if!;	United States
Environmental Protectioi
if % Agency
EPA/690/R-16/001F
Final
9-28-2016
Provisional Peer-Reviewed Toxicity Values for
oAminophenol
(CASRN 95-55-6)
Superfund Health Risk Technical Support Center
National Center for Environmental Assessment
Office of Research and Development
U.S. Environmental Protection Agency
Cincinnati, OH 45268

-------
AUTHORS, CONTRIBUTORS, AND REVIEWERS
CHEMICAL MANAGER
Q. Jay Zhao, PhD, MPH, DABT
National Center for Environmental Assessment, Cincinnati, OH
DRAFT DOCUMENT PREPARED BY
SRC, Inc.
7502 Round Pond Road
North Syracuse, NY 13212
PRIMARY INTERNAL REVIEWERS
Scott C. Wesselkamper, PhD
National Center for Environmental Assessment, Cincinnati, OH
Jeffry L. Dean II, PhD
National Center for Environmental Assessment, Cincinnati, OH
This document was externally peer reviewed under contract to:
Eastern Research Group, Inc.
110 Hartwell Avenue
Lexington, MA 02421-3136
Questions regarding the content of this PPRTV assessment should be directed to the EPA Office
of Research and Development's National Center for Environmental Assessment, Superfund
Health Risk Technical Support Center (513-569-7300).
li
o-Aminophenol

-------
TABLE OF CONTENTS
COMMONLY USED ABBREVIATIONS AND ACRONYMS	iv
BACKGROUND	1
DISCLAIMERS	1
QUESTIONS REGARDING PPRTVs	1
INTRODUCTION	2
REVIEW OF POTENTIALLY RELEVANT DATA (NONCANCER AND CANCER)	5
HUMAN STUDIES	8
ANIMAL STUDIES	8
Oral Exposures	8
Inhalation Exposures	10
OTHER DATA (SHORT-TERM TESTS, OTHER EXAMINATIONS)	10
Genotoxicity	10
Supporting Animal Toxicity Studies	15
Metabolism/Toxicokinetic Studies	17
Mode-of-Action/Mechanistic Studies	17
DERIVATION 01 PROVISIONAL VALUES	18
DERIVATION OF ORAL REFERENCE DOSES	18
DERIVATION OF INHALATION REFERENCE CONCENTRATIONS	19
CANCER WEIGHT-OF-EVIDENCE DESCRIPTOR	19
DERIVATION OF PROVISIONAL CANCER POTENCY VALUES	19
APPENDIX A. SCREENING PROVISIONAL VALUES	20
APPENDIX B. REFERENCES	32
in
o-Aminophenol

-------
COMMONLY USED ABBREVIATIONS AND ACRONYMS
a2u-g
alpha 2u-globulin
MN
micronuclei
ACGIH
American Conference of Governmental
MNPCE
micronucleated polychromatic

Industrial Hygienists

erythrocyte
AIC
Akaike's information criterion
MOA
mode of action
ALD
approximate lethal dosage
MTD
maximum tolerated dose
ALT
alanine aminotransferase
NAG
N-acetyl-P-D-glucosaminidase
AST
aspartate aminotransferase
NCEA
National Center for Environmental
atm
atmosphere

Assessment
ATSDR
Agency for Toxic Substances and
NCI
National Cancer Institute

Disease Registry
NOAEL
no-observed-adverse-effect level
BMD
benchmark dose
NTP
National Toxicology Program
BMDL
benchmark dose lower confidence limit
NZW
New Zealand White (rabbit breed)
BMDS
Benchmark Dose Software
OCT
ornithine carbamoyl transferase
BMR
benchmark response
ORD
Office of Research and Development
BUN
blood urea nitrogen
PBPK
physiologically based pharmacokinetic
BW
body weight
PCNA
proliferating cell nuclear antigen
CA
chromosomal aberration
PND
postnatal day
CAS
Chemical Abstracts Service
POD
point of departure
CASRN
Chemical Abstracts Service Registry
PODadj
duration-adjusted POD

Number
QSAR
quantitative structure-activity
CBI
covalent binding index

relationship
CHO
Chinese hamster ovary (cell line cells)
RBC
red blood cell
CL
confidence limit
RDS
replicative DNA synthesis
CNS
central nervous system
RfC
inhalation reference concentration
CPN
chronic progressive nephropathy
RfD
oral reference dose
CYP450
cytochrome P450
RGDR
regional gas dose ratio
DAF
dosimetric adjustment factor
RNA
ribonucleic acid
DEN
diethylnitrosamine
SAR
structure activity relationship
DMSO
dimethylsulfoxide
SCE
sister chromatid exchange
DNA
deoxyribonucleic acid
SD
standard deviation
EPA
Environmental Protection Agency
SDH
sorbitol dehydrogenase
FDA
Food and Drug Administration
SE
standard error
FEVi
forced expiratory volume of 1 second
SGOT
glutamic oxaloacetic transaminase, also
GD
gestation day

known as AST
GDH
glutamate dehydrogenase
SGPT
glutamic pyruvic transaminase, also
GGT
y-glutamyl transferase

known as ALT
GSH
glutathione
SSD
systemic scleroderma
GST
glutathione-S-transferase
TCA
trichloroacetic acid
Hb/g-A
animal blood-gas partition coefficient
TCE
trichloroethylene
Hb/g-H
human blood-gas partition coefficient
TWA
time-weighted average
HEC
human equivalent concentration
UF
uncertainty factor
HED
human equivalent dose
UFa
interspecies uncertainty factor
i.p.
intraperitoneal
UFh
intraspecies uncertainty factor
IRIS
Integrated Risk Information System
UFS
subchronic-to-chronic uncertainty factor
IVF
in vitro fertilization
UFd
database uncertainty factor
LC50
median lethal concentration
U.S.
United States of America
LD50
median lethal dose
WBC
white blood cell
LOAEL
lowest-observed-adverse-effect level


iv
o-Aminophenol

-------
FINAL
09-28-2016
PROVISIONAL PEER-REVIEWED TOXICITY VALUES FOR
£>-AMINOPHENOL (CASRN 95-55-6)
BACKGROUND
A Provisional Peer-Reviewed Toxicity Value (PPRTV) is defined as a toxicity value
derived for use in the Superfund Program. PPRTVs are derived after a review of the relevant
scientific literature using established Agency guidance on human health toxicity value
derivations. All PPRTV assessments receive internal review by a standing panel of National
Center for Environment Assessment (NCEA) scientists and an independent external peer review
by three scientific experts.
The purpose of this document is to provide support for the hazard and dose-response
assessment pertaining to chronic and subchronic exposures to substances of concern, to present
the major conclusions reached in the hazard identification and derivation of the PPRTVs, and to
characterize the overall confidence in these conclusions and toxicity values. It is not intended to
be a comprehensive treatise on the chemical or toxicological nature of this substance.
The PPRTV review process provides needed toxicity values in a quick turnaround
timeframe while maintaining scientific quality. PPRTV assessments are updated approximately
on a 5-year cycle for new data or methodologies that might impact the toxicity values or
characterization of potential for adverse human health effects and are revised as appropriate. It is
important to utilize the PPRTV database (http://hhpprtv.ornl.gov) to obtain the current
information available. When a final Integrated Risk Information System (IRIS) assessment is
made publicly available on the Internet (http://www.epa.gov/iris). the respective PPRTVs are
removed from the database.
DISCLAIMERS
The PPRTV document provides toxicity values and information about the adverse effects
of the chemical and the evidence on which the value is based, including the strengths and
limitations of the data. All users are advised to review the information provided in this
document to ensure that the PPRTV used is appropriate for the types of exposures and
circumstances at the site in question and the risk management decision that would be supported
by the risk assessment.
Other U.S. Environmental Protection Agency (EPA) programs or external parties who
may choose to use PPRTVs are advised that Superfund resources will not generally be used to
respond to challenges, if any, of PPRTVs used in a context outside of the Superfund program.
This document has been reviewed in accordance with U.S. EPA policy and approved for
publication. Mention of trade names or commercial products does not constitute endorsement or
recommendation for use.
QUESTIONS REGARDING PPRTVs
Questions regarding the content of this PPRTV assessment should be directed to the EPA
Office of Research and Development's National Center for Environmental Assessment,
Superfund Health Risk Technical Support Center (513-569-7300).
1
o-Aminophenol

-------
FINAL
09-28-2016
INTRODUCTION
o-Aminophenol, CASRN 95-55-6, also known as 07^/?o-aminophenol, 2-aminophenol,
o-hydroxyaniline, 2-hydroxyaniline, 2-amino-l-hydroxybenzene, o-aminohydroxybenzene, or
o-hydroxyphenylamine, is a white crystalline solid commonly used in the fur and leather industry
(under the name Oxidation Base 17 or C.I. 76520) to convert leather, fur, and hair from shades of
gray to yellow-browns (Mitchell and Waring. 2012). o-Aminophenol is also used as a chemical
intermediate for a variety of substances including pharmaceuticals (HSDB. 2011). stains and
dyes (Mitchell et al.. 2003). and heterocyclic compounds such as oxyquinolines, phenoxamines,
and benzoxazoles (Mitchell and Waring. 2012). In addition, o-aminophenol is a strong reducing
agent used in photographic developers (Mitchell et al., 2003). It readily undergoes an
oxidation/cyclization reaction in the presence of air (oxygen) and light to yield
2-aminophenoxazin-3-one (CASRN 1916-59-2) (Mitchell et al.. 2003). To avoid this
undesirable reaction, o-aminophenol is often converted to a salt, such as a hydrochloride
(CASRN 51-19-4), an acetate, or a sulfate (CASRN 67845-79-8), to increase its stability
(Mitchell et al.. 2003V
The empirical formula for o-aminophenol is C6H7NO (see Figure 1). Table 1 summarizes
the physicochemical properties of o-aminophenol. Although o-aminophenol can act as both an
acid and a base, the basic dissociation constant is larger (Mitchell et al„ 2003). As a result, it is
more common to protonate the amine group than it is to remove a proton from the hydroxy
group. o-Aminophenol does not exist as a dipolar ion (Mitchell et al.. 2003).
NH
Figure 1. o-Aminophenol Structure
2
o-Aminophenol

-------
FINAL
09-28-2016
Table 1. Physicochemical Properties of o-Aminophenol (CASRN 95-55-6)
Property (unit)
Value
Physical state
White crystals that rapidly become yellow-brown when exposed to
air and light3
Boiling point (°C)
267b
Melting point (°C)
173.5b; sublimes rapidly without decomposition at 153°C at a
reduced pressure of 11 mm Hg0-'1
Density (g/cm3)
1.328b
Vapor pressure (mm Hg)
0.0031 (extrapolated)6
pH (unitless)
NV
pKa (at 25°C)
pKai [-NH3(+i -NH2 + H(+l] = 4.66a;
pKa2 [-OH -O1 1 + H(+l] = 9.7la
Solubility in water (g/L)
20f
Octanol-water partition constant (log Kow)
0.62g
Henry's law constant (atm m3/mol at 25°C)
1.98 x 10"'(estimated)'1
Soil adsorption coefficient Koc (mL/g)
92 (estimated)'1
Atmospheric OH rate constant
(cm3/molecule-sec at 25°C)
74.2 x 10 12 (estimated)'1
Atmospheric half-life (lir)
1.7 (estimated)'1
Relative vapor density (air = 1)
NV
Molecular weight (g/mol)
109.13
aMitchell et al. (2003).
bHavnes et al. (2014).
cMitchell and Waring (2012).
do-Aminophenol is often purified via sublimation under reduced pressure.
eThe vapor pressure was extrapolated from the measured boiling point using a regression-derived equation.
fO'Neil et al. (2013).
gHSDB (2011).
hU.S. EPA (2012b).
NV = not available.
o-Aminophenol's moderate vapor pressure and estimated low Henry's law constant
indicate that the compound could volatilize from dry surfaces but is unlikely to volatilize from
water or moist surfaces. Its measured moderate water solubility and estimated low
soil-adsorption coefficient indicate that o-aminophenol is likely to leach to groundwater or
undergo runoff after a rain event. Thus, migration to groundwater is likely to compete with
light-induced oxidation/cyclization in the environment depending on local conditions (wet, dry,
shade, etc.).
A summary of available toxicity values for o-aminophenol from EPA and other
agencies/organizations is provided in Table 2.
3
o-Aminophenol

-------
FINAL
09-28-2016
Table 2. Summary of Available Toxicity Values for o-Aminophenol (CASRN 95-55-6)
Source"
Value (applicability)
Notes
Reference
Noncancer
IRIS
NV
NA
U.S. EPA (2016)
HEAST
NV
NA
U.S. EPA (2011a)
DWSHA
NV
NA
U.S. EPA (2012a)
ATSDR
NV
NA
ATSDR (2016)
IPCS
NV
NA
IPCS (2016); WHO (2016)
Cal/EPA
NV
NA
Cal/EPA (2014); Cal/EPA (2016a); Cal/EPA (2016b)
OSHA
NV
NA
OSHA (2006); OSHA (2011)
NIOSH
NV
NA
NIOSH (2016)
ACGIH
NV
NA
ACGIH (2015)
Cancer
IRIS
NV
NA
U.S. EPA (2016)
HEAST
NV
NA
U.S. EPA (2011a)
DWSHA
NV
NA
U.S. EPA (2012a)
NTP
NV
NA
NTP (2014)
IARC
NV
NA
IARC (2015)
Cal/EPA
NV
NA
Cal/EPA (2011); Cal/EPA (2016a); Cal/EPA (2016b)
ACGIH
NV
NA
ACGIH (2015)
aSources: ACGIH = American Conference of Governmental Industrial Hygienists; ATSDR = Agency for Toxic
Substances and Disease Registry; Cal/EPA = California Enviromnental Protection Agency; DWSHA = Drinking
Water Standards and Health Advisories; HEAST = Health Effects Assessment Summary Tables;
IARC = International Agency for Research on Cancer; IPCS = International Programme on Chemical Safety;
IRIS = Integrated Risk Information System; NIOSH = National Institute for Occupational Safety and Health;
NTP = National Toxicology Program; OSHA = Occupational Safety and Health Administration.
NA = not applicable; NV = not available.
Non-date-limited literature searches were conducted in June 2015 and May 2016 for
studies relevant to the derivation of provisional toxicity values for o-aminophenol
(CASRN 95-55-6). Searches were conducted using U.S. EPA's Health and Environmental
Research Online (HERO) database of scientific literature. HERO searches the following
databases: PubMed, ToxLine (including TSCATS1), and Web of Science. The following
databases were searched outside of HERO for health-related values: ACGIH, ATSDR, Cal/EPA,
U.S. EPA IRIS, U.S. EPA HEAST, U.S. EPA Office of Water (OW), U.S. EPA
TSCATS2/TSCATS8e, NIOSH, NTP, OSHA, and WHO.
4
o-Aminophenol

-------
FINAL
09-28-2016
REVIEW OF POTENTIALLY RELEVANT DATA
(NONCANCER AND CANCER)
Tables 3A and 3B provide an overview of the relevant noncancer and cancer databases,
respectively, for o-aminophenol and include all potentially relevant short-term-, subchronic-, and
chronic-duration studies. The phrase "statistical significance" and the term "significant(ly),"
used throughout the document, indicate ap-walue of < 0.05 unless otherwise noted.
5
o-Aminophenol

-------
FINAL
09-28-2016
Table 3A. Summary of Potentially Relevant Noncancer Data for o-Aminophenol (CASRN 95-55-6)
Category
Number of Male/Female,
Strain, Species, Study Type,
Study Duration
Dosimetry3
Critical Effects
NO A EL1
BMDL/
BMCLa
LOAEL1
Reference (comments)
Notesb
Human
1. Oral (mg/kg-d)
ND
2. Inhalation (mg/m3)
ND
Animal
1. Oral (mg/kg-d)a
Short-temi
5 M/0 F, rat (unspecified
strain), diet, 12 d
0, 83, 586
Targets of toxicity include
RBCs and potentially the liver
and kidney.
NDr
NDr
NDr
Eastman Kodak (1979)
(Inadequate reporting precludes
independent identification of
critical effect and
NOAEL/LOAEL.)
NPR
2. Inhalation (mg/m3)
ND
"¦Dosimetry: Values are converted to an ADD (mg/kg-day) for oral noncancer effects and an HEC (mg/m3) for inhalation noncancer effects. All repeated exposure values
are converted from a discontinuous to a continuous exposure, with the exception of values from animal developmental studies, which are not adjusted to a continuous
exposure.
bNotes: NPR = not peer reviewed.
ADD = adjusted daily dose; BMCL = benchmark concentration lower confidence limit; BMDL = benchmark dose lower confidence limit; F = female(s); HEC = human
equivalent concentration; LOAEL = lowest-observed-adverse-effect level; M = male(s); ND = no data; NDr = not determined; NOAEL = no-observed-adverse-effect
level; RBC = red blood cell.
6
o-Aminophenol

-------
FINAL
09-28-2016
Table 3B. Summary of Potentially Relevant Cancer Data for o-Aminophenol (CASRN 95-55-6)

Number of Male/Female,






Strain, Species, Study Type,


BMDL/


Category
and Duration
Dosimetry3
Critical Effects
BMCLa
Reference (comments)
Notesb
Human
1. Oral (mg/kg-d)
ND
2. Inhalation (mg/m3)
ND
Animal
1. Oral (mg/kg-d)a
Carcinogenicity
25 M/0 F, F344 rat, drinking
152
No evidence of hepatic or renal
NDr
Kurata et al. (1987)
PR

water, 52 wk (with or without

carcinogenicity or tumor




initiation with EHEN 2 wk

promotion




prior to exposure)





Carcinogenicity
6 M/0 F, S-D rat, 9 mo
14.5
No evidence of carcinogenicity
NDr
Miller and Miller (1948)
PR





(Study is considered inadequate due to lack of






controls, low animal number, short exposure






duration limited endpoint evaluation and a






single low dose that did not approach the MTD.)

2. Inhalation (mg/m3)
ND
"¦Dosimetry: The units for oral exposures are expressed as HEDs (mg/kg-day); HEDs are calculated using species-specific DAFs based on the animal:human body-weight
scaling to the 1/4 power (i.e., BW14) ratio recommended by U.S. EPA (2011b): rat:human ratio = 0.24.
bNotes: PR = peer reviewed.
BMCL = benchmark concentration lower confidence limit; BMDL = benchmark dose lower confidence limit; DAF = dosimetric adjustment factor;
EHEN = 7V-ethyl-7V-hydroxyethylnitrosamine; F = female(s); HED = human equivalent dose; M = male(s); MTD = maximum tolerated dose; ND = no data; NDr = not
determined; S-D = Sprague-Dawley.
7
o-Aminophenol

-------
FINAL
09-28-2016
HUMAN STUDIES
No data regarding the toxicity of o-aminophenol to humans following chronic or
subchronic exposure by any route have been identified. No acute oral or inhalation studies have
been located; however, methemoglobinemia was reported in humans following an intravenous
injection of o-aminophenol [Kiese and Rachor (1964) as cited by Akazawa et al. (2000)1. The
only other available human data are acute patch testing studies reporting positive reactions to
o-aminophenol in 8.3-25% of test subjects (Matsunaga et al.. 1989; Matsunaga et al.. 1988;
Katoh et al.. 1986; Yasuno. 1985).
ANIMAL STUDIES
Oral Exposures
The only repeated-dose oral studies found in the literature are an unpublished 12-day
dietary toxicity study in rats with limited data reporting (Eastman Kodak. 1979). a
tumor-promotion drinking water study in rats that evaluated limited systemic endpoints (Kurata
et al„ 1987), and a 9-month dietary study in rats that evaluated liver carcinogenicity (Miller and
Miller. 1948).
Short-Term-Duration Studies
Eastman Kodak (1979)
Groups of five male rats were fed diets containing 0, 0.1, or 1.0% o-aminophenol in diets
containing 1% corn oil for 12 consecutive days. The study authors calculated daily intakes to be
0, 83, or 586 mg/kg-day. Blood was collected for hematology (hemoglobin concentration,
hematocrit, red blood cell [RBC] morphology, white blood cell [WBC] count, and differential)
and serum chemistry (alanine aminotransferase [ALT], aspartate aminotransferase [AST],
alkaline phosphatase [ALP], lactate dehydrogenase [LDH], and glucose). At sacrifice, liver and
kidney weights were recorded, and unspecified tissues were examined for histopathological
lesions. No quantitative data were included in the study report.
No mortalities were reported. Clinical signs of toxicity observed at the 586-mg/kg-day
dose included a yellowish cast to the fur and rust-colored urine; no clinical signs of toxicity were
reported at the 83-mg/kg-day dose. Rats fed the 586 mg/kg-day diet consumed approximately
half of the amount of food as controls and did not gain any weight, while rats in the
83-mg/kg-day dose group consumed slightly less than controls but had comparable body-weight
gains. According to the study authors, decreased hemoglobin concentration and hematocrit, as
well as RBC morphology, in rats fed 586 mg/kg-day were indicative of a chemical-induced
anemia. Anemia was not observed at the 83-mg/kg-day dose. No changes in WBC counts or
differential were observed in exposed rats relative to controls. Serum chemistry values were
unaffected by treatment. Absolute liver and kidney weights were significantly reduced and
relative weights were significantly increased in the 586-mg/kg-day group, compared with
controls (percentage of changes were not reported); organ-weight effects were considered
secondary to lack of body-weight gain. The only organ-weight change reported in the
83-mg/kg-day group was a significant increase in relative liver weight, compared with control.
Histopathological changes attributed to treatment in the 586-mg/kg-day group were
mild-to-moderate diffuse hyperkeratosis in the nonglandular portion of the stomach (due to
irritation), minimal to moderate hypocellularity and congestion in the spleen, and atrophy of
mesenteric fat (secondary to reduced food intake). None of these histopathological changes were
observed in the 83-mg/kg-day group (control incidence not reported). The study authors
8
o-Aminophenol

-------
FINAL
09-28-2016
concluded that toxicity targets of o-aminophenol included the RBC and potentially the liver and
kidney.
Data reporting is inadequate for independent analysis; therefore, a
no-observed-adverse-effect level/lowest-observed-adverse-effect level (NOAEL/LOAEL)
determination was not made for this study.
Subchronic-Duration Studies
No studies evaluating adverse effects following sub chronic-duration oral exposure to
o-aminophenol have been identified.
Chronic-Duration/Carcinogenicity Studies
Kurataetal. (1987)
Seventy-five male Fischer 344 rats were divided into three groups (25/group). Two
groups were initiated with 0.1% A-ethyl-A'-hydroxyethylnitrosamine (EHEN) in the drinking
water for 2 weeks. Following the 2-week EHEN exposure, one of the groups was fed a diet
containing 0.8% o-aminophenol for 50 weeks (EHEN + o-aminophenol); the other group
continued to receive the basal diet (EHEN only). The third group was fed the 0.8%
o-aminophenol test diet for 50 weeks without EHEN-pretreatment (o-aminophenol only). All
rats were sacrificed in Week 52. At sacrifice, body, liver, and kidney weights were recorded.
Kidney and liver sections were microscopically evaluated for neoplastic lesions. Additional liver
sections were also evaluated for glutathione-»Y-transferase placental type (GST-P) positive foci
using immunohistochemistry.
Based on terminal animal numbers, it appears that one rat from EHEN + o-aminophenol
group, four rats from the EHEN-only group, and three rats from the o-aminophenol-only group
died prior to study termination, but mortality details were not provided. Body weights were
significantly decreased by 11-19% in the EHEN + o-aminophenol and o-aminophenol-only
groups, compared with the EHEN-only group. Absolute liver weights were significantly
decreased by 31-41% in the EHEN + o-aminophenol and o-aminophenol-only groups, compared
with the EHEN-only group; relative liver weight was also significantly decreased by 35% in the
o-aminophenol-only group, compared with the EHEN-only group. Absolute kidney weights
were comparable among groups, but relative kidney weights were significantly increased by
20-29% in the EHEN + o-aminophenol and o-aminophenol-only groups, compared with the
EHEN-only group. No neoplastic liver or kidney lesions were seen in the rats from the
o-aminophenol-only group. The number of hepatocellular carcinomas in the liver of rats from
the EHEN + o-aminophenol group (9/24) was significantly decreased compared with the
incidence in the EHEN-only group (15/21), suggesting that o-aminophenol inhibited liver tumor
development. Additionally, rats receiving EHEN + o-aminophenol showed significant decreases
in the incidence of GST-P positive foci and the area of the foci, compared to rats that received
EHEN alone. The incidence of neoplastic lesions in the kidney was comparable between the
EHEN + o-aminophenol group and the EHEN-only group.
Based on these results, o-aminophenol was not a carcinogenic or tumor-promoting agent
in the liver and kidneys under the conditions of this study. A NOAEL/LOAEL determination for
non-neoplastic effects (body and organ weights) was not made due to lack of an untreated
control. Based on reference body weights and water consumption values for male F344 rats in a
chronic-duration study (U.S. EPA. 1988). the estimated daily intake of o-aminophenol is
9
o-Aminophenol

-------
FINAL
09-28-2016
632 mg/kg-day, which converts to a human equivalent dose (HED) of 152 mg/kg-day using the
rat:human dosimetric adjustment factor (DAF) of 0.24 based on the animal :human body-weight
scaling to the 1/4 power (i.e., BW1/4) ratio recommended by the U.S. EPA (2011b).
Miller and Miller (1948)
A group of six male Sprague-Dawley (S-D) rats was fed a diet containing 0.117%
o-aminophenol hydrochloride for up to 9 months. A positive control group of 12 male rats was
fed a diet containing 0.06% 4-dimethylaminoazobenzene up to 9 months. It is unclear whether a
negative control group was used. The rats were sacrificed at 9 months, and the liver was
examined for gross evidence of cirrhosis and tumor incidence.
No gross evidence of liver cirrhosis or liver tumors was observed in the rats exposed to
dietary o-aminophenol hydrochloride. o-Aminophenol was not carcinogenic under the
conditions of this assay; however, confidence in this study is low due to lack of appropriate
controls, low animal numbers, limited endpoint analysis, and use of a low dose that did not
approach the maximum tolerated dose (MTD). Based on reference body weights and food
consumption values for male S-D rats in a chronic-duration study (U.S. EPA. 1988). the
estimated daily intake of o-aminophenol hydrochloride is 80.7 mg/kg-day. Based on
molar-weight ratios (o-aminophenol accounts for 75% of total molecular weight of
o-aminophenol hydrochloride), the estimated intake of o-aminophenol is 60.5 mg/kg-day, which
converts to an HED of 14.5 mg/kg-day using the rat:human DAF of 0.24 based on the
animal:human BW14 ratio recommended by the U.S. EPA (201 lb).
Reproductive/Developmental Studies
No studies evaluating the reproductive or developmental toxicity of o-aminophenol
following oral exposure have been identified.
Inhalation Exposures
No studies evaluating the toxicity of o-aminophenol following inhalation exposure have
been identified.
OTHER DATA (SHORT-TERM TESTS, OTHER EXAMINATIONS)
Genotoxicity
The genotoxicity of o-aminophenol was evaluated in vitro and in a limited number of in
vivo studies. Available studies are summarized below in Table 4. In general, the data indicate
that o-aminophenol is a weak mutagen that could cause deoxyribonucleic acid (DNA) damage
and clastogenic effects.
10
o-Aminophenol

-------
FINAL
09-28-2016
Table 4. Summary of o-Aminophenol (CASRN 95-55-6) Genotoxicity, Mutagenicity, and Clastogenicity
Endpoint
Test System
Dose/
Concentration3
Results without
Activationb
Results with
Activationb
Comments
References (notes)
Genotoxicity studies in prokaryotic organisms
Mutation
Salmonella tvphimurium
TA1535, TA1537,
TA98, and TA100
0, 10, 33, 100, 166,
333,666,
1,000 ng/plate
+
(TA100)
(TA1535,
TA1537, TA98)
+
(TA100)
(TA1535,
TA1537, TA98)
Preincubation assay: o-Aininophenol
induced a 2-3-fold increase in revertant
colonies in TA100 with and without S9
activation at >166 pg/plate. Cytotoxicity
was noted at >666 pg/plate without
activation.
Haworth et al.
(1983)
(Case Western
Reserve University)
Mutation
S. tvphimurium
TA1535, TA1537,
TA98, and TA100
0, 10,33, 100,333,
400, 666,
750 ng/plate
+
(TA100)
(TA1535,
TA1537, TA98)
+
(TA100)
(TA1535,
TA1537, TA98)
Preincubation assay: o-Aininophenol
induced a 2-3-fold increase in revertant
colonies in TA100 without activation at
>333 ng/plate and a 2-6-fold increase with
S9 activation at >100 pg/plate.
Cytotoxicity was noted at >666 pg/plate
without activation.
Haworth et al.
(1983)
(EG&G Mason
Research Institute)
Mutation
S. tvphimurium
TA1535, TA1537,
TA98, and TA100
0, 10, 33, 100, 333,
667, 1,000, 3,333,
6,667,
10,000 ng/plate

+
(TA100)
(TA1535,
TA1537, TA98)
Preincubation assay: o-Aininophenol
induced a 2-fold increase in revertant
colonies in TA100 with S9 activation at
>1,000 ng/plate. Cytotoxicity was noted at
>667 ng/plate without activation and
>6,667 ng/plate with activation.
Haworth et al.
(1983)
(SRI International)
Mutation
S. tvphimurium TA98
and TA100
Concentrations not
reported
+
+
Mutagenic activity was 2-3 times greater
than controls; unclear whether the effect
was observed in one or both strains.
Nishimura and
Osliima (1983) as
cited in U.S. EPA
(1985)
(Japanese study,
abstract only)
Mutation
S. tvphimurium TA98
and TA100
0, 25, 50, 100, 250,
500 ng/plate

+
(TA100)
(TA98)
Plate incorporation assay: o-Aininophenol
induced a 2-3-fold increase over control in
the number of revertant colonies in TA100
with S9 activation at >100 pg/plate.
Cytotoxicity in TA100 was noted at
>250 ng/plate without S9 activation.
Lavoie et al. (1979)
11
o-Aminophenol

-------
FINAL
09-28-2016
Table 4. Summary of o-Aminophenol (CASRN 95-55-6) Genotoxicity, Mutagenicity, and Clastogenicity
Endpoint
Test System
Dose/
Concentration3
Results without
Activationb
Results with
Activationb
Comments
References (notes)
Mutation
S tvphimurium G46,
TA1535, TA100,
C3076, TA1537,
D3052, TA1538, and
TA98
Plate gradient ranges:
0.1-1, 1-10, 10-100,
100-1,000 ng/inL
agar
+
(TA100)
(G46, TA1535,
C3076, TA1537,
D3052,
TA1538, TA98)
+
(TA100)
(G46, TA1535,
C3076, TA1537,
D3052,
TA1538, TA98)
Modified gradient plate test:
o-Aininophenol was mutagenic at
7-100 iig/mL agar; reporting is inadequate
to determine whether the compound was
mutagenic with metabolic activation,
without metabolic activation or under both
tested conditions.
ThoniDSon et al.
(1983)
Mutation
S. tvphimurium
TA1535, TA1537,
TA1538, TA98, and
TA100
Concentrations not
reported


Plate incorporation assay.
De Flora et al.
(1984)
Mutation
S. tvphimurium TA98
and TA100
0,0.5, 1.0,
2.0 |imol/platc (~0,
50, 100, 200 ng/plate)
+

Plate incorporation assay. No cytotoxicity
was observed.
Desawa et al.
(1979)
Mutation
S. tvphimurium TA98
and TA100
15-150 ng/plate


Plate incorporation assay.
Yoshikawa et al.
(1976) as cited in
U.S. EPA (1985)
(Japanese study)
Mutation
S. tvphimurium TA98
10-30 ng/plate


NA
Watanabe et al.
(1991) as cited in
CCRIS (1993)
(Japanese study)
Mutation
Escherichia coli
WP2and WF2«/vr.4-
Plate gradient ranges:
0.1-1, 1-10, 10-100,
100-1,000 ng/mL
agar


Modified gradient plate test.
ThoniDSon et al.
(1983)
DNA repair test
E. coli WP2 (wild-type),
WP67 (uvrApolA-),
andCM871 (uvrA-
recA-, lexA-)
Eight 2-fold dilutions
starting from
solubility or toxic
concentration
+
(CM871, WP67)

MIC was 20 ng.
De Flora et al.
(1984)
12
o-Aminophenol

-------
FINAL
09-28-2016
Table 4. Summary of o-Aminophenol (CASRN 95-55-6) Genotoxicity, Mutagenicity, and Clastogenicity
Endpoint
Test System
Dose/
Concentration3
Results without
Activationb
Results with
Activationb
Comments
References (notes)
DNA damage
(SOS
chromotest)
E. coli PQ37
Concentration not
reported
+
NV
o-Aminophenol was tested after in vitro
nitrosation via incubation with sodium
nitrate at 37°C for 1 lir prior to conducting
the SOS chromotest. The SOSIP of
o-aminophenol was 5.9 compared with
compounds that were not genotoxic
(SOSIP <0.006).
Bartsch et al.
(1991); Ohshima et
al. (1989)
Genotoxicity studies in nonmammalian eukaryotic organisms
Sex-linked
recessive lethal
mutations
Drosophila
melanogaster
0, 200 (feeding); 0,
100 (injection)
(feeding,
injection)
(feeding,
injection)
NA
Yoonetal. (1985)
Genotoxicity studies in mammalian cells—in vitro
CAs
CHO
0.1,0.2, 0.5 mM
+
NV
o-Aminophenol induced a 4-17-fold
increase in CAs. Observed CAs were
predominantly chromatid exchanges and
breaks, with a small percentage of
dicentrics and rings.
Kanava (1996)
SCE
Human lymphocytes
0, 1.6, 3.3, 6.6 ng/mL
+
NV
o-Aminophenol induced a 1.2-1.9-fold,
dose-dependent increase in SCE frequency
over control.
Kirchner and Baver
(1982)
SCE
Human fibroblasts
0,0.01,0.03,0.10,
0.3 mM
+
NV
o-Aminophenol induced a 2-fold increase
in SCE frequency at 0.10 mM.
Cytotoxicity was noted at 0.30 mM.
Wilmer et al. (1981)
SCE
CHO
0.1,0.2, 0.5 mM
+
NV
o-Aminophenol induced a 1.5-2-fold
dose-dependent increase in SCE.
Kanava (1996)
SCE
Chinese hamster V79
5-20 nM
+
NV
NA
Wild et al. (1981)
(abstract)
UDS
Primary rat hepatocytes
0.5, 1.0,5.0, 10, 50,
100, 500,
1,000 nmol/mL

NV
NA
Thompson et al.
(1983)
13
o-Aminophenol

-------
FINAL
09-28-2016
Table 4. Summary of o-Aminophenol (CASRN 95-55-6) Genotoxicity, Mutagenicity, and Clastogenicity
Endpoint
Test System
Dose/
Concentration3
Results without
Activationb
Results with
Activationb
Comments
References (notes)
Genotoxicity studies in mammals—in vivo
CAs
Ehrlich ascites tumor
cells in the mouse
Concentration not
reported


o-Aminophenol was "applied" to tumor
cells; no further details provided.
Boeaiewski and
Boeaiewska (1982)
(abstract)
SCE
Chinese hamster bone
marrow cells
5 mg/kg i.p.
—
—
Animal sacrificed 4 lir after injection.
Kirchner and Baver
(1982)
Mouse
micronucleus test
Mouse bone marrow
0.5-2.0 mM/kg i.p.
+
+
NA
Wild et al. (1981)
(abstract)
Genotoxicity studies with extracellular purified DNA
DNA cleavage
32P-5 '-end-labeled DNA
fragments from human
p53 tumor suppressing
gene and c-Ha-rav-1
proto-oncogene
0, 5 nM (without
Cu[II]); and 0, 1, 2,
and 5 |iIVI (with
Cu[II])
(without Cu[II])
+
(with Cu[II])
(without Cu[II])
+
(with Cu[II])
o-Aminophenol incubated in the presence
of Cu(II) caused DNA damage, but did not
cause damage without Cu(II).
Ohkuma and
Kawanishi (2001)
Oxidative DNA
damage
(8-oxodG
formation)
Calf thymus DNA
0, 1, 2, 5 ^M [with or
without Cu(II)]
(without Cu[II])
+
(with Cu[II])
(without Cu[II])
+
(with Cu[II])
o-Aminophenol incubated in the presence
of Cu(II) significantly increased the
formation of 8-oxodG, compared with
control. o-Aminophenol alone did not
induce oxidative DNA damage.
Ohkuma and
Kawanishi (2001)
DNA strand
break
Double-stranded /.DNA
250 nM
+
+
Exposure to o-aminophenol produced DNA
fragments between 0.6 and
0.000004 daltons.
Yamada et al.
(1985)
aLowest effective dose for positive results, highest dose tested for negative results.
b+ = positive; - = negative.
8-oxoG = 8-oxo-7,8-dihydro-29-deoxyguanosine; CA = chromosomal aberration; CHO = Chinese hamster ovary (cell line cells); Cu = copper; DNA = deoxyribonucleic
acid; i.p. = intraperitoneal; MIC = minimally inhibitory concentration; NA = not applicable; NV = not available; SCE = sister chromatid exchange;
SOSIP = SOS-inducing potency factor; UDS = unscheduled DNA synthesis.
14
o-Aminophenol

-------
FINAL
09-28-2016
Available evidence from in vitro studies indicates that o-aminophenol is a weak mutagen.
Several studies indicate that o-aminophenol is weakly mutagenic to Salmonella typhimurium
strain TA100 with and without metabolic activation. In general, induction of revertant colonies
was only two- to threefold greater than controls at noncytotoxic concentrations [Nishimura and
Oshima (1983) as cited in U.S. EPA (1985); Haworth et al. (1983); Thompson et al. (1983);
Lavoie et al. (1979)1. Other studies evaluating o-aminophenol found no evidence of
mutagenicity in S. typhimurium strain TA100 with or without metabolic activation [Yoshikawa
et al. (1976) as cited in U.S. EPA (1985); De Flora et al. (1984); Degawa et al. (1979)1.
Mutagenicity was not reported in various other S. typhimurium strains or Escherichia coli strains
WP2 or WF2//v/:4 [Watanabe et al. (1991) as cited in CCRIS (1993); Yoshikawa et al. (1976) as
cited in U.S. EPA (1985); De Flora et al. (1984); Haworth et al. (1983); Nishimura and Oshima
(1983); Thompson et al. (1983); Degawa et al. (1979); Lavoie et al. (1979)1. o-Aminophenol did
not cause sex-linked recessive lethal mutations in Drosophila melanogaster following exposure
via feeding or injection (Yoon et al.. 1985).
o-Aminophenol caused clastogenic effects in several in vitro assays without metabolic
activation, including chromosomal aberrations (CAs) in Chinese hamster ovary (CHO) cells and
sister chromatid exchanges (SCEs) in human lymphocytes, human fibroblasts, CHO cells, and
Chinese hamster V79 cells (Kan ay a. 1996; Kirchner and Bayer. 1982; Wild et al.. 1981; Wilmer
et al.. 1981). Micronuclei (MN) were induced in mouse bone marrow cells following
intraperitoneal (i.p.) injections of o-aminophenol (Wild et al„ 1981). However, in other in vivo
assays, o-aminophenol did not induce CA in Ehrlich ascites tumor cells injected into mice
(Bogaiewski and Bogaiewska. 1982) or SCE in Chinese hamster bone marrow cells (Kirchner
and Bayer. 1982).
Available data indicate that o-aminophenol is capable of causing DNA damage in vitro;
however, findings are inconsistent among different test systems. o-Aminophenol inhibited DNA
repair in E. coli strains CM871 and WP67, but not wild-type WP2 at concentrations >20 |ig/plate
(De Flora et al.. 1984). DNA damage was induced in E. coli strain PQ37 incubated with
nitrosated o-aminophenol in the SOS chromotest; o-aminophenol was not tested prior to
nitrosation (Bartsch et al.. 1991; Ohshima et al.. 1989). Unscheduled DNA synthesis was not
induced in primary rat hepatocytes exposed to o-aminophenol (Thompson et al.. 1983). In
isolated DNA samples, o-aminophenol induced strand breaks in double-stranded XDNA
(Yamada et al.. 1985). o-Aminophenol also caused DNA damage in calf thymus DNA and
32P-5'-end-labeled DNA fragments from humanp53 tumor-suppressing gene and c-Ha-ra.s -1
proto-oncogene in the presence, but not absence, of copper (Cu[II]), suggesting that oxidation of
o-aminophenol to the o-aminophenoxyl radical in the presence of Cu(II) underlies the observed
DNA damage in isolated DNA fragments (Ohkuma and Kawanishi. 2001).
Supporting Animal Toxicity Studies
Acute Toxicity
Acute studies indicate that o-aminophenol is a strong inducer of methemoglobinemia in
rats, but not mice. Methemoglobinemia was reported in rats exposed once to o-aminophenol at
an oral dose of 750 mg/kg, with methemoglobin formation of 50% in exposed rats compared
with 0.71% in controls 2 hours post-treatment (Eastman Kodak, 1979). In an i.p. injection study,
methemoglobin formation increased in a dose-related manner in rats exposed to o-aminophenol
at doses of 0.15-1.0 mM/kg (55-110 mg/kg); methemoglobin formation was -10% at the lowest
dose and -70% at the highest dose at 1 hour postinjection (data reported graphically, control data
15
o-Aminophenol

-------
FINAL
09-28-2016
not reported) (Harrison and Jollow. 1987). In contrast, methemoglobin formation rates were
much lower in mice (2-3%) following an i.p. injection of 300 mg/kg (control data not reported);
the study authors concluded that o-aminophenol was not a strong inducer of methemoglobin
formation in mice (Itoh. 1987). Itoh (1987) also reported low Heinz body formation (<4%) in
exposed mice following i.p. injection.
There is limited evidence from acute parenteral studies that o-aminophenol may cause
mild kidney toxicity. Itoh (1987) reported mild tubular vacuolated degeneration and slight
flattening of collecting tubules with dilatation in rat kidneys 1 week following an i.p. dose of
200 mg/kg (but not <150 mg/kg); however, no histological damage was observed in the kidneys
of rats 48 hours after a single intravenous injection of 2.8 mM/kg (310 mg/kg) (Calder et al..
1971). Other parenteral studies reported no changes in kidney weight or serum blood urea
nitrogen (BUN) levels (Rankin et al.. 1996; Newton et al.. 1982). although Rankin et al. (1996)
reported a transient increase in urinary protein excretion in rats exposed to o-aminophenol.
Reported oral median lethal dose (LD50) values include 951 mg/kg in male rats,
1,100 mg/kg in male and female rats, and 800 mg/kg in male mice (Eastman Kodak. 1979;
University of Miami, 1975). Animals receiving single doses >940 mg/kg showed clinical signs
of neurotoxicity, including tremors, convulsions, and central depression; it is unclear whether or
not neurotoxic effects were limited to moribund animals (University of Miami. 1975). In
anesthetized mice, the effective i.p. dose that caused 50% of mice (effective dose [ED50]) to have
a myoclonic convulsion (jerk) in response to a strong pinch was 3.42 mM/kg (376 mg/kg) (Angel
and Rogers. 1972).
Carcinogenicity
No elevated incidences of squamous metaplasia or carcinoma of the bladder were
observed in mice following implantation of cholesterol-based pellets containing o-aminophenol
in the urinary bladder, compared with mice implanted with a cholesterol pellet containing no
chemicals (Clavson et al.. 1958).
In a dermal cancer bioassay, no evidence of carcinogenicity was observed in rats
following skin exposure to a hair-dye formulation containing several chemicals, including 0.3%
o-aminophenol, for 2 years (Burnett and Goldenthal. 1988).
Reproductive/Developmental Effects
A two-generation reproduction study conducted by dermal exposure found no adverse
reproductive effects in rats following skin exposure to a hair-dye formulation containing several
chemicals, including 0.3% o-aminophenol (Burnett and Goldenthal. 1988).
Developmental effects of o-aminophenol were studied only by parenteral exposure.
Dose-related increases in frequency of litters with one or more malformed fetuses, number of
fetuses presenting with one or more malformations, and number of fetal resorptions were
observed in Syrian golden hamsters (LKV strain) following maternal exposure to
150-200 mg/kg via i.p. injection on Gestation Day (GD) 8, compared with controls; these effects
were not observed at 100 mg/kg (Rutkowski and Ferm. 1982). Induced malformations included
neural tube defects (encephalocele, exencephaly, and spina bifida), eye, limb, tail, and rib
defects, and umbilical hernia (often involving eventration of the abdominal viscera).
16
o-Aminophenol

-------
FINAL
09-28-2016
o-Aminophenol was not toxic to the dams at these doses, suggesting that the fetus may be a
sensitive target for o-aminophenol toxicity.
Metabolism/Toxicokinetic Studies
Oral absorption is very high (-95%) in laboratory animals (U.S. EPA. 1985). No
evidence of percutaneous absorption was observed through depilated abdominal skin in guinea
pigs during a 24-hour exposure in occluded conditions (Eastman Kodak. 1979). No distribution
data were located for o-aminophenol. o-Aminophenol is metabolized primarily via conjugation
at the phenolic group with sulfate or glucuronide, and the conjugated metabolites are excreted in
the urine (Elder. 1988; U.S. EPA. 1985). A-acetylation can also occur, forming an
acetamidophenol metabolite, which may be further conjugated with glucuronide prior to
excretion in the urine (see Table 5) (U.S. EPA. 1985).
Table 5. Percent Recovery of Administered Dose in the Urine over 24 Hours Following a
Single Oral Dose of o-Aminophenol in Rabbits (1,000 mg/Rabbit)a
Parent or Metabolite
Percent of Total
Parent
11
Aminophenylglucuronide
52
Aminophenolsulfate
15
Acetamidophenol
2-4
Acetamidophenylglucuronide
13
Acetamidophenylsulfate
0
aU.S. EPA (1985).
Mode-of-Action/Mechanistic Studies
Mode-of-action (MOA)/mechanistic data are consistent with the short-term-duration
Eastman Kodak (1979) in vivo study regarding toxicity to RBCs, kidney, and liver. Limited
evidence from acute human and animal studies suggests that methemoglobinemia may occur
following o-aminophenol exposure [Kiese and Rachlor (1964) as cited in Akazawa et al. (2000);
Harrison and Jollow (1987); Eastman Kodak (1979)1. o-Aminophenol also induces
methemoglobin formation in erythrocyte suspensions in vitro (Akazawa et al.. 2000; Harrison
and Jollow. 1987; Eckert and Ever. 1983; Smith et al.. 1967).
The kidney (Rankin et al.. 1996; Itoh. 1987; Newton et al.. 1982; Eastman Kodak. 1979;
Calder et al.. 1971) and liver (Eastman Kodak. 1979) may be potential targets of o-aminophenol
toxicity. In vitro, o-aminophenol was moderately toxic to renal slice cultures from male F344
and S-D rats, as evidenced by significantly reduced glutathione levels and decreased
gluconeogenesis in response to stimulation with pyruvate or 1,6-diphosphate at concentrations of
>0.1 mM (Valentovic and Ball. 1998; Valentovic et al.. 1996). Significantly increased LDH
leakage (indicative of cell death) was observed at 1 mM in slices from F344 rats only
(Valentovic and Ball. 1998; Valentovic et al.. 1996). Gluconeogenesis in response to stimulation
with pyruvate was also observed in hepatic slices from F344 rats at 2 mM o-aminophenol,
without evidence of cytotoxicity (Valentovic et al.. 1996).
17
o-Aminophenol

-------
FINAL
09-28-2016
DERIVATION OF PROVISIONAL VALUES
Tables 6 and 7 present summaries of noncancer and cancer references values,
respectively.
Table 6. Summary of Noncancer Reference Values for o-Aminophenol (CASRN 95-55-6)
Toxicity Type
(units)
Species/Sex
Critical
Effect
p-Reference
Value*
POD
Method
POD (HED)
UFc
Principal Study
Screening
subchronic p-RfD
(mg/kg-d)*
Rat/M
Increased
severity of
nephrosis
4 x 1(T2
NOAEL
12
(based on
surrogate POD)
300
Burnett et al.
(1989) as cited in
U.S. EPA (2005b)
Screening chronic
p-RfD (mg/kg-d)*
Rat/M
Increased
severity of
nephrosis
4 x 1(T3
NOAEL
12
(based on
surrogate POD)
3,000
Burnett et al.
(1989) as cited in
U.S. EPA (2005b)
Subchronic p-RfC
(mg/m3)
NDr
Chronic p-RfC
(mg/m3)
NDr
*See detailed derivation in Appendix A.
HED = human equivalent dose; M = rnale(s); NDr = not determined; NOAEL = no-observed-adverse-effect level;
POD = point of departure; p-RfC = provisional reference concentration; p-RfD = provisional reference dose;
UFC = composite uncertainty factor.
Table 7. Summary of Cancer Reference Values for o-Aminophenol (CASRN 95-55-6)
Toxicity Type (units)
Species/Sex
Tumor Type
Cancer Value
Principal Study
p-OSF (mg/kg-d) 1
NDr
p-IUR (mg/m3)-1
NDr
NDr = not determined; p-IUR = provisional inhalation unit risk; p-OSF = provisional oral slope factor.
DERIVATION OF ORAL REFERENCE DOSES
No studies were located regarding toxicity of o-aminophenol to humans by oral exposure.
Oral studies of o-aminophenol in animals were of inadequate duration and/or scope to support
derivation of a subchronic or chronic provisional reference dose (p-RfD). The only
repeated-dose study that evaluated non-neoplastic endpoints was conducted by Eastman Kodak
(19791 in which five male rats per dose were fed diets with 0, 83, or 586 mg/kg-day
o-aminophenol for 12 days. The study is limited by the use of a single sex (male), small animal
groups (five/group), and short duration (12 days). As a result of the limitations of the available
data for o-aminophenol, subchronic and chronic p-RfDs are not derived. Instead, screening
p-RfDs are derived in Appendix A using an alternative surrogate approach.
18
o-Aminophenol

-------
FINAL
09-28-2016
DERIVATION OF INHALATION REFERENCE CONCENTRATIONS
No subchronic- or chronic-duration inhalation studies of o-aminophenol in humans or
animals have been located, precluding derivation of provisional reference concentrations
(p-RfCs) for o-aminophenol based on chemical-specific data. An alternative surrogate approach
was attempted, but screening p-RfCs could not be derived due to a lack of inhalation toxicity
values for potential surrogates (see Appendix A).
CANCER WEIGHT-OF-EVIDENCE DESCRIPTOR
Table 8 identifies the cancer weight-of-evidence (WOE) descriptor for o-aminophenol.
Table 8. Cancer WOE Descriptor for o-Aminophenol (CASRN 95-55-6)
Possible WOE Descriptor
Designation
Route of Entry
Comments
"Carcinogenic to
Humans "
NS
NA
There are no human data to support this.
"Likely to Be Carcinogenic
to Humans "
NS
NA
Results from available animal studies are not sufficient
to support this, and no human data are available.
"Suggestive Evidence of
Carcinogenic Potential"
NS
NA
Results from available animal studies are not sufficient
to support this, and no human data are available.
"Inadequate Information
to Assess Carcinogenic
Potential"
Selected
Inhalation and
oral
No carcinogenicity studies are available that
evaluated oral and inhalation exposure.
"Not Likely to Be
Carcinogenic to Humans "
NS
NA
The available data do not support this.
NA = not applicable; NS = not selected; WOE = weight of evidence.
No data in humans are available to assess the carcinogenic potential of o-aminophenol.
o-Aminophenol did not cause liver tumors in rats in a 9-month dietary study by Miller and Miller
(1948); however, this bioassay is considered inadequate due to lack of appropriate control group,
low animal numbers, short duration, limited endpoint analysis, and use of a low dose that did not
approach the MTD. The only other studies evaluating the carcinogenic potential of
o-aminophenol included a drinking water tumor promotion assay and a bladder implantation
assay; neither of these limited studies indicated that o-aminophenol was a carcinogenic or
tumor-promoting agent (Kurata et al.. 1987; Clavson et al.. 1958). Genotoxicity data suggest
that o-aminophenol is a weak mutagen and may cause DNA damage and clastogenic effects.
Under the U.S. EPA cancer guidelines (U.S. EPA, 2005a), there is inadequate information to
assess the carcinogenic potential of o-aminophenol.
DERIVATION OF PROVISIONAL CANCER POTENCY VALUES
Derivation of quantitative estimates of cancer risk for o-aminophenol is precluded by the
absence of adequate carcinogenicity data for this compound.
19
o-Aminophenol

-------
FINAL
09-28-2016
APPENDIX A. SCREENING PROVISIONAL VALUES
For reasons noted in the main provisional peer-reviewed toxicity values (PPRTV)
document, it is inappropriate to derive provisional toxicity values for o-aminophenol. However,
information is available for this chemical which, although insufficient to support derivation of a
provisional toxicity value under current guidelines, may be of limited use to risk assessors. In
such cases, the Superfund Health Risk Technical Support Center summarizes available
information in an appendix and develops a "screening value." Appendices receive the same
level of internal and external scientific peer review as the PPRTV documents to ensure their
appropriateness within the limitations detailed in the document. Users of screening toxicity
values in an appendix to a PPRTV assessment should understand that there is considerably more
uncertainty associated with the derivation of an appendix screening toxicity value than for a
value presented in the main body of the assessment. Questions or concerns about the appropriate
use of screening values should be directed to the Superfund Health Risk Technical Support
Center.
APPLICATION OF AN ALTERNATIVE SURROGATE APPROACH
The surrogate approach allows for the use of data from related compounds to calculate
screening values when data for the compound of interest are limited or unavailable. Details
regarding searches and methods for surrogate analysis are presented in Wang et al. (2012).
Three types of potential surrogates (structural, metabolic, and toxicity-like) are identified to
facilitate the final surrogate chemical selection. The surrogate approach may or may not be
route-specific or applicable to multiple routes of exposure. In this document, it is limited to the
oral noncancer effects only, based on the available toxicity data. All information was considered
together as part of the final weight-of-evidence (WOE) approach to select the most suitable
surrogate both toxicologically and chemically.
Structural Surrogates (Structural Analogs)
An initial surrogate search focused on the identification of structurally similar chemicals
with toxicity values from the Integrated Risk Information System (IRIS), PPRTV, Agency for
Toxic Substances and Disease Registry (ATSDR), or California Environmental Protection
Agency (Cal/EPA) databases to take advantage of the well-characterized chemical-class
information. This was accomplished by searching EPA's DSSTox database (DSSTox. 2016) and
the National Library of Medicine's (NLM's) ChemlDplus database (ChemlDplus. 2016). Two
structural analogs to o-aminophenol were identified that have oral toxicity values:
w-aminophenol (U.S. EPA. 2006) andp-aminophenol (U.S. EPA. 2005b); no structural analogs
with inhalation toxicity values were identified. Table A-l summarizes the physicochemical
properties and similarity scores of the structural analogs. The DSSTox similarity scores for both
analogs were relatively high (>85% similar), while the ChemlDplus similarity scores were low
(<50%). Comparison of physicochemical properties of the surrogates to o-aminophenol suggests
that they will both behave in a manner analogous to o-aminophenol in the environment. Based
on this finding, it may be expected that exposure pathways and bioavailability from
environmental media might be comparable for the three compounds. Both m- and
/;-aminophenol are considered to be appropriate structural surrogates for o-aminophenol.
20
o-Aminophenol

-------
FINAL
09-28-2016
Table A-l. Comparison of Physicochemical Properties for
o-Aminophenol (CASRN 95-55-6) and Candidate Analogs3

o-Aminophenol
/w-Aminophenol
p-Aminophenol
Structure
nh2
6-°"
H2N^—-OH
XX
rr
h2n^^
CASRN
95-55-6
591-27-5
123-30-8
Molecular weight
109.13
109.13
109.13
DSSTox similarity score (%)b
100
86
89
ChemlDplus similarity score (%)°
100
<50
<50
Melting point (°C)
173.5d
123
187.5
Boiling point (°C)
267d
164 (at 11 mmHg)
284
Vapor pressure (mm Hg at 25°C)
0.0031 (extrapolated)6
0.0019 (estimated)0
0.00004 (extrapolated)
Henry's law constant (estimated)
(atm-m3/mole at 25°C)
2.0 x 10-10
2.0 x 10-10
2.0 x 10-10
Water solubility (mg/L)
20,000f
27,000
16,000
Log Iv
0.62g
0.21
0.04
pKa
pKai = 4.66h;
pKa2 = 9.71h
4.37
5.48
aData were gathered from the PHYSPROP database for each respective compound unless otherwise specified (U.S.
EPA. 2012c).
bDSSTox (DSSTox. 2016).
°ChemIDplus Advanced, similarity scores (ChemlDplus, 2016).
dHavnes et al. (2014).
eThe vapor pressure was extrapolated from the measured boiling point using a regression-derived equation.
fO'Neil et al. (2013).
gHSDB (2011).
'Mitchell et al. (2003).
Metabolic Surrogates
Oral absorption is high (78-100%) in laboratory animals for o-, m-, andp-aminophenol
(U.S. EPA. 1985). In general, metabolism and elimination pathways are the same for the
different aminophenol isomers. Each compound is conjugated at the phenolic group with sulfate
or glucuronide, and conjugated metabolites are excreted in the urine (Elder. 1988; U.S. EPA.
1985). A'-Acetylation also occurs, resulting in an acetamidophenol metabolite, which may be
further conjugated with sulfate or glucuronide prior to excretion in the urine (Elder. 1988; U.S.
EPA. 1985). Different rates for these processes lead to differences in the proportions of the
resulting urinary metabolites produced, primarily minor, but also including absence of
acetamidophenylsulfate following treatment with o-aminophenol and absence of
aminophenylsulfate following treatment with w-aminophenol (see Table A-2).
21
o-Aminophenol

-------
FINAL
09-28-2016
Table A-2. Percent Recovery in the Urine over 24 Hours Following a Single Oral Dose in
Rabbits (1,000 mg/Rabbit)a
Parent or Metabolite
0-Aminophenol
/w-Aminophenol
p-Aminophenol
Parent
11
0
2
Aminophenylglucuronide
52
59
45
Aminophenolsulfate
15
0
8
Acetamidophenol
2-4
12-19
13-25
Acetamidophenylglucuronide
13
5
16
Acetamidophenylsulfate
0
15
4
aU.S. EPA (1985).
The proximal toxicant(s) of aminophenol isomers' toxicity have not been clearly
identified, so it is unclear whether minor differences in the formation and distribution of
metabolites observed for aminophenol isomers could result in differences in toxicity. It has been
proposed that formation of an electrophilic compound, possibly a benzoquinone imine, is
necessary for ^-aminophenol to induce nephrotoxic effects (Elder. 1988). The acetylated
metabolite of ^-aminophenol is acetaminophen (A-acetyl-p-aminophenol) (Elder, 1988; U.S.
EPA. 1985); however, it is unclear if the toxic metabolite of acetaminophen
(iV-acetyl-^-benzoquinone imine [NAPQ1]) is formed following ^-aminophenol exposure. Renal
macromolecule binding assays indicate that either ^-aminophenol or a nonconjugated metabolite
is involved in renal toxicity, and inhibition or alteration of conjugation processes (such as
glucuronide formation) increases the severity of the renal lesions (Elder. 1988). Methemoglobin
formation following exposure top- and o-aminophenol is thought to be caused by cyclic
oxidation-reduction transformation of ferro- to ferrihemoglobin via an oxidized quinone imine
intermediate (Elder. 1988).
Due to a lack of significant differences in the absorption, distribution, metabolism, and
elimination of the aminophenol isomers, as well as insufficient data to identify any difference in
proximal toxicants, both m- and ^-aminophenol are considered appropriate metabolic surrogates
for o-aminophenol.
Toxicity-Like Surrogates
Table A-3 summarizes the available oral and inhalation human health assessment values
and acute toxicity data for o-aminophenol and the compounds identified as potential surrogates.
Acute lethality studies for o-aminophenol provide oral median lethal dose (LD50) values in rats
and mice. The rat LD50 data suggest that ^-aminophenol may be more acutely toxic than o- or
w-aminophenol.
22
o-Aminophenol

-------
FINAL
09-28-2016
Table A-3. Comparison of Available Human Health Assessment Values and Acute
Toxicity Data for o-Aminophenol (CASRN 95-55-6) and Candidate Analogs

o-Aminophenol
/w-Aminophenol
/7-Aminophenol
Structure

H2N^—-OH
u
rr
h2n^^
CASRN
95-55-6
591-27-5
123-30-8
Repeat-dose toxicity—oral, subchronic
POD (mg/kg-d)
NA
80
50
POD type
NA
NOAF.I.
NOAF.I.
UFC
NA
300
300
p-RfD (mg/kg-d)
NA
3 x 10-1
2 x 10-1
Critical effects
NA
Reduced body weight and tremors
Increased severity of nephrosis
Other effects
NA
Increased total bilirubin and
reticulocyte ratio; thyroid follicular
cell hypertrophy
Reduced body weight
concomitant with reduced food
intake, anemia
Species
NA
Rat (neonatal)
Rat (males, females)
Duration
NA
18 d
90 d
Route
NA
Oral (gavage)
Oral (diet)
Notes
NA
Newborn rats were more sensitive
than older rats examined in 4- and
13-wk studies. Effects noted at
higher doses in older rats included
hemolytic anemia, potential liver
toxicity (elevated ALT), and
potential kidney toxicity (increased
BUN in females, hyaline droplets
in males)
Hyperactivity and convulsions,
reductions in food consumption
and body weight, mild
hematological changes, and
embryo/fetotoxic effects
Source
NA
U.S. EPA (2006)
U.S. EPA (2005b)
Repeat-dose toxicity—oral, chronic
POD (mg/kg-d)
NA
240
50
POD type
NA
NOAF.I.
NOAEL
UFC
NA
3,000
3,000
p-RfD (mg/kg-d)
NA
8 x 10-2
2 x 10-2
Critical effects
NA
Hemolytic anemia
Increased severity of nephrosis
Other effects
NA
Thyroid follicular cell hypertrophy;
reduced body weight concomitant
with reduced food intake
Reduced body weight
concomitant with reduced food
intake, anemia
Species
NA
Rat (female)
Rat (males, females)
Duration
NA
90 d
90 d
23
o-Aminophenol

-------
FINAL
09-28-2016
Table A-3. Comparison of Available Human Health Assessment Values and Acute
Toxicity Data for o-Aminophenol (CASRN 95-55-6) and Candidate Analogs

o-Aminophenol
/w-Aminophenol
/7-Aminophenol
Route
NA
Oral (diet)
Oral (diet)
Notes
NA
NA
NA
Source
NA
U.S. EPA (2006)
U.S. EPA (2005b)
Acute toxicity
Rat oral LD50
(mg/kg)
951
924
375
Toxicity target
Tremor, cyanosis
Excitement, spastic paralysis
Muscle weakness, cyanosis
Mouse oral LD50
(mg/kg)
800
401
NA
Toxicity target
Tremor, cyanosis
Excitement, spastic paralysis
NA
Rat inhalation LC50
(mg/m3)
NA
1,162
>5
Toxicity target
NA
Excitement, spastic paralysis
NR
Mouse inhalation
LC50 (mg/m3)
NA
NA
NA
Toxicity target
NA
NA
NA
Reference
ChemlDolus (2016)
ChemlDolus (2016)
ChemlDolus (2016)
ALT = alanine aminotransferase; BUN = blood urea nitrogen; LC50 = median lethal concentration; LD50 = median
lethal dose; NA = not applicable; NOAEL = no-observed-adverse-effect level; NR = not reported; POD = point of
departure; p-RfD = provisional reference dose; UFC = composite uncertainty factor.
The repeat-dose oral database for o-aminophenol consists of a single unpublished
short-term-duration study in rats (Eastman Kodak. 1979); this study is of limited use for risk
assessment due to the use of one sex (males), small group sizes (five/group), and short duration
(12 days). Based on acute and short-term-duration animal studies, the red blood cells (RBCs)
and potentially kidney and liver are targets of o-aminophenol toxicity (Rankin et al.. 1996;
Harrison and Jollow. 1987; Itoh. 1987; Eastman Kodak. 1979).
The critical effects identified following oral exposure to w-aminophenol included
reduced body weight and tremors in neonatal rats exposed via gavage for 18 days
(lowest-observed-adverse-effect level [LOAEL] 240 mg/kg-day) and hemolytic anemia in adult
female rats exposed via diet for 90 days (LOAEL 900 mg/kg-day). Other effects noted with
repeated oral exposure included increased total bilirubin and reticulocyte ratio and thyroid
follicular cell hypertrophy, as well as potential liver and kidney toxicity thought to be secondary
to hemolytic anemia (modest increases in alanine aminotransferase [ALT], slight increases in
blood urea nitrogen [BUN] in females, and hyaline droplets in males) (U.S. EPA. 2006). The
subchronic provisional reference dose (p-RfD) of 0.3 mg/kg-day is based on a point of departure
(POD) of 80 mg/kg-day for neonatal effects in the 18-day gavage study, and the chronic p-RfD
of 8 x 10~2 mg/kg-day is based on a POD of 240 mg/kg-day for hemolytic anemia in adult
female rats (U.S. EPA, 2006).
24
o-Aminophenol

-------
FINAL
09-28-2016
The critical effect identified following oral exposure to/^-aminophenol was increased
severity of nephrosis in male and female rats exposed via diet for 90 days (LOAEL
150 mg/kg-day). Other effects noted at higher exposure levels included anemia and reduced
body weight concomitant with reduced food intake (U.S. EPA. 2005b). Both the subchronic
p-RfD of 0.2 mg/kg-day and the chronic p-RfD of 2 x 10 2 mg/kg-day are based on a POD of
50 mg/kg-day for nephrotoxic effects (U.S. EPA, 2005b).
In a comparison of relative toxicities, the subchronic POD value for /^-aminophenol is
fourfold lower than that of w-aminophenol. The lower reference value for /^-aminophenol is
based on adverse renal effects that occurred at lower doses than anemia following exposure for
90 days. In animals exposed to w-aminophenol for 90 days in the diet, anemia was observed in
the absence of renal toxicity, and in young rats in a 28-day gavage study, doses that caused
hemolytic anemia produced evidence of only mild kidney damage. Therefore, /^-aminophenol
appears to be a more potent renal toxicant than w-aminophenol. While the potency may differ
among aminophenol isomers, evidence for similar toxicity targets (kidney and RBC) suggests
that both m- and /^-aminophenol are appropriate toxicity-like surrogates for o-aminophenol for
oral exposure.
No repeat-exposure inhalation studies were available for o-, m-, or /^-aminophenol and
inhalation toxicity values were not derived.
Weight-of-Evidence Approach
A WOE approach is used to evaluate information from potential candidate surrogates as
described by Wang et al. (2012). Commonalities in structural/physicochemical properties,
toxicokinetics, metabolism, toxicity, or mode of action between potential surrogates and
chemical(s) of concern are identified. Emphasis is given to toxicological and/or toxicokinetic
similarity over structural similarity. Surrogate candidates are excluded if they do not have
commonality or demonstrate significantly different physicochemical properties and toxicokinetic
profiles that set them apart from the pool of potential surrogates and/or chemical(s) of concern.
From the remaining potential surrogates, the most appropriate surrogate (most biologically or
toxicologically relevant analog chemical) with the highest structural similarity and/or most
conservative toxicity value is selected.
The structural surrogate analysis indicated that both m- and ^-aminophenols have
relatively high similarity scores (>85%) compared to o-aminophenol based on the DSSTox
database search. A comparison of physicochemical properties of these surrogates to
o-aminophenol suggests that these surrogate chemicals share similar physicochemical properties
to o-aminophenol. Metabolic surrogate analysis indicated that, in general, the metabolism and
elimination pathways are the same for the different aminophenol isomers. However, there are
insufficient data to identify the proximal toxicant of aminophenols to inform a more refined
selection of a particular metabolic surrogate. Based on the acute and short-term-duration animal
studies with o-aminophenol, the RBC, liver, and kidney are potential toxicity targets. The two
surrogate chemicals also share the same toxicity targets (i.e., RBC and kidney), and
w-aminophenol also affects the liver; nevertheless, relative potency among the aminophenol
isomers might differ in each of these target tissues. Thus, these analyses suggest that both
candidate analogs, m- and /^-aminophenol, are acceptable as structural, metabolic, and
toxicity-like surrogates.
25
o-Aminophenol

-------
FINAL
09-28-2016
For this assessment, />-aminophenol was selected over w-aminophenol as the surrogate
for o-aminophenol for both the subchronic and chronic oral toxicity values because the POD
based on /;-aminophenol will provide a more conservative/health-protective toxicity value for
both subchronic and chronic oral exposure to o-aminophenol.
ORAL TOXICITY VALUES
Derivation of a Screening Subchronic Provisional Reference Dose
Based on the overall WOE presented in this PPRTV assessment, />-aminophenol is
selected as the surrogate for o-aminophenol for the screening subchronic p-RfD. The study used
for the subchronic p-RfD for /;-aminophenol is a 13-week (90 days) dietary study in male and
female weanling Sprague-Dawley (S-D) rats by Burnett et al. (1989) as cited in U.S. EPA
(2005b). The PPRTV report for/;-aminophenol (U.S. EPA. 2005b) described this study as
follows:
In order to investigate the effects of longer-term oral exposure, a combined
subchronic feeding, teratology, and dominant lethal study ofp-aminophenol was
conducted in rats. Groups of 40 male and 45 female weanling Sprague-Dawley
rats were fed diets containing 0, 0.07, 0.20, or 0.70% of p-aminophenol (>98.1%
purity) in their diet for 13 weeks (Burnett et al, 1989). At that time, 10 males and
10 females of each group were sacrificedfor toxicity evaluation, and 25 females
fi'om each group were removedfi'om the test diets and mated with untreated
males. After mating, the pregnant females were returned to their test diets
throughout gestation and sacrificed on gestation day 20 for fetal examinations.
Males not sacrificed at week 13 were continued on their test diets until week 20,
when 20 males fi'om each group were removedfi'om the test diets and mated with
untreatedfemales in a dominant lethal assay until their sacrifice on week 2 7. The
remaining 10 males and 10 females fi'om each group were maintained on their
test diets until sacrifice on week 17. Based on food consumption and body weight
data presented graphically in the paper, doses were approximately 0, 50, 150,
and 560 mg/kg-day in males and 0, 60, 175, and 620 mg/kg-day in females.
Animals were observed daily for general condition and monitored weekly
for signs of toxicity, body weight, andfeed consumption (Burnett et al, 1989). At
6 weeks, blood was collectedfi'om 5 males and 5 females fi'om the high dose
group (0.70%) for methemoglobin analyses. At week 12, urine was collectedfi'om
10 males and 10 females selected from each group for bacterial mutagenicity
testing; and at week 13, the same 10 rats/group were sacrificed and blood
collectedfor hematology and clinical chemistry analysis. At necropsy, the major
organs were weighed, and a complete histopathological examination was
performedfor animals fi'om the control and high dose groups. The liver, kidney,
urinary bladder, and gross lesions were also examinedfi'om animals in the
low- and mid-dose groups. The same procedures were used to collect blood and
autopsy rats sacrificed at end of the 27 week study.
Hyperactivity and convulsions were noted in a few of the females consuming
the high-dose test diet after 6 weeks on study (Burnett et al, 1989). No
treatment-related deaths occurred (one low-dose female died of unknown causes).
Food consumption was markedly lower than controls in both males andfemales
26
o-Aminophenol

-------
FINAL
09-28-2016
from the high-dose group during the first week of the study, and remained
significantly lower than controls for most of the study in both sexes. Body weights
of both males and females in this group were significantly lower than controls
throughout the study, with deficits of 10-15% in males and 15-20% in females
after week 5. Food consumption and body weight were similar to controls in the
low and mid-dose groups. Hematology analyses showed statistically significant
decreases in red blood cell counts (-10%) and hemoglobin level (-5%) in
high-dose females at 13 weeks, but not at 27 weeks. Other hematology and
clinical chemistry findings were reportedly unremarkable (data not presented in
paper). The assay for methemoglobin in high-dose rats showed no difference
from controls. Increased relative weights were observedfor several organs in
high-dose males andfemales, secondary to the decrease in body weight at this
dose. Statistically significant changes in other organ weights (increased absolute
and relative pituitary weight in low- and mid-dose females at 13 weeks, and
increased absolute heart weight in low-dose males at 13 weeks) were not
considered by the researchers to be treatment-related. No gross lesions were
seen at autopsy. Microscopic evaluation revealed nephrosis characterized by
cytoplasmic eosinophilic droplets in the tubular epithelial cells of male and
female rats of all groups, but with a dose-related increase in incidence and or
severity (Table 1). In males, the lesions were similar to glomeridonephropathy
typical of aging rats (albeit more severe in the treated groups), while in females
the droplets were smaller and intensely brown. Statistical analysis of the data in
Table 1 was performedfor this review. The Jonckheere-Terpstra trend test for
ordered categorical data showed statistically significant (p< 0.001) increases in
severity of nephrosis with increasing dose in both males andfemales, using the
13-week data for all 4 dose groups. Pairwise comparisons using the same test
showed significant (p< 0.005) differences from controls in mid- and high-dose
males and high-dose females. No other treatment-related histopathological
changes were noted. A LOAEL of 150 mg/kg-day (0.2%) andNOAEL of
50 mg/kg-day (0.07%) is identifiedfrom this study, based on increased severity of
nephrosis in males.
A similar study (Re et al., 1984) with a combined sub chronic-duration feeding and
teratology of w-aminophenol reported similar effects, but these effects occurred at a higher dose
level. In this study, four groups of 35 S-D female rats were fed diets containing 0, 0.13, 0.25, or
0.98% w-aminophenol. From reported body-weight and food consumption data, average doses
are estimated to be about 0, 120, 240, or 900 mg/kg-day.
The PPRTV report for w-aminophenol (U.S. EPA. 2006) described findings from this
study as follows:
This study identified a LOAEL of900 mg/kg-day (0.98% in the diet) based on evidence of
hemolytic anemia (increased incidence and or severity of iron-positive pigmentation in
the spleen, liver and kidneys, along with reduced red blood cell count and hemoglobin).
Body weight was lower than controls in both the mid- and high-dose groups, but the
change was minimal in the mid-dose group and was secondary to reduced food intake at
both doses (suggesting an organoleptic, rather than toxic, effect). Histological changes in
the thyroid consistent with hypertrophy (reducedfollicle size and increasedfollicular
27
o-Aminophenol

-------
FINAL
09-28-2016
epithelial cell height) were reported in both the mid- and high-dose groups (incidence
reported only as "several" animals in the mid-dose group). Thyroid hormone
measurements were not made. However, reported changes in thyroid weight were not
consistent with a hyperthyroid effect. There was an increase in relative, but not absolute
thyroid weight in the high dose group, apparently secondary to the decrease in body
weight at this dose. In the mid-dose group, absolute and relative thyroid weights were
actually reduced. Therefore, the thyroid effects are considered to be adaptive rather than
adverse. Thus, the 240 mg/kg-day dose (0.25%) is considered a NOAEL for
m-aminophenol.
Therefore, a POD based on/;-aminophenol will provide a more
conservative/health-protective toxicity value for both subchronic and chronic oral exposure to
o-aminophenol. The selection of a POD of 50 mg/kg-day is also supported by a 3-month oral
toxicity study [Fournier (1981) as cited in SCCS (2010)1 in rats treated with 50 mg/kg-day
o-aminophenol. Based on a secondary report from the Scientific Committee on Consumer Safety
(SCCS. 2010). clinical observation, body-weight gain, hematological parameters, blood
biochemistry, and urine examination revealed no difference between the treated and the control
group. Histopathological examination showed broncho-pulmonary injuries, which did not permit
to affirm the toxicity of the o-aminophenol. In addition, a developmental toxicity study
[Boutemy et al. (1981) as cited in SCCS (2010)1 in rats treated with o-aminophenol identified a
NOAEL of 70 mg/kg-day for maternal and developmental toxicity.
The SCCS report (SCCS. 2010) also reported two 30-day oral toxicity studies in rats
treated with o-aminophenol. One study [Boutemy (1989) as cited in SCCS (2010)1 identified a
LOAEL of 20 mg/kg-day (the lowest dose tested) based on increased vacuolization of the
urothelium of the bladder in males and females, and observed renal cells in the urine of male
rats. The other study [Coleman et al. (1989) as cited in SCCS (2010)1 identified a NOAEL of
5 mg/kg-day and a LOAEL of 15 mg/kg-day due to increased thyroid weight. These findings
from 30-day studies were inconsistent with the no toxicity effect found in rats treated with
50 mg/kg-day o-aminophenol for 3 months [Fournier (1981) as cited in SCCS (2010)1, which
also included a pathological examination of these target organs/tissues. The toxicity data
summarized in the SCCS report are only presented here as supportive evidence because the
report has limited information available and makes no final conclusion on the safety of
o-aminophenol (SCCS. 2010). Further, the studies cited in the SCCS report were not available
for review.
Therefore, a POD based on/;-aminophenol is considered appropriate for deriving
screening p-RfDs. The critical effect for the subchronic toxicity portion of the study of
/>-aminophenol was increased severity of nephrosis in male rats; the NOAEL of 50 mg/kg-day
for this effect was used as the POD. The subchronic p-RfD for /;-aminophenol was derived
using a composite uncertainty factor (UFc) of 300 reflecting 10-fold uncertainty factor values for
interspecies extrapolation (UFa), a 10-fold for intraspecies variability (UFh), and a 3-fold for
database uncertainties (UFd, reflecting lack of a second species in the study). Wang et al. (2012)
indicated that the uncertainty factors applied in deriving a surrogate toxicity value for the
chemical of concern should be the same as those applied to the selected analog unless additional
information is available. However, the EPA endorses body-weight scaling to the 3/4 power
(i.e., BW3/4) as a default to extrapolate toxicologically equivalent doses of orally administered
agents from all laboratory animals to humans for the purpose of deriving an RfD from effects
28
o-Aminophenol

-------
FINAL
09-28-2016
that are not portal-of-entry (U.S. EPA. 2011b). Therefore the surrogate POD was converted into
a human equivalent dose (HED) and the UFa was reduced to a 3 (rather than 10-fold) for the
derivation of a screening subchronic p-RfD for o-aminophenol. Additionally, a 10-fold UFd
(rather than 3-fold) was applied due to lack of useful repeat-dose toxicity studies for
o-aminophenol (Table A-4).
Following U.S. EPA (2011b) guidance, the POD for the 13-week study in rats exposed to
/;-aminophenol is converted to an HED through the application of a dosimetric adjustment factor
(DAF) derived as follows:
DAF = (BWa14 - BWh14)
where:
DAF = dosimetric adjustment factor
BWa = animal body weight
BWh = human body weight
Using a reference BWa of 0.25 kg for rats and a reference BWh of 70 kg for humans (U.S.
EPA. 1988). the resulting DAF is 0.24. Applying this DAF to the NOAEL identified in the
13-week rat study yields a NOAEL (HED) as follows:
POD (HED) = NOAEL (mg/kg-day) x DAF
= 50 mg/kg-day x 0.24
= 12 mg/kg-day
Using the surrogate POD (HED), the screening subchronic p-RfD for o-aminophenol is
derived as follows:
Screening Subchronic p-RfD = Surrogate POD (HED) ^ UFc
= 12 mg/kg-day -^300
= 4 x 10"2 mg/kg-day
Table A-4 summarizes the uncertainty factors for the screening subchronic p-RfD for
o-aminophenol.
29
o-Aminophenol

-------
FINAL
09-28-2016
Table A-4. Uncertainty Factors for the Screening Subchronic p-RfD for
o-Aminophenol (CASRN 95-55-6)
UF
Value
Justification
UFa
3
A UFa of 3 (10°5) is applied to account for uncertainty associated with extrapolating from
animals to humans when a cross-species dosimetric adjustment (HED calculation) is performed.
UFh
10
A UFh of 10 is applied for interindividual variability to account for human-to-human variability
in susceptibility in the absence of quantitative information to assess the toxicokinetics and
toxicodynamics of o-aminophenol in humans.
UFd
10
A UFd of 10 is applied. The repeat-dose oral database for o-aminophenol consists of a single
study that is limited due to use of one sex (males), small group sizes (five/group), and short
duration (12 d). Therefore, the POD is based on a surrogate chemical.
UFl
1
A UFl of 1 is applied because the POD is a NOAEL.
UFs
1
A UFS of 1 is applied because a subchronic-duration study was selected as the principal study.
UFC
300
Composite UF = UFA x UFH x UFD x UFL x UFS.
HED = human equivalent dose; NOAEL = no-observed-adverse-effect level; POD = point of departure;
UF = uncertainty factor.
Derivation of a Screening Chronic Provisional Reference Dose
Based on the overall surrogate approach presented in this PPRTV assessment,
/;-aminophenol is selected as the surrogate for o-aminophenol for the screening chronic p-RfD.
The chronic p-RfD for />aminophenol was derived by adding an additional UF of 10 to
extrapolate from subchronic to chronic duration (UFs); the UFc was 3,000. Similarly, the
screening chronic p-RfD for o-aminophenol is derived by factoring in a UFs of 10 to the
screening subchronic p-RfD of 4 x 10 2 mg/kg-day derived above. The screening chronic p-RfD
for o-aminophenol is derived as follows:
Screening Chronic p-RfD = Surrogate POD (HED) ^ UFc
= 12 mg/kg-day ^ 3,000
= 4 x 10"3 mg/kg-day
Table A-5 summarizes the uncertainty factors for the screening chronic p-RfD for
o-aminophenol.
30
o-Aminophenol

-------
FINAL
09-28-2016
Table A-5. Uncertainty Factors for the Screening Chronic p-RfD for
o-Aminophenol (CASRN 95-55-6)
UF
Value
Justification
UFa
3
A UFa of 3 (10"5) is applied to account for uncertainty associated with extrapolating from animals
to humans when cross-species dosimetric adjustment (HED calculation) is performed.
UFh
10
A UFh of 10 is applied for interindividual variability to account for human-to-human variability in
susceptibility in the absence of quantitative information to assess the toxicokinetics and
toxicodynamics of o-aminophenol in humans.
UFd
10
A UFd of 10 is applied. The repeat-dose oral database for o-aminophenol consists of a single
study that is limited due to use of one sex (males), small group sizes (five/group), and short
duration (12 d). Therefore, the POD is based on a surrogate chemical.
UFl
1
A UFl of 1 is applied because the POD is a NOAEL.
UFS
10
A UFS of 1 is applied because a subclironic-duration study was selected as the principal study.
UFC
3,000
Composite UF = UFA x UFH x UFD x UFL x UFS.
HED = human equivalent dose; NOAEL = no-observed-adverse-effect level; POD = point of departure;
UF = uncertainty factor.
Derivation of Screening Provisional Reference Concentrations
No subchronic- or chronic-duration inhalation studies examining the effects of candidate
analogs for o-aminophenol in humans or animals have been located (U.S. EPA. 2006. 2005b).
precluding derivation of provisional reference concentrations (p-RfCs) for o-aminophenol based
on an alternative surrogate approach.
31
o-Aminophenol

-------
FINAL
09-28-2016
APPENDIX B. REFERENCES
ACGIH (American Conference of Governmental Industrial Hygienists). (2015). 2015 TLVs and
BEIs. Based on the documentation of the threshold limit values for chemical substances
and physical agents and biological exposure indices. Cincinnati, OH.
http://www.acgih.org/forms/store/ProductFormPublic/2015-tlvs-and-beis
Akazawa. M; Takasaki. M; Tomoda. A. (2000). Oscillatory oxido-reductive reaction of
intracellular hemoglobin in human erythrocyte incubated with o-aminophenol. Tohoku J
Exp Med 192: 301-312. http://dx.doi.org/10.1620/tiem.192.301
Angel A: Rogers. KJ. (1972). An analysis of the convulsant activity of substituted benzenes in
the mouse. Toxicol Appl Pharmacol 21: 214-229.
AT SDR (Agency for Toxic Substances and Disease Registry). (2016). Minimal risk levels
(MRLs). March 2016. Atlanta, GA: Agency for Toxic Substances and Disease Registry
(ATSDR). Retrieved from http://www.atsdr.cdc.gov/mrls/index.asp
Bartsch. H; Ohshima. H; Pignatelli. B; Malaveille. C: Friesen. M. (1991). Nitrite-reactive
phenols present in smoked foods and amino-sugars formed by the Maillard Reaction as
precursors of genotoxic arenediazonium ions or nitroso compounds. In H Hayatsu (Ed.),
Mutagens in Food: Detection and Prevention (pp. 87-100). Boca Raton, FL: CRC Press,
Inc.
Bogaiewski. J; Bogaiewska. G. (1982). Comparison of determinations of chromosomal
aberrations in anaphases and metaphases. Mutat Res 173: 173-174.
Burnett. CM: Goldenthal. ES. (1988). Multigeneration reproduction and carcinogenicity studies
in Sprague-Dawley rats exposed topically to oxidative hair-coloring formulations
containing p-phenylenediamine and other aromatic amines. Food Chem Toxicol 26: 467-
474. http://dx.doi.org/doi: 10.1016/0278-6915(88)90059-2
Cal/EPA (California Environmental Protection Agency). (2011). Hot spots unit risk and cancer
potency values. Appendix A. Sacramento, CA: Office of Environmental Health Hazard
Assessment.
http://standards.nsf.org/apps/group public/download.php?document id= 19121
Cal/EPA (California Environmental Protection Agency). (2014). All OEHHA acute, 8-hour and
chronic reference exposure levels (chRELs) as of June 2014. Sacramento, CA: Office of
Health Hazard Assessment, http://www.oehha.ca.gov/air/allrels.html
Cal/EPA (California Environmental Protection Agency). (2016a). Chemicals known to the state
to cause cancer or reproductive toxicity July 15, 2016. (Proposition 65 list). Sacramento,
CA: California Environmental Protection Agency, Office of Environmental Health
Hazard Assessment, http://oehha.ca.gov/proposition-65/proposition-65-list
Cal/EPA (California Environmental Protection Agency). (2016b). OEHHA toxicity criteria
database [Database], Sacramento, CA: Office of Environmental Health Hazard
Assessment. Retrieved from http://www.oehha.ca. gov/tcdb/index.asp
Calder. IC: Funder. CC: Green. CR. (1971). Comparative nephrotoxicity of aspirin and
phenacetin derivatives. BMJ 4: 518-521.
CCRIS (Chemical Carcinogenesis Research Information System). (1993). o-Aminophenol.
CASRN: 95-55-6: National Cancer Institute. National Library of Medicine. Retrieved
from http://toxseektoxnet.nlm.nih.gov/newtoxnet/ccris.htm
ChemlDplus. (2016). ChemlDplus - a TOXNET database. Bethesda, MD: National Institutes of
Health, U.S. Library of Medicine. Retrieved from
http: //chem. si s. nlm. nih. gov/chemi dplus/
32
o-Aminophenol

-------
FINAL
09-28-2016
Clavson. DB; JulK JW; Bonser. GM. (1958). The testing of ortho-hydroxy-amines and related
compounds by bladder implantation and a discussion of their structural requirements for
carcinogenic activity. Br J Cancer 12: 222-230. http://dx.doi.org/10.1038/bic.1958.26
De Flora. S: Zanacchi. P; Camoirano. A: Bennicelli. C: Badolati. GS. (1984). Genotoxic activity
and potency of 135 compounds in the Ames reversion test and in a bacterial DNA-repair
test [Review], Mutat Res 133: 161-198. http://dx.doi.org/10.1016/0165-1110(84)90016-2
Degawa. M; Shoii. Y; Masuko. K; Hashimoto. Y. (1979). Mutagenicity of metabolites of
carcinogenic aminoazo dyes. Cancer Lett 8: 71-76. http://dx.doi.org/10.1016/03Q4-
3835(79)90025-9
DSSTox (Distributed Structure-Searchable Toxicity). (2016). DSSTox database [Database],
Research Triangle Park, NC: U.S. Environmental Protection Agency, National Center for
Computational Toxicology. Retrieved from http://www.epa.gov/ncct/dsstox/
Eastman Kodak (Eastman Kodak Company). (1979). Initial submission: Summary of the basic
toxicity of o-aminophenol with cover letter dated 09/21/92. (TSC ATS/440980.
OTS0555965. Doc #88-920010697). Rochester, NY.
https://ntrl.ntis.gov/NTRL/dashboard/searchResults.xhtml?searchQuery=OTS0555965
Eckert. KG: Ever. P. (1983). Differences in the reactions of isomeric ortho- and para-
aminophenols with hemoglobin. Biochem Pharmacol 32: 1019-1027.
Elder. RL. (1988). Final report on the safety assessment of p-Aminophenol m-Aminophenol, and
o-Aminophenol. J Am Coll Toxicol 7: 279-334.
Harrison. JH. Jr; Jollow. DJ. (1987). Contribution of aniline metabolites to aniline-induced
methemoglobinemia. Mol Pharmacol 32: 423-431.
Haworth. S: Lawlor. T; Mortelmans. K; Speck. W: Zeiger. E. (1983). Salmonella mutagenicity
test results for 250 chemicals. Environ Mutagen 5: 3-142.
http://dx.doi.org/10.1002/em.28600507Q3
Havnes. WM; Lide. PR: Bruno. TJ. (2014). 2-Aminophenol. In WM Haynes; DR Lide; TJ
Bruno (Eds.), CRC Handbook of Chemistry and Physics (95th ed., pp. 3-22). Boca Raton,
FL: Taylor & Francis Group.
HSDB (Hazardous Substances Data Bank). (2011). 2-Aminophenol. CASRN: 95-55-6. Bethesda,
MD: National Library of Medicine, National Toxicology Program.
http://toxnet.nlm.nih.gov/
IARC (International Agency for Research on Cancer). (2015). IARC Monographs on the
evaluation of carcinogenic risk to humans. Geneva, Switzerland: International Agency for
Research on Cancer, WHO. http://monographs.iarc.fr/ENG/Monographs/PDFs/index.php
IPCS (International Programme on Chemical Safety). (2016). INCHEM: Chemical safety
information from intergovernmental organizations [Database]: World Health
Organization. Canadian Centre for Occupational Health and Safety. Inter-Organization
Programme for the Sound Management of Chemicals. Retrieved from
http://www.inchem.org/
Itoh. I. (1987). Experimental studies on the toxicity of three kinds of isomeric aminophenols.
Tokyo Joshi Ika Daigaku Zasshi 57: 1655-1666.
Kanaya, N. (1996). Activation of aniline by extracts from plants and induction of chromosomal
damages in Chinese hamster ovary cells. Genes Genet Syst 71: 319-322.
http://dx.doi.org/10.1266/ggs.71.319
Katoh. J: Okuno. F; Watanabe. K; Obana. S: Sugai. T. (1986). [Allergic contact dermatitis from
hair-dyes and the incidence of positive reactions in patch tests with the ingredients]. Skin
Research 28: 149-154.
33
o-Aminophenol

-------
FINAL
09-28-2016
Kirchner. G; Bayer. U. (1982). Genotoxic activity of the aminophenols as evidenced by the
induction of sister chromatid exchanges. Hum Toxicol 1: 387-392.
Kurata. Y; Tsuda. H; Sakata. T; Yam ash it a. T; Ito. N. (1987). Reciprocal modifying effects of
isomeric forms of aminophenol on induction of neoplastic lesions in rat liver and kidney
initiated by N-ethyl-N-hydroxyethylnitrosamine. Carcinogenesis 8: 1281-1285.
http://dx.doi.Org/10.1093/carcin/8.9.1281
Lavoi e. E; Tulle v. L; Fow. E; Hoffmann. D. (1979). Mutagenicity of aminophenyl and
nitrophenyl ethers, sulfides, and disulfides. MutatRes 67: 123-131.
Matsunaga. K; Havakawa. R; Suzuki. M; Kawaguchi. K; Ogino. Y; Hirose. O. (1989). [Allergic
contact dermatitis in hairdressers and barbers causative factors and chemicals]. Skin
Research 31: 167-175.
Matsunaga, K; Hosokawa, K; Suzuki, M; Arima, Y; Havakawa, R. (1988). Occupational allergic
contact dermatitis in beauticians. Contact Derm 18: 94-96.
http://dx.doi.Org/10.llll/i.1600-0536.1988.tb02747.x
Miller. JA; Miller. EC. (1948). The carcinogenicity of certain derivatives of p-
dimethylaminozobenz in the rat. J Exp Med 87: 139-156.
Mitchell. SC: Carmichael. P; Waring. R. (2003). Aminophenols. In Kirk-Othmer encyclopedia of
chemical technology. Hoboken, NJ: John Wiley and Sons, Inc.
http://dx.doi.org/10.1002/0471238961.01130914130920Q3.a01.pub2
Mitchell. SC: Waring. RH. (2012). Aminophenols. In Ullmann's Encyclopedia of Industrial
Chemistry. Weinheim, Germany: Wiley VCH.
http://dx.doi.org/10.1002/143560Q7.a02 099
Newton. JF; Kuo. CH; Gemborys. MW: Mudge. GH; Hook. JB. (1982). Nephrotoxicity of p-
aminophenol, a metabolite of acetaminophen, in the fischer 344 rat. Toxicol Appl
Pharmacol 65: 336-344. http://dx.doi.org/10.1016/0041 -008X(82)90017-5
NIOSH (National Institute for Occupational Safety and Health). (2016). NIOSH pocket guide to
chemical hazards. Index of chemical abstracts service registry numbers (CAS No.).
Atlanta, GA: Center for Disease Control and Prevention, U.S. Department of Health,
Education and Welfare, http://www.cdc.gov/niosh/npg/npgdcas.html
Nishimura, N; Oshima, H. (1983). [Mutagenicity of pentachlorophenol, dinitro-o-cresol and their
related compounds], Sangyo Igaku 25: 510-511.
NTP (National Toxicology Program). (2014). Report on carcinogens. Thirteenth edition.
Research Triangle Park, NC: U.S. Department of Health and Human Services, Public
Health Service, http://ntp.niehs.nih.gov/pubhealth/roc/rocl3/index.html
O'Neil. MJ: Heckelman. PE; Dobbelaar. PH; Roman. KJ. (2013). o-Aminophenol. In The Merck
index (15th ed.). Cambridge, UK: The Royal Society of Chemistry.
Ohkuma. Y; Kawanishi. S. (2001). Oxidative DNA damage induced by a metabolite of
carcinogenic o-anisidine: enhancement of DNA damage and alteration in its sequence
specificity by superoxide dismutase. Arch Biochem Biophys 389: 49-56.
http://dx.doi.org/10.1006/abbi.2001.23Q2
Ohshima. H; Friesen. M; Malaveille. C: Brouet. I: Hautefeuille. A: Bartsch. H. (1989).
Formation of direct-acting genotoxic substances in nitrosated smoked fish and meat
products identification of simple phenolic precursors and phenyldiazonium ions as
reactive products. Food Chem Toxicol 27: 193-204.
34
o-Aminophenol

-------
FINAL
09-28-2016
OSHA (Occupational Safety & Health Administration). (2006). Table Z-l: Limits for air
contaminants. Occupational safety and health standards, subpart Z, toxic and hazardous
substances. (OSHA standard 1910.1000, 29 CFR). Washington, DC: U.S. Department of
Labor.
http://www.osha.gov/pls/oshaweb/owadisp.show document?p table=STANDARDS&p
id=9992
OSHA (Occupational Safety & Health Administration). (2011). Air contaminants: Occupational
safety and health standards for shipyard employment, subpart Z, toxic and hazardous
substances. (OSHA Standard 1915.1000). Washington, DC: U.S. Department of Labor.
https://www.osha.gov/pls/oshaweb/owadisp.show document?p table=STANDARDS&p
id=10286
Rankin, GO; Beers, KW; Nicoll, DW; Anestis, DK; Hong, SK; Hubbard, JL; Ball, JG;
Valentovic. MA: Brown. PI. (1996). Nephrotoxic potential of 2-amino-5-chlorophenol
and 4-amino-3-chlorophenol in Fischer 344 rats: comparisons with 2- and 4-chloroaniline
and 2- and 4-aminophenol. Toxicology 108: 109-123.
Re. TA; Loehr. RF; Rodriguez. SC: Rodwell. DE; Burnett. CM. (1984). Results of teratogenicity
testing of m-aminophenol in Sprague-Dawley rats. Fundam Appl Toxicol 4: 98-104.
Rutkowski, JV; Ferm, VH. (1982). Comparison of the teratogenic effects of the isomeric forms
of aminophenol in the Syrian golden hamster. Toxicol Appl Pharmacol 63: 264-269.
http ://dx. doi .org/10.1016/0041 -008X(82)90048-5
SCCS (Scientific Committee on Consumer Safety). (2010). Opinion on o-aminophenol. COLIPA
no A14. Directorate-General for Health & Consumers, European Commission.
http://ec.europa.eu/health/scientific committees/consumer safetv/docs/sccs o 025.pdf
Smith, RP; Alkaitis, AA; Shafer, PR. (1967). Chemically induced methemoglobinemias in the
mouse. Biochem Pharmacol 16: 317-328.
Thompson. CZ; Hill. LE; Epp. JK; Probst. GS. (1983). The induction of bacterial mutation and
hepatocyte unscheduled DNA synthesis by monosubstituted anilines. Environ Mutagen 5:
803-811. http://dx.doi.org/10.1002/em.28600506Q5
U.S. EPA (U.S. Environmental Protection Agency). (1985). Health and environmental effects
profile for aminophenols. (EPA/600/X-85/398). Cincinnati, OH: Environmental Criteria
and Assessment Office.
https://ntrl.ntis.gov/NTRL/dashboard/searchResults.xhtml?searchOuery=PB88173612
U.S. EPA (U.S. Environmental Protection Agency). (1988). Recommendations for and
documentation of biological values for use in risk assessment (pp. 1-395). (EPA/600/6-
87/008). Cincinnati, OH: U.S. Environmental Protection Agency, Office of Research and
Development, Office of Health and Environmental Assessment.
http://cfpub.epa. gov/ncea/cfm/recordisplav.cfm?deid=34855
U.S. EPA (U.S. Environmental Protection Agency). (2005a). Guidelines for carcinogen risk
assessment [EPA Report] (pp. 1-166). (EPA/630/P-03/001F). Washington, DC: U.S.
Environmental Protection Agency, Risk Assessment Forum.
http://www2.epa.gov/osa/guidelines-carcinogen-risk-assessment
U.S. EPA (U.S. Environmental Protection Agency). (2005b). Provisional Peer Reviewed
Toxicity Values for p-aminophenol (CASRN 123-30-8) [EPA Report], Cincinnati, OH.
U.S. EPA (U.S. Environmental Protection Agency). (2006). Provisional Peer Reviewed Toxicity
Values for m-aminophenol (CASRN 591-27-5) [EPA Report], Cincinnati, OH.
35
o-Aminophenol

-------
FINAL
09-28-2016
U.S. EPA (U.S. Environmental Protection Agency). (2011a). Health effects assessment summary
tables (HEAST). Washington, DC: U.S. Environmental Protection Agency, Office of
Emergency and Remedial Response, http://epa-heast.ornl.gov/heast.php
U.S. EPA (U.S. Environmental Protection Agency). (201 lb). Recommended use of body weight
3/4 as the default method in derivation of the oral reference dose (pp. 1-50).
(EPA/100/R11/0001). Washington, DC: U.S. Environmental Protection Agency, Risk
Assessment Forum, Office of the Science Advisor.
https://www.epa.gov/risk/recommended-use-bodv-weight-34-default-method-derivation-
oral-reference-dose
U.S. EPA (U.S. Environmental Protection Agency). (2012a). 2012 Edition of the drinking water
standards and health advisories [EPA Report], (EPA/822/S-12/001). Washington, DC:
Office of Water, http://www.epa.gov/sites/production/files/2015-
09/documents/dwstandards2012.pdf
U.S. EPA (U.S. Environmental Protection Agency). (2012b). Exposure Assessment Tools and
Models: Estimation Program Interface (EPI) Suite. Version 4.11. o-Aminophenol,
CASRN 95-55-6 [Fact Sheet], US Environmental Protection Agency.
http://www.epa.gov/oppt/exposure/pubs/episuite.htm
U.S. EPA (U.S. Environmental Protection Agency). (2012c). PHYSPROP. Exposure assessment
tools and models: Estimation program interface (EPI) suite version 4.11. Washington,
DC: Office of Pollution Prevention and Toxics. Retrieved from
http://www.epa.gov/oppt/exposure/pubs/episuite.htm
U.S. EPA (U.S. Environmental Protection Agency). (2016). Integrated risk information system.
IRIS assessments [Database], Washington, DC: U.S. Environmental Protection Agency,
Integrated Risk Information System. Retrieved from https://www.epa.gov/iris
University of Miami. (1975). Initial submission: Acute oral (Gavage) toxicity of pheonl, 2-
amino- in rats with cover letter dated 082892. (TSCATS/450426. OTS0571140. Doc #88-
920009484). Greensboro, NC: CIBA-GEIGY Corp.
Valentovic. MA: Ball. JG. (1998). 2-Aminophenol and 4-aminophenol toxicity in renal slices
from Sprague-Dawley and Fischer 344 rats. J Toxicol Environ Health A 55: 225-240.
Valentovic, MA; Ball, JG: Hong, SK; Rogers, BA; Meadows, MK; Harmon, RC; Rankin, GO.
(1996). In Vitro toxicity of 2- and 4-chloroaniline: comparisons with 4-amino-3-
chlorophenol, 2-amino-5-chlorophenol and aminophenols. Toxicol In Vitro 10: 713-720.
Wang. NC: Zhao. OJ: Wesselkamper. SC: Lambert. JC: Petersen. D; Hess-Wilson. JK. (2012).
Application of computational toxicological approaches in human health risk assessment.
I. A tiered surrogate approach. Regul Toxicol Pharmacol 63: 10-19.
http://dx.doi.Org/10.1016/i.vrtph.2012.02.006
WHO (World Health Organization). (2016). Online catalog for the Environmental Health
Criteria (EHC) monographs. Available online at
http://www.who.int/ipcs/publications/ehc/en/
Wild. D; King. M. -T; Eckhardt. K; Gocke. E. (1981). Mutagenic activity of aminophenols and
diphenols, and relations with chemical structure [Abstract], Mutat Res 85: 456.
Wilmer, JL; Kligerman, AD; Erexson, GL. (1981). Sister chromatid exchange induction and cell
cycle inhibition by aniline and its metabolites in human fibroblasts. Environ Mutagen 3:
627-638. http://dx.doi.org/10.1002/em.28600306Q5
Yamada. K; Shirahata. S: Murakami. H; Nishiyama. K; Shinohara. K; Omura. H. (1985). DNA
breakage by phenyl compounds. Agric Biol Chem 49: 1423-1428.
http://dx.doi.org/10.1271/bbbl961.49.1423
36
o-Aminophenol

-------
FINAL
09-28-2016
Yasuno. H. (1985). [Results of patch-testing with ingredients of hair-dye and pigmented
cosmetic dermatitis in 1983], Skin Research 27: 764-769.
http://dx.doi.Org/10.l 1340/skinresearchl959.27.764
Yoon. JS: Mason. JM; Valencia. R; Woodruff RC: Zimmering. S. (1985). Chemical
mutagenesis testing in Drosophila. IV. Results of 45 coded compounds tested for the
National Toxicology Program. Environ Mutagen 7: 349-367.
http://dx.doi.org/10.1002/em.286007031Q
37
o-Aminophenol

-------