Toluene: Toxicological Review OF
Toluene: Toxicological Review OF
Toluene: Toxicological Review OF
NCEA-S-1264
TOXICOLOGICAL REVIEW
OF
TOLUENE
August 2002
NOTICE
This document is a preliminary draft. It has not been formally released by the U.S.
Environmental Protection Agency and should not at this stage be construed to represent Agency
position on this chemical. It is being circulated for peer review on its technical accuracy and
science policy implications.
This document is a preliminary draft for review purposes only and does not constitute
U.S. Environmental Protection Agency policy. Mention of trade names or commercial products
does not constitute endorsement or recommendation for use.
FOREWORD . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
1. INTRODUCTION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
3.1. ABSORPTION/DEPOSITION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3.2. DISTRIBUTION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
3.3. METABOLISM . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
4. HAZARD IDENTIFICATION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
CONTROLS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
4.4.2. Genotoxicity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26
CHARACTERIZATION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
Modifying Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
Modifying Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
6.2.1. Noncancer/Oral . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
6.2.2. Noncancer/Inhalation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
7. REFERENCES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42
APPENDIX A. Summary of External Peer Review and Public Comments and Disposition
FOREWORD
The purpose of this Toxicological Review is to provide scientific support and rationale
for the hazard and dose-response assessment in IRIS pertaining to chronic exposure to toluene. It
is not intended to be a comprehensive treatise on the chemical or toxicological nature of toluene.
In Section 6, EPA has characterized its overall confidence in the quantitative and
qualitative aspects of hazard and dose response. Matters considered in this characterization
include knowledge gaps, uncertainties, quality of data, and scientific controversies. This
characterization is presented in an effort to make apparent the limitations of the assessment and
to aid and guide the risk assessor in the ensuing steps of the risk assessment process.
For other general information about this assessment or other questions relating to IRIS,
the reader is referred to EPA’s IRIS Hotline at 301-345-2870.
Chemical Manager
Lynn Flowers, Ph.D., D.A.B.T.
Washington, DC
Authors
Lynn Flowers, Ph.D., D.A.B.T.
NCEA
Washington, DC
North Syracuse, NY
North Syracuse, NY
Contributing Author
Karen Hogan
NCEA
Washington, DC
Reviewers
This document and summary information on IRIS have received peer review both by EPA
scientists and by independent scientists external to EPA. Subsequent to external review and
incorporation of comments, this assessment has undergone an Agency-wide review process
whereby the IRIS Program Manager has achieved a consensus approval among the Office of
Research and Development; Office of Air and Radiation; Office of Prevention, Pesticides, and
Toxic Substances; Office of Solid Waste and Emergency Response; Office of Water; Office of
Policy, Planning, and Evaluation; and the Regional Offices.
Neurotoxicology Division
Office of Water
Karen Hogan
NCEA
Pharmacokinetics Branch
NHEERL
Summaries of the external peer reviewers’ comments and the disposition of their
recommendations are in Appendix A.
This document presents background and justification for the hazard and dose-response
assessment summaries in EPA’s Integrated Risk Information System (IRIS). IRIS summaries
may include an oral reference dose (RfD), inhalation reference concentration (RfC) and a
carcinogenicity assessment.
The RfD and RfC provide quantitative information for noncancer dose-response
assessments. The RfD is based on the assumption that thresholds exist for certain toxic effects
such as cellular necrosis but may not exist for other toxic effects such as some carcinogenic
responses. It is expressed in units of mg/kg-day. In general, the RfD is an estimate (with
uncertainty spanning perhaps an order of magnitude) of a daily exposure to the human population
(including sensitive subgroups) that is likely to be without an appreciable risk of deleterious
noncancer effects during a lifetime. The inhalation RfC is analogous to the oral RfD, but
provides a continuous inhalation exposure estimate. The inhalation RfC considers toxic effects
for both the respiratory system (portal-of-entry) and for effects peripheral to the respiratory
system (extrarespiratory or systemic effects). It is generally expressed in units of mg/m3.
Conversion factor: 1 ppm = 3.77 mg/m3, 1 mg/m3 = 0.265 ppm (25oC, 760 mmHg)
Large amounts of toluene are released to the environment each year, mostly to the
atmosphere. The largest source of toluene release is during the production, transport, and use of
gasoline. In the atmosphere, toluene is degraded by reaction with hydroxyl radicals, with a
typical half-life of approximately 13 hours. Toluene has been detected in drinking water
supplies, in particular in locations with leaking underground fuel storage tanks. Toluene has
3.1. ABSORPTION/DEPOSITION
Studies quantifying oral absorption of toluene are limited, but have demonstrated nearly
100% absorption following a single oral exposure. In volunteers exposed to an infusion of 2 mg
toluene/minute for 3 hours (~5 mg/kg) via a gastric tube, absorption of toluene, measured by
monitoring exhaled air for toluene and urine for toluene metabolites, was found to be complete
(Baelum et al., 1993). Turkall et al. (1991) reported that greater than 99% of a single gavage
dose of radiolabeled toluene in rats was eliminated in the urine or expired air, indicating near-
total absorption of the exposure.
Several studies have examined the absorption of toluene during and following a single
inhalation exposure in humans. Benoit et al. (1985) reported an average retention of 83% in four
subjects exposed to 50 ppm (189 mg/m3) toluene for ~90 minutes. Carlsson (1982) reported an
average uptake (percent of inspired air) of about 55% in male subjects exposed to 300 mg/m3 for
2 hours at rest; this value dropped to 50% during the next 2 hours of exposure at rest. When the
subjects exercised, the percent uptake declined with exercise time and exercise load; the absolute
uptake (in mg toluene) increased with exercise time and exercise load (due to increased
pulmonary ventilation). Löf et al. (1990) reported a similar absorption percentage (~50%
absorbed) in groups of 10 males exposed to 3.25 mmol/m3 (~300 mg/m3) at rest for 4 hours. In a
subsequent paper, Löf et al. (1993) reported a similar absorption percentage for nine male
volunteers exposed to 194 mg/m3 for 2 hours under a light workload; during the first 20 minutes,
relative uptake averaged 55%, then slowly fell over time to a plateau of 46% after 80 minutes
(mean value 49.2%). A study by Neubert et al. (2001) found a good correlation between
measured air toluene concentrations and toluene levels in the blood of rotogravure printers at the
end of a 6-hour shift, though absorption itself was not quantified.
Toluene is absorbed through human skin slowly (Dutkiewicz and Tyras, 1968), with
absorption rates ranging from 14 to 23 mg/cm2/hour. A number of other studies have
demonstrated that percutaneous absorption can occur, though they did not quantitate the
absorption rate. Sato and Nakajima (1978) reported that 30-minute immersion of the hands of
volunteers in pure toluene resulted in a peak level of ~2 :mol toluene/L of blood, which was less
than 25% of the blood toluene level achieved by a 2-hour inhalation exposure to 100 ppm (377
3.2. DISTRIBUTION
Toluene that is absorbed into the blood is distributed throughout the body. Ameno et al.
(1989) reported that in a 51-year-old man who died from accidental oral overdose, the highest
toluene concentrations (per gram tissue) were in the liver, followed by pancreas, brain, heart,
blood, fat, and cerebrospinal fluid. However, Paterson and Sarvesvaran (1983) reported that a
16-year-old male who was found dead, presumably due to inhalation overdose of toluene, had
greater concentrations in the brain than the liver. Takeichi et al. (1986) reported similar findings
in a 20-year-old male painter who fell from a great height while working with a toluene-based
paint; the greatest concentrations upon autopsy, expressed as :g toluene/gram of tissue, were
found in the brain, followed by the liver and blood. Within the brain of a 31-year-old man who
was found dead in a room full of toluene vapor, the highest concentrations of toluene were found
in the corpus callosum, with the lowest in the caudate-putamen (Ameno et al., 1992). Thus, the
available human data suggest that more toluene accumulates in the brain than in the liver
following inhalation exposure, whereas following oral exposure, the liver contains the greatest
concentrations of toluene.
Pyykko et al. (1977) exposed groups of rats by both the oral and inhalation routes and
reported greater toluene concentrations (per gram of wet tissue) in the liver than the brain by both
exposure routes. Following inhalation exposure during which dogs were allowed to rebreathe
toluene, the liver and brain contained the highest levels (both ~190 :g/g tissue), with lesser levels
in the kidneys (Ikeda et al., 1990). Several studies have shown relationships between blood and
tissue levels of toluene, particularly for the brain (Benignus et al., 1984; Harabuchi et al., 1993).
Toluene is able to cross the placenta and enter the fetus (Ghantous and Danielsson, 1986), and
can be found in breast milk (Pellizzari et al., 1982).
3.3. METABOLISM
The main enzymatic pathways believed to be involved in toluene metabolism are shown
in Figure 1 (Nakajima and Wang, 1994; Tassaneeyakul et al., 1996; Nakajima et al., 1997;
Angerer et al., 1998; IARC, 1999). The initial step in toluene metabolism is transformation by
Toluene O
2,3-Toluene 3,4-Toluene
(CYP2E1 epoxide epoxide (GSH-S-transferase)
CYP2B6
CYP2C8
CYP1A2) CH3
CH3 CH3
H 2C SG GSH CH2OH
OH
(GSH-S-transferase) SG
(Alcohol DH
Aldehyde DH)
Conjugation glutamic acid
COOH Acyl-CoA
Acyl-CoA glutamic acid UDP-glucuronate Sulfates
(sulfotransferase) glycine
Glucuronides
glycine (UDP-Glucuronyl transferase)
H 2C S C CH CO-glucuronide
H2 CONHCH2COOH
COOH
NHCOCH3
S C CH
S-Benzyl Hippuric acid H2
mercapturic acid COOH
S-p-Toluylmercaptic acid
Studies in both humans and animals have shown that the majority of toluene in the body
is eliminated in the urine, mainly as metabolites (Löf et al., 1990, 1993; Turkall et al., 1991;
Tardif et al., 1992, 1998). As discussed above, the primary urinary metabolite of toluene is
hippuric acid, with additional metabolites (see Figure 1) resulting from minor metabolic
pathways. Elimination from the blood is rapid (Sato and Nakajima, 1978; Carlsson, 1982; Löf et
al., 1990, 1993), with three-phase elimination half times of 3, 40, and 738 minutes following a
single inhalation exposure in humans (Löf et al., 1993). A lesser, but still significant, amount of
inhaled toluene is removed in the expired air (Pellizzari et al., 1992; Monster et al., 1993).
Elimination of toluene in the expired air is greatest at time points during or immediately after
exposure, and decreases rapidly thereafter (Benoit et al., 1985). In rats, Turkall et al. (1991)
estimated that ~22% of a single oral dose is eliminated in the expired air, with the remainder
being mainly eliminated in the urine.
PBPK models are available that describe the kinetics of toluene after inhalation exposure;
two for humans (Fisher et al., 1997; Pierce et al., 1996, 1999) and two for rats (DeJongh and
Blaauboer, 1996, 1997; Tardif et al., 1993). These models are all modifications of the standard
four-compartment PBPK model developed for styrene (Ramsey and Andersen, 1984) in which:
(1) absorption into the lung blood is assumed to be dependent on the inhaled
concentration of toxicant, the concentration of toxicant in alveolar air, blood flow to the
lung, the blood/air partition coefficient, and alveolar ventilation rates;
(2) exchange of toxicant between arterial blood and tissue compartments is flow-limited;
(3) changes in the amount of toxicant in three nonmetabolizing tissue compartments
(adipose tissue, slowly perfused tissues, and rapidly perfused tissues) are described by
mass transfer differential equations with tissue volume, blood flow through the tissue
(i.e., tissue perfusion rate), arterial blood toxicant concentration, and tissue/blood
partition coefficients as explanatory variables; and
(4) changes in toxicant amount in the liver (the fourth compartment) are described by
similar differential equations that additionally include a Michaelis-Menten term for
overall rates of toxicant metabolism.
The five-compartment human model for toluene developed by Pierce et al. (1996)
includes an additional equation describing mass balance across the lung that has a Michaelis-
Menten metabolic term. A five-compartment rat PBPK model developed by DeJongh and
Blaauboer (1996) is similar in design to the Tardif et al. (1993) rat PBPK model except that it
contains an additional nonmetabolizing compartment representing the brain. The above models
have all been partially- or fully-validated using in vivo pharmacokinetic data in the appropriate
species. Another human PBPK model has been developed for volatile organic compounds that
models transfer of toxicant via lactation from a mother to a nursing infant, but in vivo
4. HAZARD IDENTIFICATION
Reports of oral exposure to toluene in humans are limited to case reports of accidental
acute ingestions. Ameno et al. (1989) reported 15 deaths by accidental oral ingestion of paint
thinner containing toluene over the period from 1977 to 1986. A case report of a 51-year old
man who died approximately 30 minutes after he had ingested a large quantity of toluene was
presented; the probable cause of death was severe central nervous system depression. Caravati
and Bjerk (1997) reported on a case of a 46-year-old man who had ingested approximately one
quart of paint thinner containing toluene. The patient presented with severe central nervous
system depression, severe abdominal pain, diarrhea, and hemorrhagic gastritis. The patient
recovered after 36 hours of supportive care. No reports of chronic oral exposure to toluene in
humans were located.
Baelum et al. (1985) investigated the effects of a 6.5-hour toluene exposure to 43 printers
with a long-term occupational exposure to a mixture of solvents including toluene and 43
controls with no history of exposure to solvents or other chemicals. The duration of employment
for the workers ranged from 9 to 25 years. Each individual was exposed only once to either 0 or
100 ppm (0 or 377 mg/m3) toluene during a 6.5-hour exposure period, preceded by a 1-hour
acclimatization period. These subjects were then subgrouped into printers exposed to toluene
(n = 20), printers exposed to air (n = 23), controls exposed to toluene (n = 21), and controls
exposed to air (n = 22). All subjects carried out a battery of tests for psychometric performance,
visual perception, and vigilance evaluation. Both printers and controls complained of nasal and
eye irritation, unacceptable air quality, and unacceptable odor level during the toluene exposure.
Signs of neurotoxicity, including moderate fatigue, sleepiness, headaches, and a feeling of
intoxication, were likewise similarly reported for both groups. A significant decrease in
performance was found for the pegboard visual motor function test in the exposed printers, but
not in the controls exposed to 100 ppm toluene. A decrease in psychometric performance,
primarily in visual perception and accuracy, was observed in toluene-exposed individuals. Acute
exposure to toluene resulted in a lower performance in 4/10 tests conducted, 3 of these 4
evaluated visual perception. The most profound difference between subjects exposed to 100 ppm
toluene and those exposed to clean air was observed in the color discrimination test; this
In a later study, Baelum et al. (1990) exposed 32 males and 39 females to clean air,
100 ppm (377 mg/m3) toluene, or a varying exposure with a TWA value of 100 ppm, but which
contained peaks of 300 ppm (1131 mg/m3) every 30 minutes for a total of 7 hours. Toluene
exposure led to significantly increased complaints about poor air quality, altered noise
perception, increased irritation of the nose and lower airways, and a feeling of intoxication, as
well as lower scores on a vigilance test. No differences were seen between subjects exposed to
the 100 ppm exposure level compared to those who experienced peaks of 300 ppm.
Andersen et al. (1983) exposed 16 young healthy subjects to a single exposure of 0, 10,
40, or 100 ppm of toluene (0, 38, 151, or 377 mg/m3) for 6 hours under controlled conditions.
Toluene exposures did not affect nasal mucus flow or lung function. At 100 ppm, but not at 10
or 40 ppm, subjects reported a subjective irritation of the eyes and nose, as well as headache,
dizziness, and feelings of intoxication. In eight tests measuring visual perception, vigilance,
psychomotor function, and higher cortical functions, no statistically significant differences were
found as a result of toluene exposure.
Forty-two college students (21 female and 21 male) were exposed to 0, 74 ppm (279
mg/m3), or 151 ppm (569 mg/m3) toluene for 7 hours over 3 days (Echeverria et al., 1989). Each
subject received all three toluene exposure levels on different days. The odor of toluene was
masked. A battery of performance tests was administered to each participant prior to starting the
exposures and again at 4 and 7 hours during the exposure; the initial test served as a control for
those tests performed during the exposure. A 5-10% decrement in performance was considered
significant if consistent with a linear trend. Test results for visual perception differed from
control values for both exposure levels. Results of a manual dexterity test differed from control
values at the higher but not the lower exposure level. Psychomotor test results were unaffected
by toluene exposure. Subjective symptomatology increased with exposure, with increasing
numbers of complaints of eye irritation, headache, and somnolence.
Zavalic et al. (1998a) examined two groups of Croatian workers occupationally exposed
to toluene for effects on color vision, relative to a group of unexposed controls. The first
exposed group (group E1) consisted of 46 workers (3 men, 43 women) employed gluing shoe
soles, while the second group (group E2) consisted of 37 workers (34 men, 3 women) employed
in a rotogravure printing press. Mean exposure times were 16.21 ±6.1 (mean ±SD) years for
group E1 and 18.34 ±6.03 years for group E2. The control group consisted of 90 workers (61
men, 29 women) who were not occupationally exposed to solvents. For all groups, smoking and
alcohol consumption information was collected. Samples of air were collected at work stations
In the high-exposure group (group E2), there were significant correlations between
toluene in air (132 ppm with a range of 66 - 250 ppm) and toluene in blood (0.0042 µg/mg with a
range of 0.0021 - 0.9422), ortho-cresol in urine (0.97 mg/g creatinine with a range of 0.26 -
4.01), and hippuric acid (1.872 g/g creatinine with a range of 0.322 - 2.875) in urine. Correlation
between toluene in air and blood for group E1 was positive, but was not statistically significant.
CCI scores on both Wednesday and Monday were significantly higher in group E2 (1.29 + 0.10
[mean ±SD] and 1.30 + 0.11, respectively) relative to both controls (1.15 + 0.10 and 1.14 + 0.10,
respectively) and to group E1 (1.17 + 0.08 and 1.18 + 0.10, respectively). CCI scores for group
E1 were not significantly different from controls at any time examined. In all groups, including
controls, a significant correlation between CCI and both age and alcohol consumption was
reported. CCI scores for those workers who consumed no alcoholic beverages at all were
significantly greater for group E1 (1.17 + 0.08 and 1.17 + 0.08, respectively) than for non-
consumers in the control group (1.13 + 0.08 and 1.13 + 0.09, respectively); however, age-
matching of these two subgroups was not reported. Given the dependence on age and alcohol
intake, the AACCI scores are considered more relevant indicators of toluene exposure than CCI
scores. AACCI scores for group E2 were significantly correlated with toluene in blood, toluene
in air, ortho-cresol in urine, and hippuric acid in urine. No statistically significant correlation
was established between AACCI scores and any marker of toluene exposure for group E1. The
AACCI scores were significantly higher (p<0.05) group E2, but not group E1, compared to
controls. Actual data points (or mean ±SD) for AACCI scores were not reported. The results
were presented graphically. This study identified a NOAEL of 32 ppm (121 mg/m3; group E1)
and a LOAEL of 132 ppm (498 mg/m3; group E2) for alterations in color vision in toluene-
exposed workers based on AACCI scores.
Further analysis of color vision loss in the same groups of workers described above
(Zavalic et al. 1998a) was carried out to compare loss in the blue-yellow and red-green ranges
(Zavalic et al. 1998b). Both blue-yellow and red-green color confusion were significantly
increased in printers, but there was no significant difference in the prevalence of either type of
color confusion between exposed and unexposed workers.
In another study, Cavalleri et al. (2000) examined a cohort of 33 rubber workers (mean
exposure duration, 117 months) and 16 referents for changes in color vision, as evaluated by the
Lanthony D-15 desaturated panel. Urine samples were taken at the end of the day and analyzed
for unmetabolized toluene. Exposed workers showed significant impairments in color vision, as
evidenced by increases in CCI or total confusion index (TOTCI) scores, relative to control
workers. However, while the indices of color vision showed linear correlations with the product
of the urinary toluene and total exposure duration, airborne levels of toluene cannot be
determined from the data presented in the manuscript. Because this study did not identify
exposure levels of toluene, instead correlating response with urinary toluene levels, no NOAEL
or LOAEL values could be identified.
Foo et al. (1990) conducted a cross-sectional study involving 30 exposed female workers
employed at an electronic assembly plant where toluene was emitted from glue. Toluene levels
reported in the study were from personal sample monitoring and reported as an 8-hour time-
weighted average (TWA), although the number of samples taken and the actual sampling period
were not given. No historical exposure values were given. Co-exposure to other solvents was
not addressed in the study. The exposed and control cohorts were matched for age, ethnicity, and
use of medications. Members of these cohorts did not use alcohol and were nonsmokers.
Medical histories were taken to eliminate any histories of central or peripheral nervous system
disorders. The average number of years (± SD) worked by the exposed population was 5.7 ± 3.2
and by the controls was 2.5 ± 2.7. Personal air samplers indicated that exposed workers breathed
mean toluene air levels of 88 ppm (332 mg/m3) as a TWA and control workers breathed a mean
of 13 ppm (49 mg/m3) (TWA). A battery of eight neurobehavioral tests were administered to all
exposed and control workers. The tests were performed midweek, before the workers reported to
their stations for the day. Group means revealed statistically significant differences in 6/8 tests;
all tests showed that the exposed workers performed poorly compared with the control cohort.
When individual test results were linearly regressed against personal exposure concentrations, the
slopes of the regression lines were all significantly nonzero. However, there was considerable
scatter among the data sets, with correlation coefficients ranging from 0.44 to 0.30. Irritation
effects were not evaluated in this study, and no clinical signs or symptoms were reported. This
study identified a LOAEL of 88 ppm of toluene (332 mg/m3) for neurobehavioral changes from
chronic exposure to toluene.
Murata et al. (1993) examined 10 rotogravure printers and 10 age-matched controls for
differences in electrocardiographic R-R intervals (CVRR and C-CVHF), the distribution of nerve
conduction velocities (DCV), and the maximal motor and sensory nerve conduction velocities
(MCV and SCV) in the median nerve. Toluene exposure was estimated to be 83 ppm (313
mg/m3), with exposure durations of 1-36 years. Blood samples for toluene analysis were taken
from the workers before electrophysiological testing during normal working hours, while urine
samples for hippuric acid analysis were taken at 5:00 pm the day following electrophysiological
analysis. CVRR and C-CVHF were significantly reduced in the toluene-exposed workers, as were
MCV in the forearm and SCV in the palm. MCV in the palm and SCV in the forearm were not
significantly different from controls. The electrophysiological data were not significantly
correlated with blood toluene or urinary hippuric acid levels, or with exposure duration. This
study identified a LOAEL of 83 ppm for alterations in electrophysiological parameters; no
NOAEL was identified.
Morata et al. (1997) examined 124 workers at a rotogravure printing facility for changes
in hearing; no control subjects were reported. Toluene levels in the air ranged from 0.14 to 919
mg/m3; co-exposure to ethanol and ethyl acetate was common. Exposure times ranged from 1 to
25 years, with a mean of 7 years. Hippuric acid in urine was utilized to assess total toluene
exposure in 109 of the workers. While a number of other variables were considered, only the age
of the subject and hippuric acid content of the urine showed significant correlations with hearing
loss. This study failed to identify a NOAEL or LOAEL.
Vrca et al. (1995, 1997) examined a group of 49 rotogravure printing workers relative to
59 controls for alterations in visual evoked potentials (VEP); a second study examined the 49
workers for changes in brain stem evoked potentials (BEAP), as measured with a brain imager.
Average length of work service for the printers was 21.4 years, and exposure concentrations
ranged from 40 to 60 ppm (151-226 mg/m3). Toluene in peripheral blood was measured
Wednesday morning before entering the work area, while urinary levels of hippuric acid and
ortho-cresol were determined both before and after the Wednesday work shift. Of the three VEP
waves examined (N75, N100, and N145), significant increases in amplitude were seen for all
three, but no differences in time of wave onset, time of wave offset, total duration of each wave,
or total duration of all waves combined were noted between the exposed and control groups
(Vrca et al., 1995). In the second study, BEAP waves P1 through P5 were examined, and there
was a significant correlation between latency of the waves and the length of exposure for all
waves except the P2 wave (Vrca et al., 1997). No correlation between wave amplitude and
exposure length was seen. Combined, these studies identify a LOAEL of 40-60 ppm for
alterations in visual- and auditory-evoked brain potentials; no NOAEL was identified.
Yin et al. (1987) reported on a cohort of over 300 solvent workers, 94 of whom (38 men,
65 women) were exposed primarily to toluene at a mean concentration of 42.8 ppm (161 mg/m3),
relative to 129 controls. Workers were co-exposed to 1.3 ppm benzene. A small but significant
decrease in hemoglobin was seen in exposed men, while a small but significant increase was seen
in exposed women. In both sexes, increased numbers of eosinophils were found in the exposed
workers relative to controls, and the values of both lactate dehydrogenase and leucine
aminopeptidase activity in the serum were decreased relative to controls. Levels of inorganic
phosphorus in the serum of exposed male workers, but not female workers, were also
significantly lower than controls. In considering the prevalence of subjective symptoms (sore
throat, headaches, and dizziness) workers were subgrouped into low (6-39 ppm, n = 28) and high
(40-123 ppm, n = 29) exposure categories. Although the prevalence of subjective symptoms was
significantly higher in the exposed workers compared with the control cohort (p<0.01), a
Eller et al. (1999) reported on the neurological effects of 98 male rotogravure printers
chronically exposed to toluene. Exposed workers were divided into workers exposed for 1-12
years (Group 1; n=30) or workers exposed for greater than 12 years (Group 2; n=49); the control
group consisted of 19 workers not exposed to toluene. Workers exposed for 12 years or under
were exposed to levels estimated at 25-32 ppm (94-121 mg/m3), though some procedures still
involve higher exposure levels for short periods of time. Workers exposed for greater than 12
years may have been exposed to levels exceeding 100 ppm (377 mg/m3) for up to 27 years. For
the scores of self-reported symptoms, the controls and Group 1 were found to be similar, while
Group 2 showed a statistically significantly higher incidence of symptoms relative to controls,
even after correction for age and alcohol consumption. In neurological tests, no differences
between Group 1 and controls were noted. Group 2 showed a statistically significantly poorer
performance, relative to the other groups, on 1 of 7 neurological tests and 2 of 5 sets of
neuropsychological tests; the tests which were significantly altered were left hand finger tapping,
retention times in the number learning test, and total time in the Bourdon-Wiersma test. This
study identified a NOAEL of 25-32 ppm and a LOAEL of >100 ppm for increases in subjective
symptoms and decreased performance in neurologic tests.
Wiebelt and Becker (1999) examined a cohort of 6830 German men from 11 rotogravure
printing plants who were exposed to toluene between 1960 and 1992. Because of an incomplete
availability of death certificates, a newly developed method was applied for the calculation of
standardized mortality ratios (SMR). Individual exposure measurements were not taken. Of the
three main work areas, two had concentrations generally lower than 30 ppm, and one was, in
general, lower than the exposure limit of 100 ppm (200 ppm before 1985). For the total cohort,
only the SMR for mental disorders, primarily alcoholism, was significantly elevated (SMR 303,
95% CI 184-541). No significant increases in cancer mortality or cause-specific cancer mortality
Svensson et al. (1990) examined the rates of cancer formation in 1020 past and present
rotogravure printers occupationally exposed to toluene for at least 3 months in one of eight
printing establishments between 1925 and 1985. Exposure levels were estimated based on
current exposure, past workplace measurements, and interviews with employees. Exposure
levels were estimated to range from 350-450 ppm until 1960, after which they steadily fell, with
a median level of ~50 ppm in 1985. Exposed workers showed no significant increase in general
mortality or from dying of malignant disease. Statistically significant excesses in tumor
incidences were seen in the gastrointestinal tract and stomach, organs that displayed SMRs of
2.06 (95% CI 1.13-3.45) and 2.72 (95% CI 1.09-5.61), respectively, when compared to those of
unexposed controls. In addition, taking all cancer formation into account, there appeared to be a
marginal excess of respiratory-tract cancers, with an SMR of 1.76 (95% CI 1.03-2.91). However,
when the latter subset was limited to those employees with greater than 5 years of potential
exposure and/or greater than 10 years latency, the resulting SMR of 1.26 (95% CI 0.57-2.38)
failed to confirm an association between exposure and response.
Anttila et al. (1998) carried out a retrospective cohort analysis of 5301 workers
(3922 male and 1379 female) monitored for biological markers of occupational exposure to
styrene, toluene, or xylene over the period of 1973-1992. Exposure was monitored from 1978 to
1983 by analysis of toluene levels in the blood. No significantly increased incidence rates of
cancer could be associated with toluene exposure.
A large number of other studies exist that examined humans exposed chronically to
toluene, mainly in an occupational setting. The majority of those studies reported concentrations
higher than those described above, and reported similar findings. Several other cohort mortality
studies also exist (Gericke et al., 2001; Gerin et al., 1998; Walker et al., 1993), but were
excluded from further analysis due to inability to account for confounding factors (i.e., smoking,
age, co-exposure to other solvents).
The primary effects described in case reports of chronic toluene abusers are on the central
nervous system, including abnormal electroencephalogram (EEG) activity, ataxia, tremors,
temporal lobe epilepsy, decreased intelligence quotient, paranoid psychosis, hallucinations,
nystagmus (involuntary eye movement), cerebral atrophy, and impaired speech, hearing, and
vision (Byrne et al., 1991; Devathasan et al., 1984; Hunnewell and Miller, 1998; King et al.,
1981; Maas et al., 1991; Meulenbelt et al., 1990; Miyagi et al., 1999; Ryu et al., 1998; Suzuki et
al., 1983). Additionally, toluene abuse during pregnancy can have dramatic effects on the
developing fetus (see section 4.3). Other known effects include effects on the kidneys, including
acidosis, necrosis, proteinuria, and tubule dysfunction (Gerkin and LoVecchio, 1998; Goodwin,
The oral toxicity of toluene was investigated in a subchronic gavage study in F344 rats
(NTP, 1990a). Groups of 10 rats/sex/group were administered toluene in corn oil at dosage
levels of 0, 312, 625, 1250, 2500, or 5000 mg/kg, 5 days/week for 13 weeks. All animals
receiving 5000 mg/kg died within the first week. One female and 8 males in the 2500 mg/kg
group died, but two of these deaths were due to gavage errors. No deaths occurred at lower
doses. Several toxic effects were noted at doses greater than or equal to 2500 mg/kg, including
prostration, hypoactivity, ataxia, piloerection, lacrimation, excessive salivation, and body
tremors. No signs of biologic significance were seen in groups receiving less than or equal to
1250 mg/kg. The only significant change in body weight was a decrease (p<0.05) for males in
the 2500 mg/kg group. There were no toxicologically significant changes in hematology or
urinalysis for any group of animals. Biochemical changes, including a significant increase
(p<0.05) in SGOT in 2500 mg/kg males and a dose-related increase in cholinesterase in females
receiving 2500 and 5000 mg/kg, were not considered to be biologically significant. There were
several pathologic findings and organ weight changes in the liver, kidney, brain, and urinary
bladder. In males, absolute and relative weights of both the liver and kidney were significantly
increased (p<0.05) at doses greater than or equal to 625 mg/kg. In females, absolute and relative
weights of the liver, kidney, and heart were all significantly increased at doses greater than or
equal to 1250 mg/kg (p<0.01 for all comparisons except p<0.05 for absolute kidney and heart
weights at 1250 mg/kg). Histopathologic lesions in the liver consisted of hepatocellular
hypertrophy, occurring at greater than or equal to 2500 mg/kg. Nephrosis was observed in rats
that died, and damage to the tubular epithelia of the kidney occurred in terminally sacrificed rats.
Histopathologic changes were also noted in the brain and urinary bladder. In the brain,
mineralized foci and necrosis of neuronal cells were observed in males and females at 2500
mg/kg and males at 1250 mg/kg. In the bladder, hemorrhage of the muscularis was seen in males
and females at 5000 mg/kg and males at 2500 mg/kg. The NOAEL in rats for this study is 223
mg/kg-day (312 mg/kg) based on liver and kidney weight changes in male rats at 446 mg/kg-day
(625 mg/kg). The toxicologic significance of these organ weight changes is strengthened by the
occurrence of histopathologic changes in both the liver and kidney at higher doses. Because the
exposure was for 5 days/week, the dose is adjusted (e.g., 312 x 5/7 = 223 mg/kg-day).
Hsieh et al. (1989) exposed groups of male CD-1 mice (five animals/group) to 0, 17, 80,
or 405 mg toluene/liter of drinking water for 4 weeks. Based on body weight and water
consumption data, the authors calculated average daily toluene doses of 0, 5, 22, or 105 mg/kg
day, respectively. Animals were weighed once per week, and food and water consumption were
monitored continuously. Water toluene concentration was determined daily, and fresh solutions
were made every three days. After 28 days, the animals were sacrificed, and body, spleen,
thymus, liver, and kidney weights were determined. Gross pathological examinations were
performed on all mice. Total erythrocytes and leukocytes were determined, and differential
leukocyte counts were measured. Splenocyte suspensions were prepared, and the
lymphoproliferative responses to the T-cell mitogens phytohemagglutinin (PHA) and
concanavalin A (Con A), the B-cell mitogen E. coli lipopolysaccharide (LPS), and the combined
mitogen pokeweed mitogen (PWM) were measured. Separate groups of animals were similarly
exposed, and were sensitized by intraperitoneal injection of sheep red blood cells (SRBC) 4 days
before the end of toluene exposure. The titer of anti-SRBC antibody in the serum collected was
used in the plaque-forming colony (PFC) assay. Toluene exposure did not result in increased
mortality or clinical signs in any exposed group. No significant changes in food or water
consumption were noted, and no gross lesions of the liver, kidney, spleen, heart, thymus, lung, or
brain were seen in any treatment group. No changes in body weight (mean ±SE) were seen as a
result of toluene exposure. Relative liver weights of toluene-exposed rats were significantly
increased (5.67 ±0.07, 6.09 ±0.11, 6.32 ±0.17, and 6.73 ±0.14 g/100 g body weight for 0, 5, 22,
and 105 mg/kg-day treatment groups, respectively) and relative thymus weights (mean ±SE) were
significantly decreased (0.019 ±0.02, 0.18 ±0.01, 0.18 ±0.02, and 0.13 ±0.02 g /100 g body
weight for 0, 5, 22, and 105 mg/kg-day treatment groups, respectively) at 105 mg/kg-day
compared to controls, but not at lower doses. The changes in organ weights at the highest dose
correspond to a 19% increase in liver weight and a 32% decrease in thymus weight relative to
controls. No changes were found in relative spleen and kidney weights at any dose. No
significant changes in hematological parameters or spleen cellularity were reported. Splenocyte
In a later study, Hsieh et al. (1990) exposed groups of male CD-1 mice for 28 days as
described above. At the end of the exposure, six discrete brain sections of the animals were
tested for endogenous levels of norepinephrine (NE), dopamine (DA), and serotonin (5-HT), as
well as their primary metabolites. No changes in body weight or clinical signs were observed.
Toluene exposure induced increases in all of the biogenic amines examined at all dose levels,
with the response generally peaking in the mid-dose group and decreasing in the high-dose
group. Significant increases of norepinephrine and its metabolite, 3-methoxy-4-hydroxymandelic
acid, were found in the midbrain of all dose groups. Significant increases in serotonin levels, but
not its metabolite (5-hydroxyindoleacetic acid), were also seen in the midbrain of all dose groups.
The unknown implications of changes in neurotransmitter levels, differences in the effects seen
in various brain sections, and the biphasic dose-response make determinations of biological
significance difficult. This study did not identify a NOAEL or LOAEL.
Wolf et al. (1956) administered groups of 10 female Wistar rats gavage doses of 0, 118,
354, or 590 mg/kg toluene dissolved in olive oil. A total of 138 doses were administered over
193 days, resulting in average doses of approximately 0, 84, 253, or 422 mg/kg-day.
Hematologic, behavioral, gross, and histopathologic examinations were conducted with no toxic
effects being reported at any dose. This study did not identify a NOAEL or LOAEL.
Fischer 344/N rats (10/sex/group) were exposed to toluene vapors at 0, 100, 625, 1250,
2500, and 3000 ppm (0, 377, 2355, 4711, 9422, and 11,307 mg/m3, respectively) 6.5 hours/day, 5
days/week (duration-adjusted to 0, 73, 455, 911, 1823, and 2187 mg/m3, respectively) for 15
weeks (NTP, 1990a). Organ weights were measured and histological examinations were
performed only on controls, 2500- and 3000-ppm groups, and animals that died before the end of
the study. Eight of 10 males exposed to 3000 ppm died, all during the second exposure week.
No females died at any exposure level. Compared to the controls, final body weights were 15
and 25% lower in the males and 15 and 14% lower in the females of the 2500- and 3000-ppm
groups, respectively. There was a concentration-related increase in the relative liver weight,
significant at 1250, 2500, and 3000 ppm in males and at 2500 and 3000 ppm in females. The
relative weights of the heart, lung, kidney, and right testis were also significantly elevated in the
2500- and 3000-ppm animals compared to those of the controls, although no histopathology was
observed in any exposure group. Toxic effects noted in a concurrently conducted gavage study
(urinary bladder hemorrhages in the two highest exposure groups) were not noted in this
subchronic inhalation study. A subsequent report on the neurologic effects of these exposures
did not indicate any neurobehavioral changes as a result of toluene exposure (Tilson, 1990), but
details of the test results are lacking. This study identified a NOAEL of 625 ppm and a LOAEL
of 1250 ppm for changes in relative liver weight in male rats.
Poon et al. (1994) exposed groups of Sprague-Dawley rats of both sexes to 0, 30, or 300
ppm toluene for 6 hours/day, 5 days/week for 4 weeks. Slightly increased relative liver weights,
but not absolute liver weights, were seen in 30 ppm males, but not in the 300 ppm males or at
any concentration in females. Females exposed to 30 ppm, but not to 300 ppm, had a mild
reduction in thyroid follicle size. A slight epithelial degeneration in the nasal conchae was noted
in 30-ppm males, but not in the higher-dose males or in either exposure group of female rats.
Because the responses occurred in the low exposure group but not the high exposure group, no
NOAEL or LOAEL could be identified from this study.
von Euler et al. (2000) exposed 30 male Sprague-Dawley rats to 80 ppm (302 mg/m3) of
toluene for 6 hours/day, 4 days/week for 4 weeks. Control animals (n=30) were similarly
exposed, but to air only. Four weeks after the last exposure, the animals were evaluated for
neurobehavioral alterations using tests for spatial learning and memory (Morris water maze),
open-field activity (open field test), and beam-walk performance. Following the conclusion of
the neurobehavioral tests, animals were examined for changes in brain morphology using
McWilliams et al. (2000) exposed groups of 8 guinea pigs to 0, 250, 500, or 1000 ppm (0,
943, 1885, or 3770 mg/m3) of toluene for 8 hours/day, 5 days/week for 1 week or 500 ppm (1885
mg/m3) for 4 weeks. At 1 and 4 weeks, animals were examined for changes in hearing by the
cubic distortion-product otoacoustic emission (CDP) technique, while after 4 weeks of exposure,
selected animals were examined histologically for changes in the cochlea. After 1 week of
exposure, a dose-related decrease in CDP amplitudes was seen, with complete recovery evident
after a 3 day rest period. A 4 week exposure to 500 ppm of toluene resulted in more severe
disturbances in hearing than were seen after 1 week, but the effects were still reversible. After 4
weeks of exposure, the cochlear cells located near the base (and high frequency) showed a loss of
succinate dehydrogenase (SDH) staining. This study identified a NOAEL of 250 ppm (943
mg/m3) and a LOAEL of 500 ppm (1885 mg/m3) for diminished startle response and histologic
alterations of the cochlea in exposed guinea pigs.
B6C3F1 mice (60/sex/group) were exposed to 0, 120, 600, or 1200 ppm (0, 452, 2261, or
4523 mg/m3, respectively) toluene 6.5 hours/day, 5 days/week (duration-adjusted to 0, 87, 47,
and 875 mg/m3, respectively) for 2 years (NTP, 1990a). Mean body weights were not
significantly different among groups and no treatment-related clinical signs were observed.
Fischer 344 rats (120/sex/group) inhaled 0, 30, 100, or 300 ppm (0, 113, 377, or 1130
3
mg/m , respectively) toluene (99.9% purity), 6 hours/day, 5 days/week (duration-adjusted to 0,
20, 67, or 202 mg/m3, respectively) for 106 weeks (CIIT, 1980; Gibson and Hardisty, 1983).
Vapor, generated by bubbling clean air through toluene, was passed through the air supply duct
and mixed with air by turbulent flow to produce the desired concentration. Hematology, blood
chemistry, and urinalysis were conducted in all groups at 6 (5/sex), 17 (5/sex), 18 (10-20/sex),
and 24 months (10/sex). Histopathology was evaluated only in the control and 300 ppm groups
at 6 (5/sex), 12 (5/sex), and 18 months (20/sex). At 24 months, histopathological examinations
were conducted in organs of all surviving animals, including the respiratory system and sections
through the nasal turbinates (number not indicated). No treatment-related non-neoplastic effects
were observed in the exposed animals. Although the male rats exposed to 300 ppm had a
significant increase in body weight compared to controls, no concentration-response was evident.
At the end of the exposure period, the female rats exposed to 100 or 300 ppm exhibited a slight
but significant reduction in hematocrit; an increase in the mean corpuscular hemoglobin
concentration was also noted but only in the females exposed to 300 ppm. Gross and
microscopic examination of tissues and organs identified no increase in neoplastic tissue or
tumor masses among treated rats when compared with controls, though because the study was
conducted at exposure levels below the MTD, the significance of this finding is less clear. The
highest concentration examined in this study, 300 ppm, is designated as a NOAEL for toxicity
remote from the respiratory tract in rats. CIIT (1980) reported that the technical and raw data
were not audited by their quality assurance group during the study period, although CIIT did
conduct a quality assessment procedure to review the data. The available pathology reports
containing these data indicate that at least the lower respiratory tract was examined.
Communication with the testing sponsor has provided information indicating that only one
section was examined from the nasal cavity of these test animals. It is not clear whether this
single section would have been sufficient to elucidate the areas of lesions noted in the NTP
(1990) study. Consequently, the designation of the 300 ppm exposure level as a NOAEL for
respiratory lesions (see NTP, 1990a) is problematic.
Toluene is believed to cause congenital defects in infants born to mothers who abused
toluene during pregnancy, though exposure levels in the available studies, if reported at all, were
very high. A detailed discussion of the high-dose effects of toluene on reproduction and
development is beyond the scope of this document; a discussion of a representative sample of the
available studies is included. In a case report study, Hersh et al. (1985) describes clinical and
morphometric characteristics common to three children whose mothers had abused toluene (but
apparently not alcohol or any other substance) for a period of 4-5 years including during their
pregnancies with the affected children. Clinical findings common to these three children
included microcephaly, CNS dysfunction, attention deficits, and developmental delay/mental
deficiency. Phenotypic similarities included a small midface, deep-set eyes, micrognathia
(smallness of the jaws), and blunting of the fingertips.
A case-referent study was conducted by McDonald et al. (1987) who examined the
history of exposure to chemicals of 301 women who had recently given birth to an infant with an
important congenital defect. An identical number of women (referents) who had given birth to
normal children were matched with respect to age, employment (hours/week), date of delivery,
and educational level. In initial matched-pair analysis, chemical exposure was higher in the cases
than in the referents (63 cases:47 referents) due to excess cardiac and miscellaneous defects. In
further analysis by chemical categories, only exposure to aromatic solvents showed a clear excess
of defects, mostly in the urinary tract. Details of these cases (n = 19) showed that toluene was
identified as the solvent in 11 of these cases.
Kostas and Hotchin (1981) exposed NYLAR mice pre- and postnatally to toluene
provided in the drinking water at concentrations of 0, 16, 80, or 400 ppm. Effects were noted in
all dosed groups on rotorod performance, measured at 45 to 55 days of age, but there was an
Gospe et al. (1994, 1996; Gospe and Zhou, 1998, 2000) conducted a series of studies
examining the effects of oral prenatal toluene exposure on the development of rats. In the first
experiment (Gospe et al., 1994), pregnant rats received 520 mg/kg in corn oil by gavage on
gestational days 6-19, and offspring were examined on gestational day 19. Toluene exposure did
not result in maternal deaths, but did result in a significant decrease in weight gain (24%
decrease) and a 12% reduction in food consumption, although this difference was not statistically
significant. No differences in number of implantations or resorptions were found between
control and exposed groups, but fetal body weights, organ weights (liver and kidney), and
placenta weights were significantly decreased in toluene-exposed animals. No gross fetal
malformations were reported, and histologic examination of the brain revealed no treatment-
related changes. In the second experiment (Gospe et al., 1996), pregnant rats received 650 mg
toluene/kg in corn oil by gavage on gestation days 6-19; both control (corn oil only) and pair-fed
controls were also examined. Fetuses were delivered and examined on day 19 of gestation.
Toluene exposure resulted in significantly decreased fetal weight, decreased organ weights
(brain, liver, heart, and kidney), and a delay in skeletal ossification. No differences were seen
between pair-fed animals and control animals, and toluene-induced changes were significantly
different from the pair-fed group as well. In the third experiment (Gospe and Zhou, 1998),
groups of pregnant rats were exposed by gavage to 650 mg toluene/kg in corn oil on gestation
days 6-19, and offspring were examined on gestation day 19, or on postnatal days 10 or 21.
Toluene exposure did not result in changes in litter size, maternal death, or maternal liver weight.
At gestational day 19, fetal body weights, as well as the weights of the heart, liver, kidney, and
brain, were significantly reduced in toluene-exposed animals. At postnatal day 10, the body,
heart, and kidney weights of prenatally-exposed weights remained significantly lower than
controls, while by postnatal day 21, no differences between control and treated animals were seen
for body or organ weights. While prenatal exposure to 650 mg toluene/kg on gestational days 6-
21 did not result in decreased organ or body weights on postnatal day 21, histologic analysis of
the brain revealed decreased neuronal packing and alterations in the patterns of staining with
bromodeoxyuridine (Gospe and Zhou, 2000), indicating compound-induced alterations in
neurogenesis and neuronal migration.
Pregnant Wistar rats and hamsters (group size not indicated) inhaled 0 or 800 mg/m3
(212 ppm) toluene vapors 6 hours/day on gestational days 14-20 (rats) or gestational days 6-11
(hamsters) (DaSilva et al., 1990). In the exposed rats, there was a significant (p<0.05) increase in
the number of litters with one or more low birth weight pups (less than 4.9 g), from 10% in the
controls to 54% in the exposed dams. A decrease (p<0.05) in the number of live pups at birth
was also noted in the litters of exposed dams. No evaluation of malformations or anomalies was
Thiel and Chahoud (1997) exposed groups of pregnant Wistar rats to 0, 300, 600, 1000,
or 1200 ppm (0, 1131, 2262, 3370, or 4524 mg/m3) toluene for 6 hours/day from day 9 to day 21
of pregnancy. At birth, the number of live and dead pups was determined, as well as mean pup
weight per litter. Postnatal weight gain was recorded weekly, and signs of physical development,
including eruption of incisors, fur development, eye opening, testes descent, and vaginal opening,
were monitored. Prior to weaning, reflex testing was performed on the offspring. After weaning,
offspring were tested for locomotor activity and discrimination learning. Toluene exposure did
not result in changes in duration of pregnancy or litter size. At the two highest doses, toluene
produced a significant decrease in maternal body weight gain and mean pup weight. High-dose
offspring had a significantly increased mortality during the suckling period (postnatal days 2-21).
Postnatal development (time of testes descent or vaginal opening) was accelerated at 600 ppm,
but was delayed at 1000 ppm of toluene or greater. No changes in neurobehavioral parameters of
the exposed offspring were noted relative to controls. With the exception of an increased mean
fertility in the 600 ppm group, the fertility of the offspring was not different from that of controls.
Dalgaard et al. (2001) exposed groups of pregnant Wistar rats to airborne concentrations
of 1200 ppm of toluene for 6 hours/day on gestational days 7 to 18, and examined male offspring
on postnatal day 110 for alterations in semen quality. No effect of toluene exposure on semen
quality was seen. In a subsequent study (published in the same manuscript), groups of pregnant
rats were exposed to 1800 ppm of toluene from gestational days 7 to 20, and the male offspring
were examined on postnatal days 11, 21, or 90. Mean body weights in exposed pups were lower
than controls on day 11, but were not significantly different on days 21 or 90. Absolute and
relative testes weights were decreased in all age groups, but the differences were not statistically
significant. Histologic analysis of the testes revealed no effects of toluene exposure in any age
group. Microscopic examination of the hippocampus revealed no changes in apoptotic
neurodegeneration in any group, whereas toluene induced a statistically significant increase in
apoptosis in the cerebellar granule layer on postnatal day 21, but not day 11 or 90.
In another study with similar exposure conditions, Hougaard et al. (1999) exposed groups
of pregnant Wistar rats to airborne levels of 1800 ppm of toluene for 6 hours/day from
gestational days 7 to 20. Body weights of exposed offspring were lowered until postnatal day 10,
after which no significant differences were noted. Neurobehavioral evaluation of the pups
revealed no effects on motor function, activity level, acoustic startle, and prepulse inhibition.
Measurement of hearing function revealed small but significant changes in male offspring.
A subsequent study by the same investigators (Hass et al., 1999) exposed groups of
pregnant Wistar rats to 1200 ppm of toluene for 6 hours/day on gestational days 7 to 18. The
exposure did not cause maternal toxicity or decreased offspring viability. As was the case with
the previous study, offspring body weights were significantly reduced through postnatal day 10,
and were not significantly different thereafter. Alterations in Morris water maze performance
were evident in female offspring at 3.5 months of age; no other changes in neurobehavioral
parameters were reported.
Ungvary and Tatrai (1985) exposed New Zealand rabbits (8-10/group) to 0, 500, or
1000 mg/m3 toluene, 24 hours/day, on gestational days 7-20, and CFLP mice (15 females/group)
to 0, 500, 1000, or 1500 mg/m3 toluene, also continuously, on gestational days 6-15. The control
groups consisted of 115 mice and 60 rabbits. All of the female mice that were exposed to 1500
mg/m3 died. In the mice exposed to 1000 mg/m3, there was an increase in fetuses with retarded
weight (29%, level of retardation not indicated) and in fetuses with skeletal retardation (12%)
compared to 7 and 5%, respectively, in the controls, which did not differ from the animals
exposed to 500 mg/m3. Of the 8 pregnant rabbits exposed to 1000 mg/m3, 2 died, 4 had
spontaneous abortions, and the remaining 2 had total litter resorption. No deaths occurred in the
10 rabbits exposed to 500 mg/m3, but 1/10 rabbits had a spontaneous abortion (as compared to
0/60 reported for the controls). No effects were seen on fetal development in rabbits exposed to
500 mg/m3.
Pregnant Charles River CD-1 mice (15-16 females/group) inhaled filtered air or 200 or
400 ppm (754 and 1508 mg/m3) toluene 7 hours/day on gestational days 7-16 (Courtney et al.,
1986). The relative liver weight in the exposed dams was reported to be significantly lower in
the two exposed groups compared to the controls, although no data were presented. A
statistically significant increase in lactate dehydrogenase activity in the brain of the dams
exposed to 400 ppm was also reported. The exposed pregnant mice did not exhibit any
significant differences in the number of implantation sites, number of live fetuses, fetal deaths, or
fetal body weight compared to the control values. A statistically significant increase over
controls in the incidence (both per litter and per fetus) of enlarged renal pelves was noted in dams
exposed to 200 ppm, but not to 400 ppm. A statistically significant alteration from controls in
the rib profile (percentage of fetuses with 1 or 2 additional/fewer ribs) was reported for fetuses
from dams exposed to 400 ppm, but not to 200 ppm. The toxicological significance of this
finding is not clear.
Ono et al. (1995) exposed groups of pregnant Sprague-Dawley rats to 0, 600, or 2000
ppm (0, 2262, or 7540 mg/m3) of toluene for 6 hours/day from days 7 to 17 of gestation, and
examined the offspring for malformations and alterations in behavioral parameters. Preweaning
tests included surface righting and negative geotaxis, while postweaning tests included an open
field test (postnatal week 4), the Biel water maze (postnatal week 6), and rotorod tests (postnatal
In a later study, Ono et al. (1996) exposed groups of male and female Sprague-Dawley
rats to 0, 600, or 2000 ppm (0, 2262, or 7540 mg/m3) of toluene for examination of effects on
fertility. Females were exposed from 14 days before mating to day 7 of gestation, while males
were exposed for 90 days, beginning at 60 days before pairing. In females exposed to 2000 ppm,
increased salivation and lacrimation were noted starting 20 days after exposure. No changes
were noted in mating behavior or fertility at either exposure level. Fetal mortality and the
number of dams with dead fetuses were both increased in the 2000 ppm animals, but these
differences were not statistically significant. In males exposed to 2000 ppm for 90 days,
increased kidney weight and decreased thymus weights were observed. Additionally, high-dose
males showed a decreased epididymal weight, though no abnormalities of the testes or
epididymis were noted histologically. Sperm counts were significantly reduced in the 2000 ppm
animals; the sperm count of the 600 ppm group was slightly decreased, but did not attain
statistical significance.
Mehta et al. (1998) exposed groups of male and female Sprague-Dawley rats to a single
gavage dose of 0, 3, 4.5, or 6 ml toluene/kg (0, 2600, 3900, or 5200 mg/kg, respectively). On
days 1 (2-3 hours after exposure), 7, and 14 post-exposure, the animal body weights were
recorded, and a functional observation battery (FOB) was conducted to detect neurobehavioral
changes. A significant, dose-dependent decrease in body weight occurred at day 7 for male rats.
Decreases in body weight gain were noted in male rats at 14 days and female rats at 7 days, but
the differences were not statistically significant. On day 1, but not on days 7 and 14, toluene-
treated rats of both sexes exhibited a dose-dependent increase in abnormal gait. The open-field
rearing scores were lower for all groups of both sexes at day 1 only, though only achieved
statistical significance in high-dose females. Horizontal motor activities were significantly lower
in both sexes at all dose levels on day 1; the values remained lower in all treated female groups
and the 3 and 4.5 ml/kg male rats on day 7, and in 3 and 4.5 ml/kg female rats on day 14. Rats of
both sexes showed increased incidences of lacrimation and/or salivation on day 1 only; the effect
was more pronounced in females.
Dyer et al. (1988) exposed groups of male Long-Evans rats to a single gavage dose of 0,
250, 500, or 1000 mg toluene/kg in corn oil. Flash-evoked potential (FEP) tests were
administered 45 minutes later as a test of the ability of the nervous system to process visual
information. Toluene exposure resulted in a significant decrease of the N3 peak of the FEP in all
dose groups, though the decrease was not dose-related. In an additional study, presented in the
same manuscript, rats were exposed to 500 mg/kg and FEP was examined at 4, 8, 16, and 30
hours post-exposure. Depression of the N3 peak remained at 8 hours post-exposure, but by 16
hours recovery appeared complete.
A number of acute animal studies have examined the neurological effects of inhaled
toluene; these studies generally reported impaired response in neurologic examinations. For
example, Rebert et al. (1989a,b) reported abnormal flash-evoked potentials in rats exposed to a
single inhalation exposure of 500-16,000 ppm toluene. Wood et al. (1983) exposed rats to
toluene levels up to 3000 ppm for 4 hours prior to behavioral evaluation, and reported that
toluene reduced performance in behavioral tests, particularly at the 1780 and 3000 ppm exposure
levels. Wood and Colotla (1990) reported a biphasic response in mice exposed to toluene for 1
hour. An increase in activity was seen at concentrations up to 1000 ppm, beyond which
decreased activity was seen. Similar results were reported by Wood and Cox (1995), with rats
exposed at concentrations up to 1000 ppm showing progressive increases in activity, with
decreasing activities at higher concentrations, up to and including 3000 ppm.
Toluene has tested negative for reverse mutation in Salmonella typhimurium, both with
and without S-9 activating mixture (Mortelmans and Riccio, 1980; Nestmann et al., 1980; Bos et
al., 1981; Litton Bionetics, Inc., 1981; Connor et al., 1985; NTP, 1990a). Toluene also tested
negative in the umu test in S. typhimurium (Nakamura et al., 1987), and was negative for reverse
mutation in E. coli (Fluck et al., 1976). NTP (1990) reported no increase in sister-chromatid
exchanges (SCE) or chromosomal aberrations in Chinese hamster ovary cells exposed to toluene.
Available studies (Gerner-Smidt and Friedrich, 1978; Richer et al., 1993) have reported no
increase in SCE in human lymphocytes exposed in vitro to toluene, even at concentrations that
inhibited cellular growth.
Dobrokhotov and Einkeev (1977) exposed male rats (strain not specified) to 610 mg/m3
toluene for 4 hours/day for 4 months, reporting a reversible increase in chromosomal gaps and
breaks in isolated bone marrow cells. Mice exposed to toluene at concentrations of 100 or 400
ppm for 6 hours/day, 5 days/week for 8 weeks showed no increase in dominant lethal mutations
(measured as pre- or post-implantation embryo loss) relative to controls (API, 1981a). BDF1
mice exposed to 500 ppm toluene for 6 hours/day, 5 days/week for up to 8 weeks showed no
increase in DNA damage, assessed by starch gel electrophoresis, relative to controls (Plappert et
al., 1994).
Published toxicity studies of oral exposure to toluene in humans are limited to case
reports of acute oral overdoses (Ameno et al., 1989; Caravati and Bjerk, 1997). Clinical effects
in these cases have included central nervous system depression, severe abdominal pain, diarrhea,
and hemorrhagic gastritis. Chronic toxicity studies of oral toluene exposure in animals are not
available. Maltoni et al. (1997) conducted a 2-year gavage study of toluene in rats; however,
While the most sensitive effects of inhalation exposure to toluene involve neurologic
endpoints, detailed reports of similar effects following prolonged oral exposure are lacking. A
report of a case study of an accidental oral overdose of toluene did report central nervous system
depression (Caravati and Bjerk, 1997), but no quantitative measure of these effects exists.
Animal studies have identified effects on the liver, and perhaps immunologic effects, as the most
sensitive effects of known toxicologic relevance. Brain lesions have been observed in orally-
exposed rats (NTP, 1990a), but these effects occurred only at exposure levels ($ 893 mg/kg-day)
higher than those affecting liver endpoints, and were not observed in mice at any exposure level.
Other effects (changes in brain neurotransmitter levels, decreased response to mitogenic
stimulation) have been reported (Hsieh et al., 1989, 1990), but their toxicological significance is
not presently clear. Liver effects have been reported to be the most sensitive following oral
toluene exposure in animals, possibly because the liver contains large amounts of the enzymes
for toluene metabolism, and the fact that orally-absorbed toluene will pass through the liver
before being distributed to the rest of the body. This observation is further supported by the fact
that studies examining the distribution of toluene following oral exposure have generally found
greater amounts of toluene per gram of tissue in the liver than in the brain (Ameno et al., 1989;
Pyykko et al., 1977). The generation of arene oxide intermediates formed in the metabolic
pathway from toluene to ortho- or para-cresol (see Figure 1) could hypothetically result in
increased toxicity at the site of metabolism, particularly at high exposure levels when other
pathways may achieve saturation. However, no data are available directly evaluating this
potential mode of action.
In summary, data on the effects of toluene in humans following oral exposure are limited
to case reports of accidental oral ingestions. Both a 4 week study in mice (Hsieh et al., 1989,
1990) and a 13 week study in rats (NTP, 1990a) identified increases in liver weight as the most
Many studies have been published examining neurological endpoints resulting from
repeated low-level toluene exposure in occupationally-exposed workers. Results from these
studies suggest that subtle neurologic effects are the most sensitive endpoints following repeated
inhalation exposure to toluene. The NOAEL/LOAEL values of several chronic human studies
are shown in Table 1. As can be seen, a number of studies of occupationally-exposed humans
have reported effect levels ranging from 40 to 132 ppm, with two studies (Zavalic et al., 1998a;
Eller et al., 1999) identifying NOAEL values as well.
There is evidence that exposure to toluene results in both transient and persistent effects
on neurologic endpoints, but the available data often do not clearly discern persistent effects from
transient effects, even on the same endpoint. For example, Baelum et al. (1985) reported that the
neurologic responses, including altered color vision, of rotogravure printers (average toluene
exposure of 9 to25 years) exposed to a single 6.5 hour exposure of 100 ppm toluene did not
differ from a control group who had not been previously exposed to toluene, suggesting that the
acute effects of toluene on color vision were transient, rather than being dependent on previous
exposure history. In contrast, Zavalic et al. (1998c) reported that analysis of color vision scores
in toluene-exposed workers on Wednesday did not differ from the scores in the same workers on
Monday, after at least 48 hours without exposure, suggesting that the effect was persistent.
Similarly, McWilliams et al. (2000) reported that guinea pigs exposed to 500 ppm toluene for up
to 4 weeks showed a reversible hearing loss, while Pryor et al. (1984) reported that hearing loss
in male rats exposed to 1000 ppm toluene or greater for up to 2 weeks was still present after a 3-
month recovery period.
The most compelling evidence for the ability of repeated toluene inhalation exposure to
produce persistent neurologic effects comes from case reports of toluene abusers, who are
generally exposed to concentrations exceeding 1000-10,000 ppm. MRI examinations of the
brain of solvent abusers (Filley et al., 1990; Rosenberg et al., 1988a, 1988b) suggest a
preferential atrophy in lipid-rich regions of the brain. Rosenberg et al. (1988a,1988b) found MRI
evidence of diffuse central nervous system demyelination in 6 toluene abusers with clinically
obvious neurological impairment, whereas Filley et al. (1990) noted that the degree of MRI-
detected white matter abnormality in 14 solvent abusers was correlated with neurological
impairment. The observed changes in MRI signals may be related to lipid compositional changes
in the white matter, since these regions are more lipid-rich than gray matter (Ameno et al., 1992).
Animal data have also suggested that respiratory tract irritation, particularly in the nasal
cavity, is a sensitive effect of toluene. However, the primary study that reported this effect (NTP,
1990a) only examined concentrations of 600 ppm and greater. A lifetime chronic (CIIT, 1980)
and 28-day subchronic (Poon et al., 1994) study have each examined exposed rats for changes in
the nasal epithelium following exposure to 300 ppm toluene, and both failed to report a
treatment-related effect. Available data from acutely-exposed humans demonstrates that nasal
and/or ocular irritation is not reported by subjects until the airborne toluene concentration reaches
100 ppm (Echeverria et al., 1989; Andersen et al., 1983), while subtle neurologic changes can be
noted at lower concentrations. Thus, it appears that neurologic effects are the most sensitive
effect observed in humans exposed to airborne toluene. Only one study in animals has reported
persistent neurobehavioral effects at concentrations known to cause similar effects in humans;
von Euler et al. (2000) reported diminished performance in the water maze test in rats 4 weeks
after exposure to 80 ppm toluene for 6 hours/day, 4 days/week for 4 weeks. Possible reasons for
the lack of animal neurologic data at low toluene concentrations include the limitations on
sample size in animal studies and the ability of human neurologic examinations to reveal more
subtle changes in neurologic endpoints relative to animal neurotoxicity assays. Benignus et al.
(1998) address the issue of apparent rat-human sensitivity differences to toluene exposure. After
On a molecular scale, little is known about mechanisms by which toluene produces acute
or residual CNS effects. The Meyer-Overton theory of partitioning of the parent compound into
membrane lipids has been widely accepted for a century (Franks and Lieb, 1985, 1987).
Recently, it has been proposed that the presence of solvent molecules in cholesterol-filled
intersticies between phospholipids and sphingolipids changes membrane fluidity, thereby altering
intercellular communication and normal ion movements (Engelke et al., 1996). It is not known if
this mechanism is involved in the chronic effects of inhaled toluene, but the observed neural
demyeliniazation in toluene abusers (Rosenberg et al., 1988a,b) would be suggestive evidence of
such a role. An alternative hypothesis is that toluene partitions into hydrophobic regions of
Under the 1986 Guidelines for Carcinogen Risk Assessment (U.S. EPA, 1986a), toluene
is classified in Group D (Not Classifiable as to Human Carcinogenicity), based on inadequate
data on the carcinogenicity of toluene in humans and inadequate evidence of carcinogenicity in
animals. This classification has not changed from the previous IRIS entry (9/7/1988). Under the
draft revised cancer guidelines (U.S. EPA, 1999), data are inadequate for an assessment of
human carcinogenic potential of toluene, because studies of humans chronically-exposed to
toluene are inconclusive, toluene was not carcinogenic in adequate inhalation cancer bioassays of
rats and mice exposed for life (CIIT, 1980; NTP, 1990a), but increased incidences of mammary
cancer and leukemia were reported in a lifetime rat oral bioassay at a dose level of 500 mg/kg
day, but not at 800 mg/kg-day (Maltoni et al., 1997). Toluene has generally not been found to be
genotoxic in short-term testing.
NTP (1990) has conducted a 2-year inhalation carcinogenicity study in F344 rats and
B6C3F1 mice, and found no evidence for carcinogenicity in either sex of either species at
exposure levels up to 1200 ppm. Another inhalation carcinogenicity study in F344 rats (CIIT,
1980; Gibson and Hardisty, 1983) likewise reported no evidence for carcinogenic effects of
toluene at exposure levels up to 300 ppm. A lifetime carcinogenicity study in Sprague-Dawley
rats by the oral route (Maltoni et al., 1997) was suggestive of potential carcinogenic effects of
toluene, but the dose-response relationships were not well defined (i.e., the 500-mg/kg animals
had considerably more tumors than those in the 800-mg/kg group) and study details were
inadequately reported.
Available studies examining the genotoxic effects of toluene have generally reported
negative results. Toluene was found to be nonmutagenic in reverse mutation assays with S.
typhimurium (Mortelmans and Riccio, 1980; Nestmann et al., 1980; Bos et al., 1981; Litton
Bionetics, Inc., 1981; Snow et al., 1981; Connor et al., 1985; Nakamura et al., 1987; NTP,
1990a) and E. coli (Fluck et al., 1976; Mortelmans and Riccio, 1980), with and without
metabolic activation. Toluene did not induce mitotic gene conversion (Litton Bionetics, Inc.,
1981; Mortelmans and Riccio, 1980) or mitotic crossing over (Mortelmans and Riccio, 1980) in
S. cerevisiae. Although Litton Bionetics, Inc. (1981) reported that toluene did not cause
increased chromosomal aberrations in bone marrow cells, several Russian studies (Lyapkalo,
1973; Dobrokhotov and Einkeev, 1977) report toluene as effective in causing chromosomal
damage in bone marrow cells of rats. There was no evidence of chromosomal aberrations in
blood lymphocytes of workers exposed to toluene only (Forni et al., 1971; Maki-Paakkanen et
al., 1980), although a slight increase was noted in workers co-exposed to toluene and benzene
(Forni et al., 1971; Funes-Craviota et al., 1977). This finding is supported by studies of cultured
Only limited data exist that examine the potential differences in susceptibility to toluene
between children and adults. Children have been demonstrated to have differences in levels of
CYP enzymes and several phase II detoxification enzymes (e.g., N-acetyl transferases, UDP
glucuronyl transferases, and sulfotransferases) relative to adults (Leeder and Kearns, 1997;
Nakajima et al., 1992; Vieira et al., 1996), as well as other physiological differences (e.g.,
children have higher brain mass per unit of body weight, higher cerebral blood flow per unit of
brain weight, and higher breathing rates per unit of body weight: see Snodgrass [1992]).
However, data on the possible contributions of these differences to potential age-related
differences are lacking.
Transfer of toluene to nursing infants from breast milk of currently exposed mothers is
expected to be a possibility because of the lipophilicity of toluene and the relatively high lipid
content of breast milk. Elimination kinetics data for nonpregnant or nonlactating humans and
rats following toluene exposure, however, indicate that most absorbed toluene is rapidly
eliminated from the body and that a much smaller portion (that which gets into adipose tissues) is
slowly eliminated (Leung and Paustenbach, 1988; Löf et al., 1993; Pierce et al., 1996, 1999;
Pellizzari et al., 1992; Rees et al., 1985). Thus, mobilization during pregnancy or lactation of
stored toluene from preconception exposure may not be a major concern.
Fisher et al. (1997) developed a human PBPK model that predicts transfer of toxicant via
lactation from a mother to a nursing infant and used the model to estimate the amount of toluene
that an infant would ingest via milk if the mother was occupationally exposed to toluene at the
ACGIH (2000) Threshold Limit Value (TLV = 50 ppm) throughout a workday. The model
predicted that such an infant would have a daily oral intake of 0.46 mg toluene/day. It should be
noted, however, that no human (or animal) studies are available regarding in vivo distribution of
toluene into breast milk or elimination kinetics from breast milk, and the Fisher et al. (1997)
PBPK model has not been validated with in vivo data.
Available studies in humans and animals have not definitively demonstrated whether sex-
related differences in the toxicity of toluene exist. Human occupational studies have failed to
report sex-related differences in effects, with the exception of the study of Plenge-Bönig and
Karmaus (1999), which reported decreased fertility in the occupationally-exposed women, but
not in occupationally-exposed men. In rats and mice exposed to toluene orally for 13 weeks
(NTP, 1990a), males of both rats and mice showed toxic effects at lower doses than females.
It has been stated that the effects of toluene exposure on color vision may be potentiated
in the elderly and consumers of alcohol (Zavalic et al., 1998a, b). It is not known if this effect is
related to the possible differences in metabolism rates under these conditions or some other
inherent property related to age or alcohol consumption. It should be noted that color vision
decreases with age (Ruddock, 1965; Bowman et al., 1984), diabetes (Matyjavri, 1992; Utku and
Atmaca, 1992) and alcohol intake (Russell et al, 1980; Mergler et al., 1988).
An RfD of 0.2 mg/kg-day was previously entered on the IRIS data base in 1990. This
value was based on the NOAEL of 223 mg/kg-day for increased relative liver and kidney weights
in rats identified by the 13 week NTP gavage study (NTP, 1990a). A total uncertainty factor of
1000 was used to account for inter- and intraspecies extrapolations, for subchronic-to-chronic
extrapolation and for limited reproductive and developmental toxicity data. Individual
uncertainty factors were not identified. The two studies by Hsieh et al. (1989, 1990) were not
discussed in the previous assessment of the RfD for toluene.
Despite its shorter duration and examination of only one sex of animals, the Hsieh et al.
(1989) study was selected as the principal study for derivation of the RfD. Decreased thymus
weight (32% decrease compared to controls) and decreased antibody response (>40%) were
observed in the Hsieh et al. (1989) study at a dose of 105 mg/kg-day. The biological relevance of
immunological effects bioassays is difficult to gauge and is the subject of some debate (Luster et
al. 1992). However, given that thymus weight and antibody response was significantly decreased,
coupled with the observed effects on host defenses following inhalation exposure (Aranyi et al.,
1985), immunological effects were identified as the primary effect with decreased thymus weight
as the critical effect. Decreased antibody response was not chosen as the critical effect due to a
lack of confidence that the dose-response relationship is representative of chemical-induced
immunological effects. Both the NTP (1990) and Hsieh et al. (1989) studies identified NOAEL
and LOAEL values for altered relative liver weight, but the LOAEL of 105 mg/kg-day and
NOAEL of 22 mg/kg-day identified by Hsieh et al. (1989) is lower than the NOAEL of 223
mg/kg-day defined by NTP (1990). However, the changes in liver weight were not correlated
with any histological or anatomical alterations, thus this endpoint is not considered a critical
effect.
An additional factor that supports the use of the Hsieh et al. (1989) study is that the
method of oral exposure, i.e., ingestion via drinking water, is considered preferable to exposure
via gavage that was utilized in the NTP (1990) study. For these reasons, and because data are
inadequate to determine which species (rat or mouse) or which mouse strain (B6C3F1 or CD-1)
is a more appropriate model for oral toluene toxicity in humans, the study with the most
conservative (i.e., health protective) NOAEL, that from Hsieh et al. (1989), was selected for
derivation of the RfD.
The RfD was derived by the benchmark dose approach (BDS. Version 1.3). The
benchmark response (BMR) was defined as the default of a change of one standard deviation
(U.S. EPA, 2000d). Benchmark analysis was performed for thymus weight changes as an
indicator of potential immunological effects. A BMDL of 41 mg/kg-day was derived and used as
the point of departure. Details of the model results are presented in Appendix B.
Several well-documented PBPK models for toluene exist for rats and humans. However,
to date they have not been applied to mice, nor have comparisons to in vivo pharmacokinetic data
in mice been utilized to validate the applicability of these models in that species. The absence of
an empirical PBPK model for mice precludes utilization of such a model in extrapolating from
mice to humans.
The BDL of 41 mg/kg-day for decreased thymus weight from the Hsieh et al. (1989)
study was utilized as the basis for the calculation of the RfD.
Total UP – 1000.
A total uncertainty factor of 1000 was applied to this effect level: 10 for extrapolation for
interspecies differences (UFA; animal to human), 10 for consideration of intraspecies variation
(UFA; human variability), and 10 for use of a subchronic study to estimate chronic effects (UFS;
duration of exposure). The total UF = 10 x 10 x 10 = 1000.
A 10-fold uncertainty factor for intraspecies differences (UFA) was used to account for
potentially sensitive human subpopulations. This UF was not reduced because of the lack of
human oral exposure information.
A 10-fold uncertainty factor was used to account for extrapolating from less than chronic
results on experimental animals when there are no useful long-term human data (UHs).
An uncertainty factor was not needed to account for extrapolating from a LOAEL to a
NOAEL because BMD modeling was used.
RfD = NOAEL ÷ UF
= 41mg kg day ÷ 1000
= 0.04 mg kg day
Confidence in the principal study is low, because while the study examined what appears
to be the most sensitive endpoint, the study was of only 28 day duration and in a single species
and sex. Confidence in the data base is rated medium due to the lack of chronic animal data and
uncertainties regarding potential discrepancies in the low dose effects between the NTP (1990)
13 week study and the Hsieh et al. (1989) 28-day study. There is low confidence in the resulting
RfD.
Two studies (Zavalic et al., 1998a; Eller et al., 1999) have identified no-effect levels in
occupationally-exposed humans as shown in Table 1 (Section 4.5.2). Eller et al. (1999) reported
no neurobehavioral effects in workers exposed to 25 to 32 ppm toluene for 1 to 12 years, while
workers exposed to greater concentrations (> 100 ppm) showed statistically significant
neurologic impairment. Zavalic et al. (1998a) reported a NOAEL of 32 ppm and a LOAEL of
132 ppm for alterations in color vision in exposed workers. A number of additional studies (see
Table 1), however, have identified effect levels for neurologic endpoints in exposed humans at
levels between 40 and 100 ppm. The available studies each have a number of limitations.
However, when considered jointly, these studies indicate that humans repeatedly exposed to
The study of Zavalic et al. (1998a) was selected as the principal study for derivation of an
inhalation RfC. It is an adequate cross-sectional study of chronically-exposed humans which
identified both a NOAEL (32 ppm) and LOAEL (132 ppm) for neurologic effects (impaired color
vision). Impaired color vision is the critical effect. Effects were correlated with both airborne
and blood toluene concentrations. The study of Eller et al. (1999) defined a similar NOAEL (25
to 32 ppm) for decreased performance in neurobehavioral and neuropsychological tests, but the
effect levels in this study were less clearly characterized. Both of these NOAEL values lie
slightly below the 40 to100 ppm range where available data (see Section 4.5.2, Table 1) suggest
that persistent neurological effects in humans chronically-exposed to toluene begin to manifest.
An RfC of 0.4 mg/m3 was previously entered on the IRIS data base in 1992. This value
was based on the LOAEL of 88 ppm (332 mg/m3) for decreased performance in neurological
tests identified by the study of Foo et al. (1990). This LOAEL was adjusted to a continuous
exposure level of 119 mg/m3, and a total uncertainty factor of 300 (10 for use of a LOAEL, 10
for intrahuman variability, and 3 for data base deficiencies, including the lack of data and well-
characterized laboratory animal exposures evaluating neurotoxicity and respiratory irritation) was
applied. While the Foo et al. (1990) study was selected as the key study for the previous RfC
derivation, a number of other studies provided evidence that neurological alterations would occur
at toluene concentrations at or near the 88 ppm LOAEL. In addition, the Foo et al. (1990) study
does not address co-exposure to other solvents and workers were exposed for a shorter period of
time (i.e., 5.7 ±3.2 years).
In addition to neurologic effects in humans, the previous RfC was also based on irritation
of the upper respiratory tract, specifically the nasal epithelium, as reported in the chronic NTP
(1990) study in rats. However, these effects occurred in rats exposed to high concentrations (600
ppm or greater) of toluene, and did not show an appreciable increase with increasing
concentration (i.e., the incidence of the lesions was greater at 600 ppm than at 1200 ppm).
Support that the nasal lesions are a high-exposure phenomenon comes from the results of a
chronic inhalation study in rats performed by CIIT (1980), which reported no effects on the nasal
epithelium of animals exposed to 300 ppm. A 28 day inhalation study in rats (30 and 300 ppm)
likewise failed to demonstrate treatment-related lesions in the nasal epithelium (Poon et al.,
1994). Acute studies in humans have demonstrated that subjective reports of irritation of the
nose and/or eyes occurs at exposure levels of 100 ppm or greater (Baelum et al., 1985, 1990;
Echeverria et al., 1989; Andersen et al., 1983), but not at exposures below 100 ppm (Echeverria
et al., 1989; Andersen et al., 1983). Because neurologic endpoints are a more sensitive endpoint
for exposed humans, they alone were selected as the critical endpoint in the previous assessment
for derivation of the inhalation RfC.
The Zavalic et al. (1998a) study examined two exposure concentrations other than
controls, and identified both a NOAEL of 32 ppm and a LOAEL of 132 ppm. Only one effect
level was identified, i.e., 132 ppm, thus limiting the study’s ability to describe the exposure-
response relationship. Actual data points and statistics are not available; data are presented in
graphical form only. Other studies of occupationally-exposed humans have identified adverse
effect levels for neurologic effects ranging from 40 to 100 ppm, further supporting a NOAEL of
32 ppm. For these reasons, the data were not modeled and a benchmark concentration (BMC)
approach was not used. A number of available PBPK models for toluene inhalation exist, but
because the Zavalic et al. (1998a) study does not require extrapolation from animals to humans,
from subchronic to chronic exposure duration, or from another route of exposure, applying these
models is unlikely to reduce the uncertainty associated with the derivation of an RfC. The
NOAEL value identified in the Zavalic et al. (1998a) study was, therefore, used for the derivation
of the RfC.
The NOAEL of 32 ppm (121 mg/m3) from the Zavalic et al. (1998a) study was adjusted
from an occupational exposure scenario to continuous exposure conditions as follows:
VEho 5days
NOAEL( ADJ ) = NOAEL × ×
VEh 7 days
10m 3 5 days
= 121mg m × 3
×
20m 3 7 days
= 43mg m 3
Where:
VEho = human occupational default minute volume (10 m3 breathed during the workday)
VEh = human ambient default minute volume (20 m3 breathed during the entire day)
August 2002
Draft-Do Not Cite or Quote
43
Total UF–10
A total uncertainty factor of 10 was applied to this effect level, i.e., 10 for consideration
of intraspecies variation (UFH; human variability).
A 10-fold uncertainty factor for intraspecies differences (UFH) was used to account for
potentially sensitive human subpopulations.
An uncertainty factor to account for extrapolating from less than chronic results was not
necessary (UFs). Workers were exposed to toluene for a mean duration of 16 - 18 years.
An uncertainty factor was not needed to account for extrapolating from a LOAEL to a
NOAEL because a NOAEL was available.
The data base for inhalation exposure to toluene is considered adequate. A single study
of reproductive effects of humans occupationally exposed to toluene (Plenge-Bönig and
Karmaus, 1999) reported no effects on male fertility, but a significant association between female
exposure and reduced fertility was found. However, exposure levels for this study were not
quantified, and confounding variables were not distributed evenly among the control and exposed
groups, which hindered statistical adjustment. Numerous animal studies have demonstrated
reproductive and developmental effects of toluene only at exposure levels which result in
maternal toxicity (e.g., decreased maternal bodyweight). In addition, a 2-generation inhalation
toxicity study is available.
Confidence in the principal study is medium. The Zavalic et al. (1998a) study is an
adequate cross-sectional study in chronically-exposed humans that examined appropriate
endpoints of concern at multiple exposure levels. However, only one effect level was identified,
thus limiting the study’s ability to describe the exposure-response relationship. Confidence in
the database is high. Chronic studies in humans exist which identify subtle neurological
alterations as a sensitive effect of long-term repeated toluene exposure at concentrations in the
range of 40 to150 ppm. In addition, numerous animal studies of the reproductive and
Not applicable.
Not applicable.
Toluene (CAS no.108-88-3) has the chemical formula C7H8 (structural formula C6H5CH3)
and a molecular weight of 92.14. At room temperature, toluene is a clear-to-amber colorless
liquid with a pungent, benzene-like odor. Toluene has a low vapor pressure which can result in
volatilization into the air. It is flammable, with a flash point of 4.4oC. Toluene is strongly
reactive with a number of chemicals, particularly nitrogen-containing compounds, and may react
with some plastics. Toluene is used as part of an additive to gasoline mixtures (BTEX) to
increase octane ratings, in benzene production, and as a solvent in paints, coatings, inks,
adhesives, and cleaners. Additionally, toluene is used in the production of nylon, plastics, and
polyurethanes. Toluene was once used as an anthelminthic agent against roundworms and
hookworms.
Data on the effects of toluene in humans following oral exposure are limited to case
reports of accidental oral ingestions. Both a 4 week study in mice (Hsieh et al., 1989, 1990) and
a 13 week study in rats (NTP, 1990a) identified changes in liver weights as the most sensitive
toxicologically relevant endpoints, with the 4 week study also reporting decreased thymus weight
and decreased antibody response at the same exposure level. The 4 week studies in mice also
noted changes in brain neurochemistry at lower exposure levels. However, these effects were not
correlated with neurobehavioral of neuroanatomical changes, thus the relevance of these more
sensitive changes is unclear.
A number of occupational studies have examined the effects of toluene exposure via
inhalation. The most sensitive effects observed in humans following inhalation exposure are
neurologic effects, including altered color vision, dizziness, fatigue, headache, and decreased
performance in neurobehavioral tests. Exposure to higher levels in humans and animals have
In mothers who inhaled very high levels of toluene during pregnancy, the children
showed a number of physical (small midface, deep-set eyes, micrognathia, and blunting of the
fingertips) and clinical (microcephaly, CNS dysfunction, attention deficits, and developmental
delay/mental deficiency) changes attributed to toluene. Animal studies of toluene inhalation
have revealed delayed neurodevelopment and decreased offspring weight, though generally only
at levels that also resulted in maternal toxicity. A series of studies in rats examining the
developmental effects of oral toluene exposure found decreases in fetal body and organ weights,
but only at levels equal to or greater than those that caused maternal toxicity. Gross
malformations were not noted at any exposure level.
6.2.1. Noncancer/Oral
There are no chronic or subchronic oral dose-response data for toluene in humans. A
single lifetime gavage study in rats (Maltoni et al., 1997) did not adequately examine noncancer
endpoints, and was not suitable for use in derivation of an RfD. A 13 week gavage study by NTP
(1990) identified a NOAEL of 223 mg/kg-day and a LOAEL of 446 mg/kg-day for increased
relative liver and kidney weights in rats. The same study reported changes in relative liver
weight at all doses of B6C3F1 mice, though changes in absolute liver weight occurred only in the
223 mg/kg-day (the lowest dose level examined) and 1786 mg/kg-day groups. Hsieh et al.
(1989) conducted a 4 week drinking water study in CD-1 mice, which reported significant
changes in relative liver and thymus weights compared to controls at 105 mg/kg-day, but not at
22 or 5 mg/kg-day; absolute liver weights were not reported. No histochemical effects on the
liver was noted in either study. The Hsieh et al. (1989) study also reported changes in ex vivo
immune response to mitogen stimulation at all exposure levels and significantly reduced
antibody response at the highest concentration. A subsequent study by the same authors (Hsieh et
al., 1990) reported neurochemical changes in the brains of CD-1 mice exposed to 5 mg/kg-day
for 4 weeks. The biological significance of the neurochemical changes cannot be determined.
Immunological effects were selected as the primary toxicological endpoint with decreased
6.2.2. Noncancer/Inhalation
Data in both humans and animals are inadequate to evaluate potential associations
between toluene exposure and human cancer. Lifetime inhalation studies in both rats and mice
(CIIT, 1980; NTP, 1990a) failed to report any increase in carcinogenicity as a result of toluene
exposure. A lifetime gavage study in rats (Maltoni et al., 1997) reported an increase in toluene-
induced tumors in both males and females, but the study was lacking in experimental detail and
the dose-response relationship was inverse at the highest dose, making the interpretation of the
study difficult. Toluene is, therefore, classified as group D, not classifiable as to human
7. REFERENCES
Abbate, C., C. Giorgianni, F. Munao, et al. 1993. Neurotoxicity induced by exposure to toluene. An
electrophysiologic study. Int. Arch. Occup. Environ. Health. 64: 389-392.
ACGIH (American Conference of Governmental Industrial Hygienists). 2000. Threshold Limit Values
for Chemical Substances and Physical Agents and Biological Exposure Indices 2000. ACGIH,
Cincinnati, OH.
Aitio, A., K. Pekari and J. Jarvisalo. 1984. Skin absorption as a source of error in biological monitoring.
Scand. J. Work. Environ. Health. 10: 317-320.
Ameno, K., C. Fuke, S. Ameno, et al. 1989. A fatal case of oral ingestion of toluene. Forensic Sci. Int.
41: 255-260.
Ameno, K., T. Kiriu, C. Fuke, et al. 1992. Regional brain distribution of toluene in rats and in a human
autopsy. Arch. Toxicol. 66: 153-156.
Andersen, I., G.R. Lundqvist, L. Molhave, et al. 1983. Human response to controlled levels of toluene
in six-hour exposures. Scand. J. Work Environ. Health. 9: 405-418.
Anderson, H.R., B. Dick, R.S. Macnair, et al. 1982. An investigation of 140 deaths associated with
volatile substance abuse in the United Kingdom (1971-1981). Hum. Toxicol. 1: 207-221.
Angerer, J. 1979. Occupational chronic exposure to organic solvents: VII. Metabolism of toluene in
man. Int. Arch. Occup. Environ. Health. 43: 63-67.
Angerer, J., M. Schildbach and A. Kramer. 1998. S-p-toluylmercapturic acid in the urine of workers
exposed to toluene: A new biomarker for toluene exposure. Arch. Toxicol. 72(2): 119-23.
Anttila, A., E. Pukkala, R. Riala, et al. 1998. Cancer incidence among Finnish workers exposed to
aromatic hydrocarbons. Int. Arch. Occup. Environ. Health. 71: 187-193.
API (American Petroleum Institute). 1981. 26-Week inhalation toxicity study of toluene in the rat.
Conducted by Bio/dynamics Inc. and Institute of Neurotoxicity, Albert Einstein College of Medicine for
API, Washington, DC.
ATSDR (Agency for Toxic Substances and Disease Registry). 2000. Toxicological Profile for Toluene.
ATSDR, U.S. Public Health Service, Atlanta, GA.
Baelum, J., M. Dossing, S.H. Hansen, et al. 1987. Toluene metabolism during exposure to varying
concentrations combined with exercise. Int. Arch. Occup. Environ. Health. 59: 281-294.
Baelum, J., G. Lundqvist, L. Molhave and N.T. Andersen. 1990. Human response to varying
concentrations of toluene. Int. Arch. Occup. Environ. Health. 62(1): 65-71.
Baelum, J., L. Molhave, S. Honore Hansen, et al. 1993. Hepatic metabolism of toluene after
gastrointestinal uptake in humans. Scand. J. Work. Environ. Health. 19(1): 55-62.
Balster, R.L. 1998. Neural basis of inhalant abuse. Drug Alcohol Dep. 512: 207-214.
Bauchinger, M., E. Schmid, J. Dresp, et al. 1982. Chromosome change in lymphocytes after
occupational exposure to toluene. Mutat. Res. 102: 439-445.
Benignus, V.A., K.E. Muller, J.A. Graham, et al. 1984. Toluene levels in blood and brain of rats as a
function of toluene level in inspired air. Environ. Res. 33: 39-46.
Benoit, F.M., W.R. Davidson, A.M. Lovett, et al. 1985. Breath analysis by API/MS human exposure to
volatile organic solvents. Int. Arch. Occup. Environ. Health. 55: 113-120.
Boey, K.W., S.C. Foo and J. Jeyaratnam. 1997. Effects of occupational exposure to toluene: a
neuropsychological study on workers in Singapore. Ann. Acad. Med. Singapore. 26(2): 84-7.
Boman, A., G. Hagelthorn and K. Magnusson. 1995. Percutaneous absorption of organic solvents
during intermittent exposure in guinea pigs. Acta. Derm. Venereol. 75(2): 114-9.
Bos, R.P., R.M.E. Brouns, R. van Doorn, et al. 1981. Non-mutagenicity of toluene, o-, m- and p-xylene,
o-methylbenzylalcohol and o-methylbenzylsulfate in the Ames assay. Mutat. Res. 88(3): 273-279.
Byrne, A., B. Kirby, T. Zibin, et al. 1991. Psychiatric and neurological effects of chronic solvent abuse.
Can. J. Psychiatry. 36: 735-738.
Campo, P., R. Lataye, B. Cossec, et al. 1998. Combined effects of simultaneous exposure to toluene and
ethanol on auditory function in rats. Neurotoxicol. Teratol. 20(3): 321-332.
Caravati, E.M. and P.J. Bjerk. 1997. Acute toluene ingestion toxicity. Ann. Emerg. Med. 30(6): 838-
839.
Carlsson, A. 1982. Exposure to toluene: Uptake, distribution and elimination in man. Scand. J. Work.
Environ. Health. 8: 43-55.
Cavalleri, A., F. Gobba, E. Nicali and V. Fiocchi. 2000. Dose-related color vision impairment in
toluene-exposed workers. Arch. Env. Health. 55(6): 399-404.
Connor, T.H., J.C. Theiss, H.A. Hanna, et al. 1985. Genotoxicity of organic chemicals frequently found
in the air of mobile homes. Toxicol. Lett. 25: 33-40.
Courtney, K.D., J.E. Andrews, J. Springer, et al. 1986. A perinatal study of toluene in CD-1 mice.
Fund. Appl. Toxicol. 6: 145-154.
Cruz, S.L., T. Mirshahi, B. Thomas, et al. 1998. Effects of the abused solvent toluene on recombinant N
methyl-D-aspartate and non-N-methyl-D-aspartate receptors expressed in Xenopus oocytes. J. Pharmacol.
Exp. Ther. 286: 334-340.
Dalgaard, M., A. Hossaini, K.S. Hougaard, et al. 2001. Developmental toxicity of toluene in male rats:
effects on semen quality, testis morphology, and apoptotic neurodegeneration. Arch. Toxicol. 75: 103-
109.
DaSilva, V.A., L.R. Malheiros and F.M.R. Bueno. 1990. Effects of toluene exposure during gestation
on neurobehavioral development of rats and hamsters. Brazil J. Med. Biol. Res. 23: 533-537.
DeJongh, J. and B.J. Blaauboer. 1996. Simulation of toluene kinetics in the rat by a physiologically
based pharmacokinetic model with application of biotransformation parameters derived independently in
vitro and in vivo. Fundam. Appl. Toxicol. 32(2): 260-8.
DeJongh, J. and B.J. Blaauboer. 1997. Evaluation of in vitro-based simulations of toluene uptake and
metabolism in rats. Toxicol. in Vitro. 11: 485-489.
Devathasan, G., D. Low, P.C. Teoh, et al. 1984. Complications of chronic glue (toluene) abuse in
adolescents. Aust. NZ J. Med. 14: 39-43.
Dobrokhotov, V.B. and M.I. Enikeev. 1977. The mutagenic action of benzene, toluene and a mixture of
these hydrocarbons in a chronic test. Gig. Sanit. 42: 32-34. (Rus) (Evaluation based on an English
translation)
Dossing, M., J.B. Aelum, S.H. Hansen, et al. 1983. Urinary hippuric acid and orthocresol excretion in
man during experimental exposure to toluene. Br. J. Ind. Med. 40: 470-473.
Dutkiewicz, T. and H. Tyras. 1968. Skin absorption of toluene, styrene and xylene by man. Br. J. Ind.
Med. 25: 243.
Dyer, R.S., M.S. Bercegeay and L.M. Mayo. 1988. Acute exposures to p-xylene and toluene alter visual
information processing. Neurotox. Teratol. 10: 147-153.
Echeverria, D., L. Fine, G. Langolf, et al. 1989. Acute neurobehavioral effects of toluene. Br. J. Ind.
Med. 46(7): 483-495.
Engelke, M., H. Tahti, and L. Vaalavirta. 1996. Perturbation of artificial and biological membranes by
organic compounds of aliphatic, alicyclic, and aromatic structure. Toxicol. In Vitro 10: 111-115.
Filley, C.M., R.K. Heaton and N.L. Rosenberg. 1990. White matter dementia in chronic toluene abuse.
Neurology. 40: 532-534.
Fisher, J., D. Mahle, L. Bankston, et al. 1997. Lactational transfer of volatile chemicals in breast milk.
American Ind. Hyg. Assoc. J. 58(6): 425-431.
Fluck, E.R., L.A. Poirier and H.W. Ruelius. 1976. Evaluation of a DNA polymerase-deficient mutant of
E. coli for rapid detection of carcinogens. Chem. Biol. Interact. 15: 219-231.
Foo, S.C., J. Jeyaratnam and D. Koh. 1990. Chronic neurobehavioral effects of toluene. Br. J. Ind. Med.
47(7): 480-484.
Forni, A., E. Pacifico and A. Limonta. 1971. Chromosome studies in workers exposed to benzene or
toluene or both. Arch. Environ. Health. 22(3): 373-378.
Franks, N.P. and W.R. Lieb. 1985. Mapping of general anaesthetic target sites provides a molecular
basis for cutoff effects. Nature. 316: 349-351.
Franks, N.P. and W.R. Lieb. 1987. Anaesthetics on the mind. Nature. 328: 113-114.
Gericke, C., B. Hanke, G. Beckmann, et al. 2001. Multicenter field trial on possible health effects of
toluene. III. Evaluation of effects after long-term exposure. Toxicology. 168: 185-209.
Gerin, M., J. Siemiatycki, M. Desy and D. Krewski. 1998. Associations between several sites of cancer
and occupational exposure to benzene, toluene, xylene, and styrene: results of a case-control study in
Montreal. Am. J. Ind. Med. 34: 144-156.
Gerkin, R.D. and F. LoVecchio. 1998. Rapid reversal of life-threatening toluene-induced hypokalemia
with hemodialysis. J. Emerg. Med. 16(5): 723-725.
Gerner-Smidt, P. and U. Friedrich. 1978. The mutagenic effect of benzene, toluene and xylene studied
by the SCE technique. Mutat. Res. 58(2-3): 313-316.
Ghantous, H. and B.R.G. Danielsson. 1986. Placental transfer and distribution of toluene, xylene and
benzene and their metabolites during gestation in mice. Biol. Res. Pregnancy Perinatol. 7(3): 98-105.
Gibson, J.E. and J.F. Hardisty. 1983. Chronic toxicity and oncogenicity bioassay of inhaled toluene in
Fischer-344 rats. Fund. Appl. Toxicol. 3: 315-319.
Gospe, S.M. Jr., D.B. Saeed, S.S. Zhou, et al. 1994. The effects of high-dose toluene on embryonic
development in the rat. Pediatr. Res. 36(6): 811-5.
Gospe, S.M. Jr., S.S. Zhou, D.B. Saeed, et al. 1996. Development of a rat model of toluene-abuse
embryopathy. Pediatr. Res. 40(1): 82-87.
Gospe, S.M. Jr. and S.S. Zhou. 1998. Toluene abuse embryopathy: Longitudinal neurodevelopmental
effects of prenatal exposure to toluene in rats. Reprod. Toxicol. 12(2): 119-126.
Gospe, S.M. Jr. and S.S. Zhou. 2000. Prenatal exposure to toluene results in abnormal neurogenesis and
migration in rat somatosensory cortex. Pediatr. Res. 47(3): 362-368.
Haglund, U., I. Lundberg and L. Zech. 1980. Chromosome aberrations and sister chromatid exchanges
in Swedish paint industry workers. Scan. J. Environ. Health. 6: 291-298.
Harabuchi, I., R. Kishi, T. Ikeda, et al. 1993. Circadian variations of acute toxicity and blood and brain
concentrations of inhaled toluene in rats. Br. J. Ind. Med. 50: 280-286.
Hass, U., S.P. Lund, K.S. Hougaard and L. Simonsen. 1999. Developmental nurotoxicity after toluene
inhalation exposure in rats. Neurotoxicol. Teratol. 21(4): 349-357.
Hersh, J.H., P.E. Podruch, G. Rogers and B. Weisskopf. 1985. Toluene embryopathy. J. Pediatr. 106:
922-927.
Hougaard, K.S., U. Hass, S.P. Lund and L. Simonsen. 1999. Effects of prenatal exposure to toluene on
postnatal development and behavior in rats. Neurotoxicol. Teratol. 21(3): 241-250.
Hsieh G.C., R.P. Sharma and R.D. Parker. 1989. Immunotoxicological evaluation of toluene exposure
via drinking water in mice. Environ. Res. 49: 93-103.
Hsieh G.C., R.P. Sharma, R.D. Parker, et al. 1990. Evaluation of toluene exposure via drinking water on
levels of regional brain biogenic monoamines and their metabolites in CD-1 mice. Ecotoxicol. Environ.
Saf. 20: 175-184.
Huang, J., N. Asaeda, Y. Takeuchi, E. Shibata, N. Hisanaga, Y. Ono and K. Kato. 1992. Dose
dependent effects of chronic exposure to toluene on neuronal and glial cell marker proteins in the central
nervous system of rats. Brit. J. Ind. Med. 49: 282-286.
Hunnewell, J. and N.R. Miller. 1998. Bilateral internuclear opthalmoplegia related to chronic toluene
abuse. J. Neuroophthalmol. 18(4): 277-280.
IARC. 1999. IARC monographs on the evaluation of carcinogenic risks to humans: Re-evaluation of
some organic chemicals, hydrazine and hydrogen peroxide. Volume 71. Part 2. Lyon, France: World
Health Organization, International Agency for Research on Cancer, 829-864.
Ikeda, N., H. Takahashi, K. Umetsu, et al. 1990. The course of respiration and circulation in
toluene-sniffing. Forensic Sci. Intern. 44: 151-158.
Inoue, O., K. Seiji, T. Watanabe, et al. 1986. Possible ethnic difference in toluene metabolism: A
comparative study among Chinese, Turkish and Japanese solvent workers. Toxicol. Lett. 34: 167-174.
Jonai, H. and M. Sato. 1988. Exposure indices for painters exposed to toluene and xylene at low
concentrations. Ind. Health. 26: 197-202.
Jone, C.M. and A.H.B. Wu. 1988. An unusual case of toluene-induced metabolic acidosis. Clin. Chem.
34(12): 2596-2599.
Kamijima, M., Y. Nakazawa, M. Yamakawa, et al. 1994. Metabolic acidosis and renal tubular injury
due to pure toluene inhalation. Arch. Environ. Health. 49(5): 410-3.
Kamijo, Y., K. Soma, I. Hasegawa, et al. 1998. Fatal bilateral adrenal hemorrage following acute
toluene poisoning: A case report. J. Toxicol. Clin. Toxicol. 36(4): 365-368.
Kaneko, T., T. Koizumi, T. Takezaki, et al. 1992. Urinary calculi associated with solvent abuse. J.
Urol. 147: 1365-1366.
Kawai, T., T. Yasugi, K. Mizunuma, et al. 1992a. Comparative evaluation of urinalysis and blood
analysis as means of detecting exposure to organic solvents at low concentrations. Int. Arch. Occup.
Environ. Health. 64: 223-234.
Kawai, T., T. Yasugi, K. Mizunuma, et al. 1992b. Monitoring of workers exposed to a mixture of
toluene, styrene and methanol vapours by means of diffusive air sampling, blood analysis and urinalysis.
Int. Arch. Occup. Environ. Health. 63: 429-435.
King, M.D., R.E. Day, J.S. Oliver, et al. 1981. Solvent encephalopathy. Br. Med. J. 283: 663-665.
Kostas, J. and J. Hotchin. 1981. Behavioral effects of low-level perinatal exposure to toluene in mice.
Neurobehav. Toxicol. Teratol. 3: 467-469.
Lee, B., S. Lee, K. Lee, et al. 1988. Dose-dependent increase in subjective symptom prevalence among
toluene-exposed workers. Ind. Health. 26(1): 11-23.
Leeder, J.S. and G.L. Kearns. 1997. Pharmacogenetics in pediatrics: Implications for practice. Pediatr.
Clin. North Am. 44(1): 55-77.
Leung, H-W. and D.J. Paustenbach. 1988. Application of pharmacokinetics to derive biological
exposure indexes from threshold limit values. Am. Ind. Hyg. Assoc. J. 49(9): 445-450.
Löf, A., M. Wallen and E.W. Hjelm. 1990. Influence of paracetamol and acetylsalicylic acid on the
toxicokinetics of toluene. Pharmacol. Toxicol. 66: 138-141.
Löf, A., E.W. Hjelm, A. Colmsjo, et al. 1993. Toxicokinetics of toluene and urinary excretion of
hippuric acid after human exposure to 2H8-toluene. Br. J. Ind. Med. 50: 55-59.
Lyapkalo, A.A. 1973. Genetic activity of benzene and toluene. Gig. Tr. Prof. Zabol. 17(3): 24-28.
(Rus.) (Evaluation based on an English translation provided by the U.S. EPA.)
Maas, E.F., J. Ashe, P. Spiegel, et al. 1991. Acquired pendular nystagmus in toluene addiction.
Neurology. 41: 282-285.
Maestri, L., S. Ghittori and M. Imbriani. 1997. Determination of specific mercapturic acids as an index
of exposure to environmental benzene, toluene, and styrene. Ind. Health. 35: 489-501.
Maltoni, C., A. Ciliberti, C. Pinto, et al. 1997. Results of long-term experimental carcinogenicity studies
of the effects of gasoline, correlated fuels, and major gasoline aromatics on rats. Ann. N.Y. Acad. Sci.
837: 15-52.
McDonald, J.C., J. Lavoie, R. Cote and A.D. McDonald. 1987. Chemical exposures at work in early
pregnancy and congenital defect: A case-referent study. Br. J. Ind. Medicine. 44: 527-533.
McWilliams, M.L., G.D. Chen and L.D. Fechter. 2000. Low-level toluene disrupts auditory function in
guinea pigs. Toxicol. Appl. Pharmacol. 167(1): 18-29.
Mehta, C.S., P.N. Sun, A. Zikarge, et al. 1998. Acute toxicity of toluene in male and female rats: A
single oral dose exposure 2-week study. Toxic Substances Mechanisms. 17: 43-55.
Meulenbelt, J., G. de Groot and T.J. Savelkoul. 1990. Two cases of acute toluene intoxication. Br. J.
Ind. Med. 47: 417-420.
Mihic, S.J., S.J. McQuilkin, E.I. Eger, et al. 1994. Potentiation of γ-aminobutyric acid type A receptor-
mediated chloride currents by novel halogenated compounds correlated with their abilities to induce
general anesthesia. Mol. Pharmacol. 46: 851-857.
Miyagi, Y., F. Shima, K. Ishido, et al. 1999. Tremor induced by toluene misuse successfully treated by a
Vim thalamotomy. J. Neurosurg. Psych. 66: 794-796.
Monster, A.C., S. Kezic, I. van de Gevel, et al. 1993. Evaluation of biological monitoring parameters for
occupational exposure to toluene. Int. Arch. Occup. Environ. Health. 65(Suppl): 159-162.
Morgan, D.L., S.W. Cooper, D.L. Carlock, et al. 1991. Dermal absorption of neat and aqueous volatile
organic chemicals in the Fischer 344 rat. Environ. Res. 55: 51-63.
Mortelmans, K.E. and E.S. Riccio. 1980. In vitro microbiological genotoxicity assays of toluene.
Prepared by SRI International, Menlo Park, CA, under Contract No. 68-02-2947 for the U.S. EPA,
Research Triangle Park, NC. p. 25.
Murata, K., S. Araki, K. Yokoyama, et al. 1993. Cardiac autonomic dysfunction in rotogravure printers
exposed to toluene in relation to peripheral nerve conduction. Ind. Health. 31(3): 79-90.
Mutti, A. and I. Franchini. 1987. Toxicity of metabolites to the dopaminergic system and the behavioral
effects of organic solvents. Br. J. Ind. Med. 44: 721-723.
Muttray, A., U. Wolff, D. Jung and J. Konietzko. 1997. Blue-yellow deficiency in workers exposed to
low concentrations of organic solvents. Int. Arch. Occup. Environ. Health. 70: 407-412.
Muttray, A., V. Wolters, D. Jung and J. Konietzko. 1999. Effects of high doses of toluene on color
vision. Neurotox. Teratol. 21(1): 41-45.
Nakajima, T. and R-S. Wang. 1994. Induction of cytochrome P450 by toluene. Int. J. Biochem.
26(12): 1333-1340.
Nakajima, T., R-S. Wang, Y. Katakura, et al. 1992. Sex-, age- and pregnancy-induced changes in the
metabolism of toluene and trichloroethylene in rat liver in relation to the regulation of cytochrome
P45011E1 and P45011C11 content. J. Pharmacol. Exp. Ther. 261(3): 869-874.
Nakajima, T., R-S. Wang, E. Elovaara, et al. 1997. Toluene metabolism by cDNA-expressed human
hepatic cytochrome P-450. Biochem. Pharmacol. 53(3): 271-277.
Nakamura, S., Y. Oda, T. Shimada, et al. 1987. SOS-inducing activity of chemical carcinogens and
mutagens in Salmonella typhimurium TA1535/pSK1002: examination with 151 chemicals. Mutat. Res.
192: 239-246.
National Research Council. 1983. Risk assessment in the Federal Government: managing the
process. Washington, DC: National Academy Press.
Nestmann, E.R., E.G.H. Lee, T.I. Matula, et al. 1980. Mutagenicity of constituents identified in pulp
and paper mill effluents using the Salmonella/mammalian-microsome assay. Mutat. Res. 79: 203-212.
Neubert, D., G. Bochert, C. Gericke, et al. 2001. Multicenter field trial on possible health effects of
toluene. I. Toluene body burdens in workers of the rotogravure indurstry. Toxicology. 168: 139-157.
Ng, T.P., S.G. Ong, W.K. Lam, et al. 1990. Urinary levels of proteins and metabolites in workers
exposed to toluene. Int. Arch. Occup. Environ. Health. 62: 43-46.
NTP (National Toxicology Program). 1990a. Toxicology and carcinogenesis studies of toluene (CAS
No. 108-88-3) in F344/N rats and B5C3F1 mice (inhalation studies). Technical Report Series No. 371.
Research Triangle Park, NC.
NTP (National Toxicology Program). 2001. Chemical Health and Safety Data. Online.
http://ntp-server.niehs.nih.gov/Main_Pages/Chem-HS.html.
Ono, A., K. Sekita, K. Ohno, et al. 1995. Reproductive and developmental toxicity studies of toluene: I.
Teratogenicity study of inhalation exposure in pregnant rats. J. Toxicol. Sci. 20(2): 109-34.
Ono, A., K. Sekita, Y. Ogawa, et al. 1996. Reproductive and developmental toxicity studies of toluene:
II. Effects of inhalation exposure on fertility in rats. J. Environ. Pathol. Toxicol. Oncol. 15(1): 9-20.
Orbaek, P. and G. Nise. 1989. Neurasthenic complaints and psychometric function of toluene-exposed
rotogravure printers. Am. J. Ind. Med. 16: 67-77.
OSHA (Occupational Safety and Health Administration). 1993. 29 CFR Part 1910. Air Contaminants;
Rule. Federal Register 58(124): 35338-35351.
Patel, R. and J. Benjamin. 1986. Renal disease associated with toluene inhalation. Clin. Toxicol. 24(3):
213-223.
Paterson, S.C. and R. Sarvesvaran. 1983. Plastic bag death: A toluene fatality. Med. Sci. Law. 23(1):
64-66.
Pelclova, D., P. Rossner and J. Pickova. 1990. Chromosome aberrations in rotogravure printing plant
workers. Mutat. Res. 245: 299-303.
Pellizzari, E.D., T.D. Hartwell, B.S.H. Haris, et al. 1982. Purgeable organic compounds in mother's
milk. Bull. Environ. Contam. Toxicol. 28: 322-328.
Pellizzari, E.D., L.A. Wallace and S.M. Gordon. 1992. Elimination kinetics of volatile organics in
humans using breath measurements. J. Expo. Anal. Environ. Epidemiol. 2(3): 341-55.
Pierce, C.H., R.L. Dills, M.S. Morgan, et al. 1996. Interindividual differences in 2H8-toluene
toxicokinetics assessed by semi-empirical physiologically based model. Toxicol. Appl. Pharmacol.
139(1): 49-61.
Pierce, C.H., T.A. Lewandowski, R.L. Dills, et al. 1999. A comparison of 1H8- and 2H8-toluene
toxicokinetics in men. Xenobiotica. 29(1): 93-108.
Pitarque, M., A. Vaglenov, M. Nosko, et al. 1999. Evaluation of DNA damage by the comet assay in
shoe workers exposed to toluene and other organic solvents. Mutat. Res. 441: 115-127.
Plenge-Bönig, A. and W. Karmaus. 1999. Exposure to toluene in the printing industry is associated with
subfecundity in women but not in men. Occup. Environ. Med. 56(7): 443-448.
Poon, R., I.H. Chu, S. Bjarnason, et al. 1994. Inhalation toxicity study of methanol, toluene, and
methanol/toluene mixtures in rats: effects of 28-day exposure. Toxicol. Ind. Health. 10(3): 231-245.
Pryor, G.T., C.S. Rebert, J. Dickinson and E.M. Feeney. 1984. Factors affecting toluene-induced
ototoxicity in rats. Neurobehav. Toxicol. Teratol. 6: 223-238.
Pyykko, K., H. Tahti and H. Vapaatalo. 1977. Toluene concentrations in various tissues of rats after
inhalation and oral administration. Arch. Toxicol. 38: 169-176.
Ramsey, J.C. and M.E. Andersen. 1984. A physiologically based description of the inhalation
pharmacokinetics of styrene in rats and humans. Toxicol. Appl. Pharmacol. 73: 159-175.
Rea, T.M., J.F. Nash, J.E., Zabik, G.S. Born and W.V. Kessler. 1984. Effects of toluene inhalation on
brain biogenic amines in the rat. Toxicology 31: 143-150.
Rebert, C.S., M.J. Matteucci and G.T. Pryor. 1989a. Acute electrophysiologic effects of inhaled toluene
on adult male Long-Evans rats. Pharmacol. Biochem. Behav. 33: 157-165.
Rebert, C.S., M.J. Matteucci and G.T. Pryor. 1989b. Multimodal effects of acute exposure to toluene
evidenced by sensory-evoked potentials from Fischer-344 rats. Pharmacol. Biochem. Behavior. 32: 757-
768.
Rees, D.C., R.W. Wood, J.P. McCormick, et al. 1985. Toxicokinetics of toluene in the rat. Scand. J.
Work. Environ. Health. 11: 301-306.
Rosenberg, N.L., B.K. Kleinschmidt-Demasters, K.A. Davis, et al. 1988a. Toluene abuse causes diffuse
central nervous system white matter changes. Ann. Neurol. 23(6): 611-614.
Rosenberg, N.L., M.C. Spitz, C.M. Filley, et al. 1988b. Central nervous system effects of chronic
toluene abuse - clinical, brainstem evoked response and magnetic resonance imaging studies.
Neurotoxicol. Teratol. 10: 489-495.
Ryu, Y.H., J.D. Lee, P.H. Yoon, et al. 1998. Cerebral perfusion impairment in a patient with toluene
abuse. J. Nucl. Med. 34(4): 632-633.
Sato, A. and T. Nakajima. 1978. Differences following skin or inhalation exposure in the absorption and
excretion kinetics of trichlorethylene and toluene. Br. J. Ind. Med. 35: 43-49.
Schmid, E., M. Bauchinger and R. Hauf. 1985. Chromosome changes with time in lymphocytes after
occupational exposure to toluene. Mutat. Res. 142: 37-39.
Snodgrass, W.R. 1992. Physiological and biochemical differences between children and adults as
determinants of toxic response to environmental pollutants. In: Guzelian, P.S., C.J. Henry, S.S. Olin, ed.
Similarities and differences between children and adults: Implications for risk assessment.
Washington,DC: International Life Sciences. pp. 35-42.
Snow, L., P. MacNair and B.C. Casto. 1981. Mutagenesis testing of toluene in Salmonella strains
TA100 and TA98. Report prepared for the U.S. EPA by Northrup Services, Inc., Research Triangle park,
NC.
Suzuki, T., S. Kashimura and K. Umetsu. 1983. Thinner abuse and aspermia. Med. Sci. Law. 23(3):
199-202.
Svensson, B.G., G. Nise, V. Englander, et al. 1990. Deaths and tumors among rotogravure printers
exposed to toluene. Br. J. Ind. Med. 47: 372-379.
Takeichi, S., T. Yamada and I. Shikata. 1986. Acute toluene poisoning during painting. Forensic Sci.
Int. 32: 109-115.
Tardif, R., G.L. Plaa and J. Brodeur. 1992. Influence of various mixtures of inhaled toluene and xylene
on the biological monitoring of exposure to these solvents in rats. Can. J. Physiol. Pharmacol. 70: 385-
395.
Tardif, R., S. Lapare, K. Krishnan, et al. 1993. Physiologically based modeling of the toxicokinetic
interaction between toluene and m-xylene in the rat. Toxicol. Appl. Pharmacol. 120: 266-273.
Tardif, R., G. Truchon and J. Brodeur. 1998. Comparison of hippuric acid and O-cresol in urin and
unchanged toluene in alveolar air for the biological monitoring of exposure to toluene in human
volunteers. Appl. Occup. Environ. Hyg. 13(2): 127-132.
Tassaneeyakul, W., D.J. Birkett, J.W. Edwards, et al. 1996. Human cytochrome P450 isolform
specificity in the regioselective metabolism of toluene and o-, m-, and p-xylene. J. Pharmacol. Exp.
Ther. 276(1): 101-108.
Taverner, D., D.J. Harrison and G.M. Bell. 1988. Acute renal failure due to interstitial nephritis induced
by 'glue sniffing' with subsequent recovery. Scott. Med. J. 33: 246-247.
Thiel, R. and I. Chahoud. 1997. Postnatal development and behaviour of Wistar rats after prenatal
toluene exposure. Arch. Toxicol. 71(4): 258-65.
Tilson, H.A. 1990. Animal neurobehavioral test battery in NTP assessment. In: Advances in
neurobehavioral toxicology: applications in environmental & occupational health. Barry L. Johnson, ED.
Lewis Publishers, Inc. pp. 403-418.
Turkall, R.M., G.A. Skowronski and M.S. Abdel-Rahman. 1991. Differences in kinetics of pure and
soil-adsorbed toluene in orally exposed male rats. Arch. Environ. Contam. Toxicol. 20: 155-160.
Ungvary, G. and E. Tatrai. 1985. On the embryotoxic effects of benzene and its alkyl derivatives in
mice, rats, and rabbits. Arch. Toxicol. Suppl. 8: 425-430.
U.S. EPA. 1984. Health Effects Assessment for Toluene. Prepared by the Office of Health and
Environmental Assessment, Environmental Criteria and Assessment Office, Cincinnati, OH, for the
Office of Emergency and Remedial Response, Washington, DC. EPA-600/X-84-188.
U.S. EPA. 1985. Drinking Water Criteria Document for Toluene. Prepared by the Office of Health and
Environmental Assessment, Environmental Criteria and Assessment Office, Cincinnati, OH, for the
Office of Drinking Water, Washington, DC. EPA/540/1-86-033.
U.S. EPA. 1986a. Guidelines for carcinogen risk assessment. Federal Register. 51(185):33992-34003.
U.S. EPA. 1986b. Guidelines for the health risk assessment of chemical mixtures. Federal Register.
51(185):34014-34025.
U.S. EPA. 1986c. Guidelines for mutagenicity risk assessment. Federal Register. 51(185): 34006-34012.
U.S. EPA. 1987. Drinking Water Criteria Document for Toluene. Prepared by the Office of Health and
Environmental Assessment, Environmental Criteria and Assessment Office, Cincinnati, OH for the
Office of Drinking Water, Washington, DC.
U.S. EPA. 1988. Recommendations for and documentation of biological values for use in risk
assessment. EPA 600/6-87/800, NTIS PB88-179874/AS.
U.S. EPA. 1991. Guidelines for developmental toxicity risk assessment. Federal Register 56:63798-
63826.
U.S. EPA. 1994a. Interim policy for particle size and limit concentration issues in inhalation toxicity:
notice of availability. Federal Register 59:53799.
U.S. EPA. 1994b. Methods for derivation of inhalation reference concentrations and application of
inhalation dosimetry. EPA/600/8-90/066F.
U.S. EPA. 1994c. Peer review and peer involvement at the U.S. Environmental Protection Agency.
Signed by the U.S. EPA Administrator, Carol A. Browner, June 7.
U.S. EPA. 1995. Use of the benchmark dose approach in health risk assessment. EPA/630/R-94/ 007.
U.S. EPA. 1996. Reproductive toxicity risk assessment guidelines. Federal Register 61(212):56274-
56322.
U.S. EPA. 1998b. Science policy council handbook: peer review. Prepared by the Office of Science
Policy, Office of Research and Development, Washington, DC. EPA/100/B-98/001.
U.S. EPA. 1999. Guidelines for carcinogen risk assessment. Review Draft, NCEA-F-0644, July 1999.
Risk Assessment Forum.
Vieira, I., M. Sonnier and T. Cresteil. 1996. Developmental expression of CYP2E1 in the human liver:
hypermethylation control of gene expression during the neonatal period. Eur. J. Biochem. 238: 476-483.
von Euler, G., S. -O. Ogren, X.M. Li, K. Fuxe and J. -A. Gustafsson. 1993. Persistent effects of
subchronic toluene exposure on spatial learning and memory, dopamine-mediated locomotor activity and
dopamine D2 agonist binding in the rat. Toxicology 77: 223-232.
von Euler, G., S. -O. Ogren, P. Eneroth, K. Fuxe and J. -A. Gustafsson. 1994. Persistent effects of 80
ppm toluene on dopamine-regulated locomotor activity and prolactin secretion in the male rat.
Neurotoxicol. 15: 621-624.
von Euler, G.: Toluene and dopaminergic transmission, in Isaacson, R.L., K.F. Jensen (eds): The
Vulnerable Brain and Environmental Risk, Toxins in Air and Water, Vol 3, New York: plenum Press,
1994, pp. 301-321.
von Euler, M., T.M. Pham, M. Hillefors, et al. 2000. Inhalation o flow concentrations of toluene induces
persistent effeccts on a learning retention task, beam-walk performance, and cerebrocortical size in the
rat. Experim. Neurol. 163: 1-8.
Vrca, A., D. Bozicevic, V. Karacic, et al. 1995. Visual evoked potentials in individuals exposed to
long-term low concentrations of toluene. Arch. Toxicol. 69(5): 337-40.
Vrca, A., D. Bozicevic, V. Bozikov, et al. 1997. Brain stem evoked potentials and visual evoked
potentials in relation to the length of occupational exposure to low levels of toluene. Acta Medica
Croatica. 51: 215-219.
Walker, J.T., T.F. Bloom, F.B. Stern, et al. 1993. Mortality of workers employed in shoe manufacturing.
Scand. J. Work Environ. Health. 19: 89-95.
Wiebelt, H. and N. Becker. 1999. Mortality in a cohort of toluene exposed employees (rotogravure
printing plant workers). J. Occup. Environ. Med. 41(12): 1134-1139.
Wolf, M.A., V.K. Rowe, D.D. McCollister, et al. 1956. Toxicological studies of certain alkylated
benzenes and benzene. Arch. Ind. Health. 14: 387-398.
Wood, R.W. and V.A. Colotla. 1990. Biphasic changes in mouse motor activity during exposure to
toluene. Fund. Appl. Toxicol. 14: 6-14.
Wood, R. and C. Cox. 1995. A repeated measures approach to the detection of the acute behavioral
effects of toluene at low concentrations. Regulatory Toxicol. Pharmacol. 33: 12-20.
Yin, S., G. Li, Y. Hu, et al. 1987. Symptoms and signs of workers exposed to benzene, toluene or the
combination. Ind. Health. 25(3): 113-130.
Zavalic, M., Z. Mandic, R. Turk, et al. 1998a. Quantitative assessment of color vision impairment in
workers exposed to toluene. Am. J. Ind. Med. 33(3): 297-304.
Zavalic, M., Z. Mandic, R. Turk, et al. 1998b. Assessment of colour vision impairment in male workers
exposed to toluene generally above occupational exposure limits. Occup. Med. 48(3): 175-180.
Benchmark dose (BMD) modeling was performed to identify the critical effect level for
derivation of the RfD for toluene. The modeling was conducted according to draft EPA
guidelines (U.S. EPA, 2000d) using Benchmark Dose Software Version 1.3 (BMDS), available
fromU.S. EPA (U.S. EPA, 2001). The BMD modeling results are summarized in Table B-1; the
output is attached. A brief discussion of the modeling results is presented below.
Since the endpoint is a continuous variable, the continuous models available with BMDS
(power, polynomial, and Hill models) were used. The hybrid model was not used, because the
hybrid model software in BMDS is still undergoing Beta-testing, and was not considered
sufficiently validated to use a BMDL from this model as the basis for the quantitative dose-
response assessment. (The hybrid modeling approach defines the benchmark response [BMR]
directly in terms of risk, as opposed to the standard approach, which defines the BMR in terms of
a change in the mean.) For all of the modeling conducted, the BMR was defined as a 1.0 SD
change in the mean, since this is the default measure recommended by the U.S. EPA (U.S. EPA,
2000d) in the absence of a clear biological rationale for selecting an alternative response level.
Table B-1: Benchmark modeling summary for relative thymus weight, Hsieh et al. (1989)
Thymus weight,
Concentration in drinking Exposure, in g/100g body weight
water (ppm) mg/kg-day Number examined (+ SE)
0 0 5 0.19 + 0.02
20 5 5 0.18 + 0.01
100 22 5 0.18 + 0.02
500 105 5 0.13 + 0.02
Goodness- Maximum
of-fit P2 residual ECSa LECSa
Continuous models p-value AIC near POD (mg/kg/d) (mg/kg/d)
* Linear model selected as best fitting model, with lowest AIC among those with adequate goodness-of-fit test results (p>0.10).
Linear model is given by:
rho is set to 0
Parameter Estimates
Dose N Obs Mean Obs Std Dev Est Mean Est Std Dev Chi^2 Res.
------ --- -------- ----------- -------- ----------- ----------
Likelihoods of Interest
Tests of Interest
to be a
dose levels.
homogeneous variance
chosen appears
BMD = 66.2745
BMDL = 40.5754