Skip to main content
Erschienen in: Breast Cancer Research 1/2018

Open Access 01.12.2018 | Research Article

The origins of breast cancer associated with mammographic density: a testable biological hypothesis

verfasst von: Norman Boyd, Hal Berman, Jie Zhu, Lisa J. Martin, Martin J. Yaffe, Sofia Chavez, Greg Stanisz, Greg Hislop, Anna M. Chiarelli, Salomon Minkin, Andrew D. Paterson

Erschienen in: Breast Cancer Research | Ausgabe 1/2018

Abstract

Background

Our purpose is to develop a testable biological hypothesis to explain the known increased risk of breast cancer associated with extensive percent mammographic density (PMD), and to reconcile the apparent paradox that although PMD decreases with increasing age, breast cancer incidence increases.

Methods

We used the Moolgavkar model of carcinogenesis as a framework to examine the known biological properties of the breast tissue components associated with PMD that includes epithelium and stroma, in relation to the development of breast cancer. In this model, normal epithelial cells undergo a mutation to become intermediate cells, which, after further mutation, become malignant cells. A clone of such cells grows to become a tumor. The model also incorporates changes with age in the number of susceptible epithelial cells associated with menarche, parity, and menopause. We used measurements of the radiological properties of breast tissue in 4454 healthy subjects aged from 15 to 80+ years to estimate cumulative exposure to PMD (CBD) in the population, and we examined the association of CBD with the age-incidence curve of breast cancer in the population.

Results

Extensive PMD is associated with a greater number of breast epithelial cells, lobules, and fibroblasts, and greater amounts of collagen and extracellular matrix. The known biological properties of these tissue components may, singly or in combination, promote the acquisition of mutations by breast epithelial cells specified by the Moolgavkar model, and the subsequent growth of a clone of malignant cells to form a tumor. We also show that estimated CBD in the population from ages 15 to 80+ years is closely associated with the age-incidence curve of breast cancer in the population.

Conclusions

These findings are consistent with the hypothesis that the biological properties of the breast tissue components associated with PMD increase the probability of the transition of normal epithelium to malignant cells, and that the accumulation of mutations with CBD may influence the age-incidence curve of breast cancer. This hypothesis gives rise to several testable predictions.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s13058-018-0941-y) contains supplementary material, which is available to authorized users.
Abkürzungen
CAF
Cancer-associated fibroblast
CBD
Cumulative exposure to breast density
ECM
Extracellular matrix
ER
Estrogen receptor
GWAS
Genome-wide association study
HGF
Hepatocyte growth factor
IGF-1
Insulin-like growth factor 1
MD
Mammographic density
MEC
Mammary epithelial cell
MMP-3
Matrix metalloproteinase 3
MMTV
Mouse mammary tumor virus
MR
Magnetic resonance
PMD
Percent mammographic density
PyVT
Polyomavirus middle T antigen
RANKL
Receptor activator of nuclear factor-κB ligand
SNP
Single-nucleotide polymorphism
TGF-β
Transforming growth factor-β
TNF
Tumor necrosis factor

Background

Percent mammographic density (PMD) is one of the strongest known risk factors for breast cancer [1]. Fibroglandular tissue attenuates X-rays more than does fat [2], and it appears white (dense) in mammograms, whereas adipose tissue appears dark. PMD, illustrated in Additional file 1: Figure S1a, refers to the area of white tissue divided by the total area of the breast in the image. The dense area and PMD are both associated positively with risk of breast cancer, and PMD is the stronger risk factor [3]. The nondense area is associated inversely with risk of breast cancer [3, 4]. The increased risk of breast cancer associated with PMD persists for at least 8–10 years after the date of the mammogram used to assess PMD [5, 6], and it cannot be explained by the “masking” of cancers by dense breast tissue [6, 7]. In addition to an increased risk of breast cancer, PMD is also associated with an increased risk of lesions that are thought to be nonobligate precursors of breast cancer [8].
Average PMD in the population decreases with increasing age [5]. A cross-sectional study of 11,000 women in 22 countries showed that average PMD declined with increasing age. Decline was present before and after menopause and was most pronounced over the menopausal transition [9]. Longitudinal data within individuals has shown average reductions in PMD of from 5% [10] to 8% [11] respectively over 10 to 5 years.
Similar variations in breast tissue composition can be seen using measures of fat and water obtained by magnetic resonance (MR). Radiologically dense breast tissue and breast water both reflect fibroglandular breast tissue (Additional file 1: Figure S1b).
Antoni et al. showed in a meta-analysis of 19 studies with a total of > 24,000 breast cancer cases [12] that, relative to women in the lowest density category, women in the highest density category had 3.1-fold (95% CI 2.2–4.2) and 3.2-fold (1.7–5.9) increased risk of estrogen receptor-positive (ER+) and ER− breast cancer, respectively. In case-only analyses, relative risks of breast tumors for ER+ versus ER− were 1.13 (95% CI 0.89–1.42) for medium versus minimal mammographic density (MD). MD remained associated with screen-detected ER+ tumors. In eight contributing studies, the association of MD did not differ by HER2 status. Variations in the distribution by age of ER+ and ER− breast cancer are likely to be influenced by factors other than MD.
Breast cancer risk increases with increasing extent of PMD, and estimates of attributable risk (which assume causality) suggest that 30–50% of breast cancer may be attributed to the most extensive categories of PMD [5, 6]. Although MD is associated with relative and attributable risks that are large compared with other risk factors for the disease, the accuracy of risk prediction in individuals is modest [13].
The mechanisms that underlie the association of PMD with risk of breast cancer are not well defined [14], and the apparent paradox that with increasing age average PMD decreases while breast cancer incidence increases remains unexplained. We have previously proposed [15] that the radiological features of the breast of PMD provide an index of cumulative exposure to events that influence the incidence of breast cancer, similar to the concept of “breast tissue aging” proposed by Pike et al. [16]. However, to date, there is only one published study to support this suggestion [10].
In this paper, we develop a testable biological hypothesis to explain the origins of breast cancer associated with mammographic density. We summarize evidence that PMD reflects the relative quantities of epithelium, stroma, and fat in the breast, and we use a two-stage model of carcinogenesis as a framework to examine how the known biological properties of these tissues may influence the transition of normal breast epithelial cells to malignant cells [17]. We expect cumulative exposure to these biological factors to contribute to the age-specific incidence of breast cancer, and we examine the relationship between estimated cumulative exposure to PMD (CBD) in the population and the age-specific incidence of breast cancer.

Methods

Two-stage model of carcinogenesis in the breast

Figure 1 shows the two-stage model described by Moolgavkar and colleagues applied to breast cancer [17]. Normal stem cells, with a birth rate (α1) divided into two daughter cells, and rate of death or differentiation (β1) can be transformed into cells of an intermediate form at a stochastic event rate (μ1) (the first mutation rate). These intermediate cells can divide into two further intermediate cells at a stochastic rate (α2) or die or differentiate at rate β2. In addition, intermediate cells can divide into one intermediate and one transformed (malignant) cell at a second stochastic event rate (μ2).
We recognize that molecular studies implicate several genetic changes in progression for breast cancer [18], and at the time of diagnosis, breast cancer cells contain multiple somatic mutations [19, 20]. In light of this, the two stages of the model may be viewed as rate-limiting steps in the process of breast carcinogenesis, with additional mutations occurring in intermediate cells that confer sustained proliferative and survival advantages to an expanding clone of cells that ultimately undergoes malignant transformation [19, 21]. We use the two-stage model solely as a framework for examining how the biological properties of the components of breast tissue associated with PMD might influence carcinogenesis in the breast.
The Moolgavkar model applied to female breast cancer is as follows:
$$ \mathrm{I}\left(\mathrm{t}\right)\approx {\upmu}_1{\upmu}_2\mathrm{F}\left(\mathrm{t}\right) $$
where It is breast cancer incidence at age t; μ1 and μ2 are the respective first and second mutation rates; and F is the susceptible cell population, modified by menarche, parity, and menopause, to age t. After assigning numerical values to μ1, μ2, and F, the model accurately predicts the age-specific incidence of breast cancer [17].

Breast tissue associated with PMD

The histologic features of the breast associated with PMD have been examined using several approaches that include randomly selected breast tissue at forensic autopsy [22], as well as a comparison of radiologically dense and nondense regions in mastectomy specimens [23] and in surgical biopsies [2426]. These approaches have shown similar results.
Selected results derived from randomly selected sections of breast tissue collected at forensic autopsy by Bartow et al. [27] are shown in Fig. 2. PMD was assessed in the BioVision (Faxitron Bioptics, Tucson, AZ, USA) image of the enucleated breast from which the section had been taken (Fig. 2a) [22]. We used quantitative microscopy in randomly selected areas of the tissue section (Fig. 2b) to measure the total, epithelial, and nonepithelial nuclear areas, which we used as an index of the number of cells (Fig. 2d), the area of Masson’s trichrome-stained collagen (Fig. 2g), and the glandular area. PMD was associated inversely with age, and after adjustment for age, positively with the nuclear area (a measure of the number of cells) (Fig. 2e) of epithelial and nonepithelial cells, glandular area, and the area of collagen (Fig. 2h). As shown in Additional file 1: Table S1, age, parity, and menopause were each associated inversely with one or more of these tissue components [22].
The stronger risk prediction seen with PMD compared with dense area suggests that the nondense area of the mammogram, which reflects fat, may provide some protection. A reduced risk of breast cancer associated with the nondense area of the mammogram has been shown in a meta-analysis by Pettersson et al. [3]. The mechanism underlying this protection is currently uncertain, but as shown in Fig. 2, breasts with low PMD are associated with fewer cells (as shown by nuclear area) and less extensive collagen, tissue components whose biological properties we show are associated with radiologically dense breast tissue and that may contribute to carcinogenesis in the breast. Further, aromatase activity in the breast is predominately in stromal preadipocytes [28] and is reduced when preadipocytes differentiate to mature adipocytes. The loss of this source of local estrogen production may contribute to the reduced risk of breast cancer associated with breast fat. As Fig. 2 shows, there can be wide variation in the number of cells (as shown by nuclear area) and the area of collagen between individuals, and it is not currently possible to assess these variations separately, or to link them to risk, using only currently available methods of imaging.

Biological properties of breast tissue associated with PMD

Breast epithelium

Breast cancer is thought to originate in the epithelial cells of the terminal ductal lobular unit [29, 30] and to be the result of the accumulation of genetic mutations [21]. The greater number of epithelial cells and greater glandular area associated with PMD may be the result of either increased cell proliferation or a reduction in the rate of cell death. Both processes increase the size of the population of susceptible epithelial cells and increase the probability of a mutation. Some breast mitogens have been associated with risk of breast cancer [3133], and proliferative activity in epithelial cells, as shown by the Ki-67 index, predicts risk of breast cancer in premenopausal women [34].
Although in adult life epithelial cells associated with PMD do not have an increased Ki-67 proliferative index [35], the greater number of epithelial cells associated with PMD in adult life may be the result of greater proliferation of progenitor cells during breast development, when susceptibility to carcinogens is also greatest [36]. The chemokine CCL2 has been detected in human mammary epithelium, and when overexpressed in mouse mammary epithelium, it induces a state of low-level inflammation that increases stromal density and risk of mammary cancer [37].

Stroma

Collagen, fibroblasts, other mesenchymal cells, and extracellular matrix (ECM) are stromal components that contribute to PMD. Selected biological properties of these components of stroma are discussed briefly in the following sections and summarized in Table 1. Some of the components of stroma considered here have multiple biological functions, and we have selected those functions that appear most likely to be relevant to the processes outlined above in the two-stage model of carcinogenesis.
Table 1
Selected biological properties of components of breast stroma
Tissue
Model
Biological effects
Two-stage model
References
Collagen
Transgenic mice (Col1a1) + PyVT transgene + MMTV promoter
2.5-fold increase in stromal collagen + 3-fold increase tumor incidence
1 + 2
[38]
Human MEC + collagen gel
Increase migration of cancer stem cells.
2
[39, 40]
Fibroblasts
Human MEC + stromal cells in mouse
Genetically modified human stromal fibroblasts promote outgrowth of benign and malignant lesions from MEC.
1 + 2
[41]
Human cells in culture + mouse models
TGF-β and HGF produced by stromal fibroblasts inhibit and stimulate, respectively, proliferation in adjacent epithelial cells.
1 + 2
[4244]
TGF-β promotes epithelial-mesenchymal transition and changes in the microenvironment that promote tumor progression.
2
[44, 45]
Cancer associated fibroblasts (CAFs)
CAFs in stroma associated with breast cancer promote tumor dissemination.
2
[42, 4649, 56]
Other cells
Aromatase in stromal preadipocytes in various models including human breast tissue
Estrogen produced by aromatase increases MEC proliferation + tumor growth.
1 + 2
[5055]
ECM
Human breast tissue
   
Proteoglycans
Overexpression of lumican and decorin in PMD and breast cancer bind growth factors.
1 + 2
[59]
MMP-3
Metalloproteinases regulate stromal matrix and the activation of growth factors.
1 + 2
[60, 61]
Stiffness
Promotes tumorigenesis and growth
 
[57, 58]
Abbreviations: ECM Extracellular matrix, MEC Mammary epithelial cell, PyVT Polyomavirus middle T antigen, MMTV Mouse mammary tumor virus, TGF-β Transforming growth factor-β, HGF Hepatocyte growth factor, MMP-3 Matrix metalloproteinase 3, PMD: Percent mammographic density
Collagen
Provenzano et al. [38] showed in a bitransgenic mouse tumor model, with both increased density of stromal collagen (Col1a1tmJae) and carrying the polyoma middle T transgene under the control of mammary-specific mouse mammary tumor virus promoter, that both epithelial cell proliferation and tumor formation were increased. Tumor formation increased approximately threefold, and tumors had a more invasive phenotype and a greater frequency of metastasis [38].
Preliminary human data suggest that periductal aligned collagen fibrils, rather than amorphous collagen, is associated with PMD [39]. Aligned collagen matrices also enhances the migration of cancer stem cells [40].
Fibroblasts
Stromal fibroblasts are the principal source of collagen and can regulate the morphogenesis of breast epithelial cells. Kuperwasser et al. showed that human stromal fibroblasts from reduction mammoplasty, immortalized with human telomerase, and implanted with normal human mammary epithelial cells (MECs) into the cleared mammary fat pad of severe combined immunodeficiency mice, resulted in the outgrowth of benign and malignant epithelial lesions [41].
Stromal fibroblasts regulate the growth of epithelial cells in part through the secretion of growth factors and chemokines [42] that include hepatocyte growth factor (HGF), insulin-like growth factor 1 (IGF-1), and transforming growth factor-β (TGF-β). HGF and IGF-1 both promote epithelial cell proliferation and tumor growth. Stromal fibroblast-derived TGF-β [43] inhibits MEC proliferation in vivo but can promote malignant behavior through diverse mechanisms that include stimulation of epithelial-mesenchymal transition [44]. TGF-β also has several effects on the microenvironment, including increasing ECM and inducing endothelial cell recruitment and proliferation, that promote tumor progression (reviewed in [45]). Fibroblasts also deposit ECM and produce collagen types I, III, and V and fibronectin (reviewed in [43]). Fibroblasts derived from disease-free breasts with extensive PMD promote adipocyte differentiation in culture and show decreased expression of CD36 [46] (see below).
The stroma associated with breast cancer contains fibroblasts (cancer-associated fibroblasts [CAFs]) that produce chemokines, growth factors, and ECM proteins, which are thought to contribute to the dissemination of malignant tumors [43, 47], and foci of fibrous tissue within invasive breast cancer are associated with an increased risk of disease recurrence [48, 49].
Other cells
Aromatase activity in the breast is a source of estrogen that may stimulate proliferation of epithelial cells and promote the growth of malignant clones [5053]. Aromatase activity in adipose tissue is expressed primarily in stromal mesenchymal preadipocytes rather than in lipid-laden adipocytes and is greatest in breast tissue where the ratio of fibroblasts to adipocytes is greatest [54], and most aromatase activity in the breast is in radiologically dense regions [50, 51, 53, 55]. The role of immune cells in PMD has received little attention to date, but Huo et al. showed in prophylactic mastectomy samples that radiologically dense areas of the breast contained fewer CD26 activated macrophages and more vimentin+/CD45 immune cells than nondense regions in the same individuals [23, 56].
Extracellular matrix
The ECM is comprised of collagens, fibronectin, laminins, polysaccharides, and proteoglycans, and it influences the changes that occur in the breast during pregnancy, lactation, involution, and tumorigenesis (see [57, 58] for reviews). Expression of the proteoglycans lumican and decorin, assessed by semiquantitative scoring of immunohistochemistry, is increased in stromal tissue associated with breast cancer and, in the absence of invasive breast cancer, in women with extensive PMD. PMD is associated with lumican and decorin scores and with duct fibrosis and collagen, but not with the tissue or ductal lobular density [59]. Proteoglycans bind growth factors, contribute to the mechanical integrity of tissues, and influence the stiffness of breast tissue that promotes tumorigenesis, tumor growth, and the invasion of malignant tissue [57, 58].
Radiologically dense breast tissue also has greater amounts of the stromal matrix regulatory protein tissue inhibitor of metalloproteinase 3 [60] that regulates stromal matrix, the activation of growth factors, and influences susceptibility to breast cancer [61]. In addition, the transmembrane receptor CD36 controls adipogenesis and deposition of the ECM. CD36-knockout mice show increased collagen and decreased fat in the mammary gland, and reduced expression of CD36 has been found to be associated with greater PMD and tumor stroma in human breast tissue [46].
Radiologically dense human breast tissue obtained from mastectomy specimens has been shown to promote the growth and progression of human carcinoma in situ xenografts in immunodeficient mice [62]. The biological properties shown in Table 1 have all, with the exception of collagen density, been observed in human cells or tissues, but only three (proteoglycan expression, matrix metalloproteinase 3 [MMP-3], and CD36 expression) have been examined to date in relation to PMD.
Genetic variants associated with histologic features
Twin and sister studies have shown that more than 60% of the variation in PMD in the population can be explained by additive genetic effects [63, 64]. Genetic variants associated with PMD dense or nondense areas are likely to be associated, directly or indirectly, with one or more of the tissue components that are reflected by these mammographic features.
Genome-wide association studies (GWASs) have identified some of the genetic variants associated with PMD. Here we limit our attention to the nine regions, comprised of eight genes and one locus on chromosome 8, shown using a two-stage design to be reproducibly associated with PMD adjusted for age and body mass index. Eight of the nine loci are also associated with the risk of breast cancer [65, 66]. Single-nucleotide polymorphisms (SNPs) near or in PRDM6 and THEM184B and the locus on chromosome 8 have been associated with PMD, and SNPs near AREG, ESR1, ZNF365, LSP1, IGF1, and SGSM3/MKL1 have all been associated with the area of dense tissue in the mammogram. The locus on chromosome 8 has also been associated with nondense area.
Although it is recognized that proximity of SNPs to genes may not identify causal genes [67, 68], functions for genes near eight of these regions were found by searching under the gene names in PubMed, the National Center for Biotechnology Information database of Genotypes and Phenotypes (dbGaP) of genotype-phenotype associations, and the GWAS Catalog (http://​www.​ebi.​ac.​uk/​gwas/​) [69]. The known functions of these eight genes of potential relevance to the components of breast tissue that are associated with PMD include the following:
  • AREG encodes amphiregulin that binds epidermal growth factor receptor, stimulates cell growth and survival, and plays a role in the development of the mammary gland [70]. Amphiregulin also promotes the growth of fibroblasts, the expression of collagen and other genes associated with the ECM, and interacts with TGF-β to stimulate fibroblast proliferation [71].
  • ESR1 encodes ER-α that mediates the physiological effects of estrogen [72]. Estrogen influences epithelial cell proliferation, and the secretion of the pituitary hormones growth hormone and prolactin that are breast mitogens [32, 7375].
  • IGF-1 encodes IGF-1 [76] and has mitogenic and antiapoptotic effects on breast epithelial cells [77]. Serum levels of IGF-1 have been associated with breast cancer risk in meta-analysis [31] and with PMD in some but not all studies (reviewed in [1]).
    Greater adult height is associated with risk of breast cancer [78] and has been positively associated with percent breast water (which, like PMD, reflects fibroglandular tissue) in young women [75] and with PMD [79] in adult women. Variants near MKL1, SGSM3, and IGF1 (in Japanese subjects) are associated with height [80, 81].
  • MKL1 is the human homologue of a murine gene (Bsac) that, when overexpressed in mice that are double-knockout for tumor necrosis factor (TNF)-associated factor, protects murine embryonic fibroblasts against cell death induced by TNF [82].
  • LSP1 [83] does not currently have any described function that is specific to breast tissue, apart from the observed associations with PMD and breast cancer.

Results

Application of two-stage model to association of PMD with breast cancer

Figure 3 summarizes how the biological properties of the tissue components associated with PMD that are summarized in Table 1 might influence the first and second stages and transitions of the two-stage model. The third column represents the growth of a clone of malignant cells to become a detectable tumor.
The probability that the first event converts a normal epithelial stem cell to an intermediate cell is expected to be proportional to the number of cells at risk, their survival time, the number of cell divisions, and the dose and duration of exposure to mutagens [84]. As shown above, the extent of PMD is associated positively with the number of epithelial cells. Further, as shown in Table 1, experimental data show that epithelial cell proliferation and survival are increased by greater density of collagen, by the growth factors associated with fibroblasts and proteoglycans, by greater stiffness of the ECM, and by the local production of estrogen by aromatase, as well as by the influence of systemic hormones and growth factors.
These factors may operate singly or, more likely, in combination to promote the expansion of the pool of normal and intermediate cells, the acquisition of additional mutations that confer proliferative and survival advantages, the transition of intermediate cells to malignancy, and the subsequent growth of a clone of malignant cells to become a detectable tumor.

Cumulative exposure to PMD and age-specific incidence of breast cancer

The postulated expansion with increasing age of the number of intermediate cells with mutations, together with continuing exposure to several components of the breast stroma that promote carcinogenesis, suggests that CBD may be related to the age-specific incidence of breast cancer [85, 86]. CBD may account for the observation that with increasing age, PMD and the total number of epithelial cells and lobular units decrease, whereas breast cancer incidence increases.

Estimated cumulative breast density in the population

We estimated CBD in the population using cross-sectional data from 4454 healthy females, predominately Caucasian and aged 15–81 years, who had participated in previous studies in which PMD was measured. Additional file 1: Table S2 shows selected characteristics of these subjects.
We measured PMD by mammography [87] (shown in Additional file 1: Figure S1a) in women over the age of 35 and by percent breast water by MR (shown in Additional file 1: Figure S1b) in those under 35 [75]. Both measures reflect fibroglandular breast tissue [88] and are strongly correlated with each other within the same individuals (rs = 0.85) [75]. We used percent breast water by MR and PMD obtained in 100 adult women to calibrate MR measures in young women to the equivalent mammographic measure (see Table 2 footnote).
Table 2
Calculation of cumulative percent mammographic density
Age (years)
No. of subjects
Median calibrated percent watera
Median percent dense area
Years
Calibrated breast density years
Calibrated cumulative breast density years
15–19
974
51.7
 
5
258.5
258.6
20–24
86
46.5
 
5
232.5
491.1
25–29
83
47.0
 
5
235.0
726.1
30–34
15
46.3
 
5
231.5
957.6
35–39
49
 
45.9
5
229.6
1187.3
40–44
405
 
40.4
5
202.0
1389.3
45–49
654
 
37.3
5
186.6
1575.9
50–54
789
 
28.4
5
141.9
1717.8
55–59
546
 
23.8
5
119.0
1836.9
60–64
324
 
23.1
5
115.3
1952.2
65–69
259
 
19.2
5
96.1
2048.3
70–74
171
 
18.5
5
92.5
2140.8
75–79
76
 
18.3
5
91.3
2232.1
80+
23
 
15.2
5
76.0
2308.1
Percent breast water was calibrated to breast density equivalent as follows: calibrated percent breast density = (80.00813 − 1365.42571)/percent breast water
aIn previous work, a random sample of 100 mothers was selected from among a total of 356 whose daughters had participated [75]. Their average age was 49.6 years (SD 4.2 years), and this and other characteristics were similar to those of mothers who did not have magnetic resonance imaging (see Table 1 in [75]). Mammographic measures of all mothers are included in the data shown in the table above
As shown in Table 2, we divided subjects into the same 5-year age categories in which breast cancer incidence in the population is reported. Median PMD decreased with increasing age and was 51.7% after calibration in the youngest group and 15.2% in the oldest. We multiplied the median PMD in each age category by the 5 years in the category to generate a variable we call “breast density years,” and we summed the product for each age group to give an estimate of CBD from ages 15 to 80+ years in the population.

Association of cumulative breast density with age-specific incidence of breast cancer

We examine in Fig. 4 the association of CBD with the age-specific incidence of invasive breast cancer in Canada. We compared the log age-specific breast cancer incidence in the Canadian population predicted for each 5-year age group using regression models, one based on log age alone, one based on log CBD alone, and one based on log age + log CBD. We used R2 (the proportion of the total variance explained) and a comparison of the observed and predicted age-incidence curve of breast cancer to assess the fit of each model. The models, the associated coefficients, and the results are shown in Additional file 1: Table S3.
As shown in Fig. 4 and Additional file 1: Table S3, we found a strong association (r2 = 0.99) between log CBD and log breast cancer incidence in the population using the following model:
$$ \mathrm{Log}\ \mathrm{I}\left(\mathrm{t}\right)\approx \log\ {\left(\mathrm{CBD}\left(\mathrm{t}\right)\right)}^{\mathrm{k}} $$
where Log It is log breast cancer incidence at age t, CBDt is the sum of median PMD in each age group (each multiplied by 5, the age interval) from age 15 to age t, and the exponent k has the estimated value of 3.5. The model based on log age alone was less strongly associated with breast cancer incidence (Additional file 1: Table S3), and the addition of log age to log CBD did not change the association with breast cancer incidence.
This model based on log CBD and the two-stage model of Moolgavkar et al. described above both accurately predict the age-specific incidence of breast cancer in the populations considered. The function Ft in the Moolgavkar model, and CBDt in the present model both reflect variations in the number of susceptible cells in the breast modified by menarche, pregnancy, and menopause [89].
The strong correlation observed between log CBD and log breast cancer incidence cannot be explained by their shared association with age. Log CBD alone was a better predictor of age-specific log breast cancer incidence than was log age, and there was no change in prediction by a model containing both CBD and age.

Discussion

The close relationship observed between log CBD and log breast cancer incidence is consistent with the hypothesis of carcinogenesis in which the accumulation of mutations or other molecular changes increases with increasing duration of exposure to PMD rather than with age. Limitations of our data include the cross-sectional observations and the ecological comparison with breast cancer incidence, as well as the small numbers at ages 30–34 and 80+ years. We estimated CBD from cross-sectional rather than longitudinal observations, using film rather than digital mammograms. However, longitudinal assessments of breast density in women aged 40 or older have shown a decline in average PMD with increasing age and menopause that is very similar to the differences seen here [10]. Further, Maskarinec et al. showed a strong association between cumulative density and age-specific breast cancer incidence in serial mammograms from 607 patients with breast cancer and 667 control subjects in the Hawaii component of the multiethnic cohort, in which the average age at first mammogram was 57 years [10]. However, the associations of cumulative density and breast cancer incidence with age were not examined [17].
CBD may also explain many of the known epidemiological associations with breast cancer risk. As shown above, the estimated size of the susceptible cell population of epithelial cells and epithelial cell proliferation are greatest at early ages and decline with increasing age. The greater amount of fibroglandular tissue, as shown by percent water, present at ages 15–18 may be related to the greater susceptibility of the breast at early ages to the effects of known exposures on risk of breast cancer, including radiation, alcohol, and smoking [36].
Early menarche is associated with an increased risk of breast cancer in later life [90] and advances the age at which fibroglandular breast tissue develops. This addition to the time of exposure will influence all estimates of PMD at later ages and will increase CBD. An early pregnancy and early menopause both reduce later risk of breast cancer and PMD [90]. The reductions in PMD associated with these events will influence all measures of PMD at later ages and reduce average CBD in parous and postmenopausal women, respectively. At least some of the effect of pregnancy in reducing risk of breast cancer has been shown to be mediated by the reduction in PMD associated with pregnancy [91].
Tamoxifen reduces PMD and risk of breast cancer, and reduction in PMD appears to predict response to adjuvant therapy with tamoxifen [92]. Progesterone as a postmenopausal replacement therapy has been shown to increase both PMD and breast cancer incidence, and the effect of progesterone on breast cancer incidence has been shown to be mediated through the effect on PMD [93]. The proliferation of mammary epithelium in response to progesterone is mediated by receptor activator of nuclear factor-κB ligand (RANKL), and increased expression of RANKL has been found to be associated with more extensive PMD in premenopausal women [94].
The biological hypothesis that we propose from the foregoing considerations is that the transition of breast epithelial cells from normal to malignant cells is completed more frequently in dense breast tissue than in nondense tissue. We propose that this transition is associated with the acquisition of mutations or other molecular changes in breast epithelial cells that increase in frequency with increasing exposure to both the amount and duration of PMD. We propose that the probability of acquiring mutations is influenced by the greater number of epithelial cells and by the several known biological properties of the stromal tissues that are associated with PMD, described in Table 1, by the amounts of such tissues, and by the duration of exposure to these influences. Proteoglycans and MMP-3 in the ECM of radiologically dense breast tissue have already been shown, in the absence of breast cancer, to be similar to those expressed in breast tissue associated with breast cancer.
Additional influences may include the greater number of stromal fibroblasts and associated chemokines associated with PMD that may, in the absence of breast cancer, resemble CAFs. CAFs can be distinguished from normal fibroblasts by markers and functional assays. Among these properties is the production of TGF-β1, which promotes epithelial-mesenchymal transition and has effects on the microenvironment that promote tumorigenesis and tumor invasion (reviewed in [45]). Epithelial-mesenchymal transition and other changes in the microenvironment may, in the absence of breast cancer, be more extensive in radiologically dense breast tissue than in nondense tissue [45].

Conclusions

PMD has reproducibly been shown to be a strong risk factor for breast cancer that may account for a substantial fraction of the disease. The biological basis for this association is currently unknown, however. We have examined potential biological mechanisms for the risk of breast cancer associated with PMD using a two-stage model of carcinogenesis as a framework.
It is understood that it is the biological properties of the breast tissues associated with PMD, not the radiological properties, that are responsible for the association of PMD with risk of breast cancer. PMD is known to be associated with a greater number of epithelial cells, greater glandular area, a greater area of collagen, and a greater number of nonepithelial cells. The known biological properties of these breast tissue components increase the probability of mutation and of transition to malignant cells. The finding that CBD in healthy subjects in the population, estimated from cross-sectional observations in healthy women, was strongly associated with the age-specific incidence of breast cancer in Canada and is consistent with the accumulation of mutations with increasing time of exposure to CBD. This biological model gives rise to a number of testable predictions concerning the properties of breast tissue associated with PMD and suggests that the radiological features of the breast may be useful in the design, sampling, analysis, and interpretation of research on the biology of breast tissues in relation to breast cancer.

Acknowledgements

We acknowledge the contribution of Dr. Alice S. Whittemore, Stanford University, who directed our attention to the Moolgavkar model.

Funding

The studies that provided data for the present paper were supported by grants from the Canadian Breast Cancer Research Alliance, the National Cancer Institute of Canada, and the National Institutes of Health (grant R01 CA082826-01). The Ontario Ministry of Health and Long-Term Care also supported this work.

Availability of data and materials

The datasets used during the present study are available from the corresponding author on reasonable request.
The data in the section of the manuscript concerned with CBD and the age-specific incidence of breast cancer were obtained in a number of separate studies for which ethics approval was obtained from the University Health Network, Sunnybrook Hospital, and Women’s College Hospital (all in Toronto), and from the Toronto District School Board, the Toronto Catholic District School Board, the York Region District School Board, the York Catholic District School Board, and the mammography screening programs of Ontario and British Columbia.
All subjects provided signed inform consent.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat Boyd NF, Martin LJ, Yaffe MJ, Minkin S. Mammographic density and breast cancer risk: current understanding and future prospects. Breast Cancer Res. 2011;13(6):223.CrossRefPubMedPubMedCentral Boyd NF, Martin LJ, Yaffe MJ, Minkin S. Mammographic density and breast cancer risk: current understanding and future prospects. Breast Cancer Res. 2011;13(6):223.CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Johns PC, Yaffe MJ. X-ray characterisation of normal and neoplastic breast tissues. Phys Med Biol. 1987;32(6):675–95.CrossRefPubMed Johns PC, Yaffe MJ. X-ray characterisation of normal and neoplastic breast tissues. Phys Med Biol. 1987;32(6):675–95.CrossRefPubMed
3.
Zurück zum Zitat Pettersson A, Graff RE, Ursin G, Santos Silva ID, McCormack V, Baglietto L, Vachon C, Bakker MF, Giles GG, Chia KS, et al. Mammographic density phenotypes and risk of breast cancer: a meta-analysis. J Natl Cancer Inst. 2014:106(5). Pettersson A, Graff RE, Ursin G, Santos Silva ID, McCormack V, Baglietto L, Vachon C, Bakker MF, Giles GG, Chia KS, et al. Mammographic density phenotypes and risk of breast cancer: a meta-analysis. J Natl Cancer Inst. 2014:106(5).
4.
Zurück zum Zitat Bertrand KA, Scott CG, Tamimi RM, Jensen MR, Pankratz VS, Norman AD, Visscher DW, Couch FJ, Shepherd J, Chen YY, et al. Dense and nondense mammographic area and risk of breast cancer by age and tumor characteristics. Cancer Epidemiol Biomarkers Prevent. 2015;24(5):798–809.CrossRef Bertrand KA, Scott CG, Tamimi RM, Jensen MR, Pankratz VS, Norman AD, Visscher DW, Couch FJ, Shepherd J, Chen YY, et al. Dense and nondense mammographic area and risk of breast cancer by age and tumor characteristics. Cancer Epidemiol Biomarkers Prevent. 2015;24(5):798–809.CrossRef
5.
Zurück zum Zitat Byrne C, Schairer C, Wolfe J, Parekh N, Salane M, Brinton LA, Hoover R, Haile R. Mammographic features and breast cancer risk: effects with time, age, and menopause status. J Natl Cancer Inst. 1995;87(21):1622–9.CrossRefPubMed Byrne C, Schairer C, Wolfe J, Parekh N, Salane M, Brinton LA, Hoover R, Haile R. Mammographic features and breast cancer risk: effects with time, age, and menopause status. J Natl Cancer Inst. 1995;87(21):1622–9.CrossRefPubMed
6.
Zurück zum Zitat Boyd NF, Guo H, Martin LJ, Sun L, Stone J, Fishell E, Jong RA, Hislop G, Chiarelli A, Minkin S, et al. Mammographic density and the risk and detection of breast cancer. N Engl J Med. 2007;356(3):227–36.CrossRefPubMed Boyd NF, Guo H, Martin LJ, Sun L, Stone J, Fishell E, Jong RA, Hislop G, Chiarelli A, Minkin S, et al. Mammographic density and the risk and detection of breast cancer. N Engl J Med. 2007;356(3):227–36.CrossRefPubMed
7.
Zurück zum Zitat McCormack VA, dos Santos SI. Breast density and parenchymal patterns as markers of breast cancer risk: A meta-analysis. Cancer Epidemiol Biomarkers Prevent. 2006;15(6):1159–69.CrossRef McCormack VA, dos Santos SI. Breast density and parenchymal patterns as markers of breast cancer risk: A meta-analysis. Cancer Epidemiol Biomarkers Prevent. 2006;15(6):1159–69.CrossRef
8.
Zurück zum Zitat Boyd NF, Jensen HM, Cooke G, Han HL. Relationship between mammographic and histological risk factors for breast cancer. J Natl Cancer Inst. 1992;84(15):1170–9.CrossRefPubMed Boyd NF, Jensen HM, Cooke G, Han HL. Relationship between mammographic and histological risk factors for breast cancer. J Natl Cancer Inst. 1992;84(15):1170–9.CrossRefPubMed
9.
Zurück zum Zitat Burton A, Maskarinec G, Perez-Gomez B, Vachon C, Miao H, Lajous M, Lopez-Ridaura R, Rice M, Pereira A, Garmendia ML, et al. Mammographic density and ageing: A collaborative pooled analysis of cross-sectional data from 22 countries worldwide. PLoS Med. 2017;14(6):e1002335.CrossRefPubMedPubMedCentral Burton A, Maskarinec G, Perez-Gomez B, Vachon C, Miao H, Lajous M, Lopez-Ridaura R, Rice M, Pereira A, Garmendia ML, et al. Mammographic density and ageing: A collaborative pooled analysis of cross-sectional data from 22 countries worldwide. PLoS Med. 2017;14(6):e1002335.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Maskarinec G, Pagano I, Lurie G, Kolonel LN. A longitudinal investigation of mammographic density: the multiethnic cohort. Cancer Epidemiol Biomarkers Prev. 2006;15(4):732–9.CrossRefPubMed Maskarinec G, Pagano I, Lurie G, Kolonel LN. A longitudinal investigation of mammographic density: the multiethnic cohort. Cancer Epidemiol Biomarkers Prev. 2006;15(4):732–9.CrossRefPubMed
11.
Zurück zum Zitat Boyd N, Martin L, Stone J, Little L, Minkin S, Yaffe M. A longitudinal study of the effects of menopause on mammographic features. Cancer Epidemiol Biomarkers Prev. 2002;11(10 Pt 1):1048–53.PubMed Boyd N, Martin L, Stone J, Little L, Minkin S, Yaffe M. A longitudinal study of the effects of menopause on mammographic features. Cancer Epidemiol Biomarkers Prev. 2002;11(10 Pt 1):1048–53.PubMed
12.
Zurück zum Zitat Antoni S, Sasco AJ, dos Santos SI, McCormack V. Is mammographic density differentially associated with breast cancer according to receptor status? A meta-analysis. Breast Cancer Res Treat. 2013;137(2):337–47.CrossRefPubMed Antoni S, Sasco AJ, dos Santos SI, McCormack V. Is mammographic density differentially associated with breast cancer according to receptor status? A meta-analysis. Breast Cancer Res Treat. 2013;137(2):337–47.CrossRefPubMed
13.
Zurück zum Zitat Chen J, Pee D, Ayyagari R, Graubard B, Schairer C, Byrne C, Benichou J, Gail MH. Projecting absolute invasive breast cancer risk in white women with a model that includes mammographic density. J Natl Cancer Inst. 2006;98(17):1215–26.CrossRefPubMed Chen J, Pee D, Ayyagari R, Graubard B, Schairer C, Byrne C, Benichou J, Gail MH. Projecting absolute invasive breast cancer risk in white women with a model that includes mammographic density. J Natl Cancer Inst. 2006;98(17):1215–26.CrossRefPubMed
14.
Zurück zum Zitat Sherratt MJ, McConnell JC, Streuli CH. Raised mammographic density: causative mechanisms and biological consequences. Breast Cancer Res. 2016;18(1):45.CrossRefPubMedPubMedCentral Sherratt MJ, McConnell JC, Streuli CH. Raised mammographic density: causative mechanisms and biological consequences. Breast Cancer Res. 2016;18(1):45.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Boyd NF, Lockwood GA, Martin LJ, Byng JW, Yaffe MJ, Tritchler DL. Mammographic density as a marker of susceptibility to breast cancer: a hypothesis. IARC Sci Publ. 2001;154:163–9.PubMed Boyd NF, Lockwood GA, Martin LJ, Byng JW, Yaffe MJ, Tritchler DL. Mammographic density as a marker of susceptibility to breast cancer: a hypothesis. IARC Sci Publ. 2001;154:163–9.PubMed
16.
Zurück zum Zitat Pike MC, Krailo MD, Henderson BE, Casagrande JT, Hoel DG. ‘Hormonal’ risk factors, 'breast tissue age’ and the age-incidence of breast cancer. Nature. 1983;303(5920):767–70.CrossRefPubMed Pike MC, Krailo MD, Henderson BE, Casagrande JT, Hoel DG. ‘Hormonal’ risk factors, 'breast tissue age’ and the age-incidence of breast cancer. Nature. 1983;303(5920):767–70.CrossRefPubMed
17.
Zurück zum Zitat Moolgavkar SH, Day NE, Stevens RG. Two-stage model for carcinogenesis: Epidemiology of breast cancer in females. J Natl Cancer Inst. 1980;65(3):559–69.PubMed Moolgavkar SH, Day NE, Stevens RG. Two-stage model for carcinogenesis: Epidemiology of breast cancer in females. J Natl Cancer Inst. 1980;65(3):559–69.PubMed
18.
Zurück zum Zitat Newburger DE, Kashef-Haghighi D, Weng Z, Salari R, Sweeney RT, Brunner AL, Zhu SX, Guo X, Varma S, Troxell ML, et al. Genome evolution during progression to breast cancer. Genome Res. 2013;23(7):1097–108.CrossRefPubMedPubMedCentral Newburger DE, Kashef-Haghighi D, Weng Z, Salari R, Sweeney RT, Brunner AL, Zhu SX, Guo X, Varma S, Troxell ML, et al. Genome evolution during progression to breast cancer. Genome Res. 2013;23(7):1097–108.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Nik-Zainal S, Van Loo P, Wedge DC, Alexandrov LB, Greenman CD, Lau KW, Raine K, Jones D, Marshall J, Ramakrishna M, et al. The life history of 21 breast cancers. Cell. 2012;149(5):994–1007.CrossRefPubMedPubMedCentral Nik-Zainal S, Van Loo P, Wedge DC, Alexandrov LB, Greenman CD, Lau KW, Raine K, Jones D, Marshall J, Ramakrishna M, et al. The life history of 21 breast cancers. Cell. 2012;149(5):994–1007.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Martincorena I, Raine KM, Gerstung M, Dawson KJ, Haase K, Van Loo P, Davies H, Stratton MR, Campbell PJ. Universal Patterns of Selection in Cancer and Somatic Tissues. Cell. 2017;171(5):1029–41. e1021CrossRefPubMedPubMedCentral Martincorena I, Raine KM, Gerstung M, Dawson KJ, Haase K, Van Loo P, Davies H, Stratton MR, Campbell PJ. Universal Patterns of Selection in Cancer and Somatic Tissues. Cell. 2017;171(5):1029–41. e1021CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.CrossRefPubMed Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646–74.CrossRefPubMed
22.
Zurück zum Zitat Li T, Sun L, Miller N, Nicklee T, Woo J, Hulse-Smith L, Tsao MS, Khokha R, Martin L, Boyd N. The association of measured breast tissue characteristics with mammographic density and other risk factors for breast cancer. Cancer Epidemiol Biomarkers Prev. 2005;14(2):343–9.CrossRefPubMed Li T, Sun L, Miller N, Nicklee T, Woo J, Hulse-Smith L, Tsao MS, Khokha R, Martin L, Boyd N. The association of measured breast tissue characteristics with mammographic density and other risk factors for breast cancer. Cancer Epidemiol Biomarkers Prev. 2005;14(2):343–9.CrossRefPubMed
23.
Zurück zum Zitat Huo CW, Chew G, Hill P, Huang D, Ingman W, Hodson L, Brown KA, Magenau A, Allam AH, McGhee E, et al. High mammographic density is associated with an increase in stromal collagen and immune cells within the mammary epithelium. Breast Cancer Res. 2015;17:79.CrossRefPubMedPubMedCentral Huo CW, Chew G, Hill P, Huang D, Ingman W, Hodson L, Brown KA, Magenau A, Allam AH, McGhee E, et al. High mammographic density is associated with an increase in stromal collagen and immune cells within the mammary epithelium. Breast Cancer Res. 2015;17:79.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Bland KI, Kuhns JG, Buchanan JB, Dwyer PA, Heuser LF, O'Connor CA, Gray LA Sr, Polk HC Jr. A clinicopathologic correlation of mammographic parenchymal patterns and associated risk factors for human mammary carcinoma. Ann Surg. 1982;195(5):582–94.CrossRefPubMedPubMedCentral Bland KI, Kuhns JG, Buchanan JB, Dwyer PA, Heuser LF, O'Connor CA, Gray LA Sr, Polk HC Jr. A clinicopathologic correlation of mammographic parenchymal patterns and associated risk factors for human mammary carcinoma. Ann Surg. 1982;195(5):582–94.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Ghosh K, Brandt KR, Reynolds C, Scott CG, Pankratz VS, Riehle DL, Lingle WL, Odogwu T, Radisky DC, Visscher DW, et al. Tissue composition of mammographically dense and non-dense breast tissue. Breast Cancer Res Treat. 2012;131(1):267–75.CrossRefPubMed Ghosh K, Brandt KR, Reynolds C, Scott CG, Pankratz VS, Riehle DL, Lingle WL, Odogwu T, Radisky DC, Visscher DW, et al. Tissue composition of mammographically dense and non-dense breast tissue. Breast Cancer Res Treat. 2012;131(1):267–75.CrossRefPubMed
26.
Zurück zum Zitat Urbanski S, Jensen HM, Cooke G, McFarlane D, Shannon P, Kruikov V, Boyd NF. The association of histological and radiological indicators of breast cancer risk. Br J Cancer. 1988;58(4):474–9.CrossRefPubMedPubMedCentral Urbanski S, Jensen HM, Cooke G, McFarlane D, Shannon P, Kruikov V, Boyd NF. The association of histological and radiological indicators of breast cancer risk. Br J Cancer. 1988;58(4):474–9.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Bartow SA, Mettler FA Jr, Black Iii WC. Correlations between radiographic patterns and morphology of the female breast. Rad Patterns Morph. 1997;13:263–75. Bartow SA, Mettler FA Jr, Black Iii WC. Correlations between radiographic patterns and morphology of the female breast. Rad Patterns Morph. 1997;13:263–75.
28.
Zurück zum Zitat Simpson ER, Clyne C, Rubin G, Boon WC, Robertson K, Britt K, Speed C, Jones M. Aromatase--a brief overview. Annu Rev Physiol. 2002;64:93–127.CrossRefPubMed Simpson ER, Clyne C, Rubin G, Boon WC, Robertson K, Britt K, Speed C, Jones M. Aromatase--a brief overview. Annu Rev Physiol. 2002;64:93–127.CrossRefPubMed
29.
Zurück zum Zitat Wellings SR, Jensen HM. On the origin and progression of ductal carcinoma in the human breast. J Natl Cancer Inst. 1973;50(5):1111–8.CrossRefPubMed Wellings SR, Jensen HM. On the origin and progression of ductal carcinoma in the human breast. J Natl Cancer Inst. 1973;50(5):1111–8.CrossRefPubMed
30.
Zurück zum Zitat Wellings SR, Jensen HM, Marcum RG. An atlas of subgross pathology of the human breast with special reference to possible precancerous lesions. J Natl Cancer Inst. 1975;55(2):231–73.PubMed Wellings SR, Jensen HM, Marcum RG. An atlas of subgross pathology of the human breast with special reference to possible precancerous lesions. J Natl Cancer Inst. 1975;55(2):231–73.PubMed
31.
Zurück zum Zitat Renehan AG, Harvie M, Howell A. Insulin-like growth factor (IGF)-I, IGF binding protein-3, and breast cancer risk: eight years on. EndocrRelat Cancer. 2006;13(2):273–8.CrossRef Renehan AG, Harvie M, Howell A. Insulin-like growth factor (IGF)-I, IGF binding protein-3, and breast cancer risk: eight years on. EndocrRelat Cancer. 2006;13(2):273–8.CrossRef
32.
Zurück zum Zitat Tworoger SS, Eliassen AH, Sluss P, Hankinson SE. A prospective study of plasma prolactin concentrations and risk of premenopausal and postmenopausal breast cancer. J Clin Oncol. 2007;25(12):1482–8.CrossRefPubMed Tworoger SS, Eliassen AH, Sluss P, Hankinson SE. A prospective study of plasma prolactin concentrations and risk of premenopausal and postmenopausal breast cancer. J Clin Oncol. 2007;25(12):1482–8.CrossRefPubMed
33.
Zurück zum Zitat Horne HN, Sherman ME, Pfeiffer RM, Figueroa JD, Khodr ZG, Falk RT, Pollak M, Patel DA, Palakal MM, Linville L, et al. Circulating insulin-like growth factor-I, insulin-like growth factor binding protein-3 and terminal duct lobular unit involution of the breast: a cross-sectional study of women with benign breast disease. Breast Cancer Res. 2016;18(1):24.CrossRefPubMedPubMedCentral Horne HN, Sherman ME, Pfeiffer RM, Figueroa JD, Khodr ZG, Falk RT, Pollak M, Patel DA, Palakal MM, Linville L, et al. Circulating insulin-like growth factor-I, insulin-like growth factor binding protein-3 and terminal duct lobular unit involution of the breast: a cross-sectional study of women with benign breast disease. Breast Cancer Res. 2016;18(1):24.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Huh SJ, Oh H, Peterson MA, Almendro V, Hu R, Bowden M, Lis RL, Cotter MB, Loda M, Barry WT, et al. The Proliferative Activity of Mammary Epithelial Cells in Normal Tissue Predicts Breast Cancer Risk in Premenopausal Women. Cancer Res. 2016;76(7):1926–34.CrossRefPubMedPubMedCentral Huh SJ, Oh H, Peterson MA, Almendro V, Hu R, Bowden M, Lis RL, Cotter MB, Loda M, Barry WT, et al. The Proliferative Activity of Mammary Epithelial Cells in Normal Tissue Predicts Breast Cancer Risk in Premenopausal Women. Cancer Res. 2016;76(7):1926–34.CrossRefPubMedPubMedCentral
35.
Zurück zum Zitat Hawes D, Downey S, Pearce CL, Bartow S, Wan P, Pike MC, Wu AH. Dense breast stromal tissue shows greatly increased concentration of breast epithelium but no increase in its proliferative activity. Breast Cancer Res. 2006;8(2):R24.CrossRefPubMedPubMedCentral Hawes D, Downey S, Pearce CL, Bartow S, Wan P, Pike MC, Wu AH. Dense breast stromal tissue shows greatly increased concentration of breast epithelium but no increase in its proliferative activity. Breast Cancer Res. 2006;8(2):R24.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Colditz GA, Frazier LA. Models of breast cancer show that risk is set by events of early life: prevention efforts much shift focus (review). Cancer Epidemiol Biomarkers Prevent. 1995;4(5):567–71. Colditz GA, Frazier LA. Models of breast cancer show that risk is set by events of early life: prevention efforts much shift focus (review). Cancer Epidemiol Biomarkers Prevent. 1995;4(5):567–71.
37.
Zurück zum Zitat Sun X, Glynn DJ, Hodson LJ, Huo C, Britt K, Thompson EW, Woolford L, Evdokiou A, Pollard JW, Robertson SA, et al. CCL2-driven inflammation increases mammary gland stromal density and cancer susceptibility in a transgenic mouse model. Breast Cancer Res. 2017;19(1):4.CrossRefPubMedPubMedCentral Sun X, Glynn DJ, Hodson LJ, Huo C, Britt K, Thompson EW, Woolford L, Evdokiou A, Pollard JW, Robertson SA, et al. CCL2-driven inflammation increases mammary gland stromal density and cancer susceptibility in a transgenic mouse model. Breast Cancer Res. 2017;19(1):4.CrossRefPubMedPubMedCentral
38.
Zurück zum Zitat Provenzano PP, Inman DR, Eliceiri KW, Knittel JG, Yan L, Rueden CT, White JG, Keely PJ. Collagen density promotes mammary tumor initiation and progression. BMC Med. 2008;6(0):11.CrossRefPubMedPubMedCentral Provenzano PP, Inman DR, Eliceiri KW, Knittel JG, Yan L, Rueden CT, White JG, Keely PJ. Collagen density promotes mammary tumor initiation and progression. BMC Med. 2008;6(0):11.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat McConnell JC, O'Connell OV, Brennan K, Weiping L, Howe M, Joseph L, Knight D, O'Cualain R, Lim Y, Leek A, et al. Increased peri-ductal collagen micro-organization may contribute to raised mammographic density. Breast Cancer Res. 2016;18(1):5.CrossRefPubMedPubMedCentral McConnell JC, O'Connell OV, Brennan K, Weiping L, Howe M, Joseph L, Knight D, O'Cualain R, Lim Y, Leek A, et al. Increased peri-ductal collagen micro-organization may contribute to raised mammographic density. Breast Cancer Res. 2016;18(1):5.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Ray A, Slama ZM, Morford RK, Madden SA, Provenzano PP. Enhanced Directional Migration of Cancer Stem Cells in 3D Aligned Collagen Matrices. Biophys J. 2017;112(5):1023–36.CrossRefPubMed Ray A, Slama ZM, Morford RK, Madden SA, Provenzano PP. Enhanced Directional Migration of Cancer Stem Cells in 3D Aligned Collagen Matrices. Biophys J. 2017;112(5):1023–36.CrossRefPubMed
41.
Zurück zum Zitat Kuperwasser C, Chavarria T, Wu M, Magrane G, Gray JW, Carey L, Richardson A, Weinberg RA. Reconstruction of functionally normal and malignant human breast tissues in mice. Proc Natl Acad Sc U S A. 2004;101(14):4966–71.CrossRef Kuperwasser C, Chavarria T, Wu M, Magrane G, Gray JW, Carey L, Richardson A, Weinberg RA. Reconstruction of functionally normal and malignant human breast tissues in mice. Proc Natl Acad Sc U S A. 2004;101(14):4966–71.CrossRef
43.
Zurück zum Zitat Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer. 2016;16(9):582–98.CrossRefPubMed Kalluri R. The biology and function of fibroblasts in cancer. Nat Rev Cancer. 2016;16(9):582–98.CrossRefPubMed
44.
Zurück zum Zitat O'Connor JW, Gomez EW. Biomechanics of TGFbeta-induced epithelial-mesenchymal transition: implications for fibrosis and cancer. Clin Transl Med. 2014;3:23.CrossRefPubMedPubMedCentral O'Connor JW, Gomez EW. Biomechanics of TGFbeta-induced epithelial-mesenchymal transition: implications for fibrosis and cancer. Clin Transl Med. 2014;3:23.CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat DeFilippis RA, Chang H, Dumont N, Rabban JT, Chen YY, Fontenay GV, Berman HK, Gauthier ML, Zhao J, Hu D, et al. CD36 repression activates a multicellular stromal program shared by high mammographic density and tumor tissues. Cancer Discov. 2012;2(9):826–39.CrossRefPubMedPubMedCentral DeFilippis RA, Chang H, Dumont N, Rabban JT, Chen YY, Fontenay GV, Berman HK, Gauthier ML, Zhao J, Hu D, et al. CD36 repression activates a multicellular stromal program shared by high mammographic density and tumor tissues. Cancer Discov. 2012;2(9):826–39.CrossRefPubMedPubMedCentral
47.
Zurück zum Zitat Folgueira MA, Maistro S, Katayama ML, Roela RA, Mundim FG, Nanogaki S, de Bock GH, Brentani MM. Markers of breast cancer stromal fibroblasts in the primary tumour site associated with lymph node metastasis: a systematic review including our case series. Biosci Rep. 2013;33(6) Folgueira MA, Maistro S, Katayama ML, Roela RA, Mundim FG, Nanogaki S, de Bock GH, Brentani MM. Markers of breast cancer stromal fibroblasts in the primary tumour site associated with lymph node metastasis: a systematic review including our case series. Biosci Rep. 2013;33(6)
48.
Zurück zum Zitat Hasebe T, Sasaki S, Imoto S, Mukai K, Yokose T, Ochiai A. Prognostic significance of fibrotic focus in invasive ductal carcinoma of the breast: a prospective observational study. Mod Pathol. 2002;15(5):502–16.CrossRefPubMed Hasebe T, Sasaki S, Imoto S, Mukai K, Yokose T, Ochiai A. Prognostic significance of fibrotic focus in invasive ductal carcinoma of the breast: a prospective observational study. Mod Pathol. 2002;15(5):502–16.CrossRefPubMed
49.
Zurück zum Zitat Mujtaba SS, Ni YB, Tsang JY, Chan SK, Yamaguchi R, Tanaka M, Tan PH, Tse GM. Fibrotic focus in breast carcinomas: relationship with prognostic parameters and biomarkers. Ann Surg Oncol. 2013;20(9):2842–9.CrossRefPubMed Mujtaba SS, Ni YB, Tsang JY, Chan SK, Yamaguchi R, Tanaka M, Tan PH, Tse GM. Fibrotic focus in breast carcinomas: relationship with prognostic parameters and biomarkers. Ann Surg Oncol. 2013;20(9):2842–9.CrossRefPubMed
50.
Zurück zum Zitat Vachon CM, Sasano H, Ghosh K, Brandt KR, Watson DA, Reynolds C, Lingle WL, Goss PE, Li R, Aiyar SE, et al. Aromatase immunoreactivity is increased in mammographically dense regions of the breast. Breast Cancer Res Treat. 2011;125(1):243–52.CrossRefPubMed Vachon CM, Sasano H, Ghosh K, Brandt KR, Watson DA, Reynolds C, Lingle WL, Goss PE, Li R, Aiyar SE, et al. Aromatase immunoreactivity is increased in mammographically dense regions of the breast. Breast Cancer Res Treat. 2011;125(1):243–52.CrossRefPubMed
51.
Zurück zum Zitat Simpson ER, Clyne CD, Rubin G, Boon WC, Robertson K, Britt K, Speed C, Jones M. Aromatase--a brief overview. Annu Rev Physiol. 2002;64(0):93–127.CrossRefPubMed Simpson ER, Clyne CD, Rubin G, Boon WC, Robertson K, Britt K, Speed C, Jones M. Aromatase--a brief overview. Annu Rev Physiol. 2002;64(0):93–127.CrossRefPubMed
52.
Zurück zum Zitat Simpson ER, McInnes KJ, Brown KA, Knower KC, Chand AL, Clyne CD, Simpson ER. Characterisation of aromatase expression in the human adipocyte cell line SGBS. Breast Cancer Res Treat. 2008;112(3):429–35.CrossRefPubMed Simpson ER, McInnes KJ, Brown KA, Knower KC, Chand AL, Clyne CD, Simpson ER. Characterisation of aromatase expression in the human adipocyte cell line SGBS. Breast Cancer Res Treat. 2008;112(3):429–35.CrossRefPubMed
53.
Zurück zum Zitat Bulun SE, Mahendroo MS, Simpson ER. Aromatase gene expression in adipose tissue: relationship to breast cancer. J Steriod Biochem Molec Biol. 1994;49(4-6):319–26.CrossRef Bulun SE, Mahendroo MS, Simpson ER. Aromatase gene expression in adipose tissue: relationship to breast cancer. J Steriod Biochem Molec Biol. 1994;49(4-6):319–26.CrossRef
54.
Zurück zum Zitat Bulun SE, Sharda G, Rink J, Sharma S, Simpson ER. Distribution of aromatase P450 transcripts and adipose fibroblasts in the human breast. J Clin Endocrinol Metabol. 1996;81(3):1273–7. Bulun SE, Sharda G, Rink J, Sharma S, Simpson ER. Distribution of aromatase P450 transcripts and adipose fibroblasts in the human breast. J Clin Endocrinol Metabol. 1996;81(3):1273–7.
55.
Zurück zum Zitat Simpson ER. Biology of aromatase in the mammary gland. J Mammary Gland Biol Neoplasia. 2000;5(3):251–8.CrossRefPubMed Simpson ER. Biology of aromatase in the mammary gland. J Mammary Gland Biol Neoplasia. 2000;5(3):251–8.CrossRefPubMed
56.
Zurück zum Zitat Lisanti MP, Tsirigos A, Pavlides S, Reeves KJ, Peiris-Pages M, Chadwick AL, Sanchez-Alvarez R, Lamb R, Howell A, Martinez-Outschoorn UE, et al. JNK1 stress signaling is hyper-activated in high breast density and the tumor stroma: connecting fibrosis, inflammation, and stemness for cancer prevention. Cell Cycle. 2014;13(4):580–99.CrossRefPubMed Lisanti MP, Tsirigos A, Pavlides S, Reeves KJ, Peiris-Pages M, Chadwick AL, Sanchez-Alvarez R, Lamb R, Howell A, Martinez-Outschoorn UE, et al. JNK1 stress signaling is hyper-activated in high breast density and the tumor stroma: connecting fibrosis, inflammation, and stemness for cancer prevention. Cell Cycle. 2014;13(4):580–99.CrossRefPubMed
58.
Zurück zum Zitat Paszek MJ, Weaver VM. The tension mounts: mechanics meets morphogenesis and malignancy. J Mammary Gland Biol Neoplasia. 2004;9(4):325–42.CrossRefPubMed Paszek MJ, Weaver VM. The tension mounts: mechanics meets morphogenesis and malignancy. J Mammary Gland Biol Neoplasia. 2004;9(4):325–42.CrossRefPubMed
59.
Zurück zum Zitat Alowami S, Troup S, Al-Haddad S, Kirkpatrick I, Watson PH. Mammographic density is related to stroma and stromal proteoglycan expression. Breast Cancer Res. 2003;5(5):R129–35.CrossRefPubMedPubMedCentral Alowami S, Troup S, Al-Haddad S, Kirkpatrick I, Watson PH. Mammographic density is related to stroma and stromal proteoglycan expression. Breast Cancer Res. 2003;5(5):R129–35.CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat Guo YP, Martin LJ, Hanna W, Banerjee D, Miller N, Fishell E, Khokha R, Boyd NF. Growth factors and stromal matrix proteins associated with mammographic densities. Cancer Epidemiol Biomarkers Prev. 2001;10(3):243–8.PubMed Guo YP, Martin LJ, Hanna W, Banerjee D, Miller N, Fishell E, Khokha R, Boyd NF. Growth factors and stromal matrix proteins associated with mammographic densities. Cancer Epidemiol Biomarkers Prev. 2001;10(3):243–8.PubMed
61.
Zurück zum Zitat Hojilla CV, Mohammed FF, Khokha R. Matrix metalloproteinases and their tissue inhibitors direct cell fate during cancer development. Br J Cancer. 2003;89(10):1817–21.CrossRefPubMedPubMedCentral Hojilla CV, Mohammed FF, Khokha R. Matrix metalloproteinases and their tissue inhibitors direct cell fate during cancer development. Br J Cancer. 2003;89(10):1817–21.CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Huo CW, Waltham M, Khoo C, Fox SB, Hill P, Chen S, Chew GL, Price JT, Nguyen CH, Williams ED, et al. Mammographically dense human breast tissue stimulates MCF10DCIS.com progression to invasive lesions and metastasis. Breast Cancer Res. 2016;18(1):106.CrossRefPubMedPubMedCentral Huo CW, Waltham M, Khoo C, Fox SB, Hill P, Chen S, Chew GL, Price JT, Nguyen CH, Williams ED, et al. Mammographically dense human breast tissue stimulates MCF10DCIS.com progression to invasive lesions and metastasis. Breast Cancer Res. 2016;18(1):106.CrossRefPubMedPubMedCentral
63.
Zurück zum Zitat Boyd NF, Dite GS, Stone J, Gunasekara A, English DR, McCredie MRE, Giles GG, Tritchler D, Chiarelli A, Yaffe MJ, et al. Heritability of mammographic density, a risk factor for breast cancer. N Engl J Med. 2002;347(12):886–94.CrossRefPubMed Boyd NF, Dite GS, Stone J, Gunasekara A, English DR, McCredie MRE, Giles GG, Tritchler D, Chiarelli A, Yaffe MJ, et al. Heritability of mammographic density, a risk factor for breast cancer. N Engl J Med. 2002;347(12):886–94.CrossRefPubMed
64.
Zurück zum Zitat Varghese JS, Thompson DJ, Michailidou K, Lindstrom S, Turnbull C, Brown J, Leyland J, Warren RM, Luben RN, Loos RJ, et al. Mammographic breast density and breast cancer: evidence of a shared genetic basis. Cancer Res. 2012;72(6):1478–84.CrossRefPubMedPubMedCentral Varghese JS, Thompson DJ, Michailidou K, Lindstrom S, Turnbull C, Brown J, Leyland J, Warren RM, Luben RN, Loos RJ, et al. Mammographic breast density and breast cancer: evidence of a shared genetic basis. Cancer Res. 2012;72(6):1478–84.CrossRefPubMedPubMedCentral
65.
Zurück zum Zitat Lindstrom S, Thompson DJ, Paterson AD, Li J, Gierach GL, Scott C, Stone J, Douglas JA, dos-Santos-Silva I, Fernandez-Navarro P, et al. Genome-wide association study identifies multiple loci associated with both mammographic density and breast cancer risk. Nat Commun. 2014;5:5303.CrossRefPubMedPubMedCentral Lindstrom S, Thompson DJ, Paterson AD, Li J, Gierach GL, Scott C, Stone J, Douglas JA, dos-Santos-Silva I, Fernandez-Navarro P, et al. Genome-wide association study identifies multiple loci associated with both mammographic density and breast cancer risk. Nat Commun. 2014;5:5303.CrossRefPubMedPubMedCentral
66.
Zurück zum Zitat Lindstrom S, Vachon CM, Li J, Varghese J, Thompson D, Warren R, Brown J, Leyland J, Audley T, Wareham NJ, et al. Common variants in ZNF365 are associated with both mammographic density and breast cancer risk. Nat Genet. 2011;43(3):185–7.CrossRefPubMedPubMedCentral Lindstrom S, Vachon CM, Li J, Varghese J, Thompson D, Warren R, Brown J, Leyland J, Audley T, Wareham NJ, et al. Common variants in ZNF365 are associated with both mammographic density and breast cancer risk. Nat Genet. 2011;43(3):185–7.CrossRefPubMedPubMedCentral
67.
Zurück zum Zitat Musunuru K, Strong A, Frank-Kamenetsky M, Lee NE, Ahfeldt T, Sachs KV, Li X, Li H, Kuperwasser N, Ruda VM, et al. From noncoding variant to phenotype via SORT1 at the 1p13 cholesterol locus. Nature. 2010;466(7307):714–9.CrossRefPubMedPubMedCentral Musunuru K, Strong A, Frank-Kamenetsky M, Lee NE, Ahfeldt T, Sachs KV, Li X, Li H, Kuperwasser N, Ruda VM, et al. From noncoding variant to phenotype via SORT1 at the 1p13 cholesterol locus. Nature. 2010;466(7307):714–9.CrossRefPubMedPubMedCentral
68.
Zurück zum Zitat Smemo S, Tena JJ, Kim KH, Gamazon ER, Sakabe NJ, Gomez-Marin C, Aneas I, Credidio FL, Sobreira DR, Wasserman NF, et al. Obesity-associated variants within FTO form long-range functional connections with IRX3. Nature. 2014;507(7492):371–5.CrossRefPubMedPubMedCentral Smemo S, Tena JJ, Kim KH, Gamazon ER, Sakabe NJ, Gomez-Marin C, Aneas I, Credidio FL, Sobreira DR, Wasserman NF, et al. Obesity-associated variants within FTO form long-range functional connections with IRX3. Nature. 2014;507(7492):371–5.CrossRefPubMedPubMedCentral
69.
Zurück zum Zitat Welter D, MacArthur J, Morales J, Burdett T, Hall P, Junkins H, Klemm A, Flicek P, Manolio T, Hindorff L, et al. The NHGRI GWAS Catalog, a curated resource of SNP-trait associations. Nucleic Acids Res. 2014;42(Database issue):D1001–6.CrossRefPubMed Welter D, MacArthur J, Morales J, Burdett T, Hall P, Junkins H, Klemm A, Flicek P, Manolio T, Hindorff L, et al. The NHGRI GWAS Catalog, a curated resource of SNP-trait associations. Nucleic Acids Res. 2014;42(Database issue):D1001–6.CrossRefPubMed
71.
Zurück zum Zitat Zhou Y, Lee JY, Lee CM, Cho WK, Kang MJ, Koff JL, Yoon PO, Chae J, Park HO, Elias JA, et al. Amphiregulin, an epidermal growth factor receptor ligand, plays an essential role in the pathogenesis of transforming growth factor-beta-induced pulmonary fibrosis. J Biol Chem. 2012;287(50):41991–2000.CrossRefPubMedPubMedCentral Zhou Y, Lee JY, Lee CM, Cho WK, Kang MJ, Koff JL, Yoon PO, Chae J, Park HO, Elias JA, et al. Amphiregulin, an epidermal growth factor receptor ligand, plays an essential role in the pathogenesis of transforming growth factor-beta-induced pulmonary fibrosis. J Biol Chem. 2012;287(50):41991–2000.CrossRefPubMedPubMedCentral
72.
Zurück zum Zitat Herrington DM, Howard TD, Hawkins GA, Reboussin DM, Xu J, Zheng SL, Brosnihan KB, Meyers DA, Bleecker ER. Estrogen-receptor polymorphisms and effects of estrogen replacement on high-density lipoprotein cholesterol in women with coronary disease. N Engl J Med. 2002;346(13):967–74.CrossRefPubMed Herrington DM, Howard TD, Hawkins GA, Reboussin DM, Xu J, Zheng SL, Brosnihan KB, Meyers DA, Bleecker ER. Estrogen-receptor polymorphisms and effects of estrogen replacement on high-density lipoprotein cholesterol in women with coronary disease. N Engl J Med. 2002;346(13):967–74.CrossRefPubMed
73.
Zurück zum Zitat Wiedemann E, Schwartz E, Frantz AG. Acute and chronic estrogen effects upon serum somatomedin activity, growth hormone, and prolactin in man. J Clin Endocrinol Metab. 1976;42(5):942–52.CrossRefPubMed Wiedemann E, Schwartz E, Frantz AG. Acute and chronic estrogen effects upon serum somatomedin activity, growth hormone, and prolactin in man. J Clin Endocrinol Metab. 1976;42(5):942–52.CrossRefPubMed
74.
Zurück zum Zitat Hankinson SE, Willett WC, Michaud DS, Manson JE, Colditz GA, Longcope C, Rosner B, Speizer FE. Plasma prolactin levels and subsequent risk of breast cancer in postmenopausal women. J Natl Cancer Inst. 1999;91(7):629–34.CrossRefPubMed Hankinson SE, Willett WC, Michaud DS, Manson JE, Colditz GA, Longcope C, Rosner B, Speizer FE. Plasma prolactin levels and subsequent risk of breast cancer in postmenopausal women. J Natl Cancer Inst. 1999;91(7):629–34.CrossRefPubMed
75.
Zurück zum Zitat Boyd N, Martin L, Chavez S, Gunasekara A, Salleh A, Melnichouk O, Yaffe M, Friedenreich C, Minkin S, Bronskill M. Breast-tissue composition and other risk factors for breast cancer in young women: a cross-sectional study. Lancet Oncol. 2009;10(6):569–80.CrossRefPubMed Boyd N, Martin L, Chavez S, Gunasekara A, Salleh A, Melnichouk O, Yaffe M, Friedenreich C, Minkin S, Bronskill M. Breast-tissue composition and other risk factors for breast cancer in young women: a cross-sectional study. Lancet Oncol. 2009;10(6):569–80.CrossRefPubMed
76.
Zurück zum Zitat Franco L, Williams FM, Trofimov S, Malkin I, Surdulescu G, Spector T, Livshits G. Assessment of age-related changes in heritability and IGF-1 gene effect on circulating IGF-1 levels. Age (Dordr). 2014;36(3):9622.CrossRef Franco L, Williams FM, Trofimov S, Malkin I, Surdulescu G, Spector T, Livshits G. Assessment of age-related changes in heritability and IGF-1 gene effect on circulating IGF-1 levels. Age (Dordr). 2014;36(3):9622.CrossRef
77.
Zurück zum Zitat Pollak M. The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nat Rev Cancer. 2012;12(3):159–69.CrossRefPubMed Pollak M. The insulin and insulin-like growth factor receptor family in neoplasia: an update. Nat Rev Cancer. 2012;12(3):159–69.CrossRefPubMed
78.
Zurück zum Zitat Zhang B, Shu XO, Delahanty RJ, Zeng C, Michailidou K, Bolla MK, Wang Q, Dennis J, Wen W, Long J, et al. Height and Breast Cancer Risk: Evidence From Prospective Studies and Mendelian Randomization. J Natl Cancer Inst. 2015;107(11) Zhang B, Shu XO, Delahanty RJ, Zeng C, Michailidou K, Bolla MK, Wang Q, Dennis J, Wen W, Long J, et al. Height and Breast Cancer Risk: Evidence From Prospective Studies and Mendelian Randomization. J Natl Cancer Inst. 2015;107(11)
79.
Zurück zum Zitat Boyd NF, Lockwood GA, Byng JW, Little LE, Yaffe MJ, Tritchler DL. The relationship of anthropometric measures to radiological features of the breast in premenopausal women. Br J Cancer. 1998;78(9):1233–8.CrossRefPubMedPubMedCentral Boyd NF, Lockwood GA, Byng JW, Little LE, Yaffe MJ, Tritchler DL. The relationship of anthropometric measures to radiological features of the breast in premenopausal women. Br J Cancer. 1998;78(9):1233–8.CrossRefPubMedPubMedCentral
80.
Zurück zum Zitat Johansson A, Marroni F, Hayward C, Franklin CS, Kirichenko AV, Jonasson I, Hicks AA, Vitart V, Isaacs A, Axenovich T, et al. Linkage and genome-wide association analysis of obesity-related phenotypes: association of weight with the MGAT1 gene. Obesity. 2010;18(4):803–8.CrossRefPubMed Johansson A, Marroni F, Hayward C, Franklin CS, Kirichenko AV, Jonasson I, Hicks AA, Vitart V, Isaacs A, Axenovich T, et al. Linkage and genome-wide association analysis of obesity-related phenotypes: association of weight with the MGAT1 gene. Obesity. 2010;18(4):803–8.CrossRefPubMed
81.
Zurück zum Zitat Okada Y, Kamatani Y, Takahashi A, Matsuda K, Hosono N, Ohmiya H, Daigo Y, Yamamoto K, Kubo M, Nakamura Y, et al. A genome-wide association study in 19 633 Japanese subjects identified LHX3-QSOX2 and IGF1 as adult height loci. Hum mol Genet. 2010;19(11):2303–12.CrossRefPubMed Okada Y, Kamatani Y, Takahashi A, Matsuda K, Hosono N, Ohmiya H, Daigo Y, Yamamoto K, Kubo M, Nakamura Y, et al. A genome-wide association study in 19 633 Japanese subjects identified LHX3-QSOX2 and IGF1 as adult height loci. Hum mol Genet. 2010;19(11):2303–12.CrossRefPubMed
82.
Zurück zum Zitat Sasazuki T, Sawada T, Sakon S, Kitamura T, Kishi T, Okazaki T, Katano M, Tanaka M, Watanabe M, Yagita H, et al. Identification of a novel transcriptional activator, BSAC, by a functional cloning to inhibit tumor necrosis factor-induced cell death. J Biol Chem. 2002;277(32):28853–60.CrossRefPubMed Sasazuki T, Sawada T, Sakon S, Kitamura T, Kishi T, Okazaki T, Katano M, Tanaka M, Watanabe M, Yagita H, et al. Identification of a novel transcriptional activator, BSAC, by a functional cloning to inhibit tumor necrosis factor-induced cell death. J Biol Chem. 2002;277(32):28853–60.CrossRefPubMed
83.
Zurück zum Zitat Hossain M, Qadri SM, Su Y, Liu L. ICAM-1-mediated leukocyte adhesion is critical for the activation of endothelial LSP1. Am J Physiol Cell Physiol. 2013;304(9):C895–904.CrossRefPubMed Hossain M, Qadri SM, Su Y, Liu L. ICAM-1-mediated leukocyte adhesion is critical for the activation of endothelial LSP1. Am J Physiol Cell Physiol. 2013;304(9):C895–904.CrossRefPubMed
84.
Zurück zum Zitat Moolgavkar SH, Day NE, Stevens RG. Two-stage model for carcinogenesis: epidemiology of breast cancer in females. J Natl Cancer Inst. 1980;65(3):559–69.PubMed Moolgavkar SH, Day NE, Stevens RG. Two-stage model for carcinogenesis: epidemiology of breast cancer in females. J Natl Cancer Inst. 1980;65(3):559–69.PubMed
85.
86.
Zurück zum Zitat Boyd NF, Martin LJ, Bronskill M, Yaffe MJ, Duric N, Minkin S. Breast tissue composition and susceptibility to breast cancer. J Natl Cancer Inst. 2010;102(16):1224–37.CrossRefPubMedPubMedCentral Boyd NF, Martin LJ, Bronskill M, Yaffe MJ, Duric N, Minkin S. Breast tissue composition and susceptibility to breast cancer. J Natl Cancer Inst. 2010;102(16):1224–37.CrossRefPubMedPubMedCentral
87.
Zurück zum Zitat Byng JW, Boyd NF, Fishell E, Jong RA, Yaffe MJ. The quantitative analysis of mammographic densities. Phys Med Biol. 1994;39(10):1629–38.CrossRefPubMed Byng JW, Boyd NF, Fishell E, Jong RA, Yaffe MJ. The quantitative analysis of mammographic densities. Phys Med Biol. 1994;39(10):1629–38.CrossRefPubMed
88.
Zurück zum Zitat Graham SJ, Ness S, Hamilton BS, Bronskill MJ. Magnetic resonance properties of ex vivo breast tissue at 1.5 T. Magn Reson Med. 1997;38(4):669–77.CrossRefPubMed Graham SJ, Ness S, Hamilton BS, Bronskill MJ. Magnetic resonance properties of ex vivo breast tissue at 1.5 T. Magn Reson Med. 1997;38(4):669–77.CrossRefPubMed
89.
Zurück zum Zitat Boyd NF, Lockwood GA, Byng JW, Tritchler DL, Yaffe MJ. Mammographic densities and breast cancer risk. Cancer Epidemiol Biomarkers Prev. 1998;7(12):1133–44.PubMed Boyd NF, Lockwood GA, Byng JW, Tritchler DL, Yaffe MJ. Mammographic densities and breast cancer risk. Cancer Epidemiol Biomarkers Prev. 1998;7(12):1133–44.PubMed
90.
Zurück zum Zitat Bernstein L. Epidemiology of endocrine-related risk factors for breast cancer. J Mammary Gland Biol Neoplasia. 2002;7(1):3–15.CrossRefPubMed Bernstein L. Epidemiology of endocrine-related risk factors for breast cancer. J Mammary Gland Biol Neoplasia. 2002;7(1):3–15.CrossRefPubMed
91.
Zurück zum Zitat Rice MS, Bertrand KA, VanderWeele TJ, Rosner BA, Liao X, Adami HO, Tamimi RM. Mammographic density and breast cancer risk: a mediation analysis. Breast Cancer Res. 2016;18(1):94.CrossRefPubMedPubMedCentral Rice MS, Bertrand KA, VanderWeele TJ, Rosner BA, Liao X, Adami HO, Tamimi RM. Mammographic density and breast cancer risk: a mediation analysis. Breast Cancer Res. 2016;18(1):94.CrossRefPubMedPubMedCentral
93.
Zurück zum Zitat Byrne C, Ursin G, Martin CF, Peck JD, Cole EB, Zeng D, Kim E, Yaffe MD, Boyd NF, Heiss G, et al. Mammographic Density Change With Estrogen and Progestin Therapy and Breast Cancer Risk. J Natl Cancer Inst. 2017;109(9) Byrne C, Ursin G, Martin CF, Peck JD, Cole EB, Zeng D, Kim E, Yaffe MD, Boyd NF, Heiss G, et al. Mammographic Density Change With Estrogen and Progestin Therapy and Breast Cancer Risk. J Natl Cancer Inst. 2017;109(9)
94.
Zurück zum Zitat Toriola AT, Dang HX, Hagemann IS, Appleton CM, Colditz GA, Luo J, Maher CA. Increased breast tissue receptor activator of nuclear factor-kappaB ligand (RANKL) gene expression is associated with higher mammographic density in premenopausal women. Oncotarget. 2017;8(43):73787–92.CrossRefPubMedPubMedCentral Toriola AT, Dang HX, Hagemann IS, Appleton CM, Colditz GA, Luo J, Maher CA. Increased breast tissue receptor activator of nuclear factor-kappaB ligand (RANKL) gene expression is associated with higher mammographic density in premenopausal women. Oncotarget. 2017;8(43):73787–92.CrossRefPubMedPubMedCentral
Metadaten
Titel
The origins of breast cancer associated with mammographic density: a testable biological hypothesis
verfasst von
Norman Boyd
Hal Berman
Jie Zhu
Lisa J. Martin
Martin J. Yaffe
Sofia Chavez
Greg Stanisz
Greg Hislop
Anna M. Chiarelli
Salomon Minkin
Andrew D. Paterson
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Breast Cancer Research / Ausgabe 1/2018
Elektronische ISSN: 1465-542X
DOI
https://doi.org/10.1186/s13058-018-0941-y

Weitere Artikel der Ausgabe 1/2018

Breast Cancer Research 1/2018 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.