Skip to main content
Erschienen in: Breast Cancer 4/2012

Open Access 01.10.2012 | Review Article

Reproductive history and breast cancer risk

verfasst von: Shunzo Kobayashi, Hiroshi Sugiura, Yoshiaki Ando, Norio Shiraki, Takeshi Yanagi, Hiroko Yamashita, Tatsuya Toyama

Erschienen in: Breast Cancer | Ausgabe 4/2012

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

The fact that reproductive factors have significant influence on the risk of breast cancer is well known. Early age of first full-term birth is highly protective against late-onset breast cancers, but each pregnancy, including the first one, increases the risk of early-onset breast cancer. Estradiol and progesterone induce receptor activator of NF-kappa B ligand (RANKL) in estrogen receptor (ER)- and progesterone receptor (PgR)-positive luminal cells. RANKL then acts in a paracrine fashion on the membranous RANK of ER/PgR-negative epithelial stem cells of the breast. This reaction cascade is triggered by chorionic gonadotropin during the first trimester of pregnancy and results in the morphological and functional development of breast tissue. On the other hand, the administration of non-steroidal anti-inflammatory drugs in the early steps of weaning protects against tumor growth through reduction of the acute inflammatory reaction of post lactation remodeling of breast tissue. This is experimental evidence that may explain the short-term tumor-promoting effect of pregnancy. The protective effect of prolonged breast feeding may also be explained, at least in a part, by a reduced inflammatory reaction due to gradual weaning. Delay of first birth together with low parity and short duration of breast feeding are increasing social trends in developed countries. Therefore, breast cancer risk as a result of reproductive factors will not decrease in these countries in the foreseeable future. In this review, the significance of reproductive history with regard to the risk of breast cancers will be discussed, focusing on the age of first full-term birth and post lactation involution of the breast.

Introduction

The classification of breast cancers according to the molecular profile of gene expression patterns has improved the fundamental understanding and the rational strategy of research and treatment of patients with breast cancer [1]. Breast cancers have been classified into 5 subtypes: luminal-A, luminal-B, Her2-enriched, basal-like, and claudin-low [2]. The alternative but practical classification using immunohistochemical staining of estrogen receptor (ER), progesterone receptor (PgR), and Her2 protein has been used worldwide [3]. Hormone dependency, per se, is a well-known important characteristic of breast cancers [4]. Therefore, the actual treatment of patients with breast cancer has often been based on the cancer subtype classification [5].
On the other hand, individual patient differences, especially regarding their hormonal environment due to age, menopausal status, pregnancy, and/or lactation, have also been considered mainly from the viewpoint of diagnosis and treatment [6]. However, the fundamental and clinical importance of postpartum involution of breast tissues as a tumor-promoting risk factor has recently received a great deal of attention [7, 8]. Expression of inflammation-related genes, such as COX2 [9], Stat3 [10], and tenascin C [11, 12], has been investigated, and the suppression of the resulting inflammatory reactions with non-steroidal anti-inflammatory drugs (NSAIDs) was reported to protect against tumor invasion and metastasis [13]. Thus, postpartum involution, together with the molecular subtypes may have a significant impact on the prognosis of patients, especially those who are premenopausal.
It is well known that pregnancy and breast feeding have dual effects on breast cancer development [14, 15]. Although early age of first full-term birth is highly protective against late occurrence of hormone-dependent breast cancers [1619], each successive pregnancy of multiparous women has a progressive effect on breast cancers irrespective of hormone dependency [20]. This review will discuss the significance of pregnancy and lactation as risk factors for breast cancer.

Age of first full-term birth

In a worldwide case–control study, MacMahon [14] was the first to report the protective effect of early age of first full-term birth against breast cancer. Compared to nulliparous women, mothers with their first full-term birth before 20 years of age had a 50 % reduced risk of breast cancer. On the other hand, those who had their first baby after age 35 had a 22 % increased risk. These relative risks were comparable across countries. The protective effect of early age of first full-term birth in parous women was similarly observed in other series from the USA [21] and Japan [22]. Except for one study from Japan [23], many reports observed a protective effect of early age of first full-term birth on hormone receptor-positive cancers [16, 18, 19, 24]. A meta-analysis of 9 cohort or case–control studies also revealed a reduced risk among patients with hormone receptor-positive cancers [17]. This protective effect was also observed in lobular carcinomas that are often positive for hormone receptors [25]. In this context, the relative protective effect of young age of first full-term birth has been observed mainly for postmenopausal women [17, 23, 24, 26].
Parity-related functional and morphological changes of breast tissue have been extensively studied by Russo and co-workers. The gene expression signature and differentiation-related chromatin remodeling in the breast tissues of parous women were distinct from those of nulliparous women [27]. Chorionic gonadotropin is secreted mostly during the first trimester of pregnancy and stimulates the ovarian granulosa-lutein cells to form the corpus luteum of pregnancy. The high levels of progesterone from the corpus luteum are critical to regulate the initial stages of the pregnancy, including breast development to prepare for a fully functional organ. Human chorionic gonadotropin (hCG) administration induced differentiation-related gene expression changes in breast epithelial cells in culture [28], and the profiles were similar in pregnancy and after hCG administration [29, 30]. On the other hand, a prolonged suppressive effect on carcinogen-induced mammary tumor formation was observed with the administration of hCG in rats [31]. A similar protective effect of hCG was observed in humans in a case–control study from Sweden [32]. Women over 50 years of age with higher blood levels of hCG during their first full-term pregnancy showed a 33 % reduced risk of breast cancer. A possible decrease in the number of mammary epithelial stem cells after pregnancy was reported in transplantation experiments [33]. Thus, the high progesterone conditions induced by the hCG seem to be essential to pregnancy-associated breast development, presumably as a result of mammary epithelial stem cell expansion.
Progesterone acts after binding to the PgR, which is in turn induced by estrogen action via ER binding. There had been, however, great controversy about the presence or absence of ER and PgR in mammary epithelial stem cells. Chimeric ER-positive and ER-negative cells were demonstrated using a biostaining technique in ER−/− mouse mammary tissue by the transplantation of cells from wild-type mice [34]. Unexpectedly, the colony forming ability that was assumed to be one of the stem cell characteristics was distinctly observed not in the ER/PgR-positive luminal cell population, but in the negative basal cell population [35]. In 2010, the detailed mechanisms of hormone action on the development of mammary tissues during pregnancy were clarified by 2 different groups [36, 37] (Fig. 1). Elevated progesterone, which is induced by the increased hCG from the trophoblast after the first trimester of pregnancy, acts on the ER/PgR-positive luminal cells and induces receptor activator of NF-kappa B ligand (RANKL). RANKL acts in a paracrine fashion on the ER/PgR-negative mammary stem cells that express RANK on their cell membrane and promotes expansion through NF-kappa B activation. This relationship of proliferation between ER/PgR-positive and -negative cells was also observed in hormone-dependent mammary carcinogenesis [38, 39] and metastasis [40]. In the case of breast cancer, RANKL may act as an autocrine fashion, or the RANK–RANKL pathway may act without the intervention of progesterone. However, there is fundamental consensus about the significance of this pathway at least in the case of breast expansion associated with pregnancy.

Post lactation involution and risk of breast cancer

Early in 1980, Woods and co-workers reported an important observation about parity and breast cancer. Among 371 consecutive patients with breast cancer, the mean age of diagnosis of parous women (57.9) was significantly younger than that of nulliparous women (63.1). Additionally, the mean age of diagnosis fell with increasing parity and almost 40 % of women with 3 or more children were diagnosed at less than 50 years of age [15]. This observation suggested the protective effect of parity on late-onset (postmenopausal) breast cancers together with a promoting effect on cancers occurring at younger ages (premenopausal). A transient postpartum increase in breast cancer was also reported in women younger than 50 years old [41], and premenopausal women [42]. A meta-analysis of 8 reports from northern European countries revealed that a higher age of first childbirth correlated with a higher risk of breast cancer between the ages of 35 and 54 [43]. The risk of breast cancer was increased until 15 years post delivery and decreased thereafter. The transient risk increase was more prominent in women with their first full-term birth at age 35 or older [44].
The worse prognosis of breast cancers diagnosed during pregnancy or lactation had been explained as being due to the difficulty of diagnosis and treatment [6, 20, 45, 46]. However, prognosis of those occurring shortly after delivery seemed to be even worse. Survival rates improved with increasing interval between their last birth and diagnosis [47]. The worse prognosis was more significant in patients younger than 35 years old [48]. The dual effect of pregnancy was also confirmed by a cohort study [49]. Very interestingly, women with higher blood hCG levels during the first trimester of their first full-term pregnancy had a higher risk of breast cancer occurring at a young age (age less than 40; low hCG vs. high hCG, 1.00 vs. 1.78) or shortly after delivery (less than 10 years; low hCG vs. high hCG, 1.00 vs. 4.33) [32].
The terminal duct lobular units (TDLUs) of breast tissue decrease in size and number during age-related involution. Histologically demonstrated spontaneous involution was correlated with a reduced risk of late-onset breast cancers [50]. However, since the breast tissues must provide for a possible next pregnancy after weaning, the post lactation involution of the breast is distinct from that of aging. The process begins with a cell death phase and progresses to the tissue remodeling phase [51]. Gene expression profiling revealed the induction of the Stat3-mediated cell death-related pathway at the initial step, followed by an acute phase inflammatory reaction [7, 52]. The involuting breast tissues accumulated fibrillar collagen and stimulated migration and metastasis of inoculated breast cancer cells [8]. The prognostic significance of tumor-associated macrophages has been reported in patients with breast cancer [53]. Accumulated collagen acts as an extracellular matrix mediator to recruit a tumor-promoting subtype of macrophage in the involuting breast tissues [54]. The macrophage and mast cell infiltration was observed at the late phase of involution with a chronic inflammatory reaction [10] which is controlled by TIMP3 via TNF expression [55].
Tenascin C is an extracellular matrix glycoprotein often upregulated in breast cancers [56]. The downregulation of tenascin C acts as a suppressor of the epithelial to mesenchymal transition, which is important in tumor invasion and metastasis [57]. Tenascin C suppresses Stat5 signaling and stimulates the WNT and NOTCH pathways. It is abundantly expressed at the metastatic front facilitating migration and metastasis of the tumor-initiating cells [12]. Significant accumulation of tenascin C together with collagen fiber in the involuting breast has been observed [11]. The administration of NSAIDs suppresses the progression of breast cancer cells through downregulation of tenascin C expression. These specific characteristics of the post lactation involution of breast tissue that make it a risk factor for cancer were reviewed by Lyons and co-workers who suggested the importance of an inflammatory reaction [9]. COX2 expression was stimulated not only breast tissue itself but also in inoculated breast cancer cells [13]. NSAIDs administration suppressed the COX2 expression and decreased the deposition of fibrillar collagen, resulting in a decline of tumor progression and metastasis.
Kreuzaler and co-workers reported a new concept about the Stat3-mediated cell death-related pathway at the initial step of the post lactation involution [58]. Stat3 induced lysosomal protease independently from the caspase-dependent apoptotic pathway and increased lysosomal membrane permeability. The increased lysosomal proteases digest cell membranes and induce cell death without nuclear fragmentation that is typical in apoptosis. They proposed, therefore, a necrosis type of new programmed cell death pathway in this physiological but special tissue remodeling procedure of post lactation involution.

Breast feeding

Additional important factors relating to pregnancy are breast feeding and number of pregnancies. A longer duration of breast feeding is correlated with a lower risk of breast cancer [16, 59]. Although the risk-reducing effect has been observed only for premenopausal women in reports of small series it was also observed for pre- and postmenopausal women in a large series [16] and in a meta-analysis of 47 reports from all over the world [59]. Periodic influence of estrogen/progesterone on breast tissue can be postponed by prolonged breast feeding or increased number of pregnancies. If this hormonal environment of the post delivery period influences the risk of breast cancer, it should be restricted to ER/PgR-positive cancers. However, the protective effect was observed in both ER/PgR-positive and -negative cancers [16]. Instead, longer duration of breast feeding reduced the risk of triple-negative cancers but not of luminal cancers defined by immunohistochemical staining in premenopausal patients [60]. Since data typically define the length of breast feeding as total lifetime duration, it was correlated with the number of pregnancies, but they were not necessarily identical [59]. The significance of the number of pregnancies as a risk-reducing factor has been controversial [59, 61]. However, the data in multiparous women limited to those with more than 3 children [26] or more than 5 children [62] showed significant risk-reducing effects. Interestingly enough, the duration of breast feeding per child was also inversely related to risk of breast cancer [63, 64]. Although it has not yet been adequately studied why the prolonged breast feeding is protective against breast cancer, these findings suggest a close relationship between slow-paced weaning and mitigation of inflammatory reaction during the involution. O’Brien and co-workers observed the coexistence of 2 kinds of lobules in the breast tissue from gradually weaning mothers, suggesting a stepwise turnover of TDLUs. One type was actively lactating and the other was involuting, as indicated by infiltration of CD45-positive acute reactive immune cells and CD68-positive macrophages [54].

Perspectives

Breast cancer incidence had been significantly lower in Japan compared to America and European countries and the rates of ER/PgR-positive breast cancers from Japanese patients were also lower, especially in postmenopausal women [6567]. The average age of first birth has increased continuously during the past 5 decades from 24.7 to 29.7. The age-specific incidence of birth has gradually shifted to a higher value and about 24 % of women who gave birth were older than 35 years of age in 2010 (Vital Statistics Japan: Ministry of Health, Labour and Welfare) (Fig. 2). These trends must have influenced the risk of postmenopausal hormone-dependent tumors later in life as well as the risk of more aggressive tumors found shortly after childbirth. Actually, the nationwide occurrence of these two events in Japan has increased significantly during the past 3 decades in number and rates from 11,000 and 20/100,000 to 54,000 and 80/100,000, respectively [68]. The rates of cancers positive for both receptors have also increased from 50 % to more than 80 % [69].
Russo and co-workers suggested a preventive effect of periodic administration of hCG early in life on late-onset breast cancers [70]. It seems, however, to be extremely difficult to demonstrate efficacy and safety concomitantly while resolving the ethical issues. On the other hand, the administration of NSAIDs from the beginning of weaning to reduce the inflammatory reaction of involution may be applicable to prevent early-onset breast cancers. NSAIDs have been used in a wide variety of conditions and the accompanying side effects and their treatment are well known. Compared to evaluating the use of hCG, evaluation of the use of NSAIDs will require shorter follow-up periods. However, the actual significance of these agents with regard to breast cancer prevention in humans is still theoretical, or at best experimental. Methods other than administration of selective estrogen receptor modulators (SERMs) were not recommended at the present stage of knowledge in a consensus statement about preventive therapy for breast cancer by experts on breast cancer prevention in 2011 [71].
The sequence of events appearing in the lactating breast from pregnancy until weaning affects the risk of breast cancer in two ways (Fig. 3a, b). First, the cell death procedure acts in a suppressive manner. Second, the expansion and tissue remodeling procedure acts in a promotive manner. Regardless of hormone dependency, the inevitable risk of breast cancer as a female mammal become obvious around 40 years of age in humans. Among them the risk of hormone-dependent cancer is influenced largely by a variety of reproductive factors and becomes a major risk at almost 50 years of age and thereafter. On the other hand, since the first full-term birth has the most prominent dual effects on the breast cancer risk, the age when it occurs is particularly important. Early age of menarche increases the risk of ER/PgR-positive cancers [17, 72], and a long interval between menarche and first birth increases the risk by 50 % in ER/PgR-positive cancers [73]. These facts suggest an inevitable accumulation of stem-progenitor cells with unrepaired DNA damage since the initiation of pubertal development. Although these cells would have increased concomitantly during the mammary proliferation and differentiation in the first full-term pregnancy, they will be largely eliminated during the programmed cell death phase of involution after weaning. This can lead to risk reduction of late-onset hormone-dependent tumors. On the other hand, the tumor-promoting environment of the remodeling phase of involution increases the risk of unspecified tumors. The baseline risk of breast cancer is very low during younger ages. The protective effect, therefore, will be emphasized and the progressive effect will be attenuated by early age of first full-term birth. On the other hand, late age of first full-term birth inevitably increases the baseline risk of unspecified breast cancers. Although multiple consecutive pregnancies at a young age protect against late-onset cancers, this pattern in the elderly increases premenopausal cancers [74]. It is, therefore, suggested that the promotive effect is offset by the protective effect in those women with births at older ages.
Factors affecting the risk of breast cancer are diverse and are not limited to reproductive history [75]. However, because of the aging of the entire population and low fertility rates, women will be indispensable to the workforce in developed countries including Japan. Such a working environment inevitably favors the delay of the first birth together with low parity and a short duration of breast feeding. Therefore, breast cancer risk due to the increasing effects of reproductive factors will not decrease in those countries in the foreseeable future. It is thus particularly important to take into account the patient’s specific reproductive history in the diagnosis and treatment of breast cancers.

Open Access

This article is distributed under the terms of the Creative Commons Attribution License which permits any use, distribution, and reproduction in any medium, provided the original author(s) and the source are credited.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 2.0 International License (https://​creativecommons.​org/​licenses/​by/​2.​0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Gynäkologie

Kombi-Abonnement

Mit e.Med Gynäkologie erhalten Sie Zugang zu CME-Fortbildungen der beiden Fachgebiete, den Premium-Inhalten der Fachzeitschriften, inklusive einer gedruckten gynäkologischen oder urologischen Zeitschrift Ihrer Wahl.

Literatur
1.
Zurück zum Zitat Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, et al. Molecular portraits of human breast tumours. Nature. 2000;406(6797):747–52.CrossRefPubMed Perou CM, Sorlie T, Eisen MB, van de Rijn M, Jeffrey SS, Rees CA, et al. Molecular portraits of human breast tumours. Nature. 2000;406(6797):747–52.CrossRefPubMed
2.
Zurück zum Zitat Prat A, Parker JS, Karginova O, Fan C, Livasy C, Herschkowitz JI, et al. Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res. 2010;12(5):R68.CrossRefPubMed Prat A, Parker JS, Karginova O, Fan C, Livasy C, Herschkowitz JI, et al. Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast Cancer Res. 2010;12(5):R68.CrossRefPubMed
3.
Zurück zum Zitat Hammond ME, Hayes DF, Wolff AC, Mangu PB, Temin S. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Oncol Pract. 2010;6(4):195–7.CrossRefPubMed Hammond ME, Hayes DF, Wolff AC, Mangu PB, Temin S. American Society of Clinical Oncology/College of American Pathologists guideline recommendations for immunohistochemical testing of estrogen and progesterone receptors in breast cancer. J Oncol Pract. 2010;6(4):195–7.CrossRefPubMed
4.
Zurück zum Zitat Yamashita H, Yando Y, Nishio M, Zhang Z, Hamaguchi M, Mita K, et al. Immunohistochemical evaluation of hormone receptor status for predicting response to endocrine therapy in metastatic breast cancer. Breast Cancer. 2006;13(1):74–83.CrossRefPubMed Yamashita H, Yando Y, Nishio M, Zhang Z, Hamaguchi M, Mita K, et al. Immunohistochemical evaluation of hormone receptor status for predicting response to endocrine therapy in metastatic breast cancer. Breast Cancer. 2006;13(1):74–83.CrossRefPubMed
5.
Zurück zum Zitat Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thurlimann B, Senn HJ, et al. Strategies for subtypes—dealing with the diversity of breast cancer: highlights of the St. Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2011. Ann Oncol. 2011;22(8):1736–47.CrossRefPubMed Goldhirsch A, Wood WC, Coates AS, Gelber RD, Thurlimann B, Senn HJ, et al. Strategies for subtypes—dealing with the diversity of breast cancer: highlights of the St. Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2011. Ann Oncol. 2011;22(8):1736–47.CrossRefPubMed
6.
Zurück zum Zitat Stensheim H, Moller B, van Dijk T, Fossa SD. Cause-specific survival for women diagnosed with cancer during pregnancy or lactation: a registry-based cohort study. J Clin Oncol. 2009;27(1):45–51.CrossRefPubMed Stensheim H, Moller B, van Dijk T, Fossa SD. Cause-specific survival for women diagnosed with cancer during pregnancy or lactation: a registry-based cohort study. J Clin Oncol. 2009;27(1):45–51.CrossRefPubMed
7.
Zurück zum Zitat Clarkson RW, Wayland MT, Lee J, Freeman T, Watson CJ. Gene expression profiling of mammary gland development reveals putative roles for death receptors and immune mediators in post-lactational regression. Breast Cancer Res. 2004;6(2):R92–109.CrossRefPubMed Clarkson RW, Wayland MT, Lee J, Freeman T, Watson CJ. Gene expression profiling of mammary gland development reveals putative roles for death receptors and immune mediators in post-lactational regression. Breast Cancer Res. 2004;6(2):R92–109.CrossRefPubMed
8.
Zurück zum Zitat McDaniel SM, Rumer KK, Biroc SL, Metz RP, Singh M, Porter W, et al. Remodeling of the mammary microenvironment after lactation promotes breast tumor cell metastasis. Am J Pathol. 2006;168(2):608–20.CrossRefPubMed McDaniel SM, Rumer KK, Biroc SL, Metz RP, Singh M, Porter W, et al. Remodeling of the mammary microenvironment after lactation promotes breast tumor cell metastasis. Am J Pathol. 2006;168(2):608–20.CrossRefPubMed
9.
Zurück zum Zitat Lyons TR, Schedin PJ, Borges VF. Pregnancy and breast cancer: when they collide. J Mammary Gland Biol Neoplasia. 2009;14(2):87–98.CrossRefPubMed Lyons TR, Schedin PJ, Borges VF. Pregnancy and breast cancer: when they collide. J Mammary Gland Biol Neoplasia. 2009;14(2):87–98.CrossRefPubMed
10.
Zurück zum Zitat Hughes K, Wickenden JA, Allen JE, Watson CJ. Conditional deletion of Stat3 in mammary epithelium impairs the acute phase response and modulates immune cell numbers during post-lactational regression. J Pathol. 2012;227(1):106–17. Hughes K, Wickenden JA, Allen JE, Watson CJ. Conditional deletion of Stat3 in mammary epithelium impairs the acute phase response and modulates immune cell numbers during post-lactational regression. J Pathol. 2012;227(1):106–17.
11.
Zurück zum Zitat O’Brien J, Hansen K, Barkan D, Green J, Schedin P. Non-steroidal anti-inflammatory drugs target the pro-tumorigenic extracellular matrix of the postpartum mammary gland. Int J Dev Biol. 2011;55(7–8–9):745–55. O’Brien J, Hansen K, Barkan D, Green J, Schedin P. Non-steroidal anti-inflammatory drugs target the pro-tumorigenic extracellular matrix of the postpartum mammary gland. Int J Dev Biol. 2011;55(7–8–9):745–55.
12.
Zurück zum Zitat Oskarsson T, Acharyya S, Zhang XH, Vanharanta S, Tavazoie SF, Morris PG, et al. Breast cancer cells produce tenascin C as a metastatic niche component to colonize the lungs. Nat Med. 2011;17(7):867–74.CrossRefPubMed Oskarsson T, Acharyya S, Zhang XH, Vanharanta S, Tavazoie SF, Morris PG, et al. Breast cancer cells produce tenascin C as a metastatic niche component to colonize the lungs. Nat Med. 2011;17(7):867–74.CrossRefPubMed
13.
Zurück zum Zitat Lyons TR, O’Brien J, Borges VF, Conklin MW, Keely PJ, Eliceiri KW, et al. Postpartum mammary gland involution drives progression of ductal carcinoma in situ through collagen and COX-2. Nat Med. 2011;17(9):1109–15.CrossRefPubMed Lyons TR, O’Brien J, Borges VF, Conklin MW, Keely PJ, Eliceiri KW, et al. Postpartum mammary gland involution drives progression of ductal carcinoma in situ through collagen and COX-2. Nat Med. 2011;17(9):1109–15.CrossRefPubMed
14.
Zurück zum Zitat MacMahon B, Cole P, Lin TM, Lowe CR, Mirra AP, Ravnihar B, et al. Age at first birth and breast cancer risk. Bull World Health Organ. 1970;43(2):209–21.PubMed MacMahon B, Cole P, Lin TM, Lowe CR, Mirra AP, Ravnihar B, et al. Age at first birth and breast cancer risk. Bull World Health Organ. 1970;43(2):209–21.PubMed
15.
Zurück zum Zitat Woods KL, Smith SR, Morrison JM. Parity and breast cancer: evidence of a dual effect. Br Med J. 1980;281(6237):419–21.CrossRefPubMed Woods KL, Smith SR, Morrison JM. Parity and breast cancer: evidence of a dual effect. Br Med J. 1980;281(6237):419–21.CrossRefPubMed
16.
Zurück zum Zitat Ursin G, Bernstein L, Lord SJ, Karim R, Deapen D, Press MF, et al. Reproductive factors and subtypes of breast cancer defined by hormone receptor and histology. Br J Cancer. 2005;93(3):364–71.CrossRefPubMed Ursin G, Bernstein L, Lord SJ, Karim R, Deapen D, Press MF, et al. Reproductive factors and subtypes of breast cancer defined by hormone receptor and histology. Br J Cancer. 2005;93(3):364–71.CrossRefPubMed
17.
Zurück zum Zitat Ma H, Bernstein L, Pike MC, Ursin G. Reproductive factors and breast cancer risk according to joint estrogen and progesterone receptor status: a meta-analysis of epidemiological studies. Breast Cancer Res. 2006;8(4):R43.CrossRefPubMed Ma H, Bernstein L, Pike MC, Ursin G. Reproductive factors and breast cancer risk according to joint estrogen and progesterone receptor status: a meta-analysis of epidemiological studies. Breast Cancer Res. 2006;8(4):R43.CrossRefPubMed
18.
Zurück zum Zitat Lord SJ, Bernstein L, Johnson KA, Malone KE, McDonald JA, Marchbanks PA, et al. Breast cancer risk and hormone receptor status in older women by parity, age of first birth, and breastfeeding: a case–control study. Cancer Epidemiol Biomarkers Prev. 2008;17(7):1723–30.CrossRefPubMed Lord SJ, Bernstein L, Johnson KA, Malone KE, McDonald JA, Marchbanks PA, et al. Breast cancer risk and hormone receptor status in older women by parity, age of first birth, and breastfeeding: a case–control study. Cancer Epidemiol Biomarkers Prev. 2008;17(7):1723–30.CrossRefPubMed
19.
Zurück zum Zitat Yang XR, Chang-Claude J, Goode EL, Couch FJ, Nevanlinna H, Milne RL, et al. Associations of breast cancer risk factors with tumor subtypes: a pooled analysis from the Breast Cancer Association Consortium studies. J Natl Cancer Inst. 2011;103(3):250–63.CrossRefPubMed Yang XR, Chang-Claude J, Goode EL, Couch FJ, Nevanlinna H, Milne RL, et al. Associations of breast cancer risk factors with tumor subtypes: a pooled analysis from the Breast Cancer Association Consortium studies. J Natl Cancer Inst. 2011;103(3):250–63.CrossRefPubMed
20.
Zurück zum Zitat Bladstrom A, Anderson H, Olsson H. Worse survival in breast cancer among women with recent childbirth: results from a Swedish population-based register study. Clin Breast Cancer. 2003;4(4):280–5.CrossRefPubMed Bladstrom A, Anderson H, Olsson H. Worse survival in breast cancer among women with recent childbirth: results from a Swedish population-based register study. Clin Breast Cancer. 2003;4(4):280–5.CrossRefPubMed
21.
Zurück zum Zitat Lee SH, Akuete K, Fulton J, Chelmow D, Chung MA, Cady B. An increased risk of breast cancer after delayed first parity. Am J Surg. 2003;186(4):409–12.CrossRefPubMed Lee SH, Akuete K, Fulton J, Chelmow D, Chung MA, Cady B. An increased risk of breast cancer after delayed first parity. Am J Surg. 2003;186(4):409–12.CrossRefPubMed
22.
Zurück zum Zitat Nagata C, Hu YH, Shimizu H. Effects of menstrual and reproductive factors on the risk of breast cancer: meta-analysis of the case–control studies in Japan. Jpn J Cancer Res. 1995;86(10):910–5.CrossRefPubMed Nagata C, Hu YH, Shimizu H. Effects of menstrual and reproductive factors on the risk of breast cancer: meta-analysis of the case–control studies in Japan. Jpn J Cancer Res. 1995;86(10):910–5.CrossRefPubMed
23.
Zurück zum Zitat Iwasaki M, Otani T, Inoue M, Sasazuki S, Tsugane S, Japan Public Health Center-based Prospective Study Group. Role and impact of menstrual and reproductive factors on breast cancer risk in Japan. Eur J Cancer Prev. 2007;16(2):116–23.CrossRefPubMed Iwasaki M, Otani T, Inoue M, Sasazuki S, Tsugane S, Japan Public Health Center-based Prospective Study Group. Role and impact of menstrual and reproductive factors on breast cancer risk in Japan. Eur J Cancer Prev. 2007;16(2):116–23.CrossRefPubMed
24.
Zurück zum Zitat Li CI, Littman AJ, White E. Relationship between age maximum height is attained, age at menarche, and age at first full-term birth and breast cancer risk. Cancer Epidemiol Biomarkers Prev. 2007;16(10):2144–9.CrossRefPubMed Li CI, Littman AJ, White E. Relationship between age maximum height is attained, age at menarche, and age at first full-term birth and breast cancer risk. Cancer Epidemiol Biomarkers Prev. 2007;16(10):2144–9.CrossRefPubMed
25.
Zurück zum Zitat Newcomb PA, Trentham-Dietz A, Hampton JM, Egan KM, Titus-Ernstoff L, Warren Andersen S, et al. Late age at first full term birth is strongly associated with lobular breast cancer. Cancer. 2011;117(9):1946–56.CrossRefPubMed Newcomb PA, Trentham-Dietz A, Hampton JM, Egan KM, Titus-Ernstoff L, Warren Andersen S, et al. Late age at first full term birth is strongly associated with lobular breast cancer. Cancer. 2011;117(9):1946–56.CrossRefPubMed
26.
Zurück zum Zitat Tamakoshi K, Yatsuya H, Wakai K, Suzuki S, Nishio K, Lin Y, et al. Impact of menstrual and reproductive factors on breast cancer risk in Japan: results of the JACC study. Cancer Sci. 2005;96(1):57–62.CrossRefPubMed Tamakoshi K, Yatsuya H, Wakai K, Suzuki S, Nishio K, Lin Y, et al. Impact of menstrual and reproductive factors on breast cancer risk in Japan: results of the JACC study. Cancer Sci. 2005;96(1):57–62.CrossRefPubMed
27.
Zurück zum Zitat Asztalos S, Gann PH, Hayes MK, Nonn L, Beam CA, Dai Y, et al. Gene expression patterns in the human breast after pregnancy. Cancer Prev Res (Phila). 2010;3(3):301–11.CrossRef Asztalos S, Gann PH, Hayes MK, Nonn L, Beam CA, Dai Y, et al. Gene expression patterns in the human breast after pregnancy. Cancer Prev Res (Phila). 2010;3(3):301–11.CrossRef
28.
Zurück zum Zitat Guo S, Russo IH, Lareef MH, Russo J. Effect of human chorionic gonadotropin in the gene expression profile of MCF-7 cells. Int J Oncol. 2004;24(2):399–407.PubMed Guo S, Russo IH, Lareef MH, Russo J. Effect of human chorionic gonadotropin in the gene expression profile of MCF-7 cells. Int J Oncol. 2004;24(2):399–407.PubMed
29.
Zurück zum Zitat Russo J, Mailo D, Hu YF, Balogh G, Sheriff F, Russo IH. Breast differentiation and its implication in cancer prevention. Clin Cancer Res. 2005;11(2 Pt 2):931s–6s.PubMed Russo J, Mailo D, Hu YF, Balogh G, Sheriff F, Russo IH. Breast differentiation and its implication in cancer prevention. Clin Cancer Res. 2005;11(2 Pt 2):931s–6s.PubMed
30.
Zurück zum Zitat Misra Y, Bentley PA, Bond JP, Tighe S, Hunter T, Zhao FQ. Mammary gland morphological and gene expression changes underlying pregnancy protection of breast cancer tumorigenesis. Physiol Genomics 2012;44(1):76–88 Misra Y, Bentley PA, Bond JP, Tighe S, Hunter T, Zhao FQ. Mammary gland morphological and gene expression changes underlying pregnancy protection of breast cancer tumorigenesis. Physiol Genomics 2012;44(1):76–88
31.
Zurück zum Zitat Russo IH, Koszalka M, Russo J. Comparative study of the influence of pregnancy and hormonal treatment on mammary carcinogenesis. Br J Cancer. 1991;64(3):481–4.CrossRefPubMed Russo IH, Koszalka M, Russo J. Comparative study of the influence of pregnancy and hormonal treatment on mammary carcinogenesis. Br J Cancer. 1991;64(3):481–4.CrossRefPubMed
32.
Zurück zum Zitat Toniolo P, Grankvist K, Wulff M, Chen T, Johansson R, Schock H, et al. Human chorionic gonadotropin in pregnancy and maternal risk of breast cancer. Cancer Res. 2010;70(17):6779–86.CrossRefPubMed Toniolo P, Grankvist K, Wulff M, Chen T, Johansson R, Schock H, et al. Human chorionic gonadotropin in pregnancy and maternal risk of breast cancer. Cancer Res. 2010;70(17):6779–86.CrossRefPubMed
33.
Zurück zum Zitat Siwko SK, Dong J, Lewis MT, Liu H, Hilsenbeck SG, Li Y. Evidence that an early pregnancy causes a persistent decrease in the number of functional mammary epithelial stem cells—implications for pregnancy-induced protection against breast cancer. Stem Cells. 2008;26(12):3205–9.CrossRefPubMed Siwko SK, Dong J, Lewis MT, Liu H, Hilsenbeck SG, Li Y. Evidence that an early pregnancy causes a persistent decrease in the number of functional mammary epithelial stem cells—implications for pregnancy-induced protection against breast cancer. Stem Cells. 2008;26(12):3205–9.CrossRefPubMed
34.
Zurück zum Zitat Mallepell S, Krust A, Chambon P, Brisken C. Paracrine signaling through the epithelial estrogen receptor alpha is required for proliferation and morphogenesis in the mammary gland. Proc Natl Acad Sci U S A. 2006;103(7):2196–201.CrossRefPubMed Mallepell S, Krust A, Chambon P, Brisken C. Paracrine signaling through the epithelial estrogen receptor alpha is required for proliferation and morphogenesis in the mammary gland. Proc Natl Acad Sci U S A. 2006;103(7):2196–201.CrossRefPubMed
35.
Zurück zum Zitat Sleeman KE, Kendrick H, Robertson D, Isacke CM, Ashworth A, Smalley MJ. Dissociation of estrogen receptor expression and in vivo stem cell activity in the mammary gland. J Cell Biol. 2007;176(1):19–26.CrossRefPubMed Sleeman KE, Kendrick H, Robertson D, Isacke CM, Ashworth A, Smalley MJ. Dissociation of estrogen receptor expression and in vivo stem cell activity in the mammary gland. J Cell Biol. 2007;176(1):19–26.CrossRefPubMed
36.
Zurück zum Zitat Asselin-Labat ML, Vaillant F, Sheridan JM, Pal B, Wu D, Simpson ER, et al. Control of mammary stem cell function by steroid hormone signalling. Nature. 2010;465(7299):798–802.CrossRefPubMed Asselin-Labat ML, Vaillant F, Sheridan JM, Pal B, Wu D, Simpson ER, et al. Control of mammary stem cell function by steroid hormone signalling. Nature. 2010;465(7299):798–802.CrossRefPubMed
37.
Zurück zum Zitat Joshi PA, Jackson HW, Beristain AG, Di Grappa MA, Mote PA, Clarke CL, et al. Progesterone induces adult mammary stem cell expansion. Nature. 2010;465(7299):803–7.CrossRefPubMed Joshi PA, Jackson HW, Beristain AG, Di Grappa MA, Mote PA, Clarke CL, et al. Progesterone induces adult mammary stem cell expansion. Nature. 2010;465(7299):803–7.CrossRefPubMed
38.
Zurück zum Zitat Schramek D, Leibbrandt A, Sigl V, Kenner L, Pospisilik JA, Lee HJ, et al. Osteoclast differentiation factor RANKL controls development of progestin-driven mammary cancer. Nature. 2010;468(7320):98–102.CrossRefPubMed Schramek D, Leibbrandt A, Sigl V, Kenner L, Pospisilik JA, Lee HJ, et al. Osteoclast differentiation factor RANKL controls development of progestin-driven mammary cancer. Nature. 2010;468(7320):98–102.CrossRefPubMed
39.
Zurück zum Zitat Gonzalez-Suarez E, Jacob AP, Jones J, Miller R, Roudier-Meyer MP, Erwert R, et al. RANK ligand mediates progestin-induced mammary epithelial proliferation and carcinogenesis. Nature. 2010;468(7320):103–7.CrossRefPubMed Gonzalez-Suarez E, Jacob AP, Jones J, Miller R, Roudier-Meyer MP, Erwert R, et al. RANK ligand mediates progestin-induced mammary epithelial proliferation and carcinogenesis. Nature. 2010;468(7320):103–7.CrossRefPubMed
40.
Zurück zum Zitat Tan W, Zhang W, Strasner A, Grivennikov S, Cheng JQ, Hoffman RM, et al. Tumour-infiltrating regulatory T cells stimulate mammary cancer metastasis through RANKL–RANK signalling. Nature. 2011;470(7335):548–53.CrossRefPubMed Tan W, Zhang W, Strasner A, Grivennikov S, Cheng JQ, Hoffman RM, et al. Tumour-infiltrating regulatory T cells stimulate mammary cancer metastasis through RANKL–RANK signalling. Nature. 2011;470(7335):548–53.CrossRefPubMed
41.
Zurück zum Zitat Bruzzi P, Negri E, La Vecchia C, Decarli A, Palli D, Parazzini F, et al. Short term increase in risk of breast cancer after full term pregnancy. BMJ. 1988;297(6656):1096–8.CrossRefPubMed Bruzzi P, Negri E, La Vecchia C, Decarli A, Palli D, Parazzini F, et al. Short term increase in risk of breast cancer after full term pregnancy. BMJ. 1988;297(6656):1096–8.CrossRefPubMed
42.
Zurück zum Zitat Layde PM, Webster LA, Baughman AL, Wingo PA, Rubin GL, Ory HW. The independent associations of parity, age at first full term pregnancy, and duration of breastfeeding with the risk of breast cancer. Cancer and Steroid Hormone Study Group. J Clin Epidemiol. 1989;42(10):963–73.CrossRefPubMed Layde PM, Webster LA, Baughman AL, Wingo PA, Rubin GL, Ory HW. The independent associations of parity, age at first full term pregnancy, and duration of breastfeeding with the risk of breast cancer. Cancer and Steroid Hormone Study Group. J Clin Epidemiol. 1989;42(10):963–73.CrossRefPubMed
43.
Zurück zum Zitat Ewertz M, Duffy SW, Adami HO, Kvale G, Lund E, Meirik O, et al. Age at first birth, parity and risk of breast cancer: a meta-analysis of 8 studies from the Nordic countries. Int J Cancer. 1990;46(4):597–603.CrossRefPubMed Ewertz M, Duffy SW, Adami HO, Kvale G, Lund E, Meirik O, et al. Age at first birth, parity and risk of breast cancer: a meta-analysis of 8 studies from the Nordic countries. Int J Cancer. 1990;46(4):597–603.CrossRefPubMed
44.
Zurück zum Zitat Lambe M, Hsieh C, Trichopoulos D, Ekbom A, Pavia M, Adami HO. Transient increase in the risk of breast cancer after giving birth. N Engl J Med. 1994;331(1):5–9.CrossRefPubMed Lambe M, Hsieh C, Trichopoulos D, Ekbom A, Pavia M, Adami HO. Transient increase in the risk of breast cancer after giving birth. N Engl J Med. 1994;331(1):5–9.CrossRefPubMed
45.
Zurück zum Zitat Albrektsen G, Heuch I, Thoresen S, Kvale G. Clinical stage of breast cancer by parity, age at birth, and time since birth: a progressive effect of pregnancy hormones? Cancer Epidemiol Biomarkers Prev. 2006;15(1):65–9.CrossRefPubMed Albrektsen G, Heuch I, Thoresen S, Kvale G. Clinical stage of breast cancer by parity, age at birth, and time since birth: a progressive effect of pregnancy hormones? Cancer Epidemiol Biomarkers Prev. 2006;15(1):65–9.CrossRefPubMed
46.
Zurück zum Zitat Mathelin C, Annane K, Treisser A, Chenard MP, Tomasetto C, Bellocq JP, et al. Pregnancy and post-partum breast cancer: a prospective study. Anticancer Res. 2008;28(4C):2447–52. Mathelin C, Annane K, Treisser A, Chenard MP, Tomasetto C, Bellocq JP, et al. Pregnancy and post-partum breast cancer: a prospective study. Anticancer Res. 2008;28(4C):2447–52.
47.
Zurück zum Zitat Whiteman MK, Hillis SD, Curtis KM, McDonald JA, Wingo PA, Marchbanks PA. Reproductive history and mortality after breast cancer diagnosis. Obstet Gynecol. 2004;104(1):146–54.CrossRefPubMed Whiteman MK, Hillis SD, Curtis KM, McDonald JA, Wingo PA, Marchbanks PA. Reproductive history and mortality after breast cancer diagnosis. Obstet Gynecol. 2004;104(1):146–54.CrossRefPubMed
48.
Zurück zum Zitat Largent JA, Ziogas A, Anton-Culver H. Effect of reproductive factors on stage, grade and hormone receptor status in early-onset breast cancer. Breast Cancer Res. 2005;7(4):R541–54.CrossRefPubMed Largent JA, Ziogas A, Anton-Culver H. Effect of reproductive factors on stage, grade and hormone receptor status in early-onset breast cancer. Breast Cancer Res. 2005;7(4):R541–54.CrossRefPubMed
49.
Zurück zum Zitat Albrektsen G, Heuch I, Hansen S, Kvale G. Breast cancer risk by age at birth, time since birth and time intervals between births: exploring interaction effects. Br J Cancer. 2005;92(1):167–75.CrossRefPubMed Albrektsen G, Heuch I, Hansen S, Kvale G. Breast cancer risk by age at birth, time since birth and time intervals between births: exploring interaction effects. Br J Cancer. 2005;92(1):167–75.CrossRefPubMed
50.
Zurück zum Zitat Milanese TR, Hartmann LC, Sellers TA, Frost MH, Vierkant RA, Maloney SD, et al. Age-related lobular involution and risk of breast cancer. J Natl Cancer Inst. 2006;98(22):1600–7.CrossRefPubMed Milanese TR, Hartmann LC, Sellers TA, Frost MH, Vierkant RA, Maloney SD, et al. Age-related lobular involution and risk of breast cancer. J Natl Cancer Inst. 2006;98(22):1600–7.CrossRefPubMed
51.
Zurück zum Zitat Watson CJ. Involution: apoptosis and tissue remodelling that convert the mammary gland from milk factory to a quiescent organ. Breast Cancer Res. 2006;8(2):203.CrossRefPubMed Watson CJ. Involution: apoptosis and tissue remodelling that convert the mammary gland from milk factory to a quiescent organ. Breast Cancer Res. 2006;8(2):203.CrossRefPubMed
52.
Zurück zum Zitat Stein T, Morris JS, Davies CR, Weber-Hall SJ, Duffy MA, Heath VJ, et al. Involution of the mouse mammary gland is associated with an immune cascade and an acute-phase response, involving LBP, CD14 and STAT3. Breast Cancer Res. 2004;6(2):R75–91.CrossRefPubMed Stein T, Morris JS, Davies CR, Weber-Hall SJ, Duffy MA, Heath VJ, et al. Involution of the mouse mammary gland is associated with an immune cascade and an acute-phase response, involving LBP, CD14 and STAT3. Breast Cancer Res. 2004;6(2):R75–91.CrossRefPubMed
53.
Zurück zum Zitat Ueno T, Toi M, Saji H, Muta M, Bando H, Kuroi K, et al. Significance of macrophage chemoattractant protein-1 in macrophage recruitment, angiogenesis, and survival in human breast cancer. Clin Cancer Res. 2000;6(8):3282–9.PubMed Ueno T, Toi M, Saji H, Muta M, Bando H, Kuroi K, et al. Significance of macrophage chemoattractant protein-1 in macrophage recruitment, angiogenesis, and survival in human breast cancer. Clin Cancer Res. 2000;6(8):3282–9.PubMed
54.
Zurück zum Zitat O’Brien J, Lyons T, Monks J, Lucia MS, Wilson RS, Hines L, et al. Alternatively activated macrophages and collagen remodeling characterize the postpartum involuting mammary gland across species. Am J Pathol. 2010;176(3):1241–55.CrossRefPubMed O’Brien J, Lyons T, Monks J, Lucia MS, Wilson RS, Hines L, et al. Alternatively activated macrophages and collagen remodeling characterize the postpartum involuting mammary gland across species. Am J Pathol. 2010;176(3):1241–55.CrossRefPubMed
55.
Zurück zum Zitat Hojilla CV, Jackson HW, Khokha R. TIMP3 regulates mammary epithelial apoptosis with immune cell recruitment through differential TNF dependence. PLoS One. 2011;6(10):e26718.CrossRefPubMed Hojilla CV, Jackson HW, Khokha R. TIMP3 regulates mammary epithelial apoptosis with immune cell recruitment through differential TNF dependence. PLoS One. 2011;6(10):e26718.CrossRefPubMed
56.
Zurück zum Zitat Hancox RA, Allen MD, Holliday DL, Edwards DR, Pennington CJ, Guttery DS, et al. Tumour-associated tenascin-C isoforms promote breast cancer cell invasion and growth by matrix metalloproteinase-dependent and independent mechanisms. Breast Cancer Res. 2009;11(2):R24.CrossRefPubMed Hancox RA, Allen MD, Holliday DL, Edwards DR, Pennington CJ, Guttery DS, et al. Tumour-associated tenascin-C isoforms promote breast cancer cell invasion and growth by matrix metalloproteinase-dependent and independent mechanisms. Breast Cancer Res. 2009;11(2):R24.CrossRefPubMed
57.
Zurück zum Zitat Nagaharu K, Zhang X, Yoshida T, Katoh D, Hanamura N, Kozuka Y, et al. Tenascin C induces epithelial-mesenchymal transition-like change accompanied by SRC activation and focal adhesion kinase phosphorylation in human breast cancer cells. Am J Pathol. 2011;178(2):754–63.CrossRefPubMed Nagaharu K, Zhang X, Yoshida T, Katoh D, Hanamura N, Kozuka Y, et al. Tenascin C induces epithelial-mesenchymal transition-like change accompanied by SRC activation and focal adhesion kinase phosphorylation in human breast cancer cells. Am J Pathol. 2011;178(2):754–63.CrossRefPubMed
58.
Zurück zum Zitat Kreuzaler PA, Staniszewska AD, Li W, Omidvar N, Kedjouar B, Turkson J, et al. Stat3 controls lysosomal-mediated cell death in vivo. Nat Cell Biol. 2011;13(3):303–9.CrossRefPubMed Kreuzaler PA, Staniszewska AD, Li W, Omidvar N, Kedjouar B, Turkson J, et al. Stat3 controls lysosomal-mediated cell death in vivo. Nat Cell Biol. 2011;13(3):303–9.CrossRefPubMed
59.
Zurück zum Zitat Collaborative Group on Hormonal Factors in Breast C. Breast cancer and breastfeeding: collaborative reanalysis of individual data from 47 epidemiological studies in 30 countries, including 50302 women with breast cancer and 96973 women without the disease. Lancet. 2002;360(9328):187–95.CrossRef Collaborative Group on Hormonal Factors in Breast C. Breast cancer and breastfeeding: collaborative reanalysis of individual data from 47 epidemiological studies in 30 countries, including 50302 women with breast cancer and 96973 women without the disease. Lancet. 2002;360(9328):187–95.CrossRef
60.
Zurück zum Zitat Gaudet MM, Press MF, Haile RW, Lynch CF, Glaser SL, Schildkraut J, et al. Risk factors by molecular subtypes of breast cancer across a population-based study of women 56 years or younger. Breast Cancer Res Treat. 2011;130(2):587–97.CrossRefPubMed Gaudet MM, Press MF, Haile RW, Lynch CF, Glaser SL, Schildkraut J, et al. Risk factors by molecular subtypes of breast cancer across a population-based study of women 56 years or younger. Breast Cancer Res Treat. 2011;130(2):587–97.CrossRefPubMed
61.
Zurück zum Zitat Alsaker MD, Opdahl S, Asvold BO, Romundstad PR, Vatten LJ. The association of reproductive factors and breastfeeding with long term survival from breast cancer. Breast Cancer Res Treat. 2011;130(1):175–82.CrossRefPubMed Alsaker MD, Opdahl S, Asvold BO, Romundstad PR, Vatten LJ. The association of reproductive factors and breastfeeding with long term survival from breast cancer. Breast Cancer Res Treat. 2011;130(1):175–82.CrossRefPubMed
62.
Zurück zum Zitat Warren Andersen S, Newcomb PA, Hampton JM, Titus-Ernstoff L, Egan KM, Trentham-Dietz A. Reproductive factors and histologic subtype in relation to mortality after a breast cancer diagnosis. Breast Cancer Res Treat. 2011;130(3):975–80. Warren Andersen S, Newcomb PA, Hampton JM, Titus-Ernstoff L, Egan KM, Trentham-Dietz A. Reproductive factors and histologic subtype in relation to mortality after a breast cancer diagnosis. Breast Cancer Res Treat. 2011;130(3):975–80.
63.
Zurück zum Zitat Chang-Claude J, Eby N, Kiechle M, Bastert G, Becher H. Breastfeeding and breast cancer risk by age 50 among women in Germany. Cancer Causes Control. 2000;11(8):687–95.CrossRefPubMed Chang-Claude J, Eby N, Kiechle M, Bastert G, Becher H. Breastfeeding and breast cancer risk by age 50 among women in Germany. Cancer Causes Control. 2000;11(8):687–95.CrossRefPubMed
64.
Zurück zum Zitat De Silva M, Senarath U, Gunatilake M, Lokuhetty D. Prolonged breastfeeding reduces risk of breast cancer in Sri Lankan women: a case–control study. Cancer Epidemiol. 2010;34(3):267–73.CrossRefPubMed De Silva M, Senarath U, Gunatilake M, Lokuhetty D. Prolonged breastfeeding reduces risk of breast cancer in Sri Lankan women: a case–control study. Cancer Epidemiol. 2010;34(3):267–73.CrossRefPubMed
65.
Zurück zum Zitat Nomura Y, Kobayashi S, Takatani O, Sugano H, Matsumoto K, McGuire WL. Estrogen receptor and endocrine responsiveness in Japanese versus American breast cancer patients. Cancer Res. 1977;37(1):106–10.PubMed Nomura Y, Kobayashi S, Takatani O, Sugano H, Matsumoto K, McGuire WL. Estrogen receptor and endocrine responsiveness in Japanese versus American breast cancer patients. Cancer Res. 1977;37(1):106–10.PubMed
66.
Zurück zum Zitat Nagai R, Kataoka M, Kobayashi S, Ishihara K, Tobioka N, Nakashima K, et al. Estrogen and progesterone receptors in human breast cancer with concomitant assay of plasma 17beta-estradiol, progesterone, and prolactin levels. Cancer Res. 1979;39(5):1834–40.PubMed Nagai R, Kataoka M, Kobayashi S, Ishihara K, Tobioka N, Nakashima K, et al. Estrogen and progesterone receptors in human breast cancer with concomitant assay of plasma 17beta-estradiol, progesterone, and prolactin levels. Cancer Res. 1979;39(5):1834–40.PubMed
67.
Zurück zum Zitat McGuire WL. Steroid receptors in human breast cancer. Cancer Res. 1978;38(11 Pt 2):4289–91.PubMed McGuire WL. Steroid receptors in human breast cancer. Cancer Res. 1978;38(11 Pt 2):4289–91.PubMed
68.
Zurück zum Zitat Matsuda T, Marugame T, Kamo KI, Katanoda K, Ajiki W, Sobue T, et al. Cancer incidence and incidence rates in Japan in 2006: based on data from 15 population-based cancer registries in the monitoring of cancer incidence in Japan (MCIJ) project. Jpn J Clin Oncol. 2012;42(2):139–47.CrossRefPubMed Matsuda T, Marugame T, Kamo KI, Katanoda K, Ajiki W, Sobue T, et al. Cancer incidence and incidence rates in Japan in 2006: based on data from 15 population-based cancer registries in the monitoring of cancer incidence in Japan (MCIJ) project. Jpn J Clin Oncol. 2012;42(2):139–47.CrossRefPubMed
69.
Zurück zum Zitat Yamashita H, Iwase H, Toyama T, Takahashi S, Sugiura H, Yoshimoto N, et al. Estrogen receptor-positive breast cancer in Japanese women: trends in incidence, characteristics, and prognosis. Ann Oncol. 2011;22(6):1318–25.CrossRefPubMed Yamashita H, Iwase H, Toyama T, Takahashi S, Sugiura H, Yoshimoto N, et al. Estrogen receptor-positive breast cancer in Japanese women: trends in incidence, characteristics, and prognosis. Ann Oncol. 2011;22(6):1318–25.CrossRefPubMed
70.
Zurück zum Zitat Kocdor H, Kocdor MA, Russo J, Snider KE, Vanegas JE, Russo IH, et al. Human chorionic gonadotropin (hCG) prevents the transformed phenotypes induced by 17 beta-estradiol in human breast epithelial cells. Cell Biol Int. 2009;33(11):1135–43.CrossRefPubMed Kocdor H, Kocdor MA, Russo J, Snider KE, Vanegas JE, Russo IH, et al. Human chorionic gonadotropin (hCG) prevents the transformed phenotypes induced by 17 beta-estradiol in human breast epithelial cells. Cell Biol Int. 2009;33(11):1135–43.CrossRefPubMed
71.
Zurück zum Zitat Cuzick J, DeCensi A, Arun B, Brown PH, Castiglione M, Dunn B, et al. Preventive therapy for breast cancer: a consensus statement. Lancet Oncol. 2011;12(5):496–503.CrossRefPubMed Cuzick J, DeCensi A, Arun B, Brown PH, Castiglione M, Dunn B, et al. Preventive therapy for breast cancer: a consensus statement. Lancet Oncol. 2011;12(5):496–503.CrossRefPubMed
72.
Zurück zum Zitat Islam T, Matsuo K, Ito H, Hosono S, Watanabe M, Iwata H, et al. Reproductive and hormonal risk factors for luminal, HER2-overexpressing, and triple-negative breast cancer in Japanese women. Ann Oncol. 2012. doi:10.1093/annonc/mdr613 Islam T, Matsuo K, Ito H, Hosono S, Watanabe M, Iwata H, et al. Reproductive and hormonal risk factors for luminal, HER2-overexpressing, and triple-negative breast cancer in Japanese women. Ann Oncol. 2012. doi:10.​1093/​annonc/​mdr613
73.
Zurück zum Zitat Li CI, Malone KE, Daling JR, Potter JD, Bernstein L, Marchbanks PA, et al. Timing of menarche and first full-term birth in relation to breast cancer risk. Am J Epidemiol. 2008;167(2):230–9.CrossRefPubMed Li CI, Malone KE, Daling JR, Potter JD, Bernstein L, Marchbanks PA, et al. Timing of menarche and first full-term birth in relation to breast cancer risk. Am J Epidemiol. 2008;167(2):230–9.CrossRefPubMed
74.
Zurück zum Zitat Kauppila A, Kyyronen P, Lehtinen M, Pukkala E. Dual effect of short interval between first and second birth on ductal breast cancer risk in Finland. Cancer Causes Control. 2012;23(1):187–93.CrossRefPubMed Kauppila A, Kyyronen P, Lehtinen M, Pukkala E. Dual effect of short interval between first and second birth on ductal breast cancer risk in Finland. Cancer Causes Control. 2012;23(1):187–93.CrossRefPubMed
75.
Zurück zum Zitat Yoshimoto N, Nishiyama T, Toyama T, Takahashi S, Shiraki N, Sugiura H, et al. Genetic and environmental predictors, endogenous hormones and growth factors, and risk of estrogen receptor-positive breast cancer in Japanese women. Cancer Sci. 2011;102(11):2065–72.CrossRefPubMed Yoshimoto N, Nishiyama T, Toyama T, Takahashi S, Shiraki N, Sugiura H, et al. Genetic and environmental predictors, endogenous hormones and growth factors, and risk of estrogen receptor-positive breast cancer in Japanese women. Cancer Sci. 2011;102(11):2065–72.CrossRefPubMed
Metadaten
Titel
Reproductive history and breast cancer risk
verfasst von
Shunzo Kobayashi
Hiroshi Sugiura
Yoshiaki Ando
Norio Shiraki
Takeshi Yanagi
Hiroko Yamashita
Tatsuya Toyama
Publikationsdatum
01.10.2012
Verlag
Springer Japan
Erschienen in
Breast Cancer / Ausgabe 4/2012
Print ISSN: 1340-6868
Elektronische ISSN: 1880-4233
DOI
https://doi.org/10.1007/s12282-012-0384-8

Weitere Artikel der Ausgabe 4/2012

Breast Cancer 4/2012 Zur Ausgabe

Update Onkologie

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