 52.9%, respectively) and for early severe toxicity clinical visits at 3 months (3.9%, 5.2%, and 5.5%, respectively). At 3-month early clinic visits, 96 (24%) of 403 of patients with vasomotor and/or joint symptoms discontinued treatment. In preplanned visits at 6 and 12 months, the protocol therapy discontinuation rates were 12% and 10%, respectively. We did not observe a difference in adherence by way of time to protocol therapy discontinuation by treatment arm for patients who did not have symptoms by 12 months (HR of exemestane to anastrozole, 1.03; 95% CI, 0.93 to 1.14; P = .54; Fig 2). Likewise, we did not observe a difference in time to treatment discontinuation among patients who experienced new or worsening symptoms at 12 months (HR, 1.06; 95% CI, 0.97 to 1.15; P = .24; Fig 3).
Fig 2.

Time to treatment discontinuation in women without new or worsening symptoms by 12 months. HR, hazard ratio.
Fig 3.

Time to treatment discontinuation in women with new or worsening symptoms by 12 months. HR, hazard ratio.
We compared baseline patient characteristics by subsequent vasomotor and joint symptoms at 12 months (Table 1); 3- and 6-month data are listed in Appendix Tables A1 and A2, respectively (online only). In each table, we report baseline characteristics for patients who had no baseline vasomotor or grade 3 or 4 joint symptoms (group 1) and for patients who did or did not have baseline symptoms (group 3). Notable significant differences (P < .001) were seen by age, with proportionately more women ≤ 55 years old having symptoms. For each time period, the exclusion of patients with grade 1 or 2 joint symptoms (group 2) did not lead to significant differences in RFS among patients with or without symptoms (data not shown).
The development of new or worsening vasomotor and joint symptoms did not significantly impact RFS in univariable assessments, at any of the evaluated time points or for any of the three groups. Specifically, for group 1 patients without baseline vasomotor or grade 3 or 4 symptoms, 3-month HR was 0.62 (95% CI, 0.33 to 1.18; P = .15; Appendix Fig A1, online only), 6-month HR was 0.80 (95% CI, 0.59 to 1.07; P = .13; Fig 4), and 12 month HR was 0.81 (95% CI, 0.63 to 1.03; P = .09; Fig 5). RFS for patients with or without baseline symptoms who subsequently developed new or worsening symptoms (group 3) at 6 and 12 months are depicted in corresponding Appendix Figures A2, A3, and A4 (online only). Likewise, in all patients (group 3), new or worsening symptoms alone did not significantly impact RFS at 3 (P = .84), 6 (P = .10), or 12 months (P = .60).
Fig 4.

Recurrence-free survival of group 1 patients without baseline vasomotor (vaso) or grade 3 or 4 joint symptoms who developed vasomotor or grade 3 or 4 joint symptoms at 6 months. HR, hazard ratio.
Fig 5.

Recurrence-free survival of group 1 patients without baseline vasomotor (vaso) or grade 3 or 4 joint symptoms who developed vasomotor or grade 3 or 4 joint symptoms at 12 months. HR, hazard ratio.
Multivariable models were stratified by use of celecoxib, aspirin, and trastuzumab and by lymph node status and prior chemotherapy. Neither treatment nor new or worsening vasomotor or joint symptoms significantly impacted RFS (P > .10) regardless of presence or absence of baseline symptoms. In all scenarios, RFS was significantly worse (P < .001), as expected, for women with T2, T3, or Tx lesions compared with those with T1 lesions and for patients who had no baseline vasomotor or grade 3 or 4 joint symptoms, at 3 months (HR, 2.22; 95% CI, 1.76 to 2.81), 6 months (HR, 2.22; 95% CI, 1.74 to 2.82), and 12 months (HR, 2.24; 95% CI, 1.68 to 2.97).
DISCUSSION
Adjuvant endocrine therapy for 5 to 10 years is recommended for most postmenopausal women with hormone receptor–positive breast cancer. The ability to predict survival outcomes based on baseline or treatment-emergent endocrine symptoms would be clinically important. Retrospective exploratory analyses of the ATAC, TEAM, and BIG 1-98 clinical trials suggested that treatment-emergent symptoms may be associated with superior survival outcomes. In contrast, our analysis of more than 7,000 MA.27 patients demonstrated no relationship between baseline or new or worsening treatment-associated symptoms and RFS (P > .10).
Compared with other investigations, a high proportion of evaluable MA.27 participants (34%) experienced baseline CTCAE vasomotor and/or joint symptoms, which may have been in part a result of a required 3-week washout period for hormone replacement therapy (HRT). HRT withdrawal is frequently associated with the appearance of rebound symptoms and improved breast cancer outcomes, which may explain the disparate results between the ATAC analysis and our study.12 The effects of withdrawal generally occur promptly and would be recorded among baseline symptoms, allowing for independent assessment of the effect of AI therapy on symptoms.12 In ATAC, which did not require an HRT washout period before random assignment, only 9.5% and 9.3% of patients who started treatment had baseline vasomotor and joint symptoms, respectively. In the TEAM trial, baseline symptom data and prior HRT use were not described.6 Approximately 18% of BIG 1-98 patients used HRT within 3 months of initiating study, and 18% of additional patients used HRT more than 3 months before the study.7
Only the 3-month symptom data were investigated in ATAC, and 55% of patients receiving either anastrozole or tamoxifen developed symptoms by 3 months. Among these ATAC participants, the emergence of vasomotor symptoms was particularly pronounced in those who had received prior HRT (P = .003). In MA.27, patient-perceived severe symptoms led to discontinuation of treatment in 24% of symptomatic participants at 3-month early clinic visits. At the preplanned 6- and 12-month visits, the discontinuation rates for those with symptoms were 12% and 10%, respectively. Prior HRT use up until, or near, the diagnosis of breast cancer could have confounded our results.
Although we did not observe the ATAC differences in cancer outcomes related to treatment-emergent symptoms, there were also important differences in the proportion of trial patients investigated. In MA.27, we investigated the symptoms for 96% of participants. Of the 6,241 ATAC patients randomly assigned to anastrozole or tamoxifen, 3,964 patients (64%) were included in the analysis, with the exclusion of patients who did not start therapy, did not have hormone receptor–positive tumors, and had prior baseline symptoms. Because inclusivity of patients was high for the TEAM (9,325 patients, 95%) and BIG 1-98 (4,798 patients, 97.5%) trials, inclusivity alone is not likely to account for intertrial differences.
An important aspect in interpreting discrepancies between MA.27 and other reports from adjuvant trials could be the method of ascertaining AEs. Patient-reported symptoms were collected for MA.27 in a standardized manner, using CTCAE version 3.0. The ATAC and TEAM trials reported using elicited symptoms or patient responses, respectively, rather than a specific/standard symptom checklist, although in the TEAM trial, the severity of symptoms was determined with National Cancer Institute Common Toxicity Criteria criteria. The BIG 1- 98 trial used CTCAE version 2.0 and reported only 38% of patients experiencing symptoms by 12 months. The prospective incorporation not only of standard AE reporting, such as the CTCAE version 3.0, but also of subjective patient-reported outcomes will allow for accurate determination of symptoms both in future trials and in clinical practice.13,14
The effects of arthralgic pain medications may have confounded results. In MA.27, we adjusted for the effects of early random assignment to celecoxib, with concomitant recording of aspirin use (81 mg/d), during the factorial period of MA.27 with stratified analyses. Details of aspirin use and use of other nonsteroidal anti-inflammatory drugs (NSAIDs), which might have been prescribed for symptoms, were not rigorously collected in ATAC; however, 12% of patients reported the use of cyclooxygenase-2 inhibitors. Use of NSAIDs was not reported in the TEAM or BIG 1-98 trials. The use of NSAIDS and cyclooxygenase-2 inhibitors in the ATAC trial could have been directed toward controlling symptoms, which may have improved adherence.
Adherence to endocrine therapy is an important component of efficacy. Overall, 31.6% of MA.27 patients discontinued treatment as a result of AEs, concomitant disease, or study refusal, with joint pain being the most frequent reason.9,13 Interestingly, despite a higher proportion of symptomatic patients in ATAC compared with MA.27, ATAC participants had a higher adherence to therapy. Specifically in ATAC, 88% of patients with symptoms and 84% without symptoms adhered to prescribed trial therapy until recurrence, compared with 70% of MA.27 participants. Although the TEAM investigators did not report treatment adherence, they performed a time-dependent Cox regression analysis for time on and off treatment. Early treatment discontinuation did not affect outcomes of patients reporting specific AEs. Moreover, patients who reported specific AEs and discontinued treatment before the designated 5-year stop date still experienced a disease-free survival benefit compared with patients who did not. Adherence to therapy was high in BIG 1-98, approaching 97%.
We recently evaluated the role of emergent symptoms in 672 premenopausal women randomly assigned to tamoxifen or placebo in the MA.12 trial. With a median follow-up time of 9.7 years, we observed only a weak association (P = .06) that women older than 50 years had more symptoms on tamoxifen; symptoms did not impact RFS (P > .90).15 Women reporting new hot flushes on tamoxifen in the Women's Healthy Eating and Living Study were more likely to benefit from tamoxifen. The authors of the Women's Healthy Eating and Living Study concluded that anti–estrogen-emergent symptoms were associated with superior therapeutic effects and improved breast cancer outcomes.
The mechanism underlying AI-associated musculoskeletal symptoms remains unknown and may include estrogen suppression and pharmacogenetic or genetic factors.16–18 Adverse effects may also be influenced by drug adherence or concomitant use of interventions alleviating symptoms.17,18 In the MA.27 genome-wide association studies of the genetic basis for musculoskeletal AEs,13 we included patients who experienced unacceptable early toxicity before the first scheduled 6-month follow-up and identified four single-nucleotide polymorphisms in the estrogen-dependent gene, T-cell leukemia (TCL1A), related to expression of cytokine interleukin-17, associated with musculoskeletal symptoms in women treated with anastrozole or exemestane.9 Another study suggested that a variant in the gene encoding for the estrogen receptor may predict AI-associated symptoms.19 Other factors may include body mass index and prior chemotherapy use, especially taxanes.2,4,20 Specific type of adjuvant chemotherapy was not recorded in MA.27; however, different agents should be approximately balanced between the two arms and are not expected to affect outcome.
A limitation of the treatment-emergent analysis of the MA.27 trial is that the original factorial design randomly assigned patients to celecoxib with stratification by aspirin use (81 mg); however, the effects of these factors and the other possible confounders of adjuvant chemotherapy and lymph node involvement were handled with stratified univariable and multivariable results. In addition, approximately 30% of MA.27 patients discontinued trial therapy by final trial follow-up at a median of 4.1 years, which may have impacted our assessment of symptoms. MA.27 collected only severe symptoms during the first 3 months; however, results for this time period were similar to those obtained at protocol-mandated follow-up at 6 and 12 months. In addition, MA.27 patients had high EFS (4-year EFS, 91% on exemestane and 91.2% on anastrozole), which would have made it more difficult to demonstrate a difference in RFS with landmark analyses where events are excluded.
In summary, the majority of studies of patients who received tamoxifen or AI reported a relationship between emergence of vasomotor and musculoskeletal symptoms and survival outcomes. Our study, examining two AIs, failed to show an association between new or worsening symptoms and outcome. Predictive factors are required to determine treatment efficacy and treatment-emergent symptoms. Future prospective analyses of symptoms in AI trials should ensure that symptoms are collected in a similar way to both the positive and negative trials to resolve the issue. Until then, treatment-emergent symptoms should be managed as effectively as possible to support treatment continuation, and it is premature to counsel patients on whether to continue or change their adjuvant AI therapy based on symptoms.
Supplementary Material
Acknowledgment
Presented, in part, at the 32nd Annual San Antonio Breast Cancer Symposium, December 10-13, 2009, San Antonio, TX, and the 47th Annual Meeting of the American Society of Clinical Oncology, June 3-7, 2011, Chicago, IL.
Glossary Terms
- anastrozole:
a third-generation nonsteroidal aromatase inhibitor that prevents the conversion of androgen to estrogen in the peripheral tissues in postmenopausal women. Because hormone-dependent breast cancer progresses with estrogen, anastrozole has been used in the treatment of breast cancer in postmenopausal women. See aromatase inhibitors.
- aromatase inhibitors:
inhibitors used in treating breast cancer in postmenopausal women. Aromatase inhibitors inhibit the conversion of androgens to estrogens by the enzyme aromatase, thus depriving the tumor of estrogenic signals. Because of decreased production of estrogen, estrogen receptors, which are important in the progression of breast cancer, cannot be activated.
Appendix
Table A1.
Baseline Characteristics of Women by Vasomotor and/or Joint Symptoms at Baseline and 3 Months
| Variable | No Baseline Symptoms* |
With or Without Baseline Symptoms† |
||||||||
|---|---|---|---|---|---|---|---|---|---|---|
| No Vasomotor or Joint Symptom at 3 Months‡ (n = 5,424) |
Vasomotor or Joint Symptom at 3 Months‡ (n = 221) |
P§ | No New or Worsening Vasomotor or Joint Symptom at 3 Months‖ (n = 6,931) |
New or Worsening Vasomotor or Joint Symptom at 3 Months‖ (n = 403) |
P§ | |||||
| No. of Patients | % | No. of Patients | % | No. of Patients | % | No. of Patients | % | |||
| Age, years | .807 | .172 | ||||||||
| ≥ 50 or missing | 5,312 | 97.9 | 216 | 97.7 | 6,751 | 97.4 | 388 | 96.3 | ||
| < 50 | 112 | 2.1 | 5 | 2.3 | 180 | 2.6 | 15 | 3.7 | ||
| Race | .388 | .945 | ||||||||
| Nonwhite | 317 | 5.8 | 16 | 7.2 | 384 | 5.5 | 22 | 5.5 | ||
| White | 5,107 | 94.2 | 205 | 92.8 | 6,547 | 94.5 | 381 | 94.5 | ||
| Performance status | .049 | .121 | ||||||||
| Other | 966 | 17.8 | 28 | 12.7 | 1,224 | 17.7 | 59 | 14.6 | ||
| Fully | 4,458 | 82.2 | 193 | 87.3 | 5,707 | 82.3 | 344 | 85.4 | ||
| Postmenopausal status | .002 | < .001 | ||||||||
| Other | 5,143 | 94.8 | 205 | 92.8 | 6,465 | 93.3 | 362 | 89.8 | ||
| ≤ 55 years old | 121 | 2.2 | 14 | 6.3 | 213 | 3.1 | 35 | 8.7 | ||
| 45-59 years old (< 12 months; HRT) | 47 | 0.9 | 1 | 0.5 | 73 | 1.1 | 2 | 0.5 | ||
| 45-59 years old (< 12 months; hysterectomy) | 113 | 2.1 | 1 | 0.5 | 180 | 2.6 | 4 | 1.0 | ||
| Most extensive surgery | .253 | .054 | ||||||||
| Partial mastectomy or missing | 3,684 | 67.9 | 142 | 64.3 | 4,739 | 68.4 | 257 | 63.8 | ||
| Mastectomy NOS | 1,740 | 32.1 | 79 | 35.7 | 2,192 | 31.6 | 146 | 36.2 | ||
| Axillary dissection | .009 | < .001 | ||||||||
| Yes or missing | 2,732 | 50.4 | 131 | 59.3 | 3,545 | 51.1 | 248 | 61.5 | ||
| No | 2,692 | 49.6 | 90 | 40.7 | 3,386 | 48.9 | 155 | 38.5 | ||
| Tumor laterality | .857 | .521 | ||||||||
| Right | 2,635 | 48.6 | 106 | 48.0 | 3,343 | 48.2 | 201 | 49.9 | ||
| Bilateral, left, and missing | 2,789 | 51.4 | 115 | 52.0 | 3,588 | 51.8 | 202 | 50.1 | ||
| T stage | .017 | .764 | ||||||||
| T1 | 3,843 | 70.9 | 173 | 78.3 | 4,984 | 71.9 | 287 | 71.2 | ||
| Other | 1,581 | 29.1 | 48 | 21.7 | 1,947 | 28.1 | 116 | 28.8 | ||
| N stage | .700 | .231 | ||||||||
| Other | 1,562 | 28.8 | 61 | 27.6 | 1,958 | 28.2 | 125 | 31.0 | ||
| N0 | 3,862 | 71.2 | 160 | 72.4 | 4,973 | 71.8 | 278 | 69.0 | ||
| Prior adjuvant chemotherapy | .882 | .048 | ||||||||
| Yes | 1,596 | 29.4 | 64 | 29.0 | 2,101 | 30.3 | 141 | 35.0 | ||
| No or missing | 3,828 | 70.6 | 157 | 71.0 | 4,830 | 69.7 | 262 | 65.0 | ||
| Prior raloxifene | .399 | .687 | ||||||||
| Yes | 84 | 1.5 | 5 | 2.3 | 103 | 1.5 | 7 | 1.7 | ||
| No, missing, unknown | 5,340 | 98.5 | 216 | 97.7 | 6,828 | 98.5 | 396 | 98.3 | ||
| Prior or concurrent radiotherapy | .294 | .138 | ||||||||
| Yes | 3,811 | 70.3 | 148 | 67.0 | 4,934 | 71.2 | 273 | 67.7 | ||
| No, missing, unknown | 1,613 | 29.7 | 73 | 33.0 | 1,997 | 28.8 | 130 | 32.3 | ||
| Fractures within 10 years before allocation | .171 | .258 | ||||||||
| Yes | 517 | 9.5 | 15 | 6.8 | 687 | 9.9 | 33 | 8.2 | ||
| No, missing, unknown | 4,907 | 90.5 | 206 | 93.2 | 6,244 | 90.1 | 370 | 91.8 | ||
| Cardiovascular history | .596 | .096 | ||||||||
| Yes | 3,043 | 56.1 | 120 | 54.3 | 3,752 | 54.1 | 201 | 49.9 | ||
| No, missing, unknown | 2,381 | 43.9 | 101 | 45.7 | 3,179 | 45.9 | 202 | 50.1 | ||
Abbreviations: HRT, hormone replacement therapy; NOS, not otherwise specified.
Patients had no baseline vasomotor symptoms or grade 3 or 4 joint symptoms (group 1).
Patients did or did not have any baseline vasomotor and/or joint symptoms (group 3).
Patients did or did not have any grade of vasomotor symptoms or grade 3 or 4 joint symptoms (group 1).
P values from a χ2 or Fisher's exact test.
Patients did or did not have any grade of new or worsening vasomotor and/or joint symptoms (group 3).
Table A2.
Baseline Characteristics of Women by Vasomotor and/or Joint Symptoms at Baseline and 6 Months
| Variable | No Baseline Symptoms* |
With or Without Baseline Symptoms† |
||||||||
|---|---|---|---|---|---|---|---|---|---|---|
| No Vasomotor or Joint Symptom at 6 Months‡ (n = 4,539) |
Vasomotor or Joint Symptom at 6 Months‡ (n = 1,081) |
P§ | No New or Worsening Vasomotor or Joint Symptom at 6 Months‖ (n = 5,379) |
New or Worsening Vasomotor or Joint Symptom at 6 Months‖ (n = 1,927) |
P§ | |||||
| No. of Patients | % | No. of Patients | % | No. of Patients | % | No. of Patients | % | |||
| Age, years | .287 | .009 | ||||||||
| ≥ 50 or missing | 4,449 | 98.0 | 1,054 | 97.5 | 5,252 | 97.6 | 1,860 | 96.5 | ||
| < 50 | 90 | 2.0 | 27 | 2.5 | 127 | 2.4 | 67 | 3.5 | ||
| Race | .561 | .292 | ||||||||
| Nonwhite | 273 | 6.0 | 60 | 5.6 | 308 | 5.7 | 98 | 5.1 | ||
| White | 4,266 | 94.0 | 1,021 | 94.4 | 5,071 | 94.3 | 1,829 | 94.9 | ||
| Performance status | .072 | .050 | ||||||||
| Other | 819 | 18.0 | 170 | 15.7 | 969 | 18.0 | 309 | 16.0 | ||
| Fully | 3,720 | 82.0 | 911 | 84.3 | 4,410 | 82.0 | 1,618 | 84.0 | ||
| Postmenopausal status | < .001 | < .001 | ||||||||
| Other | 4,328 | 95.4 | 995 | 92.0 | 5,061 | 94.1 | 1,738 | 90.2 | ||
| ≤ 55 years old | 75 | 1.7 | 60 | 5.6 | 128 | 2.4 | 120 | 6.2 | ||
| 45-59 years old (< 12 months; HRT) | 39 | 0.9 | 9 | 0.8 | 54 | 1.0 | 21 | 1.1 | ||
| 45-59 years old (< 12 months; hysterectomy) | 97 | 2.1 | 17 | 1.6 | 136 | 2.5 | 48 | 2.5 | ||
| Most extensive surgery | .815 | .377 | ||||||||
| Partial mastectomy or missing | 3,082 | 67.9 | 730 | 67.5 | 3,682 | 68.5 | 1,298 | 67.4 | ||
| Mastectomy NOS | 1,457 | 32.1 | 351 | 32.5 | 1,697 | 31.5 | 629 | 32.6 | ||
| Axillary dissection | .079 | < .001 | ||||||||
| Yes or missing | 2,271 | 50.0 | 573 | 53.0 | 2,702 | 50.2 | 1,072 | 55.6 | ||
| No | 2,268 | 50.0 | 508 | 47.0 | 2,677 | 49.8 | 855 | 44.4 | ||
| Tumor laterality | .339 | .323 | ||||||||
| Right | 2,219 | 48.9 | 511 | 47.3 | 2,619 | 51.3 | 913 | 47.4 | ||
| Bilateral, left, and missing | 2,320 | 51.1 | 570 | 52.7 | 2,760 | 51.3 | 1,014 | 52.6 | ||
| T stage | .020 | .681 | ||||||||
| T1 | 3,202 | 70.5 | 801 | 74.1 | 3,862 | 71.8 | 1,393 | 72.3 | ||
| Other | 1,337 | 29.5 | 280 | 25.9 | 1,517 | 28.2 | 534 | 27.7 | ||
| N stage | .036 | .791 | ||||||||
| Other | 1,330 | 29.3 | 282 | 26.1 | 1,521 | 28.3 | 551 | 28.6 | ||
| N0 | 3,209 | 70.7 | 799 | 73.9 | 3,858 | 71.7 | 1,376 | 71.4 | ||
| Prior adjuvant chemotherapy | .885 | < .001 | ||||||||
| Yes | 1,337 | 29.5 | 316 | 29.2 | 1,582 | 29.4 | 652 | 33.8 | ||
| No or missing | 3,202 | 70.5 | 765 | 70.8 | 3,797 | 70.6 | 1,275 | 66.2 | ||
| Prior raloxifene | .770 | .702 | ||||||||
| Yes | 70 | 1.5 | 18 | 1.7 | 82 | 1.5 | 27 | 1.4 | ||
| No, missing, unknown | 4,469 | 98.5 | 1,063 | 98.3 | 5,297 | 98.5 | 1,900 | 98.6 | ||
| Prior or concurrent radiotherapy | .846 | .730 | ||||||||
| Yes | 3,188 | 70.2 | 756 | 69.9 | 3,827 | 71.1 | 1,363 | 70.7 | ||
| No, missing, unknown | 1,351 | 29.8 | 325 | 30.1 | 1,552 | 28.9 | 564 | 29.3 | ||
| Fractures within 10 years before allocation | .106 | .511 | ||||||||
| Yes | 442 | 9.7 | 88 | 8.1 | 536 | 10.0 | 182 | 9.4 | ||
| No, missing, unknown | 4,097 | 90.3 | 993 | 91.9 | 4,843 | 90.0 | 1,745 | 90.6 | ||
| Cardiovascular history | .004 | .009 | ||||||||
| Yes | 2,581 | 56.9 | 563 | 52.1 | 2,944 | 54.7 | 988 | 51.3 | ||
| No, missing, unknown | 1,958 | 43.1 | 518 | 47.9 | 2,435 | 45.3 | 939 | 48.7 | ||
Abbreviations: HRT, hormone replacement therapy; NOS, not otherwise specified.
Patients had no baseline vasomotor symptoms or grade 3 or 4 joint symptoms (group 1).
Patients did or did not have any baseline vasomotor and/or joint symptoms (group 3).
Patients did or did not have any grade of vasomotor symptoms or grade 3 or 4 joint symptoms (group 1).
P values from a χ2 or Fisher's exact test.
Patients did or did not have any grade of new or worsening vasomotor and/or joint symptoms (group 3).
Fig A1.

Recurrence-free survival of group 1 patients without baseline vasomotor (vaso) or grade 3 or 4 joint symptoms who developed vasomotor or grade 3 or 4 joint symptoms at 3 months. HR, hazard ratio.
Fig A2.

Recurrence-free survival of group 3 patients with new or worsening vasomotor (vaso) or joint symptoms at 3 months. HR, hazard ratio.
Fig A3.

Recurrence-free survival of group 3 patients (women) with new or worsening vasomotor (vaso) or joint symptoms at 6 months. HR, hazard ratio.
Fig A4.

Recurrence-free survival of group 3 patients (women) with new or worsening vasomotor (vaso) or joint symptoms at 12 months. HR, hazard ratio.
See accompanying editorial on page 235
Support information appears at the end of this article.
Clinical trial information: NCT00066573.
Terms in blue are defined in the glossary, found at the end of this article and online at www.jco.org.
Authors' disclosures of potential conflicts of interest are found in the article online at www.jco.org. Author contributions are found at the end of this article.
Support
Supported by the NCIC Clinical Trials Group through grant support from the Canadian Cancer Society Research Institute, the US National Cancer Institute, the International Breast Cancer Study Group, and Pfizer. V.S. is supported in part by the Susan G. Komen for the Cure. P.E.G. is supported by the Avon Foundation New York.
AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
Disclosures provided by the authors are available with this article at www.jco.org.
AUTHOR CONTRIBUTIONS
Conception and design: Vered Stearns, Judith-Anne W. Chapman, Cynthia X. Ma, Matthew J. Ellis, Kathleen I. Pritchard, G. Thomas Budd, George W. Sledge, Lois E. Shepherd, Paul E. Goss
Provision of study materials or patients: Lois E. Shepherd, Paul E. Goss
Collection and assembly of data: Vered Stearns, Judith-Anne W. Chapman, Matthew J. Ellis, Jessica Kundapur, Lois E. Shepherd, Paul E. Goss
Data analysis and interpretation: Vered Stearns, Judith-Anne W. Chapman, James N. Ingle, G. Thomas Budd, Manuela Radaglio, George W. Sledge, Aurélie Le Maitre, Pedro E.R. Liedke, Lois E. Shepherd, Paul E. Goss
Manuscript writing: All authors
Final approval of manuscript: All authors
AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
Treatment-Associated Musculoskeletal and Vasomotor Symptoms and Relapse-Free Survival in the NCIC CTG MA.27 Adjuvant Breast Cancer Aromatase Inhibitor Trial
The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or jco.ascopubs.org/site/ifc.
Vered Stearns
Research Funding: Abbvie, Celgene, Merck, Novartis, Pfizer, MedImmune, Puma
Judith-Anne W. Chapman
No relationship to disclose
Cynthia X. Ma
Consulting or Advisory Role: Novartis
Research Funding: Novartis (Inst), Puma (Inst), Pfizer (Inst)
Travel, Accommodations, Expenses: Novartis
Matthew J. Ellis
Consulting or Advisory Role: AstraZeneca, Pfizer, Novartis, Genentech, Puma
Research Funding: AstraZeneca, Pfizer
Patents, Royalties, Other Intellectual Property: Bioclassifier LLC
James N. Ingle
No relationship to disclose
Kathleen I. Pritchard
Honoraria: Novartis, Roche/Genentech, GlaxoSmithKline, AstraZeneca
Consulting or Advisory Role: Roche/Genentech, Novartis, GlaxoSmithKline, Pfizer
Research Funding: Novartis (Inst), AstraZeneca (Inst), Roche/Genentech (Inst), Genomic Health (Inst)
G. Thomas Budd
Research Funding: Genentech (Inst), Threshold Pharmaceuticals (Inst), MabVax Therapeutics (Inst), ZIOPHARM Oncology (Inst)
Manuela Radaglio
No relationship to disclose
George W. Sledge
Leadership: Syndax
Stock or Other Ownership: Syndax
Honoraria: Genentech, Symphogen
Consulting or Advisory Role: Symphogen
Research Funding: Roche/Genentech
Aurélie Le Maitre
No relationship to disclose
Jessica Kundapur
No relationship to disclose
Pedro E.R. Liedke
Consulting or Advisory Role: Novartis
Travel, Accommodations, Expenses: Novartis, Roche
Lois E. Shepherd
No relationship to disclose
Paul E. Goss
No relationship to disclose
REFERENCES
- 1.Burstein HJ, Prestrud AA, Seidenfeld J, et al. American Society of Clinical Oncology clinical practice guideline: Update on adjuvant endocrine therapy for women with hormone receptor-positive breast cancer. J Clin Oncol. 2010;28:3784–3796. doi: 10.1200/JCO.2009.26.3756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Crew KD, Greenlee H, Capodice J, et al. Prevalence of joint symptoms in postmenopausal women taking aromatase inhibitors for early-stage breast cancer. J Clin Oncol. 2007;25:3877–3883. doi: 10.1200/JCO.2007.10.7573. [DOI] [PubMed] [Google Scholar]
- 3.Henry NL, Giles JT, Ang D, et al. Prospective characterization of musculoskeletal symptoms in early stage breast cancer patients treated with aromatase inhibitors. Breast Cancer Res Treat. 2008;111:365–372. doi: 10.1007/s10549-007-9774-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Mao JJ, Stricker C, Bruner D, et al. Patterns and risk factors associated with aromatase inhibitor-related arthralgia among breast cancer survivors. Cancer. 2009;115:3631–3639. doi: 10.1002/cncr.24419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Cuzick J, Sestak I, Cella D, et al. Treatment-emergent endocrine symptoms and the risk of breast cancer recurrence: A retrospective analysis of the ATAC trial. Lancet Oncol. 2008;9:1143–1148. doi: 10.1016/S1470-2045(08)70259-6. [DOI] [PubMed] [Google Scholar]
- 6.Fontein DB, Seynaeve C, Hadji P, et al. Specific adverse events predict survival benefit in patients treated with tamoxifen or aromatase inhibitors: An international tamoxifen exemestane adjuvant multinational trial analysis. J Clin Oncol. 2013;31:2257–2264. doi: 10.1200/JCO.2012.45.3068. [DOI] [PubMed] [Google Scholar]
- 7.Huober J, Cole BF, Rabaglio M, et al. Symptoms of endocrine treatment and outcome in the BIG 1-98 study. Breast Cancer Res Treat. 2014;143:159–169. doi: 10.1007/s10549-013-2792-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Goss PE, Ingle JN, Pritchard KI, et al. Exemestane versus anastrozole in postmenopausal women with early breast cancer: NCIC CTG MA. 27—A randomized controlled phase III trial. J Clin Oncol. 2013;31:1398–1404. doi: 10.1200/JCO.2012.44.7805. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Ingle JN, Schaid DJ, Goss PE, et al. Genome-wide associations and functional genomic studies of musculoskeletal adverse events in women receiving aromatase inhibitors. J Clin Oncol. 2010;28:4674–4682. doi: 10.1200/JCO.2010.28.5064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Wagner LI, Zhao F, Chapman J-A, et al. Patient-reported predictors of early treatment discontinuation: NCIC JMA.27/E1Z03 quality of life study of postmenopausal women with primary breast cancer randomized to exemestane or anastrozole. Cancer Res. 2011;71:114a. doi: 10.1007/s10549-018-4713-2. (abstr S6-2) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Solomon SD, McMurray JJ, Pfeffer MA, et al. Cardiovascular risk associated with celecoxib in a clinical trial for colorectal adenoma prevention. N Engl J Med. 2005;352:1071–1080. doi: 10.1056/NEJMoa050405. [DOI] [PubMed] [Google Scholar]
- 12.Ockene JK, Barad DH, Cochrane BB, et al. Symptom experience after discontinuing use of estrogen plus progestin. JAMA. 2005;294:183–193. doi: 10.1001/jama.294.2.183. [DOI] [PubMed] [Google Scholar]
- 13.Fellowes D, Fallowfield LJ, Saunders CM, et al. Tolerability of hormone therapies for breast cancer: How informative are documented symptom profiles in medical notes for “well-tolerated” treatments? Breast Cancer Res Treat. 2001;66:73–81. doi: 10.1023/a:1010684903199. [DOI] [PubMed] [Google Scholar]
- 14.Oberguggenberger A, Hubalek M, Sztankay M, et al. Is the toxicity of adjuvant aromatase inhibitor therapy underestimated? Complementary information from patient-reported outcomes (PROs) Breast Cancer Res Treat. 2011;128:553–561. doi: 10.1007/s10549-011-1378-5. [DOI] [PubMed] [Google Scholar]
- 15.Chapman J-AW, Shepherd LE, Le Maitre A, et al. Effect of treatment emergent symptoms on relapse free survival: NCIC CTG MA.12 a randomized placebo-controlled trial of tamoxifen after adjuvant chemotherapy in pre-menopausal women in early breast cancer. Cancer Res. 2011;71 P1-06-08. (abstr) [Google Scholar]
- 16.Burstein HJ, Winer EP. Aromatase inhibitors and arthralgias: A new frontier in symptom management for breast cancer survivors. J Clin Oncol. 2007;25:3797–3799. doi: 10.1200/JCO.2007.11.9529. [DOI] [PubMed] [Google Scholar]
- 17.Henry NL, Giles JT, Stearns V. Aromatase inhibitor-associated musculoskeletal symptoms: Etiology and strategies for management. Oncology (Williston Park) 2008;22:1401–1408. [PubMed] [Google Scholar]
- 18.Liedke PE, Goss PE. Aromatase inhibitors and musculoskeletal adverse events. Lancet Oncol. 2012;13:333–334. doi: 10.1016/S1470-2045(11)70345-X. [DOI] [PubMed] [Google Scholar]
- 19.Henry NL, Skaar TC, Dantzer J, et al. Genetic associations with toxicity-related discontinuation of aromatase inhibitor therapy for breast cancer. Breast Cancer Res Treat. 2013;138:807–816. doi: 10.1007/s10549-013-2504-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Henry NL, Azzouz F, Desta Z, et al. Predictors of aromatase inhibitor discontinuation as a result of treatment-emergent symptoms in early-stage breast cancer. J Clin Oncol. 2012;30:936–942. doi: 10.1200/JCO.2011.38.0261. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.

