You are on page 1of 16

TOXICOLOGICAL SCIENCES 120(S1), S93–S108 (2011)

doi:10.1093/toxsci/kfq329
Advance Access publication October 25, 2010

Endocrine Disruption: Historical Perspectives and Its Impact on the


Future of Toxicology Testing

Downloaded from https://academic.oup.com/toxsci/article/120/suppl_1/S93/1615160 by guest on 03 January 2021


Mary Sue Marty,1 Edward W. Carney, and Justin Craig Rowlands
Toxicology and Environmental Research and Consulting, The Dow Chemical Company, Midland, Michigan

1
To whom correspondence should be addressed at Toxicology and Environmental Research and Consulting, The Dow Chemical Company, 1803 Building,
Midland, MI 48674. Fax: (989) 638-9863. E-mail: mmarty@dow.com.

Received August 13, 2010; accepted October 15, 2010

The endocrine system is one of the body’s major toxicity, followed by coverage of key historical events in
homeostatic control systems whose aim is to maintain normal basic science, and then bring the two together to discuss
functions and development in the face of a constantly changing implications of the science on testing both now and for the
environment. Working in tandem with the nervous system, future.
which mainly is responsible for rapid and immediate responses,
the approximately 30 different glands comprising the endocrine
system tend to act in a slower and more sustained manner to EARLY HISTORY OF TOXICITY TESTING FOR
regulate processes as diverse as the female reproductive cycle, ENDOCRINE-MEDIATED EFFECTS
bone growth, cell proliferation, and psychosocial behavior.
Multiple endocrine glands also work in concert with one A casual observer might easily be led to believe that the
another to form complex feedback loops, which tightly regulate ability of exogenous substances to interfere with the endocrine
critical physiological processes. system was unknown until relatively recently, but in fact, this is
Like all homeostatic control systems, the capacity to far from true. Even the ancients were familiar with the actions
maintain physiological parameters within normal bounds is of various herbal preparations to modulate what we now know
finite, and when this capacity is exceeded by chemical to be hormonally regulated processes. For centuries, farmers
exposures, drugs, or environmental stressors, adverse have observed reproductive problems in female sheep and
consequences can ensue. In the mid-1990’s, concerns about cows grazing on pastures rich in certain clover species, which
the potential for environmental chemicals, drugs, and other later were found to contain estrogenic compounds such as
stressors to alter endocrine physiology rapidly mounted and coumestrol (Adams, 1995). As early as the 1930’s, the ability
received a great deal of attention within the toxicology of both natural and synthetic chemicals to interact with
community as well as in the public media. These circumstances endogenous hormone receptors was already well established
spurred a flurry of research on mechanisms of toxicity, new (Schueler, 1946; Sluczewski and Roth, 1948; Walker and
questions about how to deal with cumulative exposures to Janney, 1930).
endocrine-active compounds (EACs) from man-made, dietary, Starting around the 1940’s, steroidal compounds began to be
and endogenous sources, and the creation of major chemical used in the livestock industry to modulate reproductive cycles
screening and testing programs focused on endocrine-mediated and to enhance rate and efficiency of body weight gain, while
modes of toxic action. compounds such as ethinyl estradiol, mestranol, norethynodrel,
In this review, we will trace the path of knowledge regarding and megestrol acetate were being developed as contraceptive
the effects of exogenous agents on the endocrine system, the agents and to treat specific disease conditions in humans
events which gave rise to the explosion of interest in this topic (Newburgh, 1975). In parallel with these developments,
in the mid-1990’s, and how it led to a fundamentally different toxicologists began to investigate these compounds for po-
paradigm in toxicity testing as well as a great deal of basic tential adverse effects.
research on mechanisms of endocrine-mediated toxicity. We Diethylstilbestrol (DES), a ‘‘synthetic estrogen’’ with similar
first cover the history of testing for endocrine-mediated potency to 17b-estradiol, was first prescribed in 1938 to
Ó The Author 2010. Published by Oxford University Press on behalf of the Society of Toxicology. All rights reserved.
For permissions, please email: journals.permissions@oup.com
S94 MARTY, CARNEY, AND ROWLANDS

prevent miscarriage and premature births. This drug was We Threatening Our Fertility, Intelligence and Survival?—
particularly significant as the daughters of women who took A Scientific Detective Story written by Colborn et al. (1996)
DES were found to have an increased incidence of clear-cell with a forward penned by then Vice President Al Gore. These
carcinoma, a rare vaginal cancer. As a result, in 1971, the Food and other authors proposed that many EACs elicited effects at
and Drug Administration (FDA) advised physicians to stop doses far lower than toxicities caused by other modes of
prescribing DES to pregnant women. Further research has action (MOAs) and thus required special regulation. Also
revealed an increased risk for breast cancer in DES mothers around the same time were a number of more specific reports
and also possible links with reproductive tract abnormalities in alleging associations between endocrine-disrupting chemicals
DES sons and daughters (Giusti et al., 1995). It has been and declining sperm counts (Carlsen et al., 1992; Sharpe and
estimated that 5–10 million Americans received DES during Skakkebaek, 1993), increases in breast cancer (Davis et al.,
pregnancy or were exposed to the drug in utero. 1993), cryptorchism and hypospadias (Barlow et al., 1999),

Downloaded from https://academic.oup.com/toxsci/article/120/suppl_1/S93/1615160 by guest on 03 January 2021


Other episodes of endocrine-mediated toxicity also occurred and developmental abnormalities in wildlife species (Sumpter,
via unintentional exposure to hormonally active compounds, 1995). The level of concern spiked even higher following a
such as polychlorinated biphenyls (PCBs) and polychlorinated 1996 report claiming > 1000-fold level of synergy among
dibenzofurans (PCDFs). Some members of this large family of a mixture of four weakly estrogenic agents as assessed in a
structurally related congeners can act on the estrogen and yeast estrogen receptor (ER) transcriptional activation assay
arylhydrocarbon (Ah) receptor systems and/or modulate (Arnold et al., 1996). Ironically, the latter results could not be
thyroid hormone function. In Japan (1968) and Taiwan reproduced by others (Ashby et al., 1997; Ramamoorthy
(1979), a few thousand women were exposed to PCBs and et al., 1997) and a National Institutes of Health inquiry
their pyrolysis products (PCDFs) following consumption of revealed intentional falsification of the results leading to
contaminated rice oil. The offspring of these mothers exposed retraction of the original report, which had been published in
to PCBs and PCDFs tended to be smaller at birth and Science (McLachlan, 1997).
exhibited delays in neurological development (Aoki, 2001). Despite uncertainties about the scientific validity of these
Subsequent studies on PCB exposure at lower levels have reports and the true extent of the problem, both the scientific
alleged other adverse effects, although these data remain community and public reacted as if this were an approaching
somewhat controversial. tsunami. Media coverage of what would previously have been
In the environment, the presence of natural hormones in considered an esoteric science topic became mainstream
wastewater treatment outfalls in the United States was noted as headline news. Prominent government and scientific organ-
early as 1965 (Stumm-Zollinger and Fair, 1965), and this work izations such as the World Health Organization, International
expanded in 1970 to include synthetic estrogens used as birth Union of Pure and Applied Chemistry and the U.S. National
control agents (Tabak and Bunch, 1970). By the mid-1970’s, Research Council reacted by gathering endocrinologists,
scientists began to detect other pharmaceutical agents near toxicologists, and other public health professionals from
wastewater outlets in the United States (Garrison and Pope, around the world to capture their knowledge of the state-of-
1975; Hignite and Azarnoff, 1977). Concerns about dichlor- the-science and to recommend research to address the issue.
odiphenyltrichloroethane (DDT) in the environment were These efforts are documented in a series of comprehensive
highly publicized in Rachel Carson’s book, Silent Spring, review papers (IPCS, 2002; Lintelmann et al., 2003; NRC,
although a specific link to endocrine-mediated toxicity was yet 1999), which also helped spur a plethora of research on
to come. Despite the aforementioned events in both wildlife endocrine-mediated toxicity. The IPCS (2002) report was
and humans, endocrine-mediated toxicity did not garner any particularly significant in that it described a structured weight-
special attention above and beyond other mechanisms of of-evidence approach to guide the application of new research
toxicity known at the time. Instead, it tended to be regarded as for addressing causal relationships between exposure to
just one of many potential mechanisms that could lead to EACs and adverse health outcomes. In fact, application of
certain adverse outcomes, such as reproductive toxicity, weight-of-evidence approaches has failed to provide con-
developmental toxicity, or cancer. vincing support for many of the early claims that initially
sparked the issue, such as alleged links between EACs and
declining sperm counts (Fisch, 2008) and breast cancer
THE BIRTH OF ‘‘ENDOCRINE DISRUPTORS’’ (summarized at: http://epi.grants.cancer.gov/LIBCSP/ ). Re-
ported effects in aquatic species have pointed more toward
This mindset changed rather abruptly in the mid-1990’s natural and synthetic hormones (e.g., birth control pills) as
following a series of publications suggesting that pesticides and causative agents, more so than industrial chemicals as
other man-made chemicals were threatening the reproductive originally suggested (Jobling et al. 2006). Nonetheless, the
capability and intelligence of future generations of humans and term endocrine disruptor has been adopted in the vernacular
wildlife (Colborn et al., 1996, 1993; Hunter and Kelsey, 1993). of toxicologists, public health professionals, and even the
Of particular impact was a book entitled Our Stolen Future, Are general public and appears here to stay.
ENDOCRINE-MEDIATED TOXICITY S95

Somewhat unwittingly, the new emphasis on endocrine and antagonists, as well as compounds which act indirectly.
disruption also marked the beginning of a fundamental change Indirect mechanisms are plentiful and include the inhibition of
away from the traditional ‘‘outside-in’’ approach to toxicology, steroid biosynthesis or modulation of the hypothalamic-
in which adverse outcomes are identified first followed by pituitary-gonadal (HPG) axis. An even more complex example
mechanistic research to understand these effects. With begins with activation of the Ah receptor (AhR), which can
endocrine disruption came a reversed approach in which ultimately oppose estrogen signaling.
identification of mechanism came first followed by research to Ironically, some compounds bearing the very properties
define possible consequences of activating these mechanisms. that are the targets of screening can actually be beneficial. Studies
The implications of this change are quite significant and are of endocrine-disrupting AhR ligands have led to the development
discussed later in this review. of selective AhR modulators, such as ring-substituted
diindolylmethanes (DIMs), for chemotherapy of breast,

Downloaded from https://academic.oup.com/toxsci/article/120/suppl_1/S93/1615160 by guest on 03 January 2021


endometrial, pancreatic, and prostate cancer (Safe et al.,
GENESIS OF ENDOCRINE SCREENING AND TESTING 2008). Substituted furans and indoles have been licensed by a
pharmaceutical company as antitumor development compounds
At around the same time that these alarming reports were (Safe, personal communication). Some of the DIMs have been
emerging, the U.S. Safe Drinking Water Act was being updated further modified into anticarcinogenic AhR-inactive analogs,
while the Food Quality Protection Act was being created to which activate orphan nuclear receptors that induce genes
enhance protection of children and other vulnerable subpopula- associated with growth inhibition and cancer cell death.
tions. This temporal convergence led to a 1996 congressional
mandate requiring the Environmental Protection Agency (EPA) to
implement a screening and testing program to detect endocrine EPA’S ENDOCRINE DISRUPTOR SCREENING PROGRAM
disruptors. Despite the nascent state of the science at the time, TIER I ASSAYS
Congress gave EPA just 2 years to establish the new program. To
assist EPA, an advisory committee known as the Endocrine In 1998, EDSTAC released its report outlining a screening
Disruptor Screening and Testing Advisory Committee (EDSTAC) and testing program to assess the potential of compounds to
was formed to evaluate protocols for approximately 50 different interact with the endocrine system. EDSTAC recommended
endocrine assays and select a subset to comprise a ‘‘Tier I’’ that a tiered approach be adopted to examine endocrine activity
screening battery for biological activity involving the estrogen, with a focus on the potential for compounds to interact with the
androgen, and thyroid (EAT) hormone systems. EAT hormone systems. The Tier I assays were designed to
Although limiting screening to these three hormone systems screen for potential interaction with these endocrine systems
might seem arbitrary, these three appeared to have the broadest and designed to be an efficient screening process to screen
range of potential effects as well as the greatest number of a large number of chemicals for endocrine activity. In Tier I,
ligands with which they interact (Blair et al., 2000). EDSTAC opted to prioritize assays with greater sensitivity (but
Compounds that can bind steroid hormone receptors include possibly lower specificity) to avoid false-negative results.
industrial chemicals such as bisphenol A (BPA), certain EDSTAC described the importance of using a ‘‘weight-of-
alkylphenol ethoxylates, phthalates, parabens, benzophenones, evidence’’ approach to evaluate Tier I screening results. Tier II
and environmental contaminants like DDT, methoxychlor, tests, such as the multigeneration reproductive toxicity study,
some PCBs, dioxins and furans. There also are many were to be used to characterize dose-response relationships,
pharmaceuticals purposely designed to have hormonal activity, whether endocrine-mediated adverse effects for EAT occur, as
such as DES, contraceptive agents, and the selective ER- well as providing data for risk assessment. During this time
modulators (SERMs) used in the treatment of diseases such as period, EPA also took on an extensive revision of the
osteoporosis, as well as numerous plant-derived compounds multigeneration and some other Tier II tests in order to
such as genistein, daidzein, naringenin, genistin, daidzin, enhance their sensitivity to endocrine disruptors.
glycitein, and puerarin (Boue et al., 2003) and resveratrol, a The Tier I assays included in vitro, in vivo mammalian, and
constituent of red wine (Klinge et al., 2003). Finally, exposure in vivo nonmammalian screening assays. EDSTAC noted that
to synthetic and plant-derived estrogens occurs against a EACs may alter endocrine function by affecting the availability
background of endogenous estrogens, such as estradiol-17b of a hormone at a target tissue (e.g., synthesis, transport,
(E2), estrone (E1), and estriol (E3), the levels of which in metabolism, HPG/hypothalamic-pituitary-thyroid [HPT] axis
blood and tissues can vary dramatically depending on gender, function, etc.) or by affecting the cellular response to a hormone
age, and physiological state (e.g., pregnancy, pre-, or post- (e.g., receptor binding). The original Tier I battery proposed by
menopause). EDSTAC (EDSTAC, 1998) included the following: (1) ER
Beyond need to cover so many different chemistries and binding/transactivation assay, (2) androgen receptor (AR)
sources, the challenge laid before EDSTAC was further binding/transactivation assay, (3) steroidogenesis assay with
complicated by the desire to screen for both receptor agonists minced testis, (4) rodent 3-day uterotrophic assay (sc route),
S96 MARTY, CARNEY, AND ROWLANDS

TABLE 1 a positive response (i.e., if a compound achieves a PC10, which


U.S. EPA EDSP Tier I Assays is 10% of the positive control response at 1nM 17b-estradiol)
may result in numerous false-positive results and jeopardize the
predictiveness of this assay.
In vitro assays
ER binding—rat uterine cytosol
AR binding using rat prostate cytosolic preparations also is
ER—(hERa) transcriptional activation—human cell line (HeLa-9903) included in Tier I (OPPTS 890.1150), although an AR
AR binding—rat prostate cytosol transactivation assay for use in Tier I screening is still being
Steroidogenesis—human cell line (H295R) developed. The AR binding assay shares many of the same
Aromatase—human recombinant microsomes advantages and limitations as the ER binding assay.
In vivo assays
Uterotrophic (rat)
To detect compounds affecting steroidogenesis, the minced
testis assay was originally proposed. This assay was designed

Downloaded from https://academic.oup.com/toxsci/article/120/suppl_1/S93/1615160 by guest on 03 January 2021


Hershberger (rat)
Pubertal female (rat) to detect enzyme inhibition in the steroid biosynthesis pathway
Pubertal male (rat) by measuring decreased testosterone production; however, the
Amphibian metamorphosis (frog) assay was removed from consideration because of high false-
Fish short-term reproduction
positive and false-negative responses and difficulty assessing
Note.http://www.epa.gov/scipoly/oscpendo/pubs/assayvalidation/ Leydig cell cytotoxicity in the mixed population of cells
tier1battery.htm#assays. present in minced testis preparations (Powlin et al., 1998). The
steroidogenesis assay was replaced with a cell-based system
(5) rodent 20-day pubertal female assay with thyroid, (6) rodent that uses a human cell line, the H295R cells (Hecker et al.,
5- to 7-day Hershberger assay, (7) frog metamorphosis assay, 2006, 2008). This assay is designed to measure production of
and (8) fish gonadal recrudescence assay. In addition, four testosterone and 17b-estradiol by the H295R cells in the
alternate assays were identified: (1) placental aromatase assay, presence of varying concentrations of test compound to
(2) modified rodent 3-day uterotrophic (ip), (3) rodent 15-day identify alterations in steroid biosynthesis. Because of the
intact adult male assay with thyroid, and (4) rodent 20-day limited metabolic capacity of these cells, the assay primarily
thyroid/pubertal male assay. These alternative assays, if examines the ability of the parent material to interfere with
validated, could be used to replace other Tier I assays, namely, steroidogenesis. A positive result occurs if there is a significant
either the intact adult male assay or the pubertal male assay difference in steroid production in the presence of less than
would replace the pubertal female and Hershberger assays. 20% cytotoxicity.
Per the Food Quality Protection Act (1996), the endocrine Aside from steroidogenesis alterations, a separate in vitro
screening program was required to use ‘‘appropriate validated assay was developed specifically to identify compounds
test systems’’; consequently, the EPA launched the endocrine affecting aromatase (CYP19), the enzyme that converts
disruptor screening program (EDSP) validation program. androgens to estrogens. The aromatase assay examines effects
Based on results of methods development and validation work, of the parent compound on recombinant human aromatase
some of the proposed Tier I in vitro assays were adopted, activity by measuring the production of heavy water (3H2O)
whereas others have undergone some modifications. The final during the conversion of 3H-androstenedione to estrone.
list of EPA Tier I EDSP screening assays appears in Table 1. Compounds that competitively inhibit aromatase activity will
Both the ER binding and ER transactivation assays have been decrease estrone production and consequently 3H2O levels in
adopted into Tier I. The ER binding assay measures the ability a dose-related manner.
of a radiolabeled ligand ([3H]-17b-estradiol) to bind to rat Interlaboratory validation has been conducted with these
uterine cytosolic ER in the presence of increasing test material Tier I in vitro assays to verify each assay’s ability to detect
concentrations. A compound is positive for ER binding if it known EACs operating via relevant MOAs, although the size
effectively competes with [3H]-estradiol binding in a concentration- of the validation programs (i.e., number of test compounds,
related manner, and an inhibitory concentration that decreases number of laboratories, etc.) has differed by assay. One issue
radioligand binding by 50% (IC50) is obtained. The ER with the in vitro EDSP assays is that the assays have limited
competitive binding assay cannot distinguish agonists from ability to metabolize substances; thus, if a metabolite is the
antagonists. Test guidelines also were issued for the Stably EAC, it may not be detected by these assays. Options to
Transfected ER Transcriptional Activation assay using HeLa- improve metabolic capacity are under consideration. Although
9903 cells (Organization for Economic Co-operation and in vitro assays provide useful information on potential MOA
Development [OECD] TG 455; OPPTS 890.1300), although and can help clarify in vivo assay results, previous studies have
detection of agonists is all that is required. Although having suggested that in vivo data should carry more weight than
limited metabolic capability, the ER transactivation assay in vitro data when determining the need for Tier II testing (e.g.,
reportedly has good concordance with in vitro ER binding and Charles et al., 2005).
in vivo uterotrophic responses for active parent compounds The uterotrophic assay went through an extensive validation
(Takeyoshi, 2006); however, the test guideline criteria for program, which was led by the OECD Endocrine Disruptor
ENDOCRINE-MEDIATED TOXICITY S97

Testing and Assessment (EDTA) Task Force and the however, the weanling model did not detect weak antiandrogens
Validation Management Group (VMG)—Mammalian. This as consistently as the castrated adult model (Freyberger and
validation process, led by Japanese representatives on the Schladt, 2009) and therefore was not included in either the EPA
EDTA, included a detailed background document prepared by or the OECD test guideline.
the OECD (2003) and multiphase intra- and interlaboratory Initially, the 20-day female pubertal assay was recommen-
validation studies involving approximately 20 laboratories to ded, and the 20-day male pubertal assay was an alternate EDSP
show transferability, reproducibility, and reliability. This screen. Prior to initiation of the interlaboratory validation work,
validation program tested a potent estrogen (ethinyl estradiol), the exposure period for the pubertal male assay was increased
weak estrogen agonists in both a coded (blinded) and dose- from 20 to 30 days, and ultimately, both assays were adopted
response manner to evaluate sensitivity, and negative into the Tier I EDSP battery. One of the strengths of the male
compounds to verify specificity. This work culminated in and female pubertal assays is that these assays look for changes

Downloaded from https://academic.oup.com/toxsci/article/120/suppl_1/S93/1615160 by guest on 03 January 2021


a series of publications (Ashby et al., 2003; Kanno et al., in young animals during a dynamic period of endocrine
2001, 2003a,b; Owens and Köeter, 2003; Owens et al., 2003) activity. The pubertal assays are the most apical of the Tier I
and an OECD test guideline in 2007. The U.S. EPA test in vivo mammalian assays, but these assays are multimodal,
guideline was released in 2009. capable of detecting EACs that operate through a variety of
The assay uses female rats (or mice) with nonfunctional MOAs (i.e., strong and weak (anti)estrogenic and (anti)andro-
HPG axis to yield sensitive and consistent increases in uterine genic compounds, steroid biosynthesis inhibitors, aromatase
weight in response to stimulation by estrogenic materials. inhibitors, 5a-reductase inhibitors, thyroid-active agents, and
Ultimately, the test guideline outlines the application of the compounds affecting the HPG or HPT axis). Some of these
uterotrophic assay for the detection of compounds with MOAs are not detected by other Tier I in vitro or mammalian
estrogenic activity; use of this assay to identify antiestrogens assays; thus, it may be difficult to dismiss a positive result in
is supported by the test guideline, but not required. Overall, the the pubertal assays even if there is some suspicion that the
uterotrophic assay appears to perform well for the identification primary MOA is not endocrine mediated.
of estrogenic compounds, although the ovariectomized model One limitation of the pubertal assays is that it may be
has been recognized as more specific for identification of difficult to determine the MOA for potential EACs. Some
estrogenic activity than the immature model. The immature endpoints assessed in these assays (puberty onset, estrous
model, which has an intact HPG axis, can detect aromatizable cycle, and organ weights) can be altered by chemicals that
androgens and is more sensitive to body weight–mediated operate via an endocrine MOA or by nonspecific/systemic
changes in uterine weights. The alternate uterotrophic assay, toxicity. During the validation program, some compounds were
which used ip dosing, was not included in the validation not identified for their primary MOA. For example, phenobar-
because of potential localized effects of the test compounds on bital, which is a weak thyroid-active compound, did not alter
uterine tissue. thyroid weights, thyroid histopathology, or serum T4 or thyroid
The Hershberger assay also underwent an extensive multi- stimulating hormone levels in the male pubertal assay during
laboratory validation process, led by U.S. EPA through the the validation program but rather showed signs of antiandro-
OECD EDTA VMG—mammalian group (Owens et al., 2006, genicity, which included delayed preputial separation and
2007). The Hershberger assay uses castrated adult male rats decreased reproductive and AST weights (U.S. EPA, 2007).
with a nonfunctional HPG axis. Although originally proposed There remains some question regarding the specificity of the
with a 5- to 7-day exposure period, rats are treated with the pubertal assays. The Interagency Coordinating Committee on
test compound for 10 days by oral gavage or sc dosing in the Validation of Alternative Methods defines specificity as the
the adopted version of the assay. Increases in accessory sex proportion of inactive substances that are correctly identified.
tissue (AST) weights (i.e., glans penis, ventral prostate, During assay validation, 2-chloronitrobenzene (2-CNB) was
seminal vesicles with fluid, Cowper’s glands, and levator selected as a negative control chemical for the male and female
ani-bulbocavernosus muscles) indicate potential androgenicity. pubertal assays; however, 2-CNB caused thyroid effects,
To evaluate antiandrogenic potential, rats are given the delayed vaginal opening, and altered adrenal and uterine
test compound (oral gavage or sc) concurrent with 0.2 or weights in females, and delayed preputial separation, decreased
0.4 mg/kg/day testosterone propionate sc. If the test compound serum testosterone levels, decreased androgen-dependent organ
inhibits the testosterone-induced increase in AST weights, then weights, and had testis histopathological effects in males. The
the compound has potential antiandrogenic activity or is EPA selected hydroxyatrazine as an alternate negative control
a potential 5a-reductase inhibitor. Two or more organ weights compound and recently reported that it was negative in both the
must be significantly altered to yield a positive result. Overall, male and the female pubertal assays (Stoker and Zorrilla,
the Hershberger assay appears to perform well for the 2010). Only a subset of these data has been published. Once
identification of potential androgens, antiandrogens, and available, the full data set is likely to provide additional
5a-reductase inhibitors. During the Hershberger validation information on setting maximum tolerated dose levels and
studies, a weanling (non-castrated) animal model was included; interpreting pubertal assay results (i.e., provide perspective for
S98 MARTY, CARNEY, AND ROWLANDS

hydroxyatrazine results reported in Laws et al., 2003). With (i.e., advanced, delayed, or asynchronous development), thyroid
few negative compounds included in the pubertal assay histopathology is an important endpoint to confirm a specific
validation, it is unclear whether the male and female pubertal thyroid effect. To facilitate interpretation, laboratories are
assays are prone to nonspecific effects, which will produce encouraged to include positive control compounds to verify
a high frequency of false-positive results. assay performance and build historical data. Thus, the
One alternative assay proposed as part of the EDSP was the amphibian metamorphosis data should be considered in
15-day intact adult male rat assay. Approximately 30 EACs, a weight-of-evidence approach to determine potential thyroid
across a variety of MOAs, have been evaluated using the activity.
15-day intact male assay including estrogen agonists/antagonists, Originally, the fish gonadal recrudescence assay was
androgen agonists/antagonists, steroid biosynthesis inhibitors, proposed as an EDSP Tier I screening assay. In this assay,
thyroid-active compounds, and prolactin modulators. The fish were examined for compound effects on maturation from

Downloaded from https://academic.oup.com/toxsci/article/120/suppl_1/S93/1615160 by guest on 03 January 2021


validation program also examined assay specificity by evaluating a regressed state (recrudescence) in response to an increased
the impact of body weight effects on assay endpoints (O’Connor photoperiod and temperature regime. Endpoints included
et al., 2000) and including a negative control compound, allyl secondary sex characteristics, ovary and testis weight increases,
alcohol (O’Connor et al., 2008). The intact male assay was gonadosomatic index, gamete maturation, and induction of
specifically designed as an MOA screening study, whereby vitellogenin. However, feasibility studies indicated that there
researchers could compare the ‘‘fingerprint’’ of the organ weight, was high variability in the gonadal recrudescence response in
histopathology, and hormonal endpoints for an unknown continuous spawning species like the fathead minnow (Touart,
compound with data generated for a series of positive controls 2005); thus, the recrudescence assay was not included in the
to help characterize the MOA (O’Connor et al., 2002). There was EDSP. Instead, the U.S. EPA has opted for the 21-day fish
some concern regarding the use of numerous serum hormone reproduction screen (OPPTS 890.1350) in the Tier I battery as
measurements in this assay as serum hormone levels are opposed to the OECD 21-day fish assay (OECD TG 230). This
inherently variable. However, data developed during the screen uses a minimum of 96 reproductively mature adult
validation process have shown that serum hormone measure- fathead minnows (alternate models allowed per OECD)
ments are reliable with the group sizes employed (n ¼ 15 per exposed continuously for 21 days to a minimum of three test
dose). Of greater concern for the U.S. EPA was the inability of concentrations and a control. The EPA 21-day fish reproduc-
the intact male assay to reliably detect weak antiandrogens tion screen includes additional parameters not measured in the
(O’Connor et al., 1999), which would require that the assay be OECD 21-day fish screen, including fecundity, fertility,
paired with an alternative assay (e.g., Hershberger assay) for Tier gonadal histopathology, and sex steroid concentrations. In
I screening. Because of this weakness, the 15-day intact male addition, adult survival, secondary sex characteristics (such as
assay was not included in the EPA EDSP. coloration patterns and tubercle scores), reproductive behavior,
In addition to the in vitro and in vivo mammalian assays, two vitellogenin, and gonadal-somatic index (GSI) are evaluated.
nonmammalian assays also are included in the Tier I EDSP: Secondary sex characteristics, vitellogenin, and plasma sex
the amphibian metamorphosis assay and the 21-day fish steroids are more predictive of an endocrine MOA, although
reproduction screen. The amphibian metamorphosis assay the high variability seen in plasma sex steroids makes them less
(OPPTS 890.1100) provides an environmental screen to detect robust and a lower priority than vitellogenin when blood
compounds that have the potential to interfere with the HPT plasma quantities are limited. In contrast, fecundity is an
axis. Xenopus laevis tadpoles are exposed for 21 days starting indicator of general reproductive condition and not endocrine
at Nieuwkoop and Faber stage 51 (Nieuwkoop and Faber, specific. Thus, evaluations in the fish reproduction screen are
1994) and examined for mortality, developmental stage, hind a combination of endocrine-specific endpoints and apical
limb length, snout-vent length, wet weight, and thyroid endpoints that can be altered by non-endocrine MOAs. With
histopathology. Because the rate of metamorphic development these apical endpoints and the variability in some of the
is controlled by thyroid hormone in X. laevis, developmental observations, weight of the evidence will be employed in
stage (which is determined by the inspection of various determining the potential of the compound to interact with the
morphological changes associated with metamorphosis) and endocrine system of fathead minnow.
hind limb length (which is normalized by snout-vent length) are Both environmental screens must manage issues related to
used to determine if development is accelerated, asynchronous, the physicochemical properties of various test materials, the
delayed, or unaffected by the test compound after either 7 or need to develop a solvent-free test system (if possible),
21 days of exposure. Many of the endpoints assessed in this development of analytical methods for dose verification, and
assay are apical and can be altered by MOAs that are not the need to generate range-finding data to assist in dose
specific for thyroid effects. Some endpoints (e.g., snout-vent selection. Appropriate dose setting is critical to differentiate
length and wet weight) provide information on whether the systemic toxicity from genuine endocrine-mediated effects.
test compound alters growth and/or causes systemic toxicity. In 2009, the U.S. EPA released its first list of priority
If any changes in developmental stage are observed compounds for endocrine screening. The initial priority list
ENDOCRINE-MEDIATED TOXICITY S99

consisted of 67 chemicals, primarily pesticide-active ingre-


dients (58) and some pesticide inerts (9). For pesticide-active
ingredients, substantial hazard data are available because of
registration requirements. As part of the EPA EDSP program,
companies were allowed to submit other scientifically relevant
information (OSRI), describing available toxicity data for
a compound and known health risks with an emphasis on
endocrine-related effects. OSRI information for many of the
priority compounds is currently under review by the EPA;
however, it is not clear what information will be viewed as
‘‘functionally equivalent’’ to the Tier I assays. If functionally

Downloaded from https://academic.oup.com/toxsci/article/120/suppl_1/S93/1615160 by guest on 03 January 2021


equivalent data are available for a compound, companies may
be allowed to omit one or more of the Tier I EDSP assays
based on these existing data.
The goal of the Tier I EDSP is to identify substances that have
the potential to interact with the estrogen, androgen, or thyroid
hormone system for further testing in Tier II. Given that there are
apical endpoints in Tier I assays that may be altered by nonspecific
effects and there is some redundancy to detect endocrine MOAs
across different Tier I assays, it is beneficial to use a weight-of-
evidence approach to determine whether there is sufficient data to FIG. 1. This diagram illustrates some of the redundancy in Tier I EDSP
warrant Tier II testing (Fig. 1). Previous toxicity data generated assays. In a weight-of-evidence approach, a pattern of effects across related
with standardized test guidelines and/or validated methods should assays is easily interpreted (e.g., positive ER binding, ER transactivation, and
be included in this weight-of-evidence analysis. uterotrophic assays coupled with estrogenic responses in the pubertal and fish
short-term reproduction assays). However, in many cases, the pattern will not
One example of applying weight of evidence can be
be as easily discerned. If results of in vitro and in vivo assays disagree, weight
illustrated with methoxychlor, an estrogenic compound of evidence requires careful consideration of the in vivo results and previous
included in validation studies for several of the Tier I EDSP toxicity data when determining whether the compound is a potential EAC and
screening assays. It is recognized that methoxychlor is warrants Tier II testing. Aside from the expected MOA relationships depicted
metabolized in vivo to an estrogenic metabolite, 2,2-bis in this figure, other, more complex MOA relationships are not shown (e.g., the
ability to detect agents with mixed MOAs like methoxychlor or HPG axis
(p-hydroxyphenyl)-1,1,1-trichloroethane (HPTE). Thus, me-
compounds like atrazine).
thoxychlor was not included as part of the estrogen binding
assay validation work; however, other binding assays using
competitive binding to uterine cytosol ERs were negative for validation studies can be located online (http://www.epa.gov/
methoxychlor (e.g., Shelby et al., 1996). Methoxychlor was scipoly/oscpendo/).
included in the ER transactivation assay validation, wherein its Based on a weight-of-evidence approach, compounds
estimated EC50 was 1 3 105M. The authors concluded that deemed positive in Tier I will undergo Tier II testing to
methoxychlor exhibited a response curve that was consistent characterize hazards and develop data for risk assessment. The
with other weak estrogens. Methoxychlor was positive in some specific tests that will be included in Tier II have not been
AR binding assays (Charles et al., 2005), but it was negative in definitively identified. Possible tests under consideration for
the aromatase assay. In vivo, methoxychlor was positive in the inclusion in Tier II include the two-generation mammalian (rat)
uterotrophic assay (Kanno et al., 2003a,b), although it was reproductive toxicity study, the two-generation amphibian
more readily detected with oral exposure than sc injection and (frog) reproductive toxicity study, the two-generation avian
sc injection is the preferred route for administration in the EPA reproductive toxicity study, fish life cycle toxicity study, and
uterotrophic test guideline. Methoxychlor was negative in the the invertebrate (mysid) life cycle toxicity study.
Hershberger assay at doses that caused a significant increase in
liver weights (Charles et al., 2005) and positive in both the
male and the female pubertal assays. The estrogenic properties LOW-DOSE ISSUE AND MIXTURES
of methoxychlor also were identified in the fish reproduc-
tion assay. There were no data on methoxychlor’s effects in Concern over endocrine disruptors has been further
the steroidogenesis or amphibian metamorphosis assays. Based heightened because of reports that such compounds can elicit
on a weight-of-evidence approach, methoxychlor would be unique low-dose effects and exhibit nonmonotonic dose-
identified as an estrogenic compound with possibly a mixed response relationships that may not be detected in regulatory
mode of action that affects androgen-dependent endpoints. toxicity studies, which are typically conducted at higher dose
Except where indicated by other citations, results of these levels. The low-dose hypothesis has been a contentious issue
S100 MARTY, CARNEY, AND ROWLANDS

among some toxicologists. The National Toxicology Program relevant dose levels. Although risk assessment approaches for
(NTP), with support from the U.S. EPA, organized a panel to cumulative and aggregate risk have been in place for many
evaluate the scientific evidence on the low-dose effects of years (reviewed in Lipscomb et al., 2010), interest in
endocrine disruptors (Melnick et al., 2002), defining low-dose cumulative exposures to multiple EACs has received increased
effects as ‘‘biologic changes that occur in the range of human attention in recent years (Carney et al., 2010; Hotchkiss et al.,
exposures or at doses lower than those typically used in the 2008; Kortenkamp, 2008). Mixtures have been reported to
standard testing paradigm of the U.S. EPA for evaluating have additive effects (e.g., Gennings et al., 2004), less than
reproductive and developmental toxicity.’’ The panel additive effects (e.g., Charles et al., 2007), greater than additive
concluded that (1) there was evidence for low-dose effects in effects (e.g., Laetz et al., 2009), or even counteracting effects
laboratory animals, although in some cases, these effects have (e.g., Tanida et al., 2009). Overall, data indicate that the
not been replicated; (2) the shape of the dose-response curve interactions of chemicals in mixtures are dose dependent.

Downloaded from https://academic.oup.com/toxsci/article/120/suppl_1/S93/1615160 by guest on 03 January 2021


depended on the endpoint and dosing regimen used; and (3) the Using mixture data in the context of environmental risk
current reproductive/developmental toxicity testing paradigm assessment is even more complex when one considers natural
should be reviewed and possibly revised with respect to dose exposures to EACs in food and pharmaceuticals (Gaido et al.,
selection, animal models used, ages evaluated, and endpoints 1998; Safe et al., 1997). Exposures to environmental chemicals
measured. Some reasons identified for the inability to typically involve low doses of each chemical within a mixture
reproduce low-dose effects across studies included intrauterine and, given the safety factors used in risk assessment, exposures
position, strain and substrain differences in response, dietary generally below the threshold for toxicity. Thus, mixtures
levels of phytoestrogens or differences in caloric intake, research requires a focus on the behavior of mixtures at
differences in housing conditions (caging, bedding, etc.), and environmentally relevant exposures because results obtained at
seasonal variation. The statistics subgroup (Haseman et al., high-dose levels may not apply in real-world scenarios. This
2001) also identified a number of important factors that should concept also was espoused by the Expert Working Group on
be considered in the design, analysis, and interpretation of Mixtures Toxicology, which was convened by the Society of
experimental studies to help resolve low-dose issues, including Toxicology and the Society for Environmental Toxicology and
study sensitivity (power), reproducibility, control for litter Chemistry (Teuschler et al., 2002).
effect, quality control in data collection and management,
procedures for data analysis, interpretation of the significance
of biological changes, and data selectivity. Ultimately, the U.S. HISTORICAL DEVELOPMENTS IN THE BASIC SCIENCE
EPA concluded, ‘‘Until there is an improved scientific OF ENDOCRINE SIGNALING
understanding of the low-dose hypothesis, EPA believes that
it would be premature to require routine testing of substances When the history of endocrinology is written, it is likely that
for low-dose effects in the Endocrine Disruptor Screening the 20th century will be neatly divided into two epochs with the
Program’’ (U.S. EPA, 2002). first half being the era of biochemistry when the hormones were
Following the NTP low-dose report, a number of key discovered and the emphasis was on proteins and the second half
studies by Naciff et al. (2002, 2009, 2010) have examined of the 20th century, the era of genomics when the genes for the
changes in expression of up to 38,500 genes in various cell hormone and nuclear receptors (NRs) were identified. The word
lines or uterine tissues exposed to very low doses of EACs, hormone was first coined in 1905, but it was not until 1926 that
including BPA at concentrations down to 1nM. These studies the first hormones were characterized in detail when Kendeall
collectively provide support for monotonic dose-responses for and Reichstein isolated and determined the structures of
these compounds and showed negligible changes in gene cortisone and thyroxine (The Nobel Assembly at Karolinska
expression at very low-dose levels. Furthermore, several Institutet, 1950). Soon thereafter, in 1929, Adolf Butenandt and
comprehensive weight-of-evidence reviews for BPA have Edward Adelbert Doisy independently isolated and determined
concluded that nonmonotonic dose-response curves and low- the structure of estrogen (Tata, 2005). These discoveries along
dose effects from BPA are not well supported by available with other experimental observations allowed Clark to propose
data (Goodman et al., 2009; Kamrin, 2004, 2007). The Center his model for receptor-mediated processes in his publication of
for the Evaluation of Risks to Human Reproduction (Chapin the theory of occupancy. The receptor theory has been refined
et al., 2008) cited many of the issues related to using low-dose since the 1930’s along with the vocabulary used to classify
study data when evaluating BPA. Despite these reports, the chemicals that bind to NRs or ligands, which are now classified
Endocrine Society issued a scientific statement on endocrine- into agonist, antagonist, partial agonist, and inverse agonist
disrupting chemicals that included support of the low-dose based on the biological effects elicited through NRs (Table 2).
hypothesis (Diamanti-Kandarakis et al., 2009); thus, the Potential EAC’s may produce their effects through inter-
debate continues. actions with NRs. Under normal physiological conditions, the
The low-dose controversy has evolved to consider issues intracellular NRs are bound by their endogenous signaling
related to exposures to mixtures of EACs at low, environmentally molecules, or receptor ligands, such as the steroid hormones,
ENDOCRINE-MEDIATED TOXICITY S101

TABLE 2 conditional and specific to a given gene, in a specific cell and


Properties of Chemicals that Affect NRs tissue under specific conditions. For example, a given ligand
may be an agonist for a gene in a cell and at the same time be
Ligand Chemical that binds reversibly to an NR an antagonist for a different gene in the same cell; or a ligand
can be an agonist for a gene in one cell type and an antagonist
Affinity A ligand’s capacity to bind to an NR
Agonists Ligand that produce an NR conformation for the same gene in a different cell type or even the same cell
that is transcriptionally active type but under different conditions. Potency is a conditional
Antagonists Ligands that prevent agonist binding or induce measure that depends on parameters of affinity and efficacy, as
an NR conformation resulting in an action well as tissue receptor numbers and ligand bioavailability.
distinct from that produced by an agonist
Thus, the intrinsic efficacy of an EAC, whether agonist,
Partial agonists Ligands that possess weak agonist activity
antagonist, partial agonist/antagonist, or inverse agonist for

Downloaded from https://academic.oup.com/toxsci/article/120/suppl_1/S93/1615160 by guest on 03 January 2021


Inverse agonists Ligands that induce an NR conformation that
results in transcriptional repression a given NR, is dependent upon both chemical properties of the
Efficacy A measure of the capacity of the ligand to EAC and the biological environment of the NR (Jordan, 2003).
activate, prevent activation, or inactivate an NR These differences in the properties of ligands are often lost in
Potency A measure of the concentration or amount of
the discussion of EACs. An EAC’s potency is dependent on its
ligand needed to produce an effect through an NR
affinity for an NR, and some putative EACs have ER binding
affinities that are 10,000–100,000 lower than ER binding
thyroid hormones, retinoids, vitamin D, and other as yet affinities of potent, natural, or contraceptive estrogens
identified endogenous ligands. These receptor ligands either (Gutendorf and Westendorf, 2001). For example, the affinity
diffuse directly across the plasma membrane of cells or are of BPA for ERs is approximately 10,000-fold weaker than that
sometimes produced in the same cells and bind to the NR. The of estradiol (Kuiper et al., 1998). What this means in practical
commonly understood mechanism of action for NRs involves terms is that a cell would need to be exposed to 10,000
the initial ligand binding that results in receptor activation, molecules of BPA to produce the same effect through the ER
binding at specific DNA response elements in nuclear that one molecule of estradiol would produce through that same
chromatin, followed by recruitment of a variety of cofactors receptor. Furthermore, the amount of BPA that one would need
that modify the chromosomal organization and interaction with to consume, breath, or absorb through the skin would be even
the transcription complex, thereby altering the rates of greater because the absorption, distribution metabolism, and
transcription of genes. So, binding of a ligand to an NR is elimination of the BPA would limit the amount of BPA
only the first step in a complex cascade of events leading to available to the cells. Even if an EAC was highly potent in one
changes in gene expression and subsequent cell signaling. With cell type, it does not mean it will have the same efficacy in
the completion of the human genome, it is now clear that there another cell type or the same cell type under different
are some 48 NRs expressed in humans, of which only half have conditions as efficacy is independent of ligand affinity and
identified ligands such as steroid hormone receptors that instead reflects a ligand’s ability to induce a stable coactivator
constitute only a minority of this large superfamily of receptors binding site on the NR (discussed further below). Thus, two
(Gronemeyer et al., 2004). ligands can have similar potencies for inducing a response
Like the NR superfamily, coregulators are themselves a large through an NR, but the efficacy for these two ligands inducing
and mixed family of proteins and possess a varied array of the response can vary greatly and this can have a profound
enzymatic activities necessary for chromosomal maintenance impact on the ability of these ligands to produce the same
and gene regulation (Roeder, 2005). They can be categorized apical effects through the NR. The lower potencies and
based upon their wide-ranging functional properties including different efficacies of EACs relative to the natural hormones
acetyltransferases, ubiquitin ligases, ATP-coupled chromatin need to be understood and considered in the discussions of the
remodeling complexes, protein methylases, cell cycle regu- potential impact of EACs on human health and the environment.
lators, RNA helicases, and bridging proteins that facilitate It took another three decades after the elucidation of the
direct contact with components of the basal transcription structure of estrogen before the ER was isolated in 1961 by
machinery (McKenna and O’Malley, 2003). There are Elwood Jensen (Tata, 2005) and another two decades passed
currently 285 coregulators identified and many are expressed before the new tools of molecular biology allowed Pierre
differentially by cell type, tissue type, gender, developmental Chambon’s group to clone the gene for the human ER (Green
stage, and species (Lonard et al., 2007; Smith and O’Malley, et al., 1986). The 1980’s ushered in an era of rapid discovery of
2004). Moreover, coregulators can be differentially recruited to the receptor genes for most of the major hormones, which was
the same receptor by ligand-induced conformational changes in followed in the 1990’s by the cloning of novel nuclear orphan
the NR. Thus, with 48 NRs and 285 plus coregulators, the receptors for which there are no known ligands. It was also
complexity of these endocrine signaling pathways is apparent. during this period, in 1996, that Jan-Åke Gustaffson’s group
It is important to note that all these concepts of agonists, cloned a second ER, now called ERb (Kuiper et al., 1996),
antagonists, partial agonists/antagonists, or inverse agonists are requiring the observations of the previous decades of estrogen
S102 MARTY, CARNEY, AND ROWLANDS

studies to be viewed through a new lens. No doubt there will be


additional chemicals identified as ligands for some of the 48
NRs identified in humans, thereby necessitating an under-
standing of their potential for behaving as EACs.

ESTROGENS: FACTORS AFFECTING AFFINITY, POTENCY,


AND EFFICACY

One of the best understood examples of an endocrine


signaling system that brings all these concepts together is that

Downloaded from https://academic.oup.com/toxsci/article/120/suppl_1/S93/1615160 by guest on 03 January 2021


of estrogens. A half-century has passed since the initial reports
of a cognate ER, and it is now known that ERa and ERb
exist, as well as less well-understood additional subtypes. The
distribution of these two receptors is variable, with tissues such
as bone having predominantly the b isoform, uterus containing
mainly a, but many tissues expressing both a and b.
Furthermore, a and b often regulate opposing functions
providing a feedback loop for finite control of estrogen
signaling. The biological effects elicited by specific ER ligands
are highly cell and tissue specific. One of the best known FIG. 2. The actions of nuclear receptors are ligand dependent and cell
context dependent. Nuclear receptors (R) form a different tertiary structure
examples of this phenomenon is the drug tamoxifen, which has depending on the ligand that activates them (EAC-1 or EAC-2). This in turn
been used for treatment of early stage breast cancer and as causes the receptors to recruit different coregulator complexes, e.g., coactivator
a chemoprevention agent for women at high risk for this complexes or corepressor complexes that lead to gene induction or repression,
disease (Jordan, 2003). Tamoxifen is an ER antagonist in breast respectively. In Cell A, the two different ligands (EAC-1 and EAC-2) bind to
cancer but an ER agonist in the bone and uterus (Fong et al., the same receptor but induce two different tertiary structures, which in turn
cause the receptor to recruit a coactivator complex in the case of EAC-1 bound
2007). Another example is the phytoestrogen resveratrol, receptor or a corepressor complex in the case of EAC-2 bound receptor.
which shows more activity in cells that uniquely express Whereas in Cell B, the opposite occurs with EAC-1 producing a corepressor
ERb or that express higher levels of ERb than ERa and also complex recruitment through the receptor, but EAC-2 produces a coactivator
appears to exhibit estrogen antagonist activity for ERa in complex recruitment through the receptor. Thus, an individual EAC can act as
certain promoter contexts (Bowers et al., 2000). In addition, only an agonist, antagonist, partial agonist/antagonist, or inverse agonist depending
on the coregulators present in the specific cell. Although not shown, the gene
a small percentage of ER ligands are pure estrogen agonists or architecture influences nuclear receptor-coregulator recruitment as well.
antagonists. Most phytoestrogens and xenoestrogens are partial Coactivator (coactivator complex); corepressor (corepressor complex); TBP,
agonists, meaning that they are unable to induce a maximal A, B, F, pol II (RNA polymerase II holoenzyme: RNA polymerase II and
response matching that of the endogenous ligand, even at very general transcription factors); TATA (TATA box promoter site); transcription
high concentrations (Zhu, 2005). The latter compounds tend to (RNA transcript).
act as agonists under some conditions but act as antagonists
when their concentration is very high relative to more potent positive or negative stimuli from the same hormone ligand or
ligands. The research from numerous laboratories over the last different receptor ligands depending on the cell-specific
five decades has revealed the explanations for many of these coregulator availability and/or ligand-specific induced receptor
observations. conformations with differential coregulator affinities (Fig. 2).
Differences in the tissue-specific receptor agonist and Selective receptor modulators (SRMs) have been observed for
antagonist activities have long been recognized for the steroid other NRs including peroxisome proliferator–activated recep-
hormone receptors and form the basis for the development of tors, ARs, progesterone receptors, as well as the AhR
drugs for treatment of hormone-dependent diseases (Ai et al., (Gronemeyer et al., 2004; Zhang et al., 2008). Thus, SRM
2009; Nettles and Greene, 2005). Tamoxifen’s tissue-specific appears to be a common feature of ligand-activated transcrip-
activity is now thought to be because of recruitment of different tion factors that explains the observed selectivity that different
coregulators to the ER by tamoxifen in breast tissue versus ligands confer to NRs on modulation of gene expression and
uterine and bone tissue (Gronemeyer et al., 2004). Tamoxifen the resultant tissue changes both within and across tissue and
was the first of what are now called selective ER modulators or cell types (Ai et al., 2009; Zhang et al., 2008). No doubt, EACs
SERMs where the cellular response to SERMs is determined will share similar characteristics with SRMs.
by cell type– and promoter-specific differences in coregulator All the discoveries discussed above and more have been
recruitment by the ER. Thus, coregulators extend the actions of harnessed to develop in vitro and in vivo assays to identify
the same hormone receptor by producing cell type–specific chemicals with endocrine-modulating activities. The very
ENDOCRINE-MEDIATED TOXICITY S103

nature of an NR being a trans-acting transcriptional regulator effect obtained for one response to another response is
allows for their exogenous expression in numerous cell types questionable. Moreover, the problem of confounding is even
and cell lines to study the effects of potential ligands as more problematic when considering that measured parameters of
agonists, antagonists, etc. These studies have led to the in vitro high-throughput screening systems are by nature dif-
adoption of some assays into the EDSP battery of tests and ferent from those found for receptor systems in vivo. Therefore,
many others that are used in the ToxCast panel of assays. The the results from these in vitro screening systems may seldom
cloning of these receptors has also facilitated the development accurately reflect the intrinsic efficacies of NR ligands in vivo
of NR knockout (KO) mouse models that provided important and challenge the validity for extrapolating observations to
confirmatory evidence for the physiological role of hormones humans. Even if a single in vitro system was reflective of
in development, reproduction, and normal physiology as well a human response under certain conditions, the fact that the
as some unexpected observations. intrinsic efficacy of an EAC is dependent upon the cellular

Downloaded from https://academic.oup.com/toxsci/article/120/suppl_1/S93/1615160 by guest on 03 January 2021


With the advent of the KO models, it was possible to context of the NR means that multiple systems and or conditions
develop transgenic receptor knock in mouse models that would need to be employed to ensure that all the possible
express human NRs in place of their endogenous mouse conditions in vitro reflect similar conditions in humans in vivo.
receptor. The development of the ER KO mouse (ERKO Although this review only touched briefly on some of the
mouse) by the National Institute of Environmental Health key developments to date concerning endocrine-mediated
Sciences group led by Ken Korach demonstrated that both toxicity and safety assessment, it should be readily apparent
sexes of these mutant animals are infertile and show a variety that the issue has raised more questions and challenges than
of phenotypic changes, some of which are associated with the have been answered. Most likely, we are only at the very
gonads, mammary glands, reproductive tracts, and skeletal beginning of a long journey toward improved understanding of
tissues (Lubahn et al., 1993). The ERKO mice and other the basic mechanisms of endocrine-mediated toxicity and the
hormone receptor KO mouse models can be used to confirm extent to which relevant exposures to hormonally active agents
the endocrine effects of putative EACs and demonstrate impact human and environmental health. As we forge ahead,
important differences in the effects produced by endocrine this active and often controversial topic has already accelerated
chemicals compared with endogenous hormones. By knocking the science of toxicology and risk assessment and has
in the human gene for an NR into the appropriate receptor KO redirected it into some fundamentally different directions.
mouse, new transgenic mouse models are made that allow One such direction is down. This is not meant in a negative
comparison of putative EACs acting through the human sense but refers to the examination of much lower, environ-
receptor or mouse receptor. The observations from such mentally relevant dose levels than have been examined in the
studies are important to consider in the weight of evidence traditional high-dose toxicology paradigm. This new direction
for predicting the effects of EACs to humans. Future mouse was spurred by research, albeit controversial, that hormonally
models are being developed that express the human proteins for active agents might not adhere to the monotonic dose-response
multiple NRs and accessory proteins, and these will allow behavior, which has been a fundamental principle of toxicology
further refinement of the effects of EACs on human pathways. since its inception as a field of science. However, further prog-
In addition, the technology is now available to make KO rats ress in this area is possible thanks to modern technology, such
and other species, and this will be followed by the ability to as toxicogenomics, which allows one to examine for precursor
humanize these models as well, thereby furthering our ability to changes on the cellular and molecular level (Naciff et al., 2009)
understand the human relevance for EACs identified in wild- and thus help to better understand the fundamental biology
type laboratory models and in vitro assays. underlying dose-response for environmentally relevant expo-
sures. In addition, the increased utilization of human, rather than
animal, cells holds promise for enhancing human relevance
FUTURE DIRECTIONS IN ENDOCRINE DISRUPTOR even further.
SCREENING AND TESTING Another research direction points upward, namely, toward
the investigation of higher numbers of chemicals and other
Endocrinology has come far since the term hormones were stressors and their potential cumulative effects. For years,
first introduced in 1905. What started as a simple concept has toxicologists have tended to avoid studying cumulative effects
evolved into a very complex array of hormones, receptors, and because it was considered an intractable problem. This was
cofactors. Because a chemical ligand for an NR can have especially so for hormonally active agents because of the vast
multiple intrinsic efficacies (i.e., induce several NR responses) array of ligands and multiplicity of sources, which includes not
depending upon the cellular and tissue environment of the NR, only man-made chemicals but also compounds in plants,
relying upon in vitro cell and biochemical assays to define EACs endogenous hormones, and even nonchemical stressors.
has important implications. With suspect EACs possessing the However, because of new high-throughput tools, refined
potential for multiple induced NR responses, the validity for statistical methods, and economical experimental designs for
transferring inferences about the efficacy of the EAC-induced mixture studies (Gennings, 1996), questions about combined
S104 MARTY, CARNEY, AND ROWLANDS

exposures to hormonally active agents can be addressed in highlighted the tremendous impact of hepatic first pass
a scientifically rigorous and efficient manner. metabolism in converting these compounds to their
This movement toward evaluating more chemicals is also biologically inactive glucuronide forms (Teeguarden and
being combined with a fundamental change toward mechanism- Barton, 2004; Teeguarden et al., 2005). Therefore, route of
based testing (inside out) versus the traditional adverse effects- exposure can have a dramatic effect on the percentage of free,
based system (outside in) as described in the landmark report biologically active compound and consequently the effects
by the U.S. NRC Committee on Toxicity Testing and 1365 produced. The key point is that as we delve deeper into the
Assessment of Environmental Agents, Board of Environmental details of individual mechanisms, it is essential that these new
Studies and Toxicology, Institute for Laboratory Animal findings be interpreted in a holistic, physiological framework.
Research (2007) report entitled ‘‘Toxicity Testing in the 21st Fortunately, new tools in exposure modeling, physiologically
Century: A Vision and a Strategy.’’ Within this report, based pharmacokinetic modeling, and systems biology are

Downloaded from https://academic.oup.com/toxsci/article/120/suppl_1/S93/1615160 by guest on 03 January 2021


endocrine screening is highlighted as an area of toxicological increasingly available to assist with experimental design, data
concern that could benefit from the use of molecular-based integration, and data interpretation to achieve this goal.
screening methods. Progress has been made toward implementing
this approach. EPA’s ToxCast program contains numerous CONCLUSION
endocrine-related screening assays, including ER and AR
receptor binding assays, ER, AR, and thyroid receptor reporter Concerns about EACs have come to the forefront of
gene assays, an assay for thyrotropin-releasing hormone toxicology over the past 15 years. The precise impact of
receptor, and an aromatase enzymatic activity assay (Houck, EACs is difficult to discern as EACs can be beneficial or
2009). These assays are conducted in high-throughput mode, adverse, producing different effects in different tissues at
so results are generated quickly. However, there are concerns different life stages. To complicate this issue further, exposure
about relying on these in vitro and alternative assays for to low-level environmental EACs occurs in an environment in
regulatory decisions in determining potential endocrine activity which natural EACs (e.g., phytoestrogens) and/or exposures
(e.g., Bus and Becker, 2009) because of the lack of to pharmaceuticals contribute to the background hormonal
toxicokinetic considerations (absorption, distribution, metabolism, mileau. The focus on endocrine disruption represents a new
and excretion), the imperfect concordance of ToxCast assays paradigm in toxicology, wherein the MOA, rather than
with in vivo data, and achieving agreement on a mechanism- toxicological outcome, is the focus of chemical screening.
based definition of adverse. In the short run, these data may Development of the EDSP has been more complex than
contribute to the weight of evidence for some test compounds originally envisioned, and the difficulty in identifying assays,
in determining potential endocrine activity. Furthermore, if the efficiently conducting those assays, and data interpretation
vision of ‘‘Toxicology in the 21st Century’’ is realized, such an have become apparent. As endocrine issues have increased in
approach could prove to be far more efficient and cost effective scope, increased research has led to a better understanding of
than the existing EDSP assays, some of which are resource receptor biology. To take full advantage of this new
intensive and use large numbers of animals to screen for knowledge and efficiently meet the needs to screen com-
a limited number of MOAs. pounds for endocrine activity, the EDSP should be iterative
Finally, future research in this area needs to look outward, and allow for reevaluation of earlier assumptions based on the
meaning that information on specific components of the state of the science. Clearly, the past 15 years have
endocrine system (e.g., specific cell culture responses) needs demonstrated that legislative mandates for specific types of
to be interpreted in the context of whole-body physiology, toxicity testing that precede scientific understanding are
preferably that of humans. In the case of environmental difficult to implement and limit the latitude to adapt to
species effects, integrative frameworks such as the ‘‘adverse evolving science. Even so, the significant resources dedicated
outcomes pathway’’ concept have recently been developed to to developing and validating the panel of EDSP assays have
address links between events at the molecular level all the led to an improved understanding of the challenges involved
way up through adverse outcomes at the population level with implementing and interpreting EDSP assays, and these
(Ankley et al., 2010). Because the endocrine system lessons will be beneficial to eventual implementation of the
integrates responses across distant cells, tissues, and organs, EPA’s ToxCast assays and human primary cell assays as
a reductionist approach that solely focuses on isolated proposed in the National Research Council report Toxicity
components of an endocrine circuit can generate incomplete Testing in the 21st Century.
answers and may even guide public health professionals in the
wrong direction. In this same vein, basic principles of ACKNOWLEDGMENTS
toxicology and in particular, toxicokinetics, should not be
forgotten and need to be part of any research program if it is The authors would like to thank Drs James Bus, Matt
to be meaningful for risk assessment. For example, recent Lebaron, and Katie Coady for their thoughtful reviews of the
research on the toxicokinetics of BPA and genistein has manuscript and Dr David Geter for contributing Figure 1.
ENDOCRINE-MEDIATED TOXICITY S105

REFERENCES Colborn, T., Dumanoski, D., and Myers, J. P. (1996). In Our Stolen Future. Are
We Threatening Our Fertility, Intelligence and Survival?—A Scientific
Adams, N. R. (1995). Detection of the effects of phytoestrogens on sheep and Detective Story. Penguin Books USA, New York.
cattle. J. Anim. Sci. 73, 1509–1515. Colborn, T., vom Saal, F. S., and Soto, A. M. (1993). Developmental effects of
Ai, N., Krasowski, M. D., Welsh, W. J., and Ekins, S. (2009). Understanding endocrine-disrupting chemicals in wildlife and humans. Environ. Health
nuclear receptors using computational methods. Drug Discov. Today Perspect. 101, 378–384.
14, 486–494. Available at: http://www.ncbi.nlm.nih.gov/pubmed/19429508. Davis, D. L., Bradlow, H. L., Wolff, M., Woodruff, T., Hoel, D. G., and Anton-
Ankley, G. T., Bennett, R. S., Erickson, R. J., Hoff, D. J., Hornung, M. W., Culver, H. (1993). Medical hypothesis: xenoestrogens as preventable causes
Johnson, R. D., Mount, D. R., Nichols, J. W., Russom, C. L., of breast cancer. Environ. Health Perspect. 101, 372–377.
Schmieder, P. K., et al. (2010). Adverse outcome pathways: a conceptual
Diamanti-Kandarakis, E., Bourguignon, J.-P., Giudice, L. C., Hauser, R.,
framework to support ecotoxicology research and risk assessment. Environ.
Prins, G. S., Soto, A. M., Zoeller, R. T., and Gore, A. C. (2009). Endocrine-
Toxicol. Chem. 29, 730–741.
disrupting chemicals: an Endocrine Society scientific statement. Endocr.

Downloaded from https://academic.oup.com/toxsci/article/120/suppl_1/S93/1615160 by guest on 03 January 2021


Aoki, Y. (2001). Polychlorinated biphenyls, polychlorinated dibenzo-p-dioxins, Rev. 30, 293–342.
and polychlorinated dibenzofurans as endocrine disrupters—what we have
Endocrine Disruptor Screening and Testing Advisory Committee (EDSTAC).
learned from Yusho disease. Environ. Res. 86, 2–11.
(1998). Endocrine Disruptor Screening and Testing Advisory Committee
Arnold, S. F., Klotz, D. M., Collins, B. M., Vonier, P. M., Guillette, L. J., Jr.,
(EDSTAC) Final Report, August 1998. Available at: http://www.epa.gov/
and McLachlan, J. A. (1996). Synergistic activation of estrogen receptor with
endo/pubs/edspoverview/finalrpt.htm. Accessed August 13, 2010.
combinations of environmental chemicals. Science 272, 1489–1492.
Fisch, H. (2008). Declining worldwide sperm counts: disproving a myth. Urol.
Ashby, J., Lefevre, P. A., Odum, J., Harris, C. A., Routledge, E. J., and
Clin. North Am 35, 137–146.
Sumpter, J. P. (1997). Synergy between synthetic oestrogens? Nature
385, 494. Fong, C. J., Burgoon, L. D., Williams, K. J., Forgacs, A. L., and
Zacharewski, T. R. (2007). Comparative temporal and dose-dependent
Ashby, J., Owens, W., Odum, J., and Tinwell, H. (2003). The intact immature
rodent uterotrophic bioassay: possible effects on assay sensitivity of morphological and transcriptional uterine effects elicited by tamoxifen
vomeronasal signals from male rodents and strain differences. Environ. and ethynylestradiol in immature, ovariectomized mice. BMC Genomics
Health Perspect. 111, 1568–1570. 8, 151.
Barlow, S., Kavlock, R. J., Moore, J. A., Schantz, S. L., Sheehan, D. M., Food Quality Protection Act (FQPA). (1996). Public Law 104-170, 110 STAT.
Shuey, D. L., and Lary, J. M. (1999). Teratology Society Public Affairs 1489, Available at: http://www.epa.gov/scipoly/oscpendo/pubs/fqpa.pdf
Committee position paper: developmental toxicity of endocrine disruptors to Accessed August 13, 2010.
humans. Teratology 60, 365–375. Freyberger, A., and Schladt, L. (2009). Evaluation of the rodent Hershberger
Blair, R. M., Fang, H., Branham, W. S., Hass, B. S., Dial, S. L., Moland, C. L., bioassay on intact juvenile males—testing of coded chemicals and
Tong, W., Shi, L., Perkins, R., and Sheehan, D. M. (2000). The estrogen supplementary biochemical investigations. Toxicology 262, 114–120.
receptor relative binding affinities of 188 natural and xenochemicals: Gaido, K., Dohme, L., Wang, F., Chen, I., Blankvoort, B., Ramamoorthy, K.,
structural diversity of ligands. Toxicol. Sci. 54, 138–153. and Safe, S. (1998). Comparative estrogenic activity of wine extracts and
Boue, S. M., Wiese, T. E., Nehls, S., Burow, M. E., Elliott, S., Carter- organochlorine pesticide residues in food. Environ. Health Perspect.
Wientjes, C. H., Shih, B. Y., McLachlan, J. A., and Cleveland, T. E. (2003). 106(Suppl. 6), 1347–1351.
Evaluation of the estrogenic effects of legume extracts containing Garrison, A. W., and Pope, J. D. (1975). In GC/MS Analysis of Organic
phytoestrogens. J. Agric. Food Chem. 51, 2193–2199. Compounds in Domestic Wastewaters. Chemical Congress of the North
Bowers, J. L., Tyulmenkov, V. V., Jernigan, S. C., and Klinge, C. M. (2000). American Continent. Ann Arbor Science, Ann Arbor, Michigan.
Resveratrol acts as a mixed agonist/antagonist for estrogen receptors alpha Gennings, C. (1996). Economical designs for detecting and characterizing
and beta. Endocrinology 141, 3657–3667. departure from additivity in mixtures of many chemicals. Food Chem.
Bus, J. S., and Becker, R. A. (2009). Toxicity testing in the 21st century: a view Toxicol. 34, 1053–1058.
from the chemical industry. Toxicol. Sci. 112, 297–302.
Gennings, C., Carter, W. H., Jr., Carney, E. W., Charles, G. D.,
Carlsen, E., Giwercman, A., Keiding, N., and Skakkebaek, N. E. (1992). Gollapudi, B. B., and Carchman, R. A. (2004). A novel flexible approach
Evidence for decreasing quality of semen during past 50 years. BMJ for evaluating fixed ratio mixtures of full and partial agonists. Toxicol. Sci.
305, 609–613. 80, 134–150.
Carney, E., Woodburn, K. B., and Rowland, S. J. (2010). Endocrine active Giusti, R. M., Iwamoto, K., and Hatch, E. E. (1995). Diethylstilbestrol
chemicals. In Principles and Practice of Mixtures Toxicology
revisited: a review of the long-term health effects. Ann. Intern. Med.
(M. M. Mumtaz, Ed.), pp. 421–442. WILEY-VCH Verlag GmbH & Co.,
122, 778–788.
Weinheim, Germany.
Goodman, J. E., Witorsch, R. J., McConnell, E. E., Sipes, I. G., Slayton, T. M.,
Chapin, R. E., Adams, J., Boekelheide, K., Gray, L. E., Jr., Hayward, S. W.,
Yu, C. J., Franz, A. M., and Rhombert, L. R. (2009). Weight-of-evidence
Lees, P. S., McIntyre, B. S., Portier, K. M., Schnorr, T. M., Selevan, S. G.,
evaluation of reproductive and developmental effects of low doses of
et al. (2008). NTP-CERHR expert panel report on the reproductive and
bisphenol A. Crit. Rev. Toxicol. 39, 1–75.
developmental toxicity of bisphenol A. Birth Defects Res. B 83, 157–395.
Green, S., Walter, P., Greene, G., Krust, A., Goffin, C., Jensen, E., Scrace, G.,
Charles, G. D., Gennings, C., Tornesi, B., Kan, H. L., Zacharewski, T. R.,
Gollapudi, B. B., and Carney, E. W. (2007). Analysis of the interaction of Waterfield, M., and Chambon, P. (1986). Cloning of the human oestrogen
phytoestrogens and synthetic chemicals: an in vitro/in vivo comparison. receptor cDNA. J. Steroid Biochem. 24, 77–83.
Toxicol. Appl. Pharmacol. 218, 280–288. Gronemeyer, H., Gustafsson, J. A., and Laudet, V. (2004). Principles for
Charles, G. D., Kan, H. L., Schisler, M. R., Gollapudi, B. B., and Marty, M. S. modulation of the nuclear receptor superfamily. Nat. Rev. 3, 950–964.
(2005). A comparison of in vitro and in vivo EDSTAC test battery results Gutendorf, B., and Westendorf, J. (2001). Comparison of an array of in vitro
for detecting antiandrogenic activity. Toxicol. Appl. Pharmacol. 202, assays for the assessment of the estrogenic potential of natural and synthetic
108–120. estrogens, phytoestrogens and xenoestrogens. Toxicology 166, 79–89.
S106 MARTY, CARNEY, AND ROWLANDS

Haseman, J. K., Bailer, A. J., Kodell, R. L., Morris, R., and Portier, K. (2001). Kortenkamp, A. (2008). Low dose mixture effects of endocrine disrupters:
Statistical issues in the analysis of low-dose endocrine disruptor data. implications for risk assessment and epidemiology. Int. J. Androl.
Toxicol. Sci. 61, 201–210. 31, 233–240.
Hecker, M., Giesy, J. P., Jones, P. D., Higley, E. B., Newsted, J. L., and Kuiper, G. G., Enmark, E., Pelto-Huikko, M., Nilsson, S., and Gustafsson, J. A.
Mehrle, P. (2006). In Influence of Cell Passage and Freeze/Thaw Events on (1996). Cloning of a novel receptor expressed in rat prostate and ovary. Proc.
Basal Production of 17b-Estradiol and Testosterone by H295R Cells. Natl. Acad. Sci. U.S.A. 93, 5925–5930.
Interim Draft Report, Contract Code # GS-10F-0041L. ENTRIX, Inc. Kuiper, G. G., Lemmen, J. G., Carlsson, B., Corton, J. C., Safe, S. H., van der
September, 2006. Available at: www.epa.gov/endo/pubs/appendix_i-h295r_ Saag, P. T., van der Burg, B., and Gustafsson, J. A. (1998). Interaction of
culture_protocol.pdf. Accessed August 13, 2010. estrogenic chemicals and phytoestrogens with estrogen receptor beta.
Hecker, M., Giesy, J. P., and Timm, G. (2008). Multi-laboratory Validation of Endocrinology 139, 4252–4263.
the H295R Steroidogenesis Assay to Identify Modulators of Testosterone and Laetz, C. A., Baldwin, D. H., Collier, T. K., Herbert, V., Stark, J. D., and
Estradiol Production. Report, Contract Code # GS-10F-0041L. ENTRIX, Scholz, N. L. (2009). The synergistic toxicity of pesticide mixtures:

Downloaded from https://academic.oup.com/toxsci/article/120/suppl_1/S93/1615160 by guest on 03 January 2021


Inc. February, 2008. Available at: www.epa.gov/endo/pubs/h295r_validation_ implications for risk assessment and the conservation of endangered pacific
study_interim_report.pdf. Accessed August 13, 2010. salmon. Environ. Health Perspect. 117, 348–353.
Hignite, C., and Azarnoff, D. L. (1977). Drugs and drug metabolites as Laws, S. C., Ferrell, J. M., Stoker, T. E., and Cooper, R. L. (2003). Pubertal
environmental contaminants: chlorophenoxyisobutyrate and salicylic acid in development in female Wistar rats following exposure to propazine, and
sewage water effluent. Life Sci. 20, 337–341. atrazine biotransformation by-products, diamino-s-chlorotriazine and
Hotchkiss, A. K., Rider, C. V., Blystone, C. R., Wilson, V. S., Hartig, P. C., hydroxyatrazine. Toxicol. Sci. 76, 190–200.
Ankley, G. T., Foster, P. M., Gray, C. L., and Gray, L. E. (2008). Fifteen Lintelmann, J., Katayama, A., Kurihara, N., Shore, L., and Wenzel, A. (2003).
years after ‘‘Wingspread’’—environmental endocrine disrupters and human Endocrine disruptors in the environment. Pure Appl. Chem. 75, 631–681.
and wildlife health: where we are today and where we need to go. Toxicol. Lipscomb, J. C., Lambert, J. C., and Teuschler, L. (2010). Chemical mixtures
Sci. 105, 235–259. and cumulative risk assessment. In Principles and Practice of Mixtures
Houck, K. (2009). In What Can ToxCastä Tell Us about Endocrine Disruption? Toxicology (M. M. Mumtaz, Ed.). WILEY-VCH Verlag GmbH & Co.,
ISRTP Workshop: The Endocrine Disruptor Screening Program—What Can Weinheim, Germany.
Screening Results Tell Us About Potential Adverse Endocrine Effects. Lonard, D. M., Lanz, R. B., and O’Malley, B. W. (2007). Nuclear receptor
Bethesda, MD. Available at: http://www.isrtp.org/index.htm. Accessed coregulators and human disease. Endocr. Rev. 28, 575–587.
August 13, 2010. Lubahn, D. B., Moyer, J. S., Golding, T. S., Couse, J. F., Korach, K. S., and
Hunter, D. J., and Kelsey, K. T. (1993). Pesticide residues and breast cancer: Smithies, O. (1993). Alteration of reproductive function but not prenatal
the harvest of a silent spring? J. Natl. Cancer Inst. 85, 598–599. sexual development after insertional disruption of the mouse estrogen
receptor gene. Proc. Natl. Acad. Sci. U.S.A. 90, 11162–11166.
International Programme on Chemical Safety (IPCS). (2002). Global
assessment of the state-of-the-science of endocrine disruptors. Damstra T., Martin, R.-P., Wright, A. N., Miyamoto, J., and Wermuth, C. G. (1998).
Barlow, S., Bergman, A., Kavlock, R. and Van der Kraak, G., Eds., Natural and anthropogenic environmental oestrogens: the scientific basis for
WHO publication no. WHO/PCS/EDC/02.2. World Health Organization, risk assessment. Pure Appl. Chem. 70, iv.
Geneva. McKenna, N. M., and O’Malley, B. W. (2003). Nuclear receptor signaling:
Jobling, S., Williams, R., Johnson, A., Taylor, A., Gross-Sorokin, M., concepts and models. NURSA: The Nuclear Receptor Signaling Atlas.
Available at: http://www.nursa.org/flash/gene/nuclearreceptor/start.html.
Nolan, M., Tyler, C. R., van Aerle, R., Santos, E., et al. (2006). Predicted
Accessed July 15, 2010.
exposures to steroid estrogens in U.K. rivers correlate with widespread
sexual disruption in wild fish populations. Environ. Health Perspect. McLachlan, J. A. (1997). Synergistic effect of environmental estrogens: report
114(Suppl. 1), 32–39. withdrawn. Science 277, 462–463.
Jordan, V. C. (2003). Antiestrogens and selective estrogen receptor modulators Melnick, R., Lucier, G., Wolfe, M., Hall, R., Stancel, G., Prins, G., Gallo, M.,
as multifunctional medicines. 1. Receptor interactions. J. Med. Chem. Reuhl, K., Ho, S.-M., Brown, T., et al. (2002). Summary of the National
Toxicology Program’s report of the endocrine disruptors low-dose peer
46, 883–908.
review. Environ. Health Perspect. 110, 427–431.
Kamrin, M. A. (2004). Bisphenol A: a scientific evaluation. MedGenMed.
Naciff, J. M., Hess, K. A., Overmann, G. J., Torontali, S. M., Carr, G. J.,
6(3), 7.
Tiesman, J. P., Foertsch, L. M., Richardson, B. D., Martinez, J. E., and
Kamrin, M. A. (2007). The ‘‘low dose’’ hypothesis: validity and implications
Daston, G. P. (2005). Gene expression changes induced in the testis by
for human risk. Int. J. Toxicol. 26, 13–23.
transplacental exposure to high and low doses of 17{alpha}-ethynyl
Kanno, J., Onyon, L., Haseman, J., Fenner-Crisp, P., Ashby, J., and Owens, W. estradiol, genistein, or bisphenol A. Toxicol. Sci. 86, 396–416.
(2001). The OECD program to validate the rat uterotrophic bioassay to Naciff, J. M., Jump, M. L., Torontali, S. M., Carr, G. J., Tiesman, J. P.,
screen compounds for in vivo estrogenic responses: phase 1. Environ. Health Overmann, G. J., and Daston, G. P. (2002). Gene expression profile induced
Perspect. 109, 785–794. by 17alpha-ethynyl estradiol, bisphenol A, and genistein in the developing
Kanno, J., Onyon, L., Peddada, S., Ashby, J., Jacob, E., and Owens, W. female reproductive system of the rat. Toxicol. Sci. 68, 184–199.
(2003a). The OECD program to validate the rat uterotrophic bioassay. Phase Naciff, J. M., Khambatta, Z. S., Reichling, T. D., Carr, G. J., Tiesman, J. P.,
2: coded single-dose studies. Environ. Health Perspect. 111, 1550–1558. Singleton, D. W., Khan, S. A., and Daston, G. P. (2010). The genomic
Kanno, J., Onyon, L., Peddada, S., Ashby, J., Jacob, E., and Owens, W. response of Ishikawa cells to bisphenol A exposure is dose- and time-
(2003b). The OECD program to validate the rat uterotrophic bioassay. Phase dependent. Toxicology 270, 137–149.
2: dose-response studies. Environ. Health Perspect. 111, 1530–1549. Naciff, J. M., Khambatta, Z. S., Thomason, R. G., Carr, G. J., Tiesman, J. P.,
Klinge, C. M., Risinger, K. E., Watts, M. B., Beck, V., Eder, R., and Singleton, D. W., Khan, S. A., and Daston, G. P. (2009). The genomic
Jungbauer, A. (2003). Estrogenic activity in white and red wine extracts. response of a human uterine endometrial adenocarcinoma cell line to
J. Agric. Food Chem. 51, 1850–1857. 17alpha-ethynyl estradiol. Toxicol. Sci. 107, 40–55.
ENDOCRINE-MEDIATED TOXICITY S107

National Research Council (NRC) (1999). Hormonally Active Agents in the Safe, S., Connor, K., Ramamoorthy, K., Gaido, K., and Maness, S. (1997).
Environment. National Academy Press, Washington, DC. Human exposure to endocrine-active chemicals: hazard assessment prob-
National Research Council (NRC) Committee on Toxicity Testing and lems. Regul. Toxicol. Pharmacol. 26, 52–58.
Assessment of Environmental Agents, Board of Environmental Studies Safe, S., Papineni, S., and Chintharlapalli, S. (2008). Cancer chemotherapy
and Toxicology, Institute for Laboratory Animal Research. (2007). Toxicity with indole-3-carbinol, bis(3’-indolyl)methane and synthetic analogs.
Testing in the 21st Century. A Vision and a Strategy. National Academies Cancer Lett. 269, 326–338.
Press, Washington, DC. Schueler, F. W. (1946). Sex hormonal action and chemical constitution. Science
Nettles, K. W., and Greene, G. L. (2005). Ligand control of coregulator 103, 221–223.
recruitment to nuclear receptors. Annu. Rev. Physiol. 67, 309–333. Sharpe, R. M., and Skakkebaek, N. E. (1993). Are oestrogens involved in
Newburgh, R. W. (1975). Toxicology of the reproductive system. In falling sperm counts and disorders of the male reproductive tract? Lancet
Toxicology The Basic Science of Poisons. (L. J. Casarett and J. Doull, 341, 1392–1395.
Eds.), pp. 261–274. Macmillan Publishing Co., Inc, New York, NY. Shelby, M. D., Newbold, R. R., Tully, D. B., Chae, K., and Davis, V. L.

Downloaded from https://academic.oup.com/toxsci/article/120/suppl_1/S93/1615160 by guest on 03 January 2021


Nieuwkoop, P. D., and Faber, J. (1994). In Normal Table of Xenopus laevis. (1996). Assessing environmental chemicals for estrogenicity using a combi-
Garland Publishing Inc., New York, NY. nation of in vitro and in vivo assays. Environ. Health Perspect.
O’Connor, J. C., Cook, J. C., Marty, M. S., Davis, L. G., Kaplan, A. M., and 104, 1296–1300.
Carney, E. W. (2002). Evaluation of Tier I screening approaches for detecting Sluczewski, A., and Roth, P. (1948). Effects of androgenic and estrogenic
endocrine-active compounds (EACs). Crit. Rev. Toxicol. 32, 521–549. compounds on the experimental metamorphoses of amphibians. Gynecol.
O’Connor, J. C., Davis, L. G., Frame, S. R., and Cook, J. C. (2000). Evaluation Obstet. 47, 164–176.
of a Tier I screening battery for detecting endocrine-active compounds Smith, C. L., and O’Malley, B. W. (2004). Coregulator function: a key to
(EACs) using the positive controls testosterone, coumestrol, progesterone, understanding tissue specificity of selective receptor modulators. Endocr.
and RU486. Toxicol. Sci. 69, 79–91. Rev. 25, 45–71.
O’Connor, J. C., Frame, S. R., Davis, L. G., and Cook, J. C. (1999). Detection Stoker, T. E., and Zorrilla, L. M. (2010). The effects of endocrine disrupting
of the environmental antiandrogen p,p#-DDE in CD and Long-Evans rats chemicals on pubertal development in the rat: use of the EDSP pubertal
using a Tier I screening battery and a Hershberger assay. Toxicol. Sci. assays as a screen. In Endocrine Toxicology, 3rd ed. (J. C. Eldridge and
51, 44–53. J. T. Stevens, Eds.), pp. 27–81. Informa Healthcare, New York, NY.
O’Connor, J. C., Marty, M. S., Becker, R. A., Snajdr, S., and Kaplan, A. M. Stumm-Zollinger, E., and Fair, G. M. (1965). Biodegradation of steroid
(2008). Results of the negative control chemical allyl alcohol in the 15-day hormones. J. Water Pollut. Control Fed. 37, 1506–1510.
intact adult male rat screening assay for endocrine activity. Birth Defects Res. Sumpter, J. P. (1995). Feminized responses in fish to environmental estrogens.
B Dev. Reprod. Toxicol. 83, 117–122. Toxicol. Lett. 82–83, 737–742.
Organization for Economic Co-operation and Development (OECD). (2003). Tabak, H. H., and Bunch, R. L. (1970). Steroid hormones as water pollutants.
Detailed Background Review of the Uterotrophic Bioassay, OECD Series on I. Metabolism of natural and synthetic ovulation-inhibiting hormones by
Testing and Assessment, No. 38, OECD Publishing. microorganisms of activated sludge and primary settled sewage. Dev. Ind.
Owens, W., Ashby, J., Odum, J., and Onyon, L. (2003). The OECD program to Microbiol. 11, 367–376.
validate the rat uterotrophic bioassay. Phase 2: dietary phytoestrogen Takeyoshi, M. (2006). In Draft Report of Pre-validation and Inter-laboratory
analyses. Environ. Health Perspect. 111, 1559–1567. Validation for Stably Transfected Transcriptional Activation (TA) Assay to
Owens, W., Gray, L. E., Zeiger, E., Walker, M., Yamasaki, K., Ashby, J., and Detect Estrogenic Activity—The Human Estrogen Receptor Alpha Mediated
Jacob, E. (2007). The OECD program to validate the rat Hershberger Reporter Gene Assay Using hER-HeLa-9903 Cell Line. Chemicals
bioassay to screen compounds for in vivo androgen and antiandrogen Evaluation and Research Institute, Japan. Available at: http://www.oecd.
responses. Phase 2 dose-response studies. Environ. Health Perspect. 115, org/document/62/0,3343,en_2649_34377_2348606_1_1_1_1,00.htm
671–678. Accessed August 13, 2010.
Owens, W., and Koëter, H. B. (2003). The OECD program to validate the rat Tanida, T., Warita, K., Ishihara, K., Fukui, S., Mitsuhashi, T., Sugawara, T.,
uterotrophic bioassay: an overview. Environ. Health Perspect. 111, Tabuchi, Y., Nanmori, T., Qi, W.-M., Inamoto, T., et al. (2009). Fetal and
1527–1529. neonatal exposure to three typical environmental chemicals with different
Owens, W., Zeiger, E., Walker, M., Ashby, J., Onyon, L., and Gray, L. E., Jr. mechanisms of action: mixed exposure to phenol, phthalate, and dioxin
(2006). The OECD program to validate the rat Hershberger bioassay to cancels the effects of sole exposure on mouse midbrain dopaminergic nuclei.
screen compounds for in vivo androgen and antiandrogen responses. Phase 1: Toxicol. Lett. 189, 40–47.
use of a potent agonist and a potent antagonist to test the standardized Tata, J. R. (2005). One hundred years of hormones. EMBO Rep. 6, 490–496.
protocol. Environ. Health Perspect. 11, 1259–1265. Teeguarden, J. G., and Barton, H. A. (2004). Computational modeling of
Powlin, S. S., Cook, J. C., Novak, S., and O’Connor, J. C. (1998). Ex vivo and serum-binding proteins and clearance in extrapolations across life stages and
in vitro testis and ovary explants: utility for identifying steroid biosynthesis species for endocrine active compounds. Risk Anal. 24, 751–770.
inhibitors and comparison to a Tier I screening battery. Toxicol. Sci. Teeguarden, J. G., Waechter, J. M., Jr.., Clewell, H. J., III, Covington, T. R.,
46, 61–74. and Barton, H. A. (2005). Evaluation of oral and intravenous route
Ramamoorthy, K., Wang, F., Chen, I. C., Norris, J. D., McDonnell, D. P., pharmacokinetics, plasma protein binding, and uterine tissue dose metrics of
Leonard, L. S., Gaido, K. W., Bocchinfuso, W. P., Korach, K. S., and bisphenol A: a physiologically based pharmacokinetic approach. Toxicol.
Safe, S. (1997). Estrogenic activity of a dieldrin/toxaphene mixture Sci. 85, 823–838.
in the mouse uterus, MCF-7 human breast cancer cells, and yeast- Teuschler, L., Klaunig, J., Carney, E., Chambers, J., Conolly, R.,
based estrogen receptor assays: no apparent synergism. Endocrinology Gennings, C., Giesy, J., Hertzberg, R., Klaassen, C., Kodell, R., et al.
138, 1520–1527. (2002). Support of science-based decisions concerning the evaluation of the
Roeder, R. G. (2005). Transcriptional regulation and the role of diverse toxicology of mixtures: a new beginning. Regul. Toxicol. Pharmacol. 36,
coactivators in animal cells. FEBS Lett. 579, 909–915. 34–39.
S108 MARTY, CARNEY, AND ROWLANDS

The Nobel Assembly at Karolinska Institutet. (1950). The Nobel Prize in Screen in the Endocrine Disruptor Screening Program Tier-1 Battery. U.S.
physiology or medicine 1950. (Edward C. Kendall, Tadeus Reichstein, and Environmental Protection Agency, Office of Science Coordination and
Philip S. Hench). Available at: http://nobelprize.org/nobel_prizes/medicine/ Policy and Office of Research and Development, Washington, DC. Available
laureates/1950/. Accessed July 15, 2010. at: http://www.epa.gov/endo/pubs/male_pubertal_isr.pdf Accessed August
Touart, L. (2005). In The Story of the Fish Screening Assay in EPA’s Endocrine 13, 2010.
Disruptor Screening Program. Available at: http://www.epa.gov/endo/pubs/ Walker, B. S., and Janney, J. C. (1930). Estrogenic substances. II. Analysis of
edmvac/fish_screening_story.htm. Accessed April 4, 2002.
plant sources. Endocrinology 14, 389–392.
U.S. Environmental Protection Agency (U.S. EPA). (2002). EPA Statement
Zhang, S., Rowlands, C., and Safe, S. (2008). Ligand-dependent interactions of
Regarding Endocrine Disruptor Low-Dose Hypothesis. U.S. Environmental
the Ah receptor with coactivators in a mammalian two-hybrid assay. Toxicol.
Protection Agency, Washington, DC. Available at: http://www.epa.gov/
endo/pubs/edmvs/lowdosepolicy.pdf. Dated March 26, 2005. Accessed Appl. Pharmacol. 227, 196– 206. Available at: http://www.ncbi.nlm.nih.gov/
August 13, 2010. pubmed/18048071.

Downloaded from https://academic.oup.com/toxsci/article/120/suppl_1/S93/1615160 by guest on 03 January 2021


U.S. Environmental Protection Agency (U.S. EPA). (2007). Integrated Zhu, B. T. (2005). Mechanistic explanation for the unique pharmacologic
Summary Report for Validation of a Test Method for Assessment of Pubertal properties of receptor partial agonists. Biomed. Pharmacother. 59,
Development and Thyroid Function in Juvenile Male Rats as a Potential 76–89.

You might also like