EPA/635/R-07/008F
www.epa.gov/iris
vvEPA
TOXICOLOGICAL REVIEW
OF
DECABROMODIPHENYL ETHER
(BDE-209)
(CAS No. 1163-19-5)
In Support of Summary Information on the
Integrated Risk Information System (IRIS)
June 2008
U.S. Environmental Protection Agency
Washington, DC
-------
DISCLAIMER
This document has been reviewed in accordance with U.S. Environmental Protection Agency
policy and approved for publication. Mention of trade names or commercial products does not
constitute endorsement or recommendation for use.
11
-------
CONTENTS—TOXICOLOGICAL REVIEW OF
l^'^SSM'AS'^'-DECABROMODIPHENYL ETHER (CAS No. 1163-19-5)
LIST OF TABLES v
LIST OF FIGURES vi
LIST OF ACRONYMS vii
FOREWORD ix
AUTHORS, CONTRIBUTORS, AND REVIEWERS x
1. INTRODUCTION 1
2. CHEMICAL AND PHYSICAL INFORMATION 3
3. TOXICOKINETICS 4
3.1. ABSORPTION 4
3.1.1. Studies in Humans 4
3.1.2. Studies in Animals 4
3.2. DISTRIBUTION 6
3.2.1. Studies in Humans 6
3.2.1.1 Data in Human Milk 7
3.2.1.2. Data in Human Blood 7
3.2.2. Studies in Animals 9
3.3. METABOLISM 12
3.4. ELIMINATION 15
3.4.1. Half-life Estimates 17
3.5. PHYSIOLOGICALLY BASED TOXICOKINETIC MODELS 18
4. HAZARD IDENTIFICATION 19
4.1. STUDIES IN HUMANS—EPIDEMIOLOGY, CASE REPORTS, CLINICAL
TRIALS 19
4.2. SHORT-TERM, SUBCHRONIC, AND CHRONIC STUDIES AND CANCER
BIO AS SAYS IN ANIMALS—ORAL AND INHALATION 19
4.2.1. Short-term and Subchronic Studies 19
4.2.1.1. Studies in Rats 19
4.2.1.2. Studies in Mice 22
4.2.2. Chronic Studies and Cancer Bioassays 22
4.2.2.1. Study in Rats 23
4.2.2.2. Study in Mice 26
4.3. REPRODUCTIVE/DEVELOPMENTAL STUDIES—ORAL AND INHALATION 28
4.3.1. Studies in Rats 28
4.3.2. Studies in Mice 31
4.4. OTHER ENDPOINT-SPECIFIC STUDIES 35
4.4.1. Receptor Site Interactions 35
4.4.1.1. Aryl Hydrocarbon Receptor Studies 35
4.4.1.2. Estrogen Receptor Studies 37
4.4.1.3. Other Receptors 37
iii
-------
4.4.2. Immunological Studies 37
4.4.3. Genotoxicity 38
4.5. SYNTHESIS OF MAJOR NONCANCER EFFECTS 39
4.5.1. Oral 39
4.5.2. Inhalation 40
4.5.3. Mode-of-Action Information 40
4.6. EVALUATION OF CARCINOGENICITY 43
4.6.1. Summary of Overall Weight of Evidence 43
4.6.2. Synthesis of Human, Animal, and Other Supporting Evidence 44
4.6.3. Mode-of-Action Information 44
4.7. SUSCEPTIBLE POPULATIONS AND LIFE STAGES 48
4.7.1. Possible Childhood Susceptibility 48
4.7.2. Possible Gender Differences 48
5. DOSE-RESPONSE ASSESSMENTS 50
5.1. ORAL REFERENCE DOSE (RfD) 50
5.1.1. Choice of Principal Study and Critical Effect—with Rationale and
Justification 50
5.1.2. Methods of Analysis 54
5.1.2.1. Benchmark Dose Modeling 54
5.1.3. RfD Derivation 55
5.1.4. Previous RfD Assessment 58
5.2. INHALATION REFERENCE CONCENTRATION (RfC) 58
5.3. CANCER ASSESSMENT 58
5.3.1. Choice of Study/Data—with Rationale and Justification 58
5.3.2. Dose Adjustments 59
5.3.3. ExtrapolationMethod(s) 60
5.3.4. Oral Slope Factor 62
5.3.5. Previous Cancer Assessment 63
6. MAJOR CONCLUSIONS IN THE CHARACTERIZATION OF HAZARD AND
DOSE RESPONSE 64
6.1. HUMAN HAZARD POTENTIAL 64
6.2. DOSE RESPONSE 65
6.2.1. Noncancer/Oral 65
6.2.2. Cancer/Oral 66
7. REFERENCES 67
APPENDIX A: SUMMARY OF EXTERNAL PEER REVIEW AND PUBLIC
COMMENTS AND DISPOSITION A-l
APPENDIX B: BENCHMARK DOSE ANALYSIS OF NONCANCER ENDPOINTS B-l
APPENDIX C: BENCHMARK DOSE ANALYSIS OF CANCER ENDPOINTS C-l
IV
-------
LIST OF TABLES
Table 2-1. Physical properties and chemical identity of decaBDE 3
Table 4-1. Oral short-term and subchronic toxicity studies for decaBDE in laboratory
animals 20
Table 4-2. Oral chronic toxicity studies for decaBDE in laboratory animals 24
Table 4-3. Oral developmental and reproductive studies for decaBDE in laboratory
animals 29
Table 4-4. Incidence of liver neoplastic nodules and carcinomas in rats fed decaBDE
for 2 years 46
Table 4-5. Incidence of nonneoplastic or neoplastic lesions in the liver and thyroid
gland of mice fed decaBDE for 2 years 47
Table 5-1. BMDs for potential critical effect from chronic rat and mouse studies 55
Table 5-2. Cancer slope factors derived from BMDs for neoplastic effect from chronic
rat and mouse studies using the multistage model 61
Table B-l. Summary of BMD modeling results for thrombosis in the liver of male rats B-2
Table B-2. Summary of BMD modeling results for degeneration in the liver of male rats B-2
Table B-3. Summary of BMD modeling results for fibrosis in the spleen of male rats B-6
Table B-4. Summary of BMD modeling results for lymphoid hyperplasia in male rats B-7
Table B-5. Summary of BMD modeling results for centrilobular hypertrophy in the
liver of male mice B-7
Table B-6. Summary of BMD modeling results for follicular cell hyperplasia in the
thyroid of male mice B-8
Table C-l. Summary of BMD modeling results for increase in neoplastic nodules in
the liver of male rats C-l
Table C-2. Summary of BMD modeling results for increases in neoplastic nodules or
carcinomas (combined) in the liver of male rats C-4
Table C-3. Summary of BMD modeling results for increases in neoplastic nodules in
the liver of female rats C-7
Table C-4. Summary of BMD modeling results for increases in neoplastic nodules
v
-------
and carcinomas (combined) in the liver of female rats C-10
Table C-5. Summary of BMD modeling results for increases in thyroid follicular
cell hyperplasia as a key event of thyroid tumor in male mice C-13
Table C-6. Summary of BMD modeling results for increases in combined
hepatocellular adenomas or carcinomas in male mice C-16
LIST OF FIGURES
Figure 3-1. Proposed metabolic pathway for BDE-209 13
VI
-------
LIST OF ACRONYMS
AhR
AIC
ALH
ATSDR
AUC
BDE-209
bDNA
BMD
BMDL
BMDS
BMR
CALUX
CAR
CASRN
CD25
cDNA
*-^max
CYP-450
decaBDE
DRE
E2
ED
ER
EROD
FOB
fw
GC/MS
HED
hexaBDE
i.v.
IRIS
LED
LOAEL
Iw
MMP
MRL
mRNA
NAS
NOAEL
nonaBDE
NTP
aryl hydrocarbon receptor
Akaike Information Criterion
amplitude of the lateral head displacement
Agency for Toxic Substances and Disease Registry
area under the curve
decabromodiphenyl ether
branched deoxyribonucleic acid
benchmark dose
BMD 95% lower bound
benchmark dose software
benchmark response
Chemical-Activated LUciferase expression
constitutive androstane receptor
Chemical Abstracts Service Registry Number
interleukin-2-receptor a chain
complementary DNA
maximum concentration
cytochrome P-450
decabromodiphenyl ether
dioxin-responsive element
17(3-estradiol
effective dose
estrogen receptor
ethoxyresorufin O-deethylase
functional observational battery
fresh weight
gas chromatography/mass spectrometry
human equivalent dose
hexabromodiphenyl ether
intravenous
Integrated Risk Information System
95% lower confidence limit for the effective dose
lowest-observed-adverse-effect level
lipid weight
mitochondrial membrane potential
minimal risk level
messenger ribonucleic acid
National Academy of Science
no-observed-adverse-effect level
nonabromodiphenyl ether
National Toxicology Program
vn
-------
octaBDE
PBDE
PBPK
PCB
pentaBDE
PND
PROD
PXR
RfC
RfD
RSD
SXR
T3
T4
TCDD
tetraBDE
triBDE
TSH
UDPGT
UF
octabromodiphenyl ether
polybrominated diphenyl ether
physiologically based pharmacokinetic
polychlorinated biphenyl
pentabromodiphenyl ether
postnatal day
pentoxyresorufm O-deethylase
pregnane X receptor
reference concentration
reference dose
risk-specific dose
steroid X receptor
triiodothyronine
thyroxine
2,3,7,8-tetrachlorodibenzo-p-dioxin
tetrabromodiphenyl ether
tribromodiphenyl ether
thyroid stimulating hormone
uridine diphosphate glucuronyl transferase
uncertainty factor
Vlll
-------
FOREWORD
The purpose of this Toxicological Review is to provide scientific support and rationale
for the hazard and dose-response assessment in IRIS pertaining to chronic exposure to
decabromodiphenyl ether. It is not intended to be a comprehensive treatise on the chemical or
toxicological nature of decabromodiphenyl ether (BDE-209).
This health assessment deals with BDE-209 of relatively high purity (>94%) and does not
deal with earlier commercial decabromodiphenyl ether mixtures containing lower proportions of
decabromodiphenyl ether (e.g., 75% purity). In addition to BDE-209, IRIS health assessments
have also been prepared for three other polybrominated diphenyl ether congeners: tetraBDE-47,
pentaBDE-99, and hexaBDE-153. These four congeners are those for which toxicological
studies suitable for dose-response assessments were available and are the ones most commonly
found in the environment and human biological media.
The intent of Section 6, Major Conclusions in the Characterization of Hazard and Dose
Response, is to present the major conclusions reached in the derivation of the reference dose,
reference concentration and cancer assessment, where applicable, and to characterize the overall
confidence in the quantitative and qualitative aspects of hazard and dose response by addressing
the quality of data and related uncertainties. The discussion is intended to convey the limitations
of the assessment and to aid and guide the risk assessor in the ensuing steps of the risk
assessment process.
For other general information about this assessment or other questions relating to IRIS,
the reader is referred to EPA's IRIS Hotline at (202) 566-1676 (phone), (202) 566-1749 (fax), or
hotline.iris@epa.gov (email address).
IX
-------
AUTHORS, CONTRIBUTORS, AND REVIEWERS
CHEMICAL MANAGER
Joyce Morrissey Donohue, Ph.D.
Office of Water, Office of Science and Technology
Health and Ecological Criteria Division
U.S. Environmental Protection Agency
Washington, DC
AUTHORS
Joyce Morrissey Donohue, Ph.D.
Office of Water, Office of Science and Technology
Health and Ecological Criteria Division
U.S. Environmental Protection Agency
Washington, DC
Hend Galal-Gorchev, Ph.D.
Office of Water, Office of Science and Technology
Health and Ecological Criteria Division
U.S. Environmental Protection Agency
Washington, DC
Qiyu (Jay) Zhao, Ph.D.
Toxicology Excellence for JAisk Assessment
Cincinnati, OH
Bernard Gadagbui, Ph.D.
Toxicology Excellence for JAisk Assessment
Cincinnati, OH
Andrew Maier, Ph.D., Cffl, DABT
Toxicology Excellence for JAisk Assessment
Cincinnati, OH
Toxicology Excellence for JAisk Assessment staff performed work under Purchase Order
4W-1550-NATX.
OFFICE OF RESEARCH AND DEVELOPMENT CO-LEAD
Samantha J. Jones, Ph.D.
Office of Research and Development
National Center for Environmental Assessment
U.S. Environmental Protection Agency
Washington, DC
x
-------
REVIEWERS
This document has been reviewed by EPA scientists, interagency reviewers from other
federal agencies, and the public, and peer reviewed by independent scientists external to EPA. A
summary and EPA's disposition of the comments received from the independent external peer
reviewers and from the public is included in Appendix A of the Toxicological Review of
2,2',3,3',4,4',5,5',6,6'-Decabromodiphenyl ether (BDE-209).
INTERNAL EPA REVIEWERS
Linda Birnbaum, Ph.D.
Experimental Toxicology Division
National Health and Environmental Effects Research Laboratory
Office of Research and Development
Karen Hogan, M.S.
National Center for Environmental Assessment
Office of Research and Development
Tammy Stoker, Ph.D.
Reproductive Toxicology Division
National Health and Environmental Effects Research Laboratory
Office of Research and Development
EXTERNAL PEER REVIEWERS
Ahmed E. Ahmed, Ph.D.
University of Texas Medical Branch
Richard J. Bull, Ph.D.
MoBull Consulting
Lucio G. Costa, Ph.D.
University of Washington
Ralph Kodell, Ph.D.
University of Arkansas for Medical Sciences
Deborah Rice, Ph.D. (chair)
State of Maine
XI
-------
1. INTRODUCTION
This document presents background information and justification for the Integrated Risk
Information System (IRIS) Summary of the hazard and dose-response assessment of
2,2',3,3',4,4',5,5',6,6'-decabromodiphenyl ether (BDE-209). IRIS Summaries may include oral
reference dose (RfD) and inhalation reference concentration (RfC) values for chronic and other
exposure durations, and a carcinogenicity assessment.
The RfD and RfC, if derived, provide quantitative information for use in risk assessments
for health effects known or assumed to be produced through a nonlinear (presumed threshold)
mode of action. The RfD (expressed in units of mg/kg-day) is defined as an estimate (with
uncertainty spanning perhaps an order of magnitude) of a daily exposure to the human
population (including sensitive subgroups) that is likely to be without an appreciable risk of
deleterious effects during a lifetime. The inhalation RfC (expressed in units of mg/m3) is
analogous to the oral RfD, but provides a continuous inhalation exposure estimate. The
inhalation RfC considers toxic effects for both the respiratory system (portal of entry) and for
effects peripheral to the respiratory system (extrarespiratory or systemic effects). Reference
values are generally derived for chronic exposures (up to a lifetime), but may also be derived for
acute (<24 hours), short-term (>24 hours up to 30 days), and subchronic (>30 days up to 10% of
lifetime) exposure durations, all of which are derived based on an assumption of continuous
exposure throughout the duration specified. Unless specified otherwise, the RfD and RfC are
derived for chronic exposure duration.
The carcinogenicity assessment provides information on the carcinogenic hazard
potential of the substance in question and quantitative estimates of risk from oral and inhalation
exposure may be derived. The information includes a weight-of-evidence judgment of the
likelihood that the agent is a human carcinogen and the conditions under which the carcinogenic
effects may be expressed. Quantitative risk estimates are derived from the application of a low-
dose extrapolation procedure. If derived, the oral slope factor is a plausible upper bound on the
estimate of risk per mg/kg-day of oral exposure. Similarly, an inhalation unit risk is a plausible
upper bound on the estimate of risk per ug/m3 air breathed.
Development of these hazard identification and dose-response assessments for BDE-209
has followed the general guidelines for risk assessment as set forth by the National Research
Council (1983). EPA guidelines and Risk Assessment Forum Technical Panel Reports that may
have been used in the development of this assessment include the following: Guidelines for the
Health Risk Assessment of'ChemicalMixtures (U.S. EPA, 1986a), Guidelines for Mutagenicity
Risk Assessment (U.S. EPA, 1986b), Recommendations for and Documentation of Biological
Values for Use in Risk Assessment (U.S. EPA, 1988), Guidelines for Developmental Toxicity
1
-------
Risk Assessment (U.S. EPA, 1991), Interim Policy for Particle Size and Limit Concentration
Issues in Inhalation Toxicity (U.S. EPA, 1994a), Methods for Derivation of Inhalation Reference
Concentrations and Application of Inhalation Dosimetry (U.S. EPA, 1994b), Use of the
Benchmark Dose Approach in Health Risk Assessment (U.S. EPA, 1995), Guidelines for
Reproductive Toxicity Risk Assessment (U.S. EPA, 1996), Guidelines for Neurotoxicity Risk
Assessment (U.S. EPA, 1998), Science Policy Council Handbook: Risk Characterization (U.S.
EPA, 2000a), Benchmark Dose Technical Guidance Document (U.S. EPA, 2000b),
Supplementary Guidance for Conducting Health Risk Assessment of Chemical Mixtures (U.S.
EPA, 2000c), A Review of the Reference Dose and Reference Concentration Processes (U.S.
EPA, 2002), Guidelines for Carcinogen Risk Assessment (U.S. EPA, 2005a), Supplemental
Guidance for Assessing Susceptibility from Early-Life Exposure to Carcinogens (U.S. EPA,
2005b), Science Policy Council Handbook: Peer Review (U.S. EPA, 2006a), and A Framework
for Assessing Health Risks of Environmental Exposures to Children (U.S. EPA, 2006b).
The literature search strategy employed for this compound was based on the Chemical
Abstracts Service Registry Number (CASRN) and at least one common name. Any pertinent
scientific information submitted by the public to the IRIS Submission Desk was also considered
in the development of this document. The relevant literature was reviewed through November
2007.
-------
2. CHEMICAL AND PHYSICAL INFORMATION
Decabromodiphenyl ether (decaBDE or BDE-209) is a fully brominated diphenyl ether
compound (i.e., 10 bromine atoms) used as a flame retardant. The composition of commercial
decaBDE is almost exclusively the deca-substituted BDE-209 (typically >97%), the remainder
being nonabromodiphenyl ether (nonaBDE); trace amounts of octabromodiphenyl ether
(octaBDE) may be present (Schecter et al., 2003; American Chemistry Council, 2002; Hardy,
2002). The composition of older decaBDE formulations, which are no longer commercially
produced in the U.S., was approximately 77% decaBDE, 22% nonaBDE, and 1% octaBDE
(Kociba et al., 1975). Physical and chemical properties of decaBDE (>97% purity) are listed in
Table 2-1.
Table 2-1. Physical properties and chemical identity of decaBDE
CASRN
Synonyms
Physical state
Melting point, °C
Boiling point, °C
Vapor pressure at 21°C, Pa
Henry's law constant:
atm nrVmol
(Pam3mor1)at25°C
Density, g/cm3
Water solubility at 25°C, ug/L
Log Kow
Log Koc
Molecular weight
Chemical formula
Chemical structure
Physical property/chemical identity
1163-19-5
2,2',3,3',4,4',5,5',6,6'-decaBDE; BDE-209; decaBDE;
benzene, l,l'-oxybis[2,3,4,5,6,-pentabromo]-;
decabromodiphenyl oxide; decabromodiphenyl ether;
decabromobiphenyl ether; ether,
bis(pentabromophenyl)
Solid
300-310
decomposes at >320°C
4.63 x 1Q-6
1.93 x 10~8
0.04
3.0
<0.1
6.3-12.6
6.3
959.17
C12Br10O
Br Br
^v/^X ^SX^x8'
Br^x^^Br eAs^^Br
Br Br
Reference
U.S. EPA (2004)
U.S. EPA (2004);
ATSDR3 (2004)
Hardy (2002)
ECB3 (2003)
ECB (2003)
Hardy (2002)
Hardy (2002);
Cetin and Odabasi
(2005)
NASa(2000)
Hardy (2002)
Hardy (2002)
Hardy (2002)
U.S. EPA (2004)
U.S. EPA (2004)
3ATSDR = Agency for Toxic Substances and Disease Registry; ECB = European Chemicals Bureau; NAS = National
Academy of Science.
-------
3. TOXICOKINETICS
Data on the toxicokinetics of decaBDE in humans are limited to findings on the levels in
serum and maternal milk that demonstrate it is absorbed from the environment and distributed to
tissues. Several studies have been conducted to evaluate the absorption, distribution,
metabolism, and elimination of decaBDE after oral or intravenous (i.v.) dosing in rats and mice.
Absorption is low (7-26%) after oral exposure to rats. Following absorption, the highest levels
are found in the liver, muscle, and skin. This differs from studies of the lower brominated
congeners, where the highest levels are generally found in the adipose tissues. The available
metabolic data apply exclusively to rats, where both debrominated and hydroxylated-
debrominated species have been identified in plasma. In one study, the metabolites in the feces
exceeded those that could be accounted for from bile, raising the possibility that some
conversion of the parent compound may be mediated by the intestinal epithelium or microflora.
This phenomenon was not reported for the lower brominated BDE congeners. Urinary excretion
of decaBDE-209 is minimal in rats; no data are available for mice. In rats, some of the urinary
radiolabel (18%) was bound to albumin.
3.1. ABSORPTION
3.1.1. Studies in Humans
There are no direct studies of decaBDE absorption in humans. The data that demonstrate
human absorption come from measurements of decaBDE in human biological media after
anthropogenic exposures but do not provide information on the quantitative aspects of
absorption or the kinetics of tissue distribution and retention.
3.1.2. Studies in Animals
Studies on BDE-209 absorption following oral dosing demonstrate its absorption
potential. However, there were few direct measurements of absorbed dose, and absorption
estimates are based on a combination of concentrations in blood and data on excretion. Among
the several oral dosing studies, the percentage of an administered dose absorbed across the
gastrointestinal tract ranged from approximately 7-26%. In some cases it was difficult to derive
an accurate estimate of absorption because of the high proportion of the dose found in the feces
(>90%) and the high percentage that was present as metabolites in feces.
Sandholm et al. (2003) evaluated the bioavailability of unlabeled decaBDE in male
Sprague-Dawley rats. One group of rats (n = 18) was dosed by gavage with unlabeled decaBDE
(>98% purity) in dimethylamide/polyethylene glycol/water vehicle at 2 |imol/kg (1.9 mg/kg).
Another group of rats (n = 18) was dosed intravenously via the tail vein with unlabeled decaBDE
-------
at the same dose. At specific time intervals, blood samples from three rats per group were
collected according to the following schedule: group 1 was sampled 1 hour and 24 hours after
dosing; group 2 after 3 hours and 48 hours; group 3 after 6 hours and 72 hours; group 4 after
96 hours; group 5 after 120 hours; and group 6 after 144 hours. Plasma samples were extracted,
and decaBDE and its metabolites were quantified. Based on comparison to plasma levels (area
under the curve [AUC]) following i.v. injections, the oral bioavailability (the percent of the dose
reaching systemic circulation) was calculated to be 26% in the rat. The mean maximum plasma
concentration (Cmax) following oral dosing was 264 pmol/mL in the 6-hour sample.
One of the limitations of the absorption data for decaBDE is a lack of knowledge about
the mechanism of gastrointestinal absorption. Passive diffusion across the lipid membrane
appears to be restricted to lipophilic compounds with low molecular weight rather than high
molecular weight compounds such as decaBDE. Accordingly, facilitated transport and/or uptake
with dietary lipids by way of the chylomicrons may provide routes for absorption.
In a gavage study by Morck et al. (2003), male Sprague-Dawley rats (n = 8) received
3 jimol/kg (2.9 mg/kg) 14C-labeled decaBDE diluted with unlabeled compound (>98% purity,
15 Ci/mol), prepared by dissolving the decaBDE labeled/unlabeled mixture in toluene, followed
by sonication, suspension in Lutrol F127/soy phospholipone/water vehicle, and evaporation of
the toluene by nitrogen flow. Four rats were sacrificed after 3 days and the other four rats after
7 days. Urine and feces were collected at 24-hour intervals for 3 and 7 days, respectively, and
assayed for radioactivity. Two additional male Sprague-Dawley rats were bile-duct-cannulated,
treated similarly with 14C-labeled decaBDE, and sacrificed 3 days later. Bile from these rats was
collected and assayed for radioactivity.
Results indicated that in the conventional rats, about 90% of the dose was excreted in the
feces within 3 days after a single oral dose of 14C-labeled decaBDE, and the majority of this
radioactivity (65%) represented decaBDE metabolites. Radioactivity in bile collected from the
cannulated rats accounted for 10% of the fecal excretion. Almost all of the excreted
radioactivity in the bile represented metabolites, indicating that at least 10% of the decaBDE
dose had been absorbed. It is also possible that greater than 10% of the oral dose may have been
absorbed since 65% of the radioactivity excreted in the feces was in the form of metabolites.
However, interpretation of the data is difficult. The relatively large amount of metabolites
excreted in the feces could be due to a combination of biliary excretion of metabolites,
metabolism of decaBDE by the microflora in the gastrointestinal tract, metabolism in the
intestinal epithelium, and/or nonbiliary systemic secretion into the gut.
Hughes et al. (2001) conducted a study to evaluate the in vitro dermal absorption of
decaBDE in mice. In this study, the dorsal skin of female hairless (Crl:SKHl-hr-BR) mice was
removed, cut to a thickness of 255 |im, and exposed in a flow-through diffusion cell system to
carrier-free 14C-decaBDE (>98% purity; specific activity not provided) at doses of 6, 30, or
-------
60 nmol. The percent dose absorbed as measured by the amount of material in the collecting
chamber was determined at 6, 12, 18, and 24 hours. Very little compound passed through the
skin sections; the percentages of dose reaching the receptor fluid in the collecting chamber after
24 hours ranged from 0.07-0.34% and were inversely related to dose.
For all three doses used in the experiment, the largest portion of the dose was taken up
during the first 6 hours: 0.17%, 0.04%, and 0.03% of the applied 6, 30, and 60 nmol decaBDE
doses, respectively. Results also showed that the 24-hour cumulative percent of the dose
absorbed decreased with increasing applied dose, whereas the mass of chemical absorbed
increased with increasing applied dose. This result suggests saturation of uptake at the higher
doses. The authors calculated the total for the dose retained in the skin sections and transported
to the receptor fluid as 20.5%, 3.3%, and 1.9% of the applied dose for 6, 30, and 60 nmol
decaBDE, respectively. Most of the compound taken up in the 24-hour period was retained in
the skin. The authors acknowledged that the in vitro results observed in this study using mouse
skin may overestimate the amount of decaBDE that would be absorbed by human skin, given
that the mouse skin is more permeable to several chemicals, at least in vitro, than rat, pig, or
human skin.
3.2. DISTRIBUTION
BDE-209 has very low water solubility and a relatively high Kow. Accordingly, high
distribution to adipose tissue might be expected. However, as indicated by the data that follow,
that is not the case. Systemic distribution of hydrophilic metabolites, as well as molecular mass
and favored conformation, may play a role in the limited uptake by adipocytes. The low uptake
of decaBDE into tissue lipids makes it different from the less highly substituted, lower molecular
weight polybrominated diphenyl ethers (PBDEs).
3.2.1. Studies in Humans
The human data described below come from monitoring of PBDEs in human populations
rather than from measured dosing studies. The data demonstrate that humans are exposed to
PBDEs and that absorption and distribution to some tissues occur. The data do not provide
information on the quantitative aspects of absorption or the kinetics of tissue retention. Limited
data are available on the occurrence of BDE-209 in human biological media. Data, described
below, are available for human milk and blood samples and indicate a tendency for BDE-209 to
distribute to these tissues. However, thorough distribution studies have not been conducted in
humans, and therefore it is not known whether BDE-209 distributes to other tissues as well. The
number of samples examined in various studies and countries is small, and therefore the data
should not be construed as representative at the national level.
-------
3.2.1.1 Data in Human Milk
In a study conducted in 2002 of levels of PBDEs in human milk in the U.S., 47 samples
from Caucasian, African-American, and Hispanic nursing mothers 20-41 years of age and living
in Texas were analyzed for 13 PBDE congeners (Schecter et al., 2003). Mean and median total
concentrations of tribromodiphenyl ether (triBDE) through decaBDE were 74 and 34 ng/g lipid
weight (Iw). DecaBDE was present in these samples, with a mean concentration of 0.9 ng/g Iw
(1.2% of total PBDEs in the milk; median concentration not available), suggesting that some
decaBDE is absorbed, distributed to mammary tissue, and secreted in human milk during
lactation.
Milk samples were collected in 2003 from 40 first-time mothers with 8-week-old infants
and residing in urban areas in the Pacific Northwest of the U.S. (Montana, Oregon, and
Washington State) and British Columbia, Canada (She et al., 2007). Mean and median total
concentrations of 12 tri- through decaBDE congeners were 96 and 50 ng/g Iw, respectively.
These values are substantially higher than the values reported in the study of Schecter et al.
(2003) and could be due to the fact that the mothers in the later study had been nursing for
longer periods of time. BDE-47 was found at the highest level, followed by hexabromodiphenyl
ether (hexaBDE)-153 and pentabromodiphenyl ether (pentaBDE)-99 and -100. DecaBDE-209
with mean and median concentrations of 0.8 and 0.4 ng/g Iw, respectively, was a minor congener
in breast milk.
3.2.1.2. Data in Human Blood
Sjodin et al. (2001a) reported a finding of BDE-209 in serum samples collected from
12 U.S. blood donors in 1988. The median concentration was <1 pmol/g Iw with a range of
-------
14 ng/g Iw, father 23 ng/g Iw). The authors suspected house dust and breast milk to contribute
appreciably to the child and infant exposures; however, no firm conclusions can be drawn from
this study, given the small number of subjects investigated.
Sjodin et al. (1999) investigated the exposure to PBDEs of Swedish workers by
comparing blood PBDE concentrations collected in 1997 from personnel at an electronics
dismantling plant, clerks working full time in front of computer screens, and a control group of
hospital cleaning workers. Electronics dismantling involved grinding plastic goods in a
shredder. This process releases airborne particulate matter from plastic parts containing
brominated flame retardants. The investigators found decaBDE in the serum of individuals from
all three groups (19-20 male and female subjects per group). The median BDE-209
concentration in hospital cleaners was <0.7 pmol/g Iw (<9.7 ng/g), with a range of <0.3-
3.9 pmol/g Iw (<0.3-3.7 ng/g), while the median BDE-209 concentration in computer clerks was
<0.7 pmol/g Iw (<0.7 ng/g), with a range of <0.3-8.0 pmol/g Iw (<0.3-7.8 ng/g). Plasma levels
of BDE-209 were significantly higher in the electronics dismantling workers than in the other
two groups with a median BDE-209 concentration of 5.0 pmol/g Iw (4.8 ng/g, range <0.3-
9.9 pmol/g Iw [<0.3-9.5 ng/g]). The higher BDE-209 concentration in the serum of the
electronics dismantling workers was attributed to the relatively high total BDE-209
concentration in the air of the dismantling hall (mean 36 ng/m3) and at the shredder (175 ng/m3)
(Sjodin et al., 2001b). The presence of BDE-209 in the blood samples from these three groups
of workers qualitatively indicated the bioavailability of BDE-209 in humans, even though this
compound is not expected to be greatly bioavailable based on its high molecular mass. There
was no correlation between plasma levels of BDE-209 with age or fish consumption (the only
food evaluated in the study). The serum concentrations of all PBDE congeners decreased in
electronic-dismantling workers after vacation. The median decreases, standardized to 30 days of
leave, were 14% for BDE-47, -153, and -154, 30% for BDE-183, and 66% for BDE-209. These
results indicate shorter half-lives of the more highly brominated diphenyl ethers. However,
serum half-lives were not estimated by the authors.
In another Swedish study, increased BDE-209 in the serum was also found in computer
technicians who repair or partially dismantle computers (Jakobsson et al., 2002). The median
BDE-209 concentration in groups of 19-20 subjects was 1.6 pmol/g Iw (1.5 ng/g) in computer
technicians, while it was <0.7 pmol/g Iw (<0.7 ng/g) in hospital cleaners and computer clerks. A
possible source of exposure is airborne dust particulate matter to which PBDEs strongly adsorb
(Sjodin etal., 1999).
Thuresson et al. (2005) assessed the exposure to PBDEs in Swedish workers engaged in
manufacturing decaBDE flame-retarded rubber goods or electric cables. A referent group,
abattoir (slaughterhouse) workers with no occupational exposure to PBDEs, was also
investigated. The commercial decaBDE used was Saytex 102E, consisting mainly of BDE-209
-------
with traces of nonaBDEs (BDE-206, -207, and -208) and unknown octaBDE congeners.
Consumption of fatty fish was low in all three groups (median 0.5 meals/month), age ranged
between 24 and 60 years, with a median of 40 years in all three groups, and each group consisted
of approximately 20 subjects. The concentration of 12 PBDE congeners, ranging from tetra- to
decaBDE, was measured in the serum samples of all individuals participating in the study.
Elevated serum concentrations of octa-, nona-, and decaBDE were present in serum of workers
handling decaBDE flame-retarded rubber. Serum concentrations of BDE-209 were up to 50- to
100-fold higher than those of the referent group. In contrast, the serum concentrations of tetra-
to heptaBDEs were similar among rubber workers and referents.
3.2.2. Studies in Animals
No studies were identified regarding distribution of decaBDE in animals after inhalation
exposure. Several rodent oral dosing studies were identified and are described below.
Sandholm et al. (2003) evaluated the distribution of unlabeled decaBDE in male Sprague-
Dawley rats. A group of rats (n = 18) was dosed by gavage with unlabeled decaBDE (>98%
purity) in dimethylamide/polyethylene glycol 400/water vehicle at 2 |imol/kg (1.9 mg/kg).
Another group of rats (n = 18) was dosed intravenously with the same dose of decaBDE. At
specific time intervals, blood samples from three rats per group were collected for up to
144 hours. Plasma samples were extracted, and decaBDE and its metabolites were quantified.
Oral results indicated that the mean plasma Cmax was 264 pmol/mL (253 ng/g) at 6 hours after
dosing and AUC was 12 nmol x hour/mL (11.5 jig x hour/mL). The results from the i.v. study
indicated that the clearance was 0.60 mg/minute-kg and the apparent volume of distribution at
steady state was 1.4 L/kg. Concentrations in other tissues were not measured; therefore, relative
tissue distribution could not be determined. However, the identification of decaBDE in the
plasma supports other studies, suggesting wide tissue distribution.
Morck et al. (2003) evaluated the distribution of decaBDE (>98% purity) in male
Sprague-Dawley rats after oral administration. Four rats were sacrificed 3 or 7 days after
administration of 3 |imol/kg (2.9 mg/kg) of 14C-labeled and unlabeled decaBDE combined
(15 Ci/mol). Liver, adipose tissue, lung, kidney, adrenal glands, skin, muscle, spleen, testis,
thymus, heart, plasma, colon wall and contents, and small intestine wall and contents were
collected. Radioactivity was determined in all tissues. Analysis of tissues and organs for
radioactivity indicated that approximately 9% of the dose remained in the body at 3 and 7 days.
On a fresh weight (fw) adjusted basis, the concentrations at 3 and 7 days were highest in the
adrenal glands (1.25 and 0.41 nmol/g fw [1.2 and 0.4 |ig/g]), liver (0.55 and 0.20 nmol/g fw
[0.7 and 0.2 |ig/g]), kidney (0.17 and 0.07 nmol/g fw [0.16 and 0.07 |ig/g]), and heart (0.14 and
0.05 nmol/g fw [0.13 and 0.05 |ig/g]), respectively. On an Iw basis, the plasma and liver had the
highest concentrations of radiolabel (22 and 14.9 nmol/g Iw [21 and 14 |ig/g] on day 3 and
-------
8.8 and 5.3 nmol/g Iw [8.4 and 5.1 ug/g] on day 7, respectively), whereas adipose tissue, testis,
thymus, spleen, small intestine wall, skin muscle, lung, kidney, adrenal, and heart had low
concentrations. These results indicate that decaBDE was not readily distributed to lipid-rich
tissues but rather was found in plasma and blood-rich tissues, including liver, kidney, heart, and
intestinal wall.
Huwe and Smith (2007) conducted a repeat dosing study of BDE-209 during which a
group of 18 male Sprague-Dawley rats were given 0.36 ug/day BDE-209 in an oil-based
supplement added to their diets for 21 days. Eight control animals received the same diet and
oil-based supplement. At the end of dosing, groups of three animals were sacrificed on days 0,
3, 7, 10, 14, and 21 of a follow-up period that began 24 hours after the last feeding of the treated
diet. There was no collection of excreta during the feeding period. Twenty-four hours after the
last feeding, the levels of BDE-209 in liver, plasma, gastrointestinal tract, and carcass were
measured. The highest levels were found in the liver (48.2 ± 8.9 ug/g wet weight) and
gastrointestinal tract (35.9 ± 8.5 ug/g wet weight). The amounts in plasma (3.6 ± 0.9 ug/g wet
weight) and carcass (14.0 ± 4.0 ug/g wet weight) were lower. After the 21-day exposure, the
tissues of the exposed rats contained levels of BDE-209 that were 10-20 times greater than those
in control rats. In addition, hepta-, octa-, and nonaBDEs were isolated in tissues and were
apparently formed via reductive debromination of BDE-209. Two of the octaBDE congeners
(BDE-201 and -197) bioconcentrated in the tissues to a greater extent than the parent and other
debrominated metabolites. Huwe and Smith (2007) made no attempt to determine if
hydroxylated metabolites had been formed. These results differ from those for the tetra-, penta-,
and hexaBDE congeners, where there is no evidence for debromination in the absence of
hydroxylation.
Viberg et al. (2003a) studied the distribution of decaBDE in mice. NMRI male mice (n =
6-8) received a single oral dose (2.22 mg/kg) of 14C-decaBDE (40.5 uCi/kg) by gavage on
postnatal day (PND) 3, 10, or 19. The animals were sacrificed 1 and 7 days after administration
of the radiolabeled decaBDE, and the label in the brain, heart, and liver was measured. The
highest concentrations as a percentage of the administered dose were seen in the liver, followed
by the brain and heart. The authors did not state whether the radioactivity was that of the parent
compound or its metabolites.
One day after administration, the mean radioactivity in the liver of mice treated with
radiolabeled decaBDE on PND 3, 10, or 19 was 12.6%, 9.4%, or 5.8% of the administered dose,
respectively, whereas 7 days after administration, the radioactivity decreased significantly to
4.8%, 4.6%, or 0.3%, respectively. The distribution to and retention by the liver was greater for
the dosing on PNDs 3 and 10 than for the dosing on PND 19. One day after administration, mice
treated on PNDs 3 and 10 had 0.5 and 0.4% of total activity administered, respectively, in the
brain, and the radioactivity increased to 0.7 and 1.1%, respectively, 7 days after the dosing. In
10
-------
contrast, mice treated with 14C-decaBDE on PND 19 had only 0.06% of the total radioactivity
administered in the brain 1 and 7 days after administration. Significant age-dependent
differences in distribution to the liver and developing brain were noted. On the other hand, the
amount of radioactivity in the heart was minimally affected by age of the animal 24 hours after
administration (0.28, 0.31, and 0.22% of the total dose was measured in the heart 24 hours after
administration on PNDs 3, 10, and 19, respectively). After 7 days of administration, the 3-day-
old and 10-day-old animals did not have significant changes in the amounts of radioactivity in
the heart (0.34 and 0.32% of the total administered dose). The amount of radioactivity detected
in the hearts of 19-day-old mice had decreased significantly to 0.08%.
In a study by el Dareer et al. (1987) the liver of rats fed two diets containing low
(0.0277%; 25 mg/kg) or high (4.8%; 4,400 mg/kg) amounts of 14C-labeled decaBDE contained
0.45, 0.21, and 0.11% of the administered dose on days 1, 2, and 3 after administration of the
low dose and 0.007, 0.007, and 0.016% after administration of the high dose, respectively.
These data are also included in Appendix O of the National Toxicology Program (NTP) (1986)
report. These results suggest saturation in uptake or distribution. High-pressure liquid
chromatography and ultraviolet spectral analysis indicated that 81% of the radioactivity in the
liver was decaBDE rather than metabolites. The maximum percent of dose in the organs and
tissues obtained from the rats fed the lower dose of decaBDE (0.0277%) was in the following
order: liver > skin > muscle > fat > blood > gut tissue > plasma > kidneys > lungs > spleen >
brain.
Hakk et al. (2002) investigated the disposition of decaBDE in rats. Groups of four male
Sprague-Dawley rats (conventional rats) and bile-duct-cannulated rats (cannulated rats) were
administered 14C-labeled decaBDE (>97% purity; 15 mCi/mol) in Lutrol F127/soya
phospholipid/water vehicle as a single oral dose of 3 jimol/kg (2.9 mg/kg). Gastrointestinal
mucosa, kidneys, liver, and lungs were collected from the animals 72 hours following dosing.
Radioactivity content was assayed in tissue supernatants and protein-bound extracts.
Radioactivity was also quantified from pellets obtained after centrifugation of homogenized
tissues. In addition, binding of radiolabeled decaBDE and/or its metabolites to liver proteins
was determined. Analysis of liver, lung, intestinal cells, and kidney tissues of noncannulated rats
72 hours following decaBDE administration indicated that the majority (>45-80%) of the
decaBDE-derived radioactivity was associated with the membrane fraction, whereas 0-29% and
4-24% were associated with microsomal and soluble fractions, respectively.
Collectively, these studies suggest wide tissue distribution, although relative distribution
among various tissues differed across studies in adult rodents. Significant age-dependent
differences in distribution to the undeveloped brain were noted.
11
-------
3.3. METABOLISM
Oral and i.v. studies suggest that decaBDE is metabolized through oxidative
dehalogenation reactions to form phenolic metabolites and debrominated to form a
variety of nona-, octa-, and heptaBDE congeners. This conclusion is based on the
extensive presence of metabolites in the fecal matter and in blood. However, the sites of
metabolism have not been identified. At least a portion is likely to occur in the liver.
Extrahepatic metabolism may occur in the epithelium of the gastrointestinal tract or
through preabsorption metabolism of decaBDE by the intestinal microflora. Figure 3-1
provides a summary of the proposed metabolic pathway as derived from the metabolites
that have been identified in plasma, tissues, and fecal material.
It should be noted that some metabolites may be formed by intestinal microbes. The
number of bromines removed will depend on whether the starting material is the parent or a
partially debrominated metabolite. If absorption occurs with lipids via the chylomicrons, other
tissues may also participate in metabolism. The identification of the feces as the major excretory
pathway and the high percentage of metabolites present in the feces that cannot be accounted for
through biliary excretion provide support for the hypothesis that the liver may not be the only
site for metabolic conversions.
Morck et al. (2003) studied decaBDE metabolism in rats given a single oral dose by
gavage. Results indicated that in noncannulated rats about 90% of the dose was excreted in the
feces within 3 days after a single oral dose of 14C-labeled decaBDE and the majority of this
radioactivity (65%) represented decaBDE metabolites. Measurement of bile radioactivity
indicated that close to 10% of the total dose was excreted in the bile during the same period,
with almost all of the excreted dose in bile in the form of metabolites.
Analysis of radiolabeled materials from tissues of these rats at day 3 after decaBDE
administration revealed that 42% of the radioactivity in the liver represented solvent extractable
lipid-bound metabolites and 30% was in the form of extractable unconjugated metabolites (4 and
26% of which were hydroxylated and neutral metabolites, respectively). Twenty-seven percent
of the radiolabel could not be extracted and was tissue bound. Only 1% of the extractable
material was water soluble. A larger percentage (61%) of the radioactivity in the small intestine
wall was tissue bound. Lipid-bound metabolites accounted for 7% of the label, water-soluble
compounds for 11%, and unbound parent or metabolites for 20%. The percentage of the
radiolabel found as water-soluble compounds in the intestinal wall was 10 times greater than the
water-soluble metabolites in the liver, providing some support for the hypothesis that oxidative
metabolism can occur in the intestinal mucosa. Most of the radioactivity (71-80%) in the lung,
adipose tissue, and kidney was unbound parent or metabolites; 15-21% represented lipid-bound
metabolites, and 1.5-8% was tissue bound. Formation of adducts was indicative of covalent
and/or noncovalent interactions with cellular macromolecules.
12
-------
Excreted as
4
parent
compound gr'
Arene epoxide
I intermediate J
x + y = 5, 6, or ',
Figure 3-1. Proposed metabolic pathway for BDE-209.
Source: Derived fromHuwe and Smith (2007), Sandholmet al. (2003), and Morck et al. (2003).
Sandholm et al. (2003) evaluated the metabolism of unlabeled decaBDE in male rats
after gavage or i.v. injection. Blood samples were collected at specific intervals for up to 6 days.
Pooled plasma samples from all 6 days were extracted and decaBDE and its metabolites were
quantified. Analysis of the pooled samples indicated that the major neutral compound in the
plasma was unmodified decaBDE with trace amounts of three nonaBDEs. Thirteen phenolic
metabolites were determined in the plasma of both the orally and i.v. dosed rats, but only three
phenolic metabolites were present in sufficiently high concentration for further analysis. These
metabolites were characterized as a hydroxy-octaBDE, an hydroxy-nonaBDE, and an
hydroxy/methoxy hexaBDE (Figure 3-1). The relative amount of each metabolite recovered was
13
-------
not reported, but the concentration of phenolic radioactivity in the plasma collected 3 and 7 days
after oral gavage was four times higher than that of the neutral compounds (i.e., the parent or
debrominated decaBDE). The authors indicated that reductive debromination may be the first
step in the metabolic pathway of decaBDE, followed by oxidation to form phenolic metabolites.
It was also suggested that the hydroxy/methoxy metabolites were probably formed via an arene
oxide hydrolyzed to a dihydrodiol and further rearomatized followed by a methylation reaction
(Figure 3-1).
In the study conducted by Morck et al. (2003), the unconjugated phenolic fractions
isolated from the feces were examined by gas chromatography/mass spectrometry (GC/MS).
Methoxyhydroxylated penta- to heptabrominated diphenyl ethers were identified. As was the
case in the Sandholm et al. (2003) study, the methoxy and hydroxy substituents were on the same
phenyl ring when both were present. In addition, trace amounts of debrominated metabolites
and nonaBDEs were also found in the feces and bile (see Figure 3-1), indicating debromination
may have been the first step in decaBDE metabolism. A small proportion of monohydroxylated
metabolites was found in tissues and feces, indicating a role for reductive dehalogenation
followed by an oxidation step or direct oxidative dehalogenation reactions.
The plasma samples collected during the Morck et al. (2003) study were analyzed and
reported by Sandholm et al. (2003). As was the case with the plasma samples collected by
Sandholm et al. (2003), the neutral fraction was almost all BDE-209 with a small fraction of
three nonaBDEs. Neutral compounds represented 19% of the plasma total in the samples drawn
at 3 days and 21% in the samples collected at 7 days. The phenolic compounds in the plasma
could not be purified enough to allow GC/MS analysis.
Huwe and Smith (2007) identified nonhydroxylated metabolites found in feces and
tissues after 21 days dosing with 0.36 ug/day BDE-209 in an oil-based supplement added to their
diets. Only 5% of the parent dose was present in the body after 21 days of dosing. Three
nonaBDEs (206, 207, and 208), four octaBDEs (196, 197, 201, and 203), and one heptaBDE
(183) were identified in the body tissues. No attempt was made to determine if hydroxylated
metabolites were present. The results are indicative of considerable fecal excretion during the
dosing period.
Intravenous administration of 1.07 mg/kg decaBDE to rats (el Dareer et al., 1987; NTP,
1986) resulted in the major part of the dose (74%) being excreted as metabolites in feces. About
63% of the excreted material in the feces was metabolites, and the remaining 37% was
unchanged decaBDE. In these rats, 1% of the radioactivity in the bile was intact decaBDE,
while 99% of the radioactivity excreted in the bile was in the form of metabolites.
Animal studies indicated that decaBDE is a weak inducer of the phase I or phase II
xenobiotic metabolizing enzymes responsible for oxidation and conjugation of many xenobiotic
compounds (Darnerud et al., 2001). Zhou et al. (2001) conducted a study in weanling rats to
14
-------
investigate the mechanism(s) by which decaBDE interferes with thyroid hormone homeostasis.
In this study, Long-Evans female rats (eight animals/dose group) were orally administered
decaBDE (>98% purity) in corn oil at doses of 0, 0.3, 1, 3, 10, 30, 60, or 100 mg/kg-day for
4 consecutive days. Hepatic enzyme activities (ethoxyresorufm O-deethylase [EROD], a marker
for cytochrome P-450 1 Al [CYP-1 Al]; pentoxyresorufin O-deethylase [PROD], a marker for
CYP-2B1) were measured. PROD activity increased only at 1 and 30 mg/kg-day. The lack of
dose dependence was also found for EROD activity, where maximal induction was only 1.8-fold
in the 1 mg/kg-day group, indicating that there were no significant or dose-related effects on
CYP-1 Al or CYP-2B1. However, the presence of active EROD and PROD suggests that there
may be some oxidative debromination of the decaBDE during metabolism, through the activity
ofCYP-lAl and/or CYP-2B1.
Pacyniak et al. (2007) evaluated the ability of BDE-209 to induce CYP-3A11, -2B10, and
-1A1/2. Six C57BL/mice (10 weeks of age) were injected with doses of 10 or 100 umol/kg
(10 or 100 mg/kg) in corn oil for 4 days. The livers were removed 24 hours after the last dose
and the levels of messenger ribonucleic acid (mRNA) measured by Northern blot and branched
deoxyribonucleic acid (bDNA) analyses. The bDNA was considered to be the more accurate of
the two assay systems. Northern blot analysis indicated that the levels of CYP-2B10 were
induced three- and fivefold, respectively, at the two doses tested, while the bDNA results
indicated 3.6-fold and sevenfold inductions. CYP-3A11 did not show a difference with respect
to the dose administered but was induced 4-fold by the Northern blot analysis and 1.7-fold by the
bDNA analysis. The two doses of PBDE-209 induced CYP 1 Al/2 9- and 6-fold, respectively, by
Northern blot analysis and 0.3- and 0.8-fold by bDNA analysis.
3.4. ELIMINATION
As has been mentioned previously, the excretion of decaBDE and its metabolites in rats
is almost exclusively through the fecal and biliary routes. In toxicokinetic studies (Morck et al.,
2003; Hakk et al., 2002; el Dareer et al., 1987), the amounts excreted in urine have consistently
been less than 1% of the dose (about 18% bound to albumin); biliary excretion has accounted for
about 10% of the label found in the feces. There have been no studies that examined the
excretion of BDE-209 in mice. Studies by Staskal et al. (2006) using BDE-47, -99, and -153
found that urinary excretion in male mice involves binding with one or two major urinary
excretory proteins.
In one of the early toxicokinetic studies (el Dareer et al., 1987; NTP, 1986), the fate of
decaBDE in male F344 rats was investigated. The study consisted of four substudies. In
substudy one, groups of 2-month-old male F344 rats (three animals/group) were fed a standard
diet containing unlabeled decaBDE (92% purity) on days 1-7 and 9-11 and the test diet
containing 14C-labeled decaBDE (>98% purity) on day 8. The diets contained 0, 0.0250, 0.0509,
15
-------
0.250, 0.487, 2.49, or 4.99% decaBDE. Based on the rat body weight information reported in
el Dareer et al. (1987) and the average feed intake from the NTP (1986) report, the
corresponding daily doses were 0, 27, 55, 272, 512, 2396, or 4577 mg/kg. Radioactivity was
determined in feces collected daily on study days 9-12.
In substudy two, rats were fed two diets containing low (0.0277% or 25 mg/kg) or high
(4.80% or 4400 mg/kg) amounts of decaBDE or similar amounts of 14C-labeled decaBDE, using
the same protocol as in the first substudy, except that groups of three rats were killed on study
days 10, 11, or 12. Urine and feces were collected from each rat and analyzed for radioactivity.
In substudy three, a group of rats (n = 3) was injected intravenously via the tail vein with
1.07 mg/kg of 14C-labeled decaBDE; urine and feces were collected daily for 3 days and
analyzed for radioactivity 72 hours after dosing. In the fourth substudy, bile-duct-cannulated rats
(n = 5) were injected intravenously with 0.947 mg/kg of 14C-labeled decaBDE. Bile was
collected over a 4-hour period. Radioactivity was determined in each pooled bile sample.
The recovery of radioactivity in the feces ranged from 91-101% of the ingested dose in
the first feeding study and 83-86% in the second feeding study, indicating that the recovery was
not related to the dose of decaBDE or to the time of sacrifice (24, 48, or 72 hours) after
consumption of 14C-labeled decaBDE within a specific substudy. (Note: no reasons were given
for the differences in the radioactivity recovered in these two studies.) Greater than 99% of the
radioactivity was recovered in the feces and gut contents.
After i.v. dosing of 14C-labeled decaBDE (substudy three), 74% of the radioactivity was
recovered in the feces and gut contents at 72 hours. About 63% of the excreted material in the
feces was metabolites and the remaining 37% was unchanged decaBDE. In these rats, only
traces of radioactivity were noted in the urine. In bile-duct-cannulated rats, 7.2% of the
radioactivity intravenously administered appeared in the bile in 4 hours, 1% of which was intact
decaBDE (i.e., 99% of the radioactivity excreted in the bile was in the form of metabolites).
Together, these results show that the vast majority of an oral dose of decaBDE is excreted in the
feces, mostly as unabsorbed material.
Morck et al. (2003) administered orally 14C-radiolabeled decaBDE as a single 3 |imol/kg
(2.9 mg/kg) dose in soy phospholipid/Lutrol F127/water vehicle to male Sprague-Dawley rats
(i.e., conventional rats). An average of 90 and 91% of the radioactivity was excreted in the feces
at 3 and 7 days, respectively. About 71% of the total dose was excreted in the first 24 hours,
another 17% between 24 and 48 hours, and a further 2% between 48 and 72 hours. In bile-duct-
cannulated rats subjected to the same oral treatment, an average of 88% of the dose was excreted
in the feces and 9.5% in the bile within 3 days. Less than 0.1% of the radioactivity was excreted
in the urine. In the cannulated rats, 66% of the total dose was excreted in the feces in the first
24 hours and another 19% during 24-48 hours. In the first 12 hours, 4.4% of the total dose was
recovered in the bile, whereas 1.6, 2, and 0.4% of the total dose were excreted at 12-24, 24-48,
16
-------
and 48-72 hour intervals, respectively. These results indicate that excretion of decaBDE via
feces was the dominant route after the oral dose, and biliary excretion plays a major role in the
systemic elimination of absorbed decaBDE. The urinary excretion of radioactivity was
insignificant.
In the Hakk et al. (2002) study described earlier, <0.02% of the administered decaBDE
dose was excreted daily via the urine in both conventional and cannulated rats, with a total of
0.033 and 0.047%, respectively, of the administered dose excreted over the 72-hour period.
About 9% of the administered dose was excreted in the bile by 72 hours, 6% of which was
eliminated within the first 24 hours, indicating that excretion via the bile was favored over that
of urine.
About 20% of the decaBDE-derived radioactivity in urine from noncannulated rats was
protein bound at 72 hours (compared to >73% not associated with protein). Eighteen percent of
the bound material was bound to albumin, a serum protein with the ability to bind (nonspecific
protein binding) short chain fatty acids. In cannulated rats, 18.2% of the decaBDE-derived
radioactivity was unbound, and all the remaining 68.3% was associated with albumin. Two
polar metabolites were noted but not identified. Under the assumption that the bound materials
are less polar than the unbound materials, this observation supports the concept that a substantial
portion, but not all, of the metabolites in the fecal matter originate from the bile.
About 90% of the biliary radiolabel was associated with an unidentified 79-kDa protein,
with the percent of bound label decreasing from 94 to 87% in bile samples pooled at
1-24 hours, 24-48 hours, and 48-72 hours. Approximately 17% of the protein-bound biliary
radioactivity collected over the first 24 hours was parent compound (and the remainder was
unidentified metabolites); no parent compound was detected at 48 and 72 hours. The percent of
total bound label in the bile samples also declined over time. None of the label in the bile was
found to be unbound.
3.4.1. Half-life Estimates
In computer technicians, the half-life of BDE-209 is estimated to be in the range of a
week (Sjodin et al., 2003; Jakobsson et al., 2002); however, the data to support this estimate are
limited. Sjodin et al. (1999) also found that the levels of BDE-209 in the blood of
occupationally exposed Swedish workers decreased by 66% over a 30-day absence from the
workplace. These authors did not present any half-life estimate in their publication.
The half-lives of hepta- to decaBDE in human serum were estimated by using data from
occupationally exposed workers sampled before, during, and after a vacation period (Thuresson
et al., 2006). The half-lives were found to decrease with increasing bromination. The half-life
of heptaBDE-183 was 94 days, while that for BDE-209 was 15 days.
17
-------
Sandholm et al. (2003) evaluated the kinetics of unlabeled decaBDE in male rats. One
group of rats was dosed orally by gavage with 2 jimol decaBDE/kg (1.9 mg/kg). Another group
of rats was also dosed intravenously with unlabeled decaBDE at the same dose. At specific time
intervals, blood samples from three rats per group were collected according to the following
schedule: group 1 was sampled 1 hour and 24 hours after dosing, group 2 after 3 hours and
48 hours, group 3 after 6 hours and 72 hours, group 4 after 96 hours, group 5 after 120 hours, and
group 6 after 144 hours. Plasma samples were extracted and decaBDE and its metabolites were
quantified. Results from animals orally dosed with unlabeled decaBDE indicated the mean Cmax
in plasma reached 264 pmol/mL (253 ng/g) at 6 hours after dosing and AUC was
12 nmol x hour/mL (11.5 jig x hour/mL). The i.v. data indicated that the clearance was
0.60 mg/minute-kg, and the apparent volume of distribution at steady state was 1.4 L/kg.
Half-lives were determined from the slope of the total concentration-time curve by linear
regression. The disposition of decaBDE in male rats after oral dosing was described by a two-
compartment model with t1/2p and t1/2y values of 6.9 hours and 51 hours, respectively. The results
from the i.v. study were consistent with a three-compartment model with half-lives (ti/2a, ti/2p,
and ti/2y) of 1.6 hours, 12 hours, and 58 hours, respectively (Sandholm et al., 2003). This value
is lower than the 15-day half-life for humans estimated by Thuresson et al. (2006).
Huwe and Smith (2007) modeled the distribution and elimination half-lives of parent
BDE-209 and several debrominated nonhydroxylated metabolites based on assays of plasma,
excreta, liver, and carcass over a 21-day period that followed 21 days of dosing with 0.36 ug/day.
The BDE-209 data showed the best fit to a biphasic depletion pattern. The distribution half-lives
were 1.0 and 1.2 days for liver and plasma, respectively. The elimination half-lives for the same
tissues were 20.2 and 75.9 days, respectively. A plasma first order half-life for octaBDE-207
(7.9 days) was twice that of BDE-209 (3.9 days). In the liver, the first order half-life estimates
for congeners 208, 191, and 197 (~6 days) were greater than those for 206 and 207 (~1 day).
First-order estimates for 207 and 196 were intermediate (4-5 days). These values reflect the
behavior of the parent BDE-209 and the debrominated, nonhydroxylated metabolites after
absorption, distribution, and excretion but do not reflect the distribution and elimination of the
hydroxylated metabolites.
3.5. PHYSIOLOGICALLY BASED TOXICOKINETIC MODELS
Limited information is available on the absorption, distribution, metabolism, and
excretion of decaBDE in experimental animals and in humans. A model for human metabolism
has not been established. Extrapolation of results from laboratory animals to humans using
physiologically based pharmacokinetic (PBPK) models is not possible at this time.
18
-------
4. HAZARD IDENTIFICATION
4.1. STUDIES IN HUMANS—EPIDEMIOLOGY, CASE REPORTS, CLINICAL TRIALS
Studies of decaBDE levels in occupationally exposed groups or studies of decaBDE in
human biological media did not include surveillance of health endpoints.
4.2. SHORT-TERM, SUBCHRONIC, AND CHRONIC STUDIES AND CANCER
BIOASSAYS IN ANIMALS—ORAL AND INHALATION
Inhalation toxicological studies of decaBDE in experimental animals are not available.
4.2.1. Short-term and Subchronic Studies
The short-term and subchronic studies are summarized in Table 4-1.
4.2.1.1. Studies in Rats
Zhou et al. (2001) conducted a study in weanling rats to investigate the mechanism(s) by
which decaBDE interferes with thyroid hormone homeostasis. In this study, Long-Evans female
rats (eight animals/dose group) were orally administered decaBDE (>98% purity) in corn oil at
doses of 0, 0.3, 1, 3, 10, 30, 60, or 100 mg/kg-day for 4 consecutive days. Body weights were
recorded and dosing volumes adjusted daily. Animals were sacrificed 1 day after the last dose.
Serum total thyroxine (T4) and triiodothyronine (T3), serum thyroid stimulating hormone (TSH),
and hepatic enzyme activities (EROD, a marker for CYP-1A1; PROD, a marker for CYP-2B1;
and T4-uridine diphosphate glucuronyl transferase [T4-UDPGT]) were measured (see section 3.3
for data on EROD and PROD). Short-term treatment with decaBDE did not cause any visible
signs of toxicity or any effects on body-weight gain or liver-to-body-weight ratios at any dose
level. DecaBDE (up to 100 mg/kg-day) had no effect on serum T4, T3 or TSH concentration or
on hepatic UDPGT activity. Based on these observations, the highest dose of 100 mg/kg-day is
identified as the no-observed-adverse-effect level (NOAEL).
Carlson (1980) conducted a study to evaluate the induction of xenobiotic metabolism in
rats, following short-term administration of decaBDE. Groups of four male Sprague-Dawley rats
were dosed orally with 0.1 mmol/kg-day (96 mg/kg-day) of decaBDE in corn oil for 14 days.
The decaBDE was synthesized by the complete bromination of diphenyl ether and was reported
to be of "high purity." These animals were probably dosed every other day since the protocol
stated that livers were collected from animals sacrificed 24 hours after the last (seventh) dose.
19
-------
Table 4-1. Oral short-term and subchronic toxicity studies for decaBDE in laboratory animals
Study
Zhou et al.
(2001)
NTP
(1986)
NTP
(1986)
NTP
(1986)
NTP
(1986)
Species, sex, and
sample size
Rat, Long-Evans,
female weanling rats,
8/dose group
Rat, F344/N,
male and female,
5/sex/dose group
Rat, F344/N,
male and female,
10/sex/dose group
Mouse, B6C3F1,
male and female,
5/sex/dose group
Mouse, B6C3F1,
male and female,
10/sex/dose group
Route, dose, and
duration
Gavage (>98% purity);
0,0.3, 1,3, 10,30,60,
or 100 mg/kg-day; 4 days
Diet (99% purity); 0,
472, 928, 1,846, 4,569,
or 9,326 mg/kg-day in
males; 0, 538, 1,061,
2,137, 5,323, or 10,853
mg/kg-day in females; 14
days
Diet (97-99% purity); 0,
191,372,781, 1,536, or
3,066 mg/kg-day in
males; 0, 238, 504, 967,
1,955, or 3,944 mg/kg-
day in females; 13 weeks
Diet (99% purity);
0, 1,027, 2,143, 4,246,
10,536, or 20,994
mg/kg-day in males;
0, 1,146, 2,286, 4,627,
11,348, or 23,077
mg/kg-day in females;
14 days
Diet (97-99% purity);
0, 666, 1,355, 2,659,
5,278, or 10,233 mg/kg-
day in males; 0, 702,
1,437, 2,899, 5,687, or
1 1,566 mg/kg-day in
females; 13 weeks
Observed effects
No observed dose-related
effects on body weight or
liver weight or changes in T3
or T4 levels
None
None
None
None
NOAEL
100 mg/kg-day
9,326 mg/kg-day
in males;
10,853 mg/kg-
day in females
3,066 mg/kg-day
in males;
3,994 mg/kg-day
in females
20,994 mg/kg-
day in males;
23,077 mg/kg-
day in females
10,233 mg/kg-
day in males;
11, 566 mg/kg-
day in females
LOAEL3
Not identified
Not identified
Not identified
Not identified
Not identified
Comments
Survival, final mean
body weights not
adversely affected.
Clinical signs or
gross pathological
effects not noted.
Survival, final mean
body weights not
adversely affected.
Clinical signs or
gross pathological
effects not noted.
Survival, final mean
body weights not
adversely affected.
Survival, final mean
body weights not
adversely affected.
Clinical signs or
gross pathological
examination did not
show any effect.
3LOAEL = lowest-observed-adverse-effect level.
20
-------
Detoxification of O-ethyl O-p-nitrophenyl phenylphosphonothioate, methylation of
p-nitroanisole, levels of NADPH cytochrome c reductase and cytochrome P-450 (CYP-450), and
activities of UDPGT and benzo[a]pyrene hydroxylase were determined in hepatic cytosol or
microsomes. Serum sorbitol dehydrogenase measurements were also performed on blood
samples collected from the tail vein. No significant changes were observed in these enzyme
activities. DecaBDE significantly increased the liver-to-body-weight ratio, indicating liver
enlargement. Sorbitol dehydrogenase activity in the serum was not altered by decaBDE,
suggesting that liver necrosis had not occurred, although other common serum markers for liver
damage were not measured. Because of limitations in the study design, these results are not
adequate for use in the health assessment of decaBDE.
NTP (1986) conducted a 14-day study in rats exposed to decaBDE. F344/N rats (five
animals/sex/dose) were fed diets containing 0, 5,000, 10,000, 20,000, 50,000, or 100,000 ppm
decaBDE (99% purity). Based on the reported body weight information NTP (1986) and U.S.
EPA (1988) default food intake values for F344 male (0.018 kg food/day) and female (0.014 kg
food/day) rats, the corresponding estimated average daily doses were 0, 472, 928, 1,846, 4,569,
or 9,326 mg/kg-day in male rats and 0, 538, 1,061, 2,137, 5,323, or 10,853 mg/kg-day in female
rats. Animals were observed daily and were weighed on days 1, 7, and 14. At the end of the
exposure period, animals were necropsied and several organs and tissues were examined
histologically. No mortality was observed in the rats during the course of the study. Exposure to
decaBDE did not cause any clinical signs of toxicity or adversely affect the final mean body
weights. Gross pathological effects were not noted in any animal at any dose level. The results
of this study indicated a NOAEL of 9,326 mg/kg-day in male rats and 10,853 mg/kg-day in
female rats.
The subchronic effects of decaBDE (97-99% purity) on rats were also investigated in a
13-week study (NTP, 1986). Groups of F344/N rats (10/sex/dose) were administered decaBDE
in the diet at concentrations of 0, 3,100, 6,200, 12,500, 25,000, or 50,000 ppm for 13 weeks.
Based on body-weight information in the NTP (1986) report and U.S. EPA (1988) default food
intake values for F344 male (0.018 kg food/day) and female (0.014 kg food/day) rats, the
corresponding estimated average daily doses were 0, 191, 372, 781, 1,536, or 3,066 mg/kg-day in
male rats and 0, 238, 504, 967, 1,955, or 3,944 mg/kg-day in female rats. Animals were
observed twice daily and body weight, feed consumption, clinical signs, and behavior were
recorded once a week. A necropsy was performed on all animals, including those killed in
extremis, with the exception of those excessively autolyzed or cannibalized. Histologic
examination was performed on major organs and tissues from control and high-dose groups. No
mortality was observed in rats fed decaBDE, and no clinical signs of toxicity were noted.
Compound-related changes in body weight and feed consumption were not observed, and no
21
-------
gross or macroscopic pathological effects were noted in any animal examined. The results
indicate a NOAEL of 3,066 mg/kg-day in male rats and 3,944 mg/kg-day in female rats.
4.2.1.2. Studies in Mice
A 14-day study (NTP, 1986) was also conducted in mice. B6C3F1 mice (five
animals/sex/dose) were fed diets containing 0, 5,000, 10,000, 20,000, 50,000, or 100,000 ppm
decaBDE (99% purity). Based on the reported body weight information (NTP, 1986) and U.S.
EPA (1988) default food intake values for B6C3F1 male (0.0057 kg food/day) and female
(0.0048 kg food/day) mice, the estimated average daily doses were 0, 1,027, 2,143, 4,246,
10,536, or 20,994 mg/kg-day in male mice and 0, 1,146, 2,286, 4,627, 11,348, or 23,077 mg/kg-
day in female mice. Animals were observed daily and were weighed on days 1, 7, and 14.
Necropsy was performed at the end of the exposure period, and several organs and tissues were
examined histologically. Exposure to decaBDE up to 20,994 mg/kg-day in males and
23,077 mg/kg-day in females showed no effects on survival or body weight, and there were no
clinical signs of toxicity. No compound-related gross pathological effects were noted in any
animal in any group. The results of this study indicate a NOAEL of 20,994 mg/kg-day in male
mice and 23,077 mg/kg-day in female mice.
B6C3F1 mice (10 animal s/sex/dose) were fed diets containing 0, 3,100, 6,300, 12,500,
25,000, or 50,000 ppm decaBDE (97-99% purity) for 13 weeks (NTP, 1986). Based on the
mouse body weight information reported (NTP, 1986) and U.S. EPA (1988) default food intake
values for B6C3F1 male (0.0057 kg food/day) and female (0.0048 kg food/day) mice, the
corresponding estimated average daily doses were 0, 666, 1,355, 2,659, 5,278, or 10,233 mg/kg-
day in males and 0, 702, 1,437, 2,899, 5,687, or 11,566 mg/kg-day in females. Animals were
observed twice daily and body weights, feed consumption, clinical signs, and behavior were
monitored once a week. Necropsy was performed on all animals, including those killed in
extremis, with the exception of those excessively autolyzed or cannibalized. Histologic
examination was performed on the organs and tissues from control and high-dose groups. Only
one male and one female mouse fed 12,500 ppm died in the course of the study. There were no
clinical signs of toxicity, and no compound-related effects on body weight and feed consumption
were observed. No gross or macroscopic pathological effects were noted in any animal at any
dose. The results of this study indicated a NOAEL of 10,233 mg/kg-day in males and
11,566 mg/kg-day in females.
4.2.2. Chronic Studies and Cancer Bioassays
NTP (1986) investigated the relationship between ingestion of decaBDE in rats and mice
and tumor development. These chronic oral studies are summarized in Table 4-2. The decaBDE
used in both rats and mice was 94-97% pure, with no detectable brominated dioxins or furans.
22
-------
The major impurities in the decaBDE test material were isolated and identified as two
unspecified congeners of nonaBDE.
4.2.2.1. Study in Rats
Groups of 7- to 8-week-old male and female F344/N rats (50/sex/dose) were exposed to
decaBDE (94-97% purity) in the diet at concentrations of 0, 25,000, or 50,000 ppm for
103 weeks (NTP, 1986). Average daily doses of decaBDE as reported in the study were 0,
1,120, or 2,240 mg/kg-day for male rats and 0, 1,200, or 2,550 mg/kg-day for female rats. The
animals were observed twice daily and clinical signs were recorded once per week. Animals
were weighed once a week for the first 12 weeks, once per month thereafter until week 100 or
101, then every 2 weeks. Mean body weights were calculated for each group. In addition, feed
consumption, morbidity, and mortality were monitored throughout the study period. Animals
found moribund and animals that survived to the end of the study period were sacrificed.
Complete necropsy was performed on all animals, including those found dead during the course
of the study, unless they were excessively autolyzed or cannibalized. Gross and microscopic
examinations were performed on major organs or tissues.
No clinical signs of toxicity were observed in the treated rats. There were no significant
differences in the mean body weight and feed consumption between treated and control animals.
Survival of low-dose male rats was significantly lower than that of the controls after week 102,
but the decrease was not considered to be compound related because the reduction in survival
occurred late in the study and there was a lack of a dose effect.
At necropsy, several nonneoplastic changes were observed. In the liver, an increase in
the incidence of thrombosis was observed in high-dose male rats (1/50, 0/50, 9/49), with no such
increase noted in low-dose males or in any female at any dose level. A dose-dependent, but not
statistically significant, increase in the incidence of degeneration of the liver was also observed
in treated male rats at incidence rates of 13/50, 19/50, and 22/49 in the control, low-dose, and
high-dose groups, respectively. No liver degeneration was observed in female rats. In the
spleen, an increased incidence of fibrosis was seen in males at low dose (8/50) and high dose
(13/49) compared with 5/49 in controls, indicating a dose-dependent increase that was
statistically significant only in the high-dose group. Hematopoiesis was observed at an increased
incidence in the spleen of female rats (control 12/49, low dose 24/48, high dose 17/50), but the
increase was not dose dependent or statistically significant at any dose level. No such increases
were observed in males. In the mandibular lymph node, lymphoid hyperplasia increased in
males in a dose-dependent manner (4/50, 6/50, and 13/49 in the control, low dose, and high dose,
respectively), but the incidence reached statistical significance only at the high dose.
23
-------
Table 4-2. Oral chronic toxicity studies for decaBDE in laboratory animals
Study
NTP
(1986)
NTP
(1986)
Species, sex, and
sample size
Rat, F344/N,
male and female,
50/sex/dose group
Mouse, B6C3F1,
male and female,
50/sex/dose group
Route, dose, and
duration
Diet (94-97% purity);
0, 1,120, or 2,240
mg/kg-day for males; 0,
1,200, or 2,550 mg/kg-
day for females; 2 years
Diet (94-97% purity);
0,3,200, or 6,650
mg/kg-day for males; 0,
3,760, or 7,780 mg/kg-
day for females; 2 years
Observed effects
Increased incidences of thrombosis (1/50, 0/50,
9/49) and degeneration (13/50, 19/50, 22/49) in the
liver of high-dose male rats; dose-dependent
increase in splenic fibrosis in males with statistical
significance at high dose; dose-dependent increase
in lymphoid hyperplasia of the mandibular lymph
nodes in males with statistical significance at high
dose
Dose-related increase in liver neoplastic nodules
statistically significant in low- and high-dose males
and in high-dose females
Increased incidences of granulomas in the liver of
treated males (with statistical significance only at
low dose); centrilobular hypertrophy in males (with
significance at low and high doses); dose-
dependent increase in thyroid follicular cell
hyperplasia (2/50, 10/50, 19/50) in males
(significant at low and high doses); increase in the
incidence of stomach ulcers in females at high dose
Increased incidence of hepatocellular adenoma or
carcinoma (combined) in low-dose males;
marginally but not statistically significant increase
in the thyroid gland follicular cell adenomas or
carcinomas (combined) in males
NOAEL
1,120
mg/kg-day
in males
2,550
mg/kg-day
in females
Not
available in
males
3,760
mg/kg-day
in females
LOAEL3
2,240
mg/kg-day
in males
Not
identified
3,200
mg/kg-day
in males
7,780
mg/kg-day
in females
Comments
Males, but not
females,
exhibited
effects.
3LOAEL = lowest-observed adverse-effect level.
24
-------
No incidence of lymphoid hyperplasia was reported in females at any dose level. An
increased incidence of retinal degeneration was noted in low-dose female rats but not in high-
dose females or males at any dose level. This lesion was attributed to greater exposure of the
female rats to fluorescent light and was not considered to be treatment related. Acanthosis
(increased thickness of the epithelial surface) of the forestomach was observed at increased
incidence in treated males but not in females, but the incidence was not significant. Both male
and female rats demonstrated a dose-dependent decrease in the incidence of C-cell hyperplasia
in the thyroid gland (12/50, 6/49, and 2/49 in males and 14/50, 7/49, and 2/50 in females at
control, low-dose, and high-dose groups, respectively). Decreases in the C-cell hyperplasia
would not be considered biologically adverse since other thyroid effects were not observed in
these rats. No other significant pathological changes were observed.
Histopathologic examination revealed a significant increase in the incidence of neoplasia
in the rat, and several organs or tissues were adversely affected. There was a dose-dependent
increase in the incidence of neoplastic nodules in the livers of both male and female rats. The
incidence of these lesions was 1/50, 7/50, and 15/49 in males and 1/50, 3/49, and 9/50 in females
in control, low-dose, and high-dose groups, respectively. The incidence reached statistical
significance at both treatment doses in males and at the high dose in females.
At the time the NTP (1986) study was conducted, the term neoplastic nodule was used to
describe abnormal cellular masses in the livers of rats characterized by loss or distortion of
normal cellular architecture (Maronpot et al., 1986). Some of those nodules would now be
described as benign hepatocellular adenomas in rats (Wolf and Mann, 2005). However, there is
no complete equivalency between the neoplastic nodule of the past and the hepatocellular
adenoma term of today. Some of the neoplastic nodules from the NTP (1986) study might now
be classified as foci of cellular alteration or hyperplasia rather than adenomas (Maronpot et al.,
1986). Adenomas and foci of cellular alteration are considered to be preneoplastic lesions,
whereas hyperplastic lesions represent secondary nonneoplastic changes (Maronpot et al., 1986).
The assumption that the hepatic neoplastic nodules from the NTP (1986) bioassay are equivalent
to hepatic adenomas under the current NTP lexicon is a conservative interpretation of the data.
A slight increase in mononuclear cell leukemia in treated rats was observed with
incidences in males of 30/50, 33/50, and 35/50 at control, low, and high doses, respectively, and
in females of 14/50, 21/50, and 18/50 at control, low, and high doses, respectively. However, the
increase was not considered to be biologically significant because of the marginal and
insignificant increase in treated rats of both sexes and the high incidence in control rats. The
incidence of hepatocellular carcinomas was low in male and female rats and was apparently not
compound related. Other tumors observed included splenic sarcomas, pancreatic acinar cell
adenomas, carcinomas in Zymbal's gland, and osteosarcomas, but these were considered to be of
25
-------
no significance because of high historical incidence, lack of dose-response trend in the tumor
incidence, or lack of statistically significant responses.
Based on the results from this 2-year study in rats (NTP, 1986), exposure to decaBDE in
the diet did not cause compound-related effects on survival or any significant effects on body
weight or food consumption. However, treatment resulted in several nonneoplastic changes at
the high dose, including thrombosis and degeneration of the liver in males, fibrosis in the spleen
in males, and lymphoid hyperplasia in the mandibular lymph node in males. Dose-dependent
increases in the incidence of neoplastic nodules in liver of males and females provide some
evidence of carcinogenicity of decaBDE in rats. Based on these results and on increased
incidence of thrombosis and degeneration in the liver, splenic fibrosis, and lymphoid hyperplasia
of the mandibular lymph nodes, a NOAEL for systemic toxicity was 1,120 mg/kg-day and the
lowest-observed-adverse-effect level (LOAEL) was 2,240 mg/kg-day for males. Female rats
appeared to be less sensitive to the systemic toxicity of decaBDE at the doses used in this study.
Based on this finding, the NOAEL for systemic toxicity in females was 2,550 mg/kg-day, with
no LOAEL established. The dose-related increase in nonmalignant liver tumors (neoplastic
nodules) in both males and females provides some evidence of carcinogenicity of decaBDE in
rats.
4.2.2.2. Study in Mice
Groups of 9-week-old male and female B6C3F1 mice (50/sex/dose) were administered
decaBDE (94-97% purity) in the diet at doses of 0, 25,000, or 50,000 ppm for 103 weeks (NTP,
1986). Average daily doses of decaBDE as reported in the study were 0, 3,200, or 6,650 mg/kg-
day in male mice and 0, 3,760, or 7,780 mg/kg-day in female mice. The animals were observed
twice daily, and clinical signs were recorded once per week. Animals were weighed once a
week for the first 12 weeks and once per month thereafter. In addition, feed consumption,
morbidity, and mortality were monitored throughout the study period. All survivors were
sacrificed during weeks 112-113. Complete necropsy was performed on all animals, including
those found dead during the course of the study, unless they were excessively autolyzed or
cannibalized. Gross and microscopic examinations were performed on major organs or tissues.
There were no significant differences in survival among any groups of mice of either sex
at terminal sacrifice. However, loss of control male mice (presumably due to fighting) was
significant during the first 15 months of the study. There were no compound-related clinical
signs of toxicity or changes in body weight or food consumption.
At necropsy, several nonneoplastic changes were observed in tissues of treated mice. In
the liver, the incidence of granulomas in control, low-, and high-dose mice was 8/50, 22/50, and
12/50 in males and 23/50, 27/50, and 24/50 in females, respectively. Although increases were
observed in the treated animals of both sexes compared with controls, no consistent dose
26
-------
response for this effect was evident, and the effect was not associated with a statistically
significant positive trend. Thus, the toxicological significance of this effect is unclear.
Significant increases in the incidence of centrilobular hypertrophy were also observed in the
liver of male mice at high and low doses (0/50, 34/50, and 32/50 in the control, low dose, and
high dose, respectively). Although the increases were comparable in the low- and high-dose
males and were not observed in female mice at any dose level, the high incidence in the treated
males indicates that the effect was treatment related. In the thyroid gland, a dose-dependent and
statistically significant increase (at both dose levels) in the incidence of follicular cell
hyperplasia was observed in males (2/50, 10/50, and 19/50, respectively, in the control, low-
dose, and high-dose groups). In the females, the incidence was increased in the low- (9/50) and
high- (7/49) dose groups compared to 4/50 in the control group, but the increase was not dose
dependent or statistically significant at any dose level. Only high- but not low-dose female mice
exhibited a significant increase in the incidence of stomach ulcers (1/50, 1/50, and 8/50 in the
control, low-dose, and high-dose females, respectively). In males, the incidence in treated mice
was comparable with that in control mice: 5/49, 3/50, and 5/50 in the control, low-dose, and
high-dose males, respectively.
Histopathological examination revealed a significantly increased incidence of neoplasia
in mice, with the liver appearing to be the main target organ for decaBDE. In the liver, increases
(although not statistically significant) in the incidence of hepatocellular adenoma were observed
at similar rates in low- and high-dose males (4/50, 12/50, and 12/50 in control, low-dose, and
high-dose groups, respectively). Other changes observed in male mice included an increased
incidence of hepatocellular carcinoma (5/50, 14/50, and 8/50 in the control, low-dose, and high-
dose groups, respectively) that was not statistically significant at either dose. The combined
incidence of hepatocellular adenomas or carcinomas in male mice, after correction for survival,
significantly increased at low dose but not at high dose; the responses amounted to 8/50, 22/50,
and 18/50 in control, low-dose, and high-dose groups, respectively. As indicated in the study
report, these findings may have been influenced by the large number of early deaths in control
male mice compared with treated mice, thereby decreasing the significance of the hepatocellular
adenomas or carcinomas seen in the treated male mice. Consequently, the apparent statistically
significant increase in the hepatocellular adenomas and carcinomas (combined) at the high dose
(18/50 versus 8/50) based on regular rate comparison test (i.e., Fisher's exact test) was no longer
significant when the life table test or incidental tumor test was used to control for intercurrent
mortality. The trend tests from the latter two tests also failed to show statistical significance in
the combined hepatocellular adenomas or carcinomas in male rats. The incidence of
hepatocellular adenomas or carcinomas (combined) in female mice was not statistically
significant (8/50, 13/50, and 13/50 in control, low-, and high-dose mice, respectively). In the
thyroid gland, follicular cell adenomas or carcinomas (combined) were slightly, but not
27
-------
significantly, increased in treated mice of both sexes over the corresponding control mice; the
incidence was 0/50, 4/50, and 3/50 in males and 1/50, 3/50, and 3/50 in females in the control,
low-, and high-dose groups, respectively.
These results from the NTP (1986) chronic carcinogenicity study in mice show that
decaBDE treatment caused liver granulomas, liver hypertrophy, and thyroid gland follicular cell
hyperplasia in males only and stomach ulcers only in females. Males, but not females, had a
statistically significant increase in the incidence of hepatocellular adenomas and carcinomas at
the low dose but not at the high dose. Based on these results and on increased incidence of liver
granulomas and centrilobular hypertrophy and statistically significant and dose-dependent
increases in the thyroid gland follicular cell hyperplasia in male mice administered 3,200 mg/kg-
day or higher, no NOAEL for systemic effects for males was established. Female mice were less
sensitive to the systemic toxicity of decaBDE at the doses used in this study; however, the IRIS
assessment authors identified a NOAEL for a portal-of-entry effect for females of 3,760 mg/kg-
day and a LOAEL of 7,780 mg/kg-day based on an increase in the incidence of stomach ulcers.
4.3. REPRODUCTIVE/DEVELOPMENTAL STUDIES—ORAL AND INHALATION
Reproductive/developmental toxicity studies are not available in humans by the oral or
inhalation routes of exposure. No reproductive/developmental animal toxicity studies were
identified by the inhalation route of exposure. Oral reproductive and developmental studies in
experimental animals are summarized in Table 4-3.
4.3.1. Studies in Rats
Hardy et al. (2002) conducted a developmental toxicity study in rats by using a composite
of three lots of a commercial flame retardant (97.34% decaBDE and 2.66% nonaBDE and
octaBDE) produced by three manufacturers. In this study, Sprague-Dawley rats (25 mated
females per dose group) were administered decaBDE in corn oil by gavage at doses of 0, 100,
300, or 1,000 mg/kg-day during gestation days 0 through 19. Dams were observed daily for
morbidity, mortality, and signs of injury. Maternal body weight, body-weight gain, and food
consumption were monitored. Dams were sacrificed on day 20 of gestation, and liver weights,
gravid uterine weights, and the number of corpora lutea, implants, fetuses, and resorptions were
recorded. The placenta and fetuses were examined for gross abnormalities, and histologic
examinations were performed.
All dams survived decaBDE treatment until scheduled sacrifice. There were no adverse
treatment-related effects observed in maternal clinical findings, body weight, or body-weight
gain. Although a slight but statistically significant increase in food consumption was observed at
1,000 mg/kg-day at time intervals up to day 12 of gestation, the authors did not consider this
indicative of an adverse effect of treatment.
28
-------
Table 4-3. Oral developmental and reproductive studies for decaBDE in laboratory animals
Study
Hardy et al.
(2002)
Viberg et al.
(2007)
Viberg et al.
(2003a)
Tseng et al. (2006)
Rice et al. (2007)
Species, sex,
and sample size
Rat, Sprague-
Dawley,
pregnant female,
25/dose group
Rat, Sprague-
Dawley, male, 20
rats from 3-5
litters/treatment
group
Mouse, NMRI,
male, 10 mice
from 3-5 litters/
treatment group
Mouse, CD-I,
male, 50/dose
group
Mouse,
C57BL6/J, 3
males and 3
females from 1 1-
13
litters/treatment
group
Route, dose, and
duration
Gavage (97.34%
purity); 0, 100, 300, or
1,000 mg/kg-day;
gestation days 0-19
Single dose gavage
(>98% purity in 20%
fat emulsion); 0, 6.7,
or 20. 1 mg/kg on PND
3
Single oral gavage
(>99% purity in 20%
fat emulsion); 0, 2.22,
or 20.1 mg/kg (PNDs
3 and 19); 0, 1.34,
13.4, or 20.1 mg/kg
(PND 10)
Oral gavage (98%
purity in corn oil); 0,
10, 100, 500, or 1,500
mg/kg-day; PNDs 21-
70
Micropipette
administration (99.5%
purity in 20% fat
emulsion); 0, 6, or 20
mg/kg-day; PNDs 2-
15
Observed effects
No statistically significant maternal or
developmental treatment-related effects
Significant dose-related disruption in
habitation (changes in locomotion,
rearing, and total activity) at 2 months
in rats, following exposure to 6.7 and
20.1 mg/kg on PND 3
Significant dose-related disruption in
habitation (changes in locomotion,
rearing, and total activity) at 2, 4, and 6
months in mice, following exposure to
20.1 mg/kg on PND 3
Reduced amplitude of sperm lateral
head displacement, reduced sperm
mitochondria! membrane potential,
increased sperm H2O2 generation
Decreased number of male and female
pups performing the palpebral reflex on
PND 14, increased struggling behavior
on PND 20, effects on grip strength on
PNDs 14 and 16, effects on locomotor
activity on PND 70
NOAEL
1,000
mg/kg-day
Not
identified
2.22 mg/kg
100 mg/kg-
day
Not
identified
LOAEL
Not
identified
6.7 mg/kg
20.1 mg/kg
500 mg/kg-
day
6 mg/kg-day
Comments
Single dose
experiment
Single dose
experiment
29
-------
No statistically significant differences were observed in maternal absolute or relative
liver weights between treatment and control groups. At necropsy, gross examination of the dams
revealed no adverse effect of treatment with decaBDE. Number of dams with viable fetuses,
mean number of corpora lutea, number of implantation sites, percent preimplantation loss per
dam, number of viable fetuses, and gravid uterine weights were not adversely affected by
decaBDE treatment.
A statistically significant increase in the mean number of early resorptions per dam was
observed in the 1,000 mg/kg-day group compared to controls. Based on the lack of a consistent
dose response for this effect (the mean number of early resorptions per dam was 0.6, 0.6, 0.5,
and 1.4 at 0, 100, 300, and 1,000 mg/kg-day, respectively), lack of a statistically significant
positive trend associated with the effect, and the historically high incidence of this effect (0.5-
1.4) for the laboratory, these effects are not considered to be of toxicological significance.
Examination of the results indicated a marginal increase in the postimplantation loss/dam of 7
and 9% at 300 and 1,000 mg/kg-day, respectively, compared with 4% in controls and at 100
mg/kg-day. However, this effect was not associated with a statistically significant positive trend.
A slight, but statistically not significant, decrease in the percentage of viable fetuses per implant
was seen
(96, 96, 93, and 91% in the control, 100, 300, and 1,000 mg/kg-day groups, respectively). Fetal
body weights, crown-rump ratio, and fetal sex ratio were not different between treatment and
control groups. No adverse decaBDE treatment-related effects were identified during fetal
external, skeletal, or visceral examinations. DecaBDE treatment, therefore, did not produce any
evidence of maternal or developmental toxicity up to the highest dose tested of 1,000 mg/kg-day.
The NOAEL for maternal and developmental toxicity in this study was 1,000 mg/kg-day, the
highest dose tested.
BDE-209 (>98% purity) in a fat emulsion was administered by gavage to 3-day-old
Sprague-Dawley rats at 0, 6.7, or 20.1 mg/kg (Viberg et al., 2007). A total of 20 rats were
picked from three to five different litters in each treatment group. Motor activity was measured
for a 60-minute period, divided into three 20-minute periods, in rats at the age of 2 months.
Motor activity tests measured locomotion (horizontal movement), rearing (vertical movement),
and total activity (all types of vibration within the test cage [i.e., those caused by rat movements,
shaking/tremors, and grooming]). There were no clinical signs of toxicity in the BDE-209-
treated rats at any given time during the experimental period, nor was there any significant
difference in body-weight gain or adult weight between controls and rats treated with BDE-209.
In control rats, there was a distinct decrease in locomotion, rearing, and total activity at 2 months
of age, indicating habituation in response to the diminishing novelty of the test chamber over the
60-minute test period. Habituation is defined as the ability of the animals to adapt to a new
environment and is characterized as initial investigation and exploration of their surroundings
30
-------
followed by gradual acclimatization and acceptance of the new area. Two-month-old rats
exposed to 20.1 mg/kg BDE-209 on PND 3 displayed significantly less activity for all three
behavioral variables during the first 20-minute test period compared with controls, while during
the third 20-minute period (40-60 minutes) they were significantly more active than the control
animals. Rats receiving the low dose of BDE-209 (6.7 mg/kg) showed significantly increased
locomotion activity during the second 20-minute period, significantly decreased rearing activity
during the first and second 20-minute periods, and significantly higher total activity during the
first and second 20-minute periods compared with the control rats. The LOAEL in this study
was 6.7 mg/kg for significant changes in spontaneous motor behavior (locomotion, rearing, and
total activity) in 2-month-old rats given BDE-209 on PND 3.
Immediately after the spontaneous behavior tests, nicotine-induced behavior was studied
to determine whether changes in spontaneous behavior in adult rats neonatally exposed to
BDE-209 included effects on development of the cholinergic system and thereby altered the
response in the adult animal to the cholinergic agent nicotine (Viberg et al., 2007). The rats
were given a single subcutaneous injection of 80 jig nicotine base/kg, a dose known to cause an
increase in activity in experimental animals, and were immediately tested again for nicotine-
induced motor behavior with regard to locomotion, rearing, and total activity during another
60-minute period divided into three 20-minute periods.
Pair-wise testing between the nicotine-injected and saline-injected rats showed, as
expected, a significant increase in response to nicotine in the neonatally vehicle-treated rats
during the first 20-minute period (60-80 minutes) for all three variables (locomotion, rearing,
and total activity). In contrast, the nicotine-injected rats exposed to 20.1 mg/kg of BDE-209 on
PND 3 showed significantly decreased activity for all three tests (locomotion, rearing, and total
activity) during the first 20-minute period (60-80 minutes) compared with the rats neonatally
exposed to the high BDE-209 dose and injected with saline. The authors concluded that
neonatal exposure to BDE-209 on PND 3 affects adult spontaneous behavior and also affects the
cholinergic system, seen as changes in the adult rats' response to the cholinergic agent nicotine.
4.3.2. Studies in Mice
Viberg et al. (2003a) assessed the potential of decaBDE to affect the developing central
nervous system. The neurotoxic effects of decaBDE on spontaneous motor behavior of NMRI
male mice were investigated in adult animals exposed to a single oral dose as neonates. Uptake
of radiolabel by the brain of the neonatal mice orally administered 14C-labeled BDE-209 on PND
3, 10, or 19 (i.e., at different stages of neonatal mouse brain development) was also measured to
determine if there were age-related differences in tissue toxicokinetics that might correlate with
the neurodevelopmental effects evaluated.
-------
In this behavioral study, 3-day-old and 19-day-old male mice were given a single dose of
0, 2.22, or 20.1 mg/kg body weight decaBDE (purity estimated to be >99%) in a
20% (weight/weight) emulsion vehicle of egg lecithin-peanut oil and water. Ten-day-old mice
received 0, 1.34, 13.4, or 20.1 mg/kg. The spontaneous behavior test (measuring locomotion,
rearing, and total activity) was conducted in 10 mice randomly selected from the litters in each
treatment group at 2, 4, and 6 months of age. There were three to five litters (four to seven
males per litter) in each of the dose groups. The behavior variables were measured for a 60-
minute period divided into three consecutive 20-minute periods. In order to study time-
dependent changes in habituation, an habituation ratio was calculated by dividing the motor
behavior measures from the 40- to 60-minute observation period by those from the 0- to 20-
minute period and multiplying by 100 for each of the three different variables: locomotion,
rearing, and total activity. The habituation ratios from 2-, 4-, and 6-month-old mice within each
treatment group were then compared. An increase in habituation ratio indicates decreased
capability to habituate to a novel environment. Data for the three spontaneous behavior
variables and habituation ratio are only available in graphic form and could not be used for
quantitative assessment.1
Treatment with decaBDE caused no clinical signs of toxicity at any time during the
experimental period. Body weight and body-weight gain were not significantly different
between decaBDE- and vehicle-treated mice in the three different age groups. Control mice
treated on PND 3, 10, or 19 exhibited normal habituation profiles. Pair-wise testing between
adult mice exposed to 20.1 mg/kg on PND 3 and control groups indicated significant changes in
all three spontaneous behavior variables at 2, 4, and 6 months of age. For the first 20 minutes,
mice receiving 20.1 mg/kg displayed significantly less activity for locomotion, rearing, and total
activity compared with controls. During the third 20-minute period, exposure of mice to
20.1 mg/kg on PND 3 caused significantly more activity for locomotion, rearing, and total
activity than the controls at 2, 4, and 6 months. The only effect noted in mice exposed to
2.22 mg/kg was a significant decrease in total activity in the first 20-minute test period compared
with the controls at 2 months of age. However, total activity returned to control level during the
third 20-minute period. The lower dose of 2.22 mg/kg did not elicit any significant differences
in these three variables compared with controls at 4 months of age. Lower activity was observed
at 2.22 mg/kg during the first 20-minute period for the rearing variable at 6 months of age
compared with controls, again returning to control level during the third 20-minute period. Mice
exposed neonatally up to 20.1 mg on either PND 10 or 19 did not show any significant
differences in any of the variables after 2, 4, or 6 months compared with controls. The authors
Attempts to obtain numerical values and other information on the data from the authors were not successful.
32
-------
indicated that the absence of effects on spontaneous activity in mice treated on PNDs 10 and 19
suggests that there is a critical window for the induction of the observed behavioral disturbances.
The habituation ratio calculated from the three spontaneous motor behavior variables
(locomotion, rearing, and total activity) significantly increased after 2, 4, and 6 months of age in
mice exposed on PND 3 to 20.1 mg/kg. Mice exposed on PND 3 to 2.22 mg/kg did not show a
significant decrease in habituation capability with age. The decrease observed in the habituation
capability in the adult mice exposed neonatally to the high dose indicated that the neurotoxic
effect of neonatal decaBDE exposure was persistent and also worsened with age. The NOAEL
in this study was 2.22 mg BDE-209/kg, and the LOAEL was 20.1 mg/kg for significant changes
in spontaneous motor behavior and decreased habituation capability for locomotion, rearing, and
total activity, worsening with increasing age.
Neonatal exposure to 14C-decaBDE (specific activity 17.5 mCi/mmol) in mice on PND 3,
10, or 19 revealed that the labeled substance was absorbed and distributed to the brain, heart,
and liver. However, the amounts in the liver 24 hours after dosing on PNDs 3,10, and 19 were
12.6, 9.4, and 5.8%, respectively. The lower amount of BDE-209 measured as the dosing day
occurred later suggests that gastrointestinal absorption may have decreased as the
gastrointestinal tract matured over the first 19 days after birth. Only a small fraction of the dose
reached the brain or heart 24 hours after dosing, especially with dosing on day 19 (see Viberg et
al. [2003a] in section 3.2.2). However, results indicated that the amount of radioactivity
increased significantly in the brain over a 7-day period in animals exposed on PNDs 3 and 10 but
not in those exposed on PND 19. The radioactivity in the liver and heart was the same or
decreased during the 7-day period following oral administration on PNDs 3, 10, and 19. The
observed increase in radioactivity in the brain over the 7-day period suggests slow transport of
decaBDE and/or its metabolites to the brain, which may account for the fact that administration
on day 3 caused a more significant impact on habituation than administration on PND 10 or 19.
The radiolabel data showed that the amount of decaBDE in the brain 24 hours after dosing on
PNDs 3, 10, and 19 was 0.5, 0.4, and 0.06%, respectively. The radioactivity was significantly
greater 7 days after BDE-209 administration on PNDs 3 and 10 compared with 24 hours after
administration. The amount of radioactivity measured in the brain 7 days after dosing on PNDs
3 and 10 (PNDs 10 and 17) was 0.7 and 1.1%, respectively. There was no change in the amount
of radioactivity measured in the brain of animals 7 days after dosing on PND 19.
Rice et al. (2007) examined the effects of BDE-209 (99.5% purity) on developmental
milestones, sensorimotor behaviors, serum T4 levels, and locomotor activity in male and female
C57BL6/J mice administered doses of 0, 6, or 20 mg/kg-day. The BDE-209, in a 20% egg
lecithin/peanut oil/water emulsion, was administered to the pups from PNDs 2-15 by using a
micropipette to avoid gavaging neonatal animals. There were no delays in postnatal
developmental milestones (pinna detachment, incisor eruption, eye opening, vaginal opening, or
33
-------
testes descent) from BDE-209 treatment in male or female mice. There were no effects of
exposure on anogenital distance or crown-rump length.
The authors developed a special functional observational battery (FOB), suitable for
neonatal mice, to examine a series of home-cage, reflexive, and sensorimotor behaviors,
measured on PND 14, 16, 18, or 20. Three of the FOB endpoints were affected by BDE-209
exposure: palpebral reflex, forelimb grip, and struggling behavior during handling. On PND 14,
significantly fewer male and female pups in the 6 and 20 mg/kg-day groups performed the
palpebral reflex compared with controls. On PNDs 14 and 16, fewer male pups in the 20 mg/kg-
day group performed an effective forelimb grip compared with same-sex controls. Male and
female pups in the 6 mg/kg-day group struggled significantly more on PND 20, while they were
being handled for the FOB, compared with controls and the high-dose group.
Rice et al. (2007) also examined the locomotor activity of male and female mice in a
novel environment over a 2-hour period on PND 70 and at approximately 1 year of age (one
male and one female from each of 9-13 litters were evaluated). Locomotor activity declined
over the course of the 2-hour assessment in PND 70 males and in females in the control and
treated groups. However, the rate of decline was significantly different (p = 0.04) in male mice
exposed to 6 and 20 mg/kg-day compared with control animals, an effect that was most
pronounced in the high-dose males during the first 1.5 hour of the 2-hour activity session. There
was a non-dose-related decline in the locomotor activity in the female mice during the 2-hour
activity session. Unlike the males, they became hypoactive compared with the controls. There
was no effect of BDE-209 treatment on the locomotor activity of the 1-year old male and female
mice.
The effect of BDE-209 treatment on serum T4 in PND 21 offspring was also investigated
in this study. A dose-related reduction in serum T4in comparison with controls occurred in
males but not in females.
The LOAEL in this study was 6 mg/kg-day, the lowest dose tested, for decrease in the
percent of male and female pups performing the palpebral reflex on PND 14, for increased
struggling behavior of male mice on PND 20, for decreased T4 levels in male mice, and for
effects on locomotor activity of male mice on PND 70.
Tseng et al. (2006) studied the effects of BDE-209 on mouse sperm function, DNA
content, and histopathology of the testes. CD-I male mice (50/dose group) were fed decaBDE
(98% purity) by gavage in corn oil at 0, 10, 100, 500, or 1,500 mg/kg-day from PNDs 21-70.
Body weight, body-weight gain, and absolute and relative weights of the testis, epididymis,
cauda epididymis, and seminal vesicle of treated animals were not significantly different from
controls at sacrifice on PND 71. Morphology of the testicular tissues appeared normal in all
treated groups compared with controls. DNA content in testis cells was unaffected by treatment
with BDE-209. No significant differences in sperm motility (expressed as the ratio between the
34
-------
number of motile sperm and total number of sperm), sperm count, or morphology were seen
among the groups exposed to BDE-209 and the controls. Sperm motion velocity parameters
were measured, including curvilinear velocity, straight-line velocity, and amplitude of the lateral
head displacement (ALH). The only effect seen on sperm motion velocity was a significant
decrease in ALH at 500 and 1,500 mg/kg-day. Significant increase in the generation of hydrogen
peroxide in the sperm of sexually mature mice occurred at 500 and 1,500 mg/kg-day. The
mitochondrial membrane potential (MMP) of sperm cells, a predictor of sperm fertility potential,
was assessed using a lipophilic cationic compound, JC-1, which possesses the ability to
differentially label mitochondria with high and low membrane potential (Gravance et al., 2001).
Mice exposed to 500 and 1,500 mg/kg-day were found to have a significant decrease in high
MMP sperm. In addition, the MMP was negatively and significantly associated with the
generation of sperm hydrogen peroxide. The NOAEL in this study was 100 mg/kg-day and the
LOAEL 500 mg/kg-day for decrease in ALH and MMP and increased generation of hydrogen
peroxide in the sperm of adult mice.
4.4. OTHER ENDPOINT-SPECIFIC STUDIES
4.4.1. Receptor Site Interactions
Some studies of halogenated aromatic compounds show that they exert an influence on
cells by interacting with membrane receptor sites and activating cellular transcription factors.
Transcription factor complexes then initiate DNA synthesis, allowing the cell to respond to the
extracellular signal by producing a series of mRNAs that in turn produce a variety of proteins.
This process is termed signal transduction. The structural similarities between PBDEs and
polychlorinated biphenyls (PCBs) suggest that PBDEs might activate the aryl hydrocarbon
receptor (AhR) and the estrogen receptor (ER). Based on the data from the well-studied PCBs
and dioxins, the activation of these receptors is associated with immunosupression, reproductive
effects, and carcinogenesis (Klaassen, 1996; Bock, 1994), all endpoints of interest for PBDEs.
Studies of receptor interactions of BDE-209 with the AhR, ER, pregnane X receptor (PXR), and
steroid X receptor (SXR) are summarized in the sections that follow. As a group, these studies
indicate that the ability of BDE-209 to activate signal transduction responses via the receptors
studied is minimal or negligible.
4.4.1.1. Aryl Hydrocarbon Receptor Studies
Although in vivo studies investigating mechanisms of liver toxicity in animals have not
been identified, several in vitro systems have evaluated the ability of decaBDE to interact with
the AhR by using liver tissues or cell lines. Brown et al. (2004) investigated the potential of
decaBDE to bind to and activate the AhR (using a gel retardation assay) and the ability of
decaBDE to induce the expression of dioxin-responsive genes (using the reporter gene based
35
-------
Chemical-Activated Luciferase expression [CALUX] assay). The decaBDE used in this study
was 85.5% purity, and the identities of the other components present in the preparation were not
reported. Incubation of decaBDE with hepatic cytosol prepared from guinea pigs and
^9
subsequent incubation with [ P]-labeled dioxin-responsive element (DRE)-containing
oligonucleotide, followed by resolution of the ligand:AhR:[32P]DRE complex by gel retardation
analysis, indicated that decaBDE (20 jiM) stimulated AhR-DNA binding but to a much lower
level (26%) than 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). DecaBDE demonstrated some
activity in the CALUX assay. Based on the results from gel retardation and reporter gene assays,
decaBDE may induce AhR-dependent DNA binding and gene expression but with very low
potency if the activity observed is solely attributable to decaBDE. However, the low but
measurable activity may have been caused by contaminants since the test material was impure.
The possible dioxin-like effects of decaBDE or induction of genes that encode
metabolizing enzymes have also been investigated by Peters et al. (2004). AhR-mediated
induction of CYP-1 Al and -1B1 were studied in human breast carcinoma MCF7 cells by using
EROD activity as a marker for CYP-1 Al and -1B1 activity. DecaBDE (>98% purity) did not
induce EROD activity and, therefore, was not found to be an AhR agonist in these cells
incubated for 72 hours with decaBDE at concentrations that were not cytotoxic (up to 10 |iM).
Additionally, exposure of the cells to decaBDE did not induce mRNA levels of CYP-1 Al or
-1B1, indicating no effect of decaBDE on AhR gene expression.
Villeneuve et al. (2002) evaluated the dioxin-like potency of decaBDE (99% purity) at
environmentally relevant concentrations (up to 500 ng/mL) by using in vitro luciferase assays
with H4IIE-luc recombinant rat hepatoma cells. DecaBDE did not induce AhR-dependent gene
expression. The authors calculated that most PBDEs, including decaBDE, were at least
10,000 times less potent than TCDD for inducing AhR-mediated responses in vitro.
The affinity of decaBDE for rat hepatic AhR was evaluated through competitive binding
assays (Chen et al., 2001). Results indicated that decaBDE has no AhR-binding activity. The
ability of decaBDE to induce hepatic CYP-450 enzyme activity by means of EROD assays in
human, rat, chick, and rainbow trout cells was also determined in this study. Incubation of
decaBDE (of unspecified purity) with cultures of primary rat hepatocytes, primary chick embryo
hepatocytes, two human cell lines (hepatoma HepG2 and intestinal Caco-2), and one rat cell line
(hepatoma H4IIE) showed that decaBDE did not induce EROD activity in any cell line tested.
Moreover, decaBDE was almost completely inactive in a gel retardation assay, suggesting that
decaBDE did not activate the AhR.
Pullen et al. (2003) investigated the effect of decaBDE on CYP-450 activity in mouse
hepatocytes. Incubation of decaBDE (of unspecified purity) with hepatocytes isolated from
C57BL/6 mice for 24 hours did not induce CYP-1 Al (measured as EROD activity), indicating
that decaBDE does not act as a specific ligand for the AhR and, thus, exerts no effect via the
36
-------
AhR pathway as do many of the polycyclic aromatic hydrocarbons. Together, these studies
suggest that decaBDE has very limited potential to activate the AhR signal transduction
pathway, which is an important step in the mode of action through which many persistent
aromatic hydrocarbons impact cell regulation and cell maintenance (Klaassen, 1996).
4.4.1.2. Estrogen Receptor Studies
ER-mediated (estrogenic) effects of decaBDE at environmentally relevant concentrations
were investigated by Villeneuve et al. (2002) in an in vitro luciferase assay system. Incubation
of decaBDE (99% purity) at concentrations up to 500 ng/mL with MVLN recombinant human
breast carcinoma cells for 72 hours showed that decaBDE did not exhibit estrogenic effects.
Most PBDEs, including decaBDE, were at least 50,000 times less potent than l?p-estradiol (E2)
for inducing ER-mediated gene expression. This study also examined the ability of decaBDE to
displace E2 or testosterone from serum proteins in a competitive hormone displacement assay.
Incubation of decaBDE at concentrations up to 2.5 |ig/mL in the presence of 3H-estradiol or 3H-
testosterone and hormone-stripped carp serum for 15 hours indicated that decaBDE did not
cause any significant displacement of these hormones in vitro. These results suggest that
decaBDE and other PBDEs are not likely to elicit E2-mediated gene expression at the exposure
levels currently observed in fish and wildlife.
4.4.1.3. Other Receptors
Pacyniak et al. (2007) conducted a study of the PXR and its human counterpart SXR by
using HepG2 cells transvested with the appropriate complementary DNA (cDNA), the receptor
response elements, and a luciferase reporter vector. The cultured cells were exposed to 0, 0.1, 1,
10, or 100 uM concentrations of BDE-209. With the PXR there was a very slight dose-related
increase in relative luciferase activity that achieved significance for only the highest
concentration. With the SXR, there was no impact of luciferase for any concentration other than
the 100 mM preparation. Accordingly, BDE-209 appears to be a weak inducer for PXR and
SXR. The authors also compared the response in PXR knock-out mice (10-12 weeks old) with
the wild-type mice and found that CYP-3 Al 1 was induced to a greater extent in the wild-type
animals, indicating that BDE-209 seems to be a ligand for the PXR. However, even in the
knock-out mice, some CYP-3 Al 1 protein was identified, suggesting that the PXR was not the
only receptor affected by BDE-209. The authors suggested that the persisting CYP-3 Al 1 protein
could be related to activation of the constitutive androstane receptor (CAR).
4.4.2. Immunological Studies
No in vivo studies that specifically evaluated the immunosuppressive effects of decaBDE
have been identified. However, in chronic dietary studies, decaBDE has been found to induce
37
-------
histological changes in lymphoid organs, including the spleen (fibrosis) and mandibular lymph
nodes (hyperplasia) in male rats at a dose of 2,240 mg/kg-day (NTP, 1986). Although these
histopathological changes may play an important role in flagging decaBDE for immunotoxicity,
they are at the high end of the spectrum of doses evaluated, and the lack of studies to investigate
the functional effects on immune cells from exposure to decaBDE precludes complete
assessment of its immunotoxic potential.
Only one study investigated the immunotoxic effects of decaBDE on intact lymphocytes.
Pullen et al. (2003) isolated splenocytes from C57BL/6 mice and incubated the cells in culture
with decaBDE (purity not specified). DecaBDE was not cytotoxic to these cells at 3 jimol/L.
Treatment of cells for 48 hours with 3 |imol/L decaBDE did not cause attenuation of interleukin-
2-receptor a chain (CD25) expression (evaluated by immunohistochemistry and confocal
microscopy or laser scanning cytometry). The lack of attenuation of CD25 expression suggests
that decaBDE is not likely to alter a surface marker (which is essential for lymphocyte
proliferation during immune response) and may not affect the immune system in an
immunosuppressive manner. Pullen et al. (2003) also evaluated the potential of decaBDE to
alter cytokine production. Results showed that decaBDE did not have any effect on the
production of the cytokines TFN-y, IL-2, IL-6, and IL-10, indicating that decaBDE does not
interfere with the cells' ability to synthesize protein.
4.4.3. Genotoxicity
In vitro studies were conducted to evaluate the mutagenic potential of decaBDE (NTP,
1986). DecaBDE was tested in Salmonella typhimurium TA98, TA100, TA1535, or TA1537
strains up to 10,000 jig/plate in the presence or absence of exogenous metabolic system
(S9 fractions) prepared from the livers of Aroclor 1254-induced male Sprague-Dawley rats or
male Syrian hamsters. Results indicated that decaBDE was not mutagenic in the salmonella
assay systems.
The mutagenicity of decaBDE at doses of up to 10 |ig/mL was evaluated in
L5178Y/TK+ mouse lymphoma cell assay system in the absence or presence of S9 prepared
from the livers of Aroclor 1254-induced male F344/N rats. DecaBDE did not induce mutagenic
potential in this assay system. DecaBDE at doses up to 500 |ig/mL did not induce sister-
chromatid exchanges or chromosomal aberrations in Chinese hamster ovary cells in the absence
or presence of S9 prepared from the livers of Aroclor 1254-induced male Sprague-Dawley rats
(NTP, 1986).
Results from these studies provide evidence that parent decaBDE in the presence or
absence of an exogenous liver metabolic system does not react directly or indirectly with DNA
to cause gene mutations, DNA damage, or chromosomal effects.
38
-------
4.5. SYNTHESIS OF MAJOR NONCANCER EFFECTS
4.5.1. Oral
Several studies have been conducted to evaluate the effects of decaBDE and to elucidate
its mode of action. Long-term dietary studies conducted by NTP (1986) have identified
nonneoplastic liver and thyroid histopathology, such as liver thrombosis and degeneration in
male rats, and liver granulomas, centrilobular hypertrophy, and thyroid follicular cell hyperplasia
in male mice.
Neurobehavioral developmental effects on habituation were seen in adult male rats
(Viberg et al., 2007) and adult male mice (Viberg et al., 2003a) given a single dose of BDE-209
during the postnatal period. Male and female mice pups given BDE-209 during the neonatal
period showed early effects on palpebral reflex, forelimb grip, and struggling behavior.
Locomotor activity was also affected in young adult male mice (Rice et al., 2007).
Behavioral disturbances observed in the mature rats and mice exposed to decaBDE as
neonates (Rice et al., 2007; Viberg et al., 2007, 2003a) raise concern about possible
developmental neurotoxicity in children. In the study by Viberg et al. (2003a), neonatal male
mice were given a single dose of decaBDE (up to 20.1 mg/kg) in a fat emulsion on PNDs 3, 10,
or 19 and spontaneous motor behavior tests (measuring locomotion, rearing, and total activity)
were conducted in the mice at 2, 4, or 6 months of age. In addition, an habituation ratio was also
determined for each behavior variable at the tested ages. Exposure of 3-day-old mice to
20.1 mg/kg decaBDE resulted in significant dose-related changes in all three spontaneous
behavior variables at 2, 4, and 6 months of age. However, adult mice exposed neonatally up to
20.1 mg on either PND 10 or 19 did not show any significant differences in any of the variables
after 2, 4, or 6 months compared with controls. In the study in rats (Viberg et al., 2007),
significant dose-related disruption in habituation (changes in locomotion, rearing, and total
activity) was observed in 2-month-old rats, following exposure to BDE-209 at 6.7 and
20.1 mg/kg on PND 3.
Rice et al. (2007) used a similar approach in evaluating locomotor activity in groups of
9-13 male and female C57BL6/J mice exposed to 6 or 20 mg/kg-day BDE-209 from PNDs 2-15
and observed after placement in a novel environment on PND 70 during a 2-hour period. The
exposed males showed greater activity than the controls throughout the observation period.
Early in the period, the exposed male mice were hyperactive compared with controls; the
difference between the exposed animals and controls declined with time but locomotor activity
remained greater than that for the controls at the end of the 2-hour observation period. In the
case of the females, the activity of all groups was similar for the first 45 minutes of exposure but
the females became less active than the controls thereafter. The linear slope for the decline in
activity among the exposed males was significantly different from that of controls; the 20 mg/kg
group was affected to a greater extent than the 6 mg/kg group. The females on PND 70 and both
39
-------
sexes when examined at 1 year after birth did not display similar dose-related differences in
locomotor activity.
Rice et al. (2007) also examined a number of developmental and behavioral parameters
during the dosing period and for five days after dosing ceased. BDE-209 exposure did not
impact the developmental endpoints evaluated (pinnae detachment, incisor eruption, eye
opening, vaginal opening, testes descent). The behavioral parameters studied were selected as
measures of home-cage, motor, and sensorimotor behaviors. There was no apparent impact on
most measures of behavior; however, palpebral reflex, forelimb grip, and struggling during
handling appeared to be impacted by BDE-209 exposure. There was a significantly poorer
performance of the 20 mg/kg-day group in the palpebral reflex test on PND 14 and in the grip
test (males only) on PND 16. On PND 20, the struggling behavior of the group exposed to 6
mg/kg was significantly increased compared with the controls and 20 mg/kg-day group.
The most striking dose-related trend in the Rice et al. (2007) study was the locomotive
activity profile of males at PND 70. The direction of this trend differed from that observed by
Viberg et al. (2007, 2003a). In the Rice et al. (2007) study, activity in the exposed and control
mice decreased across the observation period, while in the Viberg et al. (2007, 2003a) studies
activity increased across the observation period for the exposed animals compared with the
controls. However, the exposure and testing conditions varied between the two studies and, thus,
the two sets of results are not directly comparable.
4.5.2. Inhalation
No data are available on the toxicity of decaBDE by the inhalation route of exposure.
4.5.3. Mode-of-Action Information
Impaired development of the cholinergic system during the postnatal "brain growth
spurt" period has been offered as a plausible hypothesis for the observed neurodevelopmental
impact of the PBDEs on adult responses to a novel environment (Viberg et al., 2007, 2003b).
The authors described a critical window of vulnerability in development of the cholinergic
system during postnatal development that in mice occurs in the first few weeks after birth with a
peak at PND 10 (Viberg et al., 2003b). They hypothesize that any agent that caused a deficit in
cholinergic receptors during this period could cause a hypoactive response to cholinergic
stimulants, including exposure to a novel environment in adulthood.
Viberg et al. (2007) tested this hypothesis in Sprague-Dawley rats. Two-month-old rats
that had been exposed to vehicle, 6.7 mg/kg, or 20 mg/kg BDE-209 on PND 3 were given a
subcutaneous injection of nicotine or saline solution. The vehicle control animals and animals in
the 6.7 mg/kg dose group receiving the nicotine demonstrated the typical hyperactive response of
mature rats when compared with the saline-injected rats during the 60-80 minute observation
40
-------
period. Those given the 20.1 mg/kg dose were markedly hypoactive compared with the saline
controls. These observations provide some support for the hypothesis that impaired prenatal
development of the cholinergic system may play a role in the habituation deficits observed in
adult rats and mice that were prenatally exposed to decaBDE during the critical window for
system development.
Other studies by the Eriksson/Viberg research team using PBDE compounds with four,
five, and six bromines have shown effects on locomotion and habituation that are similar to
those observed for the deca compound. However, in the study of BDE-99, when the timing of
exposure was varied, the window of greatest vulnerability occurred on day 10 rather than on
days 3 or 19 (Eriksson et al. 2002). Thus, the results for BDE-99 are different from those for
decaBDE reported in Viberg et al. (2003a). However, the data from the radiolabel component of
the study provide a rationale for the difference in the apparent window of vulnerability. Two
contributing factors are observed. First, absorption from the gastrointestinal tract decreases as
the neonatal mice age during their first few weeks of life. Thus, less of the decaBDE reaches the
liver for distribution on days 10 and 19 than on day 3. Secondly, distribution to the brain is slow.
The amount of radiolabel in the brain is higher (0.7%) when measured 7 days after dosing on
PND 3 than when measured 24 hours after dosing (0.5%). The radioactivity measured 7 days
after dosing was also higher than when measured 24 hours after dosing on PND 10 (0.4% at
24 hours; 1.1% at 7 days). Less than 0.1% of the label reaches the brain when the compound is
administered on day 19. While evidence exists that demonstrates BDE-209, as well as other
PBDEs, interact at the neurological level, data are inadequate to determine the mode of action
for BDE-209.
DecaBDE caused thyroid gland follicular cell hyperplasia and thyroid tumors in male
mice, effects that are indicative of thyroid toxicity (NTP, 1986). Based on these effects,
decaBDE may share the general property of organohalogen compounds in which in vivo
exposure in rodents results in reduction of serum total and free thyroid hormone (T4) levels
(Legler and Brouwer, 2003). Thyroid hormone disruption has been hypothesized to arise from at
least two different mechanisms, one of which is the induction of liver enzymes of different
enzyme families, including CYP-1A1 and CYP-2B (McDonald, 2002). Induction of liver
UDPGTs may increase the rate of T4 conjugation and excretion, which is believed to be a
mechanism for TCDD-induced thyroid effects. An increase in the metabolism of T4 will result
in enhanced excretion and thus a drop in its circulating levels. The other mechanism involves
alteration of thyroid homeostasis through hormone mimicry. Potential thyroid toxicants may be
metabolized by liver enzymes to metabolites that compete with the thyroid transport protein,
transthyretin, a key protein involved in the transport of T4 through the blood and into the
developing tissues in rodents.
41
-------
Zhou et al. (2001) evaluated the interference of decaBDE with thyroid hormone
homeostasis in weanling rats exposed to decaBDE (>98% purity) up to 100 mg/kg-day for
4 consecutive days. Results indicated that decaBDE did not affect hepatic enzyme activities or
serum T4, T3, TSH, and hepatic UDPGT activity at doses up to 100 mg/kg-day. Rice et al.
(2007) also evaluated T4 levels in the blood of C57BL6/J mice (1 male, 1 female/litter) on
PND 21 following exposure to 0.6 or 20 mg/kg-day BDE during PNDs 2-15. In this instance,
there was a dose-related reduction in the T4 levels for males but not females. Despite the
possibility of thyroid hormone involvement in the neurodevelopmental impact of BDE-209 on
the habituation response in male mice exposed to a single dose on PND 3, there are no mode-of-
action data that link thyroid hormones to the neurobehavioral observations reported by Viberg et
al. (2003a).
In the chronic dietary studies conducted by NTP (1986), decaBDE has been found to
induce histologic changes in lymphoid organs, including the spleen (fibrosis) and mandibular
lymph nodes (hyperplasia), in rats. These histopathologic changes could flag decaBDE as a
potential immunotoxicant. No in vivo functional immunotoxicity studies were identified.
Results from an in vitro study investigating the immunotoxic effects of decaBDE indicated that
decaBDE did not show any attenuation of CD25 expression or changes in cytokine production
(Pullen et al., 2003). As such, decaBDE is unlikely to affect the immune system in an
immunosuppressive manner.
Several in vivo and in vitro studies have evaluated decaBDE for its potential to activate
the AhR signal transduction pathway and to induce gene expression, resulting in increased phase
I and phase II metabolism. Taken together, the in vivo and in vitro studies suggest that decaBDE
has very low or no potential to bind the AhR. The inability of decaBDE to elicit CYP-450
induction or AhR binding and gene expression, indicating lack of potential to activate the AhR
signal transduction pathway, suggests that decaBDE may cause liver toxicity through a different
mechanism and that the observed liver effects are not an adaptive effect related to enzyme
induction. Results from the Morck et al. (2003) study indicated high concentration of tissue-
bound radioactivity in the liver, and the radioactivity was assumed to be bound to
macromolecules. The authors asserted that binding occurred via reactive metabolites.
Formation of such metabolites may be responsible for the liver toxicity observed on exposure to
decaBDE. However, the enzymes responsible for decaBDE metabolism and reactive
intermediates have not been identified. The existing data show tissue binding and are therefore
not adequate to elucidate a potential role in the mode of action.
A number of studies have also indicated that brominated flame retardants may interfere
with estrogen pathways by binding to and activating ERs (Legler and Brouwer, 2003). No in
vivo studies were identified that evaluated the specific estrogenic effects of decaBDE.
Furthermore, specific effects attributable to estrogenic dysregulation were not identified in the
42
-------
available animal studies, although some developmental toxicity endpoints were observed.
Mechanistic studies do not support effects of decaBDE on ER pathways.
4.6. EVALUATION OF CARCINOGENICITY
4.6.1. Summary of Overall Weight of Evidence
Under the Guidelines for Carcinogen Risk Assessment (U.S. EPA, 2005a), the descriptor
"suggestive evidence of carcinogenic potential" is appropriate for decaBDE. This descriptor of
the database is appropriate when the weight of evidence is suggestive of carcinogenicity and a
concern for potential carcinogenic effects in humans is raised, but the data are judged not
sufficient for a stronger conclusion. This descriptor covers a spectrum of evidence associated
with varying levels of concern for carcinogenicity, ranging from a positive cancer result in the
only study on an agent to a single positive cancer result in an extensive database that includes
negative studies in other species.
The weight of evidence of human carcinogenicity of decaBDE is based on (1) no studies
of cancer in humans exposed to decaBDE; (2) a statistically significant increase in incidence of
neoplastic nodules in the liver of low- and high-dose male rats and high-dose female rats; (3) a
significantly increased incidence of hepatocellular adenoma or carcinoma (combined) in male
mice at the low dose and marginally increased incidence at high dose; (4) a nonsignificantly
increased incidence of hepatocellular adenoma or carcinoma (combined) in female mice; (5) a
slightly greater (but statistically not significant) incidence of thyroid gland adenomas or
carcinomas (combined) in dosed male and female mice; (6) a significantly increased incidence
in male mice, at both doses, of follicular cell hyperplasia, considered by many as a precursor to
thyroid tumors; and (7) an apparent absence of genotoxic potential.
Based on the results of the chronic and genotoxicity studies, NTP (1986) concluded that
there was "some evidence of carcinogenicity" for male and female rats as shown by increased
incidences of neoplastic nodules of the liver in low-dose males and high-dose groups of each
sex. There was "equivocal evidence of carcinogenicity" for male mice as shown by increased
incidences of hepatocellular adenomas or carcinomas (combined) in only the low-dose group
and slight increase of thyroid gland follicular cell adenomas or carcinomas (combined) in both
dosed groups. There was "no evidence of carcinogenicity" for female mice. Several
nonneoplastic lesions were observed at increased incidences, the most notable being thyroid
gland follicular cell hyperplasia in male mice. The weight of experimental evidence is on the
strong end of the spectrum for the descriptor "suggestive evidence of carcinogenic potential,"
since there is suggestive evidence that decaBDE is carcinogenic for more than one species, sex,
and site. Based on the weight of evidence, a dose-response assessment of the carcinogenicity of
decaBDE is deemed appropriate
43
-------
4.6.2. Synthesis of Human, Animal, and Other Supporting Evidence
No information is available on the carcinogenicity of decaBDE in humans. Chronic
dietary studies of decaBDE have been conducted in rats and mice (NTP, 1986), and
carcinogenicity data are presented in Tables 4-4 and 4-5. DecaBDE caused substantial dose-
related increases in neoplastic nodules in rats with statistical significance at the high dose in both
sexes (Table 4-4). There were no statistically significant changes in hepatocellular carcinoma in
either sex. Although there was a positive trend in mononuclear cell leukemia in male rats, the
increase was marginal and not considered to be biologically significant due to the unusually high
incidence in controls.
In mice, insignificant increases in hepatocellular adenoma occurred in males at low
(3,200 mg/kg-day) and high (6,650 mg/kg-day) doses compared with controls. The incidence of
hepatocellular carcinoma was not significantly increased in male mice at either dose. In these
male mice, however, significantly increased hepatocellular adenomas or carcinomas (combined)
were observed in the low-dose group but only marginally increased in the high-dose group. An
increased incidence of hepatocellular adenoma and carcinoma was also observed in the female
mice; however, these changes were not statistically significant in either treated dose group.
There was also a slight (but not statistically significant) increase in the incidence of thyroid
tumors in mice of both sexes, but this was not observed in rats. The biological relevance of this
finding in mice is supported by the accompanying increased incidence of follicular cell
hyperplasia in male mice; the increase in hyperplasia in females was not statistically significant.
The increased incidence of neoplastic nodules of the liver observed in male and female
rats constitutes some evidence of carcinogenicity in treated rats, and this observation is
supported by the significantly increased incidence of hepatocellular adenoma and carcinoma
(combined) in low-dose male mice and nonsignificantly increased incidence in female mice.
Although the study results from each individual species/sex combination were judged to be
either "some" or "equivocal" evidence of carcinogenicity, a common target organ (liver) in two
species and both sexes indicates a potential for decaBDE to cause cancer in treated animals. In
addition to the liver cancer, slight increases in thyroid follicular cell tumors in mice were
accompanied by hyperplasia. Based on the increasing agreement that hyperplasia is a stage of
the thyroid follicular cell carcinogenic process (NAS, 2000; Hard, 1998; U.S. EPA, 1998b; Hill
et al., 1989), decaBDE would be considered as eliciting a carcinogenic effect in a second target
organ (i.e., the thyroid) in mice.
4.6.3. Mode-of-Action Information
Genotoxicity studies have found that decaBDE does not appear to be mutagenic since it
did not induce gene mutations in S. typhimurium strains or in mouse L5178Y lymphoma cells.
Also, decaBDE did not induce sister-chromatid exchanges or chromosomal aberrations in
44
-------
Chinese hamster ovary cells in the presence or absence of metabolic activation systems (NTP,
1986).
There are no other studies of the carcinogenic mode of action of decaBDE. The mode of
action is unknown.
45
-------
Table 4-4. Incidence of liver neoplastic nodules and carcinomas in rats fed decaBDE for 2 years
Dose groups ppm
mg/kg-day
Number examined
Males
0
0
50
25,000
1,120
50
50,000
2,240
50
Females
0
0
50
25,000
1,200
50
50,000
2,550
50
Liver neoplastic nodules
Overall rates
Adjusted rates
Terminal rates
Week of first observation
Historical incidence
1/50 (2%)
2.9%
1/35 (3%)
104
7/50 (14%)a
27.1%
6/24 (25%)
89
15/49(31%)a
52.7%
13/26 (50%)
87
3.5%
1/50 (2%)
2.5%
1/40 (3%)
104
3/49 (6%)
9.1%
3/33 (9%)
104
9/50 (18%)a
24.4%
7/34 (21%)
87
2.6%
Hepatocellular carcinoma
Overall rates
1/50 (2%)
1/50 (2%)
1/49 (2%)
0/50 (0%)
2/49(4.1%)
0/50 (0%)
Liver neoplastic nodule or hepatocellular carcinoma
Overall rates
Adjusted rates
Terminal rates
Week of first observation
2/50 (4%)
5.2%
1/35 (3%)
97
8/50 (16%)a
31.1%
7/24 (29%)
89
15/49 (3 l%)a
52.7%
13/26 (50%)
87
1/50 (2%)
2.5%
1/40 (3%)
104
5/49 (10%)
15.2%
5/33 (15%)
104
9/50 (18%)a
24.4%
7/34 (21%)
87
Statistically significantly different from controls (p < 0.05), based on life table tests and incidental tumor tests.
Source: NTP (1986).
46
-------
Table 4-5. Incidence of nonneoplastic or neoplastic lesions in the liver and thyroid gland of mice fed decaBDE
for 2 years
Dose groups ppm
mg/kg-day
Number examined
Males
0
0
50
25,000
3,200
50
50,000
6,650
50
Females
0
0
50
25,000
3,760
50
50,000
7,780
50
Hepatocellular adenoma
Overall rates
Adjusted rates
Terminal rates
Week of first observation
4/50 (8%)
19.0%
3/19 (16%)
81
12/50 (24%)
46.2%
11/25(44%)
100
12/50 (24%)
39.0%
7/24 (29%)
60
5/50 (10%)
16.8%
4/27 (15%)
83
10/50 (20%)
31.2%
9/31 (29%)
102
7/50 (14%)
21.9%
7/32 (22%)
103
Hepatocellular carcinoma
Overall rates
Adjusted rates
Terminal rates
Week of first observation
5/50 (10%)
20.7%
1/19 (5%)
81
14/50 (28%)
42.9%
8/25 (32%)
72
8/50 (16%)
26.8%
4/24 (17%)
76
3/50 (6%)
10.7%
2/27 (7%)
101
4/50 (8%)
12.1%
3/31 (10%)
93
7/50 (14%)
20.8%
6/32 (19%)
96
Hepatocellular adenoma or carcinoma
Overall rates
Adjusted rates
Terminal rates
Week of first observation
8/50 (16%)
33.9%
4/19 (21%)
81
22/50 (44%)a
67.7%
15/25 (60%)
72
18/50 (36%)
56.5%
11/24(46%)
60
8/50 (16%)
26.7%
6/27 (22%)
83
13/50 (26%)
39.1%
11/31 (35%)
93
13/50 (26%)
39.1%
12/32 (38%)
96
Thyroid gland
Hyperplasia, follicular cell
2/50 (4%)
10/50 (20%)a
19/50 (38%)a
4/50 (8%)
9/50 (18%)
7/49 (140/0)
Follicular cell adenoma
Overall rates
Adjusted rates
Terminal rates
Week of first observation
0/50 (0%)
0%
0/19 (0%)
3/50 (6%)
10.8%
2/25 (8%)
90
3/50 (6%)
12.5%
3/24 (13%)
103
1/50 (2%)
2.9%
0/27 (0%)
95
3/50 (6%)
7.8%
1/31(3%)
80
2/49 (4%)
6.3%
2/32 (6%)
103
Follicular cell adenoma or carcinoma (combined)
Overall rates
Adjusted rates
Terminal rates
Week of first observation
0/50
0%
0
4/50 (8%)
14.7%
3/25 (12%)
90
3/50 (6%)
12.5%
3/24 (13%)
103
1/50 (2%)
2.9%
0/27 (0%)
95
3/50 (6%)
7.8%
1/31(3%)
80
3/49 (6%)
9.4%
3/32 (9%)
103
Statistically significantly different from controls (p < 0.05), based on life table tests and incidental tumor tests.
Source: NTP (1986).
47
-------
4.7. SUSCEPTIBLE POPULATIONS AND LIFE STAGES
4.7.1. Possible Childhood Susceptibility
As discussed in section 4.3.1, a gavage study (Hardy et al., 2002) in which rats were
exposed in the diet to decaBDE (97.34% purity) during gestation days 0-19 reported no effects
on number of dams with viable fetuses, mean number of corpora lutea, number of implantation
sites, percent preimplantation loss per dam, number of viable fetuses, gravid uterine weights,
fetal body weights, crown-rump ratio, and fetal sex ratio. No effects were reported during
external, skeletal, or visceral examination of rat fetuses. The NOAEL in this study was
1,000 mg/kg-day, the highest dose tested. In a study in male mice (Tseng et al., 2006) exposed
to BDE-209 from PNDs 21-70 (see section 4.3.2), effects were seen on sperm function, DNA
content, and histopathology of the testes at 500 mg/kg-day but not at 100 mg/kg-day.
In a study in rats (Viberg et al., 2007), exposure of the animals to the lowest dose tested
of 6.7 mg/kg-day BDE-209 on PND 3 resulted in significant changes in spontaneous motor
behavior (locomotion, rearing, and total activity) in 2-month-old rats. In the study of Rice et al.
(2007), effects on the palpebral reflex, struggling behavior and locomotor activity were seen in
mice given the lowest dose of 6 mg/kg-day BDE-209 on PNDs 2-15.
Neurobehavioral alterations were observed in mature mice when male mice were
exposed to 20.1 mg/kg decaBDE as neonates on PND 3 but not on PNDs 10 and 19 (Viberg et
al., 2003a). A significant increase in the amount of radioactivity in the brain on day 7 after
exposure was noted when the neonates were exposed to 14C-decaBDE on PNDs 3 and 10 but not
on PND 19 (Viberg et al., 2003a). The increase in the radioactivity in the brain, coupled with
the behavioral disturbances on exposure to decaBDE on PND 3, suggests that differences may
exist in the absorption and distribution of decaBDE among the 3-, 10-, and 19-day-old neonates.
In addition, differences exist in the timing of the brain growth spurt in rodents (first few weeks
after birth) and humans (starts in the sixth month of gestation and continues through the first 2
years of life after birth). The significance of this difference with regard to the potential for
effects on children from exposure to BDE-209 is not clear. Moreover, whether other targets of
decaBDE (thyroid and liver) are more sensitive in children is unknown since the enzymes
responsible for decaBDE metabolism have not been identified and the mechanisms for age-
dependent kinetic differences are not completely clear. Although no studies were identified that
investigated placental transfer, decaBDE has been found in human breast milk, suggesting that
newborns and infants have a potential source of exposure via this route.
4.7.2. Possible Gender Differences
All absorption studies following oral administration of decaBDE were conducted in male
rats, precluding a comparison of kinetics in males versus females. There were no clear gender-
dependent differences in decaBDE toxicity in short-term and subchronic studies conducted in
48
-------
both sexes of rats and mice (NTP, 1986). However, long-term exposure studies to decaBDE
indicated that male rats may be more sensitive than females. Male rats exhibited nonneoplastic
lesions, including liver thrombosis and degeneration, splenic fibrosis, and lymphoid hyperplasia,
and male mice showed increased incidences of liver granulomas, hepatocellular hypertrophy,
and thyroid follicular cell hyperplasia, while female rats and mice appeared to be refractory to
these systemic effects of decaBDE at the tested doses (NTP, 1986). One exception was that
female mice had a higher incidence of stomach ulcers than their male counterparts. Although a
significantly increased incidence of liver neoplastic nodules was observed in both sexes of rats
(NTP, 1986), hepatocellular carcinomas or adenomas were not significantly increased in male
mice only. No gender-specific effects on reproductive organs were identified.
The study of Rice et al. (2007) investigated several endpoints in male and female mice.
In PND 21 offspring of mice treated with decaBDE on PNDs 2-15, a dose-related reduction in
serum T4 was observed in males but not females. Development of forelimb grip strength was
delayed at PND 16 in exposed males in the high-dose group but not in females. Struggling
behavior was more apparent in males than in females, and effects on locomotor activity were
observed on PND 70 in male mice but not in female mice.
Overall, it appears that males may be more susceptible to some effects of decaBDE
compared to females, although this is not true for all endpoints. Underlying mechanisms for the
apparent differences in susceptibility have not been identified.
49
-------
5. DOSE-RESPONSE ASSESSMENTS
5.1. ORAL REFERENCE DOSE (RfD)
5.1.1. Choice of Principal Study and Critical Effect—with Rationale and Justification
Epidemiological studies or case reports are not available for decaBDE.
Available animal studies of repeated oral exposure to decaBDE include a 4-day study in
rats (Zhou et al., 2001), 14-day studies in rats and mice (NTP, 1986), 13-week studies in rats and
mice (NTP, 1986), and 2-year studies in rats and mice (NTP, 1986). In addition, decaBDE was
also tested for its toxicity in reproductive and developmental studies (Rice et al., 2007; Viberg et
al., 2007, 2003a; Tseng et al., 2006; Hardy et al., 2002). The results from these studies are
summarized in Tables 4-1, 4-2, and 4-3. Among the available studies, several potential principal
studies for deriving an RfD were identified: the 2-year chronic studies in rats and mice (NTP,
1986) and neurodevelopmental studies in rats and mice (Rice et al., 2007; Viberg et al., 2007,
2003a).
NTP (1986) reported a study of the chronic oral toxicity and carcinogenicity of decaBDE
in rats. In the liver, an increase in the incidence of thrombosis was observed in high-dose male
rats (2,240 mg/kg-day). A dose-dependent but statistically not significant increase in
degeneration of the liver was also observed in treated male rats at incidences of 13/50, 19/50,
and 22/49 in the control, low-dose, and high-dose groups, respectively. In the spleen, increased
incidence of fibrosis was seen in males at the low dose (8/50) and high dose (13/49) compared to
5/49 in controls; the effect was statistically significant only in the high-dose group. In the
mandibular lymph node, lymphoid hyperplasia increased in males in a dose-dependent manner,
but the incidence reached statistical significance only at the high dose. Based on these results,
the NOAEL for systemic toxicity was 1,120 mg/kg-day, and the LOAEL was 2,240 mg/kg-day in
male rats based on increased incidence of thrombosis and degeneration in the liver, splenic
fibrosis, and lymphoid hyperplasia of the mandibular lymph nodes. Female rats appeared to be
refractory to the systemic toxicity of decaBDE at the doses used in this study. Therefore, the
NOAEL for systemic toxicity in females was 2,550 mg/kg-day, with no LOAEL established.
The observed systemic toxicity of decaBDE in the 2-year study in rats is supported by
observed liver effects at higher doses in a 2-year study in mice (NTP, 1986). Significant
increases in the incidence of centrilobular hypertrophy were also observed in the liver of male
mice at low and high doses (34/50, and 32/50, respectively) in comparison to controls (0/50). In
the thyroid gland, a dose-dependent and statistically significant increase (at both dose levels) in
the incidence of follicular cell hyperplasia was observed in male mice (control, 2/50; low dose,
10/50; high dose, 19/50). In the females, this incidence increased in the low- and high-dose
groups compared with the control group, but the increase was not dose dependent or statistically
50
-------
significant at any dose level. High-dose female mice exhibited a significant increase in the
incidence of stomach ulcers. Based on these results, no NOAEL for males was established. The
LOAEL, based on increased incidence of centrilobular hypertrophy and a statistically significant
and dose-dependent increase in thyroid gland follicular cell hyperplasia in male mice, was
3,200 mg/kg-day or greater. Similar to the study in rats (NTP, 1986), female mice appeared to
be refractory to the systemic toxicity of decaBDE at the doses used in this study. The study
established a NOAEL of 3,760 mg/kg-day for portal-of-entry effects for females and a LOAEL of
7,780 mg/kg-day based on a significant increase in the incidence of stomach ulcers.
Viberg et al. (2003a) reported functional neurobehavioral effects of single-dose
exposures to decaBDE. The neurotoxic effects of decaBDE on spontaneous motor behavior
were investigated in adult NMRI male mice exposed to a single oral dose of decaBDE as
neonates on PND 3, 10, or 19 (i.e., at different stages of neonatal mouse brain development).
Pair-wise testing between adult mice exposed on PND 3 and control groups indicated significant
dose-related changes in the habituation ratio calculated from three behavior variables
(locomotion, rearing, and total activity) in mice exposed to 20.1 mg/kg and evaluated at 2, 4, and
6 months of age; no statistically significant effect was seen at 2.22 mg/kg. Adult mice exposed
neonatally up to 20.1 mg/kg on either PND 10 or 19 did not show any significant differences in
any of the variables after 2, 4, or 6 months compared with controls. The authors indicated that
the absence of changes in spontaneous activity in mice treated on PNDs 10 and 19 appears to
suggest that there is a critical window for the induction of behavioral disturbances. The decrease
observed in the habituation capability in the adult mice exposed neonatally to the high dose
indicated that the neurotoxic effect of neonatal decaBDE exposure was persistent and also
worsened with age. Based on these observations, the NOAEL in this study was 2.22 mg/kg and
the LOAEL was 20.1 mg/kg. In the study in rats (Viberg et al., 2007), significant dose-related
disruption in habituation (changes in locomotion, rearing, and total activity) was observed in 2-
month-old rats, following exposure to BDE-209 at 6.7 and 20.1 mg/kg on PND 3. A NOAEL
was not identified in this study.
Rice et al. (2007) examined the effects of BDE-209 (99.5% purity) on developmental
milestones, sensorimotor behaviors, serum T4 levels and locomotor activity in male and female
C57BL6/J mice administered doses of 0, 6, or 20 mg/kg-day from PNDs 2-15. An FOB suitable
for neonatal mice was developed to examine a series of homecage, reflexive, and sensorimotor
behaviors, measured on PND 14, 16, 18, or 20. Three of the FOB endpoints were affected by
BDE-209 exposure: palpebral reflex, forelimb grip, and struggling behavior during handling. On
PND 14, significantly fewer male and female pups in the 6 and 20 mg/kg-day groups performed
the palpebral reflex compared with controls. On PNDs 14 and 16, fewer male pups in the
20 mg/kg-day group performed an effective forelimb grip compared with same-sex controls.
Male and female pups in the 6 mg/kg-day group struggled significantly more on PND 20, while
51
-------
they were being handled for the FOB, compared with controls and the high-dose group.
Locomotor activity in PND 70 males and females in the control and treated groups declined over
the course of the 2-hour assessment. However, the rate of decline was significantly different
(p = 0.04) in male mice exposed to 6 and 20 mg/kg-day compared with control animals. The
effect was most pronounced in the high-dose males during the first 1.5 hours of the 2-hour
activity session. A dose-related reduction in serum T4 in comparison with controls occurred in
PND 21 males but not in females. The LOAEL in this study was 6 mg/kg-day, the lowest dose
tested, for decrease in the percent of male and female pups performing the palpebral reflex on
PND 14, for increased struggling behavior of male mice on PND 20, for decreased T4 levels in
male mice, and for effects on locomotor activity of male mice on PND 70.
Viberg et al. (2007) administered BDE-209 by gavage to 3-day-old Sprague-Dawley rats
at 0, 6.7, or 20.1 mg/kg. A total of 20 rats were picked from three to five different litters in each
treatment group. Motor activity, measured as locomotion (horizontal movement), rearing
(vertical movement), and total activity (all types of vibration within the test cage [i.e., those
caused by rat movements, shaking/tremors, and grooming]) was evaluated for a 60-minute
period, divided into three 20-minute periods, in rats at the age of 2 months. In control rats, there
was a distinct decrease in locomotion, rearing, and total activity, indicating habituation in
response to the diminishing novelty of the test chamber over the 60-minute test period, at
2 months of age. Two-month-old rats exposed to 20.1 mg/kg BDE-209 on PND 3 displayed
significantly less activity for all three behavioral variables during the first 20-minute test period
compared with controls, while during the third 20-minute period (40-60 minutes) they were
significantly more active than the control animals. Rats receiving the low dose of BDE-209
(6.7 mg/kg) showed significantly increased locomotion activity during the second 20-minute
period, significantly decreased rearing activity during the first and second 20-minute periods,
and significantly higher total activity during the first and second 20-minute periods compared
with the control rats. The LOAEL in this study was 6.7 mg/kg for significant changes in
spontaneous motor behavior (locomotion, rearing, and total activity) in 2-month-old rats given
BDE-209 on PND 3.
Of the three neurobehavioral studies, only Viberg et al. (2003a) identified a NOAEL.
The NOAEL identified in Viberg et al. (2003a) was lower than the LOAELs identified in the
neurobehavioral studies conducted by Viberg et al. (2007) and Rice et al. (2007). The Viberg et
al. (2003a) study was therefore selected as the principal study for deriving the RfD. However,
several considerations were weighed in making this decision. Concerns regarding the study
design raise potential issues about full reliance on this study. The dosing regimen did not
include gestation and lactation exposure (U.S. EPA, 1998a); only single doses were given. The
study was conducted in male mice only. The protocol was unique and did not conform to health
effects test guidelines for neurotoxicity screening battery or developmental neurotoxicity studies
52
-------
(U.S. EPA, 1998a, c). While the study design appears to identify a developmental window of
susceptibility, it is not adequate to determine the effect of longer dosing. However, the fact that
the effects were observed after a single dose is a situation that increases concern regarding the
impact of BDE-209 on neurological development. Translating the implications of these data to
more traditional dosing regimens is problematic, particularly with regard to evaluating the
implications of in utero and postnatal exposure.
Another concern is that, based on the data provided in the published report, more than
one pup per litter were used for the behavioral testing (10 mice were randomly selected from
three to five different litters in each treatment group). Increasing the number of samples from
each litter may bias the analyses towards false positives. The observed neurobehavioral effects
may be attributable to differences in pups born to a single dam rather than related to treatment.
Another concern regarding the study design was the limited number of neurobehavioral
parameters that were assessed; the authors measured only indices related to motor activity
(locomotion, rearing, and total activity). The absence of a full FOB that evaluates neurological
and behavioral signs limits the ability to correlate the reported effects with other FOB
parameters. Data from the FOB utilized in the Rice et al. (2007) study, also in mice, mitigate
some concern related to its absence in the Viberg et al. (2003a) study.
As indicated in the Guidelines for Neurotoxicity Risk Assessment (U.S. EPA, 1998a), it is
assumed that an agent that produces detectable adverse neurotoxic effects in experimental
animal studies will pose a potential hazard to humans. For BDE-209, in the absence of human
evidence, data from experimental animal studies are used as the basis for the RfD.
While study design limitations cloud the utility of this study, several additional
considerations support the use of these data. Acute exposure to a highly lipophilic and long half-
life chemical, such as BDE-209, will result in exposure that lasts much longer than just acutely.
In addition, there are a wide variety of brain structures that have very limited critical windows
during development. These short critical windows translate to susceptible periods of exposure
that can be very short.
The concept that exposure during critical periods of development can induce functional
neurological effects later in development has been demonstrated with structurally related PBDE
congeners, including tetra-, penta-, and hexaBDEs (Kuriyama et al., 2005; Viberg et al., 2005,
2004a, b, 2003b, 2002; Ankarberg, 2003; Branch! et al., 2002; Eriksson et al., 2002, 2001). In
addition, the study of Rice et al. (2007) strengthens the evidence that links the behavioral effects
seen in mice to neonatal BDE-209 exposure. Therefore, the observed neurobehavioral effects in
the Viberg et al. (2003a) study in mice and the Viberg et al. (2007) study in rats are biologically
plausible, and exposure to BDE-209 may pose a potential hazard to humans (U.S. EPA, 1998a).
Viberg et al. (2003b) have suggested that the animals are vulnerable only during the
postnatal brain growth spurt that occurs in the first few weeks after birth in mice. In the case of
53
-------
decaBDE, the identification of PND 3 as the most vulnerable window for exposure can be
explained by the toxicokinetic data showing apparent absorption decreases during the first few
weeks of life and slow distribution to the brain when the dosing occurs on PND 3 or 10.
Although some radiolabel reaches the brain in the 24 hours after dosing, the amount increases
over the 7-day period after dosing. After dosing on PND 19, less than 0.1% of the dose was
found in the brain initially and after 7 days. These observations would explain the absence of
effects in animals exposed later in the postnatal period as adults.
Taken together, these considerations support the use of the Viberg et al. (2003 a) study as
a potential critical study for deriving the RfD for BDE-209.
5.1.2. Methods of Analysis
The published information presented by Viberg et al. (2007, 2003a) could not be used for
benchmark dose (BMD) modeling; mean values for motor activity (locomotion, rearing, and
total activity), habituation capability for 2-, 4-, and 6-month-old mice or rats, and standard
deviations are only displayed graphically, and such values cannot be read with any accuracy
from the graphs. Therefore, the NOAEL of 2.22 mg/kg is used as a point of departure for
estimating the oral RfD for BDE-209.
DecaBDE has very low water solubility and a relatively high Kow; therefore, preferential
distribution to adipose tissue might be expected. This property raises concerns for accumulation
of decaBDE in the body as a result of repeated exposure. However, limited data on the tissue
distribution of decaBDE in experimental animals indicate that it is not readily distributed to
adipose tissue (Morck et al., 2003). The limited deposition in the adipose tissue suggests that
chronic accumulation may not be significant for BDE-209. The optimal way to address this
issue would be to use a PBPK model to estimate the steady-state body burden in the lipid
compartment after repeated exposure and examine the dose-response relationship based on the
internal dose at the target tissues. No PBPK models of decaBDE were identified for humans or
animals, and data on internal doses from existing toxicity studies are not adequate to support
dose-response analysis. In the absence of these data, a dose-response analysis based on body
burden was not included in this assessment.
5.1.2.1. Benchmark Dose Modeling
Although the NOAEL from the Viberg et al. (2003a) study was selected as the critical
endpoint for the RfD derivation, the dose-response data from the NTP (1986) study were
modeled to determine the lower bound BMD (BMDL) for systemic toxicity. Details of the BMD
modeling results are presented in Appendix B and summarized in this section. The following
data sets from the NTP (1986) 2-year rat and mouse studies were selected for BMD modeling:
thrombosis in the liver, liver degeneration, fibrosis in the spleen, and lymphoid hyperplasia in
54
-------
male rats; centrilobular hypertrophy in the livers; and follicular cell hyperplasia in the thyroid of
male mice.
The BMD modeling was conducted using EPA's BMD software (BMDS) version 1.3.2.
The dose levels showing a change compared with control values exhibited responses at levels
near 10%; therefore, all of the BMD analyses were conducted with the benchmark response
(BMR) set to a 10% extra risk (U.S. EPA, 2000b). For each data set, all the dichotomous
models, including gamma, logistic, log-logistic, multistage, probit, log-probit, quantal-linear,
quantal-quadratic, and Weibull, available in the BMDS were used. BMD model fit was
evaluated based on the goodness-of-fit/> value (indicating global model fit), Akaike Information
Criterion (AIC) (indicating model fit when controlling for number of model parameters), as well
as local chi-square residual (indicating the model fit at the data point close to the preset BMR).
Based on the model output, the models providing best model fit to the data were selected to
estimate BMD10 and BMDL10. The BMD analyses results are summarized in Table 5-1.
Based on the comparison of BMD modeling results for all the potential critical effects
observed in chronic rat and mouse studies, the lowest BMDL10 is 406 mg/kg-day for liver
degeneration effect in male rats. This value is much higher than the NOAEL of 2.22 mg/kg for
the neurobehavioral changes observed in Viberg et al. (2003a). Because the graphic habituation
data from the Viberg et al. (2003a) publication could not be used for BMD modeling, the
NOAEL of 2.22 mg/kg is used as a point of departure for calculating the oral RfD.
Table 5-1. BMDs for potential critical effect from chronic rat and mouse
studies
Endpoint
Thrombosis in the liver of male rats
Liver degeneration in male rats
Splenic fibrosis in male rats
Lymphoid hyperplasia in male rats
Centrilobular hypertrophy in the liver
of male mice
Follicular cell hyperplasia in the
thyroid of male mice
Reference
NTP (1986)
NTP (1986)
NTP (1986)
NTP (1986)
NTP (1986)
NTP (1986)
NOAEL
(mg/kg-day)
1,120
2,240
1,120
1,120
N/A
N/A
LOAEL
(mg/kg-day)
2,240
N/Aa
2,240
2,240
3,200
3,200
BMD10
(mg/kg-day)
2,125
765
1,446
1,538
No model fit
1,670
BMDL10
(mg/kg-day)
1,738
406
1,001
1,165
-
1,190
3N/A = not available or cannot be estimated.
Source: NTP (1986).
5.1.3. RfD Derivation
Based on the neurobehavioral effects observed in the Viberg et al. (2003a) study, the
NOAEL of 2.22 mg/kg is used as the point of departure for calculating the RfD. The following
uncertainty factors (UFs) (U.S. EPA, 2002) are applied to the NOAEL of 2.22 mg/kg: 10 for
55
-------
extrapolating animal data to humans (UFA interspecies variability), 10 for susceptible human
subpopulation (UFH interhuman variability), and 3 for extrapolating from the single-dose to a
lifetime exposure (UFS). The total composite UF=10xlOx3 = 300.
A 10-fold UFA was used to account for laboratory animal to human interspecies
differences. Although the toxicokinetics of decaBDE in animals have been evaluated, no
adequate description of toxicokinetics of decaBDE in humans exists. The critical effect for
deriving the RfD, altered behavior due to exposure during development, is expected to be
relevant to humans. No quantitative data were identified to compare relative human and rodent
sensitivity to these changes. However, given the longer period of brain development in humans
as compared to rodents and the higher importance of cognitive function, it is appropriate to
consider that humans may be more sensitive than rodents in the absence of specific data. Based
on these considerations, the default UFA value of 10 was applied.
A default intraspecies UFH of 10 was applied to account for variations in susceptibility
within the human population (intrahuman variability). This factor accounts for the segment of
the human population that may be more sensitive than the general population to exposure to
BDE-209. A default value is warranted because insufficient information is currently available to
assess human-to-human variability in BDE-209 toxicokinetics or toxicodynamics.
A threefold UFS was used to adjust for exposure duration. For BDE-209, the principal
study identified endpoints that, for the most part, reflect specific aspects of developmental
physiology. The hypothesized window of susceptibility, proposed by the study authors, is based
on the observation that the developmental neurotoxic effects observed following exposure to
BDE-209 on PND 3 will not occur once the toxicokinetics of intestinal uptake and excretion
have matured and the animal brain is developmentally less active (outside the window of
susceptibility). The Eriksson/Viberg group has suggested that the period of maximum
vulnerability for the developing cholinergic system that coincides with the most pronounced
neurodevelopmental effects from BDE-99 exposure is from PNDs 10-14. However, for BDE-
209 the developmental neurotoxic effects were noted following exposure on PND 3 rather than
on PNDs 10 or 19. It is likely that two factors contributed to this difference in the timing for the
peak period of vulnerability. First, absorption from the gastrointestinal tract decreases as the
neonatal mice age during their first few weeks of life; thus, less of the BDE-209 reaches the liver
for distribution on PNDs 10 and 19 than on PND 3. Secondly, distribution to the brain is slow.
The amount of radiolabel in the brain is 1.4- and 2.8-fold greater 7 days after dosing compared
with 24 hours after dosing on PNDs 3 and 10. Even less reaches the brain when the BDE-209 is
administered on day 19. The UFS was viewed as a dosing duration adjustment rather than simply
a comparison of the effects of a subchronic to a chronic exposure. A threefold UFS was applied
because the critical study dosed the animals only once within the hypothesized critical window,
not because the chronic exposures would have exacerbated the impact on habituation.
56
-------
A UFLfor LOAEL-to-NOAEL extrapolation was not applied because a NOAEL was used
as the point of departure.
A UFD to account for deficiencies in the available decaBDE database was not necessary.
Available animal studies on repeated oral exposure to decaBDE include 14-day studies in rats
and mice (NTP, 1986), 13-week studies in rats and mice (NTP, 1986), and 2-year studies in rats
and mice (NTP, 1986). A developmental and behavioral toxicity study in male and female mice
was available (Rice et al., 2007), as well as a behavioral study in male rats (Viberg et al., 2007).
A 7-week study (Tseng et al., 2006) of sperm functions in mice was also available. In addition, a
standard developmental toxicity study in rats (Hardy et al., 2002) was identified for decaBDE.
No multigeneration reproductive toxicity study or other study of reproductive function is
available for pure decaBDE. This array of studies results in potential uncertainty regarding the
reproductive toxicity of decaBDE. However, this potential uncertainty is adequately accounted
for based on the following considerations. First, none of the well-conducted longer-term dosing
studies identified effects on male or female reproductive organs. Second, no developmental or
reproductive effects were observed at doses up to 1,000 mg/kg-day in rats (Hardy et al., 2002)
and 100 mg/kg-day in mice (Tseng et al., 2006). The absence of effects in the available longer-
term and developmental studies indicates that at least some aspects of reproductive organ
toxicity or function are not affected at doses much higher than those that resulted in the
neurological effects in neonates (Viberg et al., 2003a), although effects on mating and fertility
are not evaluated in these studies.
The other potential uncertainty in the database factor is immunotoxicity from exposure to
decaBDE. The potential immunotoxicity was indicated by the observations of significant
increases in spleen fibrosis and lymphoid hyperplasia in male rats treated with a high dose of
decaBDE (NTP, 1986). However, no such changes occurred in female rats or in either sex of
treated mice (NTP, 1986). In addition, an in vitro immunotoxicity study (Pullen et al., 2003)
with mouse splenocytes suggested that decaBDE is not likely to affect the immune system in an
immunosuppressive manner nor the production of the cytokines by these cells. Moreover, the
proposed point of departure is based on the developmental neurobehavioral changes and
immunotoxicity is not likely to occur at the current point of departure of 2.22 mg/kg, which is
500-fold lower than the NOAEL for histopathologic changes in rat spleen and lymphoid in the
NTP (1986) study.
Based on the choice of the critical effect and UF selections, the oral RfD for decaBDE is
calculated as follows:
RfD = NOAEL-UF
= 2.22 mg/kg - 300
= 0.007 mg/kg-day or 7 |ig/kg-day
57
-------
5.1.4. Previous RfD Assessment
An IRIS health assessment of decaBDE is available (U.S. EPA, 1989). The RfD is based
on the study of Kociba et al. (1975) in which male and female Sprague-Dawley rats (25/sex/dose
group) were treated with a commercial decaBDE (77.4% decabromodiphenyl oxide, 21.8%
nonaBDE, and 0.8% octaBDE) at doses of 0, 0.01, 0.1, or 1.0 mg/kg-day for 2 years. Parameters
examined were hematology, clinical chemistry, food consumption, organ weight, body weight,
and incidence of histopathologic lesions. No significant differences among treatment and
control groups were found. The NOAEL in this study was 1.0 mg/kg-day, the highest dose
tested. A UF of 100 was applied to the NOAEL for both the expected intra- and interspecies
variability to the toxicity of this chemical in lieu of specific data, resulting in a RfD of 0.01
mg/kg-day or 10 jig/kg-day. Confidence in the RfD was considered low (The low doses and
number of animals used in this study have been questioned as to their adequacy to determine the
carcinogenic potential of this commercial decaBDE [NTP, 1986]). The previous RfD was not
based on the NTP (1986) study because the final NTP report was not available at the time.
The National Research Council of the National Academies of Science (NAS, 2000)
derived a RfD for decaBDE based on the NTP (1986) study. The LOAEL for liver thrombosis
and degeneration in male rats was 2240 mg/kg-day and the NOAEL 1120 mg/kg-day. The
following UFs were applied: 10 for extrapolation from animals to humans, 10 for intraspecies
variation, and 3 for database deficiency. Applying this total UF of 300 to the NOAEL of
1120 mg/kg-day gives a RfD of 4.0 mg/kg-day. Confidence in this provisional RfD was
considered medium to low.
The Agency for Toxic Substances and Disease Registry (ATSDR, 2004) derived an
intermediate oral minimal risk level (MRL) of 10 mg/kg-day based on the Hardy et al. (2002)
study. No effects on any maternal or fetal endpoints were observed in any dose group. The
NOAEL in this study was the highest dose tested of 1000 mg/kg-day. Applying a total UF of 100
(10 for extrapolation from animals to humans and 10 for human variability), an intermediate oral
MRL of 10 mg/kg-day was derived.
5.2. INHALATION REFERENCE CONCENTRATION (RfC)
No data are available for deriving a RfC for decaBDE.
5.3. CANCER ASSESSMENT
5.3.1. Choice of Study/Data—with Rationale and Justification
As discussed in section 4.6, the weight of evidence of human carcinogenicity of
decaBDE is based on (1) no studies of cancer in humans exposed to decaBDE; (2) a significantly
increased incidence of neoplastic nodules in the liver of low- and high-dose male rats and high-
dose female rats; (3) a significantly increased incidence of hepatocellular adenoma or carcinoma
58
-------
(combined) in male mice at low dose and marginally increased incidence at high dose; (4) a
nonsignificantly increased incidence of hepatocellular adenoma or carcinoma (combined) in
female mice; (5) a slight (but statistically not significant) increase in incidence of thyroid gland
adenomas or carcinomas (combined) in both male and female mice; (6) a significantly increased
incidence in male mice of follicular cell hyperplasia, considered by many as a precursor to
thyroid tumors; and (7) an apparent absence of genotoxic potential. All of the data supporting
carcinogenicity were obtained from chronic studies in rats and mice (NTP, 1986).
As discussed in section 4.6, the weight of evidence suggests that decaBDE shows
"suggestive evidence of carcinogenic potential." Support for tumorigenic effects in the liver of
varying strengths is found in both sexes of rats and mice. In the NTP (1986) study, decaBDE
caused a dose-dependent increased incidence of liver neoplastic nodules in rats with statistical
significance at the high dose in both sexes (Table 4-4), indicating some evidence of
carcinogenicity in treated rats. However, there were no significant changes in hepatocellular
carcinoma in either sex. In mice, nonsignificant increases in hepatocellular adenoma or
carcinoma occurred in male mice at low (3,200 mg/kg-day) and high (6,650 mg/kg-day) doses
compared with controls. However, combined incidence of hepatocellular adenoma or carcinoma
increased significantly in the low-dose group but not in the high-dose group. Increased
incidence of hepatocellular adenoma and carcinoma was also observed in the female mice;
however, these changes were not statistically significant. Similarly, although the increases in
thyroid tumors were not statistically significant in either sex of mice, they were supported by
increases in a precursor to thyroid tumors, follicular cell hyperplasia.
Therefore, the NTP (1986) chronic studies in mice and rats were considered the basis for
the quantitative cancer assessment. Liver and thyroid tumors were considered as possible bases
for the quantitation. To select the most sensitive cancer endpoint as the point of departure to
derive a cancer risk value, benchmark modeling was conducted for each tumor endpoint or
related precursor (i.e., thyroid hyperplasia). The BMD analysis and corresponding results are
summarized in the following sections.
5.3.2. Dose Adjustments
For the chronic rat study (NTP, 1986), the original oral doses were reported as 0, 1,120,
and 2,240 mg/kg-day for males and 0, 1,200, and 2,550 mg/kg-day for females. Since there are
not enough toxicokinetic data for conducting a quantitative interspecies extrapolation, a default
dose conversion was used. Based on EPA's Guidelines for Carcinogen Risk Assessment (U.S.
EPA, 2005a), corresponding human equivalent doses (HEDs) were calculated by scaling animal
daily applied doses experienced for a lifetime in proportion to body weight raised to the 0.75
power.
59
-------
A body weight (bw)3 4 scaling factor was used to convert the points of departure in the rat
study to HEDs, in accordance with the Guidelines for Carcinogen Risk Assessment (U.S. EPA,
2005a). This procedure presumes that equal doses in these units (i.e., in mg/kg3/4-day), when
administered daily over a lifetime, will result in equal lifetime risks of the critical effect across
mammalian species (U.S. EPA, 1992). The HED may be calculated as follows (U.S. EPA,
2005a, 1992):
0.25
HED (mg/kg-day) = dose in animals (mg/kg-day) x (bwa/bwh)
where:
HED = human equivalent dose
dose = average daily dose in animal study
bwa = animal body weight (kg)
bwh = reference human body weight (70 kg)
Based on reported weekly body weight data for each dose group for male rats, average
lifetime body weights were calculated as 383.9, 385.2, and 380.7 g for the control, low-, and
high-dose groups, respectively. Using the calculated average lifetime rat body weight and a
default human body weight of 70 kg, the corresponding HEDs were calculated as 0, 305.1, and
608.3 mg/kg-day, respectively. Similarly, for female rats, average lifetime body weights were
calculated as 247.9, 246.2, and 242.1 g for the control, low-, and high-dose groups, respectively.
Based on the calculated body weight and default human body weight of 70 kg, corresponding
HEDs were calculated as 0, 292.2, and 618.4 mg/kg-day, respectively.
For the chronic mouse study (NTP, 1986), the only dose-response data on carcinogenicity
available were obtained from male mice; therefore, the dose conversion was conducted for male
mice only. The original oral doses for male mice were reported as 0, 3,200, and 6,650 mg/kg-
day. Since there are not enough toxicokinetic data for conducting a quantitative interspecies
extrapolation, a default dose conversion was used. Based on reported weekly body weight data
for each dose group, average lifetime body weights were calculated as 37.3, 37.4, and 37.0 g for
control, low-, and high-dose groups, respectively. Based on these data on average lifetime
mouse body weight and a default human body weight of 70 kg, corresponding HEDs were
calculated by scaling animal daily applied doses experienced for a lifetime in proportion to body
weight raised to the 0.75 power. The estimated HEDs are 0, 486.5, and 1,008.6 mg/kg-day,
respectively.
5.3.3. Extrapolation Method(s)
U.S. EPA BMDS (version 1.40b) was used in the cancer endpoint BMD analysis.
Following the guidelines for evaluation of data on tumorigenic responses, the cancer model
60
-------
(a multistage model with a calculation function for slope factor) was used to calculate effective
doses (EDs) for a tumor response within the range of observation and the lower confidence
estimate on that dose. The lower confidence limit of the estimated dose for a BMR similar to
the response observed in the low-dose group is used as a point of departure to estimate an oral
slope factor (U.S. EPA, 2005a, b).
All potential tumor endpoints with statistically significant responses in the rat and mouse
chronic studies (NTP, 1986) were modeled in order to identify the most sensitive point of
departure. Animals dying prior to week 53, before the first appearance of tumors, were censored
from the group totals to adjust the incidence rates for early mortality. To adjust for differential
mortality among treated male rats, censored data on neoplastic nodules or the combined
neoplastic nodules and carcinomas in rats of both sexes were also modeled. The data on
combined hepatic carcinomas and adenomas in male mice were modeled despite the fact that
they did not show a good dose-response relationship because they were significantly increased in
comparison with controls. A detailed description of the BMD modeling results is presented in
Appendix C. The estimated cancer slope factors based on estimated EDs and their 95% lower
confidence limits using the multistage model for each cancer endpoint are summarized in Table
5-2.
Table 5-2. Cancer slope factors derived from BMDs for neoplastic effect
from chronic rat and mouse studies using the multistage model
Endpoint
Neoplastic nodules in the liver
Neoplastic nodules or carcinomas (combined) in the liver
Neoplastic nodules in the liver
Neoplastic nodules or carcinomas (combined) in the liver
Follicular cell hyperplasia in the thyroid
Adenomas or carcinomas (combined) in the liver
Species
Male rat
Male rat
Female rat
Female rat
Male mouse
Male mouse
Cancer slope factor
(mg/kg-day)'1
0.0007
0.0007
0.0004
0.0005
0.0005
0.0005
Source: NTP (1986).
Neoplastic nodules and carcinomas were also modeled together because the two lesions
could have a similar mode of action. Based on the estimated ED for each cancer endpoint, the
most sensitive response is neoplastic nodule or carcinoma (combined) in the liver of male rats.
The ED12 from the lower end of the range of observation is 263 mg/kg-day, and the
corresponding 95% lower confidence limit for the effective dose (LED12) is 178 mg/kg-day. For
neoplastic nodules or carcinomas in the liver of female rats, both censored and uncensored data
resulted in higher LEDs than for this effect in male rats; therefore, the LED in females was not
used as the point of departure. Similarly, the estimated LED for the follicular cell hyperplasia in
the thyroid of male mice is also higher than that of neoplastic nodules or carcinomas (combined)
61
-------
in the male rats. Thus, the lowest LED, 178 mg/kg-day for neoplastic nodules or carcinomas
(combined) in the male rats, is selected as the point of departure for deriving an oral cancer slope
factor. However, as stated earlier (section 4.2.2.1), the term neoplastic nodule is no longer used
for rat liver tumors, and it is not possible to know if all lesions originally categorized as
neoplastic nodules were preneoplastic under current guidelines. Some may have been
nonneoplastic hyperplasia. Accordingly, there is some uncertainty in the calculated slope factor
that should be considered when it is applied in quantitative risk assessment.
5.3.4. Oral Slope Factor
Although no human studies were available, two chronic rodent studies provide suggestive
evidence of decaBDE-induced carcinogenicity. The data from these studies are adequate to
support a quantitative cancer dose-response assessment. Even though the available evidence is
suggestive of human carcinogenic potential, there is very limited information exploring the mode
of action for any of the tumors reported in the animal chronic studies. While decaBDE was not
mutagenic or genotoxic in various in vitro studies, there are no data to support alternative mode-
of-action hypotheses. In the absence of such data, a mode of action for BDE-209 could not be
determined; therefore, extrapolation from the point of departure to lower doses is conducted by
using a linear approach.
Based on a comparison of estimated EDs for all the cancer endpoints observed in rat and
mouse chronic studies (NTP, 1986), the neoplastic nodules or carcinomas (combined) in the
liver of treated male rats are the most sensitive endpoint. Therefore, the LED12 of 178 mg/kg-
day estimated for this endpoint is used as a point of departure for calculating the cancer slope
factor.
For linear extrapolation, a straight line is drawn from the point of departure expressed as
an HED to the origin to give a probability of extra risk. The slope of the line expresses extra risk
per dose unit. For linear extrapolation, the slope of the line is 0.12/LED12. The central estimate,
ED12, of exposure at 12% extra risk is 0.12/ED12. The slope of the linear extrapolation from the
central estimate ED12 is 5 x 10^, which is derived using 0.127(263 mg/kg-day). For neoplastic
nodules or carcinomas, the resulting oral cancer slope factor is 7 x 1CT4 per mg/kg-day, which is
derived using 0.127(178 mg/kg-day). Since some of the neoplastic nodules might not progress to
carcinomas, this oral risk estimate errs on the conservative side. Based on this slope factor, the
dose associated with an excess cancer risk (risk-specific dose [RSD]) value is approximately as
follows: RSD at 10"6 is 1 |ig/kg-day; RSD at 1(T5 is 10 |ig/kg-day; and RSD at 10^ is 100 |ig/kg-
day.
Doses for excess cancer risks of approximately 5 x io~6 or lower would be protective of
neurodevelopmental effects since the RfD established on the basis of these effects is 7 jig/kg-day
(see section 5.1.3). As explained above, there is some uncertainty in this slope factor because
62
-------
some of the neoplastic nodules might have been nonneoplastic hyperplasia rather than
preneoplastic foci of cellular alteration or hepatic adenomas. The combination of the
hyperplastic nodules and carcinomas for the dose-response analysis leads to a conservative but
protective slope factor.
5.3.5. Previous Cancer Assessment
The carcinogenicity of decaBDE was evaluated in IRIS (U.S. EPA, 1989). DecaBDE
was classified in Group C, "possible human carcinogen," according to EPA cancer guidelines
(U.S. EPA, 1986b). The basis of this classification was lack of human carcinogenicity data and
limited evidence of carcinogenicity in animals, namely, significantly increased incidences of
neoplastic liver nodules in male and female rats and increased incidences of hepatocellular
adenomas or carcinomas (combined) in male mice (NTP, 1986). A quantitative estimate of
carcinogenic risk from oral exposure was not derived in the IRIS assessment because the final
NTP (1986) report was not available at the time.
On the basis of the carcinogenicity studies in F344/N rats and B6C3F1 mice, NTP (1986)
concluded that there was "some evidence of carcinogenicity" for male and female rats as shown
by increased incidences of neoplastic nodules of the liver in low-dose males and high-dose
groups of both males and females. There was "equivocal evidence of carcinogenicity" for male
mice as shown by increased incidences of hepatocellular adenomas or carcinomas (combined) in
the low-dose group and of thyroid gland follicular cell adenomas or carcinomas (combined) in
both dosed groups. There was "no evidence of carcinogenicity" for female mice receiving low
or high dose of decaBDE in the diet. Several nonneoplastic lesions were observed at increased
incidences, the most notable being thyroid gland follicular cell hyperplasia in male mice.
The potential carcinogenicity of decaBDE was evaluated by NAS (2000) on the basis of
the NTP (1986) study. Based on lack of human data and limited evidence of carcinogenicity in
animals (i.e., significant increased incidences of neoplastic liver nodules in male and female rats
and increased incidences of hepatocellular adenomas or carcinomas in male mice), NAS (2000)
concluded that decaBDE is a possible carcinogen in rats but has not concluded that it is a
carcinogen in humans. Based on the incidence of hepatic neoplastic nodules in male rats (NTP,
1986), a cancer slope factor of 9 x io~4 per mg/kg-day was derived, applying the multistage
model to censored data to adjust for differential mortality among treated rats.
63
-------
6. MAJOR CONCLUSIONS IN THE CHARACTERIZATION OF
HAZARD AND DOSE RESPONSE
6.1. HUMAN HAZARD POTENTIAL
Studies of toxicokinetics of decaBDE reveal that the chemical can be absorbed by the
oral route to a limited extent, does not accumulate in tissues, and undergoes clearance, largely as
a result of metabolism in the liver and excretion in the bile.
Short-term and subchronic studies demonstrated low toxicity from oral exposure to
decaBDE with NOAELs of 3,000 mg/kg-day or higher. NTP (1986) conducted a chronic toxicity
and carcinogenicity dietary study in F344 rats. DecaBDE caused an increase in the incidence of
thrombosis in the liver in high-dose male rats (2,240 mg/kg-day). A dose-dependent, but
insignificant, increase in the incidence of degeneration of the liver was also observed in treated
male rats. In the spleen, a dose-dependent increase (statistically significant in the high-dose
group) in the incidence of fibrosis was observed in males. In the mandibular lymph node,
lymphoid hyperplasia increased in males in a dose-dependent manner, but the incidence reached
statistical significance only at the high dose. Histopathology examination also revealed a dose-
dependent increase in the incidence of neoplastic nodules in the liver in both male and female
rats. Female rats appeared to be refractory to the systemic toxicity of decaBDE at the doses used
in this study.
The observed toxicity of decaBDE in the 2-year study in rats is further supported by the
2-year mouse study conducted by NTP (1986). Significant increases in the incidence of
centrilobular hypertrophy were observed in the liver of treated male mice. In the thyroid gland, a
dose-dependent and statistically significant increase (at all dose levels) in the incidence of
follicular cell hyperplasia was observed in male mice. In the females, the incidence increased in
the low- and high-dose groups compared with the control group, but the increase was not
statistically significant at any dose level. Female mice in the high-dose group exhibited a
significant increase in the incidence of stomach ulcers. In addition, there were significant
increases in the combined incidence of hepatocellular adenomas or carcinomas at both low and
high doses in male mice. In the thyroid gland, follicular cell adenomas or carcinomas
(combined) were slightly, but not significantly, increased in treated mice of both sexes. Similar
to female rats, female mice appeared to be refractory to the systemic toxicity of decaBDE.
DecaBDE also has been shown to induce behavioral changes in several studies in mice
and rats (Viberg et al., 2007, 2003a; Rice et al., 2007). In the principal study selected, Viberg et
al. (2003a) investigated the neurotoxic effects of decaBDE on spontaneous motor behavior of
adult NMRI male mice when these animals were exposed to a single oral dose as neonates on
PND 3, 10, or 19 (i.e., at different stages of neonatal mouse brain development). Pair-wise
64
-------
testing between adult mice exposed on PND 3 and control groups indicated significant dose-
related changes in all three spontaneous behavior variables at 2, 4, and 6 months of age. Adult
mice exposed neonatally up to 20.1 mg on either PND 10 or 19 did not show any significant
differences in any of the variables. These data suggested that there was a critical window for the
induction of behavioral disturbances, and the neurotoxic effect of neonatal decaBDE exposure
was persistent and also worsened with age in male mice.
The appropriate hazard descriptor for decaBDE is "suggestive evidence of carcinogenic
potential" (U.S. EPA, 2005a, b). The weight-of-evidence of human carcinogenicity of decaBDE
is based on (1) no studies of cancer in humans exposed to decaBDE; (2) a statistically significant
increase in incidence of neoplastic nodules and a slight increase in incidence of carcinomas (not
statistically significant) in the liver of low- and high-dose male rats and high-dose female rats;
(3) a significantly increased incidence of hepatocellular adenoma or carcinoma (combined) in
male mice at the low dose and marginally increased incidence at the high dose; (4) a
nonsignificantly increased incidence of hepatocellular adenoma or carcinoma (combined) in
female mice; (5) a slightly greater (but statistically not significant) incidence of thyroid gland
adenomas or carcinomas (combined) in dosed male and female mice; (6) a significantly
increased incidence in male mice, at both doses, of follicular cell hyperplasia, considered by
many as a precursor to thyroid tumors; and (7) an apparent absence of genotoxic potential.
The weight of experimental evidence is on the strong end of the spectrum for the
descriptor "suggestive evidence of carcinogenic potential," since there is suggestive evidence
that decaBDE is carcinogenic for more than one species, sex, and site. Based on the weight of
evidence, a dose-response assessment of the carcinogenicity of decaBDE is deemed appropriate.
6.2. DOSE RESPONSE
6.2.1. Noncancer/Oral
BMD modeling was conducted for all the potential critical effects observed in the rat and
mouse chronic studies, including thrombosis in the liver, liver degeneration, fibrosis in the
spleen, and lymphoid hyperplasia in male rats, as well as centrilobular hypertrophy in the livers
and follicular cell hyperplasia in the thyroid of male mice. The data on neurobehavioral effects
in the Viberg et al. (2003a) study could not be modeled because the data were presented
graphically rather than numerically and thus were not amenable to BMD analysis. The lowest
BMDL10 in rodent chronic studies is 383 mg/kg-day for liver degeneration effect in male rats.
This value is much higher than the NOAEL level of 2.22 mg/kg for the neurobehavioral changes
observed in mice (Viberg et al., 2003a). Therefore, the NOAEL of 2.22 mg/kg is used as a point
of departure for calculating the oral RfD. A composite UF of 300 was applied to this effect level
to account for the interspecies uncertainty (10-fold), intraspecies variation (10-fold), and
65
-------
extrapolation of a single dose to a lifetime exposure (3-fold), yielding an RfD of 0.007 mg/kg-
day or 7 |ig/kg-day.
The available database included acute, short-term, subchronic, and chronic studies in two
species. Also, there are developmental and reproductive studies (Tseng et al., 2006; Hardy et al.,
2002) in addition to the critical neurobehavioral study. Confidence in the principal study
(Viberg et al., 2003a) is low because the study used a single dose during a period of
neurodevelopmental vulnerability. In addition, only limited tests on motor activity were
conducted in only male mice in the critical study by Viberg et al. (2003a). This study is
strengthened by the results of Rice et al. (2007), where behavioral effects were identified in
neonatally exposed PND 70 mice following a repeated postnatal dose protocol, and also by the
results of the Viberg et al. (2007) study in rats. The Viberg et al. (2003a) study provides
information on the most sensitive endpoint; therefore, using the NOAEL from this study as a
point of departure to calculate the RfD is expected to provide adequate protection for humans.
The overall confidence in the RfD assessment of BDE-209 is low.
6.2.2. Cancer/Oral
Although no human studies were available, two chronic rodent studies (NTP, 1986)
provide suggestive evidence of decaBDE-induced carcinogenicity. The data from these studies
are adequate to support a quantitative cancer dose-response assessment. There is very limited
information exploring the mode of action for any of the tumors reported in the animal chronic
studies. DecaBDE was not mutagenic or genotoxic in several in vitro studies, and there are no
data to support a nongenotoxic mode-of-action hypothesis. In the absence of such data,
extrapolation from the point of departure to lower doses is most appropriate using a linear
approach.
Based on a comparison of estimated EDs for all the cancer endpoints observed in rat and
mouse chronic studies (NTP, 1986), the neoplastic nodule or carcinoma (combined) in the
treated rats is the most sensitive endpoint. Therefore, the LED12 of 178 mg/kg-day estimated for
this endpoint is used as a point of departure for calculating the cancer slope factor. For
neoplastic nodules or carcinomas (combined), the resulting oral cancer slope factor is 7 x 1CT4
per mg/kg-day. This slope factor assumes that all neoplastic nodules were preneoplastic cellular
changes with the potential to become malignant. Based on this slope factor, the doses associated
with excess cancer risks of 10~4, 10~5, and 10~6 are approximately 100, 10, and 1 |ig/kg-day,
respectively. There is some uncertainty in this estimate, given the fact that the tissues identified
as neoplastic nodules may have included some lesions now categorized as nonneoplastic
hyperplasia (Maronpot et al., 1986).
66
-------
7. REFERENCES
ACC (American Chemistry Council). (2002) Voluntary Children's Chemical Evaluation Program (VCCEP). Data
summary. Decabromodiphenyl ether (decabromodiphenyl oxide, DBDPO). Prepared by the American Chemistry
Council's Brominated Flame Retardant Industry Panel, Arlington, VA; December 17. Available online at
http://regulations.gov; Document ID EPA-HQ-OPPT-2004-0085-0032 in docket "Certain polybrominated
diphenylethers; proposed significant new use rule."
Ankarberg, E. (2003) Neurotoxic effects of nicotine during neonatal brain development. Comprehensive summaries
of Uppsala Dissertations from the Faculty of Science and Technology 907. Acta Universitatis Upsaliensis, Uppsala,
Sweden.
ATSDR (Agency for Toxic Substances and Disease Registry). (2004) Toxicological profile for polybrominated
biphenyls and polybrominated diphenyl ethers. Public Health Service, U.S. Department of Health and Human
Services, Atlanta, GA. Available online at http://www.atsdr.cdc.gov/toxpro2.html.
Bock, KW. (1994) Arylhydrocarbon of dioxin receptor: biologic and toxic responses. Rev Physiol Biochem
Pharmacol 125:1-42.
Bradman A; Fenster, L; Sjodin, A; et al. (2007) Polybrominated diphenyl ether levels in the blood of pregnant women
living in an agriculture community in California. Environ Health Perspect 115(l):71-74.
Branchi, I; Alleva, E; Costa, LG. (2002) Effects of perinatal exposure to a polybrominated diphenyl ether (PBDE 99)
on mouse neurobehavioural development. Neurotoxicol 23(3):375-384.
Brown, DJ; Overmeire, IV; Goeyens, L; et al. (2004) Analysis of Ah receptor pathway activation by brominated
flame retardants. Chemosphere 55:1509-1518.
Carlson, GP. (1980) Induction of xenobiotic metabolism in rats by short-term administration of brominated diphenyl
ethers. Toxicol Lett 5:19-25.
Cetin, B; Odabasi, M. (2005) Measurement of Henry's law constants of seven polybrominated diphenyl ether (PBDE)
congeners as a function of temperature. Atmos Environ 3 9:5273-5280.
Chen, G; Konstantinov, AD; Chittim, BG; et al. (2001) Synthesis of polybrominated diphenyl ethers and their
capacity to induce CYP-1A by the Ah receptor mediated pathway. Environ Sci Technol 35:3749-3756.
Darnerud, PO; Eriksen, GS; Johannesson, T; et al. (2001) Polybrominated diphenyl ethers: occurrence, dietary
exposure, and toxicology. Environ Health Perspect 109:49-68.
ECB (European Chemicals Bureau). (2003) Bis(pentabromophenyl) ether. Summary risk assessment report.
Luxembourg, Belgium: Office for Official Publications of the European Communities. Available online at
http://ecb.jrc.it/DOCUMENTS/Existmg-Chemicals/RISK_ASSESSMENT/SUMMARY/decasum013.pdf.
el Dareer, SM; Kalin, JR; Tillery, KF; et al. (1987) Disposition of decabromodiphenyl ether in rats dosed
intravenously or by feeding. J Toxicol Environ Health 22(4):405^U5.
Eriksson, P; Jakobsson, E; Fredriksson, A. (2001) Brominated flame retardants: a novel class of developmental
neurotoxicants in our environment? Environ Health Perspect 109(9):903-908.
Eriksson, P; Viberg, H; Jakobsson, E; et al. (2002) A brominated flame retardant, 2,2',4,4',5-pentabromodiphenyl
ether: uptake, retention, and induction of neurobehavioural alterations in mice during a critical phase of neonatal brain
development. Toxicol Sci67(l):98-103.
Fischer, D; Hooper, K; Athanasiadou, M; et al. (2006) Children show highest levels of polybrominated diphenyl
ethers in a California family of four: a case study. Environ Health Perspect 114(10): 1581-1584.
67
-------
Gravance, CG; Garner, DL; Miller, MG; et al. (2001) Fluorescent probes and flow cytometry to assess rat sperm
integrity and mitochondria! function. Reprod Toxicol 15:5-10.
Hakk, H; Larsen, G; Bergman, A; et al. (2002) Binding of brominated diphenyl ethers to male rat carrier proteins.
Xenobiotica 32(12): 1079-1091.
Hard, GC. (1998) Recent developments in the investigation of thyroid regulation and thyroid carcinogenesis. Environ
Health Perspect 106(8):427^36.
Hardy, ML. (2002) A comparison of the properties of the major commercial PBDPO/PBDE product to those of
major PBB and PCB products. Chemosphere 46:717-728.
Hardy, ML; Schroeder, R; Biesemeier, J; et al. (2002) Prenatal oral (gavage) developmental toxicity study of
decabromodiphenyl oxide in rats. Int J Toxicol 21(2):83-91.
Hill, RN; Erdreich, LS; Paynter, OE; et al. (1989) Thyroid follicular cell carcinogenesis. Fund Appl Toxicol 12:629-
697.
Hughes, MF; Edwards, BC; Mitchell, CT; et al. (2001) In vitro dermal absorption of flame retardant chemicals. Food
Chem Toxicol 39(12): 1263-1270.
Huwe, JK; Smith, DJ. (2007) Accumulation, whole-body depletion, and debromination of decabromodiphenyl ether
in male Sprague-Dawley rats following dietary exposure. Environ Sci Technol 41:2371-2377.
Jakobsson, K; Thuresson, K; Rylander, L; et al. (2002) Exposure to polybrominated diphenyl ethers and
tetrabromobisphenol A among computer technicians. Chemosphere 46:709-716.
Klaassen, CD; ed. (1996) Casarett and Doull's toxicology: the basic science of poisons. 5th edition. New York, NY:
McGraw-Hill; pp. 47-18.
Kociba, RJ; Frauson, LO; Humiston, CG; et al. (1975) Results of a two-year dietary feeding study with
decabromodiphenyl oxide (DBDPO) in rats. J Combust Toxicol 2:267-285. (Cited in Darnerud et al., 2001).
Kodavanti, RS; Ward, TR; Ludewig, G; et al. (2005) Polybrominated diphenyl ether (PBDE) effects in rat neuronal
cultures: 14C-PBDE accumulation, biological effects, and structure-activity relationships. Toxicol Sci 88(1): 181-192.
Kuriyama, SN; Talsness, CE; Grote, K; et al. (2005) Developmental exposure to low dose PBDE 99: effects on male
fertility and neurobehavior in rat offspring. Environ Health Perspect 113:149-154.
Legler, J; Brouwer, A. (2003) Are brominated flame retardants endocrine disrupters? Environ Int 29:879-885.
Maronpot, RR; Montgomery, CA; Boorman, GA; et al. (1986) National Toxicology Program nomenclature for
hepatoproliferative lesions of rats. Toxicol Pathol 14(2):263-273.
McDonald, TA. (2002) A perspective on the potential health risks of PBDEs. Chemosphere 46:745-755.
Morck, A; Hakk, H; Orn, U; et al. (2003) Decabromodiphenyl ether in the rat: absorption, distribution, metabolism,
and excretion. Drug Metab Disp 31:900-907.
NAS (National Academy of Sciences). (2000) To xico logical risks of selected flame-retardant chemicals. National
Research Council, Commission on Life Sciences, Board on Environmental Studies and Technology, Committee on
Toxicology, Subcommittee on Flame-Retardant Chemicals. Washington, DC: National Academy Press.
NRC (National Research Council). (1983) Risk assessment in the federal government: managing the process.
Washington, DC: National Academy Press.
68
-------
NTP (National Toxicology Program). (1986) Toxicology and carcinogenesis studies of decabromodiphenyl oxide
(CAS No. 1163-19-5) inF344/N rats andB6C3Fl mice (feed studies). Public Health Service, U.S. Department of
Health and Human Services; NTP TR 309. Available from the National Institute of Environmental Health Sciences,
Research Triangle Park, NC, and online at http://ntp.niehs.nih.gov/ntp/htdocs/LT_rpts/tr309.pdf.
Pacyniak, EK; Cheng, X; Cunningham, MK; et al. (2007) The flame retardants, polybrominated diphenyl ethers, are
pregnane X receptor activators. Toxicol Sci 97(1):94-102.
Peters, AK; van Londen, K; Bergman, A; et al. (2004) Effects of polybrominated diphenyl ethers on basal and
TCDD-induced ethoxyresorufrn activity and cytochrome P450-1 Al expression in MCF-7, HepG2, and H4 HE cells.
Toxicol Sci 82:488-496.
Pullen, S; Boecker, R; Tieg, G. (2003) The flame retardants tetrabromobisphenol A and tetrabromobisphenol
A-bisallylether suppress the induction of interleukin-2 receptor a chain (CD25) in murine splenocytes. Toxicology
184:11-22.
Rice, DC; Reeve, EA; Herlihy, A; et al. (2007) Developmental delays and locomotor activity in the C57BL6/J mouse
following neonatal exposure to the fully-brominated PBDE, decabromodiphenyl ether. Neurotoxicol Teratol 29:511-
520.
Sandholm, A; Emanuelsson, B-M; Klasson-Wehler, E. (2003) Bio availability and half-life of decabromodiphenyl ether
(BDE-209) inrat. Xenobiotica 33(22): 1149-1158.
Schecter, A; Pavuk, M; Papke, O; et al. (2003) Polybrominated diphenyl ethers (PBDEs) in U.S. mothers' milk.
Environ Health Perspect 111(14): 1723-1729.
She, J; Holden, A; Sharp, M; et al. (2007) Polybrominated diphenyl ethers (PBDEs) and polychlorinated biphenyls
(PCBs) in breast milk from the Pacific Northwest. Chemosphere 67:8307-8317.
Sjodin, A; Hagmar, L; Klasson-Wehler, E; et al. (1999) Flame retardant exposure: polybrominated diphenyl ethers in
blood from Swedish workers. Environ Health Perspect 107(8):643-648.
Sjodin, A; Patterson, DG, Jr; Bergman, A. (200la) Brominated flame retardants in serum from U.S. blood donors.
Environ Sci Technol 35(19):3830-3833.
Sjodin, A; Carlsson, H; Thuresson, K; et al. (200 Ib) Flame retardants in indoor air at an electronics recycling plant
and at other work environments. Environ Sci Technol 35:448-454.
Sjodin, A; Patterson, DG, Jr; Bergman, A. (2003) A review on human exposure to brominated flame retardants—
particularly polybrominated diphenyl ethers. Environ Internal 29:829-839.
Staskal, DF; Hakk, H; Bauer, D; et al. (2006) Toxicokinetics of polybrominated diphenyl ether congeners 47, 99,
100, and 153 in mice. Toxicol Sci 94(l):28-37.
Thuresson, K; Bergman, A; Jakobsson, K (2005) Occupational exposure to commercial decabromodiphenyl ether in
workers manufacturing or handling flame-retarded rubber. Environ Sci Technol 39:1980-1986.
Thuresson, K; Hoglund, P; Hagmar, L; et al. (2006) Apparent half-lives of hepta- to decabrominated diphenyl ethers
in human serum as determined in occupationally exposed workers. Environ Health Perspect 114(2): 176-181.
Tseng, LH; Lee, CW; Pan, MH; et al. (2006) Postnatal exposure of the male mouse to
2,2',3,3',4,4',5,5',6,6'-decabrominated diphenyl ether: decreased epididymal sperm functions without alterations in
DNA content and histology in testis. Toxicology 224:33-43.
U.S. EPA (Environmental Protection Agency). (1986a) Guidelines for mutagenicity risk assessment. Federal
Register 51(185):34006-34012. Available online at http://www.epa.gov/iris^ackgr d.htm.
69
-------
U.S. EPA (Environmental Protection Agency). (1986b) Guidelines for carcinogen risk assessment. Federal Register
51(185):33992-34003. Available online at http://www.epa.gov/cancerguidelines.
U.S. EPA (Environmental Protection Agency). (1986c) Proliferative hepatocellular lesions of the rat: review and
future use in risk assessment. Risk Assessment Forum, Washington, DC; EPA/625/3-86/011. Available from the
National Technical Information Service, Springfield, VA, PB87-178711, and online at
http ://nepis. epa. gov/EP A/html/Pubs/pubtitleORD. htm.
U.S. EPA (Environmental Protection Agency). (1988) Recommendations for and documentation of biological values
for use in risk assessment. Environmental Criteria and Assessment Office, Office of Health and Environmental
Assessment, Cincinnati, OH; EPA/600/6-87/008. Available from the National Technical Information Service,
Springfield, VA, PB88-179874/AS, and online at http://cfpub.epa.gov/ncea/cfm/recordisplay.cfm?deid=34855.
U.S. EPA (Environmental Protection Agency). (1989) Decabromodiphenyl ether (CASRN 1163-19-5). Integrated
Risk Information System (IRIS). National Center for Environmental Assessment, Washington, DC. Available online
at http://www.epa.gov/iris.
U.S. EPA (Environmental Protection Agency). (1991) Guidelines for developmental toxicity risk assessment. Federal
Register 56:63798-63826. Available online at http://www.epa.gov/ncea/raf/rafguid.htm.
U.S. EPA (Environmental Protection Agency). (1992) Draft report: a cross-species scaling factor for carcinogen risk
assessment based on equivalence of mg/kg3/4/day. Federal Register 57(109):24152-24173.
U.S. EPA. (1994a) Interim policy for particle size and limit concentration issues in inhalation toxicity studies. Federal
Register 59(206):53799. Available from: .
U.S. EPA. (1994b) Methods for derivation of inhalation reference concentrations and application of inhalation
dosimetry. Office of Research and Development, Washington, DC; EPA/600/8-90/066F. Available from:
.
U.S. EPA (Environmental Protection Agency). (1995) Use of the benchmark dose approach in health risk assessment.
Risk Assessment Forum, Washington, DC; EPA/630/R-94/007. Available from the National Technical Information
Service, Springfield, VA, PB95-213765, and online at
http://cfpub.epa.gov/ncea/raf/raf_pubtitles.cfm?detype=document&excCol=archive.
U.S. EPA (Environmental Protection Agency). (1996) Guidelines for reproductive toxicity risk assessment. Federal
Register 61:56274-56322. Available online at http://www.epa.gov/ncea/raf/rafguid.htm.
U.S. EPA (Environmental Protection Agency). (1998a) Guidelines for neurotoxicity risk assessment. Federal
Register 63:26926-26954. Available online at http://www.epa.gov/ncea/raf/rafguid.htm.
U.S. EPA (Environmental Protection Agency). (1998b) Assessment of thyroid follicular cell tumors. Risk
Assessment Forum, Washington, DC; EPA/630/R-97/002. Available from the National Technical Information
Service, Springfield, VA, PB98-133119, and online at http://nepis.epa.gov/EPA/html/Pubs/pubtitleORD.htm.
U.S. EPA (Environmental Protection Agency). (1998c) Health effects test guidelines: neurotoxicity screening battery.
Office of Prevention, Pesticides and Toxic Substances, Washington, DC; OPPTS 870.6200; EPA 712-C-98-238.
Available online at
http://www.epa.gov/opptsfrs/publications/OPPTS_Harmonized/870_Health_Effects_Test_Guidelines/Series/870-
6200.pdf.
U.S. EPA (Environmental Protection Agency). (2000a) Science policy council handbook: risk characterization.
Office of Science Policy, Office of Research and Development, Washington, DC. EPA/100-B-00-002. Available
online at http://www.epa.gov/OSA/spc/pdfs/prhandbk.pdf.
U.S. EPA (Environmental Protection Agency). (2000b) Benchmark dose technical guidance document [external
review draft]. Risk Assessment Forum, Washington, DC; EPA/630/R-00/001. Available online at
70
-------
http ://cfpub. epa. gov/ncea/cfm/
nceapubUcation.cfm?ActType=PublicationTopics&detype=DOCUMENT&subject=BENCHMARK+DOSE&subjtype
=TITLE&excCol= Archive.
U.S. EPA. (2000c) Supplementary guidance for conducting for health risk assessment of chemical mixtures. Risk
Assessment Forum, Washington, DC; EPA/630/R-00/002. Available from: .
U.S. EPA (Environmental Protection Agency). (2002) A review of the reference dose and reference concentration
processess. Risk Assessment Forum, Washington, DC; EPA/630/P-02/002F. Available online at
http://cfpub.epa.gov/ncea/raf/raf_pubtitles.cfm?detype=document&excCol=archive.
U.S. EPA (Environmental Protection Agency). (2004) Decabromodiphenyl ether. Substance Registry System. U.S.
Environmental Protection Agency, Washington, DC. Available online at http://www.epa.gov/srs.
U.S. EPA (Environmental Protection Agency). (2005a) Guidelines for carcinogen risk assessment. Federal Register
70:17765-18717. Available online at http://www.epa.gov/cancerguidelines.
U.S. EPA (Environmental Protection Agency). (2005b) Supplemental guidance for assessing susceptibility from
early-life exposure to carcinogens. Risk Assessment Forum, Washington, DC; EPA/630/R-03/003F. Available online
at http://www.epa.gov/cancerguidelines.
U.S. EPA (Environmental Protection Agency). (2006a) Science policy council handbook: peer review. 3rd edition.
Office of Science Policy, Office of Research and Development, Washington, DC; EPA/100/B-06/002. Available
online at http://www.epa.gov/OSA/spc/2peerrev.htm.
U.S. EPA (Environmental Protection Agency). (2006b) A framework for assessing health risk of environmental
exposures to children. National Center for Environmental Assessment, Washington, DC, EPA/600/R-05/093F.
Available online at http://cfpub.epa.gov/ncea/cfm/recordisplay.cfm?deid=158363.
Viberg, H; Fredriksson, A; Eriksson, P. (2002) Neonatal exposure to the brominated flame retardant
2,2',4,4',5-pentabromodiphenyl ether causes altered susceptibility in the cholinergic transmitter system in the adult
mouse. Toxicol Sci 67(1): 104-107.
Viberg, H; Fredriksson, A; Jakobsson, E; et al. (2003a) Neurobehavioral derangements in adult mice receiving
decabromodiphenyl ether (PBDE 209) during a defined period of neonatal brain development. Toxicol Sci 76:112-
120.
Viberg, H; Frederiksson, A; Eriksson, P. (2003b) Neonatal exposure to polybrominated diphenyl ether (PBDE 153)
disrupts spontaneous behaviour, impairs learning and memory, and decreases hippocampal cholinergic receptors in
adult mice. Toxicol Appl Pharmacol 192(2):95-106.
Viberg, H; Fredriksson, A; Jakobsson, E; et al. (2004a) Neonatal exposure to the brominated flame-retardant,
2,2',4,4',5-pentabromodiphenyl ether, decreases cholinergic nicotinic receptors in hippocampus and affects
spontaneous behaviour in the adult mouse. Environ Toxicol Pharmacol 17:61-65.
Viberg, H; Fredriksson, A; Eriksson, P. (2004b) Investigations of strain and/or gender differences in developmental
neurotoxic effects of polybrominated diphenyl ethers in mice. Toxicol Sci 81:344-353.
Viberg, H; Fredriksson, A; Eriksson, P. (2005) Deranged spontaneous behavior and decrease in cholinergic
muscarinic receptors in hippocampus in the adult rat, after neonatal exposure to the brominated flame-retardant,
2,2',4,4',5-pentabromodiphenyl ether (PBDE 99). Environ Toxicol Pharmacol 20:283-288.
Viberg, H; Fredriksson, A; Eriksson, P. (2007) Changes in spontaneous behavior and altered response to nicotine in
the adult rat, after neonatal exposure to the brominated flame retardant, decabrominated diphenyl ether (PBDE 209).
Neurotoxicol 28:136-142.
71
-------
Villeneuve, DL; Kannan, K; Priest, BT; et al. (2002) In vitro assessment of potential mechanism-specific effects of
polybrominated diphenyl ethers. Environ Toxicol Chem 21(11):2431-2433.
Wolf, DC; Mann, PC. (2005) Confounders in interpreting pathology for safety and risk assessment. Toxicol Appl
Pharmacol 202:302-308.
Zhou, T; Ross, DG; DeVito, MJ; et al. (2001) Effects of short-term in vivo exposure to polybrominated diphenyl
ethers on thyroid hormones and hepatic enzyme activities in weanling rats. Toxicol Sci 61:76-82.
72
-------
APPENDIX A: SUMMARY OF EXTERNAL PEER REVIEW AND PUBLIC
COMMENTS AND DISPOSITION
The "Toxicological Review" for BDE-209 has undergone a formal external peer review
performed by scientists in accordance with EPA guidance on peer review (U.S. EPA, 2006a,
2000a). The external peer reviewers were tasked with providing written answers to general
questions on the overall assessment and on chemical-specific questions in areas of scientific
controversy or uncertainty. The external peer review for BDE-209 was conducted in concert
with the external peer review of other PBDE congeners (i.e., BDE-47, BDE-99, and BDE-153),
and some external peer review charge questions were specific to congeners other than BDE-209.
External peer reviewer comments on all of the PBDEs and the Agency response are included
below for completeness. A summary of significant comments made by the external reviewers
and EPA's responses to these comments arranged by charge question follow. In many cases the
comments of the individual reviewers have been synthesized and paraphrased in development of
Appendix A. Synthesis of comments from individual peer reviewers resulted in summaries that
combine similar statements from peer reviewers that were mentioned in conjunction with more
than one charge question. In such cases, the comment and its response have been placed under
the most relevant charge question. Some of the peer review comments were not directly related
to charge questions. Those comments are categorized as miscellaneous and placed after those
related to the charge questions. EPA also received scientific comments from the public. These
comments and EPA's responses are included in a separate section of this appendix.
The peer review of the "Toxicological Review" for BDE-209 was coupled with the
review of the documents for BDE- 47, -99, and -153. Accordingly, most of the charge questions
address all four congeners. The responses to the charge questions in this appendix apply
primarily to comments related to BDE-209. The charge to the external peer reviewers and final
external peer review report (February 2007) pertaining to the toxicological reviews of the four
polybrominated diphenyl ether congeners are available at
http://cfpub.epa.gov/ncea/cfm/recordisplay.cfm?deid=161970. The public comments received
can be found at http://www.regulations.gov/fdmspublic/component/main under the Docket EPA-
HQ-ORD-2006-0838.
EXTERNAL PEER REVIEWER COMMENTS
The reviewers made several editorial suggestions to clarify specific portions of the text. These
changes were incorporated in the document as appropriate and are not discussed further.
A-l
-------
A. General Comments
Charge Question 1. Are you aware of other published peer-reviewed toxicological studies not
included in these toxicological reviews that could be of relevance to the health assessment of
BDE-47, -99, -153, or -209?
Comment 1: Three reviewers stated that they were unaware of any other relevant studies that
would contribute to the BDE-209 IRIS assessment. One reviewer identified potentially relevant
additional literature:
Jones-Ortazo, HA; etal. (2005) Environ. Sci. Technol. 39:5121-5130
Wilford, BH; et al. (2005) Environ. Sci. Technol. 39:7027-7035
Schecter, A; et al. (2005) J. Toxicol. Environ. Health Part A 68:501-513
Kites, RA; et al. (2004) Environ. Sci. Technol. 38:4945-4949
Schecter, A; etal. (2006) Environ. Health Perspect. 114:1515-1520
Fischer, D; et al. (2006) Environ. Health Perspect. 114:1581-1584
Bradman, A; et al. (2007) Environ. Health Perspect. 115:71-74
Kodavanti, PRS; Derr-Yellin, EC. (2002) Toxicol. Sci. 68:451-457
Kodavanti, PRS; et al. (2005) Toxicol. Sci. 88:181-192
Reistad, T; Mariussen, E. (2005) Toxicol. Sci. 87:57-65
Reistad, T; et al. (2006) Arch. Toxicol. 80:785-796
Response: The Agency reviewed and evaluated the studies recommended by the reviewer and
has included the relevant studies for BDE-209. Fischer et al. (2006), Bradman et al. (2007), and
Kodavanti et al. (2005) were found to be relevant and were added to the document. The
remaining studies suggested by the reviewer fell outside the scope of the IRIS assessment (i.e.,
exposure data, commercial mixtures). Additionally, a new literature search was conducted to
ensure that recently published relevant studies are included in the IRIS assessment. Four studies
were added to the "Toxicological Review" for BDE-209 as the result of the literature search
(Huwe and Smith, 2007; Pacyniak et al., 2007; Rice et al., 2007; Thuresson et al., 2006). A
description of these studies can be found in sections 3 and 4.
B. Oral Reference Dose (RfD) Values
Charge Question 2. Have the rationale and justification for deriving RfDs on the basis of the
neurobehavioral toxicity studies been transparently and objectively described in the draft
A-2
-------
toxicological reviews ofBDE-47, -99, -153, and-209? Are there additional studies that should
be considered for deriving the RfDsfor any of the four PBDE congeners?
Comment 1: Three reviewers stated that the rationale for deriving the RfD based on the
neurobehavioral toxicity studies was clearly and transparently described. Two reviewers stated
that the neurobehavioral effects are the only toxic effects that have been observed consistently in
PBDE rodent studies. One of these reviewers noted that, although the rat and mouse bioassays
by NTP (1986) provide suitable data, the neurobehavioral studies appeared to provide the most
appropriate dose-response data on which to base the health assessment. None of the reviewers
suggested additional studies that should be considered for deriving the RfD.
Response: No response needed.
Charge Question 3. Do you agree or disagree with EPA basing the health assessment ofBDE-
47, -99, -153, and-209 to a large extent on the Eriksson/Viberg neurobehavioral studies?
Comment 1: One reviewer supported the use of the Eriksson/Viberg neurobehavioral study as
the basis for the derivation of the RfD, given the limited body of toxicological information
available. Two reviewers noted that the studies are limited by the fact that they originated from
the same laboratory. One reviewer was concerned that the experimental design of the principal
study selected more than one pup per litter, ignoring the "litter" effect. Treating littermates as
independent experimental units could confuse dose effects with litter effects. Another reviewer
was concerned with the specificity of the neurobehavioral data for developmental neurotoxicity
and suggested that independent confirmation of the endpoints is essential. One peer reviewer
identified the use of a single sex (male mice) as a limitation of the critical study that had not
been identified in the "Toxicological Review" discussion of study limitations. One of these
reviewers stated that these limitations do not hinder the derivation of the RfD for BDE-209 but
make the confidence low. Another reviewer noted that the neurobehavioral findings of this
laboratory have been corroborated in a study examining BDE-99 (Kuriyama et al., 2005). None
of the reviewers stated that the studies could not be used as the basis for the derivation of the
RfD.
Response: The "Toxicological Review" contains a detailed summary of the concerns with the
study design and methods utilized in the principal study (see section 5.1.1). A discussion of the
use of only male mice in the study by Viberg et al. (2003a) has been added to the discussion in
section 5.1.1 of the "Toxicological Review." Additionally, the neurobehavioral effects reported
in Viberg et al. (2003a) are supported by a study in male and female mice that was published by
A-3
-------
Rice et al. (2007) after the peer review, which used an expanded array of neurodevelopmental
endpoints and dosing for 14 days. The neurodevelopmental findings from Viberg et al. (2003a)
are also supported by an expanding body of literature for the BDE-47, -99 and -153 congeners
(Viberg et al., 2007, 2005, 2004a, b, 2003b, 2002; Kuriyama et al., 2005; Eriksson et al., 2002,
2001; Branch! et al., 2002) that details changes in motor and cognitive activity in rodents
following administration of single or repeated perinatal doses of PBDEs. Some of the concerns
associated with the methodology of the Eriksson/Viberg neurobehavioral studies are alleviated
by other studies (Rice et al., 2007; Kuriyama et al., 2005; Branch! et al., 2002) using more
traditional methodologies that have generated toxic effects similar to those reported by Viberg et
al. (2003a). Results reported by Viberg et al. (2003a) for BDE-209 are similar to results reported
by Eriksson et al. (2001) for BDE-47, Viberg et al. (2004b) and Kuriyama et al. (2005) for BDE-
99, and Viberg et al. (2003b) for BDE-153. Altogether, these studies support the findings of
Viberg et al. (2003a) that exposure to BDE-209 in early developmental stages can result in
lasting changes in the neurobehavioral activity of mice.
Charge Question 4. Are the Eriksson et al. (2001) (BDE-47), Viberg et al. (2004b) (BDE-99),
Viberg et al. (2003b) (BDE-153), and Viberg et al. (2003a) (BDE-209) studies appropriate for
determining the point of departure? Have the strengths and weaknesses of the Viberg and
Eriksson studies been appropriately characterized and considered?
Comment 1: All four reviewers believed that the Viberg et al. (2003a) BDE-209 study was
appropriate for determining the point of departure. One reviewer felt that these data were
appropriate as long as the document emphasizes that the neurochemical data also show
alterations in normal developmental patterns. Another reviewer noted that both Viberg et al.
(2003a) and NTP (1986) provided data appropriate for determining a point of departure, with
Viberg et al. (2003a) providing the lowest point of departure. None of the reviewers suggested
an alternative study for determining the point of departure. Two reviewers explicitly stated that
the strengths and weaknesses were identified and clearly presented.
Response: No response needed.
Charge Question 5. Have the most appropriate critical effect and point of departure been
selected? And has the rationale for the point of departure been transparently and objectively
described?
Comment 1: All four reviewers agreed with the selection of the neurobehavioral effects as
critical effects and that these effects were appropriate for identifying a point of departure. One
A-4
-------
of the reviewers felt that the neurochemical data also provided critical information and should be
presented centrally rather than as supporting data. One reviewer stated that there was no
correlation between PND of exposure and the concentration of the chemical in the brain. One
reviewer added that decreased habituation might be as appropriate as or more appropriate than
the habituation ratio as an indicator of toxicity, while another believed that the actual behavioral
data, rather than the habituation ratio, should have been presented in the document. It was not
clear for another reviewer why the actual data could not be recovered from the study authors to
allow for dose-response modeling and BMD estimation, given that the studies were published
fairly recently (2003). This reviewer recommended that the Agency attempt to recover the
neurobehavioral toxicity data from the study authors.
Response: The evidence of neurochemical interactions and the potential relationship with the
neurobehavioral effects are highlighted in the mode-of-action section of the document (section
4.5.3). The document presents the hypothesis proposed by the Eriksson/Viberg group in which
the observed effects on locomotion and habituation are related to impaired development of the
cholinergic system during the postnatal brain growth spurt; however, data are unavailable to
determine the relevance of the neurochemical effects or to establish a mode of action. This
conclusion has been added to section 4.5.3. In the case of BDE-209, the levels of radiolabel in
the brain were higher following administration of equivalent doses on PND 3 or 10 compared to
PND 19 (Viberg et al., 2003a). There are no measurements of levels of BDE-209 or the
congeners in the brain or other tissues at the time of neurobehavioral testing at 2 or 4 months to
show if any differences exist in the brain or other tissues at those time points.
The actual motor activity components (locomotion, rearing, and total activity) that gave
rise to the habituation ratios were reported in graphical form only and could not be reasonably
estimated as presented, and the Agency's attempts to obtain the raw data were unsuccessful.
Thus, the habituation ratios, rather than the actual habituation data, served as the basis for
determining the point of departure.
Comment 2: Two reviewers thought that the rationale for the point of departure had been
transparently and objectively described. Another reviewer felt the document provided clear
rationalization for the selection of the point of departure. None of the reviewers stated that the
rationale for the point of departure was not appropriately described.
Response: No response needed.
Comment 3: One reviewer noted that a BMDL would be better than a NOAEL for deriving an
RfD.
A-5
-------
Response: The Agency recognizes that a NOAEL can be limiting since it is highly dependent on
the doses selected and the sample size and does not account for the dose-response curve.
Alternatively, a BMDL is more independent of study design, takes into account the dose-
response curve, and, therefore, is preferable to a NOAEL as a point of departure. However, the
data in the principal study for BDE-209 only allows for the identification of a NOAEL. The
reporting is insufficient to provide the necessary information to perform BMD modeling and the
Agency's attempts to obtain the raw data were unsuccessful.
Charge Question 6. Have the rationale and justification for each UF selected in the draft
toxicological reviews of BDE-47, -99, -153, and-209 been transparently described? If the
selected UFs are not appropriate, what alternative UFs would you suggest and what are the
scientific rationales for those suggested? Does the database support the determinations of the
RfDs for BDE-47, -99, -153, and -209?
Note: The peer reviewers provided fairly extensive comments about the individual components
of the combined UF. For that reason the following reviewer comments and EPA responses have
been grouped by the area of uncertainty to which they apply.
Comment 1: Two reviewers agreed that the document described the rationale and justification
for each UF and another reviewer noted that the selection of the UFs was described in detail.
The fourth reviewer felt that the document did not provide much explanation or justification for
applying the default intraspecies UFH for BDE-209.
Response: There is little information available on the effects of PBDEs in humans and in the
absence of data there is no scientific rationale for moving away from the default value for the
intraspecies UFH. Additional explanation for the intraspecies UFH was added to section 5.1.3.
Comment 2: One reviewer suggested decreasing the interspecies UFA, considering the relatively
specific and sensitive nature of the neurobehavioral and neurochemical measures compared with
conventional endpoints. However, another reviewer felt that the 10-fold UFA was justifiable
based on the lack of data on the mode of action in animals and humans.
Response: The 10-fold UFA for interspecies uncertainty is retained based on the lack of mode-
of-action, pharmacokinetic, and human data that would sufficiently illustrate the similarities and
differences for the effects of the PBDEs in animals and humans. Additional explanation for
applying the default interspecies UFA was added to section 5.1.3.
A-6
-------
Comment 3: Two reviewers suggested lowering the intrahuman UFH. One of these reviewers
felt the relatively specific and sensitive nature of the neurobehavioral and neurochemical
measures compared with conventional endpoints warranted a decrease in the UFH. The other
reviewer recommended decreasing the 10-fold UFH to 3-fold based on the sensitivity of the test
species population (neonates).
Response: The 10-fold UFH for intraspecies uncertainty is retained based on the lack of
information concerning the pharmacokinetics and mode of action of the BDE-209 in humans. In
the absence of human data, the effects in potentially susceptible populations exposed to BDE-
209 cannot be determined. Additional explanation for applying the default intraspecies UFH was
added to section 5.1.3.
Comment 4: One reviewer disagreed with the treatment of a single-dose experiment as
equivalent to a subchronic exposure when applying a UF to account for differences in exposure
duration. This reviewer stated that the principal study needs to be treated as a single-dose study
and not a subchronic study. The reviewer also felt that the threefold UFS was inappropriate and
suggested raising the UFS from 3 to 10 to consider the extent to which the mother's pre-
pregnancy accumulated body burden would influence the developmental outcome, especially
since these data are unavailable. One reviewer agreed with the application of a threefold UFS,
recognizing that for the observed neurobehavioral effects the timing of exposure is more critical
than the duration of exposure. This reviewer regards the UFS as accounting for uncertainty from
lack of prenatal exposure rather than uncertainty regarding potential effects of chronic exposure.
One reviewer suggested the threefold UFS may not be necessary, considering that exposure
during a window of susceptibility indicates that chronic exposure may not necessarily result in
greater adverse effects.
Response: For BDE-209, the principal study identified endpoints that, for the most part, reflect
specific aspects of developmental physiology. The hypothesized window of susceptibility,
proposed by the study authors, is based on the observation that the developmental neurotoxic
effects observed following exposure to BDE-209 on PND 3 will not occur once the
toxicokinetics of intestinal uptake and excretion have matured and the animal brain is
developmentally less active (outside the window of susceptibility). The Eriksson/Viberg group
has suggested that the period of maximum vulnerability for the developing cholinergic system
that coincides with the most pronounced neurodevelopmental effects from BDE-99 exposure is
from PNDs 10-14. However, for BDE-209 the developmental neurotoxic effects were noted
following exposure on PND 3 rather than on PNDs 10 or 19. It is likely that two factors
A-7
-------
contributed to this difference in peak vulnerability. First, absorption from the gastrointestinal
tract decreases as the neonatal mice age during their first few weeks of life, thus less of the
BDE-209 reaches the liver for distribution on PNDs 10 and 19 than on PND 3. Secondly,
distribution to the brain is slow. The amount of radiolabel in the brain is 1.4- and 2.8-fold
greater 7 days after dosing compared with 24 hours after dosing on PNDs 3 and 10, respectively.
Even less reaches the brain when the BDE-209 is administered on day 19. The UFS was viewed
as a dosing duration adjustment rather than simply a comparison of the effects of a subchronic to
a chronic exposure. A threefold UFS was applied because the critical study dosed the animals
only once within the hypothesized critical window, not because the chronic exposures would
have exacerbated the impact on habituation.
In response to the comment about possible effects as the result of a maternal pre-
pregnancy body burden, the Agency notes that, although the principal study did not include
prenatal exposure, the maternal uptake and retention of BDE-209 during the prenatal period
would be lower than that of the pups during the postnatal period of vulnerability because of the
differences in toxicokinetics for mature versus neonatal animals. Support for the UFS is
provided by the Hardy et al. (2002) study, where BDE-209 treatment did not produce any
evidence of maternal or developmental toxicity up to the highest dose tested (1,000 mg/kg-day)
despite exposure from gestation days 0 through 19.
Comment 5: Two reviewers recommended raising the onefold database UFD to threefold. One
of these reviewers based this decision on the use of a NOAEL rather than a BMDL as the point
of departure. This reviewer felt that the absence of definitive data for dose-response modeling
for the neurobehavioral effects reflects an inadequacy in the database, although if the endpoints
could be modeled then a onefold UFD is acceptable. The other reviewer suggested raising the
UFD to account for the lack of other developmental neurotoxicity studies.
Response: EPA's practice is to apply a database UFD, generally ranging from 1-10, in the
health assessment to account for the potential for deriving an underprotective RfD as a result of
an incomplete characterization of a chemical's toxicity because of missing studies. In deciding
to apply this factor to account for deficiencies in the available data set and in identifying its
magnitude, EPA considers both the data lacking and the data available for particular organ
systems as well as life stages. The database for BDE-209 contains subchronic and chronic
toxicity studies as well as a traditional developmental study. Although the database lacks a two-
generation reproductive study, the absence of effects in the available longer-term and
developmental studies indicates that at least some aspects of reproductive organ toxicity or
function are not affected at doses much higher than those that resulted in the neurological effects
observed in neonates in the principal study. Therefore, the Agency retains the onefold database
A-8
-------
UFD. Calculating a BMDL rather than using a NOAEL for the point of departure would not
impact the use of a database UF.
Comment 6: Two reviewers believed the database supports the determination of the RfD but
stated that the overall confidence in the RfD assessment is low. Another reviewer believed the
database is very poor and suggested that the RfDs be acknowledged as temporary while waiting
for additional studies that increase confidence.
Response: The statement that the overall confidence in the RfD is low is included in the
"Toxicological Review" in section 6.2. The Agency does not develop temporary RfDs for IRIS
assessments. However, the availability of new information is one of the factors considered in
selecting a chemical for reassessment.
C. Body Burden Approach
Charge Question 7. Are there adequate data for considering body burden as an alternative
dose metric to administered doses in any of the RfD derivations?
Comment 1: All four reviewers agreed that the data were inadequate to consider body burden as
an alternative dose metric for the derivation of the RfD. Two of the reviewers stated that body
burden is a possible alternative but the data are too limited.
Response: EPA examined the data on BDE-209 to determine if a body burden approach could
be used for this congener during the development of the "Toxicological Review." It was
determined that existing half-life, exposure, metabolite and mode-of-action data could not
support a body burden calculation for this congener.
Charge Question 8. Do you agree with the rationale described in the "Toxicological Review "
of BDE-209 that the data on the window of susceptibility of the cholinergic receptors to BDE-
209 tend to minimize body burden concerns?
Comment 1: Three reviewers stated that the question was unclear. One reviewer accepted the
concept as a basis for the experimental design, given the available information. A second
reviewer stated that there was no direct evidence that BDE-209 directly affects cholinergic
receptors and suggested that the mechanism of the interaction must be complex and indirect. A
third reviewer stated that although there are no definitive data on mode of action, this hypothesis
is plausible. This reviewer acknowledges that the data on the window of susceptibility of the
A-9
-------
cholinergic receptors to BDE-209 are suggestive but believes there are too many other
possibilities for mode of action for this rationale to minimize body burden concerns.
Response: Available mode-of-action data that describe the developmental neurotoxicity of
BDE-209 are limited. The Eriksson/Viberg group, the principal study authors, have
hypothesized that the observed effects on locomotion and habituation are related to impaired
development of the cholinergic system during the postnatal brain growth spurt period based on
studies of BDE-99 (Viberg et al., 2005, 2004a) and supported by studies with BDE-153 (Viberg
et al., 2003b) and BDE-209 (Viberg et al, 2007, 2003a). They have further hypothesized that the
sensitivity of the cholinergic system occurs in the vicinity of PND 10 and have tested this
hypothesis by varying the time of dosing and observing differences in the habituation effect for
BDE-99 and BDE-209 (Viberg et al., 2007; Eriksson et al., 2002). A difference in the impact of
the time of dosing was observed between BDE-99 and BDE 209 and can be explained based on
toxicokinetic factors (see section 4.5.3). The resulting deficit in cholinergic receptors persisted
across the duration of testing and could cause an abnormal response to exposure to cholinergic
stimulants in adulthood. The following statement has been added to the mode-of-action
summary (section 4.5.3): "While evidence exists that demonstrates BDE-209, as well as other
PBDEs, interacts at the neurological level, data are inadequate to determine the mode of action
for BDE-209."
D. Carcinogenicity Assessment
Charge Question 9. Is the weight of evidence for the carcinogenicity of BDE-209 in the draft
"Toxicological Review " appropriately described? Are there additional studies that should be
included?
Comment 1: Three reviewers commented that the weight of evidence for carcinogenicity of
BDE-209 was appropriately described in the "Toxicological Review." One reviewer stated that
the carcinogenicity study was completely and methodically described. This reviewer noted that
the carcinogenicity study is over 25 years old and utilized very high doses that led to some toxic
effects and animal deaths. None of the reviewers were aware of additional studies that should be
included in the cancer assessment for BDE-209.
Response: No response needed.
Charge Question 10. Do the data support estimation of a cancer slope factor for BDE-209? If
yes, is the rationale for the quantitative analysis objectively and transparently described,
A-10
-------
considering the uncertainty in the data and the suggestive nature of the weight of evidence?
Have the rationale and justification for the use of the linear low-dose extrapolation been
objectively and transparently presented?
Comment 1: One reviewer stated that the data appear to support estimation of a cancer slope
factor, one reviewer stated that the data supports estimation, one reviewer did not specifically
answer this question but, as mentioned below, did question the appropriateness of the study EPA
used, and the fourth reviewer did not answer the question about the support for estimation of a
cancer slope factor. One reviewer felt that the data supported the weight-of-evidence
classification as "suggestive of the carcinogenic potential." One reviewer questioned the
appropriateness of the NTP (1986) study for developing the cancer slope factor based on the
utilization of very high doses and potentially limited absorption of BDE-209 and suggested that a
new bioassay be conducted. This reviewer recognized the public comments on the
carcinogenesis data, particularly the statements regarding the modest dose-response and the
change, since the completion of the study, in criteria for classifying liver lesions in rats. This
reviewer was less concerned with the scoring of neoplastic nodules as tumors because the
nodules identified in chronic studies with exposure to a single compound are less likely to
regress as compared with an initiation/promotion study. Regarding the change in classification
criteria, another reviewer noted that the data supported the estimation of a cancer slope factor for
BDE-209, with the assumption that the change in classification of the neoplastic nodules would
not appreciably change the dose response. Two reviewers explicitly stated that the rationale for
the quantitative analysis is transparently described. Two reviewers stated that the justification
for the use of the linear low-dose extrapolation is objectively presented in the BDE-209
assessment. One reviewer noted that the document did not include a rationale or justification,
other than a reference to EPA guideline documents (U.S. EPA, 2005a, b), for the use of linear
low-dose extrapolation. None of the reviewers disagreed with the estimation of the cancer slope
factor or the use of linear low-dose extrapolation.
Response: The Agency is aware of the deliberations concerning the significance of neoplastic
nodules in interpreting cancer risk. In 1986, a significant change was made regarding the NTP
nomenclature for hepatoproliferative lesions of rats: lesions previously combined under the
diagnosis of "neoplastic nodules" were replaced with three terms: "hepatocellular hyperplasia,"
"foci of cellular alteration," and "hepatocellular adenoma." The term "hyperplasia" was
reserved for proliferative lesions that were perceived to be secondary, nonneoplastic responses to
degenerative changes in the liver. Foci of cellular alteration, hepatocellular adenoma, and
hepatocellular carcinoma were considered to represent a spectrum of changes that are part of the
carcinogenic process (Maronpot et al., 1986). It is recognized that not all neoplastic nodules
A-ll
-------
have the potential to progress to malignancy: some neoplastic nodules may regress following
removal of a chemical stimulus, others may only be hyperplastic lesions, and still others may
progress to hepatocellular carcinoma. Therefore, the exact contribution of neoplastic nodules to
the overall incidence of carcinoma in the rat liver is not known with any certainty. However, an
increase in neoplastic nodules in treated rats provides some indication that the liver may be at
increased risk of cancer formation, and inclusion of the incidence of neoplastic nodules in the
assessment of cancer risk is considered to be a conservative approach (U.S. EPA, 1986c). This
change in nomenclature has been noted in the "Toxicological Review" in sections 4.2, 4.6.2, and
5.3.3 and in Table 4-2.
EPA acknowledges that toxicokinetic limitations on solubility and absorption may make
external dose a poor indicator of systemic dose. However, there are no PBPK models for BDE-
209 that might be used to estimate internal dose. Thus, the only parameter available for
modeling dose-response is the external dose.
The justification for using the linear low-dose extrapolation for deriving a cancer slope
factor for BDE-209 is based on the absence of data available to determine the mode of action for
BDE-209. This rationale is provided in section 5.3.4.
Charge Question 11. Are there alternative modeling approaches that should have been
considered instead of or in addition to the linear low-dose extrapolation approach?
Comment 1: None of the reviewers suggested that an alternative modeling approach should have
been considered. One reviewer stated that all possible modeling approaches were explained and
the model that best fit the experimental data was selected. Another reviewer noted that
consideration of other modeling approaches is not necessary, considering the dose-response data
are linear, the linear model adequately fits the data, and the data are inadequate to justify an
alternative approach.
Response: No response needed.
Miscellaneous Comments
Comment 1: Three reviewers felt that the assessment would benefit from the combination of the
individual documents for the four congeners into one comprehensive document to compare and
cohesively present the similarities and differences among the congeners.
A-12
-------
Response: The Agency has recently completed IRIS assessments for four individual PBDE
congeners: BDE-47, -99, -153, and -209 (see Foreword). These congeners were selected based
on frequent detection in human tissues and the environment, availability of animal toxicological
studies suitable for human health assessment, and their common occurrence in commercial
PBDE mixtures. Although there is some repetition in the four documents, the available database
is sufficiently different from one congener to another to support the separation of the four IRIS
assessments. However, in response to the comments from the peer reviewers, the Agency has
increased the text that compares the data on BDE-209 to those of the other congeners evaluated
using comparable methodological approaches.
Comment 2: One reviewer noted that the document failed to cite the purities of the radioactive
chemicals in most of the studies, the position of the label, location of radioactivity in the brain,
and the specific activities of the 14C compounds. Another reviewer questioned the reliance upon
the 14C data and the intermixing with direct chemical measures. The reviewer felt that the
conclusions drawn were challenging.
Response: The requested data were added to the descriptions of the pertinent studies (in section
3) when they were provided by the authors of the paper. Frequently, the position of the
radiolabel was not specified. In a few cases the radiolabel was described as "uniform,"
suggesting that all carbons carried the radiolabel. If the authors of the paper used the term
"uniform," it has been added to the discussion of the study. No change was made if the authors
of the paper did not comment on the position of the radiolabel.
Comment 3: One reviewer was concerned that the doses and concentrations of the compound
and the metabolites in biological tissues were presented in differing units (i.e., umol, ug, percent
of dose).
Response: Doses and concentrations are reported as given by the authors. If a dose was given in
molar or mole units per unit body weight, the doses have been provided parenthetically as mg/kg
body weight values. Otherwise the units are those provided in the published papers.
Comment 4: One reviewer suggested including proposed metabolic pathways to strengthen the
fact that the lower brominated congeners could be obtained from the higher brominated ones.
Response: A diagram of a proposed metabolic pathway has been included as Figure 3-1 in
section 3.3. The diagram is based on the metabolites that have been identified in several studies.
The metabolic pathway is described as "proposed" and uncertainties are indicated. The
A-13
-------
individual publications that provided data on metabolites are cited in the Figure 3-1. In cases
where the data from one study are discussed in more than one section of the "lexicological
Review," a reference to the location of prior mention of the study is provided.
Comment 5: One reviewer acknowledged that developmental neurotoxicity is consistently
observed following exposure to the PBDEs despite very different patterns of metabolism,
distribution, and persistence within the body. This reviewer recommended rationalizing the
relative potency of the PBDEs, considering the differences in the extent of metabolism.
Response: Information is currently insufficient to identify the relative potency of the four
congeners.
Comment 6: One reviewer suggested that the Agency provide conclusions on the extent of
metabolism and the presence of metabolites in excreta for the PBDEs or provide a statement if
conclusions cannot be drawn. One reviewer suggested the addition of a summary at the
beginning of the toxicokinetics section to reduce potential confusion.
Response: An overview to the toxicokinetics section and summary paragraphs have been added
to section 3.
Comment 7: One reviewer recommended improving the discussion of the possible
debromination of BDE-209 to lower brominated congeners and questioned if environmental
debromination should be mentioned.
Response: Discussion of the potential debromination of BDE-209 to lower-brominated
congeners can be found in sections 3.2.2 and 3.3, based on evidence observed in several studies
(Huwe and Smith, 2007; Morck et al., 2003; Sandholm et al., 2003; Zhou et al., 2001). Data for
environmental debromination falls outside the scope of the IRIS health assessment.
Comment 8: One reviewer recommended presenting the receptor site interaction information in
a summary table.
Response: Tables were added to the toxicological reviews for BDE-47, -99, and -153 because
there were data for multiple congeners for the tetra, penta, and hexa congeners. In the case of
BDE-209, there is only one PBDE congener and there was no need to have a table to summarize
the data.
A-14
-------
Comment 9: One reviewer felt that the evolution of exposures that are different in the U.S.
compared with other countries and the pattern of exposures are important issues and the studies
need to be presented to support or refute these observations.
Response: The Agency has provided information on exposure in the U.S. and other countries for
comparison purposes. While the Agency agrees that exposure analysis is a critical component of
risk assessment, a comprehensive presentation and analysis of exposure data are outside the
scope of the IRIS health assessment.
Comment 10: One reviewer stated that the large number of bromine atoms of the PBDEs can
impart electrophilic and lipophilic properties to the aromatic ring of the chemical and also noted
that oily vehicles (e.g., corn oil) were used in most of the in vitro and in vivo animal studies.
This reviewer was concerned the vehicle could significantly alter the distribution and tissue
uptake of the PBDEs between the oily vehicle and the biological system. These conditions could
lead to decreased absorption and distribution with subsequent alteration in metabolism and
excretion.
Response: The lipohilicity of the BDE-209 is acknowledged in the "Toxicological Review" as
part of section 3 on toxicokinetics. It will be necessary to determine if absorption occurs via the
chylomicrons along with the body lipids or via direct membrane transport before the full impact
of the vehicle on absorption, distribution, metabolism, and elimination can be determined. The
data are currently inadequate to determine the impact of the oily vehicle on the distribution and
uptake of BDE-209.
PUBLIC COMMENTS
The public commenters made several editorial suggestions to clarify specific portions of the text.
These changes were incorporated in the document as appropriate and are not discussed further.
Comment 1: One public commenter suggested that the Agency consider a body burden
approach.
Response: The Agency presented this issue to the peer reviewers in the form of a charge
question. In response to the charge question about use of a body burden approach for dose
evaluation, the peer reviewers agreed that, whereas the body burden approach might be
appropriate for some of the congeners given their lipophilicity and distribution to adipose tissue,
data to support such an approach are not presently available.
A-15
-------
Comment 2: Three public commenters from one group questioned the selection of Viberg et al.
(2003 a) as the principal study for the derivation of the RfD, and another public commenter
disagreed with the selection of Viberg et al. (2003a). Critiques of the study were submitted by
the three public commenters questioning the methods utilized by the principal study authors.
These public commenters were concerned that the purity of the radiolabeled BDE-209 in Viberg
et al. (2003a) was not reported and suggested that observed effects may have been induced by
impurities rather than the parent compound. Two of these public commenters felt that applying
the intraspecies UFH to account for sensitive populations when evidence for a sensitive
subpopulation does not exist is overly conservative. One of the public commenters noted the use
of inconsistent units of concentration within the document.
Response: The Agency has included a detailed summary of the concerns with the experimental
design and methods utilized in the principal study (see section 5.1.1). These issues were raised
during the external peer review of the BDE-209 IRIS assessment. The peer reviewers
acknowledged the limitations and concerns with the study; however, all of the peer reviewers
agreed that this study was appropriate for derivation of the RfD for BDE-209 and that its
limitations were transparently discussed in the "Toxicological Review." Additionally, the
neurobehavioral effects reported in Viberg et al. (2003a) are supported by other data for BDE-
209 (Rice et al., 2007) and by an expanding body of literature for the PBDEs (Viberg et al.,
2007, 2005, 2004a, b, 2003b, 2002; Kuriyama et al., 2005; Eriksson et al., 2002, 2001; Branch!
et al., 2002) that details changes in motor and cognitive activity in rodents following
administration of single or repeated perinatal doses of PBDEs. A summary of Rice et al. (2007)
has been added to the BDE-209 "Toxicological Review" in section 4.3.2.
In response to the radiolabel concerns, the purities and positions of the radiolabels, the
specific activities of the 14C compounds, and the locations of radioactivity in the brain were
added to the descriptions of the relevant studies (in section 3) when they were provided by the
authors of the paper. Frequently, the position of the radiolabel was not specified and/or the
radiolabel was inadequately described to allow for the detailed reporting. These data have been
added to the document where available.
The 10-fold UFH for intraspecies uncertainty is retained based on the lack of information
concerning the pharmacokinetics and mode of action of the PBDEs in humans. In the absence of
human data, the effects in potentially susceptible populations exposed to PBDEs cannot be
determined. Additional explanation for applying the default intraspecies UFH was added to
section 5.1.3.
In response to the statement regarding the inconsistent reporting of units of
concentration, the Agency notes that the doses and concentrations that are in the "Toxicological
A-16
-------
Review" are reported as given by the authors. If a dose was given in molar or mole units per unit
body weight, the doses have been provided parenthetically as mg/kg body weight values.
Otherwise the units are those provided in the published papers.
Comment 3: One public commenter suggested a revision of the absorption, distribution,
metabolism, and elimination sections (section 3) of the IRIS assessment for BDE-209.
Additionally, another public group identified weaknesses in several studies within section 3.
Response: The toxicokinetics section (section 3) has been revised and updated to reflect recently
published literature (Huwe and Smith et al., 2007; Pacyniak et al., 2007; Thuresson et al., 2006).
An introduction to the toxicokinetics information has been added to section 3. The studies
within this section have been reorganized to separate animal and human data in section 3.1.
Comment 4: One public commenter disagreed with the conclusion that the database for BDE-
209 provides "suggestive evidence of carcinogenic potential." This commenter recommended
adding a chronic study in rats administered BDE-209 (Kociba et al., 1975). Additionally, the
public commenter noted the change in nomenclature for hepatoproliferative liver lesions in rats
and the appropriateness of using the incidence of "neoplastic nodules" for quantitative cancer
risk assessment.
Response: The external peer reviewers were asked to evaluate the weight of evidence for
carcinogenic potential for BDE-209 and to determine if the database supported the estimation of
a cancer slope factor. Three of the peer reviewers explicitly stated that the database was
sufficient, and none of the reviewers disagreed with either the classification of "suggestive
evidence of carcinogenic potential" or the quantitative cancer assessment for BDE-209. Kociba
et al. (1975) administered a commercial grade BDE-209 that possesses a lower purity than the
analytical grade utilized in the majority of the studies described in the IRIS health assessment.
This study was not included for comparison since it falls outside the scope of the IRIS health
assessment for the pure BDE-209 congener. Please see the response to the external peer review
Comment 1, under Charge Question 10, regarding the change in criteria for the classification of
hepatoproliferative liver lesions.
A-17
-------
APPENDIX B: BENCHMARK DOSE ANALYSIS OF NONCANCER ENDPOINTS
The following data sets from the NTP (1986) 2-year rat and mouse studies were selected
for BMD modeling: thrombosis in the liver, liver degeneration, fibrosis in the spleen, and
lymphoid hyperplasia in male rats, as well as centrilobular hypertrophy in the liver and follicular
cell hyperplasia in the thyroid of male mice.
BMD modeling was conducted by using EPA's BMDS, version 1.3.2. All the BMD
analyses were conducted with the BMR set to a 10% response (U.S. EPA, 2000b). For each data
set, all the available dichotomous models, including gamma, logistic, log-logistic, multistage,
probit, log-probit, quantal-linear, quantal-quadratic, and Weibull, available in the BMDS were
used. BMD model fit was evaluated based on the goodness-of-fit/> value (indicating global
model fit), local chi-square residual (indicating the model fit at the data point close to the preset
BMR), as well as AIC (indicating model fit when controlling for number of model parameters).
Based on the model output, the model(s) providing best model fit to the data were selected to
estimate BMD10 and BMDL10. In some cases, the BMDS gives exactly the same results for
several different models. This occurs when model parameters are fixed at a boundary, yielding
reduced models that are identical expressions for the probability of response. For example,
when the power parameter in the Weibull or gamma model is fixed at 1 by BMDS or when the
degree of the multistage model is set to 1 by the user, these models reduce to the quantal linear
model. When this occurred, all the reduced models providing the same BMD results were
considered as one model.
Liver Thrombosis in Male Rats
The data (1/50, 0/50, and 9/49 for control, low dose, and high dose, respectively) on
thrombosis in the liver of male rats treated with decaBDE (NTP, 1986) were modeled with EPA
BMDS. The reported oral doses were 0, 1,120, and 2,240 mg/kg-day, respectively. The data
were modeled with all the dichotomous models available in the BMDS, and modeling results are
summarized in Table B-l.
Based on these results, the models that did not demonstrate significant lack of fit for the
data were obtained from three models (shown in bold in the table): gamma, log-logistic, and
Weibull. These models provided relatively high goodness-of-fit/> values and low AIC values.
Based on these considerations, an average value of the BMDs estimated from these three models
is used. For thrombosis in the livers of male rats, the average BMD10 is 2,125 mg/kg-day and the
average BMDL10 is 1,738 mg/kg-day.
B-l
-------
Table B-l. Summary of BMD modeling results for thrombosis in the liver of
male rats
Model
Gamma
Log-logistic
Logistic
Multistage
Log-probit
Probit
Quantal-linear
Quantal-quadratic
Weibull
Goodness-of-fit
p value
0.31
0.32
0.04
0.07
N/Aa
0.04
0.02
0.07
0.31
AIC
61.9754
61.9384
65.0449
66.1380
63.9383
65.8039
69.4028
66.1380
61.9384
BMD10
2,047
2,161
1,909
1,898
2,130
1,869
2,030
1,898
2,166
BMDL10
1,717
1,741
1,597
1,449.97
1,691.4
1,513.96
1,162.71
1,449.97
1,757
3N/A = not applicable.
Source: NTP (1986).
Liver Degeneration in Male Rats
The data (13/50, 19/50, and 22/49 for control, low dose, and high dose, respectively) on
degeneration in the liver of male rats treated with decaBDE (NTP, 1986) were modeled with
EPA BMDS. The reported oral doses were 0, 1,120, and 2,240 mg/kg-day, respectively. The
data were modeled with all the dichotomous models available in the BMDS, and modeling
results are summarized in Table B-2.
Table B-2. Summary of BMD modeling results for degeneration in the liver
of male rats
Model
Gamma
Log-logistic
Logistic
Multistage
Log-probit
Probit
Quantal-linear
Quantal-quadratic
Weibull
Goodness-of-fit
p value
0.83
0.89
0.71
0.83
0.51
0.72
0.83
0.42
0.83
AIC
195.178
195.148
195.269
195.178
195.555
195.259
195.178
195.779
195.178
BMD10
779
707
929
779
1,161
914
779
1,363
779
BMDL10
422
344
600
422
718
584
422
962
422
Source: NTP (1986).
Based on the goodness-of-fit/> value, none of the models demonstrated lack of fit. The
best model fit for the data was obtained from five models (shown in bold in the table): gamma,
log-logistic, multistage, quantal-linear, and Weibull. These models provided high goodness-of-
B-2
-------
fitp values and low AIC values. However, among the five models, gamma, multistage, quantal-
linear, and Weibull were reduced to a single identical model. Based on these considerations, an
average value of the BMDs estimated from the log-logistic model and the common reduced
model are used. For degeneration in the livers of male rats, the average BMD10 is 765 mg/kg-
day and the average BMDL10 is 406 mg/kg-day.
B-3
-------
Liver Degeneration in Male Rats (NTP, 1986)
10% BMR
Log-Logistic Model $Revision: 2.1 $ $Date: 2000/02/26 03:38:20
$
Input Data File: C:\BMDS\DATA\RATS.(d)
Gnuplot Plotting File: C:\BMDS\DATA\RATS.plt
Thu Nov 25 12:11:49 2004
HMDS MODEL RUN
The form of the probability function is:
P[response] = background+(1-background)/[1+EXP(-intercept-slope*Log(dose)]
Dependent variable = respl
Independent variable = Dose
Slope parameter is restricted as slope >= 1
Total number of observations = 3
Total number of records with missing values = 0
Maximum number of iterations = 250
Relative Function Convergence has been set to: le-008
Parameter Convergence has been set to: le-008
User has chosen the log transformed model
Default Initial Parameter Values
background = 0.26
intercept = -8.66331
slope = 1
Asymptotic Correlation Matrix of Parameter Estimates
( *** The model parameter(s) -slope have been estimated at a boundary point,
or have been specified by the user, and do not appear in the correlation
matrix )
background intercept
background 1 -0.69
intercept -0.69 1
B-4
-------
Parameter Estimates
Variable
background
intercept
slope
Estimate
0.262212
-8.75859
1
Std. Err.
0.0603288
0.569357
NA
NA - Indicates that this parameter has hit a bound implied by some inequality
constraint and thus has no standard error.
Analysis of Deviance Table
Model
Full model
Fitted model
Reduced model
AIC:
Log(likelihoo
d)
-95.5647
-95.5739
-97.5646
Deviance
0.0184518
3.99968
Test DF
195.148
Goodness of Fit
Dose Est._Prob
0.0000 0.2622
1120.0000 0.3726
2240.0000 0.4543
Chi-square =
Expected
13.111
18.630
22.259
DF = 1
Scaled
Observed Size
13 50
19 50
22 49
P-value = 0.8919
Benchmark Dose Computation
Specified effect = 0.1
Risk Type = Extra risk
Confidence level = 0.95
P-value
0.8919
0.1354
Residual
-0.03556
0.1081
-0.07432
BMD
BMDL
707.234
344.083
Log-Logistic Model with 0.95 Confidence Level
Log-Logistic
MD Lower Bound
0.5
0.4
0.3
0.2
BMDL
BMD
500
1000
dose
1500
2000
12:11 11/25 2004
B-5
-------
Splenic Fibrosis in Male Rats
The data (5/49, 8/50, and 13/49 for control, low dose, and high dose, respectively) on
fibrosis in the spleen of male rats treated with decaBDE (NTP, 1986) were modeled with EPA
BMDS. The reported oral doses were 0, 1,120, and 2,240 mg/kg-day, respectively. The data
were modeled with all the dichotomous models available in the BMDS, and modeling results are
summarized in Table B-3.
Table B-3. Summary of BMD modeling results for fibrosis in the spleen of
male rats
Model
Gamma
Log-logistic
Logistic
Multistage
Log-probit
Probit
Quantal-linear
Quantal-quadratic
Weibull
Goodness-of-fit
p value
N/Aa
N/A
0.92
0.69
0.87
0.88
0.69
0.80
N/A
AIC
139.172
139.172
137.183
137.336
137.197
137.195
137.336
137.233
139.172
BMDio
1,464
1,463
1,427
1,236
1,519
1,393
1,236
1,612
1,470
BMDLio
700
644
1,018
690
1,017
968
690
1,176
700
3N/A = not applicable.
Source: NTP (1986).
Based on these results, the logistic, multistage, log-probit, probit, quantal-linear, and
quantal -quadratic models did not show significant lack of fit for the data. The lowest AIC values
were obtained with three models: logistic, log-probit, and probit (shown in bold in the table).
Based on these considerations, an average value of the BMDs estimated from these three models
is used. For fibrosis in the spleen of male rats, the average BMD10 is 1,446 mg/kg-day and the
average BMDL10 is 1,001 mg/kg-day.
Lymphoid Hyperplasia in Male Rats
The data (4/50, 6/50, and 13/49 for control, low dose, and high dose, respectively) on
lymphoid hyperplasia in male rats treated with decaBDE (NTP, 1986) were modeled with EPA
BMDS. The reported oral doses were 0, 1,120, and 2,240 mg/kg-day, respectively. The data
were modeled with all the dichotomous models available in the BMDS, and modeling results are
summarized in Table B-4.
Based on these results, the models that adequately fit the data were the logistic,
multistage, probit, quantal-linear, and quantal -quadratic. The quantal -quadratic model (shown in
bold in the table) provided a relatively high goodness-of-fit/> value and the lowest AIC value.
-------
Based on these considerations, the BMD and BMDL estimates from this model are selected. For
lymphoid hyperplasia of male rats, the BMD10 is 1,538 mg/kg-day and the BMDL10 is
1,165 mg/kg-day.
Table B-4. Summary of BMD modeling results for lymphoid hyperplasia in
male rats
Model
Gamma
Log-logistic
Logistic
Multistage
Log-probit
Probit
Quantal-linear
Quantal-quadratic
Weibull
Goodness-of-fit
p value
N/Aa
N/A
0.62
0.36
N/A
0.57
0.36
0.86
N/A
AIC
127.266
127.266
125.512
126.122
127.266
125.59
126.122
125.298
127.266
BMD10
1,601
1,608
1,404
1,207
1,575
1,364
1,207
1,538
1,620
BMDL10
765
725
1,063
707
1,032
1,007
707
1,165
765
aN/A = not applicable.
Source: NTP(1986).
Centrilobular Hypertrophy in the Livers of Male Mice
The data (0/50, 34/50, and 32/50 for control, low dose, and high dose, respectively) on
centrilobular hypertrophy in the liver of male mice treated with decaBDE (NTP, 1986) were
modeled with EPA BMDS. The reported oral doses were 0, 3,200, and 6,650 mg/kg-day,
respectively. The data were modeled with all the dichotomous models available in the BMDS,
and modeling results are summarized in Table B-5.
Table B-5. Summary of BMD modeling results for centrilobular hypertrophy
in the liver of male mice
Model
Gamma
Log-logistic
Logistic
Multistage
Log-probit
Probit
Quantal-linear
Quantal-quadratic
Weibull
Goodness-of-fit
p value
0.005
0.094
0
0.005
0.004
0
0.005
0
0.005
AIC
140.455
134.65
166.133
140.455
140.591
165.095
140.455
164.89
140.455
Local chi-square
residual
2.48
1.44
3.87
0.7
2.22
4.03
2.48
5.08
2.48
BMD10
479
258
1,286
479
839
1,253
479
1,612
479
BMDL10
389
180
1,046
389
676
1,036
389
1,451
389
Source: NTP (1986).
B-7
-------
Based on the BMD modeling results from all the models used, none of the available
dichotomous models in the BMDS provided satisfactory data fit because all the goodness-of-fit
p values are less than 0. 1 . This unsatisfactory model fit is due to the nonmonotonic dose
response for this particular endpoint. Therefore, BMD and BMDL estimated from these model
fits cannot be used.
Follicular Cell Hyperplasia in the Thyroid of Male Mice
The data (2/50, 10/50, and 19/50 for control, low dose, and high dose, respectively) on
follicular cell hyperplasia in the thyroid of male mice treated with decaBDE (NTP, 1986) were
modeled with EPA BMDS. The reported oral doses were 0, 3,200, and 6,650 mg/kg-day,
respectively. The data were modeled with all the dichotomous models available in the BMDS,
and modeling results are summarized in Table B-6.
Table B-6. Summary of BMD modeling results for follicular cell hyperplasia
in the thyroid of male mice
Model
Gamma
Log-logistic
Logistic
Multistage
Log-probit
Probit
Quantal-linear
Quantal-quadratic
Weibull
Goodness-of-fit
p value
N/Aa
N/A
0.34
0.76
0.66
0.44
0.76
0.26
N/A
AIC
139.241
139.241
138.147
137.337
137.436
137.846
137.337
138.466
139.241
Local chi-square
residual
0
<0.01
0.70
-0.09
0.34
0.59
-0.25
0.96
0.00002
BMDio
2,040
2,089
2,989
1,670
2,561
2,792
1,670
3,135
2,022
BMDLio
1,196
1,008
2,430
1,190
1,977
2,265
1,190
2,598
1,196
3N/A = not applicable.
Source: NTP (1986).
Based on these results, the logistic, multistage, log-probit, probit, quantal-linear, and
quantal-quadratic models showed adequate fit of the data. The multistage and quantal-linear
models (shown in bold in the table) provided adequate goodness-of-fit p values and the lowest
AIC values. Since both models provided the same data fit, the BMDs estimated from these
models are identical. For follicular cell hyperplasia in the thyroid of male mice, the BMD10 is
1,670 mg/kg-day and the BMDLio is 1,190 mg/kg-day.
-------
APPENDIX C: BENCHMARK DOSE ANALYSIS OF CANCER ENDPOINTS
All the potential cancer endpoints in the rat and mouse chronic studies (NTP, 1986)
expressed as responses from all the animals treated were modeled, and BMD modeling results
are summarized below. All the cancer endpoints are modeled with the cancer model in the
BMDS, version 1.40b.
Neoplastic Nodules in the Liver of Male Rats
In the rat chronic study (NTP, 1986), there was a dose-dependent increase in the
incidences of neoplastic nodules in the livers of male rats. The incidences of the lesion were
1/50, 7/50, and 15/49 for control, low dose, and high dose, respectively. The original oral doses
were 0, 1,120, and 2,240 mg/kg-day, and the corresponding HEDs are 0, 305.1, and 608.3 mg/kg-
day, respectively. These incidence data on neoplastic nodules in male rats were modeled by
using the multistage model as recommended for a tumorigenic endpoint, and the modeling
results are summarized in Table C-l.
Table C-l. Summary of BMD modeling results for increase in neoplastic
nodules in the liver of male rats
Model
Multistage
Goodness-of-fit
p value
0.58
AIC
114.935
ED122
250
LED122
174
Source: NTP (1986).
The low-dose group resulted in an incidence of 7/50, which corresponds to 12.2% extra
risk. Therefore, the cancer ED at an extra risk of 12.2% was estimated to be 250 mg/kg-day, and
the corresponding LED is 174 mg/kg-day. The slope factor at the 12.2% extra risk is 0.0007 per
mg/kg-day.
C-l
-------
Neoplastic nodules in the liver of male rats (NTP, 1986)
BMR=12.2% extra risk
Cancer Model. (Version: 1.2; Date: 10/20/2005)
Input Data File: C:\BMDS140B\UNSAVED1.(d)
Gnuplot Plotting File: C:\BMDS140B\UNSAVEDl.plt
Thu Dec 01 00:38:02 2005
BMDS MODEL RUN
The form of the probability function is:
P[response] = background + (1-background)*[1-EXP(-betal*doseAl)]
The parameter betas are restricted to be positive
Dependent variable = response
Independent variable = dose
Total number of observations = 3
Total number of records with missing values = 0
Total number of parameters in model = 2
Total number of specified parameters = 0
Degree of polynomial = 1
Maximum number of iterations = 250
Relative Function Convergence has been set to: le-008
Parameter Convergence has been set to: le-008
Default Initial Parameter Values
Background = 0.0063916
Beta(l) = 0.000566746
Asymptotic Correlation Matrix of Parameter Estimates
Background Beta(l)
Background 1 -0.73
Beta(l) -0.73 1
Parameter Estimates
Variable Estimate
Background
Beta(l)
0.0190343
0.000520805
95.0% Wald Confidence Interval
Std. Err Lower Conf. Upper Conf.
Limit Limit
0.127925 -0.231695 0.269764
0.000351251 -0.000167635 0.00120924
Analysis of Deviance Table
Model Log(likelihood)
# Param's Deviance
Full model
Fitted model
Reduced model
-55.3119
-55.4674
-63.9318
3
2
1
Test
d.f.
0.311055 1
17.2399 2
P-value
0.577
0.0001805
AIC:
114.935
C-2
-------
Goodness of Fit
Dose
0.0000
305.1000
608.3000
ChiA2 = 0.30
Est._Prob
0.01^0
0.1632
0.2854
Scaled
Expected Observed
0.933 1
8.158 7
13.984 15
d.f. = 1
P-value = 0.5811
Benchmark Dose Computation
Specified effect = 0.122
Risk Type = Extra risk
Confidence level = 0.95
BMD
BMDL
BMDU
249.822
173.541
401.462
Size
49
50
49
Residual
0.070
-0.443
0.321
Taken together, (173.541, 401.462) is a 90% two-sided confidence interval for
the BMD
Cancer Slope Factor =
0.000703004
0.5
0.4
0.3
0.2
0.1
Cancer Model with 0.95 Confidence Level
Cancer
Linear extrapolation
BMDL
BMD
100
200
300
dose
400
500
600
00:38 12/01 2005
C-3
-------
Neoplastic Nodules or Carcinomas (Combined) in the Liver of Male Rats
The data (2/50, 8/50, and 15/49 for control, low dose, and high dose, respectively) on
neoplastic nodules or carcinomas (combined) in the liver of male rats treated with decaBDE
(NTP, 1986) were also modeled with BMDS. The modeling results are summarized in
Table C-2.
Table C-2. Summary of BMD modeling results for increases in neoplastic
nodules or carcinomas (combined) in the liver of male rats
Model
Multistage
Goodness-of-fit
p value
0.71
AIC
125.18
ED124
263
LED124
178
Source: NTP (1986).
The low-dose group resulted in an incidence of 8/50, which corresponds to 12.4% extra
risk. Therefore, the cancer ED at an extra risk of 12.4% was estimated to be 263 mg/kg-day, and
the corresponding LED is 178 mg/kg-day. The slope factor at the 12.4% extra risk is 0.0007 per
mg/kg-day.
C-4
-------
Neoplastic Nodules or Carcinomas (Combined) in the Liver of Male Rats (NTP,
1986)
BMR=12.4% extra risk
Cancer Model. (Version: 1.2; Date: 10/20/2005)
Input Data File: C:\BMDS140B\UNSAVED1.(d)
Gnuplot Plotting File: C:\BMDS140B\UNSAVEDl.plt
Thu Dec 01 00:51:35 2005
BMDS MODEL RUN
The form of the probability function is:
P[response] = background + (1-background)*[1-EXP(-betal*doseAl)]
The parameter betas are restricted to be positive
Dependent variable = response
Independent variable = dose
Total number of observations = 3
Total number of records with missing values = 0
Total number of parameters in model = 2
Total number of specified parameters = 0
Degree of polynomial = 1
Maximum number of iterations = 250
Relative Function Convergence has been set to: le-008
Parameter Convergence has been set to: le-008
Default Initial Parameter Values
Background = 0.0312973
Beta(l) = 0.00053218
Asymptotic Correlation Matrix of Parameter Estimates
Background Beta(l)
Background 1 -0.73
Beta(l) -0.73 1
Parameter Estimates
Variable Estimate
95.0% Wald Confidence Interval
Std. Err. Lower Conf. Limit Upper Conf.
Limit
Background 0.039023 0.127636 -0.21114 0.289186
Beta(l) 0.000503605 0.000358293 -0.000198637 0.00120585
Analysis of Deviance Table
Model Log(likelihoo # Deviance Test d.f. P-value
d) Param's
Full model -60.5216 3
Fitted -60.5898 2 0.136394 1 0.7119
model
Reduced -67.2168 1 13.3904 2 0.001237
model
AIC:
125.18
C-5
-------
Goodness of Fit
Dose
0.0000
305.1000
608.3000
Est. Prob.
0.03^0
0.1759
0.2926
Expected
1.912
8.795
14.337
Observed
2
8
15
Scaled
Size
49
50
49
Residual
0.065
-0.295
0.208
ChiA2 = 0.13
d.f. = 1
P-value = 0.7136
Benchmark Dose Computation
Specified effect = 0.124
Risk Type = Extra risk
Confidence level = 0.95
BMD
BMDL
BMDU
262.883
177.57
464.193
Taken together, (177.57 , 464.193) is a 90% two-sided confidence interval for
the BMD
Cancer Slope Factor =
0.000698317
0.5
0.4
0.3
0.2
0.1
Cancer Model with 0.95 Confidence Level
Cancer
Linear extrapolation
BMDL
BMD
0 100
00:51 12/01 2005
200
300
dose
400
500
600
C-6
-------
Neoplastic Nodules in the Liver of Female Rats
In the rat chronic study (NTP, 1986), there was also a dose-dependent increase in the
incidences of neoplastic nodules in the livers of female rats. The incidences of the lesion were
1/50, 3/49, and 9/50 for control, low dose, and high dose, respectively. The original oral doses
were 0, 1,200, and 2,550 mg/kg-day, and the corresponding HEDs are 0, 292.2, and 618.4 mg/kg-
day, respectively. The modeling results are summarized in Table C-3.
Table C-3. Summary of BMD modeling results for increases in neoplastic
nodules in the liver of female rats
Model
Multistage
Goodness-of-fit
p value
0.44
AIC
84.15
ED42
171
LED42
103
Source: NTP (1986).
The low-dose group resulted in an incidence of 3/49, which corresponds to 4.2% extra
risk. Therefore, the cancer ED at an extra risk of 4.2% was estimated to be 171 mg/kg-day, and
the corresponding LED is 103 mg/kg-day. The slope factor at the 4.2% extra risk is 0.0004 per
mg/kg-day.
C-7
-------
Neoplastic Nodules in the Liver of Female Rats (NTP, 1986)
BMR=4.2% extra risk
Cancer Model. (Version: 1.2; Date: 10/20/2005)
Input Data File: C:\BMDS140B\UNSAVED1.(d)
Gnuplot Plotting File: C:\BMDS140B\UNSAVEDl.plt
Thu Dec 01 01:12:25 2005
BMDS MODEL RUN
The form of the probability function is:
P[response] = background + (1-background)*[1-EXP(-betal*doseAl)]
The parameter betas are restricted to be positive
Dependent variable = response
Independent variable = dose
Total number of observations = 3
Total number of records with missing values = 0
Total number of parameters in model = 2
Total number of specified parameters = 0
Degree of polynomial = 1
Maximum number of iterations = 250
Relative Function Convergence has been set to: le-008
Parameter Convergence has been set to: le-008
Default Initial Parameter Values
Background = 0.00569581
Beta(l) = 0.000290683
Asymptotic Correlation Matrix of Parameter Estimates
Background Beta(l)
Background 1 -0.73
Beta(l) -0.73 1
Parameter Estimates
Confidence Interval
Variable Estimate
Background
Beta(l)
0.0173602
0.000250587
95.0% Wald
Std. Err. Lower Conf. Limit Upper Conf.
Limit
0.122542 -0.222817 0.257538
0.000317551 -0.000371802 0.000872976
Analysis of Deviance Table
Model
Full model
Fitted model
Reduced model
AIC:
Log(likelihood #
-39.7575
-40.074
-44.1226
Param's
3
2
1
Deviance
0.633004
8.73022
Test
d.f.
1
2
P-value
0.4263
0.01271
84.148
C-8
-------
Goodness of Fit
Dose
0.0000
292.2000
618.4000
ChiA2 = 0.60
Est._Prob.
0.0174
0.0867
0.1584
d.f. = 1
Scaled
Expected Observed Size
0.868 1 50
4.250 3 49
7.921 9 50
P-value = 0.4394
Residual
0.143
-0.635
0.418
Benchmark Dose Computation
Specified effect = 0.042
Risk Type = Extra risk
Confidence level = 0.95
BMD = 171.228
BMDL = 103.205
BMDU = 388.232
Taken together, (103.205, 388.232) is a 90% two-sided confidence interval for
the BMD
Cancer Slope Factor = 0.000406959
Cancer Model with 0.95 Confidence Level
0.35
0.3
0.25
0.2
0.15
0.1
0.05
0
Cancer
Linear extrapolation
BMDL
BMD
100
200
300
dose
400
500
600
01:12 12/01 2005
C-9
-------
Neoplastic Nodules and Carcinomas (Combined) in the Liver of Female Rats
The data (1/50, 5/49, and 9/50 for control, low dose, and high dose, respectively) on
neoplastic nodules or carcinomas (combined) in the liver of female rats treated with decaBDE
(NTP, 1986) were also modeled. The original oral doses were 0, 1,200, and 2,550 mg/kg-day,
and the corresponding HEDs are 0, 292.2, and 618.4 mg/kg-day, respectively. The modeling
results are summarized in Table C-4.
Table C-4. Summary of BMD modeling results for increases in neoplastic
nodules and carcinomas (combined) in the liver of female rats
Model
Multistage
Goodness-of-fit
p value
0.96
AIC
93.24
ED84
301
LED84
186
Source: NTP (1986).
The low-dose group resulted in an incidence of 5/49, which corresponds to 8.4% extra
risk. Therefore, the cancer ED at an extra risk of 8.4% was estimated to be 301 mg/kg-day, and
the corresponding LED is 186 mg/kg-day. The slope factor at the 8.4% extra risk is 0.0005 per
mg/kg-day.
C-10
-------
Neoplastic Nodules and Carcinomas (Combined) in the Liver of Female Rats
(NTP, 1986)
BMR=8.4% extra risk
Cancer Model. (Version: 1.2; Date: 10/20/2005)
Input Data File: C : \BMDS140B\UNSAVED1 . (d)
Gnuplot Plotting File: C:\BMDS140B\UNSAVEDl.plt
Thu Dec 01 01:23:38 2005
BMDS MODEL RUN
The form of the probability function is:
P [response] = background + ( 1-background) * [ 1-EXP ( -betal*doseAl ) ]
The parameter betas are restricted to be positive
Dependent variable = response
Independent variable = dose
Total number of observations = 3
Total number of records with missing values = 0
Total number of parameters in model = 2
Total number of specified parameters = 0
Degree of polynomial = 1
Maximum number of iterations = 250
Relative Function Convergence has been set to: le-008
Parameter Convergence has been set to: le-008
Default Initial Parameter Values
Background = 0.0211024
Beta(l) = 0.000288051
Asymptotic Correlation Matrix of Parameter Estimates
Background
Background 1
Beta(l) -0.75
Parameter Estimates
Beta(l)
-0.75
1
95.0% Wald Confidence Interval
Variable
Background
Beta (1)
Model
Full model
Fitted model
Estimate
0.0202022
0.000291122
Std. Err.
0.128079
0.000344031
Lower Conf. Limit
-0.230828
-0.000383167
Upper Conf.
Limit
0.271232
0.00096541
Analysis of Deviance Table
Log(likelihood) # Param's Deviance
-44.6193
-44.6208
Reduced
model
-48.6567
AIC: 93.2416
0.0030289
1
8.07484
Test
d.f.
1
2
P-value
0.9561
0.01764
C-ll
-------
Goodness of Fit
Dose
0.0000
292.2000
618.4000
Est._Prob.
0.02"02
0.1001
0.1816
Scaled
Expected Observed Size
1.010 1 50
4.905 5 49
9.081 9 50
Residual
-0.010
0.045
-0.030
ChiA2 = 0.00
d.f. = 1
P-value = 0.9560
Benchmark Dose Computation
Specified effect = 0.084
Risk Type = Extra risk
Confidence level = 0.95
BMD
BMDL
BMDU
301.382
186.131
676.695
Taken together, (186.131, 676.695) is a 90% two-sided confidence interval for
the BMD
Cancer Slope Factor = 0.000451296
Cancer Model with 0.95 Confidence Level
0.35
0.3
0.25
0.2
0.15
0.1
0.05
0
Cancer
Linear extrapolation
BMDL
BMD
100
200
300
dose
400
500
600
01:23 12/01 2005
C-12
-------
Thyroid Follicular Cell Hyperplasia in Male Mice
In the mouse chronic study (NTP, 1986), there were slight increases in follicular cell
adenomas or carcinomas (combined) accompanied by significant increase in follicular cell
hyperplasia in male mice. Because the follicular cell hyperplasia is considered a stage of the
thyroid cell carcinogenic process, this endpoint was also modeled as a cancer endpoint.
Therefore, the data (2/50, 10/50, and 19/50 for control, low dose, and high dose, respectively) on
follicular cell hyperplasia in the thyroid of male mice treated with decaBDE (NTP, 1986) were
modeled. The original oral doses were 0, 3,200, and 6,650 mg/kg-day, and the corresponding
HEDs are 0, 486.5, and 1,008.6 mg/kg-day, respectively. The modeling results are summarized
in Table C-5.
Table C-5. Summary of BMD modeling results for increases in thyroid
follicular cell hyperplasia as a key event of thyroid tumor in male mice
Model
Multistage
Goodness-of-fit
/rvalue
0.75
AIC
137.34
ED16.7
440
LED167
313
Source: NTP (1986).
The low-dose group resulted in an incidence of 10/50, which corresponds to 16.7% extra
risk. Therefore, the cancer ED at an extra risk of 16.7% was estimated to be 440 mg/kg-day, and
the corresponding LED is 313 mg/kg-day. The slope factor at the 16.7% extra risk is 0.0005 per
mg/kg-day.
C-13
-------
Thyroid Follicular Cell Hyperplasia in Male Mice (NTP, 1986)
BMD:16. 7%
Cancer Model. (Version: 1.2; Date: 10/20/2005)
Input Data File: C:\BMDS140B\UNSAVED1.(d)
Gnuplot Plotting File: C:\BMDS140B\UNSAVEDl.plt
Thu Dec 01 01:32:16 2005
BMDS MODEL RUN
The form of the probability function is:
P[response] = background + (1-background)*[1-EXP(-betal*doseAl)]
The parameter betas are restricted to be positive
Dependent variable = response
Independent variable = dose
Total number of observations = 3
Total number of records with missing values = 0
Total number of parameters in model = 2
Total number of specified parameters = 0
Degree of polynomial = 1
Maximum number of iterations = 250
Relative Function Convergence has been set to: le-008
Parameter Convergence has been set to: le-008
Default Initial Parameter Values
Background = 0.0304923
Beta(l) = 0.000434152
Asymptotic Correlation Matrix of Parameter Estimates
Background Beta(l)
Background 1 -0.72
Beta(l) -0.72 1
Parameter Estimates
95.0% Wald Confidence Interval
Variable Estimate Std. Err. Lower Conf. Limit Upper Conf.
Limit
Background 0.0386341 0.126668 -0.209631 0.2869
Beta(l) 0.000222182 -1.97658e-005 0.00085117
0.000415702
Model
Full model
Fitted model
Reduced
model
AIC:
Analysis of Deviance Table
Log(likelihood) # Param's Deviance
-66.6205
-66.6699
-76.426
0.0987661
19.6109
Test
d.f.
1
2
P-value
0.7533
<.0001
137.34
C-14
-------
Goodness of Fit
Dose
0.0000
486.5000
1008.6000
Est. Prob.
0.03^6
0.2147
0.3679
Expected
1.932
10.733
18.394
Observed
2
10
19
Scaled
Size
50
50
50
Residual
0.050
-0.252
0.178
ChiA2 = 0.10
d.f. = 1
P-value = 0.7544
Benchmark Dose Computation
Specified effect = 0.167
Risk Type = Extra risk
Confidence level = 0.95
BMD
BMDL
BMDU
439.549
313.299
685.676
Taken together, (313.299, 685.676) is a 90% two-sided confidence interval for
the BMD
Cancer Slope Factor = 0.000533037
Cancer Model with 0.95 Confidence Level
0.5
0.4
0.3
0.2
0.1
Cancer
Linear extrapolation
BMDL
BMD
0 200
01:32 12/01 2005
400 600
dose
800
1000
C-15
-------
Combined Incidence of Hepatocellular Adenomas or Carcinomas in Male Mice
In the mouse chronic study (NTP, 1986), there were significant increases in combined
hepatocellular adenomas or carcinomas in male mice. Therefore, the data (8/50, 22/50, and
18/50 for control, low dose, and high dose, respectively) on combined hepatocellular adenomas
or carcinomas of male mice treated with decaBDE (NTP, 1986) were modeled. The original
oral doses were 0, 3200, and 6650 mg/kg-day, and the corresponding HEDs are 0, 486.5, and
1008.6 mg/kg-day, respectively. The modeling results are summarized in Table C-6.
Table C-6. Summary of BMD modeling results for increases in combined
hepatocellular adenomas or carcinomas in male mice
Model
Multistage
Goodness-of-fit
/rvalue
0.03
AIC
186.5
ED333
1154
LED333
680
Source: NTP (1986).
The low-dose group resulted in an incidence of 22/50, which corresponds to 33.3% extra
risk. Therefore, the cancer ED at an extra risk of 33.3% was estimated to be 1,154 mg/kg-day,
and the corresponding LED is 680 mg/kg-day. The slope factor at the 33.3% extra risk is 0.0005
per mg/kg-day.
C-16
-------
Combined Incidence of Hepatocellular Adenomas or Carcinomas in Male Mice
(NTP, 1986)
BMR=33.3% extra risk
Cancer Model. (Version: 1.2; Date: 10/20/2005)
Input Data File: C:\BMDS140B\UNSAVED1.(d)
Gnuplot Plotting File: C:\BMDS140B\UNSAVEDl.plt
Thu Dec 01 01:47:16 2005
BMDS MODEL RUN
The form of the probability function is:
P[response] = background + (1-background)*[1-EXP(-betal*doseAl)]
The parameter betas are restricted to be positive
Dependent variable = response
Independent variable = dose
Total number of observations = 3
Total number of records with missing values = 0
Total number of parameters in model = 2
Total number of specified parameters = 0
Degree of polynomial = 1
Maximum number of iterations = 250
Relative Function Convergence has been set to: le-008
Parameter Convergence has been set to: le-008
Default Initial Parameter Values
Background = 0.235837
Beta(l) = 0.000263218
Asymptotic Correlation Matrix of Parameter Estimates
Background Beta(l)
Background 1 -0.77
Beta(l) -0.77 1
Parameter Estimates
95.0% Wald Confidence Interval
Variable Estimate Std. Err. Lower Conf. Limit Upper Conf.
Limit
Background 0.19695 0.125592 -0.0492049 0.443105
Beta(l) 0.000350822 0.00026836 -0.000175153 0.000876797
Analysis of Deviance Table
Model Log(likelihood) # Param's Deviance Test P-value
d.f.
Full model -88.9509 3
Fitted model -91.2692 2 4.63657 1 0.0313
Reduced -94.0304 1 10.159 2 0.006223
model
Goodness of Fit
Scaled
Dose Est. Prob. Expected Observed Size Residual
0.0000 0.1970 9.848 8 50 -0.657
486.5000 0.3230 16.148 22 50 1.770
1008.6000 0.4363 21.813 18 50 -1.087
C-17
-------
ChiA2 =4.75
d.f. =1
P-value = 0.0293
Benchmark Dose Computation
Specified effect = 0.333
Risk Type = Extra risk
Confidence level = 0.95
BMD
BMDL
BMDU
1154.33
680.241
3796.73
Taken together, (680.241, 3796.73) is a 90% two-sided confidence interval for
the BMD
Cancer Slope Factor = 0.000489532
0.6
0.5
0.4
0.3 :
0.2 :
0.1
Cancer Model with 0.95 Confidence Level
r -
' ^ '
>
BMDL
<
>
BMI
200
400
600
dose
800
1000
1200
01:4712/01 2005
C-18
------- |