BUTADIENE
BUTADIENE
BUTADIENE
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 policy. It is being circulated for comment on its technical accuracy and policy
implications.
This document is an external 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.
PREFACE . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xiii
1. INTRODUCTION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1-1
1.1. BACKGROUND . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1-1
1.2. SUMMARY OF PAST CARCINOGEN ASSESSMENTS OF 1,3-BUTADIENE . . 1-1
1.2.1. Summary of EPA’s Carcinogen Assessment (U.S. EPA, 1985) . . . . . . . . . . . . 1-2
1.2.2. Summary of IARC’s Evaluation of 1,3-Butadiene (IARC, 1986, 1992) . . . . . 1-5
1.2.3. Summary of the National Institute for Occupational Safety and Health
(NIOSH) Evaluation of 1,3-Butadiene (NIOSH, 1991a) . . . . . . . . . . . . . . . . . 1-7
1.2.4. California Air Resources Board (CARB, 1991) . . . . . . . . . . . . . . . . . . . . . . . . 1-8
1.2.5. Summary of Findings by U.S. Occupational Safety and Health
Administration (OSHA) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1-10
1.3. DISCUSSION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1-11
4. MUTAGENICITY . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4-1
4.1. INTRODUCTION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4-1
4.2. GENE MUTATIONS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4-1
4.3. CYTOGENETIC EFFECTSCHUMAN . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4-4
4.4. CYTOGENETIC EFFECTSCRODENT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4-5
4.5. SUMMARY . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4-6
2-1A Summary of 1,3-butadiene ambient data from the EPA Aerometric Information
Retrieval System (AIRS) for 1988 to 1991 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2-10
2-1B Summary of 1,3-butadiene ambient data from the EPA Aerometric Information
Retrieval System (AIRS) for 1992 to 1994 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2-12
2-2 Summary of 1,3-butadiene ambient data from the Urban Air Toxics Monitoring
Program (UATMP) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2-16
2-3 Summary of outdoor urban data from the National Ambient Volatile Organic
Compounds (NAVOC) Database . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2-18
2-4 Summary of air monitoring program results for 1,3-butadiene . . . . . . . . . . . . . . . . . 2-19
2-5 Summary of 1,3-butadiene ambient data from the EPA Aerometric Information
Retrieval System (AIRS) based on sampling locations . . . . . . . . . . . . . . . . . . . . . . . 2-21
2-6 Summary of 1,3-butadiene data from Table 2-2 based on sampling locations . . . . . 2-22
2-7 Summary of 1,3-butadiene data from Table 2-3 based on sampling locations . . . . . 2-22
2-8 Summary of the relative contributions to ambient 1,3-butadiene emissions given
as percent of total mg/yr . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2-24
8-1 Parameter values used in the Hattis and Wasson (1987) PBPK model . . . . . . . . . . . . 8-3
8-2 Parameter values used in the Kohn and Melnick (1993) PBPK model . . . . . . . . . . . . 8-5
8-3 Parameter values used in the Johanson and Filser (1993) PBPK model . . . . . . . . . . 8-10
8-4 Parameter values used in the Evelo et al. (1993) PBPK model . . . . . . . . . . . . . . . . . 8-14
8-5 Parameter values used in the Medinsky et al. (1993) PBPK model . . . . . . . . . . . . . . 8-17
9-1 Excess risk and 95% upper confidence limit excess risk estimates based
on the multiplicative model reported by Delzell et al., 1995 . . . . . . . . . . . . . . . . . . . 9-6
9-2 Excess risk and 95% upper confidence limit excess risk estimates based
on the power model reported by Delzell et al., 1995 . . . . . . . . . . . . . . . . . . . . . . . . . 9-6
9-3 Excess risk and 95% upper confidence limit excess risk estimates based
on the linear excess relative rate model reported by Delzell et al., 1995 . . . . . . . . . . . 9-7
9-4 Excess risk and 95% upper confidence limit excess risk estimates based
on the final square root model reported by Delzell et al., 1995 . . . . . . . . . . . . . . . . . . 9-7
9-5 Excess risk and 95% upper confidence limit excess risk estimates based
on the square root model reported by Delzell et al., 1995 . . . . . . . . . . . . . . . . . . . . . . 9-8
9-6 Observed versus predicted dose (exposure) probability P(d) of fetal weight
reduction below the 10th percentile of controls using log-logistic model . . . . . . . . . 9-34
9-7 Observed versus predicted mean fetal weight per litter using continuous model . . . . 9-34
9-8 Observed versus predicted percent of mean fetal weights per litter less
than the 5th percentile of controls (P0 = 0.05) using hybrid model . . . . . . . . . . . . . . 9-35
9-9 Observed versus predicted mean number of implants (prenatal)
using log-linear model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9-37
9-10 Observed versus predicted proportion of early and late deaths
per implantation (prenatal) using log-linear model . . . . . . . . . . . . . . . . . . . . . . . . . . 9-37
9-11 Observed versus predicted proportion of live implants (prenatal)
using log-linear model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9-38
9-12 Observed versus predicted mean number of implants (postnatal)
using log-linear model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9-38
9-13 Observed versus predicted proportion of post-implantation losses (postnatal)
using log-linear model . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9-39
9-14 Observed versus predicted mean litter size at birth using log-linear model . . . . . . . . 9-39
9-15 Observed versus predicted mean litter size at weaning using log-linear model . . . . . 9-40
9-16 Ovarian atrophy (groups 1-5) using log-logistic model . . . . . . . . . . . . . . . . . . . . . . 9-42
9-17 Uterine atrophy (groups 1-6) using quantal Weibull model . . . . . . . . . . . . . . . . . . . 9-43
9-18 Testicular atrophy (groups 1-6) using quantal Weibull model . . . . . . . . . . . . . . . . . 9-43
This Health Risk Assessment of 1,3-Butadiene has been prepared to serve as a source
document for Agencywide use. This document was developed primarily for use by the U.S.
Environmental Protection Agency’s (EPA) Office of Mobile Sources (OMS) to support decision
making regarding the Air Toxic Rule’s Section 202L2 of the Clean Air Act Amendment. Since
OMS requested that this assessment focus on mutagenicity, carcinogenicity, and
reproductive/developmental effects, an evaluation of other health hazards has not been included.
This document, therefore, is not a comprehensive health assessment. The exposure information
included here is an overview of the ambient exposures and exposure to populations adjacent to
emission sources, without any actual exposure assessment as such.
In the development of this assessment, relevant scientific literature has been
incorporated from the period July 1, 1985, through January 31, 1997. Key studies have been
evaluated to qualitatively describe the mutagenicity, reproductive/developmental effects, and
carcinogenicity of 1,3-butadiene. The assessment also includes a summary, conclusions, and
risk characterization. Measures of dose-risk relationships relevant to ambient air exposures are
discussed so that the adverse health effects can be placed in perspective with possible exposure
levels.
This document was prepared by the National Center for Environmental Assessment-
Washington Office (NCEA-W) of EPA’s Office of Research and Development. Sections of this
report were prepared by Oak Ridge National Laboratory (ORNL) under Interagency Agreement
No. DW89937638-01-0. Aparna M. Koppikar1 served as the Project Manager, providing overall
direction and technical assistance.
AUTHORS
Chapter 1: Aparna M. Koppikar, Kowetha Davidson2
Chapter 2: Chieh Wu1
Chapter 3: Kim Hoang1, Carol Forsyth2, and Robert Young2
Chapter 4: Lawrence Valcovic1
Chapter 5: Kowetha Davidson
Chapter 6: Rosmarie Faust2
Chapter 7: Aparna M. Koppikar
Chapter 8: Jennifer Jinot1, Carol Kimmel1
Chapter 9: Jennifer Jinot
Chapter 10: Aparna M. Koppikar
Chapter 11: Jennifer Jinot and Aparna M. Koppikar
CONTRIBUTORS
Thomas M. Crisp1
Dharm Singh1
Steven Bayard (now at OSHA)
Milton Siegal1
John Schaum1
Leslie Stayner3
Stephen Gilbert3
Randall Smith3
1
National Center for Environmental Assessment-Washington Office.
2
Oak Ridge National Laboratory.
3
National Institute for Occupational Safety and Health.
REVIEWERS
David Bayliss (NCEA-W)
Robert Beliles (NCEA-W)
Pam Brodowicz (OMS)
Robert Bruce (NCEA-Cin)
James Cogliano (NCEA-W)
Richard Cook (OMS)
Michael Dellarco (NCEA-W)
Gary Kimmel (NCEA-W)
William Pepelko (NCEA-W)
The authors would like to acknowledge the contributions of several people who have
made this report possible:
Theresa Konoza of NCEA-W, who was responsible for coordinating and managing the
production effort.
The CDM Group, Inc., under the direction of Kay Marshall, who was responsible for
editing, word processing, and literature searches.
1.1. BACKGROUND
1,3-Butadiene (CH2=CH-CH=CH2, CAS No. 106-99-0) is a colorless gas produced by
three different processes: (1) oxidative dehydrogenation of n-butene (the Oxo-D or O-X-D
process), (2) catalytic dehydrogenation of n-butane and n-butene (the Houdry process), and (3)
recovery from the C4 coproduct (by-product) stream from the steam cracking process used to
manufacture ethylene (the ethylene coproduct process). This noncorrosive gas has a boiling point
of 4.4 C and a vapor pressure of 1,900 mm/Hg at 20 C (Kirshenbaum, 1978). 1,3-Butadiene is
highly volatile and has a low solubility in water; thus environmental release results primarily in
atmospheric contamination. Atmospheric destruction of 1,3-butadiene occurs primarily by
photoinitiated reactions. A significant amount of destruction also occurs by the gas phase
reaction with ozone and reaction with nitrate radicals at nighttime, particularly in urban areas
(U.S. DHHS, 1992). The major photooxidation products of 1,3-butadiene are acrolein and
formaldehyde (Maldotti et al., 1980).
Approximately 12 billion pounds of 1,3-butadiene are produced annually worldwide and 3
billion pounds in the United States (Morrow, 1990; USITC, 1990). It is used as an intermediate
in the production of polymers, elastomers, and other chemicals. The major uses of 1,3-butadiene
are in the manufacture of styrene-butadiene rubber (SBR) (synthetic rubber) and of thermoplastic
resins. Elastomers of butadiene are used in the manufacture of tires, footwear, sponges, hoses
and piping, luggage, packaging, and a variety of other molded products. In addition, 1,3-
butadiene is used as an intermediate to produce a variety of industrial chemicals, including the
fungicides captan and captfol. The primary way the 1,3-butadiene is released in the environment
is via emissions from gasoline- and diesel-powered vehicles and equipment. Minor releases occur
in production processes, tobacco smoke, gasoline vapors, and vapors from the burning of plastics
as well as rubber (Miller, 1978).
1.2.3. Summary of the National Institute for Occupational Safety and Health (NIOSH)
Evaluation of 1,3-Butadiene (NIOSH, 1991a)
NIOSH (1991a) conducted a qualitative and quantitative assessment of the carcinogenicity
of 1,3-butadiene. The evaluation of animal data focused on the studies that could be used for
quantitative assessment, namely the studies using Sprague-Dawley rats (Owen et al., 1987) and
or 1.6 × 10 2 (mg/kg/day) 1 for female rats. The estimates based on applied or metabolized doses
were much lower. The data from the second mouse study were analyzed extensively; CARB
concluded that the best human cancer potency estimates based on internal doses and estimated
using the multistage model (Global 86) were 0.37 (ppm) 1 or 3.4 (mg/kg/day) 1 derived for
alveolar/bronchiolar adenoma/carcinoma in female mice. The corresponding unit risk derived
from the second mouse study was 1.6 × 10 4 (µg/m3) 1 and the exposure for the risk at 10 6 was
6.0 × 10 3 µg/m3. From their cancer potency values, CARB estimated the lifetime extra risk
associated with exposure to 1 ppb 1,3-butadiene to range from 9.8 × 10 6 to 3.7 × 10 4, which
corresponds to 10 to 370 additional cases per 1 million individuals.
1.3. DISCUSSION
Six different carcinogenicity assessments of 1,3-butadiene, done by five different agencies
in different time periods, are summarized in this chapter. The major conclusions of these
evaluations are presented in Table 1-1.
Although no apparent agreement is evident from the table among the five agencies’
assessments, in fact they are very similar. Both EPA (1985) and IARC (1986) conclude that the
carcinogenicity evidence in humans is inadequate and in animals is sufficient. But due to different
classification systems, they get different alphabetical assignments, i.e., B2 and 2B, which
correspond to “probable” and “possible” descriptors, thus appearing to be in disagreement with
each other. NIOSH and OSHA both use the dichotomous descriptors with “potential
occupational carcinogen” being the highest ranking.
NIOSH (1991a) “Potential human Range of excess risk at 1 OSHA cancer policy
health hazard with ppm is MLE of 305/10,000 classification system
respect to based on female mouse lung used.
carcinogenicity.” neoplasms to MLE of Quantitative risk is
0.03/10,000 based on heart for occupationally
hemangiosarcomas in exposed populations.
females. Data from
Melnick et al. (1990a) used
for this quantitation.
The purpose of this chapter is to present an overview of how human exposure to 1,3-
butadiene occurs. The chapter summarizes physical/chemical properties, production/use,
sources/emissions, and ambient air data. Pathways of exposure are briefly described, but no
quantitative estimates of exposure levels and numbers of people exposed are presented.
2.3.1.3. Aircraft
Human exposure to aircraft emissions is considered to be limited to the emissions that
occur during aircraft landing and take-off (LTO). Airborne aircraft are assumed to fly at
sufficiently high altitudes that their emissions do not reach the surface. This assumption is likely
to be valid for butadiene because of its short atmospheric lifetime.
Butadiene has been reported in aircraft LTO emissions from military, commercial, and
general aviation. Based on the EPA SPECIATE database, the butadiene weight percents for
aircraft LTO hydrocarbon emissions range from 1.57% for general aviation (piston engines) to
1.89% from military aircraft (jet and piston engines). The 1994 EPA report estimates that 0.1%
of the national butadiene emissions is attributable to aircraft LTO (EPA, 1994a).
2.4.1. Air
In 1989, total emissions of 1,3-butadiene to the air in the United States were estimated at
approximately 2,512 tonnes from 158 locations; total land releases were estimated at 6.7 tonnes
(U.S. National Library of Medicine, 1991).
Average concentration (ppb) Sampling sites Land use of monitor location Number of samples
Average concentration (ppb) Sampling sites Land use of monitor location Number of samples
Table 2-1B. Summary of 1,3-butadiene ambient data from the EPA Aerometric Information Retrieval System (AIRS) for 1992 to 1994
Average concentration (ppb) Sampling sites Land use of monitor location Number of samples
1992 1993 1994 1992 1993 1994
0.12 0.33 Jefferson Co., AL Residential/Rural 83 79
0.19 Jefferson Co., AL Residential/Rural 9
4.32 0.78 Tarrant City, AL Residential/Suburban 82 81
0.91 Tarrant City, AL Residential/Suburban 10
0.07 0.15 Shelby Co., AL Agricultural/Rural 50 78
0.30 0.30 Fresno, CA Residential/Suburban 30 31
0.54 Clovis, CA Residential/Urban & Center City 111
0.28 0.27 Bakersfield, CA Residential/Urban & Center City 30 9
0.35 Bakersfield, CA Commercial/Urban & Center City 23
2-12
Table 2-1B. Summary of 1,3-butadiene ambient data from the EPA Aerometric Information Retrieval System (AIRS) for 1992 to 1994 (continued)
Average concentration (ppb) Sampling sites Land use of monitor location Number of samples
1992 1993 1994 1992 1993 1994
0.06 0.08 Lemont, IL Residential/Suburban 15 13
0.10 0.11 St. Clair Co., IL St. Louis Metro Area 10 12
0.17 0.26 Kansas City, KS Industrial/Urban & Center City 11 13
0.42 0.43 Baton Rouge, LA Commercial/Urban & Center City 14 8
0.15 0.22 Glen Burnie, MD Commercial/Suburban 60 54
0.25 0.26 Essex, MD Residential/Suburban 39 58
0.18 0.17 Baltimore, MD Residential/Suburban 56 59
0.09 Baltimore, MD Industrial/Suburban 21
0.10 0.12 Baltimore, MD Industrial/Urban & Center City 50 50
0.26 0.25 0.28 Baltimore, MD Residential/Urban & Center City 58 57 48
0.09 0.10 Baltimore, MD Industrial/Urban & Center City 39 48
2-13
Average concentration (ppb) Sampling sites Land use of monitor location Number of samples
1992 1993 1994 1992 1993 1994
0.26 0.49 Camden, NJ Residential/Suburban 14 13
0.63 0.73 Newark, NJ Industrial/Urban & Center City 8 9
0.72 0.79 Plainfield, NJ Residential/Suburban 8 8
0.70 Nassau Co., NY Commercial/Suburban 9
0.19 Philadelphia, PA Residential/Suburban 38
0.08 San Antonio, TX Residential/Suburban 21
0.24 0.37 Clute, TX Residential/Suburban 29 53
0.18 0.21 Brownsville, TX Commercial/Urban & Center City 7 15
0.29 0.37 Brownsville, TX Commercial/Urban & Center City 19 59
2-14
Average concentration (ppb) Sampling sites Land use of monitor location Number of samples
1992 1993 1994 1992 1993 1994
0.44 0.81 Houston, TX Industrial/Suburban 39 54
0.24 0.64 Houston, TX Industrial/Suburban 33 58
0.05 0.39 0.73 Houston, TX Industrial/Suburban 6 37 58
0.56 Beaumont, TX Residential/Suburban 78
0.05 0.25 0.37 Beaumont, TX Residential/Suburban 7 36 59
0.10 0.23 0.44 Port Arthur, TX Residential/Suburban 6 45 57
0.32 0.43 Port Neches, TX Industrial/Suburban 18 58
5.95 Port Neches, TX Residential/Urban & Center City 23
5.22 6.11 Port Neches, TX Residential/Suburban 11 14
0.18 0.37 Corpus Christi, TX Commercial/Suburban 39 52
0.18 West Orange, TX Residential/Suburban 27
0.33 Smith Co., TX Mobile/Rural 32
2-15
AIRS
1988 level
(ppb) 0.67(1.48) 0.46(1.02)b
# Obs. 18 12
# Sites 3 2
1989
(ppb) 0.23(0.57) 0.25(0.55)b
# Obs. 399 369
# Sites 30 29
1990
1991
(ppb) 0.10(0.22) ----c
# Obs. 117
# Sites 6
1992
1993
(ppb) 0.40(0.88) 0.32(0.71)d
1994
UATMP
1989
(ppb) 0.21 (0.46) ----e
# Obs. 390
# Sites 13
1990
(ppb) 1.02(2.25) 0.12(0.27)b
# Obs. 349 293
# Sites 12 10
NAVOC
1987
(ppb) 0.34(0.75)f no data
# Obs. 9
# Sites 6
a
Alternate averages do not include data from Houston and Port Neches, TX, due to impacts from
strong point sources.
b
Average ppb from all 4-quarter data sites, excluding Houston, TX.
c
Houston, TX, was not monitored during this 4-quarter period.
d
Average ppb from all sites, excluding Houston and Port Neches, TX.
e
Port Neches, Texas, was not monitored during this 4-quarter period.
f
All urban California sites.
1992 0.07-0.45 0.13 271/6 0.05 - 0.19 0.10 154/7 0.09 - 0.29 176/5
DRAFT--DO NOT CITE OR QUOTE
0.72
1993 0.05-0.20 0.10 494/10 0.06 - 4.32 0.41 864/31 0.08 - 0.31 580/21
0.98
1994 0.07-0.37 0.18 522/11 0.06 - 1.68 0.45 1135/32 0.05 - 0.62 780/24
1.54
a
1,3-Butadiene average concentration in ppb.
Table 2-6. Summary of 1,3-butadiene data from Table 2-2 based on sampling locations
1/28/98
Suburban Urban
Table 2-7. Summary of 1,3-butadiene data from Table 2-3 based on sampling locations
2-23
Urban
Total samples/number
Year Range Averagea of locations
DRAFT--DO NOT CITE OR QUOTE
2.4.3. Water
Although 1,3-butadiene has been detected in drinking water in the United States (U.S.
EPA, 1978; Kraybill, 1980), it is not clear what happens to the chemical in the body (U.S.
DHHS, 1992). Total releases to ambient water in 1989 were estimated to be 65 tonnes (U.S.
National Library of Medicine, 1991).
2.4.4. Food
Certain cooking oils release butadiene on heating. For example, 1,3-butadiene emissions
are approximately 22-fold higher from unrefined Chinese rapeseed oil than from heated peanut
oil. Of three fatty acids tested, heated linolenic acid produced the greatest amount of 1,3-
butadiene. Although cooking oils in the U.S. are refined for purity, U.S. rapeseed oil (canola)
also emitted 1,3-butadiene (Shields et al., 1995). Also, levels of <0.2 µg/kg 1,3-butadiene were
found in retail soft margarine; the plastic tubs containing the margarine contained < 5-310 µg/kg
(Startin and Gilbert, 1984).
Figure 3-1. Some pathways in the metabolism of butadiene. Numbers in parentheses represent specific metabolic reactions for which literature
references are given in Table 3-1. Reactions (1), (2), (6), and (11) are mediated by cytochrome P-450-dependent monooxygenases. Glutathione (GSH) is a
substrate in reactions (3), (7), (9), (13), and (15), which are mediated by glutathione S-transferase or occur spontaneously. A GSH conjugate of
reaction(s) is excreted in the urine as N-acetyl-S-(1-hydroxy-3-butenyl)-L-cysteine. Enzyme-mediated GSH conjugates from reaction (7) include S-(2-
hydroxy-3-buten-1-yl)glutathione and S-(1-hydroxy-3-buten-2-yl)glutathione, which are subsequently excreted in the urine as 1-hydroxy-2-(N-
acetylcysteinyl)-3-butene and 2-hydroxy-1-(N-acetylcysteinyl)-3-butene. The GSH conjugate of reaction (9) is excreted in the urine as 1,2-dihydroxy-4-
(N-acetylcysteinyl)-butane. A=S-(2-hydroxy-3,4-epoxybut-1-yl)glutathione; B=S-(1-hydroxy-3,4-epoxybut-2-yl)glutathione; C=S-(1,2,3-trihydroxybut-4-
yl)glutathione; D=S-(1,3,4-trihydroxybut-2-yl)glutathione. The enzyme-mediated or spontaneous formation of GSH conjugates for reaction 13 form A
and B, which are excreted in the urine as C and D, respectively. Reactions (10) and (14) are mediated through the pentose phosphate pathway. Reactions
(8), (12), and (16) are mediated by epoxide hydrolase or occur spontaneously.
Sources: Dahl et al., 1990; Laib et al., 1990; Elfarra et al., 1991; Himmelstein et al., 1997.
3-3
DRAFT--DO NOT CITE OR QUOTE
1/28/98 DRAFT--DO
3-4 NOT CITE OR QUOTE
1/28/98
In vitro experiments
Wistar rat liver microsomes. (1a) Malvoisin et al. (1979a)
B6C3F1 and NMRI mouse; SD and Wistar rat, rhesus (1c) Schmidt and Loeser (1985)
monkey, and human (n = 1) postmitochondrial lung and
liver fractions.
Liver microsomes of rats (strain not stated), mice (1d) Wistuba et al. (1989)
(strain not stated), and humans ( n = 4).
3-4
microsomes.
Purified human myeloperoxidase from human (1h) Duescher and Elfarra (1992)
polymorphonuclear leukocytes and B6C3F 1 mouse liver
microsomes.
Rat (strain not stated) liver microsomes. (1i) Cheng and Ruth (1993)
SD rat, B6C3F 1 mouse, and human (n = 6) liver (1j) Duescher and Elfarra (1994)
microsomes.
B6C3F1 mouse bone marrow cells and cell lysates, (1k) Maniglier-Poulet et al. (1995)
human bone marrow cells, and purified human
myeloperoxidase.
1/28/98
SD rats. Metabolic uptake rate from untreated rats (1m) Bolt et al. (1984)
compared with that of rats pretreated with Aroclor 1254
and P-450 inhibitor.
B6C3F1 mice. Metabolic uptake rate from untreated (1n) Kreiling et al. (1986a)
rats compared with that of rats pretreated with P-450
inhibitor.
SD rats and B6C3F 1 mice. EB in exhaled breath. (1o) Kreiling et al. (1987)
SD rats and B6C3F 1 mice, pretreated with (1p) Medinsky et al. (1994)
4-methylpyrazole (P-450 inhibitor). Metabolic uptake
rate from untreated rats compared with that of rats
pretreated with P-450 inhibitor.
3-6
Nose-only exposure system SD rats and B6C3F 1 mice. EB in blood. (1q) Bond et al. (1986)
Cynomolgus monkeys. EB in blood. (1r) Dahl et al. (1990)
SD rats and B6C3F 1 mice. 1,3-butadiene and EB in (1s) Himmelstein et al. (1994)
blood.
DRAFT--DO NOT CITE OR QUOTE
SD rats and B6C3F 1 mice. 1,3-butadiene and EB in (1t) Bechtold et al. (1995)
blood.
SD rats and B6C3F 1 mice. EB in liver and lung tissues. (1u) Himmelstein et al. (1995)
SD rats and B6C3F 1 mice. EB in blood, fat, heart, (1v) Thornton-Manning et al.
liver, lung, spleen, and thymus. (1995a)
Female and male SD rats. EB in blood, femur, fat, (1w) Thornton-Manning et al.
lung, and mammary tissue. (1995b)
1/28/98
SD rat, B6C3F 1 mouse, and human liver (n = 12) and (6c) Csanády et al. (1992)
lung (n = 5) microsomes. Identified DEB only in mouse
liver microsomes.
DRAFT--DO NOT CITE OR QUOTE
(hydroxymethyl)-2-propenyl)-L-cysteine and
N-acetyl-S-(2-hydroxy-3-butenyl)-L-cysteine
in both mouse and rats.
(8) EB BD
DRAFT--DO NOT CITE OR QUOTE
In vitro experiments
Wistar rat liver microsomes. (8a) Malvoisin and Roberfroid
(1982)
SD rat liver microsomes treated with inhibitor of (8b) Bolt et al. (1983)
epoxide hydrolase. Same experiment as (1b) and (7a)
above.
SD rat, NMRI mouse, and human ( n = 1) liver (8c) Kreuzer et al. (1991)
microsomes.
1/28/98
1,2-dihydroxy-4-(N-acetylcysteinyl)-butane. Same experiments as (7f) and (8f) above. Urinary (9a) Sabourin et al. (1992)
metabolites in monkey, rat, hamster, and mouse.
1,2-dihydroxy-4-(N-acetylcysteinyl)-butane. Same experiments as (8g) above. Urinary metabolites (9b) Bechtold et al. (1994)
in humans.
N-acetyl-S-(3,4-dihydroxybutyl)-L-cysteine Same study as (3a), (7i), and (8h) above. Urinary (9c) Nauhaus et al. (1996)
in mouse and rat, N-acetyl-S-(1- metabolites.
(hydroxymethyl)-3,4-dihydroxypropyl)-L-
cysteine in mouse but not in rat.
1/28/98
In vitro experiments
Human (n = 6), SD rat, and B6C3F 1 mouse liver and (12a) Boogaard and Bond (1996)
lung microsomes.
DRAFT--DO NOT CITE OR QUOTE
(1) 1,3-Butadiene EB
Table 3-2. In vitro rates and rate constants for epoxidation, GSH conjugation, and hydrolysis reactions
involved in the metabolism of 1,3-butadiene (continued)
(6) EB DEB
Liver microsomes Mouse B6C3F1 1.4 141 (6d) Seaton et al. (1995)
Rat SD 0.41 145 (5-1,000 µM EB)
Human 0.38-1.2 304-880
1/28/98
Table 3-2. In vitro rates and rate constants for epoxidation, GSH conjugation, and hydrolysis reactions
involved in the metabolism of 1,3-butadiene (continued)
Liver cytosol Mouse B6C3F1 107 3,100 (7d) Sharer et al. (1992)
Rat SD 71 3,100 (2-10 mM EB)
Human
Purified placental GSH Human 500 10,000 (7d) Sharer et al. (1992)
S-transferase
Spontaneousg 2.01 · 10-4
(8) EB BD
Lung microsomes h Mouse B6C3F1 1.86 103 (8d) Csanády et al. (1992)
Rat SD 1.32 103 (20-200 ppm EB)
Human 3.19-7.55 103
1.65 · 10-3
3-18
a
Reaction rates are either from the reported reaction rates (units of nmol min-1 mg microsomal or cytosolic protein-1unless otherwise noted) or from maximum
reaction rate (Vmax in units of nmol min-1 mg microsomal or cytosolic protein-1 unless otherwise noted), when corresponding KMis given.
b
DRAFT--DO NOT CITE OR QUOTE
Table 3-3. Rate constants for in vivo hepatic clearance of 1,3-butadiene and EBa
(extrapolated from in vitro)
Cytosolic Microsomal
Cytochrome P-450 Epoxide glutathione conjugation of First-order
monooxygenaseb hydrolaseb,c S-transferaseb EB with GSHd hydrolysis
liver, respectively. Mouse, rat, and human liver cytosolic concentrations were 82.8, 108, and 58 mg/g liver, respectively. Liver organ
volumes for mice, rats, and humans were 6.2, 5.0, and 3.1% of body weight, respectively. In vivo hepatic clearance values (V max/KM
expressed in L/h/kg) were estimated by dividing the in vivo V max values by the apparent in vitro KM’s for the reaction.
c
Modified according to Kohn and Melnick, 1993.
d
For nonenzymic hydrolysis and reaction with glutathione, in vivo clearance was calculated using the organ fractions in footnote b.
DRAFT--DO NOT CITE OR QUOTE
To estimate the in vivo clearance for reaction with glutathione, a concentration of 10 mM GSH was used.
e
Rate constant for metabolism of EB to DEB.
(1n) Kreiling et al. (1986a) Exposure of B6C3F 1 mice to butadiene at initial Maximal metabolic uptake rate = 400
concentrations ranged from 10 to 5,000 ppm. µmol h 1 kg 1. Pretreatment of mice
with P-450 inhibitor
diethyldithiocarbamate completely
DRAFT--DO NOT CITE OR QUOTE
Kreiling et al. (1987) Exposure of B6C3F 1 mice to epoxybutene at initial Saturated metabolic uptake occurred
concentration ranging from 10 to 5,000 ppm between 100 and 500 ppm. V max = 350
(estimated from Figure 1 of Kreiling et al., 1986b). umol · h-1 · kg-1.
Abbreviations: ppm = parts per million; SD = Sprague-Dawley rat; Vmax = Michaelis-Menten enzyme kinetic constant expressing maximum metabolic rate.
a
Numbers and letters preceding reference refer to the appropriate metabolic pathway shown in Figure 3-1 and summarized in Table 3-1.
b
Reaction rates measured as uptake of epoxybutene, which could involve several reactions shown in Figure 3-1 and summarized in Table 3-1.
a
Calculated for V1 .
b
Valid for linear range of metabolism (up to 1,000 ppm for both species).
NA = not applicable.
Source: Filser and Bolt, 1981; Kreiling et al., 1990.
a
Calculated for V1 .
b
Valid for linear range of metabolism (up to 1,000 ppm for both species).
NA = not applicable.
Source: Filser and Bolt, 1981; Kreiling et al., 1987; Laib et al., 1990.
(1s) Himmelstein et al. (1994) Exposure of SD rats and B6C3F 1 mice to 1,3-Butadiene and EB pharmacokinetics characterized in
1,3-butadiene (62.5, 625, or 1,250 ppm blood by GC (butadiene) and GC-MS (EB). Butadiene
up to 6 h; blood collected at 2 to 6 h of steady-state concentrations (µM) ranged from 2.4 to 58
exposure and up to 30 min (mice) and 1.3 to 37 (rats). EB steady-state concentrations
postexposure). (µM) ranged from 0.56 to 8.6 (mice) and 0.07 to 1.3 (rats);
EB blood concentration in mice was 4 to 8 times > rats.
DRAFT--DO NOT CITE OR QUOTE
(1t) Bechtold et al. (1995) Exposure of SD rats and B6C3F 1 mice to Quantitated butadiene and EB in blood by GC-GC-MS.
1,3-butadiene (100 ppm for 4 h; blood Blood levels (µM) of butadiene were 4.1 (rat) and 2.9
collected at end of exposure). (mouse). Blood levels of EB (µM) were 0.10 (rat) and 0.38
(mouse).
(1u) Himmelstein et al. (1995) Exposure of SD rats and B6C3F 1 mice to Quantitated EB concentration by GC-MS in mouse lung 14
1,3-butadiene (625, 1,250, or 8,000 [rats times > rat lung, mouse liver 5 to 8 times > rat liver. Peak
only] ppm for 6 h; tissue samples concentrations of EB (nmol g tissue 1) during exposures
collected at 3 and 6 h of exposure and 6 were 2.6 to 3.7 (mouse lung), 0.16 to 1.3 (rat lung), 0.58 to
and 12 min postexposure). 0.93 (mouse liver), and 0.06 to 1.2 (rat liver).
1/28/98
62.5 ppm 1,3-butadiene). exposures. DEB levels were 7.7 ± 2.2 and 12.5 ± 0.8 pmol/g
in fat of rats and 265 ± 19 and 191 ± 29 pmol/g in mammary
tissue of mice after single and repeated inhalation exposures,
respectively.
(3) 3-Butenal GSH conjugates
DRAFT--DO NOT CITE OR QUOTE
(3a) Nauhaus et al. (1996) Exposure of SD rats and B6C3F 1 mice to Quantitated the urinary metabolite N-acetyl-S-(1-hydroxy-3-
1,3-[13C]-butadiene (800 ppm up to 5 h; butenyl)-L-cysteine in mouse urine using 13C-NMR. This
urine collected during exposure and for metabolite represented 3.7% of total urinary metabolites
up to 20 h postexposure). excreted by mice but was not detected in rat urine.
(6) EB DEB
(6e) Himmelstein et al. (1994) Exposure of rats and mice to 1,3- Quantitated time course of DEB in blood of mice by GC-
butadiene. Same conditions as (1s) MS. Peak concentrations of DEB in the blood of mice were
above. 0.65, 1.9, and 2.5 µM after 6 h of exposure to 62.5, 625, or
1,250 ppm 1,3-butadiene; DEB not quantitated in rats.
Detection limit = 0.13 µM.
1/28/98
(6i) Thornton-Manning et al. (1995b) Exposure of female and male SD rats to Quantitated DEB by GC-GC-MS. DEB concentrations were
1,3-butadiene. Same conditions as (1w) 3.6- to 7.1-fold greater in tissues of female rats compared
above. with tissues of male rats. Results summarized in Table 3-9.
(7) EB GSH conjugates
(7f) Sabourin et al. (1992) Exposure of SD and F344/N rats, Products identified in urine included 1-hydroxy-2-( N-
DRAFT--DO NOT CITE OR QUOTE
(9b) Bechtold et al. (1994) Same experiment as (8g) above. Quantitated 1,2-dihydroxy-4-( N-acetylcysteinyl)-butane in
human urine using isotope-dilution GC-MS. Metabolite
represents 97% of GSH conjugates derived directly from EB
(see (7g) above) or indirectly by epoxide hydrolase-mediated
hydrolysis and GSH conjugation of BD (see (8e) above).
DRAFT--DO NOT CITE OR QUOTE
Quantitated N-acetyl-S-(3,4-dihydroxybutyl)-L-cysteine in
(9c) Nauhaus et al. (1996) Exposure of rats and mice to 1,3- mouse (7.1%) and rat (26.4%) urine using 13C-NMR. N-
butadiene. Same study as (3a), (7i), and acetyl-S-(1-(hydroxymethyl)-3,4-dihydroxypropyl)-L-
(8h) above. cysteine occurred in mouse (7.1%) urine but was not
detected in rat urine. This latter metabolite was also
surmised to be a product of DEB as described in reaction 13
below.
1/28/98
Table 3-8. Comparison of 1,3-butadiene, epoxybutene, and diepoxybutane blood concentration data from
different species of laboratory animals exposed to 1,3-butadiene by inhalation
Butadiene exposure Concentration of analyte in blood
concentration (ppm) (nM analyte · ppm-1)
Mean (SE)
Mouse 7.83 (0.02) 72 (20) 68 (9) 8.5 (0.8) Bond et al. (1986)b
1,031 (13) 5.8 (0.6) 12.9 (0.5) 2.3 (0.3) Bond et al. (1986)
603 (44) 61 (7) 6.1 (0.8) 3.2 (0.4) Himmelstein et al. (1994)
3-31
1,282 (33) 45 (3) 6.7 (0.5) 2.0 (0.4) Himmelstein et al. (1994)
Rat 73.9 (0.4) 1.8 (0.2) 5.4 (0.4) 1.1 (0.2) Bond et al. (1986)b
949 (12) 3.2 (0.2) 3.5 (0.4) 0.8 (0.1) Bond et al. (1986)
DRAFT--DO NOT CITE OR QUOTE
Table 3-8. Comparison of 1,3-butadiene, epoxybutene, and diepoxybutane blood concentration data from
different species of laboratory animals exposed to 1,3-butadiene by inhalation
Butadiene exposure Concentration of analyte in blood
concentration (ppm) (nM analyte · ppm-1)
Mean (SE)a
Monkey 10.1 (0.1) 0.8 (0.4) 0.16 (0.05) 0.19 (0.06) Dahl et al. (1990)f for EB
310 (10) 1.8 (1.3) 1.6 (0.9) 0.9 (0.5) Dahl et al. (1991)f,j for DEB
a
Standard error (SE) combines variation of butadiene exposure concentration and blood concentration data.
b
Pooled mean ± SE of samples (n = 9) collected after 2, 4, and 6 h of exposure; animals removed from 1,3-butadiene exposure and exhaled 1,3-butadiene before blood
was collected; 14C-labeled analytes were recovered by vacuum-line cryogenic distillation and quantitated by liquid scintillation counting.
c
Values are means ± SE (n = 6-33 samples) for blood collected between 2 and 6 h of exposure; animals continued to inhale 1,3-butadiene as blood was collected;
1,3-butadiene was quantitated by a vial headspace equilibrium technique using GC-flame ionization detection; detection limit = 0.3 µM.
d
Values are means ± SE (n = 6) for samples collected at 4 h of exposure; animals continued to inhale 1,3-butadiene as blood was collected; analytes were recovered
by vacuum-line cryogenic distillation and analyzed by GC-GC-MS; detection limit = 0.1 µM.
DRAFT--DO NOT CITE OR QUOTE
e
One exposure was conducted at this 1,3- butadiene concentration, therefore no SE reported.
f
Blood (n = 3) collected immediately following 2 h exposure using indwelling catheter; 1,3-butadiene recovered by vacuum-line cryogenic distillation and quantitated
as 14C-labeled equivalent using liquid scintillation counting.
g
EB and DEB were recovered by extraction into methylene chloride and quantitated by GC-MS; detection limits = 0.03 µM for EB and 0.13 µM for DEB.
h
Detection limits for EB and DEB = 0.02 µM and 0.01 µM, respectively.
i
The authors exposed the monkeys to 1,3-[14C]-butadiene and vacuum-line cryogenic distillation for this compound includes14C-labeled DEB, BD, and potentially other unidentified14C-
metabolites. Since this study looked at products resulting from several reactions, it was not included in Table 3-7 (which described single reactions in the pathways).
Tissuea
Rats Mice Rats Mice
a
Mean ± SE; n = 3 or 4.
b
ND = not detected; indicates that analyte was not detected or was not above control level.
c
Includes at least one ND value.
d
As mean pmol/mg protein ± SE.
Tissuea
Males Females Males Females
a
n = 3, except for male femur, where n = 2.
b
One value was not detectable; instrument detection limit/2 was substituted to calculate the mean.
c
Statistically greater than male tissue value, p 0.05.
ND = not determined.
Source: Modified from Thornton-Manning et al., 1995b.
Uptake
Exposure
Exhalants Total metabolites
concentration
(ppm) CO2 Otherb Urine Feces recoveredc
10.1 1.5 ± 0.2 0.45 ± 0.9 ± 0.1 0.021 ± 0.005 2.88 ± 0.22
0.33
310 0.21 ± 0.04d 0.40 ± 0.8 ± 0.2 0.011 ± 0.003d 1.40 ± 0.42d
0.21
7760 0.08 ± 0.02d,e 1.00 ± 0.58 ± 0.002 ± 0.001d,e 1.65 ± 0.29d
0.35 0.06d
a
Values are mean percentage of total inhaled ± SE measured for 96 h after 2-h exposure.
b
Includes all material (except CO2) exhaled during the 2-h exposure and 96-h postexposure.
c
Mean ± SE of the sums of CO2, other, urine, and feces values for individual monkeys; does not include
residues, if any, in monkeys' bodies.
d
Significantly different from low-level exposure (p<0.05).
e
Significantly different from mid-level exposure (p<0.05).
Source: Dahl et al., 1991.
(limits of sensitivity for this assay, 100 ng/mL). The average values of M-I for exposed,
intermediately exposed, nonexposed, and outside control employees were 3,200 ± 1,600, 1,390±
550, 630 ± 190, and 320 ± 70 ng/mL, respectively. Although the levels of exposure for each
individual were not known, the urinary levels of M-I for the exposed groups were significantly
higher (p<0.05) compared with the outside control group. The implications of M-I in the urine
from individuals with no known exposure to 1,3-butadiene are not known.
In the second study that provided human data, Ward et al. (1996a) reported increased levels of
the urinary metabolite 1,2-dihydroxy-4-(N-acetyl-cysteinyl)-butane (a human urinary metabolite
also idenfified by Bechtold et al., 1994) and somatic mutations in workers at a styrene-butadiene
rubber plant. Exposure was assessed in workers from areas of higher exposures (reactor,
recovery, tank farm, laboratory) and lower exposure (blend,coagulation, bailers, shipping,
utilities, shops) using badge dosimeters; the concentration of the metabolite was measured in
urine; and the frequency of hprt mutant lymphocytes was determined
by autoradiography. The detection limit (0.25 ppm) was exceeded in 20/40 dosimeter readings
in the high-exposure group and in 0/20 readings in the low-exposure group. Sixteen high- and
nine low-exposure urine and blood samples were analyzed. Expressed as ng/mg creatinine,
metabolite concentrations were 2,363 ± 1,880 and 937 ± 583 (p<0.05), respectively, for the
4.1. INTRODUCTION
The mutagenic effects of 1,3-butadiene have been reviewed extensively (Rosenthal,
1985; de Meester, 1988; Arce et al., 1990; Norppa and Sorsa, 1993; Jacobson-Kram and
Rosenthal, 1995). The last of these reviewed publications through 1994 are on the genetic
effects associated with butadiene (and metabolites). There is extensive evidence that butadiene
and the two primary epoxide metabolites (epoxybutene and diepoxybutane) induce genotoxic
effects in a variety of in vitro and in vivo test systems. Most of the in vivo studies discussed in
the cited reviews were assays in mice and rats using cytogenetic endpoints, and the results
generally support the dichotomy in carcinogenic response where mice are more responsive than
rats. This review will focus on recently published studies performed in vivo (both somatic and
germ cell effects) with an emphasis on those studies providing information relative to the mode
of action of butadiene metabolites.
4.5. SUMMARY
The studies cited here along with the many earlier genotoxicity studies discussed in the
cited reviews provide clear evidence that 1,3-butadiene is both mutagenic and clastogenic
through its metabolism, primarily due to the mono- and diepoxide. While the difunctional DEB
is clearly more effective than the monofunctional EB for most endpoints, it is not possible to
ascribe the effects observed to one or the other when animals are exposed to butadiene. Where
both have been studied, mice are more responsive than rats, except for the recent germ cell
studies. Whether this exception is strain specific (among or between species) can only be
answered with future work.
The role of GST is also clearly established for the genotoxic effects of butadiene in
human lymphocytes. That the two glutathione S-transferases (GSTM1 and GSTT1) react
differently with the three epoxide metabolites suggests that the relative concentrations of these
metabolites will vary depending on the individual’s genotype.
2-Year studye
Ovarya 49 49 48 50 50 79
Atrophy 4 (8%) 19 (39%) 32 (67%) 42 (84%) 43 (86%) 69 (87%)
p<0.001 p<0.001 p<0.001 p<0.001 p<0.001 p<0.001
Germinal 2 (4%) 3 (6%) 8 (17%) 15 (30%) 15 (30%) 18 (23%)
epithelial p<0.001 p=0.460 p=0.017 p<0.001 p=0.010 p<0.001
hyperplasia
Angiectasis 4 (8%) 6 (12%) 3 (6%) 13 (26%) 14 (28%) 17 (22%)
p=0.259 p=0.366 p=0.606 p=0.017 p=0.021 p=0.425
Uterusa 50 49 50 49 50 78
c
Atrophy 1 (2%) 0 (0%) 1 (2%) 1 (2%) 8 (16%) 41 (53%)
a
Number of animals for which this site was examined microscopically.
b
p<0.01, pairwise comparison with controls by Fisher’s exact test.
c
Statistical tests were not conducted for these lesions.
d
p<0.05, pairwise comparison with controls by Fisher’s exact test.
e
p values for the statistical analysis (logistic regression test) for the 2-year study are presented; the value for the trend test is in the column for the control group, and the value for
pairwise comparisons of individual exposed group with the corresponding control group is in the column for the exposed groups.
Concentration (ppm)
Testesa 10 10 10 10 10 10
Absolute weight (g) 0.117 ± 0.002 0.117 ± 0.003 0.114 ± 0.003 0.103 ± 0.004b 0.102 ± 0.002b 0.059 ± 0.003b
Relative weight (mg/g 2.89 ± 0.06 2.92 ± 0.09 2.76 ± 0.09 2.87 ± 0.12 2.54 ± 0.05b 1.57 ± 0.03b
c
BW)
Testesa 10 10 10 10 10 7
Absolute weight (g) 0.116 ± 0.003 0.113 ± 0.003 0.104 ± 0.004 0.112 ± 0.003 0.100 ± 0.003b 0.071 ± 0.004b
5-6
Relative weight (mg/g 2.62 ± 0.07 2.79 ± 0.08 2.48 ± 0.04 2.66 ± 0.07 2.39 ± 0.05f 1.80 ± 0.05b
BW)
2-Year study
Testesa 50 50 50 48 49 72
a
Number of animals for which this site was examined.
b
p 0.01, pairwise comparison with controls by Williams’ or Dunnett’s test.
c
BW = body weight.
d
Statistical tests were not conducted for these lesions.
e
Testes were not examined microscopically at this concentration.
f
p<0.05, pairwise comparison with controls by Williams’ or Dunnett’s test.
Concentration (ppm)
No. of deaths 0 0 1a 0
Days 0-6 24 24 23 23
Days 6-9 13 9 1c 1c
Days 12-15 15 15 16 15
Days 15-20 54 58 61 60
a
This animal was killed in moribund state on day 19; morbidity was not related to exposure to butadiene.
b
Determined from differences in group mean body weights reported for specific days of gestation.
c
p<0.01, compared with corresponding control; analysis of variance andt test.
d
Body weight on gd 20 minus gravid uterine weight.
e
Extragestational weight minus body weight on gd 0.
f
p<0.05, compared with corresponding control; analysis of variance andt test.
Concentration (ppm)
Early resorptions 16 13 16 20
Dead fetuses/litter 0 0 0 0
a
Mean values.
b
p<0.05, Wilcoxon test.
c
p<0.01, Wilcoxon test.
Concentration (ppm)
External/visceral defects
Minor defects 76 (16.9)a 63 (23.8) 75 (24.4)b 75 (23.3)
Major defects 0 0 0 2 (0.7)
Skeletal defects
Minor defects 72 (23.2) 49 (26.9)c 45 (20.9) 43 (21.1)
Major defects 2 (0.6) 4 (2.2) 6 (2.8)b 12 (5.9)d
Variations (abnormal ossification) 267 (85.9) 164 (90.1) 185 (84.0) 199 (97.5)c
Sternebrae no. 6 152 (48.9) 107 (58.8) 126 (58.6) 147 (72.1)
a
Numbers of fetuses affected; numbers in parentheses denote the percentage of affected fetuses/fetuses examined.
b
p<0.05, Fisher’s randomization test based on frequencies of affected litters.
c
p<0.05, Wilcoxon’s test.
d
p<0.01, Fisher’s randomization test based on frequencies of affected litters.
Rat/ 0 30 6-15 20
Sprague-Dawley CD/
Inhalation 40 30 6-15 20
200 30 6-15 20
1,000 30 6-15 20
Mouse/ 0 32 6-15 18
CD-1/Inhalation
40 33 6-15 18
200 31 6-15 18
1,000 33 6-15 18
Concentration (ppm)
No. of deaths 0 0 0 0
Days 0-6 21.4 ± 1.6 21.1 ± 1.6 22.9 ± 1.3 20.1 ± 1.5
Days 6-11 25.5 ± 1.3 23.6 ± 1.3 26.6 ± 1.5 17.5 ± 1.9b
Days 11-16 29.2 ± 1.4 30.9 ± 1.7 31.7 ± 1.9 31.2 ± 2.1
Days 16-20 44.5 ± 1.8 36.7 ± 2.5 43.6 ± 2.3 43.2 ± 2.9
Gravid uterine weight (g) 73.0 ± 2.9 69.5 ± 3.5 73.9 ± 2.8 71.2 ± 4.1
Extragestational weightc (g) 289 ± 5.7 282 ± 3.9 295 ± 5.8 283 ± 3.5
Extragestational weight 47.6 ± 2.8 42.7 ± 2.2 50.9 ± 3.0 39.9 ± 3.5b
gaind (g)
Significant clinical signs None reported None reported None reported None reported
a
Mean ± standard error.
b
p<0.05, compared with corresponding control.
c
Body weight on gd 20 minus gravid uterine weight.
d
Extragestational weight minus body weight on gd 0.
Concentration (ppm)
No. implantations/dam 14.4 ± 0.55b 14.0 ± 0.71 15.3 ± 0.45 14.8 ± 0.63
No. resorptions/litter 0.46 ± 0.17 0.58 ± 0.17 0.96 ± 0.26 0.67 ± 0.14
Early resorptions/litter 0.39 ± 0.15 0.54 ± 0.16 0.88 ± 0.25 0.63 ± 0.14
Dead fetuses/litter 0 0 0 0
Fetal body weight (g) 3.49 ± 0.04 3.44 ± 0.05 3.40 ± 0.05 3.50 ± 0.06
Sex ratio (% males) 50.2 ± 2.281 52.5 ± 2.95 50.5 ± 2.77 52.5 ± 2.58
a
One rat had only one implant; this animal was excluded from statistical evaluations.
b
Mean ± standard error.
Concentration (ppm)
Malformationsa
Meningoencephalocele -- -- -- 2/1
Variations
Reduced ossification
a
Expressed as number of fetuses/number of litters; includes only those findings occurring in more than one fetus
or at more than one concentration.
b
--, no malformations observed.
Concentration (ppm)
No. of deaths 0 0 0 3
Days 0-6 2.7 ± 0.3 3.0 ± 0.3 2.5 ± 0.2 2.3 ± 0.2
Days 6-11 5.5 ± 0.4 5.8 ± 0.3 5.6 ± 0.3 4.8 ± 0.3
Days 11-16 13.3 ± 0.6b 12.7 ± 0.4 11.4 ± 0.5c 10.6 ± 0.4c
Days 16-18 5.5 ± 0.3b 5.7 ± 0.3 4.7 ± 0.4 4.8 ± 0.3
Gravid uterine weight (g) 19.3 ± 1.00b 20.3 ± 0.80 18.0 ± 0.87 16.8 ± 0.67c
Extragestational weightd (g) 35.5 ± 0.48b 35.1 ± 0.44 34.5 ± 0.46 34.1 ± 0.36c
Extragestational weight gaine 7.60 ± 0.48b 6.99 ± 0.38 6.20 ± 0.38c 5.91 ± 0.28c
(g)
a
Mean ± standard error.
b
p 0.05, significant linear trend.
c
p 0.05, pairwise comparison with corresponding control parameter.
d
Body weight on gd 18 minus gravid uterine weight.
e
Extragestational weight minus body weight on gd 0.
Concentration (ppm)
No. implantations/dam 12.7 ± 0.52 13.3 ± 0.44 13.0 ± 0.64 13.1 ± 0.43
No. resorptions/litter 1.06 ± 0.22 0.84 ± 0.22 0.67 ± 0.20 0.90 ± 0.19
Early resorptions 1.00 ± 0.23 0.58 ± 0.21 0.43 ± 0.13a 0.75 ± 0.16
Dead fetuses/litter 0 0 0 0
No. fetuses/no. litters 11.7 ± 0.66 12.5 ± 0.52 12.3 ± 0.62 12.2 ± 0.51
examined
Fetal body weight (g) 1.34 ± 0.03b 1.28 ± 0.01 1.13 ± 0.02a 1.04 ± 0.03a
Placental weight (mg) 86.8 ± 2.99b 85.4 ± 2.29 78.6 ± 3.24a 72.6 ± 1.88a
Sex ratio (% males) 51.6 ± 3.91 49.8 ± 3.06 51.5 ± 3.68 51.8 ± 3.29
a
p 0.05, pairwise comparison with corresponding control.
b
p 0.05, significant linear trend.
Concentration (ppm)
Malformationsa
Variations
Pale 2/2 -- -- --
Abnormal sternebraec,d 0.6 ± 0.9 0.4 ± 0.7 0.4 ± 0.8 0.8 ±1.3e
Supernumerary ribsc,d 1.7 ± 2.3 1.6 ± 2.1 6.0 ± 3.6e 9.9 ± 3.0e
Concentration (ppm)
Reduced ossification (all sites)c 1.7 ± 1.7 1.2 ± 1.5 2.7 ± 2.7 3.9 ± 2.6e
Phalanges -- -- -- 2/16
a
Expressed as number of fetuses/number of litters; includes only those findings occurring in more than one fetus
or at more than one concentration.
b
--, no malformations or variations noted.
c
Mean percentage per litter (mean ± SD).
d
p<0.05, linear trend, orthogonal contrast test.
e
p<0.05, Tukey’s test.
f
p<0.05, Fisher exact test (fetal incidence).
105 weeks: 2.5 mg/kg: ovarian atrophy 2.5 mg/kg for 105 weeks
Males
105 weeks: 5 mg/kg: inflammation of epididymis 5 mg/kg for 105 weeks
Butadiene Female 0.005, 0.02, 0.09, 0.36, 1.43 mmol/kg: reduced ovarian and uterine 1.43 mmol/kg Doerr et al.,
monoepoxide B6C3F1 mice 1.43 mmol/kg weights; decreased follicular counts 1995, 1996
intraperitoneally once
daily for 30 days
Female none Doerr et al.,
Sprague- 1996
Dawley rats
Butadiene Female 0.002, 0.009, 0.036, 0.14, 0.14 mmol/kg: decreased ovarian and uterine 0.14 mmol/kg Doerr et al.,
diepoxide B6C3F1 mice 0.29 mmol/kg weights; decreased follicular counts 1995, 1996
intraperitoneally once
5-25
This chapter updates the evaluation of animal studies published from 1985 through January
1997. The study by Owen et al. (1987) is not evaluated here because it was reviewed previously
in U.S. EPA (1985) as the NTP study (1984), which was subsequently published by Owen et al.
(1987).
Figure 6-1. Kaplan-Meier survival curves for male B6C3F1 mice exposed to 1,3-butadiene by
inhalation for 103 weeks.
Source: NTP, 1993.
Figure 6-2. Kaplan-Meier survival curves for female B6C3F1 mice exposed to 1,3-butadiene
by inhalation for 103 weeks.
Source: NTP, 1993.
Effects in reproductive organs are discussed in Chapter 5. Organ weights, hematological indices,
and serum chemistry were not evaluated at 103 weeks.
In the 2-year study, bone marrow atrophy was recorded in 50% of males and 14% of females
exposed to 625 ppm.
The incidence of liver necrosis was increased at the higher exposure concentrations in males
and females, occurring in 8%, 10%, 16%, 27%, 29%, and 26% of males and in 4%, 4%, 14%, 10%,
38%, and 21% of females exposed to 0 (controls), 6.25, 20, 62.5, 200, and 625 ppm, respectively.
Centrilobular hepatocellular necrosis of the liver was seen in 4% and 8% of males exposed to 62.5
and 625 ppm, respectively, and in 2%, 8%, and 9% of females exposed to 62.5, 200, and 625 ppm,
respectively. Hepatocellular necrosis was not seen in any of the concurrent controls. Liver necrosis
with no particular lobular distribution was found primarily in animals with malignant lymphoma
and hemangiosarcoma; centrilobular hepatocellular necrosis was often found in animals described
as anemic and in animals with atrial thrombi.
Myocardial mineralization, a lesion of unknown pathogenesis, occurred with increased
frequency in both sexes at 625 ppm (males, 27%; females, 14%), but was not observed in controls.
A low incidence was observed at the lower concentrations. Myocardial mineralization was also
observed in a separate stop-exposure study in which male mice were exposed to 312 ppm 1,3-
butadiene for 52 weeks or 625 ppm for 13 or 26 weeks, and observed for periods up to 103 weeks.
The incidence of myocardial mineralization for these three exposure groups was 12%, 18%, and
28%, respectively. Details of the stop-exposure study are presented in Section 3.3.
Minimal to mild olfactory epithelial atrophy occurred in females exposed to 625 ppm and
in males exposed to concentrations $20 ppm. However, the incidence in males exposed to 625 ppm
was lower than that seen in females. The olfactory epithelial lesions were unilateral at the lower
concentrations and bilateral at the higher concentrations. The lesions were similar to those seen in
the NTP (1984) study, but osseous or cartilaginous metaplasia was not observed. The investigators
considered olfactory nasal atrophy a possibly compound-related lesion.
Compared with controls, mice exposed to 1,3-butadiene exhibited increased incidences of
proliferative lesions (hyperplasia) in several organs, including the heart, lungs, forestomach, ovaries,
mammary gland, and Harderian gland (Table 6-2). Hyperplasia of the endothelium (cardiac blood
vessels), alveolar epithelium, forestomach epithelium (focal), germinal epithelium and granulosa
cells of the ovaries, mammary gland, and Harderian gland were all considered preneoplastic lesions.
Other preneoplastic lesions identified in the 2-year study were hepatocellular foci (basophilic, clear
cell, mixed cell, and eosinophilic) in female mice exposed to 1,3-butadiene. Hepatocellular foci
were observed in 16% of controls and in 29%, 38%, 24%, 10%, and 5% of females exposed to 6.25,
20, 62.5, 200, and 625 ppm, respectively. Hyperplastic lesions were also observed in separate
studies with male B6C3F1 mice using variable exposure and durations (stop-exposure experiments).
Hyperplasia in these studies occurred primarily in the endothelium (cardiac blood vessels), alveolar
epithelium, forestomach epithelium, and Harderian gland (Table 6-3).
1/28/98
Concentration (ppm)
Organ/tissue Sex
0 6.25 20 62.5 200 625
Heart, endothelium M 0/50 (0%) 1/49 (2%) 0/50 (0%) 2/48 (4%) 4/48 (8%) 5/73 (7%)
F 0/50 (0%) 2/50 (4%) 1/50 (2%) 4/49 (8%) 5/50 (10%) 8/80 (10%)
Lung, alveolar epithelium M 2/50 (4%) 9/50 (18%) 6/50 (12%) 13/49 (27%) 17/50 (34%) 12/73 (16%)
F 5/50 (10%) 5/50 (10%) 3/50 (6%) 9/50 (18%) 11/50 (22%) 11/78 (14%)
Forestomach, epithelium M 4/50 (8%) 3/50 (6%) 3/50 (6%) 5/48 (10%) 4/48 (18%) 40/72 (56%)
F 4/50 (8%) 5/49 (10%) 4/47 (9%) 7/48 (15%) 14/50 (28%) 47/79 (59%)
Ovary, germinal epithelium F 2/49 (4%) 3/49 (6%) 8/48 (17%) 15/50 (30%) 15/50 (30%) 19/79 (23%)
6-7
Ovary, granulosa cells F 1/49 (2%) 0/49 (0%) 2/48 (4%) 3/50 (6%) 3/50 (6%) 2/79 (3%)
Mammary gland F 2/50 (4%) 0/50 (0%) 2/50 (4%) 4/50 (8%) 7/50 (14%) 2/80 (3%)
DRAFT--DO NOT CITE OR QUOTE
Harderian gland M 1/50 (2%) 3/49 (6%) 4/50 (8%) 6/47 (13%) 8/47 (17%) 5/40 (13%)
F 1/50 (2%) 5/49 (10%) 9/48 (19%) 4/49 (8%) 4/49 (8%) 7/66 (11%)
6.3. CARCINOGENICITY
The first NTP mouse inhalation study of 1,3-butadiene was terminated early due to induction
of fatal neoplasms (NTP, 1984); therefore, a second study (NTP, 1993) was conducted to better
characterize the exposure-response relationship for neoplasms and nonneoplastic lesions induced
in mice by exposure to 1,3-butadiene for 2 years. The concentrations ranged from
100-fold lower (6.25 ppm) up to the lowest concentration (625 ppm) used in the first study. In
addition, stop-exposure studies were conducted to assess the relationship between concentration and
duration of exposure on the induction of neoplasms by 1,3-butadiene. Results of this study have
also been published by Melnick et al. (1990a, b, c) and Melnick and Huff (1992). Miller and
Boorman (1990) provided morphological descriptions of the neoplastic lesions induced in B6C3F1
mice by 1,3-butadiene. The results are presented here in two parts, 2-year study and stop-exposure
study.
Concentration (ppm)
Target organ Neoplastic lesion
0 6.25 20 62.5 200 625
a
All organs Malignant lymphoma (histiocytic, 4/50 (8%) 2/50 (4%) 4/50 (8%) 6/50 (12%) 2/50 (4%) 51/73 (70%)
lymphocytic, mixed, NOS, or 9.8%b 5.1% 12.2% 17.7% 4.0% 95.4%
undifferentiated) p<0.001c p=0.302N p=0.528 p=0.238 p=0.627 p<0.001
Histiocytic sarcoma 0/50 (0%) 0/50 (0%) 4/50 (8%) 5/50 (10%) 7/50 (14%) 4/73 (5%)
0.0% 0.0% 10.6% 14.3% 31.9% 10.8%
p<0.001c p=0.051 p=0.021 p<0.001 p=0.043
Malignant lymphoma or histiocytic 4/50 (8%) 2/50 (4%) 8/50 (16%) 11/50 (22%) 9/50 (18%) 55/73 (75%)
sarcoma 9.8% 5.1% 21.5% 29.6% 34.7% 95.9%
p<0.001c p=0.302N p=0.118 p=0.022 p=0.005 p<0.001
Heart Hemangiosarcoma 0/50 (0%) 0/49 (0%) 1/50 (2%) 5/48 (10%) 20/48 (42%) 4/73 (5%)
0.0% 0.0% 3.4% 19.4% 93.3% 44.6%
p<0.001c NA p=0.451 p=0.011 p<0.001 p<0.001
Lungs Alveolar/bronchiolar adenoma 18/50 (36%) 20/50 (40%) 10/50 (20%) 25/49 (51%) 21/50 (42%) 3/73 (4%)
46.9% 47.3% 28.2% 74.2% 100.0% 59.4%
6-10
Alveolar/bronchiolar adenoma, 21/50 (42%) 23/50 (46%) 19/50 (38%) 31/49 (63%) 35/50 (70%) 3/73 (4%)
adenocarcinoma, or carcinoma 54.9% 54.5% 53.6% 87.9% 100.0% 59.4%
p<0.001c p=0.552N p=0.276 p<0.001 p<0.001 p<0.001
Forestomach Squamous cell papilloma 1/50 (2%) 0/50 (0%) 0/50 (0%) 1/50 (2%) 7/50 (14%) 2/73 (3%)
2.5% 0.0% 0.0% 4.5% 51.7% 40.0%
p<0.001d p=0.535N p=0.486N p=0.739 p=0.012 p=0.446
Squamous cell papilloma or 1/50 (2%) 0/50 (0%) 0/50 (0%) 1/50 (2%) 8/50 (16%) 4/73 (5%)
carcinoma 2.5% 0.0% 0.0% 4.5% 54.5% 51.8%
p<0.001c p=0.481N p=0.545N p=0.679 p<0.001 p<0.001
Liver Hepatocellular adenoma 13/50 (26%) 13/50 (26%) 19/50 (38%) 16/48 (33%) 23/48 (48%) 5/72 (7%)
32.1% 31.3% 52.1% 57.0% 92.2% 100.0%
p=0.042d p=0.552 p=0.158 p=0.261 p=0.008 p=0.253
Hepatocellular carcinoma 11/50 (22%) 16/50 (32%) 16/50 (32%) 17/48 (35%) 26/48 (54%) 1/72 (2%)
26.0% 36.6% 44.8% 58.3% 100.0% 50.0%
p=0.036d p=0.142 p=0.389 p=0.088 p<0.001 p=0.347
Table 6-4. Incidence of primary neoplasms in male B6C3F1 mice exposed to 1,3-butadiene
by inhalation for 103 weeks (continued)
1/28/98
Concentration (ppm)
Target organ Neoplastic lesion
0 6.25 20 62.5 200 625
Liver (cont.) Hepatocellular adenoma or 21/50 (42%) 23/50 (46%) 30/50 (60%) 25/48 (52%) 33/48 (69%) 5/72 (7%)
carcinoma 47.9% 53.0% 70.1% 79.2% 100.0% 100.0%
p=0.067d p=0.375 p=0.078 p=0.185 p=0.030 p=0.450
Harderian gland Adenoma 6/50 (12%) 7/50 (14%) 8/50 (16%) 19/50 (38%) 30/50 (60%) 6/73 (8%)
14.8% 17.3% 25.8% 63.4% 95.4% 100.0%
p<0.001d p=0.497 p=0.395 p<0.001 p<0.001 p=0.264
Carcinoma 0/50 (0%) 1/50 (2%) 1/50 (2%) 3/50 (6%) 2/50 (4%) 0/73 (0%)
0.0% 2.6% 4.2% 11.7% 6.3% 0.0%
p=0.720d p=0.522 p=0.425 p=0.086 p=0.352 NA
Adenoma or carcinoma 6/50 (12%) 7/50 (14%) 9/50 (18%) 20/50 (40%) 31/50 (62%) 6/73 (8%)
14.8% 17.3% 29.5% 64.9% 95.5% 100.0%
p<0.001d p=0.497 p=0.217 p<0.001 p<0.001 p=0.002
Preputial gland Carcinoma 0/50 (0%) 0/50 (0%) 0/50 (0%) 0/50 (0%) 5/50 (10%) 0/73 (0%)
6-11
a
Overall rate: number of tumor-bearing animals/number of animals examined.
DRAFT--DO NOT CITE OR QUOTE
b
Survival-adjusted rate. Kaplan-Meier estimated neoplasm incidence at the end of the study after adjustment for intercurrent mortality.
c
Life table test. Beneath the control incidence are the p values associated with the trend test. Beneath the dosed group incidence are the P values corresponding to pairwise comparison between
the control and dosed groups. The life table analysis regards neoplasms in animals dying prior to terminal kill as being (directly or indirectly) the cause of death.
d
Logistic regression test. This test regards the neoplasms as nonfatal.
Alveolar/bronchiolar 0/50 (0%) 5/50 (10%) 11/50 (22%) 9/50 (18%) 19/49 (39%) 8/78 (10%)
adenocarcinoma or carcinoma 0.0% 13.3% 42.9% 40.8% 100.0% 100.0%
p<0.001c p=0.029 p<0.001 p<0.001 p<0.001 p<0.001
Alveolar/bronchiolar adenoma, 4/50 (8%) 15/50 (30%) 19/50 (38%) 24/50 (48%) 25/49 (51%) 22/78 (28%)
DRAFT--DO NOT CITE OR QUOTE
Concentration (ppm)
Target organ Neoplastic lesion
0 6.25 20 62.5 200 625
Liver (cont.) Hepatocellular adenoma or 15/49 (31%) 14/49 (29%) 15/50 (30%) 19/50 (38%) 16/50 (32%) 2/80 (3%)
carcinoma 39.3% 34.3% 45.5% 74.8% 91.7% 100.0%
p=0.497d p=0.504N p=0.441 p=0.027 p=0.008 p=0.302
Ovary Benign granulosa cell tumor 1/49 (2%) 0/49 (0%) 1/48 (2%) 6/50 (12%) 6/50 (12%) 6/79 (8%)
2.8% 0.0% 3.2% 28.5% 100.0% 27.1%
p=0.030d p=0.517N p=0.735 p=0.026 p=0.020 p=0.303
Malignant granulosa cell tumor 0/49 (0%) 0/49 (0%) 0/48 (0%) 3/50 (6%) 2/50 (4%) 0/79 (0%)
0.0% 0.0% 0.0% 19.3% 54.2% 0.0%
p=0.068d NA NA p=0.046 p=0.037 NA
Benign or malignant granulosa cell 1/49 (2%) 0/49 (0%) 1/48 (2%) 9/50 (18%) 8/50 (16%) 6/79 (8%)
tumor 2.8% 0.0% 3.2% 42.9% 100.0% 27.1%
p=0.006d p=0.517N p=0.735 p=0.001 p=0.001 p=0.303
Mammary gland Adenoacanthoma 0/50 (0%) 1/50 (2%) 2/50 (4%) 6/50 (12%) 4/50 (8%) 0/80 (0%)
0.0% 2.9% 7.7% 32.5% 13.6% 0.0%
6-13
Malignant mixed tumor 0/50 (0%) 0/50 (0%) 0/50 (0%) 0/50 (0%) 0/50 (0%) 4/80 (5%)
0.0% 0.0% 0.0% 0.0% 0.0% 29.4%
p<0.001c NA NA NA NA p=0.003
Harderian gland Adenoma 8/50 (16%) 10/50 (20%) 6/50 (12%) 15/50 (30%) 20/50 (40%) 9/80 (11%)
20.8% 29.2% 20.7% 61.0% 89.3% 45.2%
p=0.046d p=0.356 p=0.511N p=0.016 p=0.001 p=0.176
Carcinoma 0/50 (0%) 1/50 (2%) 1/50 (2%) 0/50 (0%) 1/50 (2%) 0/80 (0%)
0.0% 2.7% 2.3% 0.0% 50.0% 0.0%
p=0.873Nd p=0.493 p=0.631 NA p=0.085 NA
Adenoma or carcinoma 8/50 (16%) 10/50 (20%) 7/50 (14%) 15/50 (30%) 20/50 (40%) 9/80 (11%)
20.8% 29.2% 22.5% 61.0% 89.3% 45.2%
p=0.061d p=0.356 p=0.575N p=0.016 p=0.001 p=0.176
a
b
Overall rate; number of tumor-bearing animals/number of animals examined.
c
Survival-adjusted rate. Kaplan-Meier estimated neoplasm incidence at the end of the study after adjustment for intercurrent mortality.
Life table test. Beneath the control incidence are the p values associated with the trend test. Beneath the dosed group incidence are the p values corresponding to pairwise comparison between
d
the control and dosed groups. The life table analysis regards neoplasm in animals dying prior to terminal kill as being (directly or indirectly) the cause of death.
Logistic regression test. This test regards the neoplasms as nonfatal.
NA = not applicable; no tumors in these groups.
NOS = not otherwise specified.
N = incidence in dose group is lower than in control group.
Source: NTP, 1993.
Table 6-6. Incidence of primary neoplasms in male B6C3F1 mice exposed to 1,3-butadiene
by inhalation for 9 months and 15 months
1/28/98
Concentration (ppm)
Target organ Neoplastic lesion
0 6.25 20 62.5 200 625
All organs Malignant 9 months 1/10
lymphoma
(histiocytic,
lymphocytic,
mixed, NOS, or 15 months 2/7
undifferentiated)
Heart Hemangiosarcoma 9 months
a
Overall rate: number of tumor-bearing animals/number of animals examined.
b
Survival-adjusted rate. Kaplan-Meier estimated neoplasm incidence at the end of the study after adjustment for intercurrent mortality.
c
Life table test. Beneath the control incidence are the p values associated with the trend test. Beneath the dosed group incidence are the p values corresponding to pairwise comparison
between the control and dosed groups. The life table analysis regards neoplasms in animals dying prior to terminal kill as being (directly or indirectly) the cause of death.
d
Logistic regression test. This test regards the neoplasms as nonfatal.
Concentration (ppm)
Target organ Neoplastic lesion
0 6.25 20 62.5 200 625
All organs Malignant lymphoma 9 months 1/8
(lymphocytic, mixed,
NOS, or undifferentiated 15 months 1/10 1/10 0/2
cell type)
Heart Hemangiosarcoma 9 months
15 months 1/10 2/2
Lungs Alveolar/bronchiolar 9 months 2/10 1/8
adenoma,
adenocarcinoma, or 15 months 3/10 3/10 ½
carcinoma
Forestomach Squamous cell papilloma 9 months
or carcinoma 15 months 1/10 2/10 ½
Liver Hepatocellular adenoma 9 months
or carcinoma 15 months 1/10 1/10 0/10 1/10 3/10 ½
Ovary Benign or malignant 9 months 1/10
granulosa cell tumor 15 months 1/10 4/10 ½
Mammary gland Adenoacanthoma, 9 months
adenocarcinoma,
carcinoma, or malignant 15 months 2/10 ½
mixed tumor
Harderian gland Adenoma or carcinoma 9 months 1/5
15 months 2/9 1/1 1/1 1/10 3/10 0/2
a
Overall rate; number of tumor-bearing animals/number of animals examined.
b
Survival-adjusted rate. Kaplan-Meier estimated neoplasm incidence at the end of the study after adjustment for intercurrent mortality.
c
Life table test. Beneath the control incidence are the p values associated with the trend test. Beneath the dosed group incidence are the p values corresponding to pairwise comparison
between the control and dosed groups. The life table analysis regards neoplasm in animals dying prior to terminal kill as being (directly or indirectly) the cause of death.
d
Logistic regression test. This test regards the neoplasms as nonfatal.
Figure
6 - 3 .
Kaplan-
Meier
survival
curves
f o r
m a l e
mice in the stop-exposure inhalation study of 1,3-butadiene.
Source: NTP, 1993.
Table 6-8. Incidence of primary neoplasms in male B6C3F 1 mice exposed to 1,3-butadiene by
inhalation in the stop-exposure study
Parameters 0 ppm 200 ppm 625 ppm 312 ppm 625 ppm
Duration of exposures (weeks) 103 40 13 52 26
Total exposure (ppm weeks) 0 8,000 8,000 16,000 16,000
Target organ Neoplastic lesion
Hematopoietic Lymphocytic malignant lymphoma 2/50a (4%) 6/50 (12%) 17/50 (34%) 4/50 (8%) 30/50 (60%)
4.7%b 26.7% 35.8% 100.0% 81.5%
-- p=0.033c p<0.001 p=0.034 p<0.001
Lymphoma (mixed or NOS) 2/50 (4%) 2/50 (4%) 5/50 (10%) 4/50 (8%) 3/50 (6%)
5.3% 7.8% 34.8% 58.0% 43.3%
-- p=0.382c p=0.010 p=0.005 p=0.002
Histiocytic sarcoma 0/50 (0%) 5/50 (10%) 2/50 (4%) 7/50 (14%) 2/50 (4%)
0.0% 21.3% 28.9% 43.0% 15.6%
-- p=0.006c p=0.011 p<0.001 p=0.036
Malignant lymphoma or histiocytic 4/50 (8%) 13/50 (26%) 24/50 (48%) 15/50 (30%) 35/50 (70%)
sarcoma 9.8% 46.8% 72.1% 100.0% 91.2%
-- p<0.001c p<0.001 p<0.001 p<0.001
6-19
Malignant lymphoma (lymphocytic, mixed, 4/50 (8%) 8/50 (16%) 22/50 (44%) 8/50 (16%) 33/50 (66%)
or NOS) 9.8% 32.4% 58.2% 100.0% 89.5%
-- p=0.023c p<0.001 p<0.001 p<0.001
DRAFT--DO NOT CITE OR QUOTE
Heart Hemangiosarcoma 0/50 (0%) 15/50 (30%) 7/50 (14%) 33/50 (66%) 13/50 (26%)
0.0% 76.2% 61.8% 100.0% 100.0%
-- p<0.001c p<0.001 p<0.001 p<0.001
Lungs Alveolar/bronchiolar adenoma 18/50 (36%) 24/50 (48%) 17/50 (34%) 26/50 (52%) 12/50 (24%)
46.9% 94.3% 85.3% 100.0% 100.0%
-- p=0.015d p=0.044 p=0.001 p<0.001
Alveolar/bronchiolar 5/50 (10%) 22/50 (44%) 18/50 (36%) 16/50 (32%) 11/50 (22%)
adenocarcinoma or carcinoma 14.3% 89.5% 87.7% 100.0% 100.0%
-- p<0.001c p<0.001 p<0.001 p<0.001
Table 6-8. Incidence of primary neoplasms in male B6C3F1 mice exposed to 1,3-butadiene by
inhalation in the stop-exposure study (continued)
Concentration (duration of exposure)
1/28/98
Parameters 0 ppm 200 ppm 625 ppm 312 ppm 625 ppm
Duration of exposures (weeks) 103 40 13 52 26
Total exposure (ppm weeks) 0 8,000 8,000 16,000 16,000
Target organ Neoplastic lesion
Alveolar/bronchiolar adenoma, 21/50 (42%) 36/50 (72%) 28/50 (56%) 32/50 (64%) 17/50 (34%)
adenocarcinoma, or carcinoma 54.9% 100.0% 100.0% 100.0% 100.0%
-- p<0.001c p<0.001 p<0.001 p<0.001
Liver Hepatocellular adenoma 13/50 (26%) 27/49 (55%) 19/49 (39%) 19/50 (38%) 11/50 (22%)
32.1% 91.1% 91.1% 100.0% 100.0%
-- p<0.001d p=0.042 p=0.045 p=0.284
Hepatocellular carcinoma 11/50 (22%) 14/49 (29%) 14/49 (29%) 10/50 (20%) 4/50 (8%)
26.0% 50.3% 90.9% 74.6% 50.5%
-- p=0.530d p=0.142 p=0.453 p=0.393
Hepatocellular adenoma or carcinoma 21/50 (42%) 33/49 (67%) 24/49 (49%) 24/50 (48%) 13/50 (26%)
47.9% 93.4% 94.4% 100.0% 100.0%
-- p=0.004d p=0.063 p=0.169 p=0.561
6-20
Forestomach Squamous cell papilloma 1/50 (2%) 3/50 (6%) 4/50 (8%) 4/50 (8%) 4/50 (8%)
2.5% 21.4% 28.3% 100.0% 20.1%
-- p=0.195d p=0.260 p=0.181 p=0.301
DRAFT--DO NOT CITE OR QUOTE
Squamous cell carcinoma 0/50 (0%) 0/50 (0%) 4/50 (8%) 5/50 (10%) 6/50 (12%)
0.0% 0.0% 51.6% 33.1% 40.9%
-- NA p<0.001d p<0.001 p<0.001
Squamous cell papilloma or carcinoma 1/50 (2%) 3/50 (6%) 7/50 (14%) 9/50 (18%) 10/50 (20%)
2.5% 21.4% 56.6% 100.0% 52.8%
-- p=0.065c p<0.001 p<0.001 p<0.001
Harderian gland Adenoma 6/50 (12%) 26/50 (52%) 20/50 (40%) 28/50 (56%) 13/50 (26%)
14.8% 87.9% 94.3% 100.0% 100.0%
-- p<0.001d p=0.001 p<0.001 p=0.046
Table 6-8. Incidence of primary neoplasms in male B6C3F 1 mice exposed to 1,3-butadiene by
inhalation in the stop-exposure study (continued)
Parameters 0 ppm 200 ppm 625 ppm 312 ppm 625 ppm
Duration of exposures (weeks) 103 40 13 52 26
Total exposure (ppm weeks) 0 8,000 8,000 16,000 16,000
Target organ Neoplastic lesion
Carcinoma 0/50 (0%) 2/50 (4%) 4/50 (8%) 2/50 (4%) 0/50 (0%)
0.0% 5.6% 38.8% 51.5% 0.0%
-- p=0.397d p=0.190 p=0.006 NA
Adenoma or carcinoma 6/50 (12%) 27/50 (54%) 23/50 (46%) 30/50 (60%) 13/50 (26%)
14.8% 88.3% 100.0% 100.0% 100.0%
-- p<0.001d p<0.001 p<0.001 p=0.046
Kidney Renal tubule adenoma 0/50 (0%) 4/48 (8%) 1/50 (2%) 3/49 (6%) 1/50 (2%)
0.0% 17.4% 14.3% 27.8% 6.3%
-- p=0.073d p=0.273 p=0.075 p=0.731
Braine Malignant glioma 0/50 (0%) 0/50 (0%) 2/50 (4%) 0/50 (0%) 1/50 (2%)
Malignant neuroblastomas 0/50 (0%) 0/50 (0%) 2/50 (4%) 0/50 (0%) 0/50 (0%)
6-21
Preputial gland Carcinoma 0/50 (0%) 1/50 (2%) 4/50 (8%) 4/50 (8%) 3/50 (6%)
0.0% 10.0% 16.9% 100.0% 100.0%
-- p=0.368c p=0.039 p<0.001 p=0.002
DRAFT--DO NOT CITE OR QUOTE
Adenoma or carcinoma 0/50 (0%) 1/50 (2%) 5/50 (10%) 4/50 (8%) 3/50 (6%)
0.0% 10.0% 22.9% 100.0% 100.0%
-- p=0.368c p=0.013 p<0.001 p=0.002
Zymbal’s gland Adenoma or carcinoma 1/50 (2%) 1/50 (2%) 2/50 (4%) 0/50 (0%) 2/50 (4%)
2.9% 4.8% 8.8% 0.0% 37.3%
-- p=0.531c p=0.178 p=0.998N p=0.009
Table 6-8. Incidence of primary neoplasms in male B6C3F 1 mice exposed to 1,3-butadiene by
inhalation in the stop-exposure study (continued)
a
b
Overall rate, number of tumor-bearing animals/number of animals examined.
Survival-adjusted rate. Kaplan-Meier estimated neoplasm incidence at the end of the study after adjustment for intercurrent mortality.
c
Life table test. The p values for pairwise comparison of exposed groups with controls are beneath the exposed group incidence. N refers to negative association with control group. The life
table analysis regards neoplasms in animals dying prior to terminal kill as being (directly or indirectly) the cause of death.
d
e
Logistic regression test. This test regards the neoplasms as nonfatal.
No statistical analysis.
NA = not applicable.
NOS = not otherwise specified.
N = incidence in dose group is lower than in control group.
Source: NTP, 1993a.
Lymphocytic lymphomas of thymic origin occurred at a markedly increased incidence in
mice exposed to 625 ppm for 13 or 26 weeks. According to the life table test, the incidence of
lymphocytic lymphoma was also significantly increased in mice exposed to 200 ppm for 40 weeks
or 312 ppm for 52 weeks. The incidence of histiocytic sarcomas was significantly increased (life
table test) in mice in all stop-exposure groups as well.
The lower incidences of lymphocytic lymphomas at 200 ppm (40 weeks) and 312 ppm (52
weeks) compared to 625 ppm for 13 and 26 weeks, respectively, demonstrate that the concentration
of 1,3-butadiene is a greater contributing factor in the development of this lesion than the duration
of exposure, i.e., a high concentration for a short duration is more effective than a lower
concentration of longer duration. A comparison of the 200 ppm (40 weeks) versus the 625 ppm (13
weeks) and of the 312 ppm (52 weeks) versus the 625 ppm (26 weeks) lymphocytic lymphoma
results using a life table test confirms that the higher concentration/shorter duration regimen is
significantly more effective than the lower concentration/longer duration regimen within each
cumulative exposure grouping (p=0.005 for 8,000 ppm·weeks and p<0.001 for 16,000 ppm·weeks)
after survival differences are taken into account.
As observed in the 2-year study, lymphocytic lymphomas occurred very early after exposure
started: as early as 23 weeks in the group exposed to 625 ppm for 26 weeks and as early as 24
weeks in the group exposed to 625 ppm for 13 weeks. This lesion accounted for 24 and 17,
respectively, of the first 25 deaths occurring in these groups by weeks 45 and 79, respectively.
Therefore, early deaths due to lymphocytic lymphoma would have a tremendous negative effect on
the incidence of late-developing lesions.
Hemangiosarcomas of the heart, which also accounted for some of the early deaths, were
significantly increased in most stop-exposure groups compared with the controls. The highest
incidence, which was about twice as high as that of other groups, occurred in the group exposed to
312 ppm, followed by the groups exposed to 200 ppm and 625 ppm (26 weeks). The lowest
incidence occurred in the group exposed to 625 ppm for 13 weeks. Hemangiosarcomas appeared
at about 9 months in the 200, 312, and 625 ppm (26-week) stop-exposure groups. Comparison (life
table test) of groups having the same total exposures showed that the incidences of
hemangiosarcomas in mice exposed to 625 ppm were significantly lower than that of the
corresponding group exposed to 312 ppm (p=0.032) but not 200 ppm. The incidences of
hemangiosarcomas in both 625-ppm stop-exposure groups were higher than that in the 625-ppm 2-
year exposure group, probably due to longer survival of the stop-exposure groups.
The incidences of neoplastic lesions of the lung (alveolar/bronchiolar adenoma,
adenocarcinoma, or carcinoma) were significantly elevated in each exposure group. The highest
incidence occurred in the 200-ppm stop-exposure group, followed by the 312-, 625- (13 weeks),
and 625-ppm (26 weeks) groups. The adenomas developed after week 47 and the adenocarcinomas
and carcinomas developed after week 53; the late appearance of these lesions relative to lymphocytic
lymphomas probably accounted for the lowest incidence of lung neoplasms occurring in 625 ppm
(26 weeks) group. A life table analysis suggested the incidence of lung lesions in the 625 ppm (26
weeks) group was significantly greater than in the 312 ppm (52 weeks) group (p=0.013), but no
difference was detected between the 200 ppm (40 weeks) and 625 ppm (13 weeks) groups.
Mice exposed to 200 ppm 1,3-butadiene for 40 weeks had significantly increased incidences
of hepatocellular adenomas and adenomas/carcinomas combined; the incidences of hepatocellular
carcinomas analyzed alone were not significantly increased. Exposure to 1,3-butadiene at 312 ppm
or 625 ppm (13 or 26 weeks) did not increase the incidence of hepatocellular neoplasms of any type.
The earliest detection of these neoplasms was 67 weeks for the 625 ppm (13 weeks), 57 weeks for
the 200 ppm, 47 weeks for the 312 ppm, and 45 weeks for the 625 ppm (26 weeks) stop-exposure
groups. A logistic regression analysis found no differences between the 200 ppm and 625 ppm (13
weeks) or the 312 ppm and 625 ppm (26 weeks) groups.
A low incidence of squamous cell papillomas of the forestomach occurred in each of the
groups, and squamous cell carcinomas were seen in mice exposed to 312 ppm or 625 ppm for 13
and 26 weeks. The incidences of squamous cell papillomas were not significantly greater than
controls for any group, but the incidences of squamous cell carcinomas were significantly greater
by the life table test, which is considered to be the appropriate test (NTP, 1993) for these fatal
neoplasms. A life table analysis also revealed a statistically significant exposure-rate effect for the
squamous cell carcinomas in both of the total exposure groupings (p=0.019 for 8,000 ppm·weeks
and p=0.015 for 16,000 ppm·weeks), suggesting that the higher concentration/shorter duration
exposures were more potent.
The incidence of adenomas of the Harderian gland was significantly greater in each exposure
group than in the controls by a logistic regression test. A low incidence of Harderian gland
carcinomas occurred in mice exposed to 200 ppm for 40 weeks (not significant), 312 ppm for 52
weeks (p=0.006), and 625 ppm for 13 weeks (not significant). No Harderian gland carcinomas were
observed in the controls or in mice exposed to 625 ppm for 26 weeks. A logistic regression analysis
did not detect any exposure-rate effects.
Other neoplasms occurred at low incidence in the stop-exposure studies; they were
considered to be related to exposure because of their low spontaneous incidences in NTP historical
control male mice. These neoplasms occurred in the kidney, brain, Zymbal’s gland, and preputial
gland. The incidences of these neoplasms are also summarized in Table 6-8.
Renal tubule neoplasms occurred in historical male control mice; the range was 0 to 1%.
The small number of these neoplasms in each of the exposure groups are considered to be related
to administration of 1,3-butadiene because the incidences were greater than the upper range for
historical controls.
Brain neoplasms, including two neuroblastomas and two malignant gliomas, observed in
male mice exposed to 625 ppm for 13 weeks and one malignant glioma observed in male mice
exposed to 625 ppm for 26 weeks may have been related to 1,3-butadiene exposure. Brain
neoplasms are rare in untreated B6C3F1 mice; none have been reported in 574 NTP historical
control male mice. Furthermore, a low incidence of gliomas was also reported in the previous NTP
(1984) study. For these reasons, the brain neoplasms are considered exposure-related lesions.
A low incidence of preputial gland carcinomas occurred in the exposed groups in the stop-
exposure studies, and none were seen in controls. Compared with the incidence in concurrent
controls, the combined incidences of preputial gland tumors (adenoma and carcinoma) were
significant in male mice exposed to 312 ppm (52 weeks) and to 625 ppm (13 and 26 weeks) by the
life table test. Preputial gland carcinomas were not reported in a survey of NTP historical control
mice, further indicating that these neoplasms are probably related to exposure to 1,3-butadiene.
One male exposed to 200 ppm for 40 weeks, two males exposed to 625 ppm for 13 weeks,
and two males exposed to 625 ppm for 26 weeks developed Zymbal’s gland carcinomas. This lesion
did not occur in male mice exposed to 312 ppm for 52 weeks; one control male, however, developed
an adenoma. The combined incidence of Zymbal’s gland adenomas and carcinomas in animals
exposed to 625 ppm for 26 weeks was significantly increased compared with controls by the life
table test. Zymbal’s gland neoplasms are rare spontaneous neoplasms that had not been observed
in any NTP historical controls before the only occurrence of this adenoma in the control male mice
for these studies.
To summarize the results of the stop-exposure study pertaining to the relationship between
exposure level and duration of exposure: For lymphocytic lymphomas, there is strong evidence that
higher concentration/shorter duration exposures are more potent than the lower concentration/longer
duration exposures for both the 8,000 ppm-weeks and 16,000 ppm-weeks total exposure groupings.
There is also some evidence for a similar exposure-rate effect for forestomach squamous cell
carcinomas in both total exposure groupings. Any exposure-rate effects at other sites are less clear,
especially because it is difficult to distinguish a small apparent increased potency effect of higher
concentration/shorter duration exposures from an effect of longer potential postexposure follow-up
times following the shorter-duration exposures.
This updated review presents the evaluation of studies published from 1985 through
January 1997. The follow-up proposed by Lemen et al. (1990) of the cohort studied by
Meinhardt et al. (1982) and Downs et al. (1992), an abstract submitted for the International
Symposium are not reviewed in this evaluation. Lemen et al. (1990) did not present any results,
while no details of study design and analysis were available for Downs et al. (1992). Since
1985, investigators have conducted studies of workers who produce 1,3-butadiene as a raw
material (monomer production) or who use 1,3-butadiene in styrene-butadiene rubber (SBR)
production (polymer production).
1
Residual lymphohematopoietic cancers include ICD codes 200, 202, 203, 208, and 209.
2
It was not explained in the paper how the cohort was reduced to 2,582 from 2,586.
7.1.1.3. Divine et al., 1993: Cancer Mortality Among Workers at a Butadiene Production
Facility
This update added another 5 years of follow-up to the earlier cohort of monomer workers
(Divine, 1990). Cohort inclusion criteria remained the same but were extended from December
31, 1979, to December 31, 1990. This yielded additional workers resulting in a total cohort of
2,749 individuals. The four exposure groups were similar to those used in earlier studies with
slight changes as follows: (1) The background exposure group (included office utility,
warehouse, and transportation workers, N = 347). This group was called the low-exposure
group in the previous two studies (Downs et al., 1987; Divine, 1990). (2) The low-exposure
group (included workers from operating units, planners and engineers, welders, carpenters, and
workers from brick masons, N = 958). This group was a combination of some of the low-
exposure and all of the unknown exposure group from the previous two studies. (3) The
nonroutine exposure group (included skilled maintenance workers such as pipefitters, tinsmiths,
instrument and electrical workers, and insulators, N = 865). (4) The routine exposure group
(included process, lab, storage, and transport workers, N = 1056). Although the last two
categories appeared to be the same as in the earlier two studies, the change in the number of
individuals in these categories was not explained in the paper. For this study, the investigators
reviewed the results of the IH data and information obtained from the plant personnel and found
that the main difference between the routine and nonroutine exposure groups was in the
frequency and not the intensity of exposure.
Monson’s computer program (Monson, 1974) was used for the analysis of this study also.
All the analytical methods included use of white male death rates of the U.S. population (since
there were very few blacks in the study, they were assumed to be white for the analysis) and
calculation of person-years. The follow-up procedures and acquisition of death certificates were
the same as in an earlier study by Divine (1990).
A total of 83,591 person-years was accrued. At the end of the follow-up period, 1,660
individuals were still alive, 38 were lost to follow-up, and 1,051 were deceased (death
certificates were obtained for 1,036 individuals).
7.1.1.4. Divine and Hartman, 1996: Mortality Update of Butadiene Production Workers
This recent follow-up of the same cohort added 46 more individuals to the cohort (2,795)
by extending the inclusion criteria and the follow-up period through December 31, 1994. The
person-years accrued increased to 85,581. Of 2,795 individuals, 999 were still alive, 574 were
lost to follow-up (28 known to be alive), and 1,222 were deceased (death certificates were
obtained for 1,202 individuals). The follow-up procedures and analytical techniques (for SMR
analysis) were the same as for earlier studies. The exposure categories also remained the same
for this follow-up.
Based on IH data available since 1980, each employee’s potential exposure to butadiene
was estimated by separating the employee’s work history by job categories into 1-year segments.
Two variables were used to calculate the estimated exposure (job categories and calendar time
periods). There were six exposure classes based on job categories: 0, 1, 2, 3, 4, and 5 with 0,
0.1, 0.2, 0.3, 0.4, and 0.5 weights (wt), respectively, and five calendar time periods: <1946 (wt
= 10), 1946-59 (wt = 8), 1960-76 (wt = 4), 1977-85 (wt = 2), and 1986-94 (wt = 1). The
cumulative exposure was obtained for each individual by summing up the scores for all the years
of employment. These exposure estimates were used to conduct survival analyses for: (1) total
lymphohematopoietic cancer, (2) lymphosarcoma, (3) non-Hodgkin’s lymphoma, (4) multiple
myeloma, and (5) leukemias.
Three different models were used for the survival analysis, i.e., a Cox proportional
hazard model with a time-dependent estimate of cumulative exposure, a person-time logistic
regression model with a time-dependent estimate of cumulative exposure, and a nested case-
control model using conditional logistic regression. Each case had 10 matched controls by date
of birth
(+2 years). The selection of controls without replacement was from noncases at the time of the
occurrence of each case.
1. Mortality Analysis:
A total of 614 employees comprised the cohort. The follow-up period was from 1948 to
December 31, 1989. Vital status was assessed from company records, SSA, master beneficiary
files, and the National Death Index (NDI). Death certificates were obtained for all the deceased
workers and coded by a trained nosologist according to the revision of the ICD in effect at the
time of death. Mortality rates of Harris County, TX, were used to compute the age-, race-, and
calendar year-adjusted SMRs, using the Occupational Cohort Mortality Analysis Program
(OCMAP) from the University of Pittsburgh.
A total of 7,232 person-years were accrued. Of 614 employees, 589 were still alive, 1
was lost to follow-up, and 24 were dead. No excess mortality, either for total deaths or total
cancers (including cause-specific cancers), was observed.
2. Morbidity Analysis:
Original cohort members who were active at some time between January 1, 1982, and
December 31, 1989, qualified for the morbidity study. Morbidity data were obtained from the
Shell Health Surveillance System. The follow-up period was from 1982 to 1991. Causes of
morbidity were coded according to the 9th revision clinical modification of the ICD. Morbidity
cohort
Follow-up extended through
1985 SS SMR = 269 for prewar
subcohort
Comparison group U.S.
population SS SMR = 561 for routinely
exposed for less than 10
years
population
Divine and Hartman (1996) Update of the cohort from Based on IH data and work Results of SMR analysis 52 years follow-up
Divine et al. (1993) histories using were similar as earlier
6 exposure classes studies Exposure estimation useful
Cohort increased to 2,795 as the 5 calendar periods
inclusion period extended Survival analysis failed to Major limitation is no exposure
through December 31, 1994 Individual exposures were show any SS excess in any estimation available in prewar
estimated for each worker cause-specific cancer subcohort, which has the SS
Follow-up extended through lymphosarcoma excess
1994 Three different models used
DRAFT--DO NOT CITE OR QUOTE
U.S. population
Kanawha County population
SS = Statistically significant.
SNS = Statistically nonsignificant.
NSMR = National standard mortality ratios.
LSMR = Local standard mortality ratios.
IH = Industrial hygiene.
Because the findings of the published study are essentially the same, it will not be reviewed
again.
7.2.1.2. Matanoski et al., 1989: Epidemiologic Data Related to Health Effects of 1,3-Butadiene
Matanoski et al., 1990: Mortality of a Cohort of Workers in the Styrene-Butadiene
Polymer Manufacturing Industry (1943-1982)
These two publications essentially reported the same updated reanalysis of the earlier
cohort. In addition, Matanoski et al. (1989) also presented the results of the nested case-control
study in this population. Three methodological differences in the original analysis (Matanoski et
al., 1987) and the reanalysis presented in these two publications should be noted: extension of
follow-up through 1982, fewer workers whose vital status was unknown (3.4% vs.
6.6% in the earlier report), and deletion of workers from the Canadian plant who had relatively
short-term exposure (i.e., workers who had worked for less than 10 years or who had not
reached the age of 45 during employment). Analytical methods were essentially unchanged
from the earlier analysis.
In addition to information received from the SSA and the Motor Vehicle Administration,
follow-up through local plant beneficiary records and the National Death Index was done to
assess the vital status of the study cohort. Follow-up procedures for Canadian workers were
similar to the earlier study. Death certificates were obtained from the local health departments.
The total cohort was reduced from 13,920 to 13,422 in this study. Of 12,113 workers for whom
the vital status was successfully traced, 23% (2,784) were still working in the plants, 53.4%
(6,472) were alive but not working in the plants, 20.2% (2,441) had died, and vital status was
unknown for 3.4% (416). The racial distribution was 75% whites, 10% blacks, 15% unknown
(presumed to be white for the analysis), and less than 1% other.3 Death certificates were
obtained for 97.2% of the deceased individuals and were coded by a trained senior nosologist,
using the eighth revision of the ICD.
Data analyses were done by using age, race, calendar time, and cause-specific U.S.
population rates. A modified life-table program by Monson (1974) was used. The person-years
were calculated through December 31, 1982. The first-year work experience was omitted from
person-years because one of the inclusion criteria was that an individual had to have worked for
at least 1 year. A total of 251,431 person-years were accrued, of which 226,475 were
contributed by whites.
3
The percentages, which are quoted from the paper, add up to 101. This is due to the rounding of the numbers by the
authors of the paper.
7.2.1.3. Matanoski et al., 1989: Epidemiologic Data Related to Health Effects of 1,3-Butadiene
Santos-Burgoa et al., 1992: Lymphohematopoietic Cancer in Styrene-Butadiene
Polymerization Workers
To elucidate the separate contributions of 1,3-butadiene and styrene to the cancers
identified in the updated cohort study, a nested case-control study of this cohort of SBR workers
was conducted using estimates of exposure to 1,3-butadiene and to styrene for each job.
Fifty-nine cases and 193 controls (matched for duration of work) were included in the analysis.
Among the case group were 26 cases of leukemia; 18 of other lymphatic cancers, which included
10 multiple myelomas and 7 non-Hodgkin’s lymphomas; 8 Hodgkin’s lymphomas; and 6
lymphosarcomas.
Cases (workers who had lymphohematopoietic cancer as either the underlying or
contributory cause of death on death certificates) arose from the original eight plants with the
same selection criteria for the eligibility of that cohort (13,422), with the exception of the
Canadian plant. For the Canadian plant, the restriction of either 10 years of work or those who
had reached age 45 during employment was dropped from the selection of cases, which added
two more cases to lymphohematopoietic cancers. Another four cases were added in which
7.2.2.2. Macaluso et al., 1996: Leukemia and Cumulative Exposure to Butadiene, Styrene, and
Benzene Among Workers in the Synthetic Rubber Industry
A cohort mortality study conducted in synthetic rubber workers by Delzell et al. (1996)
(Section 7.2.2.1) had a component of exposure estimation. The exposures to 1,3-butadiene,
styrene, and benzene were estimated by Macaluso et al. (1996).
An exposure estimation was conducted on each and every worker based on detailed work
histories, work area/job specification, IH monitoring survey records, IH recommendations,
various records from the plants, historical aerial pictures, use of protective and safety equipment,
walk-through surveys, and interviews with plant management as well as long-term employees in
specific areas/jobs. The quantitative exposure estimation was based on process analysis, job
analysis, and exposure estimation. The job-exposure matrices (JEMs) were computed for 1,3-
butadiene, styrene, and benzene, which were linked to work histories of each employee.
Quantitative estimates of exposure to 1,3-butadiene and styrene were based on
background exposure plus task-specific exposure, using multiple exposure and point source
models, respectively. Input variables for these models were derived from several information
sources described earlier. Limited validation of exposure estimates was attempted by comparing
the available IH data from the 1970s and 1980s as well as actually measuring the air
concentrations of 1,3-butadiene and styrene under controlled conditions. The latter method
showed a good agreement among the methods of sampling, while the comparison of IH data
indicated overestimations of 1,3-butadiene exposure.
For each job, 8-h time-weighted average (TWA) intensities and the number of peak
exposures (15-min exposures over 100 ppm) were calculated. Based on job exposures, a JEM
database was developed that was linked with individual work histories to develop individual
quantitative work exposure estimates. For each individual, the exposure indices were multiplied
by the length of employment in that particular process or job and were added up for the total
employment period in various jobs to estimate the cumulative exposure.
Mortality analysis was done by calculating the SMRs and risk ratios (RR) using
estimated quantitative exposures to 1,3-butadiene, styrene, and benzene. Both cumulative ppm-
Matanoski et al. Nested case-control study Exposure to 1,3- For 1,3-butadiene: One of the strengths is attempt was
(1989) butadiene and styrene made to estimate actual exposure
DRAFT--DO NOT CITE OR QUOTE
Matanoski et Same as nested case- Exposure estimation Similar results with new Verification of cause of death
al. (1993) control study done based on controls
measurements New set of controls validates earlier
A new set of 3 controls provided by seven Reanalysis of cohort data for results
per case plants, IISRP, and three plants
DRAFT--DO NOT CITE OR QUOTE
NIOSH
Cause of death verified SS SMR = 163 for all LHC
by hospital records SS SMR = 181 for leukemia
and aleukemia
Cohort data reanalysis
1/28/98
Table 7-2. Epidemiologic studies of the health effects of exposure to 1,3-butadiene Cpolymer production
(continued)
1,3-Butadiene
Authors Population studied exposure Results Strengths and limitations
assessment
Macaluso et al. Cohort of seven U.S. and Exposure estimation Adjusted RRs for Methods used and efforts made for
(1996) one Canadian SBR conducted based on cumulative exposure to 1,3- exposure estimation are best efforts
workers mortality study several information butadiene of 0, <1, 1-19, 20- to date
sources including IH 79 and 80 + ppm-years
Worked for at least 1 were 1, 2.0, 2.1, 2.4, and Misclassification with respect to job
year between January 1, Quantitative exposure 4.5. may be possible but unlikely to be
1943, and January 1, estimates on Trend test was SS only in leukemia deaths
1992 background, task-
7-28
Delzell et al. Same as Macaluso et al. Same as Macaluso et Ever-hourly workers showed Same as Macaluso et al. (1996)
(1996) (1996) al. (1996) for leukemia
SS SMR = 143 for all ever- Cause of death not verified
Analysis by ever- hourly workers
hourly and never- SS SMR = 227 for blacks Histologic typing of leukemia not
hourly SS SMR = 187 for 1985+ available, thus leading to
year of death misclassification
Analysis by process SS SMR = 200 for 1950-59,
7-29
SS = Statistically significant.
SMR = Standard mortality ratio.
IH = Industrial hygiene.
RR = Risk ratios.
OR = Odds ratio.
LHC = Lymphohematopoietic cancers.
7-30
DRAFT--DO NOT CITE OR QUOTE
even at the lowest dose of 6.25 ppm (NTP, 1993). Occupational populations are exposed to
butadiene in the production/recovery of butadiene monomer and production of resins and
plastics. Exposure to this colorless, odorless gas is entirely via inhalation due to its extremely
volatile nature. The general population is exposed to butadiene in ambient air, the major sources
of its release in ambient air being automotive exhaust and cigarette smoke. Its potential to cause
cancer in humans has become an important public health issue.
Butadiene becomes diluted in ambient air and is eliminated by photooxidation. Thus it is
difficult to study the health effects of exposure to butadiene in the general population. Since
exposure to butadiene is ubiquitous in the general population, "unexposed" reference populations
used in occupational cohort studies are likely to contain a substantial number of individuals who
are exposed to butadiene nonoccupationally. Furthermore, the issue of health measurement is
complicated by the fact that occupational cohorts tend to be healthier than the overall general
population and have below average mortality, which is referred to as the "healthy worker effect."
Thus the standard mortality ratios observed in occupational cohorts, computed using the general
population as the reference group, are underestimations of real risk.
In conclusion, some of the causality criteria apply to monomer workers and occurrence
of lymphosarcoma while all the criteria apply well for leukemia among SBR workers. Based on
strength of association, dose-response relationship, specificity of cancer (leukemiaCspecific cell
type is not known at this time), and biological plausibility, there is sufficient evidence to
consider 1,3-butadiene a known human carcinogen.
8.1. INTRODUCTION
Several physiologically based pharmacokinetic (PBPK) models of 1,3-butadiene
metabolism and disposition have been developed to attempt to explain the interspecies
differences in the potency and site specificity of the carcinogenic response between mice and rats
and to provide a corresponding dosimetric basis for quantitatively extrapolating carcinogenic
potency from rodents to humans (Hattis and Wasson, 1987; Hallenbeck, 1992; Kohn and
Melnick, 1993; Johanson and Filser, 1993; Evelo et al., 1993; Medinsky et al., 1994). PBPK
models use species-specific physiological parameters such as alveolar ventilation rates and blood
flow rates, chemical-specific distribution parameters such as blood:air and tissue:blood partition
coefficients, and species- and chemical-specific metabolic rates to elucidate the
pharmacokinetics (i.e., the uptake, distribution, metabolism, and excretion) of a chemical.
Ideally, such models provide species-specific target tissue doses of the toxicologically
active form(s) of the chemical. Carcinogenic risks from bioassay data can then be extrapolated
to humans on the basis of equivalent effective doses, reducing some of the uncertainties that
occur when interspecies extrapolation is based simply on exposure to the parent compound,
especially when nonlinear physiological processes are involved. Assumptions must still be made
to the effect that the mechanisms of action of the active form(s) of the compound at the target
tissue(s) are the same across species and that the tissues of different species are equally sensitive.
If these assumptions are not valid, pharmacodynamic data and modeling are required for more
precise risk assessment.
PBPK models that fall short of describing target tissue doses of the active form(s) of a
chemical may still be useful for improving the dosimetric basis of interspecies extrapolation for
quantitative risk assessment. For example, it is well established that metabolic activation of
1,3-butadiene is probably necessary for its carcinogenic action (Chapter 4). Therefore, a PBPK
model describing the production and disposition of 1,2-epoxy-3-butene (EB), the first product of
metabolic activation of 1,3-butadiene, may be able to provide a better dose metric than the
default methodology of using exposure to 1,3-butadiene itself.
This chapter reviews and analyzes the six PBPK models for 1,3-butadiene that are
currently available and assesses their usefulness for quantitative risk assessment of 1,3-butadiene
based on interspecies extrapolation. Each of these PBPK models assumes, for simplicity, that
the transfer of 1,3-butadiene to tissues is blood flow-limited, that each tissue compartment is
"well mixed," and that tissue concentrations are in equilibrium with the venous blood
concentration leaving the tissue.
a
Awake.
b
Asleep.
c
The blood:air partition coefficient of 0.35 is the "best estimate" value from "fitting" the model. The
tissue:blood partition coefficients (P) are from functions of the blood:air partition coefficient for which the
"best estimate" value of 0.35 was used. Partition coefficients are assumed to be the same across species.
d
From Kreiling et al. (1986b).
e
From allometric scaling of the rodent values.
f
"Best estimate" from "fitting" the model.
Subscripts f, m, and lvr designate the fat, muscle, and liver and vessel-rich compartments (tissues), respectively.
Q: tissue blood flow rate.
V: tissue volume.
P: tissue:blood partition coefficient.
Biochemical parametersd
Liver V cyt1 (nmol/h/mg) 155.4 35.4 70.8
Liver Km cyt1 (mM) 0.002 0.00375 0.00514
Liver V cyt2 (nmol/h/mg) 12
Liver Km cyt2 (mM) 0.0156
Liver V EH (nmol/h/mg) 347.4 148.8 1,110
Liver Km EH (mM) 1.59 0.26 0.58
Liver V GST (nmol/h/mg) 30,000 14,460 2,706
Liver Km GST (mM) 35.3 13.8 10.4
Liver micro prot (mg/L) 11,600 16,800 14,500
Liver cyto prot (mg/L) 82,800 108,000 58,000
Lung V cyt1 (nmol/h/mg) 138.6 9.6 9
Lung Km cyt1 (mM) 0.00501 0.00775 0.002
Lung k hydr (h-1/mg) 0.1116 0.0792 0.1914
Lung V GST (nmol/h/mg) 6,380 2,652
Lung Km GST (mM) 36.5 17.4
Lung k GST (h-1/mg) 0.1536
Lung micro prot (mg/L) 3,000 3,000 3,000
Lung cyto prot (mg/L) 82,800 108,000 58,000
a
Compartment volumes are given as fractions of body weight; compartment blood flow rates are given as
fractions of cardiac output.
b
Human cardiac output at rest: 336 L/h; human ventilation rate at rest: 240 L/h.
c
Lung:air and tissue:blood; assumed same for all species.
d
Data from Csanády et al. (1992).
Physiological data
Body weight (g) Standard animal 25 250
Simulations 27.5 157.5-217.5a
b
Partition coefficients
DRAFT--DO NOT CITE OR QUOTE
a
Depending on experiment simulated.
DRAFT--DO NOT CITE OR QUOTE
b
Tissue:blood and blood:air; assumed same for all species.
c
From Filser (1992).
d
Obtained by best fit.
e
Kreuzer et al. (1991).
f
Average of literature data.
g
Deutschmann and Laib (1989).
h
Kreiling et al. (1988).
Physiological parameters
Blood:air 0.894
Fat:blood 32.362
Liver:blood 2.675
Muscle:blood 1.871
Kidney:blood 1.690
Lung:blood 1.272
Brain:blood 2.355
Vessel rich:bloodb 2.02
Metabolic parameters
a
Same for all species.
b
Mean value of kidney:blood and brain:blood.
Physiological parameters:
Alveolar ventilation (L/hr/kg)a 17 41
Cardiac output (L/hr/kg)b 17 41
Body weight (kg)c 0.215-0.475 0.028-0.035
Tissue concentrations
Liver microsomal concentration (mg/g liver) 35 35
Lung microsomal concentration (mg/g lung) 20 20
Liver cytosolic concentration (mg/g liver)d 108 82.8
a
Obtained by optimization.
b
Ventilation/perfusion = 1.
c
Depending on experiment simulated.
d
From Csanády et al. (1992), with Vmax values scaled to whole organ using above microsomal concentrations.
EB: 1,2-epoxy-3-butene.
8.3. SUMMARY
Pharmacokinetic modeling of 1,3-butadiene has not yet elucidated the reasons for the
interspecies differences in carcinogenic response between mice and rats. It appears that either
the PBPK models are not sufficiently sophisticated to adequately model the relevant
pharmacokinetics (e.g., the models may need to incorporate the production and disposition of
DEB) or a pharmacodynamic component(s) (e.g., DNA susceptibility or repair) is required to
accurately correlate dose to response.
Furthermore, uncertainties in the existing PBPK models and data make them unreliable
for use in risk assessment. Serious uncertainties exist pertaining to the model structures,
parameter values, and validation. For example, there are discrepancies among the models and
X
(1) Multiplicative: RR = e
(2) Power: RR = e [ln(1+X)]
(3) Linear Excess: RR = 1 + X
(4) Polynomial Excess: RR = 1 + 1 Xp + 2 Xq +....
a
Adapted from Table 67 in Delzell et al. (1995). Results presented are adjusted for age, calendar year, years since
models are slight. The authors expressed a preference for the square root model as the best
model based on its goodness of fit and its simplicity. This model was refined into a "final
model" by omitting styrene and race because the effect of these variables on the estimated
parameter for 1,3-butadiene exposure was considered to have been minimal. In addition, certain
age, calendar year, and years since hire categories were collapsed for the final model for similar
reasons. The final model is summarized in Table 9-2. The relationship between cumulative 1,3-
butadiene exposure and leukemia mortality was highly statistically significant in this model
(p=0.002).
Linear term
(1,3-butadiene ppm- 0.17 0.10 9.41 (1) 0.002
years)0.5
a
This table is an adaptation of Table 68 in Delzell et al. (1995).
b
S.E. is the standard error of the parameter estimate.
c
Chi-square ( 2) and degrees of freedom (d.f.) based on the likelihood ratio statistic.
takes into account the effects of competing causes of death.1 U.S. age-specific mortality rates
for all race and gender groups combined (NCHS, 1993) were used to specify the leukemia and
all-cause background rates in the actuarial program. Exposures to 1,3-butadiene were assumed
to be continuous for the entire lifetime, and the risks were computed up to age 85. The
occupational 1,3-butadiene exposures in the epidemiologic study were converted to continuous
environmental exposures by multiplying the occupational exposure estimates by a factor to
account for differences in the number of days exposed per year (365/240 days) and another
1
This program is an adaptation of the approach that was previously used in BEIR IV.
Health Risks of Radon and Other Internally Deposited Alpha Emitters. National Academy Press,
Washington, DC, 1988, pp. 131-134.
2
The maximum reported SMR was 13.33. This SMR was based on two leukemia deaths
among black men from plant #2 with at least 10 years of work (not all of which was salaried) and
at least 20 years of elapsed time since hired. (See Table 29 of Delzell et al., 1995.)
Figur
e 9-2. Excess risk and 95% upper confidence limit excess risk estimates based on the
power model reported by Delzell et al., 1995.*
* Power model: RR= e [1v(1+X)]
Figure 9-4. Excess risk and 95% upper confidence limit excess risk estimates based on the
final square root model reported by Delzell et al., 1995.*
* Final square root model: RR=1 + x½
Ratios are also presented in Table 9-5 that were calculated by dividing the excess risk (p)
by the corresponding LECp for each model. Each ratio is the slope of the line segment
connecting the point (LECp, p) with the origin. Based on the LEC1, these ratios vary by
approximately one order of magnitude from 0.016 to 0.15. If these LEC1-based ratios were used
to calculate the concentration corresponding to a 1 in a million excess lifetime risk by linear
interpolation3, the values would range from 7 to 64 parts per trillion. The final model presented
by Delzell et al. (1995) would yield a corresponding exposure level of 12 parts per trillion.
Table 9-4. MLEs of parts per million continuous exposure concentrations
associated with varying excess risk levels with one-sided 95% lower confidence
3
Linear interpolation between the origin and the point (LECp, p) is also referred to as
“linear extrapolation.”
a
The ratio is the excess risk (p/100%) divided by the one-sided lower 95% confidence limit on the exposure
estimate (LECp).
as well. Moreover, for a given linear extrapolation, the ratios in Table 9-5 show that the
sensitivity of the result to the choice of excess risk level varies considerably for these models,
with the linear model being least sensitive and the two square root models being most sensitive.
Of the two square root models, however, the final relative rate model could be advantageous to
the other model if the omitted parameters for the effects of race and styrene exposure are
unnecessary.
For the suggested default approach under the proposed guidelines revisions, the EC01
level is chosen because that is within the observable response range of leukemia deaths. At the
EC01 level, the different models provide dose estimates ranging from 0.45 ppm to 1.16 ppm and
the 95% LCLs on dose ranging from 0.066 to 0.64. Without specific directions for choice from
the proposed guidelines, potency estimates based on each of the models examined by Delzell et
al. are presented in Table 9-6.
The cancer potency estimates using EC01s as the point of departure range from 8.7 ×
-3
10 /ppm (linear model) to 0.022/ppm (final square root model). The square root model was the
model preferred by Delzell et al. based on goodness of fit and simplicity; thus they chose that
model for various refinements, resulting in the final square root model. The cancer potency
estimates based on LEC01s range from 0.016/ppm to 0.15/ppm, with the final square root model
yielding 0.083/ppm while the linear model yields 0.019/ppm. Although the proposed Guidelines
do not offer explicit guidance on choice of model, it may be appropriate in this particular case to
use the final square root model to obtain the point of departure because this model benefits from
the refinements performed by Delzell et al.
As the estimates of choice, the MLEs of both the potency and EC01 are chosen. The main
reason for this choice is that these estimates are based on human data from a large, well-
conducted study. Although EPA has historically used upper-limit potency estimates for animal-
to-human extrapolations, these upper limits derive their use more from computational
instabilities of the MLEs in the quantitative risk models used. Human-to-human extrapolations
typically use a simpler linear model form that does not have these instabilities. Furthermore, the
human data inherently engender far less uncertainty in the risk estimates, so one may have more
confidence in the use of MLEs from human data than from animal data.
9.2.2.1. Quantal
When EPA estimates cancer risks for humans from rodent bioassay data, the risk
estimates are generally calculated from the incidence of rodents of the most sensitive species,
strain, and sex bearing tumors at any of the sites displaying treatment-attributable increases. In
the case of 1,3-butadiene, so many sites demonstrated significant tumor increases attributable to
1,3-butadiene that background levels of tumor-bearing animals obfuscate the effects of 1,3-
butadiene when all these tumor sites are combined. Therefore, risk estimates were derived from
the incidence of female (most sensitive sex) mice with malignant lymphomas, heart
hemangiosarcomas, lung tumors (alveolar/bronchiolar adenomas or carcinomas), mammary
gland tumors (carcinomas, adenocanthomas, or malignant mixed tumors), or benign or
malignant ovary granulosa cell tumors (Table 9-7). These sites were considered to be the most
relevant sites with low background tumor incidence. Most of the impact on the low-dose linear
extrapolation is from the lung tumor response, because the lung tumor incidences show the
Administered exposure
(ppm) Control 6.25 20 62.5 200
Human equivalent
exposure (ppm) 0 1.1 3.6 11 36
Number of mice with
tumorsa 6/50 19/49 26/50 31/50 46/49
Number of mice at riskb
a
Lymphocytic lymphomas, heart hemangiosarcomas, alveolar/bronchiolar adenomas or carcinomas, mammary
gland
tumors (carcinomas, adenocanthonomas, malignant mixed tumors), or benign or malignant ovary granulosa cell
tumors.
b
Female mice surviving to the time of the first significant tumor, which was a lymphocytic lymphoma at day 203.
largest increases at the lowest exposures. The 625 ppm exposure group was not included in the
dose-response analysis because all of the mice were dead by week 65, and the tumor response
was already virtually saturated in the 200 ppm exposure group. Note also that mice that died
before the time of observation of the first tumor were considered to be not at risk and were
excluded from the incidence denominators.
Human equivalent exposures were based on ppm 1,3-butadiene exposure, adjusted for
continuous daily exposure (e.g., 6.25 ppm × 6/24 × 5/7 = 1.12 ppm). No attempt was made to
adjust for internal doses of reactive 1,3-butadiene metabolites because the PBPK data were
inadequate to develop reliable PBPK models (Chapter 8). No adjustments were made for 1,3-
butadiene absorption because there are no adequate human data. Furthermore, there is no reason
to expect nonlinearities in absorption at the lowest exposures (at least < 625 ppm).
A 95% upper-limit incremental lifetime unit cancer risk (extra risk) for humans was
calculated from the incidence data in Table 9-7 using the LMS model. The multistage model has
the form:
where P(d) represents the lifetime risk (probability) of cancer at dose d, and parameters qi 0,
for I=0, 1, ..., k. Extra risk over the background tumor rate is defined as
Thus, the incremental unit cancer risk estimate (95% UCL) for humans calculated from the
mouse 1993 NTP inhalation bioassay results is 0.10 per ppm for continuous lifetime inhalation
exposure to 1,3-butadiene. The MLE of risk appears to be nearly linear between 1 ppm and 1
ppb and is about 0.075 per ppm 1,3-butadiene exposure.
Under EPA’s proposed new cancer risk assessment guidelines (U.S. EPA, 1996), unit
cancer risk estimates for genotoxic chemicals, such as 1,3-butadiene, would be derived by
straight linear extrapolation to 0 from the LED10 (estimated 95% UCL on the dose corresponding
to a 10% cancer risk). Using the LEC10 generated for the LMS model by GLOBAL86 yields a
unit cancer risk of 0.10/1.0 ppm = 0.10 per ppm, the same as the q1*. Using the EC10 yields
0.10/1.4 7.1 × 10-2 per ppm.
MLE of risk for a dose of 1 ppm = 7.4 × 10-2
MLE of risk for a dose of 1 ppb = 7.6 × 10-5
MLE of dose for a risk of 0.10 (EC10) = 1.4 ppm
95% UCL on dose for a risk of 0.10 (LEC10) = 1.0 ppm
The unit cancer risk estimate (95% UCL) derived above is intended to depict a plausible
upper limit on the risk of developing any 1,3-butadiene-attributable tumor over a full (70-year)
lifetime. However, using the quantal incidence data for total tumor-bearing mice in each
exposure group does not fully characterize the cancer potency reflected by the mouse bioassay
9.2.2.2. Time-to-Tumor
The mouse inhalation bioassay results demonstrate different dose-response relationships
for different tumor sites. To assess the characteristics of the dose-response relationships for
different tumor sites, time-to-tumor analyses were performed to adjust for competing mortality
from cancer at other sites.
Time-to-tumor analyses were conducted from the individual mice data, including the 9-
month and 15-month interim sacrifice data, for sites demonstrating an increased cancer
incidence. Benign and malignant tumors were combined for sites where appropriate. Thus
time-to-tumor analyses were performed for lung alveolar/bronchiolar adenomas or carcinomas;
lymphocytic lymphomas; heart hemangiosarcomas; hepatocellular adenomas or carcinomas;
Harderian gland adenomas or carcinomas; forestomach squamous cell papillomas or carcinomas;
malignant or benign ovary granulosa cell tumors (female); and mammary gland
adenocanthomas, carcinomas, or malignant mixed tumors (female). Preputial gland carcinomas
in male mice were not analyzed because not all the tissues were examined microscopically.
Data from the 625 ppm exposure groups were excluded from analysis because of
excessive early mortality, as in the quantal analysis discussed above. In addition, data from
interim sacrifices for specific sites were excluded for dose groups for which it appeared that
complete histopathological examination for that site was not performed on the entire interim
sacrifice group.
Human equivalent exposures were based on ppm 1,3-butadiene exposure, adjusted for
continuous daily exposure, as described above.
The general model used for the time-to-tumor (or time-to-response) analyses was the
multistage Weibull model, which has the form
where P(d,t) represents the probability of a tumor (or other response) by age t (in bioassay
weeks) for dose d (human equivalent exposure), and parameters z 1, t0 0, and qi 0 for i=0, 1, ...,
k, where k = the number of dose groups - 1. The parameter t0 represents the time between when
Tissue Q0 Q1 Q2 Q3 Z
Tissue Q0 Q1 Q2 Z
Table 9-11. Human unit cancer risk estimates (extra risk, computed for risks of
10 -6 ) based on male mouse tumor incidences, w/o 625 ppm group using multistage
Weibull time-to-tumor model
Lymphocytic lymphoma 6.437 × 10-3 2.220 × 10-3 46.6 16.1 6.224 × 10-3
Heart hemangiosarcoma 0.01266 4.040 × 10-3 12.0 7.59 0.01318
Lung 0.1023 0.06998 1.48 1.01 0.09890
Liver 0.04447 0.02720 3.80 2.33 0.04300
Forestomach 4.258 × 10-3 1.660 × 10-5 19.2 13.3 7.517 × 10-3
Harderian gland 0.07402 0.05398 1.92 1.40 0.07157
Histiocytic sarcoma 0.02162 0.01394 7.42 4.78 0.02090
where the standard deviation is the square root of the variance, summing the variances across
tumor sites to obtain the variance of the sum of the MLEs, and calculating the 95% UCL on the
sum from the variance of the sum using the same formula. The resulting 95% UCL on the unit
potency for the total unit risk was 0.38/ppm. In comparison, summing the q1*s across the female
mouse tumor sites yielded 0.50/ppm.
The unit potencies were also summed using a Monte Carlo analysis and the software
Crystal Ball version 4.0 (Decisioneering, Denver, CO). Normal distributions were assumed for
the unit potency for each tumor site, with the mean equal to the MLE and as calculated from
the above formula. A distribution around the sum of the MLEs was then generated by
simulating the sum of unit potencies picked from the distributions for each tumor site (according
to probabilities determined by those distributions) 10,000 times. The mean for the sum and the
95th percentile on the distribution were the same as the sum of MLEs and 95% UCL calculated
above, as they should be. However, a sensitivity analysis based on the contribution to variance
revealed that variability associated with the unit potency estimate for the histiocytic sarcomas
was contributing more than 83% of the variance on the sum, and some of the simulated sums
were negative (the distributions for the unit potency estimates were not constrained for the
summation analyses). Excluding the histiocytic sarcomas yielded the same MLE of total risk of
9.2.3. Discussion
Based on the analyses discussed above, the best estimate for an upper bound on human
extra cancer risk from continuous lifetime exposure to 1,3-butadiene derived from animal data is
about 0.3/ppm. This estimate reflects the time-to-tumor response as well as the exposure-
response relationships for the multiple tumor sites (excluding histiocytic sarcomas) in the most
sensitive species and sex (the female mouse). Histiocytic sarcomas were excluded because they
introduced excessive variance into the upper bound while contributing only negligibly to the
MLE of total unit risk.
The greatest source of uncertainty in this estimate is from the interspecies extrapolation
of risk from the mouse to humans. The two rodent species for which bioassay data were
availableCthe mouse and the ratCvaried significantly in their carcinogenic responses to 1,3-
butadiene, in terms of both site specificity and degree of response (Chapter 6). The mouse and
rat also exhibit substantial quantitative differences in their metabolism of 1,3-butadiene to
potentially reactive metabolites (Chapter 3). Unfortunately, existing pharmacokinetic models
have been unable to explain the species differences in carcinogenic response (Chapter 8), and it
is likely that there are pharmacodynamic as well as pharmacokinetic differences between the
mouse and rat with respect to their sensitivities to 1,3-butadiene.
The mouse was the more sensitive species to the carcinogenic effects of 1,3-butadiene
exposure and, hence, the more conservative (public health protective) for the extrapolation of
risk to humans. In addition, the mouse appears to be the more relevant species for extrapolation
to humans in terms of site specificity, as 1,3-butadiene induces tumors of the
lymphohematopoietic system in both mice and humans. Melnick and Kohn (1995) further
suggest that the genetic mutations observed in 1,3-butadiene-induced mouse tumors are
analogous to genetic alterations frequently observed in human tumors.
In addition to uncertainties pertaining to the relevance of the rodent models to human
risk, there is uncertainty in quantitatively scaling the animal risks to humans. Ideally, a PBPK
model for the internal dose of the reactive metabolite(s) would decrease some of the quantitative
uncertainty in interspecies extrapolation; however, current PBPK models are inadequate for this
purpose (Chapter 8). In vitro metabolism data for humans suggest that interhuman variability in
the capacity to metabolically activate 1,3-butadiene nearly spans the range between rats and mice
(Chapter 3).
Minimal Mild Moderate Minimal Mild Moderate Minimal Mild Moderate Marked
0.00 0 0 0 0 0 0 1 (2) 2 (4) 1 (2) 0
6.25 ppm -- -- -- 0 0 0 0 15 (31) 4 (8) 0
20.00 ppm -- -- -- 1 (10) 0 0 1 (2) 23 (48) 8 (17) 0
62.50 ppm 0 0 0 1 (10) 7 (70) 1 (10) 3 (6) 18 (36) 21 (42) 0
200.00 ppm 0 0 9 (90) 0 1 (10) 6 (60) 0 9 (18) 34 (18) 0
625.00 ppm 0 0 8 (100) 0 0 2 (100) 0 19 (24) 47 (59) 3 (4)
Table 9-15. NTP chronic study (1993) (continued)
1/28/98
0 10 0 10 0 50 1 (2)
6.25 ppm -- -- 1 0 49 0
20 ppm -- -- 10 0 50 1 (2)
0 10 0 10 0 50 1 (2)
20 ppm -- -- 1 0 50 4(8)
where P(d, s) is the probability of a low-weight fetus at dose d and litter size s, and the
parameters , , , 1 , and 2 are estimated by methods of maximum likelihood. In order to get
an acceptable fit, an intercept parameter (d0) was included in the model (sometimes referred to as
a threshold parameter, i.e., the point at which the model can no longer distinguish from
background). The parameter constraints were: d0 0; 1; 0 - 1 s 1.
Fetal weight also was modeled as the average of mean fetal weights per litter using the
continuous power model (Allen et al., 1994b). The THWC model software (ICF Kaiser
International, KS Crump Group) was used for this purpose. Several cutoff values were used,
based on Kavlock et al. (1995):
(1) a 5% reduction in mean fetus weight/litter from the control mean,
*
The EC is the effective (exposure) concentration associated with a given level of risk,
5% in this case.
**
The LEC is the lower confidence limit on the effective concentration associated with a
given level of risk. The LEC is also known as the benchmark concentration.
m(d) = + d,
where m(d) is the mean of the mean fetus weight/litter for dose d, and , , and are parameters
estimated by maximum likelihood methods. The parameter constraints were: 0; 1.
2
Goodness of fit was determined by a test for the log-logistic model, and by an F test
for the continuous power model (U.S. EPA, 1995, Appendix A). The model was considered to
provide an acceptable fit if the p value was greater than 0.05 and a graphical display of the data
showed a good fit of the model.
A third approach used to model fetal weight data was the hybrid approach proposed by
Gaylor and Slikker (1990) and further developed by Crump (1995). The BENCH_C model
software (ICF Kaiser International, KS Crump Group) was used for this purpose. This approach
uses all of the information contained in the original observations by modeling changes in mean
response as a function of exposure concentration, but defines ECs and LECs in terms of
probability of response. The continuous data are fit using a model that incorporates parameters
from the quantal model. Several models are possible within the software for both continuous
data and quantal risk estimates. For this study, the log-logistic model (not including litter size)
was used for the quantal risk estimates and the following model for the continuous portion of the
hybrid model:
where N is the standard normal distribution function, m(d) is the mean response at dose d, is
the standard deviation of the response fixed for all dose groups, and and k are the log-logistic
model parameters estimated by the maximum likelihood method. The parameter constraints
were: k 1; 0.
Crump (1995) indicated that a background rate (P0) of 5% and an EC corresponding to
10% additional risk corresponds to a change from the control mean of 0.61 SD. Since a change
in mean fetal weight/litter of 0.5 SD corresponded on average to a NOAEL in studies by
Kavlock et al. (1995), a P0 of 0.05 and an EC10 (10% additional risk) were used here.
Log-logistic (1-4)a Individual fetal 5th percentile EC05 = 46.85 LEC05 = 0.079
weight 27.02
a
Exposure levels modeled in each case are shown in parentheses.
Figure 9-7. Observed versus predicted mean fetal weight per litter using continuous
model.
This model was used because of the wide spacing of doses and the lack of linearity in the dose-
response relationship. The data were limited in that only two exposure levels in addition to
controls were used, and the exposure levels differed by two orders of magnitude.
Although a statistically significant effect was noted at 12.5 ppm and 1,250 ppm for the
incidence of late deaths in the original paper (Anderson et al., 1993), the response in late deaths
at the higher exposure level was lower than at 12.5 ppm, probably because there were so many
early deaths at the higher level. For the same reason, the incidence of congenital abnormalities
was higher at 12.5 ppm than at 1,250 ppm. When early and late deaths were combined, a
consistently increasing response with increasing exposure level was seen. When combined, the
incidence in the controls was increased from 0 to 13 (4.68% of total implants), while in the 12.5
ppm group the incidence increased from 7 (2.29%) to 23 (7.52%).
a
Table 9-17. ECs and LECs for male-mediated developmental toxicity
Prenatal data Postnatal data
Post- Litter
No. Early and Live No. implantation size at Litter size
Estimate implants late deaths implants implants loss birth at weaning
Figure 9-10. Observed versus predicted proportion of early and late deaths per implantation (prenatal)
using log-linear model .
Figure 9-12. Observed versus predicted mean number of implants (postnatal) using log-linear model.
Figure 9-14. Observed versus predicted mean litter size at birth using log-linear model.
groups, no other endpoints showed a statistically significant increase at 12.5 ppm by pairwise comparison.
However, there was a trend toward an increase in the incidence of early and late fetal deaths and percent
postimplantation loss, and a decrease in percent live implants and litter size at birth and at weaning in the 12.5
ppm exposure group. Given the overall effect seen on development in this study, the NOAEL for most endpoints
was considered to be much closer to 12.5 ppm than to 1,250 ppm. Since litter size at birth and at weaning showed
the lowest ECs and LECs, these endpoints will be used for calculation of an RfC.
where P(d) is the probability of response at exposure level d and , , and are parameters that are estimated
from the observed dose-response data. Parameter constraints were 0; 0; > 0. The model was run to
2
determine the probability of additional risk. Goodness of fit was determined by a test. The model was
considered to provide an acceptable fit if the p value was greater than 0.05 and a graphical display of the data
showed a good fit of the model.
Table 9-18 gives the results of fitting the log-logistic model to the 2-year ovarian atrophy data for
exposure groups 1-5 and 1-4. The model gave a poor fit for all six exposure groups, because of leveling off of the
response at exposures above 62.5 ppm (36 ppm adjusted for continuous exposure). The best fit of the model was
for exposure groups 1-4, although the model also fit exposure groups 1-5 well (Figure 9-16; exposures adjusted for
continuous exposure), and the EC10s and LEC10s obtained for groups 1-4 and 1-5 were similar. As expected,
LEC10s were
lowest for ovarian atrophy at 2 years. Moderate ovarian atrophy at 2 years also was modeled using the quantal
Weibull model with exposure groups 1-5 or 1-4. The EC10 and LEC10 were higher than those for all lesions.
Ovarian atrophy data for all six exposure groups at 9 and 15 months were fit using the quantal Weibull or log-
logistic model.
Table 9-18. ECs and LECs for ovarian, uterine, and testicular atrophy using the quantal Weibull
and log-logistic models a
Endpoint Model NOAEL/LOAEL EC 10 LEC 10 p-Value
Ovarian atrophy - 2 years Log-logistic (1-5)b 1.1 ppm (LOAEL) 0.32 0.22 0.11
0.29c 0.21c
0.24c 0.17c
Ovarian atrophy - 2 year Quantal Weibull (1-5) 1.1 ppm 3.02 2.35 0.55
Moderate lesions only
Ovarian atrophy - 15 mos Log-logistic (1-6) 1.1 ppm 2.10 0.72 0.66
Ovarian atrophy - 9 mos Quantal Weibull (1-6) 11 ppm 20.04 9.95 0.83
Uterine and testicular atrophy data also were modeled using the quantal Weibull model. The quantal
Weibull model resulted in an acceptable fit of the 2-year uterine atrophy and
testicular atrophy data (Table 9-18 and Figures 9-17 and 9-18; exposures adjusted for continuous exposure).
However, the EC10s and LEC10s were much higher for these endpoints than for 9-month, 15-month or 2-year
ovarian atrophy data. LEC10s were estimated because it has been shown (Allen et al., 1994b) that, for quantal
responses, the LEC10 is near or below the range of detectable responses. Also, the Proposed Guidelines for
Carcinogen Risk Assessment (EPA, 1996) propose use of an LED10 as the default point of departure for low-dose
extrapolation, and use of an LEC10 as a default for noncancer estimation of an RfC would be consistent with this
approach.
Although some 9- and 12-month interim sacrifice data were available for ovarian, uterine, and testicular
atrophy (Table 9-15), these were less than ideal for modeling because smaller numbers of animals were killed and
not all dose groups were represented. In addition, some animals died or became moribund and were killed before
the 2-year death time point. To account for the variability in time of death, time-to-response analyses were done
using the multistage Weibull model as discussed in Section 9.2.2.2. Exposures were adjusted to the equivalent
Figure 9-18. Testicular atrophy (groups 1-6) using quantal Weibull model .
Table 9-19. Parameters for Weibull time-to-response model used to model reproductive effects
observed in mice based on ppm butadiene exposure 1
625 ppm
group
Response included Q0 Q1 Q2 Z
Ovarian atrophy no 4.86 × 10-6 7.06 × 10-6 - 2.21
-7 -6
yes 9.01 × 10 1.32 × 10 - 2.58
-5 -5
Uterine atrophy no 6.73 × 10 5.28 × 10 - 1.0
yes 9.08 × 10-5 9.74 × 10-6 1.31 × 10-6 1.0
-4 -5
Testicular no 4.28 × 10 2.24 × 10 - 1.0
atrophy -4 -4
yes 1.60 × 10 1.52 × 10 - 1.0
1
Each response was considered to be incidental with induction time, T0=0. See Section 9.2.2.2 on time-to-tumor
modeling of the mouse carcinogenicity data for a discussion of the Weibull model structure and selection.
Acute/
subchronic- LOAEL-to-
NOAEL Interspecies Intraspecies to-chronic NOAEL Risk RfC based on
(or LOAEL) EC 10 LEC 10 uncertainty uncertainty uncertainty uncertainty reduction NOAEL RfC based on
Effect (ppm) (ppm) (ppm) factor factor factor factor factor a (ppm) LEC 10 (ppm)
Decreased 28 14 3 10 1b 3 0.14
fetal weight
40 (LOAEL)
(10 d, 6h/d, 3 10 1b 10 0.13
GD 6-15)
2.2 (LOAEL)
(10 week, 3 10 3 10 0.002
adjusted to 24
9-48
h/d)
1.1 (LOAEL)
(2 year, 1.5e 10 1 10 0.007
adjusted to 24
DRAFT--DO NOT CITE OR QUOTE
h/d)
36
(2 year, 3 10 1 1 1.2
adjusted to 24
h/d)
a
To decrease risk to below what would be a detectable level, analogous to the LOAEL-to-NOAEL uncertainty factor.
b
Although from acute study, only exposure during gestation is assumed to be relevant to fetal weight.
cAdjusted to 24-h daily exposure.
d
Adjusted to chronic continuous exposure.
e
There is strong evidence that ovarian atrophy is caused specifically by the metabolite 1,2:3,4-diepoxybutane, and humans are thought to produce less of this metabolite than mice, although their relative
sensitivity to the metabolite is unknown (see text).
gestation are relevant. Although dominant lethal effects appear to occur with exposure during a
specific time period of spermatogenesis (i.e., only certain stages of developing sperm appear
susceptible), chronic exposure might result in continuous induction of these effects, so a factor
of 3 was used.
Under the NOAEL/LOAEL approach, the NOAEL is defined as the exposure level for
which there is no observed adverse effect, although it is circumscribed by the detection limit of
the study. For endpoints for which there is no NOAEL, an uncertainty factor of 10 is typically
used to attempt to extrapolate from the LOAEL to a level at which there are presumed to be no
detectable effects. In the benchmark dose approach, the typical "point of departure" corresponds
to a 10% increased response level, which is explicitly not a no-effect level. In this risk
assessment, a risk reduction factor of 3 was used to extrapolate to a level below which no
detectable effects would be expected, analogous to the LOAEL-to-NOAEL uncertainty factor.
Final guidance on this methodology is still being developed by EPA.
In addition to the sample RfCs presented in Table 9-21 for lifetime 1,3-butadiene
exposure, two RfCs were calculated for subchronic exposure. An RfCDT of 0.14 ppm for
developmental toxicity from short-term exposures was calculated for decreased fetal weight,
using the same factors depicted in Table 9-21. This RfCDT is identical to the sample RfC
calculated for decreased fetal weight because no subchronic-to-chronic uncertainty factor was
used in that calculation. Finally, an RfC for subchronic exposure was calculated for the
decreased litter size endpoints from the subchronic dominant lethal study. Using the LEC10 of
0.15 ppm and uncertainty factors of 3 for interspecies extrapolation, 10 for intraspecies
variability, and 3 for risk reduction (analogous to the LOAEL-to-NOAEL uncertainty factor), as
described above, yields an R/D RfC for subchronic exposure of 0.0015 ppm.
9.4.3. Discussion
The EC10s in Table 9-21 suggest that the dominant lethal (male-mediated) effect is the
most sensitive reproductive/developmental endpoint (i.e., the "critical" endpoint), and thus
should be the basis for the final R/D RfC. The dominant lethal effect also yields the lowest
sample RfC of 0.5 ppb. To arrive at the final R/D RfC, a further uncertainty factor of 3 is used
to account for the lack of comprehensive reproductive testing, especially the absence of a
multigenerational study. This final calculation yields an R/D RfC of 0.15 ppb.
There are substantial uncertainties in estimating low-exposure human risks for
reproductive and developmental effects observed in animals exposed to high concentrations of
an agent. It is generally believed that there is a nonlinear low-dose exposure-response
relationship for noncancer effects, and perhaps a threshold, although this is difficult to
demonstrate empirically. The shape of this low-dose exposure-response relationship is unclear,
9.4.4. Conclusions
In conclusion, an R/D RfC of 0.15 ppb was calculated for the critical endpoint of the
dominant lethal effect of decreased litter size at birth (or at weaning), based on mouse data. This
reference concentration, the uncertainties discussed above notwithstanding, is presumed to
represent a daily exposure to humans that is likely to be without an appreciable risk of
reproductive or developmental effects during a lifetime.
10.1. EVALUATION
1,3-Butadiene is a colorless, odorless chemical that exists in ambient air in gaseous form.
This extremely volatile chemical is very slightly soluble in water and is not found in soil and food.
Thus, exposure to 1,3-butadiene is mainly via inhalation. Increased mortality from leukemias and
lymphomas was observed among male workers occupationally exposed to 1,3-butadiene in
polymer and monomer production, respectively. No information is available in females. The data
from one Canadian and seven U.S. polymer production plants show that exposure to 1,3-
butadiene is causally associated with occurrence of leukemias (cell type is not known at this time).
Two lifetime inhalation studies in mice and one lifetime inhalation study in rats found
occurrence of malignant tumors in multiple sites in both mice and rats. Increased occurrence of
lymphomas in a 1-year inhalation study in Swiss mice indicated that the presence of retrovirus was
not an essential factor for the development of 1,3-butadiene-induced lymphomas.
Once inhaled, 1,3-butadiene is distributed throughout the body. The relative distribution
of 1,3-butadiene in different organs is unknown at this time. 1,3-butadiene is metabolized by
oxidation to a monoepoxide, diepoxide, and epoxy diol. Which metabolite(s) is responsible for
the causation of cancer is still uncertain. Differences in measured concentration levels of these
metabolites in mice and rats do not provide an explanation for the differences observed in
malignancies in these two species. All three of these metabolites have been shown to be
mutagenic in vivo and in vitro.
10.2. CONCLUSION
Based on the overall evidence from human, animal, and mutagenicity studies, 1,3-
butadiene is concluded to be a known human carcinogen.
11.1. INTRODUCTION
The U.S. Environmental Protection Agency (EPA) first published a health assessment of
1,3-butadiene in 1985. The 1985 assessment concluded that 1,3-butadiene was a possible human
carcinogen and calculated an upper bound cancer potency estimate of 0.25/ppm based on mouse
data. Since then, a number of new studies on 1,3-butadiene have been completed in various
disciplines such as epidemiology, toxicology, and pharmacokinetics. The purpose of this effort
was to review the new information and determine if any changes were needed to the earlier
conclusions.
This reassessment is intended to serve as a source document for risk assessors inside and
outside the Agency. Its development, however, was prompted primarily by a request from EPA’s
Office of Mobile Sources (OMS) to support decision making regarding the Air Toxic Rule’s
Section 202 (l) (2) of the Clean Air Act Amendment. The scope of the document has been limited
to address only the health effects specifically requested by OMS: carcinogenicity, mutagenicity,
and reproductive/developmental toxicity. Similarly, a detailed exposure assessment was not
requested and not conducted. For background purposes, however, some exposure information
has been included.
The major findings of this report are as follows. First, sufficient evidence exists to
consider 1,3-butadiene a known human carcinogen. The evidence for this includes findings in
epidemiologic studies as well as clear evidence that 1,3-butadiene is an animal carcinogen and is
metabolized into genotoxic metabolites by experimental animals and humans.
Second, based on linear modeling of human data, the best estimate of lifetime extra cancer
risk from continuous 1,3-butadiene exposure is about 9 × 10-3/ppm, or 9 × 10-6/ppb. In other
words, it is estimated that 9 persons in 1 million exposed to 1 ppb 1,3-butadiene continuously for
their lifetimes would develop cancer as a result of their exposure. Lower cumulative exposures
are expected to result in risks that are proportionately lower.
Third, although there are no human data on reproductive or developmental effects, a
variety of such effects have been observed in mice and rats exposed to 1,3-butadiene. A
reproductive/developmental reference concentration (RfC) of 0.05 ppb was calculated based on
the critical reproductive effect of reduced litter sizes, reflecting increased prenatal mortality,
observed among the offspring of male mice exposed to 1,3-butadiene.
Fourth, there are insufficient data to determine if children or other special subpopulations
are differentially affected by exposure to 1,3-butadiene. Heavy smokers are likely to be more
heavily exposed than the general population.
Number of
workers,
Plants dates studied Authors Approach Significant findings
7 U.S. and 1 15,000, Delzell et al., Cohort study Excess mortality due to
Canadian 1943-1994 1996 using leukemia;
polymer Macaluso et al., quantitative leukemia risk increased
production plants 1996 exposure with increasing
(UAB cohort)a estimates for exposure level
each worker
7 U.S. and 1 13,500, Matanoski and Cohort studies Excess mortality due to
Canadian 1943 - 1985 Schwartz, 1987 using qualitative lumpho- hematopoietic
polymer Matanoski et al., exposures; cancers;
production plants 1989, 1990, and case-control leukemia risk increased
(JHU cohort)a 1993 study using with increasing
Santos-Burgoa et estimated exposure level in case-
al., 1992 quantitative control study
exposures for
each case and
control
1 U.S. monomer 2,800, Downs et al., Cohort studies Excess mortality due
production plant 1943-1994 1987 using qualitative to lymphosarcoma
(Texaco cohort) Divine, 1990 exposures, last in prewar workers
Divine et al., 1993 study made
Divine and quantitative
Hartman, 1996 exposure
estimates
3 U.S. monomer 364, Ward et al., 1995 Cohort study Excess mortality due to
production plants 1940-1990 and 1996a using qualitative lymphosarcoma in
(Union Carbide exposures World War II workers
cohort)
a
Six U.S. plants and one Canadian plant were common in Johns Hopkins University (JHU) and University of
Alabama, Birmingham (UAB) studies.
1
One Canadian plant and six U.S. plants were common in the JHU and UAB studies.
a
Relative odds is the ratio of the frequency of exposure to 1,3-butadiene in cases to the frequency of exposure to
1,3-butadiene in controls, where both the cases and controls are from the same occupational cohort.
Upper bound on
Species Sex Tumor sites/types potency (ppm -1)
M Leydig cell, pancreatic exocrine cell, Zymbal 4.2 × 10-3
a
Rat gland
F Mammary gland, thyroid follicular cell, Zymbal 5.6 × 10-2
gland
M Lymphocytic lymphomas, histiocytic sarcomas,
b
Mouse heart hemangiosarcomas, lung, forestomach, 0.22
Harderian gland, liver, preputial gland
F Lymphocytic lymphomas, heart
hemangiosarcomas, lung, forestomach, Harderian 0.29
gland, liver, ovary, mammary gland
M Lymphocytic lymphomas 6.4 × 10-3
F Lymphocytic lymphomas 2.4 × 10-2
a
From U.S. EPA’s 1985 assessment; linearized multistage model.
b
Based on 1993 NTP study; Weibull multistage time-to-tumor model.
lymphocytic lymphomas in mice, because this was the tumor type in rodents most analogous to
the lymphohematopoietic cancers observed in workers exposed to 1,3-butadiene.
In both rodent species, females are apparently more sensitive than males, as evidenced by
the higher risk estimates. The “best estimate” (i.e., MLE from the linear model) of 8.7 × 10-3/ppm
for extra cancer risk from the human (male) leukemia data exceeds the upper bound estimates
based on the male rat data and on the male mouse data for lymphocytic lymphomas, and is 25
times lower than the upper bound estimate based on all male mouse tumors.
Human health risk estimates based on extrapolation from high-quality epidemiologic
results are preferable to those based on rodent data because they avoid the uncertainties inherent
in extrapolating across species and, typically, the human exposures in epidemiologic studies are
closer to anticipated environmental exposures than the high exposures used in animal studies, thus
reducing the extent of low-dose extrapolation. In the case of 1,3-butadiene, while the rat
exposures were far in excess of human exposures, the lowest EXPOSURE in the 1993 NTP
mouse study (4.7 ppm, 8 h TWA) is within the range of occupational exposures (0.7-1.7 ppm
median and 39-64 ppm max 8 h TWAs for work-area groups). However, interspecies differences
Epidemiology
The medical records for the leukemia cases in the studies by Delzell et al. and
Macaluso et al. should be reviewed to verify the cell types of leukemias.
Further follow-up of these studies is recommended because it will give an opportunity
to observe whether any noncancer effects, such as cardiovascular, or any cancers with
a longer latency period are associated with exposure to 1,3-butadiene.
All epidemiologic studies to date have examined male cohorts. Some butadiene
production facilities around the world (e.g., China) employ women in their
laboratories. If the number of women in these facilities is large enough, a
reproductive/developmental study would help determine if female workers are at risk
of reproductive effects or if exposed fetuses are at risk of developmental effects.
Toxicology
Elucidation of the mechanisms responsible for the interspecies differences in sensitivity
to 1,3-butadiene could assist in resolving questions about the human risk for
reproductive effects and for cancer at sites for which the Delzell et al. study may have
had insufficient power to detect an effect.
Molecular biology
Once the mechanisms of 1,3-butadiene-induced health effects are better understood,
information on polymorphisms in human metabolic enzymes (or DNA repair enzymes,
etc.) could help define sensitive subpopulations.
The unit cancer risk estimate has been changed from 0.25/ppm (upper bound based on
mouse data) to 0.009/ppm (best estimate based on linear modeling and extrapolation
of human data).
For the first time, an RFC (0.15 ppb) is calculated for reproductive/developmental
effects.
Acquavella, JF. (1989) The paradox of butadiene epidemiology. Exp Pathol 37:114.
Adler, ID; Anderson, D. (1994) Dominant lethal effects after inhalation exposure to 1,3-butadiene. Mutat Res
309:295-297.
Adler, ID; Cao, J; Filser, JG; et al. (1994) Mutagenicity of 1,3-butadiene inhalation in somatic and germinal cells
of mice. Mutat Res 309:307-314.
Adler, ID; Filser, JG; Gassner, P; et al. (1995) Heritable translocations induced by inhalation exposure of male
mice to 1,3-butadiene. Mutat Res 347:121-127.
Agency for Toxic Substances and Disease Registry (ATSDR) (1992) Toxicological profile for 1,3-butadiene. TP-
91/07, PB93-11690. Public Health Service, U.S. Department of Health and Human Services, Atlanta, GA.
Albertini, RJ; Castle, KL; Borcherding, WR. (1982) T-cell cloning to detect the mutant 6-thioguanine-resistant
lymphocytes present in human peripheral blood. Proc Natl Acad Sci USA 79:6617-6621.
Allen, BC; Kavlock, RJ; Kimmel, CA; et al. (1994a) Dose-response assessment for development toxicity: III.
Statistical models. Fundam Appl Toxicol 23:496-509.
Allen, BC; Kavlock, RJ; Kimmel, CA; et al. (1994b) Dose-response assessment for developmental toxicity: II.
Comparison of generic benchmark dose estimates with NOAELs. Fundam Appl Toxicol 23:487-495.
American Congress of Government Industrial Hygienists (ACGIH). (1994) World-wide limits for toxic and
hazardous chemicals in air, water, and soil. ACGIH Pub. No. 9565.
Anderson, EL; U.S. EPA Carcinogen Assessment Group. (1983) Quantitative approaches in use to assess cancer
risk. Risk Anal 3:277-295.
Anderson, D; Edwards, AJ; Brinkworth, MH. (1993) Male-mediated F1 effects in mice exposed to 1,3-butadiene.
In: Butadiene and styrene: assessment of health hazards. IARC Scientific Publications. Vol. 127. Sorsa, M;
Peltonen, K; Vainio, H; et al., eds. Lyon, France: International Agency for Research on Cancer, pp. 171-181.
Anderson, D; Edwards, AJ; Brinkworth, MH. (1995) The detection of dominant lethal mutations and foetal
malformations, tumours and chromosome damage in the offspring of male mice treated sub-chronically with
butadiene. BIBRA Report No. 1060/5/95.
Andjelkovich, D; Taulbee J; Symons, M. (1976) Mortality experience of a cohort of rubber workers, 1964-1973. J
Occup Med 18:387-394.
Andjelkovich, D; Taulbee, J; Symons, M. (1977) Mortality of rubber workers with reference to work experience. J
Occup Med 19:397-405.
Arce, GT; Vincent, DR; Cunningham, MJ; et al. (1990) In vitro and in vivo genotoxicity of 1,3-butadiene.
Environ Health Perspect 86:75-78.
Armitage, P. (1971) Statistical methods in medical research. New York, NY: John Wiley and Sons, pp. 362-365.
Au, WW; Bechtold, WE; Wharton, EB, Jr.; et al. (1995) Chromosome aberrations and response to gamma-ray
challenge in lymphocytes of workers exposed to 1,3-butadiene. Mutat Res 334:125-130.
Bailer, AJ; Portier, CJ. (1988) Effects of treatment-induced mortality and tumor-induced mortality on tests for
carcinogenicity in small samples. Biometrics 44:417-431.
Barnes, DG; Dourson, M; U.S. EPA Reference Dose Work Group. (1988) Reference dose (RfD): Description and
use in health risk assessments. Reg Toxicol Pharmacol 8:471-86.
Beaumont, JJ; Breslow, NE. (1981) Power considerations in epidemiologic studies of vinyl chloride workers. Am
J Epidemiol 114:725-734.
Bechtold, WE; Strunk, MR; Chang, I-Y; et al. (1994) Species differences in urinary metabolite ratios between
mice, rats, and humans. Toxicol Appl Pharmacol 127:44-49.
Bechtold, WE; Strunk, MR; Thornton-Manning, JR; et al. (1995) Analysis of butadiene, butadiene monoxide and
butadiene diepoxide in blood by gas chromatography/gas chromatography/mass spectroscopy. Chem Res Toxicol
8:182-187.
Bernadini, S; Pelin, K; Peltonen, K; et al. (1996) Induction of sister chromatid exchange by 3,4-epoxybutane-1,2-
diol in cultured human lymphocytes of different GSTT1 and GSTM1 genotypes. Mutat Res 361:121-127.
Bolt, HM; Schmeidel, G; Filser, JG; et al. (1983) Biological activation of 1,3-butadiene to vinyl oxirane by rat
liver microsomes and expiration of the reactive metabolite by exposed rats. J Cancer Res Clin Oncol 106:112-116.
Bolt, HM; Filser, JG; Störmer, F. (1984) Inhalation pharmacokinetics based on gas uptake studies. Arch Toxicol
55:213-218.
Bond, JA; Csanády, GA. (1991) Species and organ differences in the in vitro metabolism of butadiene to
butadiene monoepoxide. Proc Am Assoc Cancer Res 32:119.
Bond, JA; Dahl, AR; Henderson, RF; et al. (1986) Species differences in the disposition of inhaled butadiene.
Toxicol Appl Pharmacol 84:617-627.
Bond, JA; Dahl, AR; Henderson, RF; et al. (1987) Species differences in the distribution of butadiene in tissues.
Am Ind Hyg Assoc J 48:867-872.
Boogaard, PJ; Bond, JA. (1996) The role of hydrolysis in the detoxication of 1,2:3,4-diepoxybutane by human, rat,
and mouse liver and lung in vitro. Toxicol Appl Pharmacol 141:617.
Boogaard, PJ; Sumner, SCJ; Bond, JA. (1996) Glutathione conjugation of 1,2:3,4-diepoxybutane in human liver
and rat and mouse liver and lung in vitro. Toxicol Appl Pharmacol 136:307-316.
Brunnemann, KD; Kagan, MR; Cox, JE; et al. (1990) Analysis of 1,3-butadiene and other selected gas-phase
components in cigarette mainstream and sidestream smoke by gas chromatography-mass selective detection,
Carcinogenesis 11:1863.
Budavari, S, ed. (1989) The Merck index, 11th ed. Rahway, NJ:Merck & Co., pp. 230-231.
California Air Resources Board. (1991) Proposed identification of 1,3-butadiene as a toxic air contaminant.
Prepared by the Office of Environmental Health Hazard Assessment, Sacramento, CA.
Campbell, DL; Mangino, J. (1994) Evaluation and improvement of the Puget Sound toxic air contaminants
emissions inventory. Prepared by Radian Corporation for the Office of Research and Development, U.S.
Environmental Protection Agency, Washington, DC, under EPA contract no. 68-D1-0031.
Carpenter, CP; Shaffer, CB; Weil, CS; et al. (1944) Studies on the inhalation of 1,3-butadiene; with a comparison
of its narcotic effect with benzol, toluol and styrene, and a note on the elimination of styrene by the human. J Ind
Hyg Toxicol 26:69-78.
Checkoway, H; Williams, TM. (1982) A hematology survey of workers at a styrene-butadiene synthetic rubber
manufacturing plant. Am Ind Hyg Assoc J 43:164-169.
Cheng, X; Ruth, JA. (1993) A simplified methodology for quantitation of butadiene metabolites. Application of
the study of butadiene metabolism by rat liver microsomes. Drug Metab Dispos 21:121-124.
Citti, L; Gervasi, PG; Turchi, G; et al. (1984) The reaction of 3,4-epoxy-1-butene with deoxyguanosine and DNA
in vitro: synthesis and characterization of the main adducts. Carcinogenesis 5:47-52.
Cochrane, JE; Skopek, TR. (1993) Mutagenicity of 1,3-butadiene and its epoxide metabolites in human TK6 cells
and splenic T cells isolated from exposed B6C3F1 mice. In: Butadiene and styrene: assessment of health hazards.
IARC Scientific Publications. Vol. 127. Sorsa, M; Peltonen, K; Vainio, H; et al., eds. Lyon, France: International
Agency for Research on Cancer, pp. 195-204.
Cochrane, JE; Skopek, TR. (1994a) Mutagenicity of butadiene and its epoxide metabolites: I. Mutagenic potential
of 1,2-epoxybutene, 1,2,3,4-diepoxybutane and 3,4-epoxy-1,2-butanediol in cultured human lymphoblasts.
Carcinogenesis 15:713-717.
Cochrane, JE; Skopek, TR. (1994b) Mutagenicity of butadiene and its epoxide metabolites: II. Mutational spectra
of butadiene, 1,2-epoxybutene, and diepoxybutane at the hprt locus in splenic T cells from exposed B6C3F1 mice.
Carcinogenesis 15:719-723.
Cole, P; Delzell, E; Acquavella, J. (1993) Exposure to butadiene and lymphatic and hematopoietic cancer.
Epidemiology 4:86.
Concawe. (1987) A survey of exposures to gasoline vapour. Report No. 4/87. de Hague: Concawe.
Cowles, SR; Tsai, SP; Snyder, PJ; et al. (1994) Mortality, morbidity, and haematologic results from a cohort of
long-term workers involved in 1,3-butadiene monomer production. Occup Environ Med 51:323-329.
Cox, DR. (1972) Regression models and life tables. J R Stat Soc B34:187-220.
Crisp, TM. (1992) Organization of the ovarian follicle and events in its biology: oogenesis, ovulation or atresia.
Mutat Res 296:89-106.
Crump, KS. (1995) Calculation of benchmark doses from continuous data. Risk Anal 15:79-89.
Crump, KS. (1984) A new method for determining allowable daily intakes. Fundam Appl Toxicol 4:854-871.
Csanády, GA; Bond, JA. (1991) Species differences in the biotransformation of 1,3-butadiene to DNA-reactive
epoxides: role in cancer risk assessment. Chemical Industry Institute of Toxicology, CIIT Activities 11:1-8.
Dahl, AR; Birnbaum, LS; Bond, JA; et al. (1987) The fate of isoprene inhaled by rats: comparison to butadiene.
Toxicol Appl Pharmacol 89:237-242.
Dahl, AR; Bechtold, WE; Bond, JA; et al. (1990) Species differences in the metabolism and disposition of inhaled
1,3-butadiene and isoprene. Environ Health Perspect 86:65-69.
Dahl, AR; Sun, DJ; Birnbaum, LS; et al. (1991) Toxicokinetics of inhaled 1,3-butadiene in monkeys: comparison
to toxicokinetics in rats and mice. Toxicol Appl Pharmacol 110:9-19.
de Meester, C; Poncelet, F; Roberfroid, M; et al. (1980) The mutagenicity of butadiene towards Salmonella
typhimurium. Toxicol Lett 6:125-130.
Del Monte, M; Citti, L; Gervasi, PG. (1985) Isoprene metabolism by liver microsomal monooxygenases.
Xenobiotica 15:591-597.
Delzell, E; Sathiakumar, N; Macaluso, M. (1995) A follow-up study of synthetic rubber workers. Final report
prepared under contract to International Institute of Synthetic Rubber Producers.
Delzell, E; Sathiakumar, N; Hovinga, M. (1996) A follow-up study of synthetic rubber workers. Toxicology
113:182-189.
Deutschmann, S. (1988) Vergleichende Untersuchungen zur Depletion von zellulärem Glutathion durch 1,3-
Butadien bei Maus und Ratte. Thesis. Ruhr-Universität, Bochum, Germany.
Deutschmann, S; Laib, RJ. (1989) Concentration-dependent depletion of non-protein sulfhydryl (NPSH) content in
lung, heart and liver tissue of rats and mice after acute inhalation exposure to butadiene. Toxicol Lett 45:175-183.
Divine, BJ. (1990) An update on mortality among workers at a 1,3-butadiene facility— preliminary results.
Environ Health Perspect 86:119-128.
Divine, BJ; Hartman, CM. (1996) Mortality update of butadiene production workers. Toxicology 113:169-181.
Divine, BJ; Wendt, JK; Hartman, CM. (1993) Cancer mortality among workers at a butadiene facility. In:
Butadiene and styrene: assessment of health hazards. IARC Scientific Publications. Vol. 127. Sorsa, M; Peltonen,
K; Vainio, H; et al., eds. Lyon, France: International Agency for Research on Cancer, pp. 345-362.
Doerr, JK; Hooser, SB; Smith, BJ; et al. (1995) Ovarian toxicity of 4-vinylcyclohexene and related olefins in
B6C3F1 mice: role of diepoxides. Chem Res Toxicol 8:963-969.
Doerr, JK; Hollis, EA; Sipes, IG. (1996) Species difference in the ovarian toxicity of 1,3-butadiene epoxides in
B6C3F1 mice and Sprague-Dawley rats. Toxicology 113:128-136.
Downs, TD; Crane, MM; Kim, KW. (1987) Mortality among workers at a butadiene facility. Am J Ind Med
12:311-329.
Downs, T; Pier, S; Crane, M. (1992) Cause-specific mortality in a cohort of 1,000 ABS workers. Abstr Symp.
Butadiene and styrene: assessment of health hazards. Espoo, Finland.
Duescher, RJ; Elfarra, AA. (1994) Human liver microsomes are efficient catalysts for 1,3-butadiene oxidation:
evidence for major roles by cytochrome P450 2A6 and 2E1. Arch Biochem Biophys 311:342-349.
Dunnick, JK; Eustis, SL; Piegorsch, WW; et al. (1988) Respiratory tract lesions in F344/N rats and B6C3F 1 mice
after inhalation exposure to 1,2-epoxybutane. Toxicology 50:69-82.
Elfarra, AA; Duescher, RJ; Pasch, CM. (1991) Mechanisms of 1,3-butadiene oxidations to butadiene monoxide
and crotonaldehyde by mouse liver microsomes and chloroperoxidase. Arch Biochem Biophys 286:244-251.
Elfarra, AA; Sharer, JE; Duescher, RJ. (1995) Synthesis and characterization of N-acetyl-L-cysteine S-conjugates
of butadiene monoxide and their detection and quantitation in urine of rats and mice given butadiene monoxide.
Chem Res Toxicol 8:68-76.
Evelo, CTA; Oostendorp, JGM; ten Berge, WF; et al. (1993) Physiologically based toxicokinetic modeling of 1,3-
butadiene lung metabolism in mice becomes more important at low doses. Environ Health Perspect 101:496-502.
Filser, JG. (1992) The closed chamber technique—uptake, endogenous production, excretion, steady-state kinetics
and rates of metabolism of gases and vapors. Arch Toxicol 66:1-10.
Filser, JG; Bolt, HM. (1984) Inhalation pharmacokinetics based on gas uptake studies. Arch Toxicol 55:219-223.
Filser, JG; Althaler, B; Welter, HF; et al. (1992) Metabolism of 1,3-butadiene in microsomes from livers of mouse,
rat, and man. Naunyn-Schmiedeberg's Arch Toxicol Suppl 345:R31.
Fiserova-Bergerova, V; Diaz, ML. (1986) Determination and prediction of tissue gas partition coefficients. Int
Arch Occup Environ Health 58:75-87.
Frank, PM. (1978) Introduction to system sensitivity theory. New York: Academic Press.
Frome, EL; Checkoway, H. (1985) Use of Poisson regression models in estimating incidence rates and ratios. Am J
Epidemiol 121(2):309-22.
Gart, JJ; Chu, KC; Tarone, RE. (1979) Statistical issues in interpretation of chronic bioassay for carcinogenicity. J
Natl Cancer Inst 62:957-974.
Gaylor, DW; Slikker, W. (1990) Risk assessment for neurotoxic effects. Neurotoxicology 11:211-218.
Gervasi, PG; Longo, V. (1990) Metabolism and mutagenicity of isoprene. Environ Health Perspect 86:85-87.
Gervasi, PG; Citti, L; Del Monte, M; et al. (1985) Mutagenicity and chemical reactivity of epoxide intermediates
of the isoprene metabolism and other structurally related compounds. Mutat Res 156:77-82.
Gibson, RW; Bross, IDJ; Lilienfeld, AM; et al. (1968) Leukemia in children exposed to multiple risk factors. New
Engl J Med 279:906-909.
Goodrow, T; Reynolds, S; Maronpot, R; et al. (1990) Activation of K-ras by codon 13 mutations in C57BL/6 ×
C3HF1 mouse tumors induced by exposure to 1,3-butadiene. Cancer Res 50:4818-4823.
Hackett, PL; Sikov, MR; Mast, TJ; et al. (1987a) Inhalation developmental toxicology studies of 1,3-butadiene in
the rat (final report). Richland, WA: Pacific Northwest Laboratory; PNL Report No. PNL-6414 UC-48; NIH
Report No. NIH-401-ES-41031; 104 p. Prepared for NIEHS, NTP, under a Related Services Agreement with the
U.S. Department of Energy under contract DE-AC06-76RLO-1830.
Hackett, PL; Sikov, MR; Mast, TJ; et al. (1987b) Inhalation developmental toxicology studies: teratology study of
1,3-butadiene in mice (final report). Richland, WA: Pacific Northwest Laboratory; PNL Report No. PNL-6412
UC-48; NIH Report No. NIH-401-ES-40131; 92 p. Prepared for NIEHS, NTP, under a Related Services
Agreement with the U.S. Department of Energy under contract DE-AC06-76RLO-1830.
Hackett, PL; McClanahan, BJ; Brown, MG; et al. (1988a). Sperm-head morphology study in B6C3F 1 mice
following inhalation exposure to 1,3-butadiene (final technical report). Richland, WA: Pacific Northwest
Laboratory; PNL Report No. PNL-6459 UC-48; NIH Report No. NIH-Y01-ES-70153; 51 p. Prepared for NIEHS,
NTP, under a Related Services Agreement with the U.S. Department of Energy under contract DE-AC06-76RLO-
1830.
Hackett, PL; McClanahan, BJ; Mast, TJ; et al. (1988b). Dominant lethal study in CD-1 mice following inhalation
exposure to 1,3-butadiene (final technical report). Richland, WA: Pacific Northwest Laboratory; PNL no. PNL-
6545 UC-408; NIH no. NIH-Y01-ES-70153; 85 p. Prepared for NIEHS, NTP, under a Related Services Agreement
with the U.S. Department of Energy under contract DE-AC06-76RLO-1830.
Haley, TJ, Jr. (1978) Chloroprene (2-chloro-1,3-butadiene)Cwhat is the evidence for its carcinogenicity? Clin
Toxicol 13:153-170.
Hallenbeck, W. (1992) Cancer risk assessment for the inhalation of 1,3-butadiene using physiologically based
pharmacokinetic modelling. Bull Environ Contam Toxicol 49:66-70.
Hayes, RB; Xi, L; Bechtel, W; et al. (1996) hprt Mutation frequency among workers exposed to 1,3-butadiene in
China. Toxicology 113:100-105.
Hazleton Laboratories Europe, Ltd. (HLE). (1981) 1,3-Butadiene: inhalation teratogenicity study in the rat (final
report). Harrowgate, England: Hazleton Labs; addendum no. 2788-522/3.
Himmelstein, MW; Turner, MJ; Asgharian, B; et al. (1994) Comparison of blood concentrations of 1,3-butadiene
and butadiene epoxides in mice and rats exposed to 1,3-butadiene by inhalation. Carcinogenesis 15:1479-1486.
Himmelstein, MW; Asgharian, B; Bond, JA. (1995) High concentrations of butadiene epoxides in livers and lungs
of mice compared to rats exposed to 1,3-butadiene. Toxicol Appl Pharmacol 132:281-288.
Himmelstein, MW; Acquavella, JF; Recio, L; et al. (1997) Toxicology and epidemiology of 1,3-butadiene. Crit
Rev Toxicol 27(1):1-108.
Howe, RB; Van Landingham, C. (1986) GLOBAL86: a computer program to extrapolate quantal animal toxicity
data to low doses. EPA Contract No. 68-01-6826. KS Crump and Company, Ruston, LA. Unpublished.
Huff, JE; Melnick, RL; Solleveld, HA; et al. (1985) Multiple organ carcinogenicity of 1,3-butadiene in B6C3F 1
mice after 60 weeks of inhalation exposure. Science 227:548-549.
International Agency for Research on Cancer (IARC). (1982) IARC monographs on the evaluation of carcinogenic
risk of chemicals to humans. Vol. 28. The rubber industry. Lyon, France: International Agency for Research on
Cancer.
International Agency for Research on Cancer (IARC). (1986) IARC monographs on the evaluation of carcinogenic
risk of chemicals to humans. Vol. 39. Some chemicals used in plastics and elastomers butadiene. Lyon, France:
International Agency for Research on Cancer, pp. 155-179.
International Agency for Research on Cancer (IARC). (1992) IARC monographs on the evaluation of carcinogenic
risks to humans. Vol. 54. 1,3-Butadiene. Occupational exposures to mists and vapours from strong inorganic acids
and other industrial chemicals. Lyon, France: International Agency for Research on Cancer, pp. 237-287.
International Institute of Synthetic Rubber Producers (IISRP). (1982) 1,3-Butadiene: inhalation teratogenicity in
the rat (final report with cover letter dated 08/11/82). Report no. 2788-522/3; submission 8EHQ-0382-0441.
Harrowgate, England: Hazleton Laboratories Europe, Ltd.
Irons, RD. (1990) Studies on the mechanism of 1,3-butadiene-induced leukemogenesis: the potential role of
endogenous murine leukemia virus. Environ Health Perspect 86:49-55.
Irons, RD; Smith, CN; Stillman, WS; et al. (1986a) Macrocytic-megaloblastic anemia in male B6C3F 1 Swiss mice
following chronic exposure to 1,3-butadiene. Toxicol Appl Pharmacol 83:95-100.
Irons, RD; Smith, CN; Stillman, WS; et al. (1986b) Macrocytic-megaloblastic anemia in male NIH Swiss mice
following repeated exposure to 1,3-butadiene. Toxicol Appl Pharmacol 85:450-455.
Irons, RD; Stillman, WS; Cloyd, MW. (1987) Selective activation of endogenous ecotropic retrovirus in
hematopoietic tissues of B6C3F1 mice during the preleukemic phase of 1,3-butadiene exposure. Virology 161:457-
462.
Irons, RD; Cathro, HP; Stillman, WS; et al. (1989) Susceptibility to 1,3-butadiene-induced leukemogenesis
correlates with endogenous ecotropic retroviral background in the mouse. Toxicol Appl Pharmacol 101:170-176.
Jacobson-Kram, D; Rosenthal, S. (1995) Molecular and genetic toxicology of 1,3-butadiene. Mutat Res 339:121-
130.
Jelitto, B; Vangala, RR; Laib, RJ. (1989) Species differences in DNA damage by butadiene: role of
diepoxybutane. Arch Toxicol 13 (suppl.):246-249.
Johanson, G; Filser, JG. (1992) Experimental data from closed chamber gas uptake studies in rodents suggest
lower uptake rate of chemical than calculated from literature values on alveolar ventilation. Arch Toxicol 66:291-
295.
Johanson, G; Filser, JG. (1993) A physiologically based pharmacokinetic model for butadiene and its metabolite
butadiene monoxide in rat and mouse and its significance for risk extrapolation. Arch Toxicol 67:151-163.
John, EM; Savitz, DA; Sandler, DP. (1991) Prenatal exposure to parents’ smoking and childhood cancer. Am J
Epidemiol 133:123-132.
Kaplan, EL; Meyer, P. (1958) Nonparametric estimation from incomplete observations. J Am Stat Assoc 53:457-
481.
Kelsey, KT; Wiencke, JK; Ward, J; et al. (1995) Sister-chromatid exchanges, glutathione S-transferase theta
deletion and cytogenetic sensitivity to diepoxybutane in lymphocytes from butadiene monomer production
workers. Mutat Res 335:267-273.
Kirshenbaum, I. (1978) Butadiene. In: Kirk-Othmer encyclopedia of chemical technology, 3rd ed., vol. 4. Mark,
HF; Othmer, DF; Overberger, CG; et al., eds. New York, NY: John Wiley and Sons, pp. 313-337.
Kohn, MC; Melnick, RL. (1993) Species differences in the production and clearance of 1,3-butadiene metabolites:
a mechanistic model indicates predominantly physiological, not biochemical, control. Carcinogenesis 14:619-628.
Kosaric, N; Duvnjak, Z; Farkas, A; et al. (1987) Ethanol. In: Ullmann’s encyclopedia of industrial chemistry, 5th
Rev. Ed., Vol. A9. Gerhartz, W, ed. New York, NY:VCH Publishers, p. 590.
Kraybill, HF. (1980) Evaluation of public health aspects of carcinogenic biorefractories in drinking water. Prev
Med 9:212-218.
Kreiling, R; Laib, RJ; Bolt, HM. (1986a) Alkylation of nuclear proteins and DNA after exposure of rats and mice
to [1,4-14C]1,3-butadiene. Toxicol Lett 30:131-136.
Kreiling, R; Laib, RJ; Filser, JG; et al. (1986b) Species differences in butadiene metabolism between mice and
rats evaluated by inhalation pharmacokinetics. Arch Toxicol 58:235-238.
Kreiling, R; Laib, RJ; Filser, JG; et al. (1987) Inhalation pharmacokinetics of 1,2-epoxybutene-3 reveal species
differences between rats and mice sensitive to butadiene induced carcinogenesis. Arch Toxicol 61:7-11.
Kreiling, R; Laib, RJ; Bolt, HM. (1988) Depletion of hepatic non-protein sulfhydryl content during exposure of
rats and mice to butadiene. Toxicol Lett 41:209-214.
Kreuzer, P; Kessler, W; Welter, HF; et al. (1991) Enzyme specific kinetics of 1,2-epoxybutene-3 in microsomes
and cytosol from livers of mouse, rat, and man. Arch Toxicol 65:59-67.
Krewski, D; Crump, KS; Farmer, J; et al. (1983) A comparison of statistical methods for low dose extrapolation
utilizing time-to-tumor data. Fundam Appl Toxicol 3:140-160.
Laib, RJ; Filser, JG; Kreiling, R; et al. (1990) Inhalation pharmacokinetics of 1,3-butadiene and 1,2-epoxybutene-
3 in rats and mice. Environ Health Perspect 86:57-63.
Laib, RJ; Tucholski, M; Filser, JG; et al. (1992) Pharmacokinetic interaction between 1,3-butadiene and styrene in
Sprague-Dawley rats. Arch Toxicol 66:310-314.
Legator, MS; Au, WW; Ammenheuser, M; et al. (1993) Elevated somatic cell mutant frequencies and altered
DNA repair responses in nonsmoking workers exposed to 1,3-butadiene. In: Butadiene and styrene: assessment of
health hazards. IARC Scientific Publications. Vol. 127. Sorsa, M; Peltonen, K; Vainio, H; et al., eds. Lyon,
France: International Agency for Research on Cancer, pp. 253-263.
Lemen, RA; Meinhardt, TJ; Crandall, MS; et al. (1990) Environmental epidemiologic investigations in the
styrene-butadiene rubber production industry. Environ Health Perspect 86:103-106.
Ligocki, MP. (1993) Review of sources and emission estimates for 1,3-butadiene. SYSAPP-93/074. Prepared by
Systems Applications International, Inc., San Rafael, CA.
Linet, MS. (1985) The Leukemias: epidemiologic aspects. Monographs in epidemiology and biostatistics. Vol. 6.
New York: Oxford University Press, p. 16.
Löfgren, L; Berglund, P; Nordlinder, R; et al. (1991) Selective assessment of C2-Co alkenes in air by adsorption
sampling and gas chromatography. Int J Environ Anal Chem 45:39-44.
Löfroth, G; Burton, RM, Forehand, L; et al. (1989) Characterization of environmental tobacco smoke. Environ Sci
Technol 23:610.
Lyman, WJ; Reehl, WF; Rosenblatt, DH. (1990) Handbook of chemical property estimation methods.
Washington, DC: American Chemical Society.
Macaluso, M; Larson, R; Delzell, E. (1996) Leukemia and cumulative exposure to butadiene, styrene and benzene
among workers in the synthetic rubber industry. Toxicology 113:190-202.
Maldotti, A; Chiorboli, C; Bignozzi, CA; et al. (1980) Photooxidation of 1,3-butadiene containing systems: rate
constant determination for the reaction of acrolein with OH radicals. Int J Chem Kinet 12:905-913.
Malvoisin, E; Lhoest, G; Poncelet, F; et al. (1979a) Identification and quantitation of 1,2-epoxybutene-3 as the
primary metabolite of 1,3-butadiene. J Chromatogr 178:419-425.
Malvoisin, E; Evrard, E; Roberfroid, M; et al. (1979b) Determination of Kováts retention indices with a capillary
column and electron capture detection: application to the assay of the enzymatic conversion of 3,4-epoxy-1-
butene into diepoxybutane. J Chromatogr 179:81.
Malvoisin, E; Mercier, M; Roberfroid, M. (1981) Enzymatic hydration of butadiene monoxide and its importance
in the metabolism of butadiene. Biol Reactive Intermed II, 136A:437-444.
Maniglier-Poulet, C; Cheng, X; Ruth, JA; et al. (1995) Metabolism of 1,3-butadiene to butadiene monoxide in
mouse and human bone marrow cells. Chem-Biol Interact 97:119-129.
Maronpot, RR. (1987) Ovarian toxicity and carcinogenicity in eight recent National Toxicology Program studies.
Environ Health Perspect 73:125-130.
Matanoski, GM; Schwartz, L. (1987) Mortality of workers in styrene-butadiene polymer production. J Occup Med
29:675-680.
Matanoski, GM; Schwartz, L; Sperrazza, J; et al. (1982) Mortality of workers in the styrene-butadiene rubber
polymer manufacturing industry. A report prepared under contract to International Institute of Synthetic Rubber
Producers.
Matanoski, GM; Santos-Burgoa, C; Zeger, SL; et al. (1989) Epidemiologic data related to health effects of
1,3-butadiene. In: Assessment of inhalation hazards. Mohr, U; Bates, DV; Dungworth, DL; et al., eds. New York,
NY: Springer-Verlag, pp. 201-214.
Matanoski, GM; Santos-Burgoa, C; Schwartz, L. (1990) Mortality of a cohort of workers in the styrene-butadiene
polymer manufacturing industry (1943-1982). Environ Health Perspect 86:107-117.
Matanoski, GM; Francis, M; Correa-Villasenor, A. (1993) Cancer epidemiology among styrene-butadiene rubber
workers. In: Butadiene and styrene: assessment of health hazards. Scientific Publications. Vol. 127. Sorsa, M;
Peltonen, K; Vainio, H; et al., eds. Lyon, France: International Agency for Research on Cancer, pp. 363-374.
Mattison, DR; Plowchalk, DR; Meadows, MJ; et al. (1990) Reproductive toxicity: male and female reproductive
systems as targets for chemical injury. Med Clin N Am 74:391-411.
McAlister, RA; Moore, WH; Rice, J; et al. (1989) Nonmethane organic compound monitoring program final
report volume II: urban air toxics monitoring program, 1988. EPA-450/4-89-005. Office of Air Quality Planning
and Standards, U.S. Environmental Protection Agency.
McAlister, RA; Moore, WH; Rice, J; et al. (1990) Urban air toxics monitoring program, 1989. EPA-450/4-91-001.
Research Triangle Park, NC: Office of Air Quality Planning and Standards, U.S. Environmental Protection
Agency.
McAlister, RA; Epperson, DL; Jongleux, RF. (1991) Urban air toxics monitoring program aldehyde results, 1989.
EPA-450/4-91-006. Research Triangle Park, NC: Radian Corp. and Office of Air Quality Planning and Standards,
U.S. Environmental Protection Agency.
McCabe, RW; Siegl, O; Chun, W; et al. (1992) Speciated hydrocarbon emissions from the combustion of single
component fuels. II. Catalyst effects. J Air Waste Manage Assoc 42:1071-1077.
McMichael, AJ; Spirtas, R; Kupper, LL. (1974) An epidemiologic study of mortality within a cohort of rubber
workers, 1964-1972. J Occup Med 16:458-464.
McMichael, AJ; Spirtas, R; Gamble, JF; et al. (1976) Mortality among rubber workers: relationship to specific
jobs. J Occup Med 18:178-185.
Medinsky, MA; Leavens, TL; Csanády, GA; et al. (1994) In vivo metabolism of butadiene by mice and rats: a
comparison of physiological model predictions and experimental data. Carcinogenesis 15:1329-1340.
Meinhardt, TJ; Lemen, RA; Crandall, MS; et al. (1982) Environmental epidemiologic investigations of the
styrene-butadiene rubber industry: mortality patterns with discussion of the hematopoietic and lymphatic
malignancies. Scand J Work Environ Health 8:250-259.
Melnick, RL; Huff, JE. (1992) 1,3-Butadiene: toxicity and carcinogenicity in laboratory animals and in humans.
Rev Environ Contam Toxicol 124:111-144.
Melnick, RL; Huff, JE. (1993) 1,3-Butadiene induces cancer in experimental animals at all concentrations from
6.25 to 8,000 parts per million. In: Butadiene and styrene: assessment of health hazards. IARC Scientific
Melnick, RL; Kohn, MC. (1995) Mechanistic data indicate that 1,3-butadiene is a human carcinogen.
Carcinogenesis 16:157-163.
Melnick, RL; Huff, JE; Chou, BJ; et al. (1990a) Carcinogenicity of 1,3-butadiene in C57BL/6 × C3HF 1 mice at
low exposure concentrations. Cancer Res 50:6592-6599.
Melnick, RL; Huff, JE; Bird, MG; et al. (1990b) Symposium overview: toxicology, carcinogenesis, and human
health aspects of 1,3-butadiene. Environ Health Perspect 86:3-5.
Melnick, RL; Huff, JE; Roycroft, JH; et al. (1990c) Inhalation toxicology and carcinogenicity of 1,3-butadiene in
B6C3F1 mice following 65 weeks of exposure. Environ Health Perspect 86:27-36.
Melnick, RL; Sills, RC; Roycroft, JH; et al. (1994) Isoprene, an endogenous hydrocarbon and industrial chemical,
induces multiple organ neoplasia in rodents after 26 weeks of inhalation exposure. Cancer Res 54:5333-5339.
Meng, Q; Recio, L; Reilly, AA; et al. (1996) The in vivo mutagenic potential of 1,3-butadiene, evaluated at the
hprt locus of T-cells, is greater in mice than in rats exposed concurrently by inhalation. Abstract presented at the
Twelfth Health Effects Institute Annual Conference.
Miller, LM. (1978) Investigations of selected potential environmental contaminants: butadiene and its oligomers.
U.S. Environmental Protection Agency, Washington, DC. EPA-560/2-78-008.
Miller, RA; Boorman, GA. (1990) Morphology of neoplastic lesions by 1,3-butadiene in B6C3F 1 mice. Environ
Health Perspect 86:37-48.
Monson, RR. (1974) Analysis of relative survival and proportional mortality. Comput Biomed Res 7:325-332.
Monson, RR; Nakano, KK. (1976) Mortality among rubber workers. I. White male union employees in Akron,
Ohio. Am J Epidemiol 103:284-296.
Morrow, NL. (1990) The industrial production and use of 1,3-butadiene. Environ Health Perspect 86:7-8.
National Institute for Occupational Safety and Health (NIOSH). (1984) 1,3-Butadiene. Current Intelligence
Bulletin 41, Publication No. 84-105.U.S. Department of Health and Human Services, Cincinnati, OH.
National Institute for Occupational Safety and Health (NIOSH). (1991a) A quantitative assessment of the risk of
cancer associated with exposure to 1,3-butadiene, based on a low dose inhalation study in B6C3F 1 mice.
Submitted to the Occupational Safety and Health Administration (OSHA) for Butadiene Docket (#H-041). U.S.
Department of Health and Human Services, Washington, DC.
National Institute for Occupational Safety and Health (NIOSH). (1991b) Testimony from the National Institute for
Occupational Safety and Health on the Occupational Safety and Health Administration's proposed rule on
occupational exposure to 1,3-butadiene. U.S. Department of Health and Human Services, Washington, DC.
National Toxicology Program (NTP), U.S. Public Health Service, U.S. Department of Health and Human
Services. (1984) Toxicology and carcinogenesis studies of 1,3-butadiene (CAS No. 106-99-0) in B6C3F 1 mice
(inhalation studies). NTP TR 288, NIH Pub. No. 84-2544. Research Triangle Park, NC.
National Toxicology Program (NTP), U.S. Public Health Service, U.S. Department of Health and Human
Services. (1985) Quarterly report from Lovelace Research Institute, January 1 through March 31, 1985.
Interagency agreement 22-Y01-ES-0092. (L. Birnbaum, NTP Project Officer).
National Toxicology Program (NTP), U.S. Public Health Service, U.S. Department of Health and Human
Services. (1989) Toxicology and carcinogenesis studies of 4-vinyl-1-cyclohexene diepoxide (CAS No. 106-87-6)
in F344/N rats and B6C3F1 mice (dermal studies). NTP TR 362, NIH Pub. No. 90-2817. Research Triangle Park,
NC.
National Toxicology Program (NTP), U.S. Public Health Service, U.S. Department of Health and Human
Services. (1993) Toxicology and carcinogenesis studies of 1,3-butadiene (CAS No. 106-99-0) in B6C3F 1 mice
(inhalation studies). NTP TR 434. Research Triangle Park, NC.
Nauhaus, SK; Fennell, TR; Asgharian, B; et al. (1996) Characterization of urinary metabolites from Sprague-
Dawley rats and B6C3F1 mice exposed to [1,2,3,4-C-13]butadiene. Chem Res Technol 9:764-773.
Nordlinder, RG; Nilsson, RI; Buskhe, AB. (1996) 1,3-butadiene in gasoline: an analytical conclusion. J Occup
Environ Med 38:463.
Norppa, H; Sorsa, M. (1993) Genetic toxicology of 1,3-butadiene and styrene. In: Butadiene and styrene:
assessment of health hazards. IARC Scientific Publications. Vol. 127. Sorsa, M; Peltonen, K; Vainio, H; et al.,
eds. Lyon, France: International Agency for Research on Cancer, pp. 185-193.
Norppa, H; Hirvonen, A; Jarventaus, H; et al. (1995) Role of GSTT1 and GSTM1 genotypes in determining
individual sensitivity to sister chromatid exchange induction by diepoxybutane in cultured human lymphocytes.
Carcinogenesis 16:1261-1264.
Osterman-Golkar, SM; Kautianen, A; Bergmark, E; et al. (1991) Hemoglobin adducts and urinary mercapturic
acids in rats as biological indicators of butadiene exposure. Chem Biol Interactions 80:291-302.
Osterman-Golkar, SM; Bond, JA; Ward, JB, Jr; et al. (1993) Use of hemoglobin adducts for biomonitoring
exposure of 1,3-butadiene. In: Butadiene and styrene: assessment of health hazards. IARC Scientific Publications.
Vol. 127. Sorsa, M; Peltonen, K; Vainio, H; et al., eds. Lyon, France: International Agency for Research on
Cancer, pp. 127-134.
Owen, PE; Glaister, JR. (1990) Inhalation toxicity and carcinogenicity of 1,3-butadiene in Sprague-Dawley rats.
Environ Health Perspect 86:19-25.
Owen, PE; Pullinger, DH; Glaister, JR; et al. (1985) 1,3 Butadiene: two-year inhalation toxicity/carcinogenicity
study in the rat (abstract no. P34). In: 26th Congress of the European Society of Toxicology, 16-19 June, Kuopio,
University of Kuopio. Hanhijärvi, H., ed. p. 69.
Owen, PE; Glaister, JR; Gaunt, IF; et al. (1987) Inhalation toxicity studies with 1,3-butadiene. 3. Two-year
toxicity/carcinogenicity study in rats. Am Ind Hyg Assoc J 48(5):407-413.
Percy, C; Stanek, E; Gloeckler L. (1981) Accuracy of cancer death certificates and its effects on cancer mortality
statistics. Am J Public Health 71:242-250.
Peter, H; Wiegand, HJ; Bolt, HM; et al. (1987) Pharmacokinetics of isoprene in mice and rats. Toxicol Lett 36:9-
14.
Portier CJ; Bailer AJ. (1989) Testing for increased carcinogenicity using a survival-adjusted quantal response test.
Fundam Appl Toxicol 12:731-737.
Portier, CJ; Hedges, JC; Hoel, DG. (1986) Age-specific models of mortality and tumor onset for historical control
animals in the National Toxicology Program’s carcinogenicity experiments. Cancer Res 46:4372-4378.
Recio, L; Goldsworthy, TL. (1995) The use of transgenic mice for studying mutagenicity induced by 1,3-
butadiene. Toxicol Lett 82:607-612.
Recio, L; Cochrane, J; Simpson, D; et al. (1990) DNA sequence analysis of in vivo hprt mutation in human T-
lymphocytes. Mutagenesis 5:505-510.
Reynolds, SH; Stowers, SJ; Patterson, RM; et al. (1987) Activated oncogenes in B6C3F1 mouse liver tumors:
implications for risk assessment. Science 237:1309-1316.
Robinson, JP; Wiley, JA; Piazza, T; et al. (1989) Activity patterns of California residents and their implications
for potential exposure to pollution. Draft final report. California Air Resources Board, Sacramento, CA.
CARB-46-177-33.
Rosenthal, SL. (1985) The reproductive effects assessment group's report on the mutagenicity of 1,3-butadiene
and its reactive metabolites. Environ Mutagen 7:933-945.
Rothman, K; Boice, J. (1982) Epidemiologic analysis with a programmable calculator. Boston, MA:
Epidemiology Resources, Inc., pp. 11-17.
Sabourin, PJ; Burka, LT; Bechtold, WE; et al. (1992) Species differences in urinary butadiene metabolites;
identification of 1,2-dihydroxy-4-(N-acetylcysteinyl) butane, a novel metabolite of butadiene. Carcinogenesis
13:1633-1638.
Sandberg, DV; Pickford, SG; Darley, EF. (1975) Emissions from slash burning and the influence of flame
retardant chemicals. J Air Pollut Control Assoc 25:278-281.
Santodonato, J. (1985) Monograph on human exposure to chemicals in the workplace: 1,3-butadiene (PB86-
147261). Washington, DC: U.S. National Technical Information Service.
Sax, NI; Lewis, RJ, Sr. (1987) Hawley’s condensed chemical dictionary, 11th ed. New York, NY: Van Nostrand
Reinhold, p. 177.
Schottenfeld, D; Fraumeni, JF, Jr. (1996) Cancer epidemiology and prevention. 2nd ed. New York: Oxford Univ
Press, p. 856.
Seaton, MJ; Follansbee, MH; Bond, JA. (1995) Oxidation of 1,2-epoxy-3-butene to 1,2:3,4-diepoxybutane by
cDNA expressed human cytochromes P450 2E1 and 3A4 and human, mouse and rat liver microsomes.
Carcinogenesis 16:2287-2293.
Shah, JJ; Heyerdahl, EK. (1988) National ambient volatile organic compounds database update (project report).
U.S. Environmental Protection Agency, Atmospheric Sciences Research Laboratory, Research Triangle Park, NC.
EPA/600/3-88/010(a).
Sharer, JE; Duescher, RJ; Elfarra, AA. (1992) Species and tissue differences in the microsomal oxidation of 1,3-
butadiene and the glutathione conjugation of butadiene monoxide in mice and rats. Drug Metab Dispos 20:658-
664.
Shields, PG; Xu, GX; Blot, WJ; et al. (1995) Mutagens from heated Chinese and U.S. cooking oils. J Natl Cancer
Inst 87:836-841 [abstract from Toxline 95:258350].
Siematycki, J; ed. (1991) Risk factors for cancer in the workplace. Boca Raton, FL: CRC Press.
Sjöblom, T; Lähdetie, J. (1996) Micronuclei are induced in rat spermatids in vitro by 1,2,3,4-diepoxybutane but
not by 1,2-epoxy-3-butene and 1,2-dihydroxy-3,4-epoxybutane. Mutagenesis 11:525-528.
Soderkvist, P; Goodrow, T; Cochran, C; et al. (1992) Characterization of p53 mutations and allelic losses on
chromosome 4, 11, and 14 in butadiene-induced tumors of B6C3F1 mice. J Cell Biochem 16B:138.
Sorsa, M; Autio, K; Demopoulos, NA; et al. (1994) Human cytogenetic biomonitoring of occupational exposure to
1,3-butadiene. Mutat Res 309:321-326.
Sorsa, M; Osterman-Golkar, S; Peltonen, K; et al. (1996) Assessment of exposure to butadiene in the process
industry. Toxicology 113:77-83.
SRI International, 1993. 1993 directory of chemical producers--USA. Menlo Park,CA: SRI International.
Startin, JR; Gilbert, J. (1984) Single ion monitoring of butadiene in plastics and foods by coupled mass
spectrometry-automatic headspace gas chromatography. J Chromatogr 294:427-430.
Stjernfeldt, M; Berglund, K; Lindsten, J; Ludrigsson, J. (1986) Maternal smoking during pregnancy and risk of
childhood cancer. Lancet 1(8494):1350-1352.
Sun, JD; Dahl, AR; Bond, JA; et al. (1989a) Characterization of hemoglobin adduct formation in mice and rats
after administration of [14C]butadiene or [14C]isoprene. Toxicol Appl Pharmacol 100:86-95.
Sun, JD; Dahl, AR; Bond, JA; et al. (1989b) Metabolism of inhaled butadiene to monkeys: comparison to rodents.
Exp Pathol 37:133-135.
Tates, AD; van Dam, FJ; de Zwart, FA; et al. (1996) Biological effect monitoring in industrial workers from the
Czech Republic exposed to low levels of butadiene. Toxicology 113:91-99.
Thier, R; Müeller, M; Taylor, JB; et al. (1995) Enhancement of bacterial mutagenicity of bifunctional alkylating
agents by expression of mammalian glutathione S-transferase. Chem Res Toxicol 8:465.
Thornton-Manning, JR; Dahl, AR; Bechtold, WE; et al. (1995a) Disposition of butadiene monoepoxide and
butadiene diepoxide in various tissues of rats and mice following a low-level inhalation exposure to 1,3-butadiene.
Carcinogenesis 16:1723-1731.
Thornton-Manning, JR; Dahl, AR; Bechtold, WE; et al. (1995b) Gender differences in the metabolism of 1,3-
butadiene in Sprague-Dawley rats following low-level inhalation exposure. Carcinogenesis 16:2875-2878.
Thornton-Manning, JR; Bechtold, WE; Griffith, RF; et al. (1996) Tissue concentrations of butadiene epoxides in
rodents following multiple and single exposures to 1,3-butadiene (BD) by inhalation. (Abstract.) Toxicologist
30:215.
Thurmond, LM; Lauer, LD; House, RV; et al. (1986) Effect of short-term inhalation exposure to 1,3-butadiene on
murine immune functions. Toxicol Appl Pharmacol 86:170-179.
Travis, CC. (1988) Carcinogen risk assessment: contemporary issues in risk analysis. Vol. 3. New York, NY:
Plenum Press.
U.S. Department of Health and Human Services (U.S. DHHS), Public Health Service. (1992) Toxicological
profile for 1,3-butadiene.
U.S. Department of Labor, Occupational Safety and Health Administration (OSHA). (1990, August 10)
Occupational exposure to 1,3-butadiene; proposed rule and notice of hearing. Federal Register 55:32736-32776.
U.S. Department of Labor, Occupational Safety and Health Administration (OSHA). (1996, November 4)
Occupational exposure to 1,3-butadiene; final rule. Federal Register 61(214):56745-56795.
U.S. Environmental Protection Agency. (1978) Interim primary drinking water regulations. Federal Register
43:20135-29150.
U.S. Environmental Protection Agency. (1985) Mutagenicity and carcinogenicity assessment of 1,3-butadiene.
Office of Health and Environmental Assessment, Office of Research and Development, Washington, DC.
EPA/600/8-85/004F.
U.S. Environmental Protection Agency. (1986, Sept. 24) Guidelines for carcinogen risk assessment. Federal
Register 51(185):33992-34003.
U.S. Environmental Protection Agency. (1989a) AIRS user's guide, volumes I-VII. Office of Air Quality Planning
and Standards, Research Triangle Park, NC.
U.S. Environmental Protection Agency. (1989b) 1989 Urban Air Toxics Monitoring Program. Office of Air
Quality Planning and Standards, Research Triangle Park, NC. EPA/450/4-91-001.
U.S. Environmental Protection Agency. (1989c) 1989 Urban Air Toxics Monitoring Program; aldehyde results.
Office of Air Quality Planning and Standards, Research Triangle Park, NC. EPA/450/4-91-006.
U.S. Environmental Protection Agency (1990b) 1990 Urban Air Toxics Monitoring Program carbonyl results.
Office of Air Quality Planning and Standards, Research Triangle Park, NC. EPA/450/4-91-025.
U.S. Environmental Protection Agency. (1991) Non-road engine and vehicle emission study. Office of Mobile
Sources, Ann Arbor, MI. EPA 21A-2001.
U.S. Environmental Protection Agency. (1992) Compilation of air pollutant emission factors (AP-42). Supplement
E, October 1992. U.S. Environmental Protection Agency.
U.S. Environmental Protection Agency. (1993a) Motor vehicle-related air toxics study. Office of Mobile Sources,
Ann Arbor, MI. EPA/420-R-93-005.
U.S. Environmental Protection Agency. (1993b) The Integrated Air Cancer Project, Boise Study; research to
improve risk assessment of area sources, woodstoves and mobile sources. Office of Research and Development.
Presentation to the Office of Mobile Sources, August 1993.
U.S. Environmental Protection Agency. (1994a) Locating and estimating air emissions from sources of 1,3-
butadiene. (September 1994 draft.) Office of Air Quality Planning and Standards, Research Triangle Park, NC.
U.S. Environmental Protection Agency. (1994b) Memo from Rich Cook, Office of Mobile Sources to Dennis
Beauregard, Office of Air Quality Planning and Standards. November 9, 1994.
U.S. Environmental Protection Agency. (1995) The use of the benchmark-dose approach in health risk assessment.
Office of Research and Development, Washington, DC. EPA/630/R-94/007.
U.S. Environmental Protection Agency. (1996, April 23) Proposed guidelines for carcinogen risk assessment.
Federal Register 61(79):17960-18011.
U.S. International Trade Commission (USITC). (1990) Synthetic organic chemicals; United States production and
sales. 1989. Pub. 2338. Washington, DC: Government Printing Office.
U.S. National Library of Medicine. (1991) Toxic Chemical Release Inventory (TRI) Data Bank, Bethesda, MD.
Uuskula, M; Jarventaus, H; Hirvonen, A; et al. (1995) Influence of GSTM1 genotype on sister chromatid
exchange induction by styrene-7,8-oxide and 1,2-epoxy-3-butene in cultured human lymphocytes. Carcinogenesis
16:947-950.
Van Duuren, BL; Nelson, N; Orris, L; et al. (1963) Carcinogenicity of epoxides, lactones, and peroxy compounds.
J Natl Cancer Inst 31:41-55.
Van Duuren, BL; Langseth, L; Orris, L; et al. (1966) Carcinogenicity of epoxides, lactones, and peroxy
compounds. IV. Tumor response in epithelial and connective tissue in mice and rats. J Natl Cancer Inst 37:825-
838.
Verschueren, K. (1983) Handbook of environmental data on organic chemicals, 2nd ed. New York, NY:Van
Nostrand Reinhold, pp. 295-297.
Ward, DE; Hao, WM. (1992) Air toxic emissions from burning of biomass globallyCpreliminary estimates. Paper
92-98.03 Presented at the 85th Annual Meeting of the Air and Waste Management Association, Kansas City, MO.
Ward, EM; Fajen, JM; Ruder, A. (1995) Mortality study of workers in 1,3-butadiene production units identified
from a chemical workers cohort. Environ Health Perspect 103:598-603.
Ward, JB, Jr; Ammenheuser, MM; Bechtold, WE. (1996a) Assessment of butadiene exposure in a rubber plant
using urine metabolite and HPRT mutant frequency as biological markers of exposure and effect. (Abstract.)
Environ Molec Mutag 27(suppl. 27):73.
Ward, JB, Jr.; Ammenheuser, MM; Wharton, EB, Jr.; et al. (1996b) Biological monitoring for mutagenic effects
of occupational exposure to butadiene. Toxicology 113:84-90.
Ward, EM; Fajen, JM; Ruder, A. (1996c) Mortality study of workers employed in 1,3-butadiene production units
identified from a large chemical workers cohort. Toxicology 113:157-168.
Weast, RC, ed. (1989) CRC handbook of chemistry and physics, 70th ed. Boca Raton, FL:CRC Press, p. C-160.
Wijnberg, L; Faoro, R. (1989) Urban air toxics monitoring programCresults of aldehyde monitoring fiscal year
1988. PEI Associates and the U.S. Environmental Protection Agency, Office of Air Quality Planning and
Standards. Research Triangle Park, NC. September 1989.
Wistuba, D; Nowotny, H-P; Trágerf, O; et al. (1989) Cytochrome P-450-catalyzed asymmetric epoxidation of
simple prochiral and chiral aliphatic alkenes: species dependence and effect of enzyme induction of
enantioselective oxirane formation. Chirality 1:127.
Xiao, Y; Tates, AD (1995) Clastogenic effects of 1,3-butadiene and its metabolites 1,2-epoxybutene and 1,2,3,4-
diepoxybutane in splenocytes and germ cells of rats and mice in vivo. Environ Mol Mutagen 26:97-108.