ArticlePDF Available

Traditional Breast Cancer Risk Factors in Relation to Molecular Subtypes of Breast Cancer

Authors:

Abstract

At least four major categories of invasive breast cancer have been reproducibly identified by gene expression profiling: luminal A, luminal B, HER2-type, and basal-like. These subtypes have been shown to differ in their outcome and response to treatment. Whether this heterogeneity reflects the evolution of these subtypes through distinct etiologic pathways has not been clearly defined. We evaluated the association between traditional breast cancer risk factors and risk of previously defined molecular subtypes of breast cancer in the Nurses' Health Study. This analysis included 2,022 invasive breast cancer cases for whom we were able to obtain archived breast cancer tissue specimens. Tissue microarrays (TMAs) were constructed, and slides were immunostained for estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), cytokeratin 5/6 (CK5/6), and epidermal growth factor receptor (EGFR). Using immunostain results in combination with histologic grade, cases were grouped into molecularly defined subtypes. We used Cox proportional hazards models to estimate hazard ratios (HRs) and 95% confidence intervals (CIs). We observed differences in the association between risk factors and subtypes of breast cancer. In general, many reproductive factors were most strongly associated with the luminal A subtype, although these differences were not statistically significant. Weight gain since age 18 showed significant differences in its association with molecular subtypes (P-heterogeneity = 0.05) and was most strongly associated with the luminal B subtype (P-trend 0.001). Although there was not significant heterogeneity for lactation across subtypes, an inverse association was strongest for basal-like tumors (HR = 0.6, 95% CI 0.4-0.8; P-heterogeneity = 0.88). These results support the hypothesis that different subtypes of breast cancer have different etiologies and should not be considered as a single group. Identifying risk factors for less common subtypes such as luminal B, HER2-type and basal-like tumors has important implications for prevention of these more aggressive subtypes.
Traditional Breast Cancer Risk Factors in Relation to Molecular
Subtypes of Breast Cancer
Rulla M. Tamimi1,2, Graham A. Colditz3, Aditi Hazra1,2, Heather J. Baer1,2, Susan E.
Hankinson1,2, Bernard Rosner1, Jonathan Marotti4, James L. Connolly4, Stuart J. Schnitt4,
and Laura C. Collins4
1Channing Laboratory, Department of Medicine, Brigham and Women’s Hospital and Harvard
Medical School, Boston, MA, 02115
2Department of Epidemiology, Harvard School of Public Health, Boston, MA, 02115
3Department of Surgery, Washington University School of Medicine, St. Louis, MO 63110
4Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School,
Boston, MA 02115
Abstract
Background—At least four major categories of invasive breast cancer have been reproducibly
identified by gene expression profiling: luminal A, luminal B, HER2-type and basal-like. These
subtypes have been shown to differ in their outcome and response to treatment. Whether this
heterogeneity reflects the evolution of these subtypes through distinct etiologic pathways has not
been clearly defined.
Methods—We evaluated the association between traditional breast cancer risk factors and risk of
previously defined molecular subtypes of breast cancer in the Nurses’ Health Study. This analysis
included 2,022 invasive breast cancer cases for whom we were able to obtain archived breast
cancer tissue specimens. Tissue microarrays (TMAs) were constructed and slides were
immunostained for estrogen receptor (ER), progesterone receptor (PR), human epidermal growth
factor receptor 2 (HER2), cytokeratin 5/6 (CK5/6), and epidermal growth factor receptor (EGFR).
Using immunostain results in combination with histologic grade, cases were grouped into
molecularly defined subtypes. We used Cox proportional hazards models to estimate hazard ratios
(HRs) and 95% confidence intervals (CIs).
Results—We observed differences in the association between risk factors and subtypes of breast
cancer. In general, many reproductive factors were most strongly associated with the luminal A
subtype, although these differences were not statistically significant. Weight gain since age 18
showed significant differences in its association with molecular subtypes (p-heterogeneity=0.05)
and was most strongly associated with the luminal B subtype (p-trend 0.001). Although there was
not significant heterogeneity for lactation across subtypes, an inverse association was strongest for
basal-like tumors (HR=0.6, 95%CI 0.4–0.8; p-heterogeneity=0.88).
Conclusions—These results support the hypothesis that different subtypes of breast cancer have
different etiologies and should not be considered as a single group. Identifying risk factors for less
common subtypes such as luminal B, HER2-type and basal-like tumors has important implications
for prevention of these more aggressive subtypes.
Corresponding Author: Rulla M. Tamimi, Channing Laboratory, 181 Longwood Avenue, Boston, MA 02115, Telephone: (617)
525-0862, rulla.tamimi@channing.harvard.edu.
NIH Public Access
Author Manuscript
Breast Cancer Res Treat. Author manuscript; available in PMC 2013 January 1.
Published in final edited form as:
Breast Cancer Res Treat
. 2012 January ; 131(1): 159–167. doi:10.1007/s10549-011-1702-0.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Keywords
breast cancer; risk factors; molecular subtype; ER; PR; HER2
INTRODUCTION
Recent studies using microarray technology and unsupervised cluster analysis have provided
new insights into the classification of invasive breast cancers [1–4]. These studies have
resulted in the identification of several breast cancer subtypes that vary in their gene
expression signatures and clinical outcome. The molecularly distinct breast cancer
subgroups identified to date include luminal subtypes A and B (both of which are hormone
receptor-positive), the HER2-type, and a group known as basal-like cancers [1–4]. Although
prognosis and response to treatment has been shown to vary according to these subtypes, it
is unclear if classifying breast cancer in this way may help us to understand better the
etiology of breast cancer. Examination of breast cancer risk factors in relation to these
subtypes may offer the potential to build on traditional classification of tumor types based
on ER and PR status [5, 6] and extend insights into etiology.
Immunohistochemical staining of paraffin sections using antibody panels has been shown to
be a reasonable, albeit imperfect, surrogate for molecular classification of invasive breast
cancers as categorized by gene expression profiling studies [4, 7–10]. Antibodies against
estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor
2 (HER2), cytokeratin 5/6 (CK5/6) and epidermal growth factor receptor (EGFR) have been
particularly useful for this purpose in populations in which material for expression profiling
is not available or for which studying very large numbers of cases would not be feasible.
The heterogeneity of breast tumors may be a reflection of distinct etiologic pathways. For
example, risk factors that influence estrogen levels (i.e, circulating hormones,
postmenopausal hormone use and postmenopausal adiposity), are associated with ER-
positive, but not ER- negative breast cancer [5, 6, 11]. The additional markers used to
classify the molecular phenotypes beyond ER and PR may allow for greater refinement of
tumor subtypes and allow for identification of distinct etiologic pathways. Because luminal
A and B tumor subtypes are both hormone receptor positive and together represent the
majority of breast cancer cases it is not surprising that the majority of identified breast
cancer risk factors are those associated with hormonal exposures. By examining risk factors
in relation to more homogenous subtypes we may identify novel risk factors for less
common subtypes of breast cancer. In the current study, we examine the association between
traditional breast cancer risk factors and the risk of the previously defined molecular
subtypes of breast cancer.
MATERIALS AND METHODS
Study population
Study Design and Population—The Nurses’ Health Study was initiated in 1976 when
121,700 U.S. registered nurses ages 30–55 returned an initial questionnaire. The cohort has
been followed by mailed questionnaires biennially to update exposure information and
ascertain non-fatal incident diseases. Information on body mass index (BMI), reproductive
history, age at menopause, and postmenopausal hormone (PMH) use as well as diagnosis of
cancer and other diseases are updated every two years through questionnaires. The follow-
up rate among this cohort was over 90% through 1996[12].
Tamimi et al. Page 2
Breast Cancer Res Treat. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Breast cancer case confirmation
All women reporting incident diagnoses of cancer were asked for permission to review their
medical records to confirm the diagnosis and to classify cancers as in situ or invasive, by
histologic type, size, and presence or absence of metastases. To identify cases of cancer in
nonrespondents who died, death certificates for all deceased participants and medical
records for the incident cancers were obtained. Following medical record review, 99% of
self-reported breast cancers were confirmed.
Breast cancer tissue block collection and tissue microarray (TMA) construction
Detailed description of the tissue block collection and TMA construction have been
described previously [13, 14]. Briefly, we constructed tissue microarrays from 3,093 cancers
and positive lymph nodes from 2,897 participants. TMAs were constructed in the Dana
Farber Harvard Cancer Center Tissue Microarray Core Facility, Boston, MA. Three 0.6-mm
cores were obtained from each breast cancer and were inserted into the recipient TMA
blocks. We excluded from the current analysis participants with positive lymph nodes only
(n=25), lobular carcinoma in situ (n=31), in situ carcinomas with both ductal and lobular
features (n=13), ductal carcinoma in situ (n=272), and additional rare tumor types including
malignant phyllodes tumors, neuroendocrine carcinoma and angiosarcoma (n=10).
Immunohistochemical analysis
We performed immunohistochemical staining for ER, PR, HER2, CK5/6, and EGFR on 5-
μm paraffin sections cut from the TMA blocks. Immunostains for each marker were
performed in a single staining run on a Dako Autostainer (Dako Corporation, Carpinteria,
CA). These particular biomarkers were selected for analysis because they have been
commonly used as surrogates to classify invasive breast cancers according to their molecular
phenotypes [4, 7–10]. Sources and dilutions of the primary antibodies used in this study are
listed in Appendix 1. Immunostains for ER, PR, HER2, CK 5/6 and EGFR were performed
using methods described in detail previously [13, 15]. Appropriate positive and negative
controls were included in all staining runs.
Immunostained TMA slides were evaluated for ER and PR expression, HER2 protein over-
expression, and expression of CK5/6 and EGFR in each core. Tumor cells that showed any
nuclear staining for ER or PR were considered ER-positive or PR-positive respectively,
whereas all ER-negative or PR-negative cases showed complete absence of tumor cell
staining in all tissue cores. Of note, low positive ER or PR (1–10% of tumor cell nuclei
staining) and positive ER or PR (>10% of tumor cell nuclei staining) were collapsed into a
single “positive” category for the purposes of this analysis. Tumor cells were considered
positive for HER2 protein over-expression when more than 10% of the cells showed
moderate or strong membrane staining (2+ and 3+). The results of analyses in which HER2
positivity was defined as 3+ were very similar to those presented with a definition of 2+ and
3+. Cases were considered basal CK-positive or EGFR-positive if any cytoplasmic and/or
membranous staining was detected in the tumor cells, even if focal. These latter criteria are
similar to those previously used for scoring these markers in invasive basal-like cancers [4,
7, 8].
Classification of Molecular Subtypes
Immunostained TMA sections were reviewed under a microscope and visually scored for
each individual tissue core as described above. We classified a case as positive if there was
staining in any of the three cores from that case and negative if there was no
immunostaining present. Based on RNA expression data [1–3] and previous large scale
epidemiologic studies [16, 17] we used a panel of immunohistochemical markers to classify
Tamimi et al. Page 3
Breast Cancer Res Treat. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
the tumors into molecular subtypes. Cases that were ER-positive and/or PR-positive and
HER2-negative and histologic grade 1 and 2 were classified as luminal A cancers; cases that
were either a.) ER-positive and/or PR-positive and HER2-positive or b.) ER-positive and/or
PR-positive, HER2-negative and histologic grade 3 were classified as luminal B; cases that
were ER-negative, PR-negative, and HER2-positive were classified as HER2 type; and cases
that were negative for ER, PR, and HER2 and positive for CK 5/6 and/or EGFR were
categorized as basal-like. Cases that lacked expression of all 5 markers were considered
“unclassified”. Of the invasive tumors on tissue microarrays, 2249 could be classified into
one of these 5 molecular subtypes.
Statistical Analysis
Information on breast cancer risk factors was obtained from biennial questionnaires. Women
who reported a diagnosis of cancer other than nonmelanoma skin cancer were excluded at
baseline and from subsequent follow-up analysis. Person-time for each participant was
calculated from the date of return of the 1976 questionnaire to the date of breast cancer
diagnosis, date of any other cancer diagnosis (not including nonmelanoma skin cancer),
death from any cause or June 1, 1998, which ever came first.
The primary analysis used incidence rates with person-months in the denominator.. Person-
time for each participant was calculated from the date of return of the 1976 questionnaire to
the date of breast cancer diagnosis, date of any other cancer diagnosis (not including
nonmelanoma skin cancer), death from any cause or June 1, 1998, whichever came first. The
primary analysis used incidence rates with person-months in the denominator. For each
woman, person-months were allocated to each exposure category, beginning in 1976 and
updated every two years. We used Cox proportional hazards models to estimate hazard
ratios (RRs) and 95% confidence intervals (CIs). Multivariate analysis included age at
menopause, family history of breast cancer in a first-degree relative, personal history of
benign breast disease (BBD), body mass index (BMI) at age 18, weight change since age 18,
age at menarche, parity/age at first birth, alcohol consumption, menopausal status/PMH use,
lactation, and smoking. These variables were considered because they are either well
established risk factors for breast cancer [18] or have been reported to be associated with a
particular molecular phenotype of breast cancer[16, 17].
To determine if the association between exposures is differentially associated with tumor
subtypes, we used competing risks models[19, 20]. Specifically, this approach uses the data
augmentation method described by Lunn and McNeil [21] to create a separate observation
for each subject for each type of outcome and then stratifies on event type, allowing for
estimation of separate associations of each risk factor with the relative hazard of each type
of outcome [20]. Likelihood ratio tests were conducted to compare models assuming
different associations of exposures with each subtype of tumor to models assuming the same
association with all types; a significant p value for this test of heterogeneity would indicate
that the associations are different for the different tumor subtypes. All analyses were
conducted with SAS version 9.2 (SAS, Cary, North Carolina). All statistical tests were two-
sided and p-values <0.05 were considered statistically significant. This study was approved
by the Committee on the Use of Human Subjects in Research at Brigham and Women’s
Hospital.
RESULTS
During the course of follow-up, 2,022 invasive breast cancer cases were identified that could
be classified into one of five molecular phenotypes. Based on immunostaining data from the
five markers used in conjunction with histologic grade (which was used as a surrogate for
Ki67 proliferation index), 1,267 tumors were classified as luminal A; 321 were luminal B;
Tamimi et al. Page 4
Breast Cancer Res Treat. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
113 were HER2; 226 were basal-like and 95 tumors were unclassifiable (ER-/PR-/HER2-/
EGFR-/CK5/6-). In addition, 3,549 breast cancers occurred among women for whom we
were unable to classify into these phenotypes due to lack of either tissue availability or
staining results for one or more of the five markers. Compared with women with tissue
specimens, the women for whom we were unable to obtain specimens were very similar
with respect to breast cancer risk factors and tumor characteristics [13].
The mean age at diagnosis ranged from 55.3 years for women with basal like tumors to 58.0
years for women with luminal A tumors (Table 1). In general, the luminal A tumors were
smaller, less likely to have nodal involvement, were of lower stage and were more often of
lower histologic grade than the other four molecular subtypes. The luminal B and basal like
tumors were more often high grade, while the HER2 type was most likely to be stage III/IV
(33.0%) and the unclassified type was most likely to be metastatic at diagnosis (8.1%)
relative to the other subtypes.
We observed differences in the association between breast cancer risk factors and molecular
subtypes of breast cancer (Table 2). In general, reproductive risk factors for breast cancer
including age at menarche, parity, and age at first birth tended to be most strongly associated
with the luminal A subtype, although there was no evidence of statistical heterogeneity
across the subtypes. For example, compared to nulliparous women, having three or more
children was inversely associated with luminal A breast cancer (HR=0.7, 95%CI 0.5–1.0; p-
trend=0.01). However, this inverse association was not observed among the other subtypes.
We found that hormonal exposures in later adult life exhibited the greatest heterogeneity in
association with these molecular subtypes. Weight gain since age 18 was positively
associated with both luminal A (p-trend=0.05) and B tumor (p-trend=0.001) subtypes, but
not with the ER- subtypes (p-heterogeneity=0.05). The association between weight gain
since age 18 and luminal B tumors was significantly stronger than the association with
luminal A tumors (p-heterogeneity=0.0007). There was suggestive evidence that the
association between postmenopausal hormone use and breast cancer may also vary by
subtype (p-heterogeneity=0.08). We observed a significant association between estrogen
only hormone therapy (HR=1.4, 95%CI 1.1–1.7) and estrogen plus progestin therapy
(HR=1.5, 95%CI 1.2–2.0) and risk of luminal A tumors, but not luminal B tumors (P-
heterogeneity=0.004). Estrogen plus progestin therapy was also associated with basal-like
(HR=1.8, 95% CI 1.0–3.4) and unclassified (HR=2.9, 95% CI 1.1–7.6) tumors, but not
luminal B or HER2-type.
Family history of breast cancer was differentially associated with breast cancer subtypes
(p=0.01). Having one first degree relative with breast cancer was significantly associated
with luminal A and B subtypes only, while having two first degree relatives with breast
cancer was associated with an increased risk of luminal A (HR=2.3, 95% CI 1.3–4.2),
HER2-type (HR=2.5, 95% CI 0.3–18.1), and basal-like (HR=2.9, 95% CI 0.7–11.7) tumors,
and not the others.
There was little evidence that the association between the other risk factors we considered
differed by molecular subtypes. Having a prior benign breast disease was associated with a
20–70% increased risk of all breast cancer subtypes (p-heterogeneity=0.69). BMI at age 18
was inversely associated with luminal A, basal-like and unclassified subtypes of breast
cancer (p-heterogeneity=0.49). Although there were no significant differences in the
association between lactation across molecular subtypes, an inverse association was
strongest for the basal-like tumors (HR=0.6, 95%CI 0.4–0.9; p-heterogeneity=0.88).
Tamimi et al. Page 5
Breast Cancer Res Treat. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
DISCUSSION
In this study of over 2,000 breast cancer cases, we found significant differences in the
association between breast cancer risk factors and molecular subtypes of tumors. As
expected many reproductive risk factors including age at menarche, parity, age at first birth,
age at menopause, and postmenopausal hormone use were associated with luminal A
tumors, the most common type of breast of cancer. In general, hormonally related risk
factors in later adult life demonstrated the most heterogeneity. The association between
postmenopausal hormone use and luminal A tumors was significantly stronger than the
relation with luminal B tumors (the other ER+ subtype). Interestingly, weight gain since age
18 was more strongly associated with luminal B tumors than luminal A tumors.
Unexpectedly, we found that some hormonal factors were associated with hormone receptor
negative subtypes. For example, age at menopause was significantly associated with the
HER2-type and unclassified subtype, and current estrogen plus progestin hormone use was
strongly associated with both basal-like and unclassified tumor types.
A number of other studies have evaluated the association between breast cancer risk factors
and tumor subtypes, although only a handful have evaluated markers beyond ER, PR and
HER2. The Polish Breast Cancer Study (n=804 breast cancer cases) also found that most
established breast cancer risk factors were associated with luminal A tumors [16]. However,
they reported that age at menarche was more strongly inversely associated with basal-like
tumors than luminal A tumors (p-heterogeneity =0.0009), which we did not observe in the
current study. Similar to our study, the Polish Breast Cancer Study found that having a
family history of breast cancer was a risk factor for almost all subtypes although the
magnitude of the effect was greatest for basal-like and HER2-type breast cancers.
The Carolina Breast Cancer Study (CBCS) (n=1424 breast cancer cases) also examined the
association between risk factors for molecular subtypes of breast cancer in a case-control
study of both Caucasian and African American women[17]. Millikan et al. found that
increasing parity was associated with reduced risk of luminal A tumors and an increased risk
of basal-like tumors. Our results with respect to parity are consistent with this finding. In
addition, The CBCS reported an inverse association between lactation and basal-like tumors.
Although there was no significant heterogeneity between lactation and subtype in our study,
we did find a strong inverse association between lactation and basal-like tumors. For women
with total breast feeding of 4+ months, we found a 40% reduced risk of basal-like breast
cancer, which is in line with the 30% reduced risk observed in the CBCS.
In addition, studies examining risk factors in relation to tumors classified with information
on ER, PR and HER2 only have also been conducted. A combined study of the LACE and
Pathways studies within Kaiser Permanente Northern California examined breast cancer risk
factors in relation to subtypes defined by ER, PR, and HER2. In this study of 2544 invasive
breast cancer cases, Kwan et al [22]found that relative to luminal A cases (ER+ and/or PR+/
HER2-), luminal B cases (ER+ and/or PR+/HER2+) were less likely to consume alcohol and
use HRT. Breast feeding for at least four months was associated with a lower risk of triple
negative cases (ER-/PR-/HER2-) compared with luminal A. Similarly, two other studies
Phipps et al [23](n=1130 total cases) and Gaudet et al [24](n=890 total cases), also reported
an inverse association between breastfeeding for 6 or more months and triple negative breast
tumors.
Of interest, a number of risk factors in our study did not demonstrate heterogeneity across
tumor subtypes including age at menarche, BMI at age 18, previous BBD, and alcohol
consumption. It is possible that these factors are having a similar effect on risk across the
different subtypes and this may be indicating how these factors are affecting breast cancer
Tamimi et al. Page 6
Breast Cancer Res Treat. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
etiology. For example, having a prior BBD may indicate having early proliferative lesions
which could have developed through a number of different pathways. BBD is believed to be
a general marker of breast cancer risk, and thus may reflect the culmination of many risk
factors and not be specific to any one pathway. It is also possible that we may not have had
enough power to detect the difference across subtypes for some exposures.
Our classification of tumor subtypes was similar although not identical to those used in
previous epidemiologic studies [16, 17]. Both of the prior studies utilized
immunohistochemical markers to define molecular subtypes, while we also incorporated
histologic grade. Others have shown that the distinction between luminal A and B tumors
can be refined by adding the proliferation marker Ki67 to ER, PR, and HER2[25]. Given
that Ki67 data were not available for our cases, we used histologic grade as a surrogate for
proliferation rate given the close correlation between proliferation rate and histologic grade.
Thus, our definitions for luminal A and B are different than the two previous studies, but
more in keeping with the most recently proposed classification scheme[25]. This may limit
our ability to compare across studies and explain some of the differences observed.
The results of the current study taken together with the previous studies suggest that many of
the traditional breast cancer risk factors are associated with luminal A tumors, and that there
may be some differences with other subtypes. The associations with luminal A tumors is not
surprising since these are the most common tumor type and may also reflect the selection of
exposures we have focused in the current study. We have chosen to examine traditional
breast cancer risk factors; these were initially identified because they are the risk factors
shown to be most commonly associated with breast cancer. The majority of these risk
factors have also been shown to be associated with the most common type of breast cancer,
namely ER+ breast cancers. While it is reassuring that these risk factors are associated with
the luminal A subtype, it does not help us to further our understanding of risk factors
associated with ER- subtypes. Adding the additional markers and dividing the cases into
smaller subsets limits our power to detect associations for the rare subtypes. One of the most
consistent findings from this study and other studies is the inverse association between
breast feeding and triple negative or basal like tumors. This finding supports the hypothesis
that molecular classification of tumors may help us to better understand etiology and/or
provide insights into the mechanisms by which these less common molecular subtypes
develop.
This study has a number of strengths including the large study population with over 2,000
invasive breast cancer cases, the prospectively collected nature of the exposure variables,
and uniform staining and scoring of molecular markers. Despite the large number of cases,
we were still limited by the number of less common subtypes in particular the HER2-type
and the unclassified tumors. Thus, our power to detect significant differences by these
subtypes was limited. In addition, we lacked adequate power to examine these associations
among premenopausal breast cancer cases. It is worth noting that the frequency of receptor
status positivity and molecular subtype frequency among invasive tumors in our study
population was very similar to other populations suggesting that samples included in this
study are representative of the overall US population.
In conclusion, in this study we found that traditional breast cancer risk factors demonstrated
different relations with the molecular subtypes of breast cancer. In general, many of the
reproductive factors were most strongly associated with the luminal A subtype. In addition,
we confirmed a previously reported strong inverse association between lactation and basal-
like tumors. It is unclear whether classifying breast tumors according to the molecular
phenotypes (ie. subtypes that are known to have prognostic importance) permits
identification of differences in risk factor profiles. Additional work to determine if the
Tamimi et al. Page 7
Breast Cancer Res Treat. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
differences that have been observed are due to single markers or to the molecular phenotype
is necessary. It remains to be seen whether non-traditional breast cancer risk factors may
exist that are associated with less common tumor subtypes. Identifying risk factors for these
less common subtypes such as HER2 and basal-like tumors, which also have a poorer
prognosis, has important implications for prevention of these tumor subtypes.
Acknowledgments
Funding/Support: Supported by GlaxoSmithKline (WE234 (EPI40307)); Public Health Service Grants CA087969,
and SPORE in Breast Cancer CA089393, from the National Cancer Institute, National Institutes of Health,
Department of Health and Human Services and Breast Cancer Research Fund. Dr. Graham Colditz is supported in
part by an American Cancer Society Cissy Hornung Clinical Research Professorship.
We thank the participants and staff of the Nurses' Health Study cohort, for their valuable contributions. We thank
the following state cancer registries for their help: AL, AZ, AR, CA, CO, CT, DE, FL, GA, ID, IL, IN, IA, KY, LA,
ME, MD, MA, MI, NE, NH, NJ, NY, NC, ND, OH, OK, OR, PA, RI, SC, TN, TX, VA, WA, WY. The study
sponsors had no role in the design of the study; the collection, analysis, and interpretation of the data; the writing of
the manuscript; or the decision to submit the manuscript for publication.
References
1. Perou CM, Jeffrey SS, van de Rijn M, Rees CA, Eisen MB, Ross DT, Pergamenschikov A,
Williams CF, Zhu SX, Lee JC, et al. Distinctive gene expression patterns in human mammary
epithelial cells and breast cancers. Proc Natl Acad Sci U S A. 1999; 96(16):9212–9217. [PubMed:
10430922]
2. Sorlie T, Perou CM, Tibshirani R, Aas T, Geisler S, Johnsen H, Hastie T, Eisen MB, van de Rijn M,
Jeffrey SS, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with
clinical implications. Proc Natl Acad Sci U S A. 2001; 98(19):10869–10874. [PubMed: 11553815]
3. Sorlie T, Tibshirani R, Parker J, Hastie T, Marron JS, Nobel A, Deng S, Johnsen H, Pesich R,
Geisler S, et al. Repeated observation of breast tumor subtypes in independent gene expression data
sets. Proc Natl Acad Sci U S A. 2003; 100(14):8418–8423. [PubMed: 12829800]
4. Brenton JD, Carey LA, Ahmed AA, Caldas C. Molecular classification and molecular forecasting of
breast cancer: ready for clinical application? J Clin Oncol. 2005; 23 (29):7350–7360. [PubMed:
16145060]
5. Colditz GA, Rosner BA, Chen WY, Holmes MD, Hankinson SE. Risk factors for breast cancer
according to estrogen and progesterone receptor status. J Natl Cancer Inst. 2004; 96(3):218–228.
[PubMed: 14759989]
6. Ma H, Bernstein L, Pike MC, Ursin G. Reproductive factors and breast cancer risk according to
joint estrogen and progesterone receptor status: a meta-analysis of epidemiological studies. Breast
Cancer Res. 2006; 8(4):R43. [PubMed: 16859501]
7. Nielsen TO, Hsu FD, Jensen K, Cheang M, Karaca G, Hu Z, Hernandez-Boussard T, Livasy C,
Cowan D, Dressler L, et al. Immunohistochemical and clinical characterization of the basal-like
subtype of invasive breast carcinoma. Clin Cancer Res. 2004; 10(16):5367–5374. [PubMed:
15328174]
8. Abd El-Rehim DM, Ball G, Pinder SE, Rakha E, Paish C, Robertson JF, Macmillan D, Blamey RW,
Ellis IO. High-throughput protein expression analysis using tissue microarray technology of a large
well-characterised series identifies biologically distinct classes of breast cancer confirming recent
cDNA expression analyses. Int J Cancer. 2005; 116(3):340–350. [PubMed: 15818618]
9. Abd El-Rehim DM, Pinder SE, Paish CE, Bell J, Blamey RW, Robertson JF, Nicholson RI, Ellis IO.
Expression of luminal and basal cytokeratins in human breast carcinoma. J Pathol. 2004; 203(2):
661–671. [PubMed: 15141381]
10. van de Rijn M, Perou CM, Tibshirani R, Haas P, Kallioniemi O, Kononen J, Torhorst J, Sauter G,
Zuber M, Kochli OR, et al. Expression of cytokeratins 17 and 5 identifies a group of breast
carcinomas with poor clinical outcome. Am J Pathol. 2002; 161(6):1991–1996. [PubMed:
12466114]
Tamimi et al. Page 8
Breast Cancer Res Treat. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
11. Missmer SA, Eliassen AH, Barbieri RL, Hankinson SE. Endogenous estrogen, androgen, and
progesterone concentrations and breast cancer risk among postmenopausal women. J Natl Cancer
Inst. 2004; 96(24):1856–1865. [PubMed: 15601642]
12. Colditz GA, Hankinson SE. The Nurses' Health Study: lifestyle and health among women. Nat Rev
Cancer. 2005; 5(5):388–396. [PubMed: 15864280]
13. Tamimi RM, Baer HJ, Marotti J, Galan M, Galaburda L, Fu Y, Deitz AC, Connolly JL, Schnitt SJ,
Colditz GA, et al. Comparison of molecular phenotypes of ductal carcinoma in situ and invasive
breast cancer. Breast Cancer Res. 2008; 10(4):R67. [PubMed: 18681955]
14. Collins LC, Marotti JD, Baer HJ, Tamimi RM. Comparison of estrogen receptor results from
pathology reports with results from central laboratory testing. J Natl Cancer Inst. 2008; 100(3):
218–221. [PubMed: 18230800]
15. Collins LC, Botero ML, Schnitt SJ. Bimodal frequency distribution of estrogen receptor
immunohistochemical staining results in breast cancer: An analysis of 825 cases. Am J Clin
Pathol. 2005; 123:16–20. [PubMed: 15762275]
16. Yang XR, Sherman ME, Rimm DL, Lissowska J, Brinton LA, Peplonska B, Hewitt SM, Anderson
WF, Szeszenia-Dabrowska N, Bardin-Mikolajczak A, et al. Differences in risk factors for breast
cancer molecular subtypes in a population-based study. Cancer Epidemiol Biomarkers Prev. 2007;
16(3):439–443. [PubMed: 17372238]
17. Millikan RC, Newman B, Tse CK, Moorman PG, Conway K, Smith LV, Labbok MH, Geradts J,
Bensen JT, Jackson S, et al. Epidemiology of basal-like breast cancer. Breast Cancer Res Treat.
2008; 109(1):123–139. [PubMed: 17578664]
18. Hankinson, S.; Hunter, DJ. Breast Cancer. In: Adami, HOHD.; Trichopoulos, D., editors. Textbook
of Cancer Epidemiology. New York: Oxford University Press; 2002. p. 301-339.
19. Glynn RJ, Rosner B. Methods to evaluate risks for composite end points and their individual
components. J Clin Epidemiol. 2004; 57(2):113–122. [PubMed: 15125620]
20. Glynn RJ, Rosner B. Comparison of risk factors for the competing risks of coronary heart disease,
stroke, and venous thromboembolism. Am J Epidemiol. 2005; 162(10):975–982. [PubMed:
16207808]
21. Lunn M, McNeil D. Applying Cox regression to competing risks. Biometrics. 1995; 51 (2):524–
532. [PubMed: 7662841]
22. Kwan ML, Kushi LH, Weltzien E, Maring B, Kutner SE, Fulton RS, Lee MM, Ambrosone CB,
Caan BJ. Epidemiology of breast cancer subtypes in two prospective cohort studies of breast
cancer survivors. Breast Cancer Res. 2009; 11(3):R31. [PubMed: 19463150]
23. Phipps AI, Malone KE, Porter PL, Daling JR, Li CI. Reproductive and hormonal risk factors for
postmenopausal luminal, HER-2-overexpressing, and triple-negative breast cancer. Cancer. 2008;
113(7):1521–1526. [PubMed: 18726992]
24. Gaudet MM, Press MF, Haile RW, Lynch CF, Glaser SL, Schildkraut J, Gammon MD, Douglas
Thompson W, Bernstein JL. Risk factors by molecular subtypes of breast cancer across a
population-based study of women 56 years or younger. Breast Cancer Res Treat. 2011 Jun 11.
[Epub ahead of print].
25. Cheang MC, Chia SK, Voduc D, Gao D, Leung S, Snider J, Watson M, Davies S, Bernard PS,
Parker JS, et al. Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J
Natl Cancer Inst. 2009; 101(10):736–750. [PubMed: 19436038]
Tamimi et al. Page 9
Breast Cancer Res Treat. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Tamimi et al. Page 10
Table 1
Tumor characteristics according to breast cancer phenotypes among women with invasive breast cancer, Nurses’ Health Study (1976–1996).
Characteristic Luminal A Luminal B HER2 Basal-like Unclassified
N (%) 1267 (62.7) 321 (15.9) 113 (5.6) 226 (11.2) 95 (4.7)
Mean age at diagnosis, yrs 58.0 56.5 56.1 55.3 55.6
Median age at diagnosis, yrs 59.0 57.0 56.0 56.0 55.0
Tumor size1
0.1 to 1.0 cm 323 (26.7) 47 (15.3) 18 (17.0) 27 (12.9) 19 (21.6)
1.1 to 2.0 cm 503 (41.6) 116 (37.8) 27 (25.5) 74 (35.2) 31 (35.2)
2.1 to 4.0 cm 281 (23.2) 100 (32.6) 44 (41.5) 87 (41.4) 24 (27.3)
4.1+ cm 103 (8.5) 44 (14.3) 17 (16.0) 22 (10.5) 14 (15.9)
Missing 57 14 7 16 7
Lymph node status1
No Nodes 787 (66.3) 165 (55.4) 47 (43.9) 119 (56.9) 50 (57.5)
1–3 Nodes 234 (19.7) 65 (21.8) 28 (26.2) 54 (25.8) 14 (16.1)
4–9 Nodes 86 (7.2) 38 (12.8) 14 (13.1) 22 (10.5) 9 (10.3)
10+ Nodes 55 (4.6) 23 (7.7) 13 (12.2) 7 (3.4) 7 (8.1)
Metastatic at diagnosis 26 (2.2) 7 (2.4) 5 (4.7) 7 (3.4) 7 (8.1)
Missing 79 23 6 17 8
Stage at diagnosis1
I/II 1026 (84.6) 230 (74.7) 71 (67.0) 173 (80.8) 66 (73.3)
III/IV 187 (15.4) 78 (25.3) 35 (33.0) 41 (19.2) 24 (26.7)
Missing 54 13 7 12 5
Grade2
Well differentiated 366 (28.9) 8 (2.7) 3 (2.8) 7 (3.1) 16 (16.8)
Int. differentiated 901 (71.1) 57 (18.9) 54 (49.5) 57 (25.3) 37 (39.0)
Poorly differentiated 0 237 (78.5) 52 (47.7) 161 (71.6) 42 (44.2)
Missing 0 19 4 1 0
Histology1
Invasive ductal 1002(79.4) 300 (94.0) 111 (99.1) 210 (94.6) 79 (85.9)
Invasive lobular 180 (14.3) 10 (3.1) 0 3 (1.4) 8 (8.7)
Breast Cancer Res Treat. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Tamimi et al. Page 11
Characteristic Luminal A Luminal B HER2 Basal-like Unclassified
N (%) 1267 (62.7) 321 (15.9) 113 (5.6) 226 (11.2) 95 (4.7)
Invasive ductal and lobular 70 (5.6) 5 (1.6) 0 0 2 (2.2)
Invasive not specified 10 (0.8) 4 (1.3) 1 (0.9) 9 (4.1) 3 (3.3)
Missing 5 2 1 4 3
Race
White 1209 (95.4) 304 (94.7) 109 (96.5) 210 (92.9) 90 (94.7)
Black 11 (0.9) 7 (2.2) 1 (0.9) 8 (3.5) 2 (2.1)
American Indian 2 (0.2) 0 0 0 0
Asian 4 (0.3) 0 1 (0.9) 2 (0.9) 0
Hawaiian 0 0 0 1 (0.4) 0
Other 31 (2.5) 8 (2.5) 0 4 (1.8) 3 (3.2)
Multiracial 10 (0.8) 2 (0.6) 2 (1.8) 1 (0.4) 0
1Information obtained from pathology records
2Information obtained from centralized pathology review of slides
Breast Cancer Res Treat. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Tamimi et al. Page 12
Table 2
Relative risk1 of invasive breast cancer molecular subtypes breast cancer according to breast cancer risk factors
Luminal A Luminal B HER2 Basal Unclassified
Cases 1267 321 113 226 95
Person-years 2008898 2009767 2009968 2009865 2009986
Age at menarche
<12 1.0 (ref) 1.0 (Ref) 1.0 (Ref) 1.0 (Ref) 1.0 (Ref)
12 1.0 (0.8–1.1) 1.1 (0.8–1.5) 1.4 (0.8–2.3) 0.8 (0.5–1.1) 0.8 (0.4–1.3)
13 0.8 (0.7–1.0) 1.1 (0.8–1.5) 0.9 (0.5–1.5) 0.7 (0.5–1.0) 0.5 (0.3–0.9)
14 0.7 (0.6–0.8) 1.0 (0.7–1.5) 0.9 (0.4–1.8) 0.8 (0.5–1.3) 0.6 (0.3–1.2)
14+ 0.7 (0.5–0.9) 1.0 (0.6–1.6) 1.1 (0.5–2.3) 0.8 (0.5–1.3) 0.8 (0.4–1.7)
P for Trend 0.002 0.14 0.31 0.72 0.11
P-Heterogeneity 0.92
BMI at age 18
<20 1.0 (ref) 1.0 (ref) 1.0 (ref) 1.0 (Ref) 1.0 (Ref)
20–21.9 0.9 (0.7–1.0) 1.0 (0.8–1.3) 0.6 (0.4–1.0) 0.8 (0.5–1.1) 0.5 (0.3–0.9)
22.0–23.9 0.8 (0.7–1.0) 0.8 (0.6–1.2) 0.7 (0.4–1.3) 0.9 (0.6–1.3) 0.6 (0.3–1.2)
24.0–26.9 0.6 (0.5–0.8) 1.0 (0.6–1.5) 0.5 (0.2–1.1) 0.7 (0.4–1.3) 0.8 (0.4–1.8)
27- 0.5 (0.4–0.8) 0.7 (0.3–1.4) 0.6 (0.2–1.7) 0.4 (0.1–1.1) 0.2 (0.0–1.2)
P for Trend <0.0001 0.56 0.27 0.04 0.02
P-Heterogeneity 0.49
Weight Gain since 18
Loss <2 kg 0.9 (0.6–1.1) 0.9 (0.5–1.7) 1.1 (0.4–2.9) 0.8 (0.4–1.7) 3.1 (0.8–11.9)
Stable 1.0 (ref) 1.0 (Ref) 1.0 (ref) 1.0 (ref) 1.0 (Ref)
Gain 2.1–5kg 0.9 (0.7–1.1) 0.9 (0.5–1.5) 0.6 (0.2–1.6) 1.4 (0.8–2.5) 3.1 (0.9–10.8)
Gain 5.1–10kg 0.9 (0.7–1.1) 1.1 (0.7–1.8) 1.4 (0.6–3.0) 1.1 (0.6–2.0) 2.6 (0.7–8.8)
Gain 10.1–20kg 0.9 (0.8–1.2) 1.3 (0.8–2.1) 1.3 (0.6–2.7) 1.2 (0.7–2.1) 3.5 (1.1–11.5)
Gain 20.1–25kg 0.9 (0.7–1.3) 1.2 (0.7–2.1) 1.3 (0.5–3.3) 1.0 (0.5–2.2) 2.4 (0.6–9.6)
Gain 25+ kg 1.1 (0.8–1.4) 1.5 (0.9–2.5) 1.2 (0.5–3.2) 1.5 (0.8–2.8) 2.5 (0.6–9.6)
P for trend 0.05 0.001 0.31 0.11 0.35
Breast Cancer Res Treat. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Tamimi et al. Page 13
Luminal A Luminal B HER2 Basal Unclassified
P-Heterogeneity 0.05
Parity
Nulliparous 1.0 (Ref) 1.0 (Ref) 1.0 (Ref) 1.0 (Ref) 1.0 (Ref)
1 child 0.8 (0.6–1.3) 2.2 (0.8–5.9) 1.1 (0.3–4.2) 0.6 (0.2–1.8) 1.4 (0.3–8.2)
2 children 0.7 (0.5–1.1) 1.2 (0.5–3.0) 1.0 (0.3–3.5) 0.9 (0.3–2.6) 1.1 (0.2–5.4)
3+ children 0.7 (0.5–1.0) 1.1 (0.5–2.6) 0.7 (0.2–2.3) 1.1 (0.4–2.9) 1.4 (0.3–6.0)
P for trend 0.01 0.01 0.12 0.04 (+) 0.65
P-Heterogeneity 0.60
Age at first birth
Per 1 year increase 1.018 (1.007–1.030) 0.994 (0.964–1.024) 1.024 (0.991–1.059) 1.002 (0.968–1.037) 1.006 (0.959–1.056)
P-Heterogeneity 0.36
Age at Menopause
Per year increase 1.039 (1.021–1.058) 1.067(1.024–1.111) 1.075 (1.006–1.150) 1.012 (0.972–1.053) 1.089 (1.006–1.179)
P-Heterogeneity 0.10
Previous BBD
No 1.0 (Ref) 1.0 (ref) 1.0 (ref) 1.0 (Ref) 1.0 (Ref)
Yes 1.4 (1.2–1.5) 1.7 (1.3–2.2) 1.2 (0.8–1.9) 1.5 (1.1–2.0) 1.5 (0.9–2.4)
P-Heterogeneity 0.69
Menopausal status/PMH Use
Premenopausal 1.2 (1.0–1.5) 1.5 (1.0–2.2) 1.1 (0.6–2.1) 1.1 (0.7–1.9) 1.7 (0.8–3.7)
Post Never Use 1.0(REF) 1.0 (Ref) 1.0 (Ref) 1.0 (Ref) (Ref)
Post Past Use 0.9 (0.8–1.1) 1.2 (0.8–1.7) 1.3 (0.7–2.4) 0.8 (0.5–1.4) 1.5 (0.7–3.2)
Post Current E only 1.4 (1.1–1.7) 1.0 (0.7–1.7) 1.1 (0.6–2.3) 1.4 (0.8–2.3) 2.0 (0.9–4.4)
Post Current E+P 1.5 (1.2–2.0) 1.3 (0.8–2.4) 0.3 (0.0–2.2) 1.8 (1.0–3.4) 2.9 (1.1–7.6)
P-Heterogeneity 0.08
Alcohol Consumption
None 1.0 (Ref) 1.0 (Ref) 1.0 (Ref) 1.0 (Ref) 1.0 (Ref)
<5 g/week 1.1 (1.0–1.3) 0.8 (0.6–1.1) 1.1 (0.6–1.8) 0.8 (0.6–1.2) 0.5 (0.3–1.0)
5–10 1.0 (0.8–1.3) 1.1 (0.7–1.7) 1.3 (0.7–2.7) 0.7 (0.4–1.3) 0.9 (0.5–1.9)
Breast Cancer Res Treat. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Tamimi et al. Page 14
Luminal A Luminal B HER2 Basal Unclassified
10–15 1.2(1.0–1.5) 0.8 (0.5–1.3) 1.4 (0.7–2.8) 0.7 (0.4–1.2) 0.6 (0.3–1.3)
15+ 1.3 (1.0–1.6) 1.0 (0.7–1.5) 1.4 (0.7–2.8) 0.8 (0.5–1.4) 0.8 (0.4–1.6)
P-for trend 0.04 0.91 0.13 0.31 0.58
P-Heterogeneity 0.32
Lactation
Never 1.0 (Ref) 1.0 (Ref) 1.0 (ref) 1.0 (Ref) 1.0 (Ref)
0–3 months 0.8 (0.7–0.9) 1.0 (0.7–1.4) 0.8 (0.4–1.3) 0.8 (0.6–1.2) 1.0 (0.6–1.8)
4+ months 0.8 (0.7–1.0) 0.8 (0.6–1.1) 0.9 (0.6–1.5) 0.6 (0.4–0.9) 0.6 (0.4–1.1)
P-Heterogeneity 0.88
Family History of breast cancer
None 1.0 (Ref) 1.0 (ref) 1.0 (Ref) 1.0 (Ref) 1.0 (Ref)
1 1st degree relative 1.6 (1.4–1.9) 1.5 (1.0–2.1) 1.5 (0.8–2.6) 1.0 (0.6–1.6) 0.9 (0.4–1.9)
2+ 1st degree relat 2.3 (1.3–4.2) -- 2.5 (0.3–18.1) 2.9 (0.7–11.7) --
P-Heterogeneity 0.01
1Mutually adjusted for all variables in the table
Breast Cancer Res Treat. Author manuscript; available in PMC 2013 January 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Tamimi et al. Page 15
Appendix 1
Sources and dilutions of primary antibodies used in this study.
Antibody Clone Manufacturer Dilution
ER 1D5 Dako 1:200
PR PgR 636 Dako 1:50
HER2 A0485 (rabbit polyclonal) Dako 1:400
CK 5/6 D5/16B4 Dako 1:50
EGFR 2-18C9 Dako pre-diluted (pharmDX kit)
ER=estrogen receptor; PR=progesterone receptor; HER2=human epidermal growth factor receptor 2; CK5/6=cytokeratin 5/6; EGFR=epidermal
growth factor receptor
Breast Cancer Res Treat. Author manuscript; available in PMC 2013 January 1.
... The risk factors for breast cancer include amongst others exposure to endogenous and exogenous female sex hormones. Hormonal risk factors are associated with hormone receptor-positive and luminal A-like subtypes [8][9][10]. Less is known about the risk factors for the remaining intrinsic-like subtypes. ...
... We did not find previous reports suggesting inverse associations between body fatness and luminal B-like breast cancer as we did for overweight duration, intensity, and weight gain from overweight. To our knowledge, most studies have reported non-significant results [8,19,52,53], and one study reported an increased risk of the luminal B-like subtype among women with obesity compared with normal-weight women [14]. Variations in the study design, age, measure of exposure, sample size, and subtype definition may explain these discrepancies. ...
... While many studies have addressed weight change in relation to the risk of subtypes of breast cancer [8,16,17,20,[54][55][56][57], to our knowledge, this is the first study to assess associations between BMI trajectories and breast cancer subtypes. Previous studies on life-course fluctuations of body fatness in relation to breast cancer risk used trajectories of perceived body silhouettes [28][29][30][31]. ...
Article
Full-text available
Background Body fatness is a dynamic exposure throughout life. To provide more insight into the association between body mass index (BMI) and postmenopausal breast cancer, we aimed to examine the age at onset, duration, intensity, and trajectories of body fatness in adulthood in relation to risk of breast cancer subtypes. Methods Based on self-reported anthropometry in the prospective Norwegian Women and Cancer Study, we calculated the age at onset, duration, and intensity of overweight and obesity using linear mixed-effects models. BMI trajectories in adulthood were modeled using group-based trajectory modeling. We used Cox proportional hazards models to calculate hazard ratios (HRs) with 95% confidence intervals (CIs) for the associations between BMI exposures and breast cancer subtypes in 148,866 postmenopausal women. Results A total of 7223 incident invasive postmenopausal breast cancer cases occurred during follow-up. Increased overweight duration and age at the onset of overweight or obesity were associated with luminal A-like breast cancer. Significant heterogeneity was observed in the association between age at overweight and overweight duration and the intrinsic-like subtypes ( p heterogeneity 0.03). Compared with women who remained at normal weight throughout adulthood, women with a descending BMI trajectory had a reduced risk of luminal A-like breast cancer (HR 0.54, 95% CI 0.33–0.90), whereas women with ascending BMI trajectories were at increased risk (HR 1.09; 95% CI 1.01–1.17 for “Normal-overweight”; HR 1.20; 95% CI 1.07–1.33 for “Normal-obesity”). Overweight duration and weighted cumulative years of overweight and obesity were inversely associated with luminal B-like breast cancer. Conclusions In this exploratory analysis, decreasing body fatness from obesity in adulthood was inversely associated with overall, hormone receptor-positive and luminal A-like breast cancer in postmenopausal women. This study highlights the potential health benefits of reducing weight in adulthood and the health risks associated with increasing weight throughout adult life. Moreover, our data provide evidence of intrinsic-like tumor heterogeneity with regard to age at onset and duration of overweight.
... Nineteen of the seventy-five studies reported data that could not be meta-analyzed, resulting in fifty-six studies included in meta-analysis. Four cohort studies [16][17][18][19] that reported hazard ratios to evaluate the association of reproductive factors and subtypes were combined with case-control studies in the meta-analysis. ...
... Forty-six studies evaluated the association between age at menarche and BC subtypes (twenty-four case-control studies [9, 22, 28, 36, 37, 39-42, 44, 46, 48, 53, 57, 61, 63, 65, 66, 72, 79, 80, 83, 85, 86], fourteen case-only studies [20, 27, 30, 35, 43, 59, 69, 71, 74-77, 81, 89], three casecontrol/case-case studies [24,52,60], and five cohort studies [17,18,55,70,88]), and were included in the systematic review. Among the cohort and case-control studies, later age at menarche was associated with lower risk of BC in the majority of studies regardless of subtype [9, 17, 18, 22, 24, 28, 36, 37, 39-42, 44, 46, 48, 52, 53, 55, 57, 60, 61, 63, 65, 66, 70, 72, 79, 80, 83, 85, 86, 88], of which fourteen studies [17,18,24,39,40,42,52,57,63,72,79,83,85,86] were luminal A, nine studies [17,24,39,42,52,57,63,85,86] were luminal B, twelve studies [17,22,24,39,42,44,53,63,65,79,83,85] were HER2, and eighteen studies were TNBC [9,18,22,37,39,40,46,52,53,57,60,63,65,66,72,79,85,86]. ...
... Forty-six studies evaluated the association between age at menarche and BC subtypes (twenty-four case-control studies [9, 22, 28, 36, 37, 39-42, 44, 46, 48, 53, 57, 61, 63, 65, 66, 72, 79, 80, 83, 85, 86], fourteen case-only studies [20, 27, 30, 35, 43, 59, 69, 71, 74-77, 81, 89], three casecontrol/case-case studies [24,52,60], and five cohort studies [17,18,55,70,88]), and were included in the systematic review. Among the cohort and case-control studies, later age at menarche was associated with lower risk of BC in the majority of studies regardless of subtype [9, 17, 18, 22, 24, 28, 36, 37, 39-42, 44, 46, 48, 52, 53, 55, 57, 60, 61, 63, 65, 66, 70, 72, 79, 80, 83, 85, 86, 88], of which fourteen studies [17,18,24,39,40,42,52,57,63,72,79,83,85,86] were luminal A, nine studies [17,24,39,42,52,57,63,85,86] were luminal B, twelve studies [17,22,24,39,42,44,53,63,65,79,83,85] were HER2, and eighteen studies were TNBC [9,18,22,37,39,40,46,52,53,57,60,63,65,66,72,79,85,86]. Among the case-only studies [20, 24, 27, 30, 35, 43, 52, 59, 60, 69, 71, 74-77, 81, 89], compared to luminal A, later age of menarche was associated with lower risk of luminal B in seven studies [20,24,30,43,52,69,89], HER2 subtype in six studies [20,30,59,74,76,89], and TNBC in eight studies [27,30,52,60,71,74,76,89]. Sung et.al. ...
Article
Full-text available
Background Associations between reproductive factors and breast cancer (BC) risk vary by molecular subtype (i.e., luminal A, luminal B, HER2, and triple negative/basal-like [TNBC]). In this systematic review and meta-analysis, we summarized the associations between reproductive factors and BC subtypes. Methods Studies from 2000 to 2021 were included if BC subtype was examined in relation to one of 11 reproductive risk factors: age at menarche, age at menopause, age at first birth, menopausal status, parity, breastfeeding, oral contraceptive (OC) use, hormone replacement therapy (HRT), pregnancy, years since last birth and abortion. For each reproductive risk factor, BC subtype, and study design (case–control/cohort or case-case), random-effects models were used to estimate pooled relative risks and 95% confidence intervals. Results A total of 75 studies met the inclusion criteria for systematic review. Among the case–control/cohort studies, later age at menarche and breastfeeding were consistently associated with decreased risk of BC across all subtypes, while later age at menopause, later age of first childbirth, and nulliparity/low parity were associated with increased risk of luminal A, luminal B, and HER2 subtypes. In the case-only analysis, compared to luminal A, postmenopausal status increased the risk of HER2 and TNBC. Associations were less consistent across subtypes for OC and HRT use. Conclusion Identifying common risk factors across BC subtypes can enhance the tailoring of prevention strategies, and risk stratification models can benefit from subtype specificity. Adding breastfeeding status to current BC risk prediction models can enhance predictive ability, given the consistency of the associations across subtypes.
... Breast cancer is a heterogeneous tumor that can be classified into four major molecular subtypes: luminal-like (luminal A and luminal B), HER2-positive, and basal-like. 1 The prognosis varies significantly among these molecular subtypes, prompting extensive research into the molecular diagnosis of breast cancer. 2 HER2 overexpression, which arises from the amplification of the ERBB2/neu proto-oncogene located on the centromere of chromosome 17 (CEN17), is found in approximately 15%-20% of breast cancers. ...
Article
Full-text available
Background We aimed to predict human epidermal growth factor receptor 2 (HER2) 2+ status in patients with breast cancer by constructing and validating machine learning models utilizing ultrasound (US) radiomics and clinical features. Methods We analyzed 203 breast cancer cases immunohistochemically determined as HER2 2+ and used fluorescence in situ hybridization (FISH) as the confirmation method. From each case, the study analyzed 840 extracted radiomics features and 11 clinicopathologic features. Cases were randomly split into training ( n = 141) and validation sets ( n = 62) at a 7:3 ratio. Univariate logistic regression analysis was first performed on the 11 clinicopathologic characteristics. The least absolute shrinkage and selection operator (LASSO) and decision tree (DT) techniques were employed for post‐feature selection. Finally, 19 radiomics features were utilized in logistic regression (LR) and Naive Bayesian (NB) classifiers. Model performance was gauged using the area under the receiver operating characteristic curve (AUC), accuracy, sensitivity, and specificity. Results Our models exhibited notable diagnostic efficacy in differentiating HER2‐positive from negative breast cancer cases. In the validation sets, the LR model outperformed the NB model with an AUC of 0.860 and accuracy of 83.8% compared to NB's AUC of 0.684 and accuracy of 79.0%. The LR model demonstrated higher sensitivity (92.3% vs. 46.2%) while the NB model had a better specificity (91.8% vs. 63.3%) in the validation set. Conclusions Machine learning models grounded on radiomics efficiently predicted IHC HER2 2+ status in breast cancer patients, suggesting potential enhancements in clinical decision‐making for treatment and management.
... YOBC is also etiologically distinct from postmenopausal breast cancer with, for example, obesity associated with increased risk in post-and decreased risk in premenopausal women [8][9][10]. Additionally, breast cancer tumor subtypes are associated with distinct clinical treatments and outcomes, and burgeoning evidence suggests different etiologies by subtype [11][12][13][14][15], including for YOBC [16]. ...
Article
Full-text available
Purpose The role of alcohol in young-onset breast cancer (YOBC) is unclear. We examined associations between lifetime alcohol consumption and YOBC in the Young Women’s Health History Study, a population-based case–control study of breast cancer among Non-Hispanic Black and White women < 50 years of age. Methods Breast cancer cases (n = 1,812) were diagnosed in the Metropolitan Detroit and Los Angeles County SEER registry areas, 2010–2015. Controls (n = 1,381) were identified through area-based sampling and were frequency-matched to cases by age, site, and race. Alcohol consumption and covariates were collected from in-person interviews. Weighted multivariable logistic regression was conducted to calculate adjusted odds ratios (aOR) and 95% confidence intervals (CI) for associations between alcohol consumption and YOBC overall and by subtype (Luminal A, Luminal B, HER2, or triple negative). Results Lifetime alcohol consumption was not associated with YOBC overall or with subtypes (all ptrend ≥ 0.13). Similarly, alcohol consumption in adolescence, young and middle adulthood was not associated with YOBC (all ptrend ≥ 0.09). An inverse association with triple-negative YOBC, however, was observed for younger age at alcohol use initiation (< 18 years vs. no consumption), aOR (95% CI) = 0.62 (0.42, 0.93). No evidence of statistical interaction by race or household poverty was observed. Conclusions Our findings suggest alcohol consumption has a different association with YOBC than postmenopausal breast cancer—lifetime consumption was not linked to increased risk and younger age at alcohol use initiation was associated with a decreased risk of triple-negative YOBC. Future studies on alcohol consumption in YOBC subtypes are warranted.
... Female breast cancer (BC) is the most common cancer worldwide regardless of sex [1] and in Scotland accounts for 28.8% of cancer cases [2]. BC is comprised of multiple molecular subtypes, each of which have their own prognosis, treatment, and aetiology [3][4][5][6][7][8][9][10][11][12][13]. Socioeconomic disparities in BC incidence and survival have been described in Scotland [6,14] and multiple countries around the world [15][16][17][18]. ...
Article
Full-text available
Background Women from socioeconomically deprived areas have lower breast cancer (BC) incidence rates for screen-detected oestrogen receptor (ER) + tumours and higher mortality for select tumour subtypes. We aimed to determine if ipsilateral breast cancer recurrence (IBR) differs by Scottish Index of Multiple Deprivation (SIMD) quintile and tumour subtype in Scotland. Methods Patient data for primary invasive BC diagnosed in 2007–2008 in Scotland was analysed. Manual case-note review for 3495 patients from 10 years post-diagnosis was used. To determine the probability of IBR while accounting for the competing risk of death from any cause, cumulative incidence functions stratified by ER subtype and surgery were plotted. Multivariable Cox Proportional Hazards models were used to estimate the association of SIMD accounting for other predictors of IBR. Results Among 2819 ER + tumours, 423 patients had a recurrence and 438 died. SIMD was related to death (p = 0.018) with the most deprived more likely to have died in the 10-year period (17.7% vs. 12.9%). We found no significant differences by SIMD in prognostic tumour characteristics (grade, TNM stage, treatment, screen-detection) or risk of IBR. Among 676 patients diagnosed with ER- tumours, 105 died and 185 had a recurrence. We found no significant differences in prognostic tumour characteristics by SIMD except screen detection with the most deprived more likely than the least to have their tumours detected from screening (46.9% vs. 28%, p = 0.03). Among patients with ER- tumours, 50% had mastectomy and the most deprived had increased 5-year IBR risk compared to the least deprived (HR 3.03 [1.41–6.53]). Conclusions IBR is not a major contributor to mortality differences by SIMD for the majority of BC patients in our study. The lack of inequities in IBR are likely due to standardised treatment protocols and access to healthcare. The association with socioeconomic deprivation and recurrence for ER- tumours requires further study.
... However, consistent evidence indicates that breastfeeding is associated with a lower risk of HER2+ breast cancer [28,37]. The existing evidence on the relationship between breastfeeding and luminal B breast cancer risk is primarily based on larger prior studies, which do not demonstrate a significant association [37][38][39][40]. However, a study (N=476) reported a reduced risk of luminal B breast cancer among women who breastfed [OR=1.89 ...
Article
Breastfeeding has been extensively studied in relation to breast cancer risk. The results of the reviewed studies consistently show a decreased risk of breast cancer associated with breastfeeding, especially for 12 months or longer. This protective effect is attributed to hormonal, immunological, and physiological changes during lactation. Breastfeeding also appears to have a greater impact on reducing breast cancer risk in premenopausal women and specific breast cancer subtypes. Encouraging breastfeeding has dual benefits: benefiting infants and reducing breast cancer risk long-term. Healthcare professionals should provide evidence-based guidance on breastfeeding initiation, duration, and exclusivity, while public health policies should support breastfeeding by creating enabling environments. This review examines the existing literature and analyzes the correlation between breastfeeding and breast cancer risk.
... However, consistent evidence indicates that breastfeeding is associated with a lower risk of HER2+ breast cancer [28,37]. The existing evidence on the relationship between breastfeeding and luminal B breast cancer risk is primarily based on larger prior studies, which do not demonstrate a significant association [37][38][39][40]. However, a study (N=476) reported a reduced risk of luminal B breast cancer among women who breastfed [OR=1.89 ...
Article
Full-text available
Breastfeeding has been extensively studied in relation to breast cancer risk. The results of the reviewed studies consistently show a decreased risk of breast cancer associated with breastfeeding, especially for 12 months or longer. This protective effect is attributed to hormonal, immunological, and physiological changes during lactation. Breastfeeding also appears to have a greater impact on reducing breast cancer risk in premenopausal women and specific breast cancer subtypes. Encouraging breastfeeding has dual benefits: benefiting infants and reducing breast cancer risk long-term. Healthcare professionals should provide evidence-based guidance on breastfeeding initiation, duration, and exclusivity, while public health policies should support breastfeeding by creating enabling environments. This review examines the existing literature and analyzes the correlation between breastfeeding and breast cancer risk.
Article
Background Associations between reproductive factors and risk of breast cancer differ by subtype defined by joint estrogen receptor (ER), progesterone receptor (PR), and HER2 expression status. Racial and ethnic differences in the incidence of breast cancer subtypes suggest etiologic heterogeneity, yet data are limited because most studies have included non-Hispanic White women only. Methods We analyzed harmonized data for 2,794 breast cancer cases and 4,579 controls, of whom 90% self-identified as African American, Asian American or Hispanic. Questionnaire data were pooled from three population-based studies conducted in California and data on tumor characteristics were obtained from the California Cancer Registry. The study sample included 1,530 luminal A (ER-positive and/or PR-positive, HER2-negative), 442 luminal B (ER-positive and/or PR-positive, HER2-positive), 578 triple-negative (TN; ER-negative, PR-negative, HER2-negative), and 244 HER2-enriched (ER-negative, PR-negative, HER2-positive) cases. We used multivariable unconditional logistic regression models to estimate subtype-specific ORs and 95% confidence intervals associated with parity, breast-feeding, and other reproductive characteristics by menopausal status and race and ethnicity. Results Subtype-specific associations with reproductive factors revealed some notable differences by menopausal status and race and ethnicity. Specifically, higher parity without breast-feeding was associated with higher risk of luminal A and TN subtypes among premenopausal African American women. In contrast, among Asian American and Hispanic women, regardless of menopausal status, higher parity with a breast-feeding history was associated with lower risk of luminal A subtype. Among premenopausal women only, luminal A subtype was associated with older age at first full-term pregnancy (FTP), longer interval between menarche and first FTP, and shorter interval since last FTP, with similar OR estimates across the three racial and ethnic groups. Conclusions Subtype-specific associations with reproductive factors overall and by menopausal status, and race and ethnicity, showed some differences, underscoring that understanding etiologic heterogeneity in racially and ethnically diverse study samples is essential. Breast-feeding is likely the only reproductive factor that is potentially modifiable. Targeted efforts to promote and facilitate breast-feeding could help mitigate the adverse effects of higher parity among premenopausal African American women.
Article
Full-text available
To compare diffusion-kurtosis imaging (DKI) and diffusion-weighted imaging (DWI) parameters of single-shot echo-planar imaging (ss-EPI) and readout-segmented echo-planar imaging (rs-EPI) in the differentiation of luminal vs. non-luminal breast cancer using histogram analysis. One hundred and sixty women with 111 luminal and 49 non-luminal breast lesions were enrolled in this study. All patients underwent ss-EPI and rs-EPI sequences on a 3.0T scanner. Histogram metrics were derived from mean kurtosis (MK), mean diffusion (MD) and the apparent diffusion coefficient (ADC) maps of two DWI sequences respectively. Student’s t test or Mann–Whitney U test was performed for differentiating luminal subtype from non-luminal subtype. The ROC curves were plotted for evaluating the diagnostic performances of significant histogram metrics in differentiating luminal from non-luminal BC. The histogram metrics MKmean, MK50th, MK75th of luminal BC were significantly higher than those of non-luminal BC for both two DWI sequences (all P<0.05). Histogram metrics from rs-EPI sequence had better diagnostic performance in differentiating luminal from non-Luminal breast cancer compared to those from ss-EPI sequence. MK75th derived from rs-EPI sequence was the most valuable single metric (AUC, 0.891; sensitivity, 78.4%; specificity, 87.8%) for differentiating luminal from non-luminal BC among all the histogram metrics. Histogram metrics of MK derived from rs-EPI yielded better diagnostic performance for distinguishing luminal from non-luminal BC than that from ss-EPI. MK75th was the most valuable metric among all the histogram metrics.
Article
Full-text available
The article provides an overview of the built-in tools for proxying traffic in mobile testing using Charles. The article also covers situations and problems that inevitably arise in testing engineers' daily routine, which can be solved by using the appropriate tools. This article is especially useful for professionals – practicing test engineers involved in testing mobile applications on iOS and Android
Article
Full-text available
cDNA microarrays and a clustering algorithm were used to identify patterns of gene expression in human mammary epithelial cells growing in culture and in primary human breast tumors. Clusters of coexpressed genes identified through manipulations of mammary epithelial cells in vitro also showed consistent patterns of variation in expression among breast tumor samples. By using immunohistochemistry with antibodies against proteins encoded by a particular gene in a cluster, the identity of the cell type within the tumor specimen that contributed the observed gene expression pattern could be determined. Clusters of genes with coherent expression patterns in cultured cells and in the breast tumors samples could be related to specific features of biological variation among the samples. Two such clusters were found to have patterns that correlated with variation in cell proliferation rates and with activation of the IFN-regulated signal transduction pathway, respectively. Clusters of genes expressed by stromal cells and lymphocytes in the breast tumors also were identified in this analysis. These results support the feasibility and usefulness of this systematic approach to studying variation in gene expression patterns in human cancers as a means to dissect and classify solid tumors.
Article
Full-text available
Differences in incidence, prognosis, and treatment response suggest gene expression patterns may discern breast cancer subtypes with unique risk factor profiles; however, previous results were based predominantly on older women. In this study, we examined similar relationships in women ≤ 56 years, classified by immunohistochemical staining for estrogen receptor, progesterone receptor, and human epidermal growth factor receptor-2 for 890 breast cancer cases and 3,432 frequency-matched population-based controls. Odds ratios (OR) and 95% confidence intervals (CI) for tumor subtypes were calculated using multivariate polytomous regression models. A total of 455 (51.1%) tumors were considered luminal A, 72 (8.1%) luminal B, 117 (13.1%) non-luminal HER-2/neu+, and 246 (27.6%) triple negative. Triple negative tumors were associated with breast feeding duration (per 6 months: OR = 0.76, 95% CI 0.64-0.90). Among premenopausal women, increasing body size was more strongly associated with luminal B (OR = 1.73, 95% CI 1.07-2.77) and triple negative tumors (OR = 1.67, 95% CI 1.22-2.28). A history of benign breast disease was associated only with increased risk of luminal A tumors (OR = 1.89, 95% CI 1.43-2.50). A family history of breast cancer was a risk factor for luminal A tumors (OR = 1.93, 95% CI 1.38-2.70) regardless of age, and triple negative tumors with higher risks for women <45 (OR = 5.02, 95% CI 2.82-8.92; P for age interaction = 0.005). We found that little-to-no breastfeeding and high BMI were associated with increased risk of triple negative breast cancer. That some risk factors differ by molecular subtypes suggests etiologic heterogeneity in breast carcinogenesis among young women.
Article
Full-text available
The aim of this study was to describe breast tumor subtypes by common breast cancer risk factors and to determine correlates of subtypes using baseline data from two pooled prospective breast cancer studies within a large health maintenance organization. Tumor data on 2544 invasive breast cancer cases subtyped by estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 (Her2) status were obtained (1868 luminal A tumors, 294 luminal B tumors, 288 triple-negative tumors and 94 Her2-overexpressing tumors). Demographic, reproductive and lifestyle information was collected either in person or by mailed questionnaires. Case-only odds ratios (ORs) and 95% confidence intervals (CIs) were estimated using logistic regression, adjusting for age at diagnosis, race/ethnicity, and study origin. Compared with luminal A cases, luminal B cases were more likely to be younger at diagnosis (P = 0.0001) and were less likely to consume alcohol (OR = 0.74, 95% CI = 0.56 to 0.98), use hormone replacement therapy (HRT) (OR = 0.66, 95% CI = 0.46 to 0.94), and oral contraceptives (OR = 0.73, 95% CI = 0.55 to 0.96). Compared with luminal A cases, triple-negative cases tended to be younger at diagnosis (P < or = 0.0001) and African American (OR = 3.14, 95% CI = 2.12 to 4.16), were more likely to have not breastfed if they had parity greater than or equal to three (OR = 1.68, 95% CI = 1.00 to 2.81), and were more likely to be overweight (OR = 1.82, 95% CI = 1.03 to 3.24) or obese (OR = 1.97, 95% CI = 1.03 to 3.77) if premenopausal. Her2-overexpressing cases were more likely to be younger at diagnosis (P = 0.03) and Hispanic (OR = 2.19, 95% CI = 1.16 to 4.13) or Asian (OR = 2.02, 95% CI = 1.05 to 3.88), and less likely to use HRT (OR = 0.45, 95% CI = 0.26 to 0.79). These observations suggest that investigators should consider tumor heterogeneity in associations with traditional breast cancer risk factors. Important modifiable lifestyle factors that may be related to the development of a specific tumor subtype, but not all subtypes, include obesity, breastfeeding, and alcohol consumption. Future work that will further categorize triple-negative cases into basal and non-basal tumors may help to elucidate these associations further.
Article
Full-text available
Background Gene expression profiling of breast cancer has identified two biologically distinct estrogen receptor (ER)-positive subtypes of breast cancer: luminal A and luminal B. Luminal B tumors have higher proliferation and poorer prognosis than luminal A tumors. In this study, we developed a clinically practical immunohistochemistry assay to distinguish luminal B from luminal A tumors and investigated its ability to separate tumors according to breast cancer recurrence-free and disease-specific survival.
Article
Immunohistochemical analysis is used routinely to determine the estrogen receptor (ER) status of breast cancers in paraffin sections. However, lack of standardization has raised concerns that weakly ER+ tumors often are classified erroneously as ER-. To determine the frequency of weakly ER+ tumors, we reviewed ER immunostains of 825 breast cancers. For each case, we estimated the proportion of ER+ tumor cells and also determined an Allred score (which results in scores of 0 or 2 through 8, based on staining intensity and proportion of positive cells). In 817 cases (99.0%), tumor cells showed complete absence of staining or staining in 70% or more of the cells. Similarly, 818 cases (99.2%) exhibited Allred scores of 0 or of 7 or 8. Thus, with the immunohistochemical method used in our laboratory, ER staining is essentially bimodal. The overwhelming majority of breast cancers are either completely ER- or unambiguously ER+, and cases with weak ER immunostaining are rare.
Article
Molecular profiling studies have identified subtypes of breast cancer that can be approximately classified by estrogen receptor (ER), progesterone receptor (PR), and HER-2/neu (HER-2) expression. These molecular subtypes are prognostically significant, but to the authors' knowledge, differences in their etiologic profiles have not been established. Reproductive factors may plausibly be differentially correlated with the risk of different breast cancer subtypes because these factors are presumed to impact exposure to endogenous sex hormones. The authors pooled 2 population-based, case-control studies of breast cancer in women ages 55 to 79 years for an analysis including 1476 controls and 1023 cases of luminal breast cancer, 39 cases of HER-2-overexpressing breast cancer, and 78 cases of triple-negative breast cancer. Polytomous logistic regression was used to compare each case group with controls. Associations varied by molecular subtype. Early age at menarche was only found to be associated with risk of HER-2-overexpressing disease (odds ratio [OR] of 2.7; 95% confidence interval [95% CI], 1.4-5.5), whereas breastfeeding for > or =6 months was only found to be protective for luminal and triple-negative disease (OR of 0.8 [95% CI, 0.6-1.0] and OR of 0.5 [95% CI, 0.3-0.9], respectively). Both late age at menopause and the use of estrogen plus progestin hormone therapy were only found to be associated with risk of luminal disease (OR of 1.6 [95% CI, 1.1-2.2] and OR of 1.7 [95% CI, 1.3-2.1], respectively). No differences in risks associated with parity or age at first live birth were observed by subtype. Certain reproductive factors may have a greater impact on the risk of certain molecular subtypes of disease compared with others. Future studies that further define the etiology of breast cancer subtypes will add to the biologic understanding of this disease.