Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Aug 1.
Published in final edited form as: Breast Cancer Res Treat. 2016 Jul 22;159(1):109–118. doi: 10.1007/s10549-016-3918-5

Impact of Race, Ethnicity and BMI on Achievement of Pathologic Complete Response Following Neoadjuvant Chemotherapy for Breast Cancer: A Pooled Analysis of Four Prospective Alliance Clinical Trials (A151426)

Erica T Warner 1, Karla V Ballman 2, Carrie Strand 3, Judy Boughey 4, Aman U Buzdar 5, Lisa A Carey 6, William M Sikov 7, H Partridge 8,9
PMCID: PMC5011019  NIHMSID: NIHMS805454  PMID: 27449492

Abstract

Purpose

Previous studies demonstrated poor response to neoadjuvant systemic therapy (NST) for breast cancer among black women and women who are overweight or obese but this may be due to chemotherapy under dosing. We assessed associations of race, ethnicity and body mass index (BMI) with pathologic complete response (pCR) in clinical trial populations.

Methods

1797 women enrolled in four NST trials (CALGB 40601, 40603; ACOSOG Z1041, Z1071) were included. Tumor subtypes were defined by estrogen receptor (ER) and HER2 status. Logistic regression generated odds ratios (OR) and 95% confidence intervals (CI) for the associations of race, ethnicity, and BMI with pCR adjusting for subtype, study arm, lymph node status, tumor size, and tumor grade.

Results

253 (14.1%) were black, 199 (11.1%) Hispanic, 520 (28.9%) overweight, and 743 (41.4%) obese. Compared to whites, Blacks and Hispanics were more likely to be obese and Blacks were more likely to have triple-negative cancer. pCR rates differed significantly by tumor subtype. In multivariate analyses, neither race (black vs. white: OR: 1.18, 95% CI: 0.85–1.62) nor ethnicity (Hispanic vs. non-Hispanic: OR: 1.30, 95% CI: 0.67–2.53) were significant predictors of pCR overall or by subtype. Overweight and obese women had lower pCR rates in ER+/HER2+, but higher pCR rates in ER−/HER2+ cancers.

Conclusions

There was no difference in breast pCR according to race or ethnicity. Overall, there was no major difference in pCR rates by BMI. These findings suggest that pCR with optimally dosed NST is a function of tumor, rather than patient, biology.

Keywords: breast cancer, race, ethnicity, body mass index, pathologic complete response

Introduction

Obesity [1,2], and black race [3,4] have each been independently associated with poor breast cancer outcomes. The relationship of Hispanic ethnicity with breast cancer outcomes has been inconsistent with some studies suggesting equal or superior outcomes when compared to non-Hispanic whites, while others suggest worse outcomes [5,6,3]. Black race, Hispanic ethnicity, and obesity are each associated with a higher incidence of more aggressive tumor biology (high grade, hormone receptor negative, high proliferation fraction, more lymphovascular invasion) [5,7,8]. Differences in tumor subtype likely contribute to observed poorer outcomes, but residual disparities remain [9]. This could be due, at least in part, to differences in response to therapy [10]. In addition, differences in quality of chemotherapy, including treatment delays [11,12] and suboptimal dosing [13] may contribute to disparate breast cancer outcomes according to race, ethnicity, and body mass index (BMI). Given the high prevalence of overweight and obesity among black and Hispanic women in the US, it is possible that race and BMI may interact. In an effort to address these disparities, it is important to investigate whether, independent of their measurable tumor characteristics, and in a setting of standardized therapeutic regimens, overweight, obesity, or black race are associated with response to therapy.

Achievement of a pathologic complete response (pCR) to neoadjuvant systemic therapy (NST) can be defined in a number of ways, but all definitions include the absence of residual invasive cancer in the breast (in-breast pCR). Some definitions also require the absence of invasive and in-situ disease and include the axillary nodes [14]. While the absence of invasive and in-situ disease in the breast and the axillary nodes is most strongly associated with favorable prognosis [15], in-breast pCR is highly correlated with pCR in the axillary nodes [16]. Regardless of definition, pCR rates differ greatly across tumor subtypes with the highest rates observed among patients with triple negative or estrogen receptor (ER)-negative, human epidermal growth factor receptor-2 (HER2)-positive (ER−/HER2+) cancers while very low pCR rates are seen in patients with ER+/HER2− tumors [17]. In an era of personalized medicine, NST often allows women with locally advanced breast cancer to become candidates for breast conserving surgery[18] and permits in vivo assessment of treatment response [19]. Women achieving pCR have better event-free and overall survival than those who do not, though this association is also subtype-dependent [15]. The greatest survival benefits of pCR appear to occur in patients with triple negative and ER−/HER2+ tumors [20]. Because of its demonstrated association with survival, the Food and Drug Administration allows pCR to be used as a surrogate endpoint for accelerated approval in pharmaceutical trials [21].

Given the observed disparities in survival according to race, ethnicity, and BMI and the association of pCR with survival, the purpose of this study was to examine the association of race, ethnicity and BMI with pCR in clinical trial populations. Our sample includes multiple trials in which chemotherapy dosing, which may vary considerably between patient populations outside of trials [22,23], was standardized and rigorously accounted for. Using data on 1,797 women participating in four Alliance for Clinical Trials in Oncology (Alliance) clinical trials we evaluated the proportion of women achieving in-breast pCR according to race, ethnicity, and BMI. We examine these factors overall, and stratified by subtypes defined by ER status and HER2.

Methods

Study population

This analysis pools data from four neoadjuvant chemotherapy clinical trials conducted by the members of the Alliance (Table 1), Cancer and Leukemia Group B (CALGB) and American College of Surgeons Oncology Group (ACOSOG) are now part of the Alliance. Two trials (CALGB 40601 and ACOSOG Z1041) were limited to patients with HER2+ cancers, while one (CALGB 40603) enrolled only patients with triple-negative cancers [2426]. The fourth trial (ACOSOG Z1071) included all breast cancer subtypes of breast cancer; it was designed to evaluate the false-negative rate for sentinel lymph node surgery following neoadjuvant chemotherapy in women initially presenting with biopsy-proven node-positive breast cancer [27]. All trials were approved by the appropriate medical ethics committees. All patients gave written informed consent for study participation and data collection.

Table 1.

Trials included in pooled analysis

Study Number Title Accrual Subtype Study Open Date Study Close Date Reference
CALGB 40603 Paclitaxel With or Without Carboplatin and/or Bevacizumab Followed By Doxorubicin and Cyclophosphamide in Treating Patients With Breast Cancer That Can Be Removed by Surgery 454 Triple Negative May 2009 August 2013 Sikov WM et al. (2014) JCO [26]
CALGB 40601 Paclitaxel and Trastuzumab With or Without Lapatinib in Treating Patients With Stage II or Stage III Breast Cancer That Can Be Removed by Surgery 305 HER2+ December 2008 January 2014 Carey LA et al. (2015) JCO [25]
ACOSOG Z1041 Definitive analysis of randomized neoadjuvant trial comparing 5-fluorouracil, epirubicin and cyclophosphamide (FEC) followed by paclitaxel plus trastuzumab with paclitaxel plus trastuzumab followed by FEC plus trastuzumab in HER2+ operable breast cancer 282 HER2+ September 2007 December 2011 Buzdar AU et al. (2013) Lancet Oncol [24]
ACOSOG Z1071 A Phase II Study Evaluating The Role of Sentinel Lymph Node Surgery and Axillary Lymph Node Dissection Following Preoperative Chemotherapy in Women with Node Positive Breast Cancer (T1–4, N1–2, M0) at Initial Diagnosis 756 Any July 2009 July 2011 Boughey JC et al., (2013) JAMA [51]

Data collection and statistical analysis

Data collection and statistical analyses were conducted by the Alliance Statistics and Data Center. Race and ethnicity were self-reported and BMI was collected by institutional study coordinators from chemotherapy orders. Breast cancer tumor subtypes were assigned on the basis of clinical estrogen receptor (ER) and HER2 status as determined locally. ER was assessed by standard criteria using immunohistochemical (IHC) staining. HER2 was assessed using either IHC or fluorescence in situ hybridization (FISH). Pathologic complete response was defined as the absence of residual invasive disease in the breast (ypT0/is). This was the primary endpoint for all trials except Z1071, which as noted previously, evaluated a nodal question but collected breast and nodal pCR rates on participants.

Analyses included 1797 women from the four trials. We used logistic regression models to examine the relationship between race, ethnicity BMI and response to neoadjuvant chemotherapy. We evaluated the association between race (Black or White), ethnicity (Hispanic or Non-Hispanic) and BMI (<20, 20–24, 25–29, ≥30 kg/m2) with pCR in the breast following NST. We tested for potential interaction between race and BMI using a cross product term. We present models overall and stratified by subtypes defined by ER status and HER2 (ER+/HER2−, ER+/HER2+, ER−/HER2+, and triple negative). Models are adjusted for subtype (overall model only), study arm, lymph node status, and tumor grade. Tumor size was not collected in Z1041. We performed a sensitivity analysis by running models with and without tumor size included (using a missing indicator). As results did not materially differ, we present results without adjustment for tumor size. Statistical tests were two-sided, and p < 0.05 was considered significant. Statistical analyses were performed by Alliance statisticians at the Alliance Statistics and Data Center using SAS version 9.3 (SAS Institute, Cary, NC) on a dataset frozen on January 13, 2016.

Results

Patient Characteristics (Table 2)

Table 2.

Patient characteristics according to race and body mass index

Racea Ethnicity BMI (kg/m2)b
White n=1417 Black n=253 Other n=127 p-value Hispanic Non-Hispanic n=1476 p-value <20 n=66 20–24 n=468 25–29 n=520 ≥30 n=743 p-value
Age 0.03 0.04 <0.0001
 Mean (SD) 49.7 (10.8) 49.4 (9.6) 47.2 (10.8) 48.4 (11.8) 49.8 (10.4) 45.9 (10.4) 47.4 (10.9) 50.5 (10.4) 50.5 (10.4)
BMI <0.0001 0.002
 Mean (SD) 29.1 (6.8) 31.9 (6.0) 27.2 (5.5) 30.6 (7.1) 29.3 (6.6)
Race, n (%) <0.0001 <0.0001
 White 166 (83.4) 1177 (79.7) 54 (3.8) 398 (28.1) 414 (29.2) 551 (38.9)
 Black 8 (4.0) 226 (15.3) 2 (0.8) 32 (12.6) 67 (26.5) 152 (60.1)
 Other 25 (12.6) 73 (4.9) 10 (7.9) 38 (29.9) 39 (30.7) 40 (31.5)
Ethnicity, n (%) <0.00001 0.003
 Non-Hispanic 1177 (83.1) 226 (89.3) 73 (57.5) 54 (3.7) 389 (26.4) 425 (28.8) 608 (41.2)
 Hispanic 166 (11.7) 8 (3.2) 25 (19.7) 2 (1.0) 44 (22.1) 55 (27.6) 98 (49.2)
 Unknown 74 (5.2) 19 (7.5) 29 (22.8) 10 (8.2) 35 (28.7) 40 (32.8) 37 (30.3)
HER2+, n (%) 589 (41.6) 74 (29.2) 64 (50.4) 0.0006 108 (54.3) 572 (38.8) 0.0003 31 (4.3) 205 (28.2) 209 (28.7) 282 (38.8) 0.08
ER+, n (%) 671 (47.7) 96 (37.9) 56 (44.8) 0.02 97 (48.7) 674 (45.9) 0.6 31 (5.9) 217 (41.5) 242 (46.3) 333 (6.3) 0.88
Subtype, n (%) 0.0001 <0.0001 0.68
 ER+/HER2− 343 (24.4) 53 (20.9) 21 (16.8) 38 (19.2) 361 (24.6) 12 (2.9) 105 (25.2) 124 (29.7) 176 (42.2)
 ER+/HER2+ 328 (23.3) 43 (17.0) 35 (28.0) 59 (29.8) 313 (21.4) 19 (4.7) 112 (27.6) 118 (29.1) 157 (38.7)
 ER−/HER2+ 255 (18.1) 31 (12.3) 27 (21.6) 49 (24.7) 252 (17.2) 12 (3.8) 90 (28.8) 87 (27.8) 124 (39.6)
 Triple 480 (34.1) 126 (49.8) 42 (33.6) 52 (26.3) 540 (36.8) 23 (3.5) 155 (23.9) 186 (28.7) 284 (43.8)
 Negative
Node pos, n (%) 266 (18.8) 63 (24.9) 34 (26.8) 0.05 35 (17.6) 291 (19.7) 0.006 11 (3.0) 96 (26.4) 107 (29.5) 149 (41.0) 0.92
Grade 3, n (%) 859 (60.6) 166 (65.6) 94 (74.0) 0.03 126 (63.3) 910 (61.7) 0.1 43 (3.8) 304 (27.2) 317 (28.3) 455 (40.7) 0.37
Tumor Sizec 0.06 0.05 0.09
 Mean (SD) 3.3 (2.3) 3.1 (2.4) 3.8 (2.5) 3.1 (1.9) 3.3 (2.3) 3.9 (3.5) 3.1 (2.1) 3.4 (2.4) 3.3 (2.3)
Trial, n (%) <0.0001 <0.0001 0.13
 40601 243 (17.1) 27 (10.7) 35 (27.6) 23 (11.6) 249 (16.9) 15 (4.9) 94 (30.8) 92 (30.2) 104 (34.1)
 40603 331 (23.4) 89 (35.2) 34 (26.8) 36 (18.1) 373 (25.3) 11 (2.4) 115 (25.3) 132 (29.1) 196 (43.2)
 Z1041 232 (16.4) 31 (12.3) 19 (15.0) 64 (32.2) 212 (14.4) 10 (3.5) 78 (27.7) 72 (25.5) 122 (43.3)
 Z1071 611 (43.1) 106 (41.9) 39 (30.7) 76 (38.2) 642 (43.5) 30 (4.0) 181 (23.9) 224 (29.6) 321 (42.5)
a

Column percents presented.;

b

Row percents presented;

c

Tumor size not collected in Z1041

Of the 1797 women included in analyses, 1177 (83.1%) were white, 253 (14.1%) were black, 127 (7.1%) were listed as ‘other’, 199 (11.1%) were Hispanic, and 520 (28.9%) were overweight (BMI 25–29) and 743 (41.3%) were obese (BMI ≥ 30 kg/m2). Information on ER, PR, and HER2 status was available for 1784 (99.3 %) patients. The mean age of overweight and obese women (50.5 years) was higher than for normal weight women (47.4 years) (p <0.0001). Compared to white women, a higher percentage of black women had triple-negative cancers (49.8% vs. 34.1%), with consequent reductions in the percentage of black women with ER+ and HER2+ cancers (p=0.0001). A slightly higher percentage of black women had clinically node-positive disease at study entry (24.9%, vs. 18.8% for white women; p=0.05). Compared to white women, black women had a higher mean BMI (31.9 kg/m2 vs. 29.1 kg/m2; p <0.0001) and were more likely to be obese (60.1% vs. 38.9%; p <0.0001). Hispanic women were also more likely to be obese than non-Hispanic women (49.2% vs. 41.2%; p=0.0003). Aside from a slight increase in the percentage of patients with triple-negative cancers who were obese (43.8% vs. 40.2% for other subtypes), there were no major differences in the distribution of BMI among the subtypes (p=0.68).

pCR rates, Race, Ethnicity (Tables 3 & 4)

Table 3.

Pathologic complete response in four Alliance clinical trials according to race, ethnicity and BMI (N=1672)

Na %pCR OR (95% CI)b
Race
 White 1381 550 (39.8) 1.0 (reference)
 Black 245 99 (40.4) 1.18 (0.85, 1.62)
Ethnicity
 Non-Hispanic 1441 570 (39.6) 1.0 (reference)
 Hispanic 194 90 (46.4) 1.22 (0.86, 1.72)
BMI (kg/m2)
 <20 64 33 (51.6) 1.60 (0.89, 2.86)
 20–24 454 198 (43.6) 1.0 (reference)
 25–29 510 198 (38.8) 0.86 (0.64, 1.16)
 ≥30 722 277 (38.4) 0.82 (0.62, 1.08)
 p-trend c 0.094
Subtype
 ER+/HER2− 416 66 (15.9) 1.0 (reference)
 ER+/HER2+ 399 152 (38.1) 2.16 (1.32, 3.51)
 ER−/HER2+ 311 195 (62.7) 5.48 (3.40, 8.85)
 Triple Negative 611 285 (46.6) 2.56 (1.75, 3.75)
a

125 women excluded due to missing values of variables;

b

mutually adjusted for all variables in table plus trial arm, tumor grade, lymph node status, tumor size, and subtype;

c

p-trend is from a test of linear trend of an ordinal variable defined by the median of each category.

Table 4.

Pathologic complete response according to breast cancer subtype in four Alliance clinical trials

ER+/HER2− (N=416) ER+/HER2+ (N=406) ER−/HER2+ (N=313) Triple Negative (N=648)
N % pCR OR (95% CI)a N % pCR OR (95% CI)a N % pCR OR (95% CI)a N % pCR OR (95% CI)a
Race
 White 343 51 (14.9) 1.0 (reference) 323 125 (38.7) 1.0 (reference) 254 156 (61.4) 1.0 (reference) 450 211 (46.9) 1.0 (reference)
 Black 52 9 (17.3) 1.04 (0.41, 2.66) 42 18 (42.9) 1.42 (0.66, 3.08) 30 18 (60.0) 0.65 (0.27, 1.59) 121 54 (44.6) 1.09 (0.70, 1.72)
Ethnicity
 Non-Hispanic 361 53 (14.7) 1.0 (reference) 307 120 (39.1) 1.0 (reference) 250 153 (61.2) 1.0 (reference) 513 238 (46.4) 1.0 (reference)
 Hispanic 38 7 (18.4) 1.24 (0.45, 3.38) 59 26 (44.1) 1.30 (0.67, 2.53) 49 32 (65.3) 0.97 (0.47, 2.03) 47 24 (51.1) 1.32, (0.68, 2.54)
BMI (kg/m2)
 <20 12 3 (25.0) 1.75 (0.32, 9.52) 19 9 (47.4) 1.03 (0.34, 3.13) 12 10 (83.3) 4.49 (0.84, 24.1) 21 11 (52.4) 1.32 (0.50, 3.48)
 20–24 104 18 (17.3) 1.0 (reference) 109 52 (47.7) 1.0 (reference) 90 49 (54.4) 1.0 (reference) 145 74 (51.0) 1.0 (reference)
 25–29 124 20 (16.1) 1.27 (0.54, 2.98) 116 30 (25.9) 0.36 (0.19, 0.67) 87 62 (71.3) 2.24 (1.09, 4.62) 178 83 (46.6) 0.81, (0.50, 1.31)
 ≥30 176 25 (14.2) 0.97 (0.43, 2.18) 155 61 (39.4) 0.68 (0.38, 1.22) 122 74 (60.7) 1.35 (0.72, 2.54) 267 117 (43.8) 0.72 (0.46, 1.14)
 p-trend b 0.88 0.010 0.082 0.40
a

mutually adjusted for all variables in table plus trial arm, tumor grade, and lymph node status

b

p-trend is from a test for linear trend of an ordinal variable defined by the median of each category

Overall, the pCR rate was significantly higher in patients with ER+/HER2+ (38.1%; OR: 2.16, 95% CI: 1.32–3.51), ER−/HER2+ (62.7%; 5.48: 3.40–8.85) and triple-negative (46.6%; 2.56: 1.75–3.75) cancers than in patients with ER+/HER2− cancers (15.9%). In total, 40.4% (99/245) of black women achieved pCR as compared to 39.8% (550/1381) of white women. Adjusting for subtype and other potential confounders, we observed no significant association between black race and pCR (OR: 1.18, 95% CI: 0.85–1.62). In analyses stratified by tumor subtype, we also saw no significant differences in pCR rates between black and white women (Table 4). Hispanic ethnicity was also not associated with pCR, either overall (Table 3) or when stratified by tumor subtype (Table 4). Overall, 46.4% (90/194) of Hispanic women achieved pCR compared to 39.6% (570/1441) of non-Hispanic women. Adjusting for subtype and other potential confounders, we found no significant association between Hispanic ethnicity and pCR (OR: 1.22, 95% CI 0.86–1.72). In analyses stratified by tumor subtype, we also saw no significant differences in pCR rates between Hispanic and non-Hispanic women (Table 4).

BMI and pCR (Tables 3 & 4)

Compared to normal weight women (BMI 20–24), in whom the overall pCR rate was 43.6%, the small cohort (n= 66, 3.7% of the study population) of underweight women (BMI <20) had a non-significantly higher pCR rate of 51.6% (OR 1.60: 95% CI 0.89–2.86), while overweight (38.8%; OR 0.86: 0.64–1.16) and obese (38.4%; OR 0.82: 0.62–1.08) had non-significantly lower pCR rates. In multivariable adjusted models, there was a statistically non-significant, inverse association between BMI and pCR (p-trend=0.09; Table 4). In adjusted models there was a significant inverse association between BMI and pCR in ER+/HER2+ patients (p-trend=0.01); in this subtype, overweight and obese women had pCR rates of 25.9% (30/116) and 39.4% (61/155) compared to 47.7% (52/109) and 47.4% (9/19) for normal and underweight women, respectively. In contrast, in ER−/HER2+ patients pCR rates were higher in overweight (71.3%; 62/87), obese women (60.7%; 74/122) and underweight women (83.3%; 10/12) women compared to normal weight women (54.4%; 49/90), resulting in a non-significant positive association between BMI and pCR (p-trend=0.82) for that subtype. We saw no significant differences in pCR rates according to BMI among women with ER+/HER2− or triple negative tumors. There was no interaction between race and BMI (p=0.91).

Discussion

In this large pooled analysis of four randomized clinical trials including 1797 patients, we examined whether race, Hispanic ethnicity and/or BMI were associated with in-breast pathologic complete response to neoadjuvant chemotherapy and HER2−targeted therapy in patients with HER2+ cancers. We observed no difference in pCR rates according to race or ethnicity overall or when stratified by tumor subtype. We observed no overall differences according to BMI, but did see trends associating increasing BMI with lower pCR rates in ER+/HER2+ patients and higher pCR rates in ER−/HER2+ patients.

In our study pCR rates were similar in black and white women as well as in Hispanics and non-Hispanics. Several previous studies have examined the association between race and pCR with some finding no difference by race [28,10], while others found lower pCR rates among black women.[29,30] Chavez-MacGregor and colleagues found no differences in pCR rates between blacks, whites and Hispanics, overall or within tumor subtypes, in a retrospective analysis of 2074 patients treated at the M.D. Anderson Cancer Center between 1994 and 2008, in which the reported pCR rates (overall = 12.5%, HER2+ 22.6%, triple-negative 19.4%, hormone receptor-positive 5%) were much lower than in our population. 28 This lower observed pCR rates is at least in part because of differences in pCR definition (in-breast vs. breast and axilla) and use of combination therapy (i.e., trastuzumab) in our trials. In a more recent paper, Killelea and colleagues, using data from the National Cancer Data Base (NCDB), found similar pCR rates across racial groups overall, but when stratified by tumor subtype non-Hispanic black women had a non-significantly lower pCR rate for ER−/HER2+ tumors (42.6% vs. 53.9%; OR: 0.72, 95% CI: 0.46–1.14) and a significantly lower pCR rate for triple-negative (36.6%; 416/1138) tumors (36.6% vs. 42.8%; OR 0.73, 95% CI: 0.59–0.89) compared to white women [29]. There were no significant differences according to Hispanic ethnicity. The different results in the present study and that by Killelea et al. may be due to our use of clinical trial populations. The setting of randomized clinical trials provides several important advantages including standardized enrollment requirements, uniform assessment of disease characteristics such as stage, tumor phenotype and other factors associated with prognosis. Treatment is also standardized, carefully monitored, and facilitated, eliminating variation in care that may occur outside of a trial. While the NST regimen for patients enrolled on Z1071 was not specified in the protocol, most received an anthracycline and a taxane in the neoadjuvant setting, and since the treating physicians at the participating institutions were familiar with treatment guidelines from other cooperative group trials, we considered it highly likely that the treatment plans utilized followed similar guidelines (standard chemotherapy dosing using actual rather than ideal body weight to calculate body surface area, for example).

Though most of the available data is from the adjuvant setting, there is evidence that black women with early stage breast cancer start chemotherapy later, are more likely to receive a lower than standard dose and lower relative dose intensity, and are more likely to have dose reductions in the first and subsequent treatment cycles compared to whites [31,32]. These differences could not be explained by racial differences in BMI, comorbid conditions or hematological factors [33]. There is also little evidence of greater levels of acute toxicity in black women receiving neoadjuvant or adjuvant chemotherapy [34]. It is possible that the lower pCR rates among black women in the NCDB are due to dose reductions, treatment delays, or differences in chemotherapy regimens received. Taken together, our results suggest that racial differences in pCR identified in retrospective studies may not be caused by biology.

Our results suggest that, overall, in-breast pCR rates are not significantly affected by BMI in appropriately dosed patients, though there was a non-significant trend towards lower pCR rates with a higher BMI (p-trend=0.094). In another retrospective analysis from the M. D. Anderson database, Litton and colleagues found that overweight (OR = 0.59, 95% CI: 0.37–0.95) or the combination of overweight and obese (OR = 0.67, 95% CI: 0.45–0.99) women had significantly lower pCR rates compared to normal/underweight women.35 Results were not stratified by tumor subtype [35]. Among Chinese women, patients with BMI ≥ 25 kg/m2 were 55% less likely to achieve pCR compared to those with BMI <25 kg/m2. Differences were largest among postmenopausal and HR− patients [36]. Several other studies have associated lower BMI with higher rates of pCR [37]. Weight gain during neoadjuvant chemotherapy has not been associated with pCR [38], except perhaps in subgroups of women who were normal/underweight or postmenopausal at baseline, where it was associated with higher pCR rates [39].

In our study, the impact of BMI on pCR appeared to differ by tumor subtype. Specifically, in overweight and obese women we saw lower pCR rates in ER+/HER2+ cancers and higher pCR rates in ER−/HER2+ cancers. However confidence intervals were wide and it is possible that result was due to chance. Potential mechanisms relating overweight and obesity to lower pCR rates and poorer survival include: 1) higher circulating estrogen levels among obese postmenopausal women as compared to leaner women and higher estrogen levels when on aromatase inhibitor therapy [40], 2) higher levels of circulating insulin, adiponectin, c-peptide, leptin and other metabolic hormones [41], and 3) higher circulating inflammatory cytokines which may reduce apoptosis and increase proliferation among obese women [42,43]. In some settings chemotherapy may not be dosed appropriately leading to under dosing of obese women, and this would have a greater impact on black women where we observed higher rates of obesity compared to whites [44,45]. Chemotherapy under dosing is less of a concern in our clinical trials because weight-based dosing is required [26,25,24].

Our study has several important strengths and limitations. While our analysis included almost 1800 women, dividing the population by tumor subtype, race, ethnicity and BMI limited our statistical power. Our outcome was in-breast pCR (ypT0/Tis), while many other studies have used pCR in the breast and axillary nodes (ypT0/Tis ypN0) pCR as their endpoint, as it has a stronger association with disease-free survival [46,20]. However, in order to explain the difference in results between our study and those using ypT0/Tis ypN0 race or BMI would have to only affect the likelihood of achieving pCR in the axilla, but not in the breast. This seems unlikely since most women that achieve pCR in-breast also do so in the axilla [15]. Assigning subtype by ER and HER status alone ignores certain molecular heterogeneity within those subtypes that may have a major impact on tumor response and achievement of pCR [47,48]. While the treatment regimens received across our included trials were not uniform, within each trial with the exception of Z1071, patients received standardized therapy. Clinical trial participants may not be representative of the general population. For example, clinical trial participants are generally younger than patients in the community [49]. Lack of racial and ethnic diversity is problematic in many clinical trials; however, our sample was 14% black and 11% Hispanic, reasonably similar to their distribution in the United States. Lastly, we did not look at long-term outcomes associated with pCR according to race, ethnicity or BMI. Studies that have done this suggest that among women that achieve pCR outcomes such as disease-free survival and overall survival are similar, but disparities persist among women that do not [28,50]. Further research is necessary to tease apart the molecular characteristics and post-neoadjuvant treatment and behavioral/environmental exposures that contribute to these disparities among non-responders.

In conclusion, our study found no differences in breast pCR according to race or Hispanic ethnicity and no significant impact of BMI on the overall pCR rate. These findings suggest that achievement of pCR with optimally dosed NST is largely a function of tumor, rather than patient, biology. It further suggests that yet-to-be-defined biologic factors that may influence which patients within each subtype achieve a pCR are also little influenced by race, ethnicity and BMI; if these patient characteristics determined those factors we would expect to see more substantial differences in the pCR rates among our subgroups. Analysis of other studies utilizing similarly effective NST regimens are warranted to see if they also demonstrate lower pCR rates in ER+/HER2+ and higher pCR rate in ER−/HER2+ overweight and obese women that could generate hypotheses for the etiology of these observations.

Acknowledgments

The authors would like to acknowledge Dr. Olwen Hahn for her contributions to the included studies. Research reported in this publication was supported by the National Cancer Institute of the National Institutes of Health under the Award Number UG1CA189823 (to the Alliance for Clinical Trials in Oncology NCORP Grant), U10CA180790, U10CA180838, U10CA180858, U10CA180867. Dr. Warner was supported by NCI grant K01CA188075. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. The study sponsors had no role in study design, collection, analysis, and interpretation of data, writing the report, or the decision to submit the report for publication. Dr. Warner had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

Footnotes

Ethical Approval

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards.

Conflict of Interests

The authors declare that they have no conflict of interests.

References

  • 1.Sparano JA, Wang M, Zhao F, Stearns V, Martino S, Ligibel JA, Perez EA, Saphner T, Wolff AC, Sledge GW, Jr, Wood WC, Fetting J, Davidson NE. Obesity at diagnosis is associated with inferior outcomes in hormone receptor-positive operable breast cancer. Cancer. 2012 doi: 10.1002/cncr.27527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Ligibel JA, Cirrincione CT, Liu M, Citron M, Ingle JN, Gradishar W, Martino S, Sikov W, Michaelson R, Mardis E, Perou CM, Ellis M, Winer E, Hudis CA, Berry D, Barry WT. Body Mass Index, PAM50 Subtype, and Outcomes in Node-Positive Breast Cancer: CALGB 9741 (Alliance) J Natl Cancer Inst. 2015;107(9) doi: 10.1093/jnci/djv179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Warner ET, Tamimi RM, Hughes ME, Ottesen RA, Wong YN, Edge SB, Theriault RL, Blayney DW, Niland JC, Winer EP, Weeks JC, Partridge AH. Racial and Ethnic Differences in Breast Cancer Survival: Mediating Effect of Tumor Characteristics and Sociodemographic and Treatment Factors. J Clin Oncol. 2015;33(20):2254–2261. doi: 10.1200/JCO.2014.57.1349. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Albain KS, Unger JM, Crowley JJ, Coltman CA, Jr, Hershman DL. Racial disparities in cancer survival among randomized clinical trials patients of the Southwest Oncology Group. Journal of the National Cancer Institute. 2009;101(14):984–992. doi: 10.1093/jnci/djp175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Banegas MP, Li CI. Breast cancer characteristics and outcomes among Hispanic Black and Hispanic White women. Breast cancer research and treatment. 2012;134(3):1297–1304. doi: 10.1007/s10549-012-2142-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Ooi SL, Martinez ME, Li CI. Disparities in breast cancer characteristics and outcomes by race/ethnicity. Breast Cancer Res Treat. 2011;127(3):729–738. doi: 10.1007/s10549-010-1191-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Parise CA, Bauer KR, Brown MM, Caggiano V. Breast cancer subtypes as defined by the estrogen receptor (ER), progesterone receptor (PR), and the human epidermal growth factor receptor 2 (HER2) among women with invasive breast cancer in California, 1999–2004. The breast journal. 2009;15(6):593–602. doi: 10.1111/j.1524-4741.2009.00822.x. [DOI] [PubMed] [Google Scholar]
  • 8.Sweeney C, Bernard PS, Factor RE, Kwan ML, Habel LA, Quesenberry CP, Shakespear K, Weltzien EK, Stijleman IJ, Davis CA, Ebbert MT, Castillo A, Kushi LH, Caan BJ. Intrinsic subtypes from PAM50 gene expression assay in a population-based breast cancer cohort: differences by age, race, and tumor characteristics. Cancer Epidemiol Biomarkers Prev. 2014;23(5):714–724. doi: 10.1158/1055-9965.EPI-13-1023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.O'Brien KM, Cole SR, Tse CK, Perou CM, Carey LA, Foulkes WD, Dressler LG, Geradts J, Millikan RC. Intrinsic breast tumor subtypes, race, and long-term survival in the Carolina Breast Cancer Study. Clinical cancer research : an official journal of the American Association for Cancer Research. 2010;16(24):6100–6110. doi: 10.1158/1078-0432.CCR-10-1533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Tichy JR, Deal AM, Anders CK, Reeder-Hayes K, Carey LA. Race, response to chemotherapy, and outcome within clinical breast cancer subtypes. Breast cancer research and treatment. 2015;150(3):667–674. doi: 10.1007/s10549-015-3350-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Chavez-MacGregor M, Clarke CA, Lichtensztajn DY, Giordano SH. Delayed Initiation of Adjuvant Chemotherapy Among Patients With Breast Cancer. JAMA Oncol. 2016;2(3):322–329. doi: 10.1001/jamaoncol.2015.3856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Gagliato DeM, Gonzalez-Angulo AM, Lei X, Theriault RL, Giordano SH, Valero V, Hortobagyi GN, Chavez-Macgregor M. Clinical impact of delaying initiation of adjuvant chemotherapy in patients with breast cancer. J Clin Oncol. 2014;32(8):735–744. doi: 10.1200/JCO.2013.49.7693. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Gurney H. How to calculate the dose of chemotherapy. Br J Cancer. 2002;86(8):1297–1302. doi: 10.1038/sj.bjc.6600139. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Untch M, Konecny GE, Paepke S, von Minckwitz G. Current and future role of neoadjuvant therapy for breast cancer. Breast. 2014;23(5):526–537. doi: 10.1016/j.breast.2014.06.004. [DOI] [PubMed] [Google Scholar]
  • 15.von Minckwitz G, Untch M, Blohmer JU, Costa SD, Eidtmann H, Fasching PA, Gerber B, Eiermann W, Hilfrich J, Huober J, Jackisch C, Kaufmann M, Konecny GE, Denkert C, Nekljudova V, Mehta K, Loibl S. Definition and impact of pathologic complete response on prognosis after neoadjuvant chemotherapy in various intrinsic breast cancer subtypes. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2012;30(15):1796–1804. doi: 10.1200/JCO.2011.38.8595. [DOI] [PubMed] [Google Scholar]
  • 16.Kuerer HM, Newman LA, Smith TL, Ames FC, Hunt KK, Dhingra K, Theriault RL, Singh G, Binkley SM, Sneige N, Buchholz TA, Ross MI, McNeese MD, Buzdar AU, Hortobagyi GN, Singletary SE. Clinical course of breast cancer patients with complete pathologic primary tumor and axillary lymph node response to doxorubicin-based neoadjuvant chemotherapy. J Clin Oncol. 1999;17(2):460–469. doi: 10.1200/JCO.1999.17.2.460. [DOI] [PubMed] [Google Scholar]
  • 17.Houssami N, Macaskill P, von Minckwitz G, Marinovich ML, Mamounas E. Meta-analysis of the association of breast cancer subtype and pathologic complete response to neoadjuvant chemotherapy. Eur J Cancer. 2012;48(18):3342–3354. doi: 10.1016/j.ejca.2012.05.023. [DOI] [PubMed] [Google Scholar]
  • 18.Golshan M, Cirrincione CT, Sikov WM, Berry DA, Jasinski S, Weisberg TF, Somlo G, Hudis C, Winer E, Ollila DW Oncology AfCTi. Impact of neoadjuvant chemotherapy in stage II–III triple negative breast cancer on eligibility for breast-conserving surgery and breast conservation rates: surgical results from CALGB 40603 (Alliance) Ann Surg. 2015;262(3):434–439. doi: 10.1097/SLA.0000000000001417. discussion 438–439. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.von Minckwitz G, Blohmer JU, Costa SD, Denkert C, Eidtmann H, Eiermann W, Gerber B, Hanusch C, Hilfrich J, Huober J, Jackisch C, Kaufmann M, Kümmel S, Paepke S, Schneeweiss A, Untch M, Zahm DM, Mehta K, Loibl S. Response-guided neoadjuvant chemotherapy for breast cancer. J Clin Oncol. 2013;31(29):3623–3630. doi: 10.1200/JCO.2012.45.0940. [DOI] [PubMed] [Google Scholar]
  • 20.Cortazar P, Zhang L, Untch M, Mehta K, Costantino JP, Wolmark N, Bonnefoi H, Cameron D, Gianni L, Valagussa P, Swain SM, Prowell T, Loibl S, Wickerham DL, Bogaerts J, Baselga J, Perou C, Blumenthal G, Blohmer J, Mamounas EP, Bergh J, Semiglazov V, Justice R, Eidtmann H, Paik S, Piccart M, Sridhara R, Fasching PA, Slaets L, Tang S, Gerber B, Geyer CE, Jr, Pazdur R, Ditsch N, Rastogi P, Eiermann W, von Minckwitz G. Pathological complete response and long-term clinical benefit in breast cancer: the CTNeoBC pooled analysis. Lancet. 2014;384(9938):164–172. doi: 10.1016/S0140-6736(13)62422-8. [DOI] [PubMed] [Google Scholar]
  • 21.(CDER) USDoHaHSFaDACfDEaR. Guidance for Industry Pathological Complete Response in Neoadjuvant Treatment of High-Risk Early-Stage Breast Cancer: Use as an Endpoint to Support Accelerated Approval 2014 [Google Scholar]
  • 22.Griggs JJ, Culakova E, Sorbero ME, van Ryn M, Poniewierski MS, Wolff DA, Crawford J, Dale DC, Lyman GH. Effect of patient socioeconomic status and body mass index on the quality of breast cancer adjuvant chemotherapy. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2007;25(3):277–284. doi: 10.1200/JCO.2006.08.3063. [DOI] [PubMed] [Google Scholar]
  • 23.Griggs JJ, Liu Y, Sorbero ME, Jagielski CH, Maly RC. Adjuvant chemotherapy dosing in low-income women: the impact of Hispanic ethnicity and patient self-efficacy. Breast Cancer Res Treat. 2014;144(3):665–672. doi: 10.1007/s10549-014-2869-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Buzdar AU, Suman VJ, Meric-Bernstam F, Leitch AM, Ellis MJ, Boughey JC, Unzeitig G, Royce M, McCall LM, Ewer MS, Hunt KK American College of Surgeons Oncology Group i. Fluorouracil, epirubicin, and cyclophosphamide (FEC-75) followed by paclitaxel plus trastuzumab versus paclitaxel plus trastuzumab followed by FEC-75 plus trastuzumab as neoadjuvant treatment for patients with HER2-positive breast cancer (Z1041): a randomised, controlled, phase 3 trial. The Lancet Oncology. 2013;14(13):1317–1325. doi: 10.1016/S1470-2045(13)70502-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Carey LA, Berry DA, Cirrincione CT, Barry WT, Pitcher BN, Harris LN, Ollila DW, Krop IE, Henry NL, Weckstein DJ, Anders CK, Singh B, Hoadley KA, Iglesia M, Cheang MC, Perou CM, Winer EP, Hudis CA. Molecular Heterogeneity and Response to Neoadjuvant Human Epidermal Growth Factor Receptor 2 Targeting in CALGB 40601, a Randomized Phase III Trial of Paclitaxel Plus Trastuzumab With or Without Lapatinib. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2015 doi: 10.1200/JCO.2015.62.1268. JCO.2015.62.1268 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Sikov WM, Berry DA, Perou CM, Singh B, Cirrincione CT, Tolaney SM, Kuzma CS, Pluard TJ, Somlo G, Port ER, Golshan M, Bellon JR, Collyar D, Hahn OM, Carey LA, Hudis CA, Winer EP. Impact of the addition of carboplatin and/or bevacizumab to neoadjuvant once-per-week paclitaxel followed by dose-dense doxorubicin and cyclophosphamide on pathologic complete response rates in stage II to III triple-negative breast cancer: CALGB 40603 (Alliance) Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2015;33(1):13–21. doi: 10.1200/JCO.2014.57.0572. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Boughey JC, Suman VJ, Mittendorf EA, Ahrendt GM, Wilke LG, Taback B, Leitch AM, Kuerer HM, Bowling M, Flippo-Morton TS, Byrd DR, Ollila DW, Julian TB, McLaughlin SA, McCall L, Symmans WF, Le-Petross HT, Haffty BG, Buchholz TA, Nelson H, Hunt KK Alliance for Clinical Trials in O. Sentinel lymph node surgery after neoadjuvant chemotherapy in patients with node-positive breast cancer: the ACOSOG Z1071 (Alliance) clinical trial. JAMA. 2013;310(14):1455–1461. doi: 10.1001/jama.2013.278932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Chavez-Macgregor M, Litton J, Chen H, Giordano SH, Hudis CA, Wolff AC, Valero V, Hortobagyi GN, Bondy ML, Gonzalez-Angulo AM. Pathologic complete response in breast cancer patients receiving anthracycline- and taxane-based neoadjuvant chemotherapy: evaluating the effect of race/ethnicity. Cancer. 2010;116(17):4168–4177. doi: 10.1002/cncr.25296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Killelea BK, Yang VQ, Wang SY, Hayse B, Mougalian S, Horowitz NR, Chagpar AB, Pusztai L, Lannin DR. Racial Differences in the Use and Outcome of Neoadjuvant Chemotherapy for Breast Cancer: Results From the National Cancer Data Base. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2015;33(36):4267–4276. doi: 10.1200/JCO.2015.63.7801. [DOI] [PubMed] [Google Scholar]
  • 30.Balmanoukian A, Zhang Z, Jeter S, Slater S, Armstrong DK, Emens LA, Fetting JH, Wolff AC, Davidson NE, Jacobs L, Lange J, Tsangaris TN, Zellars R, Gabrielson E, Stearns V. African American women who receive primary anthracycline- and taxane-based chemotherapy for triple-negative breast cancer suffer worse outcomes compared with white women. J Clin Oncol. 2009;27(22):e35–37. doi: 10.1200/JCO.2008.21.5509. author reply e38–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Griggs JJ, Sorbero ME, Stark AT, Heininger SE, Dick AW. Racial disparity in the dose and dose intensity of breast cancer adjuvant chemotherapy. Breast Cancer Res Treat. 2003;81(1):21–31. doi: 10.1023/A:1025481505537. [DOI] [PubMed] [Google Scholar]
  • 32.Griggs JJ, Culakova E, Sorbero ME, Poniewierski MS, Wolff DA, Crawford J, Dale DC, Lyman GH. Social and racial differences in selection of breast cancer adjuvant chemotherapy regimens. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2007;25(18):2522–2527. doi: 10.1200/JCO.2006.10.2749. [DOI] [PubMed] [Google Scholar]
  • 33.Daly B, Olopade OI. A perfect storm: How tumor biology, genomics, and health care delivery patterns collide to create a racial survival disparity in breast cancer and proposed interventions for change. CA Cancer J Clin. 2015;65(3):221–238. doi: 10.3322/caac.21271. [DOI] [PubMed] [Google Scholar]
  • 34.Han HS, Reis IM, Zhao W, Kuroi K, Toi M, Suzuki E, Syme R, Chow L, Yip AY, Glück S. Racial differences in acute toxicities of neoadjuvant or adjuvant chemotherapy in patients with early-stage breast cancer. Eur J Cancer. 2011;47(17):2537–2545. doi: 10.1016/j.ejca.2011.06.027. [DOI] [PubMed] [Google Scholar]
  • 35.Litton JK, Gonzalez-Angulo AM, Warneke CL, Buzdar AU, Kau SW, Bondy M, Mahabir S, Hortobagyi GN, Brewster AM. Relationship between obesity and pathologic response to neoadjuvant chemotherapy among women with operable breast cancer. J Clin Oncol. 2008;26(25):4072–4077. doi: 10.1200/JCO.2007.14.4527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Chen S, Chen CM, Zhou Y, Zhou RJ, Yu KD, Shao ZM. Obesity or overweight is associated with worse pathological response to neoadjuvant chemotherapy among Chinese women with breast cancer. PLoS One. 2012;7(7):e41380. doi: 10.1371/journal.pone.0041380. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Elsamany S, Alzahrani A, Abozeed WN, Rasmy A, Farooq MU, Elbiomy MA, Rawah E, Alsaleh K, Abdel-Aziz NM. Mammographic breast density: Predictive value for pathological response to neoadjuvant chemotherapy in breast cancer patients. Breast. 2015;24(5):576–581. doi: 10.1016/j.breast.2015.05.007. [DOI] [PubMed] [Google Scholar]
  • 38.Bao J, Borja N, Rao M, Huth J, Leitch AM, Rivers A, Wooldridge R, Rao R. Impact of weight change during neoadjuvant chemotherapy on pathologic response in triple-negative breast cancer. Cancer medicine. 2015;4(4):500–506. doi: 10.1002/cam4.388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Kogawa T, Fouad TM, Wei C, Masuda H, Kai K, Fujii T, El-Zein R, Chavez-MacGregor M, Litton JK, Brewster A, Alvarez RH, Hortobagyi GN, Valero V, Theriault R, Ueno NT. Association of Body Mass Index Changes during Neoadjuvant Chemotherapy with Pathologic Complete Response and Clinical Outcomes in Patients with Locally Advanced Breast Cancer. Journal of Cancer. 2015;6(4):310–318. doi: 10.7150/jca.10580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Ligibel JA, Winer EP. Aromatase inhibition in obese women: how much is enough? Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2012;30(24):2940–2942. doi: 10.1200/JCO.2012.43.7244. [DOI] [PubMed] [Google Scholar]
  • 41.Ligibel J. Obesity and breast cancer. Oncology (Williston Park, NY) 2011;25(11):994–1000. [PubMed] [Google Scholar]
  • 42.Goodwin PJ, Ennis M, Bahl M, Fantus IG, Pritchard KI, Trudeau ME, Koo J, Hood N. High insulin levels in newly diagnosed breast cancer patients reflect underlying insulin resistance and are associated with components of the insulin resistance syndrome. Breast cancer research and treatment. 2009;114(3):517–525. doi: 10.1007/s10549-008-0019-0. [DOI] [PubMed] [Google Scholar]
  • 43.Irwin ML, Duggan C, Wang CY, Smith AW, McTiernan A, Baumgartner RN, Baumgartner KB, Bernstein L, Ballard-Barbash R. Fasting C-peptide levels and death resulting from all causes and breast cancer: the health, eating, activity, and lifestyle study. Journal of clinical oncology : official journal of the American Society of Clinical Oncology. 2011;29(1):47–53. doi: 10.1200/JCO.2010.28.4752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Lyman GH. Weight-based chemotherapy dosing in obese patients with cancer: back to the future. J Oncol Pract. 2012;8(4):e62–64. doi: 10.1200/JOP.2012.000606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Lyman GH, Sparreboom A. Appropriate chemotherapy dosing in obese patients with cancer. Nat Rev Clin Oncol. 2013;10(11):664. doi: 10.1038/nrclinonc.2013.108-c2. [DOI] [PubMed] [Google Scholar]
  • 46.Mougalian SS, Hernandez M, Lei X, Lynch S, Kuerer HM, Symmans WF, Theriault RL, Fornage BD, Hsu L, Buchholz TA, Sahin AA, Hunt KK, Yang WT, Hortobagyi GN, Valero V. Ten-Year Outcomes of Patients With Breast Cancer With Cytologically Confirmed Axillary Lymph Node Metastases and Pathologic Complete Response After Primary Systemic Chemotherapy. JAMA Oncol. 2015:1–9. doi: 10.1001/jamaoncol.2015.4935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Keenan T, Moy B, Mroz EA, Ross K, Niemierko A, Rocco JW, Isakoff S, Ellisen LW, Bardia A. Comparison of the genomic landscape between primary breast cancer in African American versus white women and the association of racial differences with tumor recurrence. Journal of Clinical Oncology:JCO. 2015 doi: 10.1200/JCO.2015.62.2126. 2015.2062. 2126. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Masuda H, Baggerly KA, Wang Y, Zhang Y, Gonzalez-Angulo AM, Meric-Bernstam F, Valero V, Lehmann BD, Pietenpol JA, Hortobagyi GN, Symmans WF, Ueno NT. Differential response to neoadjuvant chemotherapy among 7 triple-negative breast cancer molecular subtypes. Clin Cancer Res. 2013;19(19):5533–5540. doi: 10.1158/1078-0432.CCR-13-0799. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Kalata P, Martus P, Zettl H, Rödel C, Hohenberger W, Raab R, Becker H, Liersch T, Wittekind C, Sauer R, Fietkau R, Group GRCS. Differences between clinical trial participants and patients in a population-based registry: the German Rectal Cancer Study vs. the Rostock Cancer Registry. Dis Colon Rectum. 2009;52(3):425–437. doi: 10.1007/DCR.0b013e318197d13c. [DOI] [PubMed] [Google Scholar]
  • 50.Loibl S, Jackisch C, Lederer B, Untch M, Paepke S, Kümmel S, Schneeweiss A, Huober J, Hilfrich J, Hanusch C. Outcome after neoadjuvant chemotherapy in young breast cancer patients: a pooled analysis of individual patient data from eight prospectively randomized controlled trials. Breast cancer research and treatment. 2015;152(2):377–387. doi: 10.1007/s10549-015-3479-z. [DOI] [PubMed] [Google Scholar]
  • 51.Boughey JC, McCall LM, Ballman KV, Mittendorf EA, Ahrendt GM, Wilke LG, Taback B, Leitch AM, Flippo-Morton T, Hunt KK. Tumor biology correlates with rates of breast-conserving surgery and pathologic complete response after neoadjuvant chemotherapy for breast cancer: findings from the ACOSOG Z1071 (Alliance) Prospective Multicenter Clinical Trial. Annals of Surgery. 2014;260(4):608–614. doi: 10.1097/SLA.0000000000000924. discussion 614–606. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES