My Homepage

Environmental exposures and mammary gland development: state of the science, public health implications, and research recommendations.

OBJECTIVES: Perturbations in mammary gland (MG) development may

increase risk for later adverse effects, including lactation impairment,

gynecomastia (in males), and breast cancer. Animal studies indicate that

exposure to hormonally active agents leads to this type of developmental

effect and related later life susceptibilities. In this review we

describe current science, public health issues, and research

recommendations for evaluating MG development.

DATA SOURCES: The Mammary Gland Evaluation and Risk Assessment

Workshop was convened in Oakland, California, USA, 16--17 November 2009,

to integrate the expertise and perspectives of scientists, risk

assessors, and public health advocates. Interviews were conducted with

18 experts, and seven laboratories conducted an MG slide evaluation

exercise. Workshop participants discussed effects of gestational and

early life exposures to hormonally active agents on MG development, the

relationship of these developmental effects to lactation and cancer, the

relative sensitivity of MG and other developmental end points, the

relevance of animal models to humans, and methods for evaluating MG

effects.



SYNTHESIS: Normal MG development and MG carcinogenesis demonstrate

temporal, morphological, and mechanistic similarities among test animal

species and humans. Diverse chemicals, including many not considered

primarily estrogenic, alter MG development in rodents. Inconsistent

reporting methods hinder comparison across studies, and relationships

between altered development and effects on lactation or carcinogenesis

are still being defined. In some studies, altered MG development is the

most sensitive endocrine end point.



CONCLUSIONS: Early life environmental exposures can alter MG

development, disrupt lactation, and increase susceptibility to breast

cancer. Assessment of MG development should be incorporated in chemical

test guidelines and risk assessment.

KEY WORDS: breast cancer, carcinogen susceptibility, development,

endocrine disruptors, human health, lactation, mammary gland, risk

assessment, rodent model, whole mount. Environ Health Perspect

119:1053-1061 (2011). doi:10.1289/ehp. 1002864 [Online 22 June 2011]



The trend toward earlier breast development initiation in U.S.

girls (Euling et al. 2008) may put them at increased risk of later life

outcomes such as breast cancer--already the most common cancer in U.S.

women (American Cancer Society 2010) and a leading cause of death for

U.S. women in midlife (Brody et al. 2007). Although many factors, such

as nutritional status and body size, may contribute to maturation trends

(Kaplowitz 2008) and breast cancer (Renehan et ai. 2008), environmental

chemicals have been hypothesized to contribute as well (Birnbaum and

Fenton 2003; Brody et al. 2007; Euling et al. 2008). Animal studies

demonstrate that early life exposure to hormonally active agents can

lead to effects on mammary gland (MG) development, impaired lactation,

and increased susceptibility to cancer (Fenton 2006). However, the

influence of environmental exposures on breast development outcomes is

poorly understood, as is the relationship between breast development,

lactational deficits, and breast cancer. Few chemicals coming into the

marketplace are evaluated for these effects. The findings in animal

studies raise concerns that perturbations to human breast development

may increase the risk for later life adverse effects including lactation

impairment, gynecomastia (in males), and breast cancer in either sex.



This review is the result of the Mammary-Gland Evaluation and Risk

Assessment Workshop held in Oakland, California, USA, on 16-17 November

2009 to review and discuss recent findings of altered MG development

after gestational/perinatal exposure to certain endocrine-disrupting

chemicals (EDCs). Workshop participants included research scientists



from multiple disciplines, public health advocates, and risk assessors.

Many of the participants are leading internationally recognized MG

experts.



The goal of the workshop was to improve assessment of MG

developmental end points and their integration into human health risk

assessment. Workshop participants discussed current research on the

effects of developmental exposures to EDCs on MG development, the

relationship of these effects to later life lactation and cancer

outcomes, relative sensitivity of MG development and other developmental

reproductive end points, relevance of effects in animal models to

humans, and MG assessment in current toxicology protocols. Data gaps and

research recommendations were identified at the workshop and through

interviews with 18 risk assessors and toxicologists. Presentation slides

and other workshop materials are available online (Silent Spring

Institute 2011). In conjunction with the workshop, experts from seven

laboratories in Canada, the United States, and Argentina participated in

a round-robin evaluation of MG whole mounts.



Factors Affecting Mammary Gland Development

The female MG undergoes most of its development postnatally,

achieving a fully differentiated state late in pregnancy. This process

includes numerous events that can be disrupted by exposure to EDCs.

Gestation, puberty, and pregnancy are the critical periods during which

EDC exposure may most affect MG development (Fenton 2006). Critical

events include mammary bud development in the fetus, exponential

epithelial outgrowth during puberty, and the rapid transition to

lactational competency that occurs during late pregnancy (Figure 1).

These stages occur in both rodents and humans.



Normal MG development. Normal female MG development involves a

well-orchestrated sequence of events marked by extensive proliferation

at puberty and by proliferation and differentiation during pregnancy.

This process is regulated by hormones, growth factors, and stromal factors and is similar between rodents and humans, although rodent MG

development is more completely described (Kleinberg et al. 2009; Medina

2005). Female human MG development begins with budding and branching

between 6 and 20 weeks of gestation, yielding at birth a primitive gland

composed of ducts ending in ductules. During childhood, MG growth keeps

pace with overall body growth; at puberty it accelerates dramatically.



In rodents, the epithelial bud is formed at the site of the nipple

around gestation days (GDs) 12-16, and by birth the epithelium has

entered the fat pad and formed a ductal tree. The fat pad and mammary

epithelium grow at the same pace as the body for the first 2-3 weeks of

life, and just before puberty, an exponential growth phase begins. In

rodents, and presumably in humans, this phase of ductal development is

characterized by formation of terminal end buds (TEBs), which lead the

epithelial extension through the fat pad, leaving behind a network of

branched ducts. After the fat pad is filled, TEBs differentiate into

terminal ductal structures, namely, terminal ductal lobular units in

humans, lobules and alveolar buds in rats, and terminal ducts in mice.

In humans and rodents, additional MG proliferation and regression events

occur with each luteal phase of the ovulatory cycle, and at pregnancy

there is significant differentiation of the terminal structures with

lobularalveolar development (Kleinberg et al. 2009; Russo and Russo

2004a, 2004b). Mammary epithelial growth also occurs in male rats and

men, whereas male mice lack mammary epithelium. Male mice and rats do

not normally possess nippies because androgens during gestation induce

regression. Retained nipples in male rats is a characteristic effect of

prenatal antiandrogen exposure (Foley et al. 2001).



Assessment of altered MG development. Whole mounts and other

techniques. Early life treatment with some hormonally active agents

results in altered development of the MG in male and female rodents.

Although laboratories vary in their methods for reporting altered MG

development, the primary approach has been morphological assessment of

the entire fourth or fifth abdominal MG fat pad mounted flat on a slide,

fixed, stained, defatted, and permanently affixed to the slide as a

"whole mount." Whole mounts allow an assessment of total and

relative abundance of mammary terminal ductal structures (i.e., TEBs,

terminal ducts, alveolar buds, and lobules), extension of the epithelial

cells through the fat pad, and branching patterns and density at

different times during development (e.g., Fenton et al. 2002; see also

Supplemental Material, Table 1 (doi: 10.1289/ehp. 1002864)]. A common

measurement in mammary whole mounts is the number of TEBs. A TEB is a

teardrop-shaped duct end with a diameter of about 100 um in the rat

compared with about 70 um for a terminal duct (Russo and Russo 1978).



Several rodent studies have reported altered MG development after

prenatal, neonatal, or peripubertal exposure to a range of hormonally

active agents, including pharmaceutical hormones, dietary constituents,

and EDCs [see Supplemental Material, Table 1 (doi: 10.1289/ehp.

1002864)]. These studies typically include histopathological evaluation

of MG whole mounts of developing animals and report morphological

features such as branching, extent of growth, and relative proportion of

structures (e.g., TEBs, lobules, and terminal ducts). Other studies

report changes in morphology or immunohistochemistry of tissue sections

or gene expression in tissue homogenates. Methods and data reporting

vary between laboratories, making it difficult to compare findings

across studies. More uniform approaches will facilitate progress;

however, unanticipated end points should continue to be reported because

this field is still developing.



Morphological changes reflect timing of assessment. Because normal

development involves a well-characterized, consistent progression of

types and ratios of terminal structures and extension through the fat

pad, alterations are sometimes reported as accelerated or delayed

development relative to controls (e.g., Moon et al. 2007). Some agents

alter the pace at which differentiation occurs, leading to an increased

or decreased number of TEBs depending on timing of assessment. If a

perinatally administered EDC causes accelerated development, the number

of TEBs in the treated group will be higher than that in vehicle-treated

controls at weaning [postnatal day (PND) 21] because of increased

proliferation, but lower in early adulthood (PNDs 45-50) because of

accelerated differentiation, as is seen after exposure to estrogens (Hovey et al. 2005). In the case of an EDC, such as dioxin, that causes

delayed development, reduced differentiation leads to a higher number of

TEBs in early adulthood and a longer period during which TEBs are

present (Brown et al. 1998; Fenton et al. 2002). The number of TEBs

present in the gland also depends on the number of ducts in the gland.

Therefore, the number of TEBs at a particular time point can be altered

by changes to the extent of growth as well as to the pace of

differentiation. For example, if the overall number of ducts is

decreased by an environmental exposure, then the overall number of TEBs

in the gland will be decreased compared with those in controls at any

time point, as demonstrated for perfluorooctanoic acid (PFOA) exposures

in mice (White et al. 2009). Some reports have not differentiated

between changes in TEB number due to overall gland size and those due to

altered developmental pace. Evaluation at multiple time points and

consideration of the total number of terminal ends, as well as the

absolute number of TEBs, alleviate this problem and convey the relative

number of structures.



Steroid hormones. In one of the first studies of neonatal exposure

to estrogen, progesterone, or both in mice, Jones and Bern (1979)

reported irreversible adult MG effects, including secretory stimulation,

dilated ducts, and abnormal lobuloalveolar development. Perinatal

treatment with estrogens such as estradiol or diethylstilbestrol (DES)

has been reported to produce accelerated development, characterized by

increased pubertal TEB density, and to promote ductal proliferation

during the peripubertal period in both rats and mice (Fielden et al.

2002; Hilakivi-Clarke et al. 1997; Hovey et al. 2005; Tomooka and Bern

1982; Warner 1976). In addition, Doherty et al. (2010) reported that

prenatal DES exposure in mice altered expression in MG of genes that may

be important in tumorigencsis. Ovariectomy has been reported to diminish

or obviate the effect of neonatal ovarian steroids on mouse MG

development (Jones and Bern 1979; Mori et al. 1976), and strain

differences in sensitivity have also been reported (Mori et al. 1976;

Yang et al. 2009). In rats exposed continuously beginning at conception,

oral ethinyl estradiol exposure induced ductal hyperplasia in male rat

MGs by PND50, and this effect was less apparent in rats assessed later

in life (Latendresse et al. 2009). Thus, morphological changes in MG

reflect timing of exposure as well as timing of assessment, and so both

of these variables must be considered when comparing results across

studies. Supplemental Material, Table 1 (doi: 10.1289/ehp. 1002864)

compiles the methods and findings of studies that have evaluated the

effects of hormone, dietary, or chemical exposures during the prenatal,

neonatal, or peripubertal periods on MG development up to 10 weeks.

Several additional endocrine-sensitive end points commonly assessed to

indicate relative sensitivity are also included in the table.



In addition, whereas perinatal steroid hormone exposure alters

proliferation and TEB number, peripubertal exposure that occurs after

proliferation has begun affects mainly the differentiation of TEBs into

mature structures. For example, pubertal DES treatment in rats increased

the pace of lobule formation and decreased the number of terminal ducts

and TEBs compared with vehicle-treated controls just after puberty (Odum

et al. 1999). Prepubertal DES treatment of rats (on PNDs 23-29) resulted

in fewer TEBs, terminal ducts, and alveolar buds, with a concomitant

increase in the more differentiated lobules, overall suggesting a faster

differentiation pace (Brown and I.amartiniere 1995). Treatment of

postpubcrtal rodents with steroids or human chorionic gonadotropin increases differentiation of the MG in a manner thought to mimic

development during pregnancy (Russo and Russo 2004b; Sivaraman et al.

1998).



Phytoestrogens. Effects of treatment with phytoestrogens such as

genistein are similar to those observed after estrogen reccpror agonist

exposure; perinatal exposure can lead to increased proliferation, and

peripubertal exposure can lead to accelerated differentiation (reviewed

by Warri et al. 2008). For example, Hilakivi-Clarke et al. (1998) and

Padilla-Banks et al. (2006) showed increased TEBs after perinatal

genistein treatment, and Cotroneo et al. (2002) showed accelerated

development in MG after prepubertal exposure, as indicated by increased

TEBs and ductal branching at an early time point, compared with

untreated animals. After gestational and lactational genistein exposure,

You et al. (2002) observed enhanced glandular differentiation at

weaning, and males were more sensitive to the effect than females. Male

rats in a multigenerational genistein feeding study also showed ductal

hyperplasia at PND50, a surprisingly early life stage for these effects

(Latendresse et al. 2009). Effects on MG development have also been

observed after perinatal exposure to other phytoestrogens, including

zearalanone and resveratrol, and to flaxseed [see Supplemental Material,

Table 1 (doi: 10.1289/ehp. 1002864)].



Environmental chemicals. Altered MG development after perinatal

exposure has also been observed for numerous EDCs, including atrazine,

bisphenol A (BPA), dibutylphthalate, dioxin, methoxychlor, nonylphenol,

poly-brominated diphenyl ethers, and PFOA. Changes include delayed MG

development, ductal hyperplasia, alveolar hypoplasia, reduced apoptosis

in TEBs, altered gene or protein expression, increased or decreased

numbers of terminal ducts or lobules, and accelerated alveolar

differentiation [see Supplemental Material, Table 1 (doi: 10.1289/ehp.

1002864)], as well as increased MG tumors after carcinogen challenge

(Brown et al. 1998; Durando et al. 2007; Jenkins et al. 2007). In

addition, late-gestational treatment with Ziracin, a candidate

antibacterial drug, induced hypoplasia (ducts without any acinar development) in rats (Poulet et al. 2005).



Critical exposure windows and reversibility. Studies of the

ubiquitous industrial pollutant dioxin and the high-use herbicide

atrazine have investigated critical periods of exposure associated with

MG effects. Atrazine delayed MG development when exposure occurred

around GD17-19 but had less of an effect after earlier 3-day windows,

and dioxin exposure at GDI5, but not after GDI9, led toMG

underdevelopment (Fenton et al. 2002; Rayner et al. 2005). More recent

studies on the industrial surfactant PFOA (White et al. 2009)

demonstrate a similar critical period. The heightened sensitivity during

this time period is attributed to the formation of the mammary bud and

initial branching that occurs during late pregnancy.



As discussed above, exposure timing and dose influence the pattern

of MG changes (Warri et al. 2008).



Although numerous studies have shown persistent effects on the MG,

few have evaluated whether the changes could be reversible. For example,

in utero exposure to dioxin, Zlfacin, PFOA, or BPA led to permanent

changes in the adult MG (Fenton et al. 2002; Poulet et al. 2005;

Vandenberg et al. 2007; White et al. 2009). In contrast, effects of

genistein and ethinyl estradiol in male MG appeared to reverse after

treatment withdrawal (Latendresse et al. 2009). It is unclear whether

the persistence of alterations reflects the biological half-life and

lipophilicity of the chemical or epigenctic changes, and this may differ

by compound.



Mechanisms. It is striking that MG developmental changes have been

observed after exposure to diverse agents, including estrogens,

androgens, antiandrogens, thyroid-active chemicals, and aryl hydrocarbon

receptor agonists. Data do not indicate a similar mode of action for

atrazine, but PFOA, bro-minated diphenyl ethers, and dioxin all have

been shown to induce a phenotypically similar response of delayed MG

development after neonatal exposures [see Supplemental Material, Table 1

(doi: 10.1289/ehp. 1002864)]. Novel mechanisms continue to be

discovered. In a recent study, Doherty et al. (2010) found that in utero

exposure of mice to DES or BPA increased protein expression and

functional activity' of the histone methyltransferase enhancer of

zeste homolog 2 (EZH2) in the MG. EZH2 has been linked to breast cancer

risk and epigenctic regulation of tumorigenesis. Its up-regulation is a

potential mechanism through which in utero exposure to these chemicals

may produce epigenetic changes leading to increased breast cancer risk

(Doherty et al. 2010).



Sex differences. The few studies that have evaluated effects on

male MG have indicated that male rats could be more sensitive. For

example, one study found altered MG in males, but not females, treated

with methoxycior during gestation (You et al. 2002), and MG effects of

genistein and ethinyl estradiol have been reported in males at lower

doses than in females (Delclos et al. 2001; Latendresse et al. 2009).

Study of sex differences in responsiveness can provide information about

mechanisms of action for the test agents. Although male mice lack

mammary epithelia, there are transgenic mouse models in which mammary

epithelial growth can be induced in males (Li et al. 2002). Mouse models

are needed to study some chemicals, such as PFOA, whose pharmacokinetics

in mice and humans are most similar.





Table 1. Female MG outcomes after developmental environmental

exposures: rodent-human concordance for selected agents.



Environmental Human study Animal study

factor MG outcomes MG outcomes

Development Lactation Cancer Development

risk



Hormonal [DELTA] [DELTA] [DELTA] [DELTA]

milieu:

dosing

(animals) or

surrogates

(humans)



DES [DELTA](c) [DELTA]

Genistein / [DELTA] [DELTA] A

soy



DDT/DDE [DELTA](c) [DELTA] [DELTA]

Dioxins / [DELTA] [DELTA](d) [DELTA]

furans



Environmental

factor

Lactation Cancer

susceptibility



Hormonal [DELTA] [DELTA]

milieu: (EE (2))-dams)(a)

dosing --

(animals) or (EE (2))-offspring(b)

surrogates

(humans)



DES [DELTA] (Dams) [DELTA]

Genistein / [DELTA] (Dams)

soy

[DELTA] (Offspring) [DELTA]



DDT/DDE -(Dams)

Dioxins / [DELTA] (Dams) [DELTA]

furans



Abbreviations:--, no effect on this end point; A, at least one study

has reported an association between the exposure and altered outcomes

[see details and citations in Supplemental Material, Table 2 (doi:

10.1289/ehp. 1002864)]; DDE, dichlorodiphenyldichloroethylene; DDT,

dichlorodiphenyltrichloroethane; DMBA, dimethylbenzanethrene;

EE (2), ethinyl-estradiol. Examples of concordance between

rodents and humans for MG effects are included here. In some cases,

findings are mixed or conflicting; in human studies, exposure

measures are often imprecise. (a)Lactation effect in animals dosed

continuously or during pregnancy and/or lactation. (b)Lactation

effect in animals dosed only in utero and/or preweaning.

(c)Conflicting findings.[d]Exposure may not have been during early

life/development.

The Organisation for Economic Co-operation and Development (OECD)

guidelines for subchronic oral toxicity testing (OECD 2008) include

evaluation of the male, but not female, MG as an optional end point. In

some studies using these guidelines, the male MG appears to be among the

most sensitive end points evaluated (Okazaki et al. 2001), and at least

one such study has found it to be the most sensitive end point in males

(Andrews et al. 2002).



Consequences of Altered Mammary Gland Development



Developmental exposures to certain EDCs can lead to MG

developmental effects, lactational deficits, or cancer, but little is

known about the relationships between the developmental and adult end

points. The morphological changes in MG development, particularly

effects on TEBs, suggest the potential for functional outcomes such as

lactational insufficiency, altered pubertal timing, preneoplasia, or

increased susceptibility to carcinogens (Fenton 2006). Table 1 and

Supplemental Material, Table 2 (doi: 10.1289/ehp. 1002864) show these

types of effects occurring across rodent and human studies for selected

compounds for which there are data. However, there are data gaps

regarding the relationships among the various MG outcomes because a)

only a handful of chemicals have been studied; b) there has not been a

standard procedure to assess MG developmental changes; c) MG assessment

in multigcnerational studies has been limited; and d) few studies

include full assessment of dose response.



Carcinogenesis. Reported changes in patterns of breast development

in U.S. girls (reviewed by Euling et al. 2008) raise concerns about

whether earlier onset of breast development is associated with breast

cancer or other adult diseases, because earlier menarche is an

established risk factor for breast cancer (Kelsey et al. 1993).

Furthermore, studies in humans and rodent models demonstrate that

hormonal factors that affect MG development also influence

susceptibility to carcinogens.



Hormonal factors alter susceptibility to carcinogens. Ovarian,

pituitary, and placental hormones, which vary by life stage and with

pregnancy events, are important determinants of breast cancer

susceptibility in humans and rodents (Russo and Russo 2004b). In both

mice and rats treated with chemical carcinogens, hormone withdrawal

(ovariectomy) inhibits tumor development, whereas hormone

supplementation increases the incidence of adenocarcinoma. In humans,

removal of ovaries by 35 years of age dramatically reduces breast cancer

risk (Eisen et al. 2005; Trichopoulos et al. 1972), and antiestrogens

are effective in breast cancer treatment and chemoprevention (Vogel et

al. 2010).



Susceptibility to carcinogens depends on life stage. The influence

of life stage on susceptibility to carcinogen exposure has been

demonstrated in rats and humans. For example, ionizing radiation is

maximally potent as a human breast carcinogen when exposure occurs

during childhood or adolescence (Henderson et al. 2010; Land 1995); this

observation is consistent with findings in rodents (Imaoka et al. 2009).

The increased tumor response from carcinogen exposure early in life is

attributed to the ptesence of proliferating and undifferentiated

structures such as TEBs, which are present during the pubertal mammary

epithelial expansion and display elevated DNA synthesis compared with

other MG structures (Kleinberg et al. 2009). TEBs are considered the

most vulnerable MG target structure for carcinogen exposure (Medina

2007; Russo and Russo 1996). In animals and humans, tumor response from

carcinogen exposure is highest when exposure occurs during adolescence,

when TEBs are still abundant (Henderson et al. 2010; Imaoka et al. 2009;

Land 1995; Russo and Russo 2004b). As a result, there is concern that

exposures to xenobiotics that increase the number or longevity of

proliferating TEBs might increase susceptibility to breast cancer

(Birnbaum and Fenton 2003; Fenton 2006). After the pubertal growth spurt and throughout adult life, it is the terminal ductal structures that

give rise to breast cancers (Kleinberg et al. 2009; Medina 2007; Russo

and Russo 2004b). During pregnancy, differentiation of terminal

structures increases, and this differentiation has been hypothesized to

account for lower MG sensitivity to carcinogens post-pregnancy (Russo

and Russo 2004b).



Early life exposures to (noncarcinogenic) chemicals may affect

response to carcinogens in later life. Experimental models involving

carcinogen challenge have been used widely to demonstrate that hormones

and growth factors influence MG development, differentiation, and

carcinogenesis; these models could readily be extended to evaluate

increased cancer risk from early life environmental exposures. These

models have been used, for example, to investigate potential

chemopreventive agents that accelerate MG differentiation (e.g., by

mimicking pregnancy hormones) and decrease tumor susceptibility

(Cotroneo et al. 2002; Kleinberg et al. 2009; Russo and Russo 1996).

Rodent models used in these studies include dimethylbenz [a] anthracene (DMBA) and nitrosomethylurea (NMU) challenge in rats and mice, and the

mouse mammary tumor virus (MMTV) model. More recently, genetically

modified mouse models have been used to study mammary tumors that are

comparable with human breast tumors in their latency, histotypes, and

endocrine responsiveness (Cardiff et al. 2000; Kamiya et al. 1995;

Medina 2007; Russo and Russo 2004b; Thompson and Singh 2000).



In rodents, early life exposure to hormon-ally active agents

affects MG tumor formation in carcinogen-challenge models. For example,

neonatal estrogen (or androgen) treatment of mice (MMTV model) or rats

(DMBA model) induced MG developmental changes and increased tumors

(Lopez et al. 1988; Mori et al. 1976, 1979). In addition, early life

exposures to genistein (Hilakivi-Clarke et al. 1998, 1999), alcohol

(Hilakivi-Clarke et al. 2004), dioxin (Brown et al. 1998; Desaulniers et

al. 2001; Jenkins et al. 2007), and oral BPA (Jenkins et al. 2009)

caused increased MG tumor multiplicity and decreased latency after DMBA

challenge at PND50. These effects were accompanied by altered MG

development observed in whole mounts (genistein, alcohol, dioxin) or

altered protein expression (BPA). Lifetime exposures (beginning

pre-natally) to genistein, ethinyl estradiol, and BPA have been reported

to alter MG development and increase incidence of preneoplastic lesions

in the MG, with a stronger effect in early adulthood than at 2 years of

age, when MG histopathology is typically performed (Latendresse et al.

2009; Murray et al. 2007;



Vandenberg et al. 2008). Short-term genistein treatment during the

peripubertal period reduces MG tumors after carcinogen challenge,

whereas perinatal or lifetime exposure seems to increase them, although

studies are not consistent (reviewed by Warri et al. 2008). Similarly,

both gestational (Brown et al. 1998) and prepubertal (Desaulniers et al.

2001) dioxin exposure caused increased MG tumors after carcinogen

challenge, whereas later life exposure decreased spontaneous MG tumors

(Kocibaetal. 1978).



In humans, maternal factors that affect the fetal hormone

environment also appear to affect later breast cancer risk in daughters,

possibly by imprinting the developing MG, thereby altering future tissue

responsiveness to hormonal stimulation (e.g., altering estrogen receptor levels or sensitivity) or to genotoxic insult (e.g., by increasing cell

proliferation or diminishing differentiation). The hypothesis that in

utero endocrine-related factors influence breast cancer risk of a

daughter is supported by epidemiology studies that have found d)

preeclampsia associated with reduced breast cancer risk in offspring and

b) high birth weight correlated with higher breast cancer risk (Hoover

and Troisi 2001; Troisi et al. 2007; Xue and Michels 2007). In addition,

there is some evidence that in utero exposure to DES is associated with

higher breast cancer risk in women [Palmer et al. (2006); however,

Verloop et al. (2010) did not find an association] and with increased MG

tumor incidence in rats (Rothschild et al. 1987). Furthermore, the

single epidemiologic study of the EDC dichlorodiphenyltrichloroethane (DDT) that used prospective measures of adolescent/young adult exposure

in relation to breast cancer risk (Cohn et al. 2007) found significant

associations, whereas many studies in which DDT or its metabolite

dichlorodiphenyldichloro-ethylene (DDE) were measured in older women did

not observe an association with breast cancer.



Pregnancy is another critical window corresponding to a time of

extensive MG proliferation and differentiation. DES exposure in pregnant

women has been associated with increased breast cancer risk in the

mother as well as her daughter (Tirus-Ernstoff et al. 2001). A study of

DMBA-challenged rats fed a high-fat diet during pregnancy showed an

increase in circulating estrogen during pregnancy and increased mammary

tumors (Hilakivi-Clarke et al. 1996).



Lactation. The American Association of Pediatrics (AAP) recommends

that all infants receive breast milk during the first 6 months (AAP

1997) and, further, that they are fed breast milk exclusively during

this time (AAP 2005), because of the numerous demonstrated benefits of

breast-feeding. Although data are limited, reports estimate that 3-6

million mothers are unable to produce milk or have difficulty

breast-feeding each year (Lew et al. 2009). The reasons for this remain

unclear, especially given that lactation insufficiency can be the result

of psychosocial as well as biological factors. However, environmental

chemicals are one candidate explanation for inability to initiate and/or

sustain breast-feeding (Neville and Walsh 1995).



Impaired lactation may be associated with altered MG development

(decreased or unresponsive breast tissue) and/or endocrine disruption

(improper hormonal support for lactation). Critical windows include

pregnancy and lactation as well as puberty and the prenatal/perinatal

period. As such, exposure to an EDC during pregnancy has the potential

to disrupt lactation in the mother and the daughter. In human studies,

strong early findings of associations between serum DDE and shortened

lactation in two populations have been only partly replicated, and few

other agents have been studied (Cupul-Uicab et al. 2008; Gladen and

Rogan 1995; Rogan et al. 1987). In rodents, impaired lactation has been

observed in conjunction with altered MG development in one or more

generations after gestational exposure to dioxin (Vorderstrasse et al.

2004), PFOA (White et al. 2007, 2009), atrazine (Rayner et al. 2005),

BPA (California Office of Environmental Health Hazard Assessment 2009;

Matsumoto et al. 2004), genistein [National Toxicology Program (NTP)

2008], and the candidate pharmaceutical Ziracin (Poulet et al. 2005).

For example, atrazine fed to rats during gestation induced MG

developmental changes in offspring, characterized by stunted

development, and when these rats were bred, their offspring

(second-generation) had significantly reduced weight gain, suggesting

insufficient milk production (Rayner et al. 2005). In an example of

effects on lactation in the dam, exposure of pregnant mice to PFOA

decreased pup weight and survival, diminished differentiation/growth of

dam MG, and induced some alterations in gene expression for milk

proteins, which taken together suggest effects on lactation in the

exposed dams (White et al. 2007, 2009).



Treatment during pregnancy has the potential to affect lactation in

both the dam and offspring. Impaired lactation in the dams is typically

identified because of decreased pup weight or survival, and impaired

lactation in the offspring can be determined only in multigenerational

studies where offspring are followed through successful reproduction and

lactation (Makris 2011). The rodent models and assessment methods used

in guideline studies arc not adequate for identifying effects on

lactation because the surrogate markers of pup weight and postnatal

survival are not sensitive or specific indicators of impaired lactation

(Makris 2011).



Human Health Risk



Assessment Issues



MG assessment in chemical test guidelines. MG development can be

affected after early exposure to EDCs in rodents. However, few guideline

studies for testing environmental chemicals include prenatal or early

life dosing, and MG end points are limited primarily to indirect or

surrogate observations during lactation and to clinical and pathological

evaluation of adult mammary tissue (Makris 2011). For example, the

standard 2-year rodent cancer bioassay, initiating treatment in young

adult animals, is likely to be less sensitive to carcinogens than if

developmental exposures were used, and it cannot provide information on

altered susceptibility to carcinogens induced by early life exposures

affecting MG development (Hovey et al. 2002; Medina 2007; Rudel et al.

2007; Russo and Russo 2004b; Singh et al. 2000; Thayer and Foster 2007).



To strengthen MG assessment and chemical testing, it is a priority

to enhance histopathological evaluation of MG development (e.g., using

longitudinal rather than transverse sectioning so that a larger tissue

plane is evaluated), increase attention to evaluation of male MG tissue,

and incorporate early life exposures in rodent subchronic and

chronic/carcinogenicity studies. Consistent with these recommendations,

the NTP has begun including gestational and lactational dosing in rats

assigned to subchronic and carcinogenicity studies and is taking steps

to include early life male and female MG whole-mount preparations and

longitudinal MG sectioning in reproductive assessment and cancer studies

(NTP 2010; Thayer and Foster 2007). Use of these expanded protocols will

facilitate linking altered MG development with later life outcomes.





Table 2. MG as a sensitive end point of endocrine disruption after

developmental exposures in rodents.(a)



Compound Study Species, MG effect

exposure type(b)

timing

Females



BPA Jenkins et al. Rat, Proliferation(d)

2009 postnatal

(lactation)



Murray et al- Rat, Hyperplasia(e)

2007 prenatal



Munoz-de-Toro Mouse, Morphology(f),

et al. 2005 perinatal proliferation(d)

DDT Brown and Rat, Proliferation(d)

Lamartiniere peripubertal



1995

Genistein Fritz etal. Rat, prenatal Morphology(f)

1998 and

postnatal



Padilla-Banks Mouse, Morphology(f)

et al. 2006 neonatal

Males



Genistein Delclos etal. Rat, prenatal Size(g);

2001 and hyperplasia(e)

postnatal



Compound MG effect Basis for

Females LOEL inclusion(c)



BPA 250[micro]g

No effects on

/ kg / age of V0,

day body weight,

serum

progesterone,

or serum

estradiol at

250 [micro]g / kg

/ day (highest

dose tested)

2.5[micro]g No effects on

/ kg / body weight,

day age of V0,

litter size,

or sex ratio

at this or

higher doses

(2.5-1,000 ug

/ kg / day).

25 ng / kg No effects on

/ day plasma

estradiol at

first

proestrus at

this or higher

dose (250

ng/kg / day)



DDT 50 ng / kg Single-dose

/ day study; no

effects on

body weight or

uterine-

ovarian

weight



Genistein 25 No effects on

mg/kg/day body weight,

uterine

weight, AGD,

estrous

cyclicity, or

age at V0 at

this or higher

dose (250 mg /

kg / day)

0.5 Effects on

mg/kg/day ability to

deliver live

pups and

estrous

cyclicity at

50 mg / kg /

day (but not

at either 0.5

or 5 mg / kg /

day)



Males



Genistein 25 ppm Effects on

ventral

prostate

weight,

pituitary

weight, age of

eye opening

and age of ear

unfolding at

1,250 ppm



Abbreviations: AGD, anogenital distance; LOEL, lowest observed effect

level. (a)For inclusion, a study must have assessed other end points

in addition to MG; findings are based on statistically significant

effects observed. (b)Ali effects are relative to negative controls;

effects on protein or gene expression are omitted. For more detail,

see Supplemental Material, Table 1 (doi: 10.1289/ehp. 1002864).

(c)See articles for further study methods and results. (d)Changes in

markers of proliferation/mitotic activity (e.g., cell cycle marker

proliferating cell nuclear antigen, cell number). (e)Changes in

numbers or sizes of hyperplastic structures. (f)Changes in

numbers/ratios of structures, branching, and so on for given

developmental stage. (g)Changes in the area or weight of gland.

As a potential addition to some toxicity test guidelines, MG

whole-mount assessment can demonstrate morphological changes in

development and differentiation and define the temporal and spatial

progression of epithelial development. Another important reason to

include MG assessments in screening-level toxicology studies is to

ensure that MG effeers are identified and can be evaluated in more

comprehensive studies. Specifically, data generated using whole mounts

may be used to trigger further assessment, such as: a) sectioning tissue

blocks, b) evaluating subsequent (e.g., F(1)) generations for

lactational impairment, c) maintaining a population longer on study for

spontaneous neoplasia evaluation, or d) evaluating altered tumor

susceptibility using a carcinogen-challenge protocol. Furthermore, a

whole mount may be the only indication of abnormal development in the

male MG, which is sensitive to very low doses in some studies (Delclos

et al. 2001; Latendresse et al. 2009). However, currently there are no

standardized whole-mount procedures, and consideration of these data in

chemical risk assessment has been limited. The OECD test guideline for

an extended one-generation reproductive toxicity study (OECD 2010) could

be revised to include assessment of MG development using whole mounts

and/or more thorough histopathology (Hvid et al. 2010). In addition, MG

assessment of males and females could be added to the U.S. EPA Endocrine

Disruptor Screening Program (EDSP) pubertal development protocols (U.S.

EPA 2009a, 2009b). MG developmental assessment could also be extended to

include females in the OECD Test Guideline 407 pubertal protocol (OECD

2008).



Adding MG whole-mount procedures to EDSP or OECD test guidelines

has raised concerns that a) these assays could be redundant to

endocrine-sensitive end points assessed [e.g., anogenit.al distance,

timing of vaginal opening (VO), circularing hormones, and estrous cyclicity], and b) that the procedure is too difficult to be

consistently executed across laboratories. However, in some cases MG

effects have been observed at lower doses than other EDC outcomes (Table

2), and there is concern that EDSP assays, which identify chemicals

affecting estrogen, androgen, or thyroid activities, may not be

sensitive to the many mechanisms that can affect breasr development. It

is reasonable, therefore, to include the MG whole mount in screening

studies, at least on a provisional basis, to see if the information

gathered is redundant or unique.



Human relevance of rodent models. Rodent models have been widely

used to characterize the influence of susceptibility factors (e.g.,

ovarian, pituitary, and placental hormones; life-stage and reproductive

events) on malignant transformation of the MG, and parallels between

rodent and human MG structures and pathologies have been enumerated

(Medina 2007; Russo and Russo 2004b; Singh et al. 2000). Although a few

findings in the context of chemicals testing have gener-ared concern

about human relevance (reviewed by Rudel et al. 2007), an extensive body

of breast cancer research demonstrates similarities between rodent and

human MG development and carcinogenesis. These studies indicate that

rodent mammary tumors mimic the diversity of human breast cancers with

respect to important initiation processes, histopathology, hormone

dependence, and host-target cell interactions (Boylan and Calhoon 1983;

Imaoka et al. 2009; Medina 2007; Rudland et al. 1998; Russo et al. 2000;

Russo and Russo 1993, 2004a; Singh et al. 2000). In general, research

indicates greater cross-species similarities for MG development and

cancer than for human menstrual and rodent estrous cyclicity or for

human puberty and rodent VO--end points currently included in many EDC

test protocols (U.S. EPA 2011).



An expert panel on MG tumors concluded that existing rodent models

are useful as screening tools for identifying potential breast

carcinogens (Thayer and Foster 2007). Further, the majority of chemicals

that are positive for mammary tumors in the rodent cancer bioassay have

some evidence of geno-toxicity and many are mulrisite carcinogens,

supporting relevance to humans (Rudel et al. 2007). Although there are

many similarities in the hormonal control of lactation across species,

less is known about the utility of the rodent as a model for predicting

chemical effects on human lactation. In any case, many risk assessment

guidelines operate on the principle that animal effects are considered

relevant to humans in the absence of data to indicate otherwise (U.S.

EPA 1991, 1996, 2005).



A related issue is the consideration of carcinogen-challenge models

as indicators of altered carcinogen susceptibility. DMBA and NMU are

primary breast-specific carcinogens that have been widely used in

experiments designed to assess the alteration of the tumor response by

hormones or other factors (Kamiya et al. 1995; Medina 2007; Singh et al.

2000). Despite the long-standing use of such carcinogen challenge

experiments to assess effects of hormonal or developmental alterations

on tumor susceptibility, the protocols are not common in chemical

toxicity assessment. Risk assessors have not considered data from

carcinogen challenge experiments because of concerns about the protocol

representing a chemical mixture study and about the presumed lack of

relevance of DMBA or NMU exposure to humans. However, a number of

consistent findings of increased susceptibility have been observed in

human and rodent studies across multiple MG end points for endogenous

hormonal factors, DES, genistein, and dioxin, among others (Table 1).

Models that consider the interactive effects of endogenous hormones and

carcinogenic factors across multiple life stages are likely to be more

relevant to human health than those with simpler design, because they

better reflect the human experience.





Table 3. Priority questions, current views, and issues for improving

risk assessment for MG effects.(a)



Priority Current views Outstanding

question for issues

risk

assessment

application



Are the rat Current knowledge Lack of

and mouse suggests that the information

adequate rat and mouse are about human

models for reasonable pubertal

human MG surrogates. development;

development? mechanisms

may differ

among

species.



What is the In utero exposure Few EDC

sensitivity of in some studies studies

MG leads to assess both

developmental developmental MG

effects? effects at doses development

similar to or and another

lower than other sensitive end

developmental and point of ED;

reproductive end there is a

points. lack of human

data to

address dose

response and

a lack of

standardized

MG

development

protocol and

assessment

criteria.



Are MG These changes in Varied

developmental MG are considered definitions

changes adverse because of

adverse? they represent "adversity,"

alterations in depending on

growth and scientific

development(b)and discipline

may be a risk and context.

factor for

lactation and/or

cancer outcomes.



(a)Based on the majority viewpoint of the experts at the Mammary

Gland Evaluation and Risk Assessment Workshop. (b)According to

guidance from the U.S. EPA (1991,1996).

Relative sensitivity of MG effects. A limited set of studies

provide evidence that MG alterations may be more sensitive to some EDCs

than arc other hormonally responsive end points (Table 2). To precisely

determine the relative sensitivity of EDC effects requires studies that

include MG as part of a larger set of endocrine-sensitive end points. Of

the studies that simultaneously evaluated MG morphology and at least one

other EDC-sensitive end point after developmental dosing, a subset has

detected effects on the MG at dose levels or during exposure periods

that did not elicit observable changes in other end points.



Adversity of MG developmental changes. Hormonal factors either

increase or decrease MG tumor susceptibility, and both transient and

permanent effects have been observed on MG development. This raises the

question of what types of alterations to MG development should be

considered adverse. In the context of regulatory evaluation of

chemicals, one point of view is that MG developmental changes reflect

altered growth and development, effects considered adverse by the U.S.

EPA Developmental Toxicity Risk Assessment Guidelines (U.S. EPA 1991).

For comparison, there is also controversy in the risk assessment

community about whether other common markers of altered pubertal timing

(e.g., VO, preputial separation) have human relevance. These end points

have nevertheless been considered adverse, as they are responsive to

endogenous sex steroids, which are important regulators of sexual

development conserved across mammalian species. By this reasoning,

altered MG growth and develop-menr, which is known to have human

relevance, should be considered adverse as well. The question of

adversity was discussed by experts gathered at the workshop in the

context of risk assessment. In spite of the outstanding questions, the

majority perspectives among experts advance the view that MG development

and subsequent effects represent a public health outcome of concern and

are a priority for future research and assessment. Priority questions,

current views, and outstanding issues for risk assessment are summarized

in Table 3.



An important question is whether MG developmental alterations are

plausibly related to increased tumor susceptibility by d) epi-genetic

imprinting of tissue, b) alteration of stem cell populations, or c)

increased number or ontological duration of TEBs or other structures

known to be more vulnerable to carcinogens. Some experts suggest that

such agents should themselves be considered carcinogens. Indeed, the

International Agency for Research on Cancer (IARC) deems an agent

carcinogenic if it is "capable of increasing the incidence of

malignant neoplasms, reducing their latency, or increasing their

severity or multiplicity" (IARC 2006). The U.S. EPA defines an

effect as adverse if it "reduces the organism's ability to

respond to an additional environmental challenge" (U.S. EPA 2010).

Applying these definitions, compounds that cause cancer, either alone or

in combination with other factors at a variety of points in a biological

chain of events leading to tumor formation, may reasonably be considered

carcinogens, including chemicals that increase susceptibility to cancer.

Even if such agents are not designated as carcinogens, their profound

impacts should encourage the risk assessment community to consider the

increase in cancer susceptibility as an adverse effect and therefore to

characterize doses required to elicit the effect. In any case, applying

this approach to risk assessment requires a better understanding of the

relationship between altered MG development and carcinogen

susceptibility.



Conclusions and Research Recommendations



Research demonstrates many similarities between humans and rodents

in normal and perturbed MG development and carcinogenesis. In both

humans and rodents, developmental exposure to hormones affects MG

development and carcinogen susceptibility, and these findings are the

basis for ongoing research to identify chemo-preventative agents in

humans and to determine how EDCs may alter breast cancer risk, pubertal

timing, or lactation. EDCs with diverse mechanisms of action, including

many not considered primarily estrogenic, alter MG development in

rodents. In some cases, altered MG development can be the most sensitive

endocrine end point.



The lack of consistent methods for evaluating and reporting MG

changes makes it difficult to compare findings across studies, hindering

consideration of MG developmental effects in rodent removal services risk assessment. Continued

progress will require consistent approaches across laboratories, along

with a discussion of unique findings and unanticipated effects. In

addition, the relationships between altered development and effects on

lactation or carcinogenesis are still being defined. Addressing these

research needs [detailed in Supplemental Material (doi: 10.1289/ehp.

1002864)] is a priority, and enhanced chemical testing and risk

assessment are needed to characterize these effects.



Major research initiatives under way include The National

Children's Study, which has a number of EDC hypotheses proposed for

testing in its longitudinal study (Lewin Group 2003), and the NIEHS Breast Cancer and the Environment Research Centers (NIEHS 2010), which

have ongoing human studies focusing on the relationship between

environmental exposures and age of breast development onset and which

also support experimental animal research in this area. Research

priorities identified at the workshop [provided in detail in

Supplemental Material (doi: 10.1289/ehp. 1002864)] include further

development and validation of the MG whole-mount protocol, research to

establish the relationship between effects on MG development and later

life outcomes, and issues relevant to use of these data in risk

assessment.



REFERENCES



AAP (American Academy of Pediatrics). 1997. Breastfeeding and the

use of human milk. Pediatrics 100(6):1035-1039.



AAP (American Academy of Pediatrics). 2005. Policy statement:

breastfeeding and the use of human milk. Pediatrics 115(2):496-506.



American Cancer Society. 2010. Cancer Facts and Figures 2010.

Atlanta, GA:American Cancer Society. Available:

http://www.cancer.org/acs/groups/content/@nho/documents/document/acspc-024113.pdf [accessed 17 June 2011].



Andrews P, Freyberger A, Hartmann E, Eiben R, Loot I, Schmidt U, et

al. 2002. Sensitive detection of the endocrine effects of the estrogen

analogue ethinylestradiol using a modified enhanced subacute rat study

protocol (OECD Test Guideline no. 407). Arch Toxicol 76(4): 194-202.



Birnbaum LS, Fenton SE. 2003. Cancer and developmental exposure to

endocrine disruptors. Environ Health Perspect 111:389-394.



Boylan ES, Calhoon RE. 1983. Transplacental action of

diethyl-stilbestrol on mammary carcinogenesis in female rats given one

or two doses of 7,12-dimethylbenz(a)anthracene. Cancer Res

43(101:4879-4884.



Brody JG, Rudel RA, Michels KB, Moysich KB, Bernstein L, Attfield

KR, et al. 2007. Environmental pollutants, diet, physical activity, body

size, and breast cancer: where do we stand in research to identify

opportunities for prevention? Cancer 109(suppl 121:2627-2634.



Brown NM, Lamartiniere CA. 1995. Xenoestrogens alter mammary gland

differentiation and cell proliferation in the rat. Environ Health

Perspect 103:708-713.



Brown NM, Manzolillo PA, Zhang JX, Wang J, Lamartiniere CA. 1998.

Prenatal TCDD and predisposition to mammary cancer in the rat.

Carcinogenesis 19(91:1623-1629.



California Office of Environmental Health Hazard Assessment. 2009.

Meeting State of California Office of Environmental Health Hazard

Assessment. Proposition 65: Developmental and Reproductive Toxicant Identification Committee [meeting transcript, 15 July 2009). Available:

http://oehha.ca.gov/prop65/public_meetings/pdf/DARTICTranscript71509.

pdf [accessed 17 June 2011].



Cardiff RD, Anver MR, Gusterson BA, Hennighausen L, Jensen RA,

Merino MJ, et al. 2000. The mammary pathology of genetically engineered

mice: the consensus report and recommendations from the Annapolis

meeting. Oncogene 19(81:968-988.



Cohn BA, Wolff MS, Cirillo PM, Sholtz Rl. 2007. DDT and breast

cancer in young women: new data on the significance of age at exposure.

Environ Health Perspect 115:1406-1414.



Cotroneo MS, Wang J, Fritz WA, Eltoum IE, Lamartiniere CA. 2002.

Genistein action in the prepubertal mammary gland in a chemoprevention

model. Carcinogenesis 23(9):1467-1474.



Cupul-Uicab LA, Gladen BC, Hernandez-Avila M, Weber JP, Longnecker

MP. 2008. DDE, a degradation product of DDT, and duration of lactation

in a highly exposed area of Mexico. Environ Health Perspect 116:179-183.



Delclos KB, BucciTJ, Lomax LG, Latendresse JR, Watbritton A, Weis

CC, et al. 2001. Effects of dietary genistein exposure during

development on male and female CD (Sprague-Dawley) rats. Reprod Toxicol

15(61:647-663.



Desaulniers D, Leingartner K, Russo J, Perkins G, Chittim BG,

Archer MC, et al. 2001. Modulatory effects of neonatal exposure to TCDD,

or a mixture of PCBs, p,p'-DDT, and p-p'-DDE, on

methylnitrosourea-induced mammary tumor development in the rat. Environ

Health Perspect 109:739-747.



Doherty LF, Bromer JG, Zhou Y, Aldad TS, Taylor HS. 2010. In utero

exposure to diethylstilbestrol (DES) or bisphenol-A (BPA) increases EZH2

expression in the mammary gland: an epigenetic mechanism linking

endocrine disruptors to breast cancer. Harm Cancer 1:146-155.



Durando M, Kass L, Piva J, Sonnenschein C, Soto AM, Luque EH, et

al. 2007. Prenatal bisphenol A exposure induces preneoplastic lesions in

the mammary gland in Wistar rats. Environ Health Perspect 115:80-86.



Eisen A, Lubinski J, Klijn J, Moller P, Lynch HT, Offit K, et al.

2005. Breast cancer risk following bilateral oophorectomy in BRCA1 and

BRCA2 mutation carriers: an international case-control study. J Clin

Oncol 23(30):7491-7496.



Enoch RR, Stanko JP, Greiner SN, Youngblood GL, Rayner JL, Fenton

SE. 2007. Mammary gland development as a sensitive end point after acute

prenatal exposure to an atrazine metabolite mixture in female Long-Evans

rats. Environ Health Perspect 115-.541-547.



Euling SY, Selevan SG, Pescovitz OH, Skakkebaek NE. 2008. Role of

environmental factors in the timing of puberty. Pediatrics 121(suppl

3):S167-S171.



Fenton SE. 2006. Endocrine-disrupting compounds and mammary gland

development: early exposure and later life consequences. Endocrinology

147(suppl 6):S18--S24.



Fenton SE, Hamm JT, Birnbaum LS, Youngblood GL. 2002. Persistent

abnormalities in the rat mammary gland following gestational and

lactational exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD).

Toxicol Sci 67(1):63-74.



Fielden MR, Fong CJ, Haslam SZ, Zacharewski TR. 2002. Normal

mammary gland morphology in pubertal female mice following in utevo and

lactational exposure to genistein at levels comparable to human dietary

exposure, Toxicol Lett 133(2-3):181-191.



Foley J, Dann P, Hong J, Cosgrove J, Dreyer B, Rimm D, et al. 2001.

Parathyroid hormone-related protein maintains mammary epithelial fate

and triggers nipple skin differentiation during embryonic breast

development. Development 128(4):513-525.



Fritz WA, Coward L, Wang J, Lamartiniere CA. 1998. Dietary

genistein: perinatal mammary cancer prevention, bioavailability and

toxicity testing in the rat. Carcinogenesis 19(12):2151-2158.



Gladen BC, Rogan WJ. 1995. DDE and shortened duration of lactation

in a northern Mexican town. Am J Public Health 85(4).504-508.



Henderson TO, Amsterdam A, Bhatia S, Hudson MM, Meadows AT, Neglia

JP, et al. 2010. Systematic review: surveillance for breast cancer in

women treated with chest radiation for childhood, adolescent, or young

adult cancer. Ann Intern Med 152(7):444-455.



Hilakivi-Clarke L, Cabanes A, de Assis S, Wang M, Khan G, Shoemaker

WJ, et al. 2004. In utero alcohol exposure increases mammary

tumorigenesis in rats. Br J Cancer 90(11):2225-2231.



Hilakivi-Clarke L, Cho E, Clarke R. 1998. Maternal genistein

exposure mimics the effects of estrogen on mammary gland development in

female mouse offspring. Oncol Rep 5(3):609-616.



Hilakivi-Clarke L, Cho E, Onojafe I, Raygada M, Clarke R. 1999.

Maternal exposure to genistein during pregnancy increases

carcinogen-induced mammary tumorigenesis in female rat offspring. Oncol

Rep 6(5): 1089-1095.



Hilakivi-Clarke L, Cho E, Raygada M, Kenney N. 1997. Alterations in

mammary gland development following neonatal exposure to estradiol,

transforming growth factor alpha, and estrogen receptor antagonist

IC1182,780. J Cell Physiol 170(3):279-289.



Hilakivi-Clarke L, Onojafe I, Raygada M, Cho E, Clarke R, Lippman

ME. 1996. Breast cancer risk in rats fed a diet high in n-6

polyunsaturated fatty acids during pregnancy. J Natl Cancer Inst

88(24):1821-1827.



Hoover RN, Troisi RJ. 2001. Understanding mechanisms of breast

cancer prevention. J Natl Cancer Inst 93(15):1119-1120.



Hovey RC, Asai-Sato M, Warri A, Terry-Koroma B, Colyn N, Ginsburg

E, et al. 2005. Effects of neonatal exposure to diethylstilbestrol,

tamoxifen, and toremifene on the BALB/c mouse mammary gland. Biol Reprod

72(2):423-435.



Hovey RC, Trott JF, Vonderhaar BK. 2002. Establishing a framework

for the functional mammary gland: from endocrinology to morphology. J

Mammary Gland Biol Neoplasia 7(1):17-38.



Hvid H, Thorup I, Oleksiewicz MB, Sjogren I, Jensen HE. 2010. An

alternative method for preparation of tissue sections from the rat

mammary gland. Exp Toxicol Pathol 63(4):317-324;

doi:10.1016/j.etp.2010.02.005 [Online 2 March 2010).



IARC (International Agency for Research on Cancer). 2006. Preamble

to the IARC Monographs (amended January 2006). Available:

http://monographs.iarc.fr/ENG/Preamble/index.php [accessed 17 June

2011].



Imaoka T, Nishimura M, leuka D.Daino K.Takabatake T, Okamoto M,

etal. 2009. Radiation-induced mammary carcinogenesis in rodent models:

what's different from chemical carcinogenesis? J Radiat Res (Tokyo)

50(4):281-293.



Jenkins S, Raghuraman N, Eltoum I, Carpenter M, Russo J,

Lamartiniere CA. 2009. Oral exposure to bisphenol A increases

dimethylbenzanthracene-induced mammary cancer in rats. Environ Health

Perspect 117:910-915.



Jenkins S, Rowell C, Wang J, Lamartiniere CA. 2007. Prenatal TCDD

exposure predisposes for mammary cancer in rats. Reprod Toxicol

23(31:391-396.



Jones LA, Bern HA. 1979. Cervicovaginal and mammary gland

abnormalities in BALB/cCrgl mice treated neonatally with progesterone

and estrogen, alone or in combination. Cancer Res 39(7 Pt 1):2560-2567.



Kamiya K, Yasukawa-Barnes J, Mitchen JM, Gould MN, Clifton KH.

1995. Evidence that carcinogenesis involves an imbalance between

epigenetic high-frequency initiation and suppression of promotion. Proc

Natl Acad Sci USA 92(5):1332-1336.



Kaplowitz PB. 2008. Link between body fat and the timing of

puberty. Pediatrics 121(suppl 3):S208-S217.



Kelsey JL, Gammon MD, John EM. 1993. Reproductive factors and

breast cancer. Epidemiol Rev 15(1):36-47.



Kleinberg DL, Wood TL, Furth PA, Lee AV. 2009. Growth hormone and

insulin-like growth factor-l in the transition from normal mammary

development to preneoplastic mammary lesions. Endocr Rev 30(1):51-74.



Kociba RJ, Keyes DG, Beyer JE, Carreon RM, Wade CE, Dittenber DA,

et al. 1978. Results of a two-year chronic toxicity and oncogenicity study of 2,3,7,8-tetrachlorodibenzo-p-dioxin in rats. Toxicol Appl

Pharmacol 46(2):279-303.



Land CE. 1995. Studies of cancer and radiation dose among atomic

bomb survivors. The example of breast cancer. JAMA 274(5):402-407.



Latendresse JR, Bucci TJ, Olson G, Mellick P, Weis CC, Thorn B, et

al. 2009. Genistein and ethinyl estradiol dietary exposure in

multigenerational and chronic studies induce similar proliferative

lesions in mammary gland of male Sprague-Dawley rats. Reprod Toxicol

28(3):342-353.



Lew BJ, Collins LL, O'Reilly MA, Lawrence BP. 2009. Activation

of the aryl hydrocarbon receptor during different critical windows in

pregnancy alters mammary epithelial cell proliferation and

differentiation. Toxicol Sci 111(1):151-162.



Lewin Group. 2003. Summary of Hypotheses Identified through The

Lewin Group's Targeted Literature Review (02SG0067). Available:

http://www.nationalchildrensstudy.gov/about/organization/advisorycommittee/2003Sep/Pages/NCS-Summary-of-Hyptheses.pdf [accessed 19 November

2010].



Li X, Warri A, Makela S, Ahonen T, Streng T, Santti R, et al.



EDCs and altered mammary gland development 2002. Mammary gland

development in transgenic male mice expressing human P450 aromatase.

Endocrinology 143(101:4074-4083.



Lopez J, Ogren L, Verjan R, Talamantes F. 1988. Effects of

perinatal exposure to a synthetic estrogen and progestin on mammary

tumorigenesis in mice. Teratology 38(2):129-134.



Makris SL. 2011. Current assessment of the effects of environmental

chemicals on the mammary gland in guideline rodent studies by the U.S.

Environmental Protection Agency, Organisation tor Economic Co-operation

and Development, and National Toxicology Program. Environ Health

Perspect 119:1047-1052.



Matsumoto C, Miyaura C, Ito A. 2004. Bisphenol-A suppresses the

growth of newborn pups through insufficient supply of maternal milk in

mice. J Health Sci 50(31:315-318.



Medina D. 2005, Mammary developmental fate and breast cancer risk.

Endocr Relat Cancer 12(31:483-495.



Medina D. 2007. Chemical carcinogenesis of rat and mouse mammary

glands. Breast Dis 28:63-68.



Moon HJ, Han SY, Shin JH, Kang IH, Kim TS, Hong JH, et al. 2007.

Gestational exposure to nonylphenol causes precocious mammary gland

development in female rat offspring. J Reprod Dev 53(2):333-344.



Mori T, Bern HA, Mills KT, Young PN. 1976. Long-term effects of

neonatal steroid exposure on mammary gland development and tumorigenesis

in mice. J Natl Cancer Inst 57(5):1057-1062.



Mori T, Nagasawa H, Bern HA. 1979. Long-term effects of perinatal

exposure to hormones on normal and neoplastic mammary growth in rodents:

a review. J Environ Pathol Toxicol 3(1-2):191-205.



Munoz-de-Toro M, Markey CM, Wadia PR, Luque EH, Rubin BS,

Sonnenschein C, et al. 2005. Perinatal exposure to bisphenol-A alters

peripubertal mammary gland development in mice. Endocrinology 146(9):

4138-4147.



Murray TJ, Maffini MV, Ucci AA, Sonnenschein C, Soto AM. 2007.

Induction of mammary gland ductal hyperplasias and carcinoma in situ following fetal bisphenol A exposure. Reprod Toxicol 23(3):383-390.



Neville MC, Walsh CT. 1995. Effects of xenobiotics on milk

secretion and composition. Am J Clin Nutr 61(suppl 3):687S-694S.



NIEHS (National Institute of Environmental Health Sciences). 2010.

Breast Cancer and the Environment Research Centers. Available:

http://www.niehs.nih.gov/research/supported/centers/breast-cancer/index.cfm [accessed 17 June 2011].



NTP (National Toxicology Program). 2008. Multigenerational

Reproductive Toxicology Study of Genistein (CAS NO. 446-72-0) in

Sprague-Dawley Rats (Feed Study). TR 539. Research Triangle Park,

NC:NTP. Available: http://ntp. niehs.nih.gov/files/539_FINAL_WEB_508.pdf

[accessed 17 June 2011].



NTP (National Toxicology Program). 2010. NTP Study Types:

Toxicology/Carcinogenicity. Available: http://ntp.niehs.nih.

gov/INDEX33B7.HTM?objectid=72015DAF-BDB7-CEBA-F9A7F9CAA57DD7F5 [accessed

21 June 2011].



Odum J, Pyrah IT, Soames AR, Foster JR, Van Miller JP, Joiner RL,

et al. 1999. Effects of p-nonylphenol (NP) and diethylstilboestrol (DES)

on the Alderley Park (Alpk) rat: comparison of mammary gland and uterus

sensitivity following oral gavage or implanted mini-pumps. J Appl

Toxicol 19(5):367-378.



OECD (Organisation for Economic Co-operation and Development).



2008. OECD Guideline for Testing of Chemicals. Repeated Dose 28-Day Oral

Toxicity Study in Rodents. TG 407. Paris:OECD.



OECD (Organisation for Economic Co-operation and Development).

2010. OECD Guideline for the Testing of Chemicals. Draft Proposal for an

Extended One-Generation Reproductive Toxicity Study. Parrs.OECD.

Available: http://www.oecd.org/dataoecd/23/10/46466062.pdf [accessed 17

June 2011].



Okazaki K, Okazaki S, Nishimura S, Nakamura H, Kitamura Y, Hatayama

K, etal. 2001. A repeated 28-day oral dose toxicity study of

methoxychlor in rats, based on the 'enhanced OECD test guideline

407' for screening endocrine-disrupting chemicals. Arch Toxicol

75(9):513-521.



Padilla-Banks E, Jefferson WN, Newbold RR. 2006. Neonatal exposure

to the phytoestrogen genistein alters mammary gland growth and

developmental programming of hormone receptor levels. Endocrinology

147(10):4871-4882.



Palmer JR, Wise LA, Hatch EE, Troisi R, Titus-Ernstoff L,

Strohsnitter W, et al. 2006. Prenatal diethylstilbestrol exposure and

risk of breast cancer. Cancer Epidemiol Biomarkers Prev 15(8):1509-1514.



Poulet FM, Veneziale R, Vancutsem PM, Losco P, Treinen K, Morrissey

RE. 2005. Ziracin-induced congenital urogenital malformations in female

rats. Toxicol Pathol 33(3):320-328.



Rayner JL, Enoch RR, Fenton SE. 2005. Adverse effects of prenatal

exposure to atrazine during a critical period of mammary gland growth.

Toxicol Sci 87(1):255-266.



Renehan AG, Tyson M, Egger M, Heller RF, Zwahlen M. 2008. Body-mass

index and incidence of cancer: a systematic review and meta-analysis of

prospective observational studies. Lancet 371 (9612):569-578.



Rogan WJ, Gladen BC, McKinney JD, Carreras N, Hardy P, Thullen J,

et al. 1987. Polychlorinated biphenyls (PCBs) and dichlorodiphenyl

dichloroethene (DDE) in human milk: effects on growth, morbidity, and

duration of lactation. Am J Public Health 77(10):1294-1297.



Rothschild TC, Boylan ES, Calhoon RE, Vonderhaar BK. 1987.

Transplacental effects of diethylstilbestrol on mammary development and

tumorigenesis in female ACI rats. Cancer Res 47(16):4508-4516.



Rudel RA, Attfield KR, Schifano JN, Brody JG. 2007. Chemicals

causing mammary gland tumors in animals signal new directions for

epidemiology, chemicals testing, and risk assessment for breast cancer

prevention. Cancer 109(suppl 12):2635-2666.



Rudland PS, Barraclough R, Fernig DG, Smith JA. 1998. Growth and

differentiation of the normal mammary gland and its tumours. Biochem Soc

Symp 63:1-20.



Russo IH, Russo J. 1978. Developmental stage of the rat mammary

gland as determinant of its susceptibility to

7,12-dimethylbenz[a]anthracene. J Natl Cancer Inst 61(6):1439-1449,



Russo IH, Russo J. 1996. Mammary gland neoplasia in long-term

rodent studies. Environ Health Perspect 104:938-967.



Russo J, Hu YF, Yang X, Russo IH. 2000. Developmental, cellular,

and molecular basis of human breast cancer. J Natl Cancer

lnstMonogr(27):17-37.



Russo J, Russo IH. 1993. Development pattern of human breast and

susceptibility to carcinogenesis. Eur J Cancer Prev 2(suppl 3):85-100.



Russo J, Russo IH. 2004a. Development of the human breast.

Maturitas 49(1):2-15.



Russo J, Russo IH. 2004b. Molecular Basis of Breast Cancer. New

York:Springer.



Silent Spring Institute. 2011. Silent Spring Institute Organizes

Scientific Workshop on Mammary Gland Evaluation and Risk Assessment.

Available: http://www.silentspring.org/mammary-gland-workshop [accessed

4 March 2011).



Singh M, McGinley JN, Thompson H J. 2000. A comparison of the

histopathology of premalignant and malignant mammary gland lesions

induced in sexually immature rats with those occurring in the human. Lab

Invest 80(2):221-231.



Sivaraman L, Stephens LC, Markaverich BM, Clark JA, Krnacik S,

Conneely OM, et al. 1998. Hormone-induced refractoriness to mammary

carcinogenesis in Wistar-Furth rats. Carcinogenesis 19(9):1573-1581.



Thayer KA, Foster PM. 2007. Workgroup report: National Toxicology

Program workshop on Hormonally Induced Reproductive Tumors--Relevance of

Rodent Bioassays. Environ Health Perspect 115:1351-1356.



Thompson HJ, Singh M. 2000. Rat models of premalignant breast

disease. J Mammary Gland Biol Neoplasia 5(4):409--420.



Titus-Ernstoff L, Hatch EE, Hoover RN, Palmer J, Greenberg ER,

Ricker W, et al. 2001. Long-term cancer risk in women given

diethylstilbestrol (DES) during pregnancy. Br J Cancer 84(f):126-133.



Tomooka Y, Bern HA. 1982. Growth of mouse mammary glands after

neonatal sex hormone treatment. J Natl Cancer Inst 69(6):1347-1352.



Trichopoulos D, MacMahon B, Cole P. 1972. Menopause and breast

cancer risk. J Natl Cancer Inst 48(3):605-613.



Troisi R, Potischman N, Hoover RN. 2007. Exploring the underlying

hormonal mechanisms of prenatal risk factors for breast cancer: a review

and commentary. Cancer Epidemiol Biomarkers Prev 16(9):1700-1712.



U.S. EPA (U.S. Environmental Protection Agency). 1991. Guidelines

for Developmental Toxicity Risk Assessment. Fed Reg 56: 63798-63826.

EPA/600/FR-91/Q01. Washington, DC:U.S. EPA. Available:

http://oaspub.epa.gov/eims/eimscomm. getfile?p_download_id=4560 [accessd

17 June 2011].



U.S. EPA (U.S. Environmental Protection Agency). 1996. Guidelines

for Reproductive Toxicity Risk Assessment. 630/R-96/009. Washington,

DC:U.S. EPA.



U.S. EPA (U.S. Environmental Protection Agency). 2005. Guidelines

for Carcinogen Risk Assessment. EPA/630/P-03/001F. Washington, DC:U.S.

EPA. U.S. EPA (U.S. Environmental Protection Agency). 2009a. OPPTS 890.1450. Pubertal Development and Thyroid Function in Intact

Juvenile/Peripubertal Female Rats. EPA 740-C-09-009. Washington, DC:U.S.

EPA.



U.S. EPA (U.S. Environmental Protection Agency). 2009b. OPPTS

890.1500: Pubertal Development and Thyroid Function in Intact

Juvenile/Peripubertal Male Rats. EPA 740-C-09-012. Washington, OC:U.S.

EPA.



U.S. EPA (U.S. Environmental Protection Agency). 2010. Integrated

Risk Information System. Available: http://www.epa.gov/IRIS/ [accessd 17

June 2011]. U.S. EPA (U.S. Environmental Protection Agency). 2011.

Endocrine Disruptor Screening Program assay status table. Available:

http://www.epa.gov/endo/pubs/assayvalidation/status.htm [accessed 22

June 2011].



Vandenberg LN, Maffini MV, Schaeberle CM, Ucci AA, Sonnenschein C,

Rubin BS, et al. 2008. Perinatal exposure to the xenoestrogen

bisphenol-A induces mammary intraductal hyperplasias in adult CD-I mice.

Reprod Toxicol 26(3-4):210-219.



Vandenberg LN, Maffini MV, Wadia PR, Sonnenschein C, Rubin BS, Soto

AM. 2007. Exposure to environmentally relevant doses of the xenoestrogen

bisphenol-A alters development of the fetal mouse mammary gland.

Endocrinology 148(1):116-127.



Verloop J, van Leeuwen FE, Helmerhorst TJ, van Boven HH, Rookus MA.

2010. Cancer risk in DES daughters. Cancer Causes Control

21(7):999-1007.



Vogel VG, Costantino JP, Wickerham DL, Cronin WM, Cecchini RS,

Atkins JN, et al. 2010. Update of the National Surgical Adjuvant Breast

and Bowel Project Study of Tamoxifen and Raloxifene (STAR) P-2 Trial:

Preventing Breast Cancer. Cancer Prev Res (Phila) 3(61:696-706.



Vorderstrasse BA, Fenton SE, Bohn AA, Cundiff JA, Lawrence BP.

2004. A novel effect of dioxin: exposure during pregnancy severely

impairs mammary gland differentiation. Toxicol Sci 78(2):248-257.



Warner MR. 1976. Effect of various doses of estrogen to BALB/cCrgl

neonatal female mice on mammary growth and branching at 5 weeks of age.

Cell Tissue Kinet 9(5):429-438.



Warri A, Saarinen NM, Makela S, Hilakivi-Clarke L. 2008. The role

of early life genistein exposures in modifying breast cancer risk. Br J

Cancer 98(9):1485-1493.



White SS, Calafat AM, Kuklenyik Z, Villanueva L, Zehr RD, Helfant

L, et al. 2007. Gestational PFOA exposure of mice is associated with

altered mammary gland development in dams and female offspring. Toxicol

Sci 96(1):133-144.



White SS, Kato K, Jia LT, Basden BJ, Calafat AM, Hines EP, et al.

2009. Effects of perfluorooctanoic acid on mouse mammary gland

development and differentiation resulting from cross-foster and

restricted gestational exposures. Reprod Toxicol 27(3-4):289-298.



Xue F, Michels KB. 2007. Intrauterine factors and risk of breast

cancer: a systematic review and meta-analysis of current evidence.

Lancet Oncol 8(12):1088-1100.



Yang C, Tan YS, Harkema JR, Haslam SZ. 2009. Differential effects

of peripubertal exposure to perfluorooctanoic acid on mammary gland

development in C57BI/6 and Balb/c mouse strains. Reprod Toxicol

27(3-41:299-306.



You L, Casanova M, Bartolucci EJ, Fryczynski MW, Dorman DC, Everitt

Jl, et al. 2002. Combined effects of dietary phytoestrogen and synthetic

endocrine-active compound on reproductive development in Sprague-Dawley

rats: genistein and methoxychlor. Toxicol Sci 66(1):91-104.



Ruthann A.Rudel,(1) Suzanne E. Fenton, (2) Janet M. Ackerman, (1)

Susan Y. Euling, (3) adn Susan L. Makris(3)



(1)Silent Spring Institute, Newton, Massachusetts, USA; (2)National

Toxicology Program, National Institute of Environment Health Sciences,

National Institutes of Health, Department of Health and Human Services,

Research Triangle Park, North Carolina, USA; (3)National Center for

Environmental Assessment, U.S. Environmental Protection Agency,

Washington, DC, USA



Address correspondence to R. Rudel, Silent Spring Institute, 29

Crafts St., Newton, MA 02458 USA. Telephone: (617) 332-4288, ext 214.

Fax: (617) 332-4284. E-mail: rudel@silcntspring.org



Supplemental Material is available online (doi: 10.1289/ehp.

1002864 via http://dx.doi.org/).



We thank our workshop participants for their contributions to this

project [sec Supplemental Material, p. 2 (doi: 10.1289/ehp. l002864)].

We also thank C. Reed for careful review of the tables in Supplemental

Material.



This workshop was supported by the California Breast Cancer

Research Program, U.S. Environmental Protection Agency (U.S. EPA),

National Institute or Environmental Health Sciences (NIEHS), National

Toxicology Program (NTP), and Silent Spring Institute.



The views expressed in this paper are those of the authors and do

not necessarily reflect the views or policies of the U.S. EPA or NIEHS.



R.A.R. and J.M.A. are employed at Silent Spring Institute, a

scientific research organization dedicated to studying environmental

factors in women's health. The Institute is a 501(c)3 public chanty funded by federal grants and contracts, foundation grants, and private

donations, including donations from breast cancer organizations. The

authors declare they have no actual or potential competing financial

interests.



Received 16 August 2010; accepted 17 May 2011.

This website was created for free with Own-Free-Website.com. Would you also like to have your own website?
Sign up for free