Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Jun 14.
Published in final edited form as: Nat Rev Cancer. 2015 Feb 12;15(4):248–254. doi: 10.1038/nrc3896

Triple-negative breast cancer in African-American women: disparities versus biology

Eric C Dietze 1, Christopher Sistrunk 2, Gustavo Miranda-Carboni 3, Ruth O’Regan 4, Victoria L Seewaldt 5
PMCID: PMC5470637  NIHMSID: NIHMS861862  PMID: 25673085

Abstract

Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype that disproportionately affects BRCA1 mutation carriers and young women of African origin. There is evidence that African-American women with TNBC have worse clinical outcomes than women of European descent. However, it is unclear whether survival differences persist after adjusting for disparities in access to health-care treatment, co-morbid disease and income. It remains controversial whether TNBC in African-American women is a molecularly distinct disease or whether African-American women have a higher incidence of aggressive biology driven by disparities: there is evidence in support of both. Understanding the relative contributions of biology and disparities is essential for improving the poor survival rate of African-American women with TNBC.


Triple-negative breast cancer (TNBC) represents a diverse group of cancers that are characterized by lack of expression of the oestrogen receptor (ER) and progesterone receptor (PR) and absence of ERBB2 (also known as HER2) amplification1. There is a high prevalence of TNBC in women of African descent and women who carry a mutated copy of the BRCA1 gene2. The basal subset of TNBC is characterized by the expression of basal-type cytokeratin 5 and cytokeratin 6 and high expression of epidermal growth factor receptor (EGFR), and frequently exhibits aggressive clinical behaviour2. Women with TNBC have a high frequency of metastasis to the lung, liver and brain, and survival is generally poor3.

There is evidence that African-American women with TNBC have worse clinical outcomes compared with women of European descent who have the disease. However, published reports of survival outcomes for African-American women with TNBC, relative to European-American women, are conflicting410. Furthermore, it is unclear whether survival differences persist after adjusting for disparities in access to health-care treatment, co-morbid disease and income. African-American women experience lack of access to breast cancer screening, lack of access to oncology care and delays in treatment11. There is strong evidence that disparities in health-care provision, co-morbid disease and income affect the stage of presentation and survival of African-American women with TNBC11,12. Emerging data also indicate that disparities may drive aggressive biology in African-American women with TNBC. It is essential that we understand the potential molecular mechanism by which biology and disparities may intersect to drive the aggressive TNBC subtypes in African-American women: such knowledge may lead to the development of models that specifically capture the risk of women of African descent for TNBC, to increased access to effective early detection and to the implementation of health policies to eliminate disparities.

Demographics and prognosis

TNBC is a distinct subtype

In 2001, Perou and colleagues used cDNA microarray profiling to identify breast cancer subtypes2,13,14. These subtypes were subsequently integrated with traditional hormone-receptor-based clinical subtyping to define four integrated subtypes: basal-like (ER, PR, no ERBB2 amplification, cytokeratin 5+, cytokeratin 6+ and EGFR+); ERBB2+ (ERBB2 amplified) and ER and PR (ERBB2+, ER and PR); luminal A (ER+ and/or PR+, no ERBB2 amplification); and luminal B (ER+ and/or PR+, and ERBB2+). Over 75% of TNBCs express basal markers and cluster with the basal-like subtype by gene expression profiling2,13,14.

Demographics

Many studies show that premenopausal African-American and African women have a high prevalence of TNBC relative to women of European descent (TABLE 1). The Carolina Breast Cancer Study showed that the highest prevalence of the basal-like subtype of TNBC (39%; 38/97 invasive cancers) occurred in premenopausal African-American women. This was substantially higher than the prevalence of TNBC observed in post-menopausal African-American women (14%; 14/99 invasive cancers) or American women of European descent (16%; 48/300 invasive cancers) (p < 0.001 for both comparisons)3. The high frequency of TNBC in African-American women has also been observed in population-based studies in Philadelphia15, Boston16, Georgia4 and Michigan17 (TABLE 1).

Table 1.

Incidence of TNBC in population-based studies of black women of African and European origins

Geographical location TNBC incidence Mean age at diagnosis Study Refs
Ghana 82% (37/45) 48.0 +/− 6.4 years Population-based; consecutive; Komfo Anokye Hospital, Kumasi, Ghana; Jan 2007–Dec 2008 17
Soweto, South Africa 20.4 (209/1,092) 55.3 +/− 14.3 years Population-based; consecutive; Soweto; 2006–2012 21
North Carolina, USA 39% AA (52/196); 16% EA (48/300) 46 years (basal subtype); 52 years (luminal A subtype) Population-based; case–control; North Carolina Central Cancer registry; Mar 1993–Dec 1996 3
Philadelphia, Pennsylvania, USA 20.8% AA (59/283); 10.4% EA (203/2,230) Not stated Population-based; SEER registry; 1990–2000 15
Boston, Massachusetts, USA 30% AA (52/177); 19% EA (13/148) No difference in age Population-based; consecutive; Boston University Hospital, Massachusetts; Mar 1998–Nov 2006 16
Georgia, USA 46.4% AA (56/116); 21.8% EA (79/360) AA women were a younger age at diagnosis Population-based; consecutive; 1990–1992 4
Michigan, USA 26.4% AA (107/405); 16.0% EA (122/763) 60.8 +/− 13.7 years (AA); 62.4 +/− 13.7 years (EA) Population-based; consecutive; Henry Ford Hospital; Jan 2001–Dec 2007 17

AA, African American; EA, European American; SEER, Surveillance, Epidemiology, and End Results; TNBC, triple-negative breast cancer.

Several studies show that TNBC occurs in sub-Saharan and West African women at a higher frequency and a younger age than in African-American women1719. Although breast cancer incidence rates among sub-Saharan African women are low (10–40 per 100,000), these women experience higher mortality rates (5–20 per 100,000) and are more often diagnosed at a younger age than either African-American or European-American women (peak incidence: 35–45 years). In a population-based study that evaluated African-American and European-American women with invasive breast cancer at Henry Ford Hospital in Detroit, Michigan, USA, and Ghanaian women with invasive breast cancer at Komfo Anokye Hospital in Kumasi, Ghana, 83% (n = 507) of Ghanaian women had TNBC, whereas 41.9% (n = 507) of African-American women and 15.4% of European-American women had TNBC. Most of the Ghanaian women (83%) presented with advanced, high-grade tumours17. One of the strengths of this study was that all biopsy samples underwent central pathology review and analysis of expression of ERBB2, ER and PR at the University of Michigan in Ann Arbor, USA. A second study evaluated a collection of 507 invasive breast cancers from six sites in Nigeria and Senegal; most of the tumours were basal-like or unclassified subtypes (27% and 28%, respectively), 29% were luminal A or B, 15% were the ERBB2+ and ER subtype, and 1% were unclassified20. However, not all studies of African women show an increased incidence of TNBC or a younger age of presentation. In a population-based study of 1,218 consecutive women (91% black) diagnosed with invasive breast cancer from 2006 to 2012 at a public hospital in Soweto, South Africa, the age-specific incidence of TNBC in black South African women was similar to that in African-American women21.

Prognosis

African-American women have higher overall breast cancer mortality compared with women of European descent3,8,22. Contributing factors include disparities in income, barriers to screening, differences in treatment, higher stage of disease at diagnosis and increased incidence of TNBC2327. Although Western nations have a higher incidence of breast cancer in comparison to developing nations, mortality is highest in developing nations. The factors influencing a woman’s survival are affected by cultural and economic barriers to care and vary greatly throughout the world27.

Newman and her colleagues28 conducted a meta-analysis of over 13,000 African-American women with breast cancer compared with 75,000 women of European descent. In this study, African-American identity was associated with a statistically significant, nearly 30% higher mortality rate (mortality hazard, 1.27; 95% confidence interval, 1.18–1.38). Although African-American identity is associated with poor prognosis, it is unclear whether poor survival is due to differences at the molecular level, beyond an increased frequency of TNBC3.

Studies of the impact of race on survival in women with TNBC have yielded mixed results. Dean-Colomb et al.10 compared transcriptional profiles of tumours of 98 women with TNBC who received cytotoxic chemotherapy and observed no differences in pathologic complete response (pCR) rates by ethnicity or race, and no differences in gene expression between African-American and Caucasian women. Dawood et al.9 investigated the impact of race on pCR in 471 women with TNBC who were receiving anthracycline-based chemotherapy and showed that race did not significantly affect outcome. Shen et al.8 reported shorter survival rates among African-American women compared with women of European descent (n = 6,054). Woodward et al.7 explored the effect of race on survival outcome in patients treated at the University of Texas MD Anderson Cancer Center in Houston, USA, and reported that, despite a uniform distribution of treatment, African-American race was independently associated with poorer survival. Albain’s group6 found that African-American women with early-stage premenopausal breast cancer participating in Phase III Southwest Oncology Group (SWOG) trials had a significantly worse prognosis than age- and prognosis-matched European-American women. Bauer et al.5 and Lund et al.4 also reported a worse survival for African-American women with TNBC after controlling for socioeconomic factors, treatment delay and breast cancer receptor expression (FIG. 1). Taken together, these studies provide evidence that even after controlling for treatment disparities, biological differences may contribute to the poor survival of women with TNBC. However, the data are inconsistent, and molecular studies are needed to better define potential biological differences.

Figure 1. TNBC in African-American compared with European-American women.

Figure 1

a| Age-related incidence of triple-negative breast cancer (TNBC) by race, shown as a percentage of overall breast cancer incidence. b | Survival of African-American women based on breast cancer subtype (oestrogen receptor (ER)+, progesterone receptor (PR)+, ERBB2 versus ER, PR, ERBB2). c | TNBC survival rate based on race. d | Adjusted odds ratios (ORs) for race (African Americans compared with European Americans) in the Atlanta study4. Data for parts ad are derived from Lund et al.4 with kind permission from Springer Science and Business Media. CI, confidence interval.

Genetic risk

Frequency of BRCA1 mutations in African-American women with TNBC is low relative to women of European descent

BRCA1 is a tumour suppressor gene and has a key role in homology-directed repair of DNA double-strand breaks2931. Most breast cancers (69%) in women with BRCA1 mutations are TNBCs32. Although the incidence of TNBC is high in African-American women, several studies show that the incidence of germline BRCA1 mutations is low relative to the incidence in women of European descent. In a study of 155 high-risk families evaluated at the University of Chicago in Illinois, USA, African Americans had a lower rate of deleterious germline BRCA1 mutations compared with non-Hispanic, non-Jewish Caucasians (27.9% versus 46.2%, respectively) but a higher rate of sequence variations (44.2% versus 11.5%, respectively; p < 0.001)33,34. Deleterious mutations in BRCA1 were highest for Ashkenazi-Jewish women (69.0%)33. Similarly, whereas 50% of Caucasian non-Ashkenazi-Jewish women with TNBC had germline BRCA1 mutations, fewer than 20% of African-American women with TNBC had germline BRCA1 mutations35. This suggests that another genetic mechanism (or mechanisms) beyond germline mutation of BRCA1 may promote TNBC in African-American women.

A diverse spectrum of BRCA1 mutations and sequence variations unique to African women has been reported3642. A complete review of the founder mutations identified in African-American women is beyond the scope of this article (see Oluwagbemiga et al.36 for a full review). However, although many sequence variations of BRCA1 have been found in women of African descent, only a small number of the mutations result in known structural defects that block BRCA1 function (for example, protein-truncating mutations)3742, and these known structural mutations occur in women of African origin at a lower rate than is observed in women of European descent37.

BROCA profiling tests for 18–40 genes (in addition to BRCA) that repair DNA. These new ‘panel tests’ use a combination of targeted genomic capture and next-generation sequencing and are becoming widely available. It is anticipated that such panel tests will provide important insights into the genetic mutations that contribute to TNBC risk. BROCA profiling of 249 African-American women with breast cancer found that approximately 20% carried an inherited abnormality in at least 1 of 18 genes associated with breast cancer susceptibility; 56 of 249 women (22%) had at least one clinically relevant mutation43. Most women carried a single mutation, and most mutations (79%) were in BRCA1 and BRCA2. However, 21% of the mutations were in other cancer-associated genes, including ataxia telangiectasia mutated (ATM), checkpoint kinase 2 (CHEK2; also known as CHK2), partner and localizer of BRCA2 (PALB2) and PTEN. Mutations were most common in patients with a second primary cancer in the breast (49%), TNBC (30%) and/or a family history of either breast or ovarian cancer in a close relative (30%), and/or in patients diagnosed before the age of 45 years (27%)43.

A recent study in women of European descent identified that loss-of-function mutations in PALB2 are an important cause of hereditary breast cancer44. Another recent study evaluated the contribution of germline PALB2 mutations in 279 African-American women with breast cancer; novel monoallelic truncating mutations were identified in three patients (1.08%) together with 50 sequence variants. This study showed that rare PALB2 mutations accounted for a small but substantial proportion of breast cancer in women of African descent45.

Genome-wide studies

Most genome-wide association studies (GWASs) have been carried out in European-ancestry populations; no risk variants for TNBC have been identified solely in women of African ancestry. As the Black Women’s Health Study matures (see Further information), it is anticipated that GWASs focusing on women of African ancestry will provide important insights into the genetic basis of TNBC in African-American women. GWASs of African-American women identified a common risk variant at the telomerase reverse transcriptase (TERT)CLPTM1-like (CLPTM1L) locus on chromosome 5p15 (odds ratio (OR) = 1.25; p = 1.1 × 10−9) that was present at a greater frequency in African-American women than in women of European ancestry and was significantly associated with TNBC in women less than 50 years old (OR = 1.48; p = 1.9 × 10−9)46. A genetic variant in the LOC643714 gene has also been identified that predicts a 23% increased risk for breast cancer in African-American women but not in women of European ancestry (OR = 1.23; 95% confidence interval, 1.05–1.44)47. A nested case–control study of breast cancer in the Black Women’s Health Study (1,199 cases/1,948 controls) identified two single-nucleotide polymorphisms — rs10069690 in 5p13.33 (TERT gene) and rs8170 in 19p13.11 — that were significantly associated with TNBC in African-American women, with higher ORs relative to controls48.

Biology of TNBC

Unique biology versus higher frequency of poor prognosis subtypes

As outlined above, disparities in treatment, co-morbid disease, income and access to health care all affect survival. It is unclear whether women of African descent have worse survival owing to an increased frequency of TNBC subtypes that have poor prognosis or whether TNBC in women of African descent has a uniquely aggressive biology. Studies comparing biological differences between TNBC in African-American and European-American women have yielded conflicting results. Some studies have been complicated by the combined analysis of multiple breast cancers49,50. A key emerging area of study is the potential interaction between disparities and signalling pathways that are known to promote aggressive biology and genomic instability (FIGS 2,3).

Figure 2. Proposed model of how disparities might drive signalling pathways associated with aggressive biology in TNBC.

Figure 2

Disparities in income, lack of access to fresh vegetables and healthy food, unsafe neighbourhoods, and lack of exercise can promote co-morbid diseases such as obesity and diabetes, which in turn may drive phosphoprotein signalling pathways associated with aggressive biology in triple-negative breast cancer (TNBC). Obesity and diabetes result in increased levels of circulating insulin and tissue cytokines, such as interleukin-6 (IL-6), tumour necrosis factor (TNF), leptin, chemokine (C-C motif) ligand 2 (CCL2) and transforming growth factor-β (TGFβ), which activate signalling networks that are known to promote epithelial cell proliferation and genomic instability, including PI3K–AKT, signal transducer and activator of transcription 3 (STAT3)–nuclear factor-κB (NF-κB), WNT–microRNA 34 (miR34)–p53, and Aurora A–polo-like kinase (PLK)52,5759,62. Obesity and accompanying tissue inflammation increases tissue factors, such as hypoxia-inducible factor 1α (HIF1α), that promote angiogenesis and contribute to aggressive biology. Recent studies provide evidence that chronic tissue inflammation and cyclooxygenase 2 (COX2) activation promote changes in collagen structure that result in the activation of focal adhesion kinase (FAK) and AKT network signalling79,81.

Figure 3. Proposed model of genetic and epigenetic mechanisms that link unique DNA sequences and disparities with genomic instability, loss of BRCA1 function and aggressive TNBC biology in women of African descent.

Figure 3

Over 60% of Ashkenazi Jewish women with triple-negative breast cancer (TNBC) have a mutation in BRCA1. By contrast, less than 20–25% of African-American women with TNBC have a germline DNA mutation. Although many sequence variations of BRCA1 have been found in women of African descent, only a small number of the mutations are protein truncating3742, and these protein-truncating mutations occur in women of African origin at a lower rate than is observed in women of European descent37. On-going studies are testing for germline mutations in other genes that regulate BRCA1-associated DNA repair (for example, partner and localizer of BRCA2 (PALB2)) and/or signalling pathways that promote epigenetic loss of BRCA1 function. As the Black Women’s Health Study matures, it is anticipated that genome-wide association studies focusing on women of African ancestry will provide important insights into the genetic basis of TNBC in African-American women. Active areas of research aim to link disparities in access, environmental exposures, neighbourhood safety and obesity with epigenetic mechanisms that promote genomic instability. These emerging areas of research have important potential to identify important epigenetic mechanisms that link disparities with genomic instability. For example, emerging data indicate that low vitamin D levels may affect DNA repair82. African-American women have low vitamin D levels owing to increased skin pigmentation83,84; lack of safe neighbourhoods further contributes to low vitamin D levels85. Blue boxes: strong mechanistic evidence exists in TNBC, but specifically linking this evidence with TNBC in women of African descent is a work in progress. Tan boxes: areas of on-going investigation for which published data are currently weak or lacking. EZH2, enhancer of zeste homologue 2; IL-6, interleukin-6; miRNA, microRNA; SNP, single-nucleotide polymorphism; STAT3, signal transducer and activator of transcription 3.

The Cancer Genome Atlas

The Cancer Genome Atlas recently analysed primary breast cancers from 627 women, 53 of whom (8.5%) were African American51. Relative to breast cancers in women of European descent, breast cancers from African-American women showed increased expression of members of the p53, BRCA1, Aurora A, Aurora B and polo-like kinase signalling networks52.

Comparisons of TNBC arising in African-American versus European-American women have yielded varied results. The gene expression profiles of the disease have been shown to be highly similar between these two groups53. In a study by Lindner et al. comparing a total of 128 TNBC tumour samples from European-American women (54%) and African-American women (39%) — the remaining 7% were of Hispanic origin — the transcriptional profiles from African-American women demonstrated a gene expression signature that was consistent with increased loss of BRCA1 expression, increased activation of insulin-like growth factor 1 receptor (IGF1R) and increased expression of vascular endothelial growth factor-activated genes, as compared with the transcriptional profiles of European-American women54. Although these studies directly compared TNBC in African-American women versus European-American women, they did not control for body mass index. Increased expression of IGF1R may be attributable to obesity (which is on average higher in African-American women) and increased insulin signalling.

EZH2 network signalling

Enhancer of zeste homologue 2 (EZH2) is a member of the Polycomb group (PcG) family. PcG family members form multimeric protein complexes that maintain the transcriptional repressive state of genes over successive cell generations5557. EZH2 overexpression is observed in TNBC and is associated with poor prognosis in women of European descent55,57. EZH2 has been shown to induce AKT-dependent genomic instability and block BRCA1 function58. There is emerging evidence that EZH2 may also have a role in the aggressive biology of TNBC in African women. A joint study by Kleer and Newman59 investigated the role of EZH2 expression in 100 invasive breast cancers obtained from Ghanaian women. Of the 100 invasive carcinomas, 89% were ductal, 2% were lobular and 9% were metaplastic; 30% of the tumours were basal-type TNBC. Nuclear EZH2 overexpression was significantly associated with the basal-like subtype of TNBC (p = 0.03) in women of African descent, and was seen in 42% of basal-type TNBC cases in women of African descent. Similarly, in a set of 295 patients with breast cancer from the Netherlands, 40.3% of cases showed high EZH2 expression and a significant association with basal-type cancer (p < 0.0001)60. Therefore, EZH2 might play a part in TNBC regardless of patient ethnicity.

WNT

WNT genes encode a family of signalling molecules that are involved in embryologic development and progenitor cell renewal, and WNT–β-catenin signalling is known to be activated in TNBC arising in women of all races61. Studies specifically investigating WNT–β-catenin signalling in TNBC in women of African descent are limited. A pilot study comparing expression of WNT10B and the WNT10B target gene high mobility group AT-hook 2 (HMGA2) in TNBC from women living in Los Angeles (California, USA), Chicago (Illinois, USA), Berlin (Germany) and North Carolina (USA) shows that WNT10B and HMGA2 expression are increased in TNBCs in both women of African descent (15/17 (88%) and 15/17 (88%), respectively) and women of European descent (33/45 (73%) and 36/45 (80%), respectively)62. Differential gene expression studies comparing stage- and age-matched TNBC in women of African origin (n = 31) and women of European origin (n = 13) showed that dysregulated genes associated with the WNT–β-catenin pathway were significantly enriched (fold-change >3.0; p < 0.05) in women of African origin compared with the women of European descent, suggesting that activation of this pathway may contribute to the more aggressive phenotype of TNBC in women of African origin63.

Progenitor-like cells

Breast cancers have been reported to contain a subpopulation of cells with the ability to self-renew and undergo differentiation to phenotypically diverse populations of tumour cells64. Cell surface markers associated with self-renewal capacity include CD44 and aldehyde dehydrogenase 1 (ALDH1; also known as retinal dehydrogenase 1 and ALDH1A1) and low or no CD24 expression6466. A study by Nalwoga et al.67 tested for ALDH1 in 192 breast cancers from women of African descent. ALDH1 expression was detected in 88 breast cancer specimens from Uganda (48%) and was associated with p53 (p = 0.034) and basal-type markers (p = 0.008)67. For comparison, 19% and 30% of breast tumours were ALDH1+ in tissue collected from non-African women in Michigan (USA) and France, respectively65.

A link to aggressive biology?

Survival has improved in European-American but not in African-American women

Women of European descent diagnosed with breast cancer experienced increased survival rates in 1999–2005 relative to 1991–1998 (REF. 68). By contrast, African-American women did not experience improved breast cancer survival during this same time period68. Poor survival rates for African-American women are attributed to advanced disease presentation, co-morbid disease11, disparities in income, lack of access to breast cancer screening and oncology care, and delays in treatment11. African-American women experience an unequal burden of co-morbid disease, including obesity, diabetes and hypertension69.

There is a clear association between unequal living standards and increased levels of co-morbid disease. Disparities that promote disease include unsafe neighbourhoods, lack of access to grocery stores, stress and exposures to environmental carcinogens70. There is increasing evidence that disparities in health and co-morbid diseases may play a part in defining the biology of TNBC in women of African descent, but more work is needed before definite conclusions can be drawn (FIG. 4). Currently, the molecular link between disparities and TNBC is poorly defined. Emerging areas of research include obesity, tissue inflammation and the role of disparities in altering phosphoprotein signalling (FIG. 2) and genomic instability (FIG. 3).

Figure 4. Proposed model depicting the intersection of disparities and aggressive biology in African-American women with TNBC.

Figure 4

Lack of access to health care and cultural barriers result in delays in diagnosis, delays in treatment, lack of completion of therapy and lack of follow-up for screening tests and treatment, culminating in women presenting with breast cancer at an advanced stage and having poor survival rates. Income inequality results in unequal health insurance and lack of access to genetic testing and state-of-the-art screening for triple-negative breast cancer (TNBC), such as breast magnetic resonance imaging (MRI). Co-morbid diseases such as obesity, diabetes and hypertension complicate therapy and can result in treatment delays. Obesity increases tissue inflammation and the production of tissue cytokines and growth factors that promote aggressive cancer biology. Signalling networks activated by insulin, such as AKT–mTOR, are associated with aggressive cancer biology. Unsafe neighbourhoods and an unequal burden of environmental exposures contribute to stress, lack of access to healthy food (food deserts, which in turn promotes obesity), lack of exercise and exposure to carcinogens and heavy metals (such as cadmium, lead and arsenic) that are thought to promote abnormal imprinting and epigenetic changes8689, which in turn may promote aggressive biology. EMT, epithelial–mesenchymal transition.

Obesity and tissue inflammation

The Carolina Breast Cancer Study demonstrated that high body mass index and high waist/hip ratio increased the risk of the basal subset of TNBC in premenopausal African-American women3. Several studies, including a recent meta-analysis, have shown that obesity is a general risk factor for TNBC in premenopausal women7173. Because there is a higher incidence of obesity in African-American women and obesity predicts poor survival, it is hypothesized that obesity is a potential driver of aggressive TNBC biology in African-American women. There are many potential biological mechanisms by which obesity might increase the incidence of aggressive subtypes of TNBC in African and African-American women (FIG. 2). Obese women have tissue inflammation associated with increased circulating levels of insulin and inflammatory cytokines, including interleukin-6 (IL-6), IL-8, tumour necrosis factor (TNF) and leptin. IL-6 and IL-8 activate signal transducer and activator of transcription 3 (STAT3)–nuclear factor-κB (NF-κB) and EZH2 signalling, and predict poor prognosis in women with TNBC74,75.

Pregnancy, involution and inflammation

Approximately 21% of African-American women breastfeed in comparison to 37% of European-American women76. A lack of breastfeeding increases the risk of women developing TNBC7678. After lactation, or parturition in the absence of nursing, the mammary gland undergoes involution79. Using mice, Schedin and colleagues have shown that breast tissue involution following lactation results in tissue inflammation and activation of cyclooxygenase 2 (COX2; also known as prostaglandin G/H synthase 2), and wound-healing programmes that promote the deposition of high-risk crosslinked, fibrillar collagen79. Aligned fibrillar collagen predicts poor survival in European-American women with invasive breast cancer80. Further studies are needed to better understand the molecular link between pregnancy, parity and TNBC in African-American and African women.

Summary and future directions

Less than 20–25% of African-American women with TNBC have a germline BRCA1 mutation. Future challenges include defining the molecular signalling pathways that promote TNBC in women of African descent. It will be important to distinguish whether women of African descent have an increased incidence of TNBC subtypes that have a poor prognosis or whether there are unique biological factors that promote aggressive biology.

Studies of African women provide evidence that TNBC is a disease of young women of African descent and suggest that environmental and/or genetic factors may influence the age of onset and subtype frequency in different populations. Defining the molecular link between environmental exposures, disparities and TNBC in African-American and African women is a complex undertaking. Key areas include the identification of windows of vulnerability (such as environmental exposures in utero, during puberty and during pregnancy), the role of obesity in promoting aggressive TNBC biology and the determination of whether environmental disparities, such as unsafe neighbourhoods, stress and toxic-waste dumping, play a part in aggressive biology.

Acknowledgments

This work was supported by US National Institutes of Health/National Cancer Institute (NIH/NCI) grants R01CA155664, R01CA158668, R01CA170851 (to V.L.S.) and CA155664-03S1 (to C.S.), a Susan G. Komen Breast Promise Award (KG091020; to V.L.S.) and a V-Foundation Award (to V.L.S.). This work was also funded by a gift from F. Stanback and A. Stanback. The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Footnotes

Competing interests statement

The authors declare no competing interests.

FURTHER INFORMATION

Black Women’s Health Study: http://www.bu.edu/bwhs/

ALL LINKS ARE ACTIVE IN THE ONLINE PDF

Contributor Information

Eric C. Dietze, Duke University, Durham, North Carolina 27710, USA

Christopher Sistrunk, Duke University, Durham, North Carolina 27710, USA.

Gustavo Miranda-Carboni, University of Tennessee Health Science Center, Memphis, Tennessee 90095, USA.

Ruth O’Regan, Winship Cancer Institute, Emory University and Grady Memorial Hospital, Atlanta, Georgia 30322, USA.

Victoria L. Seewaldt, Duke University, Durham, North Carolina 27710, USA

References

  • 1.Foulkes WD, Smith IE, Reis-Filho JS. Triple-negative breast cancer. N Engl J Med. 2010;363:1938–1948. doi: 10.1056/NEJMra1001389. [DOI] [PubMed] [Google Scholar]
  • 2.Sorlie T, et al. Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA. 2001;98:10869–10874. doi: 10.1073/pnas.191367098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Carey LA, et al. Race, breast cancer subtypes, and survival in the Carolina Breast Cancer Study. JAMA. 2006;295:2492–2502. doi: 10.1001/jama.295.21.2492. [DOI] [PubMed] [Google Scholar]
  • 4.Lund MJ, et al. Race and triple negative threats to breast cancer survival: a population-based study in Atlanta, GA. Breast Cancer Res Treat. 2009;113:357–370. doi: 10.1007/s10549-008-9926-3. [DOI] [PubMed] [Google Scholar]
  • 5.Bauer KR, Brown M, Cress RD, Parise CA, Caggiano V. Descriptive analysis of estrogen receptor (ER)-negative, progesterone receptor (PR)-negative, and HER2-negative invasive breast cancer, the so-called triple-negative phenotype: a population-based study from the California cancer registry. Cancer. 2007;109:1721–1728. doi: 10.1002/cncr.22618. [DOI] [PubMed] [Google Scholar]
  • 6.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. J Natl Cancer Inst. 2009;101:984–992. doi: 10.1093/jnci/djp175. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Woodward WA, et al. African-American race is associated with a poorer overall survival rate for breast cancer patients treated with mastectomy and doxorubicin-based chemotherapy. Cancer. 2006;107:2662–2668. doi: 10.1002/cncr.22281. [DOI] [PubMed] [Google Scholar]
  • 8.Shen Y, et al. Are there racial differences in breast cancer treatments and clinical outcomes for women treated at M. D. Anderson Cancer Center? Breast Cancer Res Treat. 2007;102:347–356. doi: 10.1007/s10549-006-9337-2. [DOI] [PubMed] [Google Scholar]
  • 9.Dawood S, et al. Triple receptor-negative breast cancer: the effect of race on response to primary systemic treatment and survival outcomes. J Clin Oncol. 2009;27:220–226. doi: 10.1200/JCO.2008.17.9952. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Dean-Colomb W, et al. Transcriptional profiles of triple receptor-negative breast cancer: are Caucasian, Hispanic, and African-American women different? J Clin Oncol. 2008;26:S22014. [Google Scholar]
  • 11.Vona-Davis L, Rose DP. The influence of socioeconomic disparities on breast cancer tumor biology and prognosis: a review. J Womens Health (Larchmt) 2009;18:883–893. doi: 10.1089/jwh.2008.1127. [DOI] [PubMed] [Google Scholar]
  • 12.Danforth DN. Jr Disparities in breast cancer outcomes between Caucasian and African American women: a model for describing the relationship of biological and nonbiological factors. Breast Cancer Res. 2013;15:208. doi: 10.1186/bcr3429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Perou CM, et al. Distinctive gene expression patterns in human mammary epithelial cells and breast cancers. Proc Natl Acad Sci USA. 1999;96:9212–9217. doi: 10.1073/pnas.96.16.9212. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Perou CM, et al. Molecular portraits of human breast tumours. Nature. 2000;406:747–752. doi: 10.1038/35021093. [DOI] [PubMed] [Google Scholar]
  • 15.Morris GJ, et al. Differences in breast carcinoma characteristics in newly diagnosed African-American and Caucasian patients: a single-institution compilation compared with the National Cancer Institute’s Surveillance, Epidemiology, and End Results database. Cancer. 2007;110:876–884. doi: 10.1002/cncr.22836. [DOI] [PubMed] [Google Scholar]
  • 16.Stead LA, et al. Triple-negative breast cancers are increased in black women regardless of age or body mass index. Breast Cancer Res. 2009;11:R18. doi: 10.1186/bcr2242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Stark A, et al. African ancestry and higher prevalence of triple-negative breast cancer: findings from an international study. Cancer. 2010;116:4926–4932. doi: 10.1002/cncr.25276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Fregene A, Newman LA. Breast cancer in sub-Saharan Africa: how does it relate to breast cancer in African-American women? Cancer. 2005;103:1540–1550. doi: 10.1002/cncr.20978. [DOI] [PubMed] [Google Scholar]
  • 19.Boyle P. Triple-negative breast cancer: epidemiological considerations and recommendations. Ann Oncol. 2012;23(Suppl 6):vi7–vi12. doi: 10.1093/annonc/mds187. [DOI] [PubMed] [Google Scholar]
  • 20.Huo D, et al. Population differences in breast cancer: survey in indigenous African women reveals over-representation of triple-negative breast cancer. J Clin Oncol. 2009;27:4515–4521. doi: 10.1200/JCO.2008.19.6873. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.McCormack VA, et al. Breast cancer receptor status and stage at diagnosis in over 1,200 consecutive public hospital patients in Soweto, South Africa: a case series. Breast Cancer Res. 2013;15:R84. doi: 10.1186/bcr3478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Chlebowski RT, et al. Ethnicity and breast cancer: factors influencing differences in incidence and outcome. J Natl Cancer Inst. 2005;97:439–448. doi: 10.1093/jnci/dji064. [DOI] [PubMed] [Google Scholar]
  • 23.Bradley CJ, Given CW, Roberts C. Race, socioeconomic status, and breast cancer treatment and survival. J Natl Cancer Inst. 2002;94:490–496. doi: 10.1093/jnci/94.7.490. [DOI] [PubMed] [Google Scholar]
  • 24.Ayanian JZ, Kohler BA, Abe T, Epstein AM. The relation between health insurance coverage and clinical outcomes among women with breast cancer. N Engl J Med. 1993;329:326–331. doi: 10.1056/NEJM199307293290507. [DOI] [PubMed] [Google Scholar]
  • 25.McWhorter WP, Mayer WJ. Black/white differences in type of initial breast cancer treatment and implications for survival. Am J Public Health. 1987;77:1515–1517. doi: 10.2105/ajph.77.12.1515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Furberg H, Millikan R, Dressler L, Newman B, Geradts J. Tumor characteristics in African American and white women. Breast Cancer Res Treat. 2001;68:33–43. doi: 10.1023/a:1017994726207. [DOI] [PubMed] [Google Scholar]
  • 27.Servick K. Breast cancer: a world of differences. Science. 2014;343:1452–1453. doi: 10.1126/science.343.6178.1452. [DOI] [PubMed] [Google Scholar]
  • 28.Newman LA, et al. Meta-analysis of survival in African American and white American patients with breast cancer: ethnicity compared with socioeconomic status. J Clin Oncol. 2006;24:1342–1349. doi: 10.1200/JCO.2005.03.3472. [DOI] [PubMed] [Google Scholar]
  • 29.Hall JM, et al. Linkage of early-onset familial breast cancer to chromosome 17q21. Science. 1990;250:1684–1689. doi: 10.1126/science.2270482. [DOI] [PubMed] [Google Scholar]
  • 30.Yoshida K, Miki Y. Role of BRCA1 and BRCA2 as regulators of DNA repair, transcription, and cell cycle in response to DNA damage. Cancer Sci. 2004;95:866–871. doi: 10.1111/j.1349-7006.2004.tb02195.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Antoniou AC, Easton DF. Models of genetic susceptibility to breast cancer. Oncogene. 2006;25:5898–5905. doi: 10.1038/sj.onc.1209879. [DOI] [PubMed] [Google Scholar]
  • 32.Mavaddat N, et al. Pathology of breast and ovarian cancers among BRCA1 and BRCA2 mutation carriers: results from the Consortium of Investigators of Modifiers of BRCA1/2 (CIMBA) Cancer Epidemiol Biomarkers Prev. 2012;21:134–147. doi: 10.1158/1055-9965.EPI-11-0775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Nanda R, et al. Genetic testing in an ethnically diverse cohort of high-risk women: a comparative analysis of BRCA1 and BRCA2 mutations in American families of European and African ancestry. JAMA. 2005;294:1925–1933. doi: 10.1001/jama.294.15.1925. [DOI] [PubMed] [Google Scholar]
  • 34.Olopade OI, et al. Breast cancer genetics in African Americans. Cancer. 2003;97:236–245. doi: 10.1002/cncr.11019. [DOI] [PubMed] [Google Scholar]
  • 35.Greenup R, et al. Prevalence of BRCA mutations among women with triple-negative breast cancer (TNBC) in a genetic counseling cohort. Ann Surg Oncol. 2013;20:3254–3258. doi: 10.1245/s10434-013-3205-1. [DOI] [PubMed] [Google Scholar]
  • 36.Oluwagbemiga LA, Oluwole A, Kayode AA. Seventeen years after BRCA1: what is the BRCA mutation status of the breast cancer patients in Africa? — a systematic review. SpringerPlus. 2012;1:83. doi: 10.1186/2193-1801-1-83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Szabo CI, King MC. Population genetics of BRCA1 and BRCA2. Am J Hum Genet. 1997;60:1013–1020. [PMC free article] [PubMed] [Google Scholar]
  • 38.Yawitch TM, van Rensburg EJ, Mertz M, Falkson CI. Absence of commonly recurring BRCA1 mutations in black South African women with breast cancer. S Afr Med J. 2000;90:788. [PubMed] [Google Scholar]
  • 39.Gao Q, et al. Protein truncating BRCA1 and BRCA2 mutations in African women with pre-menopausal breast cancer. Hum Genet. 2000;107:192–194. doi: 10.1007/s004390000342. [DOI] [PubMed] [Google Scholar]
  • 40.Fackenthal JD, et al. Complete allelic analysis of BRCA1 and BRCA2 variants in young Nigerian breast cancer patients. J Med Genet. 2005;42:276–281. doi: 10.1136/jmg.2004.020446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Zhang B, et al. Evidence for an ancient BRCA1 mutation in breast cancer patients of Yoruban ancestry. Fam Cancer. 2009;8:15–22. doi: 10.1007/s10689-008-9205-9. [DOI] [PubMed] [Google Scholar]
  • 42.Zhang J, Fackenthal JD, Huo D, Zheng Y, Olopade OI. Searching for large genomic rearrangements of the BRCA1 gene in a Nigerian population. Breast Cancer Res Treat. 2010;124:573–577. doi: 10.1007/s10549-010-1006-9. [DOI] [PubMed] [Google Scholar]
  • 43.Churpek JE, et al. Inherited mutations in breast cancer genes in African-American breast cancer patients revealed by targeted genomic capture and next-generation sequencing. J Clin Oncol. 2013;31:CRA1501. [Google Scholar]
  • 44.Antoniou AC, et al. Breast-cancer risk in families with mutations in PALB2. N Engl J Med. 2014;371:497–506. doi: 10.1056/NEJMoa1400382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Zheng Y, Zhang J, Niu Q, Huo D, Olopade OI. Novel germline PALB2 truncating mutations in African American breast cancer patients. Cancer. 2012;118:1362–1370. doi: 10.1002/cncr.26388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Haiman CA, et al. A common variant at the TERT-CLPTM1L locus is associated with estrogen receptor–negative breast cancer. Nature Genet. 2011;43:1210–1214. doi: 10.1038/ng.985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ruiz-Narvaez EA, et al. Polymorphisms in the TOX3/LOC643714 locus and risk of breast cancer in African-American women. Cancer Epidemiol Biomarkers Prev. 2010;19:1320–1327. doi: 10.1158/1055-9965.EPI-09-1250. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Palmer JR, et al. Genetic susceptibility loci for subtypes of breast cancer in an African American population. Cancer Epidemiol Biomarkers Prev. 2013;22:127–134. doi: 10.1158/1055-9965.EPI-12-0769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Field LA, et al. Identification of differentially expressed genes in breast tumors from African American compared with Caucasian women. Cancer. 2012;118:1334–1344. doi: 10.1002/cncr.26405. [DOI] [PubMed] [Google Scholar]
  • 50.Martin DN, et al. Differences in the tumor microenvironment between African-American and European-American breast cancer patients. PLoS ONE. 2009;4:e4531. doi: 10.1371/journal.pone.0004531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature. 2012;490:61–70. doi: 10.1038/nature11412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Stewart PA, Luks J, Roycik MD, Sang QX, Zhang J. Differentially expressed transcripts and dysregulated signaling pathways and networks in African American breast cancer. PLoS ONE. 2013;8:e82460. doi: 10.1371/journal.pone.0082460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Sturtz LA, Melley J, Mamula K, Shriver CD, Ellsworth RE. Outcome disparities in African American women with triple negative breast cancer: a comparison of epidemiological and molecular factors between African American and Caucasian women with triple negative breast cancer. BMC Cancer. 2014;14:62. doi: 10.1186/1471-2407-14-62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Lindner R, et al. Molecular phenotypes in triple negative breast cancer from African American patients suggest targets for therapy. PLoS ONE. 2013;8:e71915. doi: 10.1371/journal.pone.0071915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Kleer CG, et al. EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc Natl Acad Sci USA. 2003;100:11606–11611. doi: 10.1073/pnas.1933744100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.De Brot M, Rocha RM, Soares FA, Gobbi H. Prognostic impact of the cancer stem cell related markers ALDH1 and EZH2 in triple negative and basal-like breast cancers. Pathology. 2012;44:303–312. doi: 10.1097/PAT.0b013e3283534bcb. [DOI] [PubMed] [Google Scholar]
  • 57.Hussein YR, et al. Clinical and biological relevance of enhancer of zeste homolog 2 in triple-negative breast cancer. Hum Pathol. 2012;43:1638–1644. doi: 10.1016/j.humpath.2011.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Gonzalez ME, et al. Histone methyltransferase EZH2 induces Akt-dependent genomic instability and BRCA1 inhibition in breast cancer. Cancer Res. 2011;71:2360–2370. doi: 10.1158/0008-5472.CAN-10-1933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Pang J, et al. Invasive breast carcinomas in Ghana: high frequency of high grade, basal-like histology and high EZH2 expression. Breast Cancer Res Treat. 2012;135:59–66. doi: 10.1007/s10549-012-2055-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Pietersen AM, et al. EZH2 and BMI1 inversely correlate with prognosis and TP53 mutation in breast cancer. Breast Cancer Res. 2008;10:R109. doi: 10.1186/bcr2214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.King TD, Suto MJ, Li Y. The Wnt/β-catenin signaling pathway: a potential therapeutic target in the treatment of triple negative breast cancer. J Cell Biochem. 2012;113:13–18. doi: 10.1002/jcb.23350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Wend P, et al. WNT10B/β-catenin signalling induces HMGA2 and proliferation in metastatic triple-negative breast cancer. EMBO Mol Med. 2013;5:264–279. doi: 10.1002/emmm.201201320. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Getz J, et al. Differential gene expression in key oncolytic pathways between node-matched Caucasian-American, African-American, and East African triple-negative breast cancer patients. Cancer Res. 2014;74:2368. [Google Scholar]
  • 64.Al-Hajj M, Wicha MS, Benito-Hernandez A, Morrison SJ, Clarke MF. Prospective identification of tumorigenic breast cancer cells. Proc Natl Acad Sci USA. 2003;100:3983–3988. doi: 10.1073/pnas.0530291100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Ginestier C, et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell. 2007;1:555–567. doi: 10.1016/j.stem.2007.08.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Fillmore CM, Kuperwasser C. Human breast cancer cell lines contain stem-like cells that self-renew, give rise to phenotypically diverse progeny and survive chemotherapy. Breast Cancer Res. 2008;10:R25. doi: 10.1186/bcr1982. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Nalwoga H, Arnes JB, Wabinga H, Akslen LA. Expression of aldehyde dehydrogenase 1 (ALDH1) is associated with basal-like markers and features of aggressive tumours in African breast cancer. Br J Cancer. 2010;102:369–375. doi: 10.1038/sj.bjc.6605488. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Silber JH, et al. Characteristics associated with differences in survival among black and white women with breast cancer. JAMA. 2013;310:389–397. doi: 10.1001/jama.2013.8272. [DOI] [PubMed] [Google Scholar]
  • 69.Black SA. Diabetes, diversity, and disparity: what do we do with the evidence? Am J Public Health. 2002;92:543–548. doi: 10.2105/ajph.92.4.543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Hood E. Dwelling disparities: how poor housing leads to poor health. Environ Health Perspect. 2005;113:A310–A317. doi: 10.1289/ehp.113-a310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Lee E, et al. Characteristics of triple-negative breast cancer in patients with a BRCA1 mutation: results from a population-based study of young women. J Clin Oncol. 2011;29:4373–4380. doi: 10.1200/JCO.2010.33.6446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Pierobon M, Frankenfeld CL. Obesity as a risk factor for triple-negative breast cancers: a systematic review and meta-analysis. Breast Cancer Res Treat. 2013;137:307–314. doi: 10.1007/s10549-012-2339-3. [DOI] [PubMed] [Google Scholar]
  • 73.Kwan ML, et al. Epidemiology of breast cancer subtypes in two prospective cohort studies of breast cancer survivors. Breast Cancer Res. 2009;11:R31. doi: 10.1186/bcr2261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Creighton CJ, et al. Residual breast cancers after conventional therapy display mesenchymal as well as tumor-initiating features. Proc Natl Acad Sci USA. 2009;106:13820–13825. doi: 10.1073/pnas.0905718106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Hartman ZC, et al. Growth of triple-negative breast cancer cells relies upon coordinate autocrine expression of the proinflammatory cytokines IL-6 and IL-8. Cancer Res. 2013;73:3470–3480. doi: 10.1158/0008-5472.CAN-12-4524-T. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Hall IJ, Moorman PG, Millikan RC, Newman B. Comparative analysis of breast cancer risk factors among African-American women and White women. Am J Epidemiol. 2005;161:40–51. doi: 10.1093/aje/kwh331. [DOI] [PubMed] [Google Scholar]
  • 77.Shinde SS, et al. Higher parity and shorter breastfeeding duration: association with triple-negative phenotype of breast cancer. Cancer. 2010;116:4933–4943. doi: 10.1002/cncr.25443. [DOI] [PubMed] [Google Scholar]
  • 78.Palmer JR, et al. Parity and lactation in relation to estrogen receptor negative breast cancer in African American women. Cancer Epidemiol Biomarkers Prev. 2011;20:1883–1891. doi: 10.1158/1055-9965.EPI-11-0465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Lyons TR, et al. Postpartum mammary gland involution drives progression of ductal carcinoma in situ through collagen and COX-2. Nature Med. 2011;17:1109–1115. doi: 10.1038/nm.2416. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Conklin MW, et al. Aligned collagen is a prognostic signature for survival in human breast carcinoma. Am J Pathol. 2011;178:1221–1232. doi: 10.1016/j.ajpath.2010.11.076. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Lu P, Weaver VM, Werb Z. The extracellular matrix: a dynamic niche in cancer progression. J Cell Biol. 2012;196:395–406. doi: 10.1083/jcb.201102147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Gonzalez-Suarez I, et al. A new pathway that regulates 53BP1 stability implicates cathepsin L and vitamin D in DNA repair. EMBO J. 2011;30:3383–3396. doi: 10.1038/emboj.2011.225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Harris SS, Dawson-Hughes B. Seasonal changes in plasma 25-hydroxyvitamin D concentrations of young American black and white women. Am J Clin Nutr. 1998;67:1232–1236. doi: 10.1093/ajcn/67.6.1232. [DOI] [PubMed] [Google Scholar]
  • 84.Clemens TL, Adams JS, Henderson SL, Holick MF. Increased skin pigment reduces the capacity of skin to synthesise vitamin D3. Lancet. 1982;1:74–76. doi: 10.1016/s0140-6736(82)90214-8. [DOI] [PubMed] [Google Scholar]
  • 85.Lancaster KJ, Bermudez OI. Beginning a discussion of nutrition and health disparities. Am J Clin Nutr. 2011;93:1161S–1162S. doi: 10.3945/ajcn.110.003459. [DOI] [PubMed] [Google Scholar]
  • 86.Murphy SK, Huang Z, Hoyo C. Differentially methylated regions of imprinted genes in prenatal, perinatal and postnatal human tissues. PLoS ONE. 2012;7:e40924. doi: 10.1371/journal.pone.0040924. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ho SM, et al. Environmental epigenetics and its implication on disease risk and health outcomes. ILAR J. 2012;53:289–305. doi: 10.1093/ilar.53.3-4.289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Pozharny Y, Lambertini L, Clunie G, Ferrara L, Lee MJ. Epigenetics in women’s health care. Mt Sinai J Med. 2010;77:225–235. doi: 10.1002/msj.20176. [DOI] [PubMed] [Google Scholar]
  • 89.Smeester L, et al. Imprinted genes and the environment: links to the toxic metals arsenic, cadmium, lead and mercury. Genes (Basel) 2014;5:477–496. doi: 10.3390/genes5020477. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES