£% FQA United States
Environmental Protection
§ % Agency
EPA/690/R-06/006F
Final
3-01-2006
Provisional Peer Reviewed Toxicity Values for
Mono-, Di- and Tri- Butyltin Compounds
(Various CASRN)
Derivation of Subchronic and Chronic Oral RfDs
Superfund Health Risk Technical Support Center
National Center for Environmental Assessment
Office of Research and Development
U.S. Environmental Protection Agency
Cincinnati, OH 45268

-------
Acronyms and Abbreviations
bw	body weight
cc	cubic centimeters
CD	Caesarean Delivered
CERCLA	Comprehensive Environmental Response, Compensation and Liability Act
of 1980
CNS	central nervous system
cu.m	cubic meter
DWEL	Drinking Water Equivalent Level
FEL	frank-effect level
FIFRA	Federal Insecticide, Fungicide, and Rodenticide Act
g	grams
GI	gastrointestinal
HEC	human equivalent concentration
Hgb	hemoglobin
i.m.	intramuscular
i.p.	intraperitoneal
i.v.	intravenous
IRIS	Integrated Risk Information System
IUR	inhalation unit risk
kg	kilogram
L	liter
LEL	lowest-effect level
LOAEL	lowest-observed-adverse-effect level
LOAEL(ADJ)	LOAEL adjusted to continuous exposure duration
LOAEL(HEC)	LOAEL adjusted for dosimetric differences across species to a human
m	meter
MCL	maximum contaminant level
MCLG	maximum contaminant level goal
MF	modifying factor
mg	milligram
mg/kg	milligrams per kilogram
mg/L	milligrams per liter
MRL	minimal risk level
1

-------
MTD
maximum tolerated dose
MTL
median threshold limit
NAAQS
National Ambient Air Quality Standards
NOAEL
no-observed-adverse-effect level
NOAEL(ADJ)
NOAEL adjusted to continuous exposure duration
NOAEL(HEC)
NOAEL adjusted for dosimetric differences across species to a human
NOEL
no-observed-effect level
OSF
oral slope factor
p-IUR
provisional inhalation unit risk
p-OSF
provisional oral slope factor
p-RfC
provisional inhalation reference concentration
p-RfD
provisional oral reference dose
PBPK
physiologically based pharmacokinetic
PPb
parts per billion
ppm
parts per million
PPRTV
Provisional Peer Reviewed Toxicity Value
RBC
red blood cell(s)
RCRA
Resource Conservation and Recovery Act
RDDR
Regional deposited dose ratio (for the indicated lung region)
REL
relative exposure level
RfC
inhalation reference concentration
RfD
oral reference dose
RGDR
Regional gas dose ratio (for the indicated lung region)
s.c.
subcutaneous
SCE
sister chromatid exchange
SDWA
Safe Drinking Water Act
sq.cm.
square centimeters
TSCA
Toxic Substances Control Act
UF
uncertainty factor

microgram
(.imol
micromoles
voc
volatile organic compound
11

-------
3-1-2006
PROVISIONAL PEER REVIEWED TOXICITY VALUES FOR
MONO-, DI- AND TRI- BUTYLTIN COMPOUNDS (Various CASRN)
Derivation of a Subchronic and Chronic Oral RfD
Background
On December 5, 2003, the U.S. Environmental Protection Agency's (EPA's) Office of
Superfund Remediation and Technology Innovation (OSRTI) revised its hierarchy of human
health toxicity values for Superfund risk assessments, establishing the following three tiers as the
new hierarchy:
1.	EPA's Integrated Risk Information System (IRIS).
2.	Provisional Peer-Reviewed Toxicity Values (PPRTV) used in EPA's Superfund
Program.
3.	Other (peer-reviewed) toxicity values, including:
~	Minimal Risk Levels produced by the Agency for Toxic Substances and Disease
Registry (ATSDR),
~	California Environmental Protection Agency (CalEPA) values, and
~	EPA Health Effects Assessment Summary Table (HEAST) values.
A PPRTV is defined as a toxicity value derived for use in the Superfund Program when
such a value is not available in EPA's Integrated Risk Information System (IRIS). PPRTVs are
developed according to a Standard Operating Procedure (SOP) and are derived after a review of
the relevant scientific literature using the same methods, sources of data, and Agency guidance
for value derivation generally used by the EPA IRIS Program. All provisional toxicity values
receive internal review by two EPA scientists and external peer review by three independently
selected scientific experts. PPRTVs differ from IRIS values in that PPRTVs do not receive the
multi-program consensus review provided for IRIS values. This is because IRIS values are
generally intended to be used in all EPA programs, while PPRTVs are developed specifically for
the Superfund Program.
Because science and available information evolve, PPRTVs are initially derived with a
three-year life-cycle. However, EPA Regions (or the EPA HQ Superfund Program) sometimes
request that a frequently used PPRTV be reassessed. Once an IRIS value for a specific chemical
becomes available for Agency review, the analogous PPRTV for that same chemical is retired. It
should also be noted that some PPRTV manuscripts conclude that a PPRTV cannot be derived
based on inadequate data.
1

-------
3-1-2006
Disclaimers
Users of this document should first check to see if any IRIS values exist for the chemical
of concern before proceeding to use a PPRTV. If no IRIS value is available, staff in the regional
Superfund and RCRA program offices are advised to carefully review the information provided
in this document to ensure that the PPRTVs used are appropriate for the types of exposures and
circumstances at the Superfund site or RCRA facility in question. PPRTVs are periodically
updated; therefore, users should ensure that the values contained in the PPRTV are current at the
time of use.
It is important to remember that a provisional value alone tells very little about the
adverse effects of a chemical or the quality of evidence on which the value is based. Therefore,
users are strongly encouraged to read the entire PPRTV manuscript and understand the strengths
and limitations of the derived provisional values. PPRTVs are developed by the EPA Office of
Research and Development's National Center for Environmental Assessment, Superfund Health
Risk Technical Support Center for OSRTI. Other EPA programs or external parties who may
choose of their own initiative to use these PPRTVs are advised that Superfund resources will not
generally be used to respond to challenges of PPRTVs used in a context outside of the Superfund
Program.
Questions Regarding PPRTVs
Questions regarding the contents of the PPRTVs and their appropriate use (e.g., on
chemicals not covered, or whether chemicals have pending IRIS toxicity values) may be directed
to the EPA Office of Research and Development's National Center for Environmental
Assessment, Superfund Health Risk Technical Support Center (513-569-7300), or OSRTI.
INTRODUCTION
No RfDs for monobutyltin, dibutyltin or tributyltin compounds other than tributyltin
oxide are available on IRIS (U.S. EPA, 2006), the HEAST (U.S. EPA, 1997a) or the Drinking
Water Standards and Health Advisories (U.S. EPA, 2002). An RfD for tributyltin oxide [bis(tri-
n-butyltin)oxide; CASRN 56-35-9] is available on IRIS (U.S. EPA, 1997b, 2006). This RfD of
3E-4 mg/kg-day is based on a NOAEL of 0.025 mg/kg-day and LOAEL of 0.25 mg/kg-day for
immunosuppression in rats exposed via the diet for 18 months (Vos et al., 1990). The RfD was
estimated from a BMDL of 0.03 mg/kg-day for immunosuppression and an uncertainty factor of
100. ATSDR (2003) established a chronic-duration oral MRL of 3E-4 mg/kg-day for tributyltin
oxide based on the NOAEL of 0.025 for immunosuppression in rats (Vos et al.,1990). ATSDR
(2003) also established an intermediate-duration oral MRL of 5E-3 mg/kg-day for dibutyltin
2

-------
3-1-2006
dichloride based on a LOAEL of 5 mg/kg-day for immunological effects in rats exposed via the
diet for six weeks (Seinen et al, 1977a).
The CARA database (U.S. EPA, 1991, 1994) does not list any documents covering
organotin compounds. A NIOSH (1976) Criteria Document for organotin compounds, Poison
Information Monographs on dibutyltin dichloride (WHO, 1993) and tributyl tin compounds
(WHO, 1994), an Environmental Health Criteria document on tributyl tin compounds (WHO,
1990) and a review of the biological activity of organotin compounds (Snoeij et al., 1987) were
consulted for relevant information. In addition, the NTP (2003a) background document for
testing of methyltin and butyltin compounds, the management status documents for monobutyltin
trichloride (NTP, 2003b) and dibutyltin diacetate (NTP, 2003c) and the health and safety report
for dibutyltin acetate (NTP, 2003d) were also consulted. IARC (2003) has not reviewed butyltin
compounds. In addition to the above, several documents on tributyltin oxide, which is not a
subject of this issue paper because it already has an RfD on IRIS, were consulted for relevant
information: the IRIS document (U.S. EPA, 2006) and Toxicological Review (U.S. EPA, 1997b)
and a Concise International Chemical Assessment Document (WHO, 1999).
Computer literature searches of TOXLIT (1965-1992), TOXLINE (1965-1992), CHEM
ID, RTECS (through August, 1992), and TSCATS databases were conducted for monobutyltin
oxide and dibutyltin oxide in August, 1992. An update literature search of TOXLINE,
MEDLINE, EMIC, HSDB, DART, and RTECS was performed in March, 1995 for dibutyltin and
tributyltin compounds. More recently, update literature searches were conducted on
monobutyltin, dibutyltin and tributyltin compounds for the period from 1994 to August 2003 in
the following databases: TOXLINE (including NTIS and BIOSIS updates), CANCERLIT,
MEDLINE, CCRIS, GENETOX, HSDB, EMIC/EMICBACK, DART/ETICBACK, RTECS and
TSCATS. An additional literature search was conducted through September 2004 which
produced no new data.
Monosubstituted organotins have had limited application as stabilizers in PVC films.
Dialkylorganotin compounds such as dibutyltin are used in the chemical industry as heat
stabilizers in the production of PVC, curing agents for silicon rubber, and catalysts in the
production of polyurethane. Tributyltin compounds are used mainly for their biocidal properties
as molluscicides, fungicides, insecticides and miticides. Tetrasubstituted organotin compounds
are mainly used as intermediates in the preparation of other organotin compounds (Boyer, 1989;
Bulten and Meinema, 1991; Magos, 1986; Nicklin and Robson, 1988; NIOSH, 1976; WHO,
1980).
Toxicity of organotin compounds is somewhat determined by the nature and number of
groups bound to tin (Bulten and Meinema, 1991). In general, toxicity decreases as the number of
linear carbons increases, such that triethyltin is more toxic than trioctyltin (Magos, 1986; NIOSH,
3

-------
3-1-2006
1976). Also, toxicity decreases as the number of substitutions decrease, for example, triethyltin
chloride is more toxic than monoethyltin trichloride (Magos, 1986; WHO, 1980).
Table 1 gives the butyltin compound molecular weights and associated factors for
converting exposures (mg/kg-day) from the administered compound to the relevant butyltin
moiety on which each RfD is based.
Table 1. Molecular weights and dose-conversion factors for butyltin compounds
Compound
MW
moiety
conversion
factora
fraction
Snb
butyltin moiety
175.74
-
0.675
monobutyltin trichloride
282.10
0.623
0.421
dibutyltin moiety
232.79
-
0.510
dibutyltin dichloride
303.70
0.767
0.391
dibutyltin oxide
248.79
0.936
0.477
dibutyltin diacetate
350.83
0.664
0.339
tributyltin moiety
289.83
-
0.410
tributyltin chloride
325.28
0.891
0.362
tributyltin oxide
305.83
0.948
0.388
a MW of corresponding moiety divided by MW of compound
b for conversion to or from tin mass (dose expressed in terms of Sn in some studies)
REVIEW OF THE PERTINENT LITERATURE
Human Studies
No information was located regarding the effects of butyltin compounds in humans
following oral exposure. Early reports summarized by WHO (1980) and Boyer (1989) provide
information regarding the dermal effects of some of these compounds. Dermal lesions caused by
4

-------
3-1-2006
di- and tributyltin compounds in laboratory workers and process workers handling these
compounds were described as typical acute skin burns developing 1 to 8 hours after exposure.
This could be prevented by washing the skin immediately after exposure. A single dermal
application of undiluted dibutyltin maleate, dibutyltin oxide, dibutyltin dilaurate, dibutyltin
diacetate, or tetrabutyltin on the back of the hands of volunteers did not cause irritation.
However, within 8 hours of a single application of dibutyltin dichloride, tributyltin chloride,
tributyltin acetate, tributyltin laurate, or bis(tributyltin) oxide, the exposed area of the skin
reddened. Follicular inflammation and pruritus accompanied this effect. Irritant contact
dermatitis was reported in painters that came in contact with a paint containing 0.6%
bis(tributyltin)oxide. The effect was characterized by severe itching, redness, swelling and
blistering, and confined to the areas of the skin directly exposed to the paint.
Animal Studies
Monobutyltin Compounds
Information on the oral toxicity of monobutyltin compounds in animals comes from
developmental toxicity studies in rats exposed by gavage to monobutyltin trichloride (CASRN
1118-46-3) (Noda et al., 1992a; Ema et al., 1995a; Ema and Harazono, 2001); synonyms for this
compound are butyltin trichloride and n-butyltin trichloride. No subchronic or chronic duration
studies were located.
Noda et al. (1992a) exposed groups of 13-16 pregnant Wistar rats to 0, 50, 100, 200, or
400 mg/kg-day monobutyltin trichloride by gavage in olive oil on gestational days 7-17. These
doses are equivalent to 30.2, 60.3, 120.6 and 241.2 mg monobutyltin/kg-day, respectively. Body
weights, food consumption and clinical signs were recorded daily. The dams were killed on
gestational day 20. Endpoints examined at termination included the weight of the maternal
thymus, the number of corpora lutea, the numbers of living and dead fetuses, the number of
resorptions, fetal weight, fetal sex, and external malformations; half of the fetuses in each litter
were examined for skeletal and half for visceral anomalies. No significant alterations in maternal
body weight gain, food consumption or thymus weights were observed. In the offspring, no
consistent alterations in the incidence of external, visceral, or skeletal alterations were observed.
This study identifies a NOAEL of 400 mg/kg-day for developmental or maternal effects of
monobutyltin trichloride in rats (equivalent to 248 mg monbutyltin/kg-day); a LOAEL was not
identified.
Higher doses were tested by Ema et al. (1995a). In this study, groups of 6-11 pregnant
female Wistar rats were given monobutyltin trichloride at doses of 0, 1000, 1500 or 2000 mg/kg-
day by gavage in olive oil on gestational days (GD) 7 and 8. Maternal body weight was
monitored up to GD 20; results were reported for the periods GD 0-7, 7-9, 9-20 and 0-20. At
termination on GD 20, uteri were examined for the numbers of resorptions and dead and live
5

-------
3-1-2006
fetuses. All live fetuses were sexed, weighed and examined for external malformations,
including those of the oral cavity; two thirds of the fetuses were examined for skeletal
malformations and the rest for visceral malformations. Maternal mortality was significantly
increased at >1500 mg/kg-day (0/10, 0/10, 5/11 and 6/6 for the control and low-to-high dose
groups, respectively). Most deaths occurred within two days of administration; necropsy of all
dead rats revealed hemorrhages in the stomach. A loss in maternal body weight was observed in
all treated groups for the period GD 7-9, which includes the two days of treatment; body weight
gain was significantly reduced in these groups for the remainder of the study. Total litter
resorption occurred in 1/6 litters at 1500 mg/kg-day. Also at 1500 mg/kg, fetal body weights
were significantly reduced (male and female). The incidence of malformations was not increased
at any dose. This study identifies a LOAEL for monobutyltin trichloride of 1000 mg/kg
(equivalent to 620 mg monobutyltin/kg-day) for maternal toxicity (reduced body weight on days
of treatment and reduced body weight gain overall); a maternal NOAEL was not identified. The
NOAEL for fetal toxicity of butyltin trichloride administered to rats on GD 7 and 8 was 1000
mg/kg-day and the LOAEL was 1500 mg/kg-day (equivalent to 930 mg monobutyltin/kg-day) for
reduced body weight in male and female offspring.
Ema and Harazono (2001) evaluated the effect of monobutyltin trichloride (purity 95%)
administered by gavage in olive oil on early pregnancy in rats. Groups of 16 pregnant female
Wistar rats received butyltin trichloride at doses of 0, 56, 226, or 903 mg/kg-day on gestational
days (GD) 0-3 or 4-7. Maternal body weight and food consumption were recorded daily; results
for these parameters were reported for the periods GD 0-4 and GD 4-20 for dams treated on GD
0-3 and for the periods GD 0-4, GD 4-8 and GD 8-20 for dams treated on GD 4-7. Dams were
sacrificed on GD 20 and examined for corpora lutea and the numbers of resorptions and dead and
live fetuses in the uterus. Live fetuses were examined for sex, weight and external
malformations. Similar results were observed for the two treatment schedules. No maternal
mortality or other clinical sign of toxicity was observed. In dams that received 903 mg/kg-day,
food consumption and body weight gain were significantly reduced only for the reporting period
that included days of treatment, that is, GD 0-4 for dams treated on GD 0-3, and GD 4-8 for
dams treated on GD 4-7; in the latter group a slight body weight loss was observed. The only
treatment-related developmental effect was a significantly lower body weight in female fetuses at
the 903 mg/kg-day dose on either schedule. This study identifies a NOAEL of 226 mg/kg-day
and a LOAEL of 903 mg/kg-day for maternal effects (reduced food consumption and body
weight gain) and fetal toxicity (reduced female body weight) for butyltin trichloride administered
to rats on GD 0-3 or GD 4-7. The maternal and fetal NOAELs and LOAELs are equivalent to
140 and 560 mg monobutyltin/kg-day.
Dibutyltin Compounds
Several studies have examined the toxicity of dibutyltin compounds in rodents following
subchronic (Seinen et al., 1977a,b; Bartalini, 1959; Barnes and Stoner, 1958; Gaunt et al., 1968)
6

-------
3-1-2006
and chronic (NCI, 1979) administration. Noda et al. (1992a, 1992b, 1993) and Ema et al. (1991,
1992) have studied the developmental toxicity of several dibutyltin compounds in rats.
Seinen et al. (1977a) administered dibutyltin dichloride (CASRN 683-18-1; purity >98%)
in the diet at levels of 0, 50, or 150 ppm to weanling Wistar-derived rats (10/sex/dose level) for 2
weeks. The dietary levels correspond to 0, 5, and 15 mg/kg-day, respectively, using the standard
food consumption rate of 10% of body weight per day for young rats. At termination, animals
were necropsied. Body weights and the weights of the thymus, spleen, popliteal lymph node,
liver, kidneys, and adrenals were recorded; the same organs were examined for histopathology.
Six rats in the 150 ppm group died during the second week. Significant and dose-related changes
in males and females included decreased body weight and decreased relative weights of spleen,
thymus, and popliteal lymph nodes. Relative liver weight was increased in 150 ppm males only.
Necropsy revealed a marked reduction in thymus size in all treated rats. In addition, rats that
died early and two male and two female survivors in the 150 ppm group had thickened and
dilated bile ducts and yellowish discolored livers. No liver histopathology was seen at 50 ppm,
but severe proliferation of bile duct epithelial cells and bile ductules, associated with
pericholangiolitis and periportal fibrosis was observed in 150 ppm rats. Depletion of
lymphocytes in the thymic cortex was observed at 50 ppm and was nearly complete at 150 ppm;
lymphocyte depletion was also observed in thymus-dependent areas of the spleen (periarteriolar
lymphocyte sheets) and popliteal lymph node (paracortical areas). This study identifies a
LOAEL of 50 ppm for immunotoxicity (depletion of thymic cortex lymphocytes) in rats exposed
to di-n-butyltin dichloride for two weeks (equivalent to 3.84 mg dibutyltin/kg-day), with
hepatotoxicity at higher doses. A NOAEL was not identified.
Seinen et al. (1977b) conducted a series of experiments evaluating immune system
functions in weanling Wistar rats (groups of 5-10 of one sex) fed dibutyltin dichloride in the diet
at concentrations of 0, 50, or 150 ppm for 4-6 weeks. Using the standard food consupmtion
factor of 10% of body weight per day for young rats, these dietary concentrations correspond to
approximate doses of 0, 5, and 15 mg/kg-day. Thymus-dependent cellular immunity was
evaluated in an allograft rejection experiment; two donor tail-skin grafts (from Wistar-derived
WAG xBFj hybrid rats) were transferred to groups of 5-9 seven-week-old inbred male WAG
rats that were dietarily exposed to dibutyltin dichloride for 6 weeks (after weaning at week 3 to
week 9). Terminal body weights were significantly reduced (by 28.5% compared to controls)
and allograft (i.e., from other animals) rejection times were significantly increased by 1.3 days,
indicating impaired thymus-dependent cellular immunity, in the 150 ppm group; dose-related
changes were also observed in the 50 ppm group, but were not statistically significant. A second
experiment evaluated thymus-dependent humoral immunity; groups of 7-8 female rats were
immunized with sheep red blood cells five days before the end of a 4-week dietary exposure. In
these rats, a statistically significant decrease in body weight gain (-6.6% compared to controls)
was observed in the 150 ppm group. Statistically significant immunological effects in females
included reductions in the number of spleen cells and the number of antibody-producing cells per
7

-------
3-1-2006
whole spleen in the 50 and 150 ppm groups and in the hemagglutination titer in the 150 ppm
group. A third experiment evaluated thymus-independent humoral immunity; groups of 10 male
rats were immunized i.v. with E. coli 38 days after dietary exposure. No significant alterations in
hemagglutinating antibody titers were observed; however, 4/10 rats in the 150 ppm group died
(authors noted that deaths were probably the result of endotoxin shock). Considering the three
experiments, the most sensitive LOAEL for dibutyltin dichloride was 50 ppm (equivalent to 3.84
mg dibutyltin/kg-day) for reduced thymus-dependent humoral immunity (reduction in the
numbers of spleen cells and antibody-producing cells per spleen) in female rats exposed in the
diet for 4 weeks. A NOAEL was not identified.
Seinen et al. (1977b) also examined the allograft rejection process in groups of 5-10
neonatal Wistar rat pups that received gavage doses of 0, 1, or 3 mg/kg-day dibutyltin dichloride
in arachis oil three times per week, for nine weeks, beginning the second day after birth.
Significant dose-related decreases in terminal body weights (10-12% lower than controls) were
observed in the pups treated with 1 or 3 mg/kg-day dibutyltin dichloride. Allograft rejection
times were significantly prolonged by 0.6 and 2.6 days, respectively, in the 1 and 3 mg/kg-day
groups. It is conceivable that a generalized stress in these animals, as evidenced by the body
weight loss, contributed to the immunotoxicity. However, a direct effect of dibutyltin on
immune suppression is highly likely given its well established effect on cell-mediated immune
response. The lowest gavage dose of 1 mg/kg-day dibutyltin dichloride, administered 3
days/week, was a LOAEL for reduced body weight and suppressed thymus-dependent cellular
immunity (delayed allograft rejection) in the pups exposed for nine weeks after birth; averaged
over a week, the LOAEL for dibutyltin dichloride during the postnatal period was 0.43 mg/kg-
day (equivalent to a daily dose of 0.33 mg dibutyltin/kg-day). The study did not identify a
NOAEL.
Bartalini (1959) orally administered 2.5 mg dibutyltin oxide/kg-day to 7 rats for 60 days
(the study was published in Italian with an English summary). Weight gain was not affected by
treatment, and the animals showed no signs of toxicity throughout the study. Hematologic
testing at sacrifice revealed a slight increase in red blood cell count. Histological examination
was limited to the liver and kidneys. Liver changes were described as mild, and consisted of
signs of nuclear hypertrophy and cytoplasmic vacuolation. Changes in the kidneys were
restricted to the renal tubules and were indicative of a degenerative process followed by signs of
regeneration. The lack of comparison of the incidences of hepatic and renal lesions to a control
group precludes identifying a NOAEL or LOAEL for this study.
Gaunt et al. (1968) fed groups of CFE rats (16/sex/group) diets containing 0, 10, 20, 40,
or 80 ppm dibutyltin dichloride for 90 days. From data on body weight and food consumption,
Gaunt et al. (1968) estimated that these dietary levels provided an average of 0, 0.5, 1.0, 2.0, or
4.0 mg dibutyltin dichloride/kg-day. Endpoints examined included body weight and food
consumption, hematological parameters, clinical chemistry (AST, ALT, amylase), urinalysis, and
8

-------
3-1-2006
gross (with special reference to the bile duct and pancreas) and microscopic examination of
major organs and tissues. Effects were limited to a slight reduction in body weight gain at 4.0
mg/kg-day (<9%), and a slight but significant decrease in blood hemoglobin concentration at 4.0
mg/kg-day in females at week 6 and in males at week 13. Since the observed changes were
minor, it appears that the 4.0 mg/kg-day dibutyltin dichloride dose (equivalent to 3 mg
dibutyltin/kg-day) represents a NOAEL in this study.
In a study conducted by Barnes and Stoner (1958), albino rats (6/group, sex distribution
not specified) were administered dibutyltin dichloride in the diet at levels of 0, 20, 50, 75, or 100
ppm for up to 6 months. These levels correspond to 0, 1.0, 2.5, 3.8, or 5 mg/kg-day dibutyltin
dichloride, using a food consumption factor of 0.05 for adult rats. The results are described with
few details and can be summarized as follows: doses of 50 ppm or greater significantly reduced
weight gain and food intake. After 6 months of treatment, 7 out of 10 rats in the 50 ppm group
had bile duct damage characterized by thickening and dilation of the duct, and fibrosis of the
pancreas. Mortality was reported in the 100 ppm group during the first 4 weeks, but survivors
exhibited no clinical signs of toxicity. At sacrifice (6 months), all the surviving rats in the 100
ppm group had some bile duct damage. The LOAEL for body weight and bile duct changes is 50
ppm dibutyltin dichloride and the NOAEL is 20 ppm (equivalent to 1.9 and 0.75 mg
dibutyltin/kg-day, respectively).
NCI (1979) conducted chronic feeding bioassays with dibutyltin diacetate in rodents.
Fischer 344 rats (50/sex/group) were given a basal diet containing 66.5 or 133 ppm time-
weighted average dietary levels of dibutyltin diacetate for 78 weeks followed by a 26-week
observation period on the basal diet; control rats (20/sex) received the basal diet throughout the
study. Based on a food consumption factor of 0.05, it can be estimated that the rats were dosed
with 3.3 or 6.7 mg/kg-day. Animals were inspected twice daily for clinical signs of toxicity.
Body weights were recorded weekly for the first six weeks, every two weeks for the next twelve
weeks and at monthly intervals thereafter. Food consumption data were collected only at
monthly intervals from 20 percent of the animals in each group. All animals found dead or
euthanized prematurely or killed at 104 weeks were necropsied; microscopic examinations were
conducted on gross lesions and all major tissues and organs. Most tissue samples were lost for
17/50 high-dose female rats. Survival at 104 weeks was 85, 78, and 52% for males, and 74, 84,
and 64% for females in the control, low- and high-dose groups, respectively; the decrease was
statistically significant for high-dose males. Dose-related depression of body weight gain
compared to controls was observed in male rats throughout the experiment; mean body weights
were lower in high-dose female rats for most of the study, but not significantly. No other clinical
signs were recorded. Calculi of the bile ducts were observed in 0/20 control, 0/42 low-dose and
10/50 high-dose males, and in 0/19 control, 1/49 low-dose and 3/33 high-dose females. There
was a significant dose-related increase in the incidence of pneumonia in male rats, possibly
suggesting a compromised immune system. However, no histological alterations were observed
in the spleen, thymus or lymph nodes. The average low dose of 3.3 mg/kg-day (equivalent to 2.3
9

-------
3-1-2006
mg dibutyltin/kg-day) is aNOAEL and 6.7 mg/kg-day (equivalent to 4.4 mg dibutyltin/kg-day) is
an FEL for accelerated mortality, decreased body weight gain and increased bile duct calculi in
males rats.
B6C3F1 mice (50/sex/species/group) were exposed to 76 or 152 ppm time-weighted
average dietary levels of dibutyltin diacetate for 78 weeks followed by a 26-week observation
period; control mice (20/sex) received the basal diet throughout the study (NCI, 1979). Based on
a food consumption factor of 0.1 for adult mice, it can be estimated the male mice were dosed
with 7.6 or 15.2 mg/kg-day. Mice were analyzed as described above for rats. Survival was 95,
96, and 86% for male mice and 95, 90, and 58% for female mice in the control, low-, and high-
dose groups, respectively. The decreased survival in high-dose female mice was statistically
significant. Body weight gain was suppressed in high-dose female mice after week 60, but not in
other groups. No other clinical signs were recorded. Degenerative and necrotic changes in the
liver were absent in controls and were dose-related in treated mice; however, the incidence of
these lesions was low, less than 10% at the high dose. The average low dose of 7.6 mg/kg-day
(equivalent to 5.0 mg dibutyltin/kg-day) is aNOAEL and 15.2 mg/kg-day (equivalent to 10.1 mg
dibutyltin/kg-day) is a FEL for accelerated mortality and reduced body weight gain in female
mice exposed to dibutyltin diacetate.
Seinen et al. (1977a,b) also conducted dietary assays in mice. Weanling male Swiss mice
(8-10 per group) were exposed for four weeks to 0, 50, or 150 ppm dietary concentrations of
dibutyltin dichloride (0, 7.5, or 23 mg/kg-day, estimated using a food consumption factor of 0.15
for young mice). Body weights and the weights of the thymus, spleen, and liver were recorded.
No clinical signs, body weight effects, or effects on thymus or spleen weights were observed in
treated mice. No significant alteration in the antibody response to sheep red blood cells was
observed. Thus, the NOAEL for immunotoxicity of dibutyltin dichloride is 23 mg/kg-day in
mice exposed for 4 weeks (equivalent to 17.6 mg dibutyltin/kg-day).
A number of developmental toxicity studies have been conducted in rats exposed to
dibutyltin compounds. Studies using standard exposure periods - on or close to gestational days
6-15 (GD 6-15) - have been conducted in rats exposed by gavage to dibutyltin diacetate (Noda et
al., 1992a) or dibutyltin dichloride (Ema et al., 1991; Farr et al., 2001). Follow-up experiments
were conducted to identify vulnerable periods during gestation for exposure to dibutyltin
diacetate (Noda et al., 1992b) or dibutyltin dichloride (Ema et al., 1992, 1995a, 1996; Ema and
Harazono, 2000). Noda et al. (1993) compared the developmental toxicity of several dibutyltin
compounds administered at a selected dibutyltin dose on GD 8. Seinen et al. (1977a) evaluated
immunotoxicity in rat pups exposed to dibutyltin dichloride during gestation and lactation and
subsequently for several weeks via the diet. As presented in detail below, the standard
developmental toxicity studies conducted by Noda et al. (1992a) and Farr et al. (2001) provide
evidence that teratogenicity of dibutyltin, primarily external and skeletal malformations, occurs
only at doses that are toxic to dams. Conversely, Ema et al. (1991) reported malformations at the
10

-------
3-1-2006
maternal no-effect level; however, as this study did not investigate changes in the maternal
thymus, the maternal no-effect level may have been overestimated. The Noda et al. (1992a)
study identifies a NOAEL and LOAEL of 1.7 and 5.0 mg/kg-day for dibutyltin diacetate, and the
Ema et al. (1991) study identifies a NOAEL and LOAEL of 2.5 and 5.0 mg/kg-day for dibutyltin
dichloride.
Noda et al. (1992a) administered by gavage 0, 1.7, 5.0, 10.0, or 15.0 mg/kg-day di-n-
butyltin diacetate in olive oil to groups of 13-16 pregnant Wistar rats on gestational days 7-17.
The dams were killed on gestational day 20. A significant decrease in maternal body weight was
observed in the 15.0 mg/kg-day group (without a significant alteration in food intake) and
decreased thymus weights were observed in the 5.0, 10.0, and 15.0 mg/kg-day groups. In the 15
mg/kg-day group, a significant decrease in the number of dams with living fetuses, an increase in
the number of dams with total resorptions, and an increase in the incidence of dead or resorbed
fetuses in the early stage were observed. Decreases in male and female fetal body weights were
observed in the 10.0 and 15.0 mg/kg-day groups. Significant increases in the number of fetuses
and litters with skeletal malformations (primarily anomaly of mandibular fixation, fused ribs and
fused thoracic vertebral arches) and external malformations (primarily cleft mandible, cleft lower
lip, ankyloglossia and/or schistoglossia, and anury or vestigial tail) were observed in the 10.0 and
15.0 mg/kg-day groups. In the 5.0, 10.0, and 15.0 mg/kg-day groups, dose-related significant
increases in the number of fetuses and litters with skeletal variations (primarily cervical ribs)
were observed. No visceral alterations were observed. Thus, this study identifies a NOAEL of
1.7 mg/kg-day and LOAEL of 5.0 mg/kg-day for developmental effects and maternal toxicity in
rats exposed to di-n-butyltin diacetate (equivalent to 1.1 and 3.3 mg dibutyltin/kg-day,
respectively).
Follow-up studies by these researchers showed that the critical period of exposure for
production of these effects by dibutyltin diacetate was early in organogenesis (GD7-9) (Noda et
al., 1992b) and that similar effects were produced by equimolar doses of other dibutyltin
compounds (dibutyltin maleate, dibutyltin dilaurate, dibutyltin oxide, and dibutyltin dichloride)
(Noda et al., 1993).
Dibutyltin dichloride was used in another series of studies. Ema et al. (1991)
administered by gavage 0, 2.5, 5.0, 7.5, or 10.0 mg/kg-day dibutyltin dichloride in olive oil on
gestational days 7-15 to groups of 12 pregnant Wistar rats. In the 7.5 and 10.0 mg/kg-day
groups, significant increases in maternal deaths, decreases in body weight gain, and decreases in
food consumption were observed. The mean time to death was 8 and 6 days after the start of
dosing in the 7.5 and 10.0 mg/kg-day groups, respectively. The cause of death was not reported,
but it was noted that hemorrhage in the stomach was observed in most of the dams dying early.
Significant increases in the number of resorbed or dead fetuses per litter and the number of post-
implantation losses per litter were observed in the 7.5 mg/kg-day group. Decreases in male and
female fetal body weights and placental weights were observed in the 5.0, 7.5, and 10.0 mg/kg-
11

-------
3-1-2006
day groups. An increase in the number of litters with external malformations (primarily cleft
mandible and ankyloglossia) and skeletal malformations (primarily mandibular defects, fusion
and/or absence of cervical vertebral arches, and fusion of ribs) were observed in the 5.0, 7.5, and
10.0 mg/kg-day offspring. No significant alterations in the incidence of visceral malformation
were observed in these groups. No external, skeletal, or visceral malformations were observed in
the control or 2.5 mg/kg-day groups. This study identifies a NOAEL of 2.5 mg/kg-day and
LOAEL of 5.0 mg/kg-day (equivalent to 1.9 and 3.8 mg dibutyltin/kg-day) for developmental
effects and a NOAEL of 5.0 mg/kg-day and FEL of 7.5 mg/kg-day (equivalent to 3.8 and 5.7 mg
dibutyltin/kg-day) for maternal effects following administration of dibutyltin dichloride on GD 7-
15.
Other studies by these researchers investigated the effect of dosing with dibutyltin
dichloride at different times during gestation. Early organogenesis (GD7-8) was identified as the
most sensitive period for induction of post-implantation loss and teratogenesis (Ema et al., 1992,
1995a, 1996). Exposure earlier in gestation produced an increase in preimplantation loss (Ema
and Harazono, 2000).
Farr et al. (2001) also evaluated developmental toxicity of dibutyltin dichloride
administered to pregnant female Wistar rats (25/group) by gavage in olive oil at doses of 0, 1,
2.5, 5 or 10 mg/kg-day on GD 6-15. Maternal endpoints included food consumption and body
weight gain (both reported for the period GD 6-16), thymus weight, number of pregnant females,
number of females with total litter loss and the number of females with viable fetuses.
Pregnancy outcomes were evaluated on GD 20 for corpora lutea, implantations, viable fetuses,
early and late resorptions, dead fetuses, and fetal weight; all fetuses were examined for external
malformations and then equally divided into two groups for examination of visceral or skeletal
malformations. The only maternal effects were statistically significant reductions in food
consumption, body weight gain, and thymus weight following treatment at 10 mg/kg-day. No
statistically significant effects were noted in litter/fetal parameters although, at 10 mg/kg-day,
there was a slight increase in the number of single defects/malformations (4 affected fetuses/262
from 3/20 litters vs 1/269 control fetuses). The investigators considered one malformation,
ankyloglossia, to be significant because of its rarity and because other studies on dibutyltin
dichloride had reported increased incidences of this defect at higher doses. This study identifies
a NOAEL of 5 mg/kg-day and a LOAEL of 10 mg/kg-day (equivalent to 3.8 and 7.7 mg
dibutyltin/kg-day, respectively) for maternal toxicity (reductions in food consumption, body
weight gain, and thymus weight) and teratogenicity (slightly increased incidence of defects,
including one instance of ankyloglossia) in rats exposed to dibutyltin dichloride.
Seinen et al. (1977a) evaluated immune function in rats exposed pre- and post-natally to
dibutyltin dichloride (purity >98%) by dietary exposure. Groups of 5-6 pregnant Wistar rats
were fed diets containing 0, 50, or 150 ppm dibutyltin dichloride throughout pregnancy (starting
on gestational day 2) and lactation; after weaning, exposure of offspring continued through the
12

-------
3-1-2006
diet up to postnatal day 39. Using the U.S. EPA (1988) reference values for body weights and
food consumption in Wistar rats and the reported body weights for offspring at termination, the
calculated doses for dams during gestation were 0, 5 and 15 mg/kg-day and the doses for
offspring were 0, 5 or 16 mg/kg-day. In the exposed offspring, significant decreases were
observed in body weight (in males at >50 ppm and females at 150 ppm), the number of spleen
cells, and the number of antibody-producing cells per 1 million spleen cells (in males at 150 ppm
and females at >50 ppm) and per whole spleen. A significant decrease in hemagglutination titer
was also observed in both sexes at 150 ppm. The 50 ppm dietary level (equivalent to 5 mg/kg-
day dibutyltin dichloride or 3.8 mg dibutyltin/kg-day) was a LOAEL for suppressed thymus-
dependent humoral immunity in rats exposed both prenatally and postnatally (via milk and diet)
to dibutyltin dichloride. A NOAEL was not identified.
Tributyltin Compounds
Oral (dietary) toxicity studies for tributyltin compounds other than tributyltin oxide have
been conducted in rats: a subchronic study on tributyltin acetate (Barnes and Stoner, 1958), and
short-term immunotoxicity assays (Bressa et al., 1991; Snoeij et al., 1985), a two-generation
reproductive toxicity assay (Ogata et al., 2001; Omura et al., 2001), and developmental toxicity
assays on tributyltin chloride (Ema et al., 1995a, 1995b, 1996; Harazono et al., 1996).
In a study conducted by Snoeij et al. (1985), groups of 10 male weanling Wistar rats were
fed a diet containing 0, 15, 50, or 150 ppm tributyltin chloride (purity >98%) in the diet for 2
weeks. These dietary concentrations correspond to doses of approximately 0, 1.5, 5, or 15
mg/kg-day using a food intake factor of 0.1 for young rats. Body and brain weights were
significantly decreased at 50 ppm; in this group food intake was reduced by about 25%. Relative
spleen weight was significantly reduced at >2 mg/kg-day, and relative thymus weight was
significantly reduced at >7 mg/kg-day (61% reduction at 21 mg/kg-day). Relative liver weight
was increased at >7 mg/kg-day. Severe and dose-related (>7 mg/kg-day) lymphocyte depletion
was observed in cell suspensions prepared from thymus glands. The spleen showed no
lymphocyte depletion or extramedullary hematopoiesis. There was a dose-related (>7 mg/kg-
day) decrease in the amount of iron per spleen, but the iron concentration was not altered. A
dose-related increase in the number of erythrocytes situated as rosettes around mononuclear cells
was observed in the splenic medullary sinuses. No morphological changes were noticed in the
livers. Snoeji et al. (1985) also exposed a group of 6 male weanling rats to 0 or 100 ppm
tributyltin trichloride (0 or 10 mg/kg-day, calculated in same manner as 2 week study) in the diet
for 4 weeks. A decrease in body weight and a decrease in relative thymus weight were observed.
A severe reduction in lymphocyte density was observed in the thymic cortex. The authors noted
that lymphocyte depletion was more severe in the rats treated for 4 weeks as compared to those
treated for 2 weeks. After 4 weeks on the tributyltin chloride diets, the rats were fed the basal
diet for 1-8 weeks. Thymus weight was similar to control values after 1 week, although body
weights remained depressed for 3 weeks. When 100 ppm tributyltin chloride was fed to
13

-------
3-1-2006
adrenalectomized rats for 13 days, thymic atrophy was observed, suggesting that this effect was
not due to stress. The lowest dietary level of 15 ppm in the 2-week study, equivalent to a dose
of 1.5 mg/kg-day for tributyltin chloride (equivalent to 1.3 mg tributyltin/kg-day), was a LOAEL
for reduced relative spleen weight. A NOAEL was not identified.
Bressa et al. (1991) examined the immunotoxic effects of bis(tributyltin)oxide (TBTO)
and tributyltin chloride in rats. Groups of 4 male Wistar rats (0.190-0.200 kg) were fed diets
containing 5 ppm pure bis(tributyltin)oxide, commercial bis(tributyltin)oxide (80% pure), or
tributyltin chloride; groups of 8 rats were fed 25 ppm of each chemical, and a group of 8 animals
served as control. Treatment lasted 4 weeks. From the average tin consumption, provided by the
authors, it can be estimated that the 5 and 25 ppm dietary levels of pure oxide provided doses of
0.432 and 1.46 mg bis(tributyltin) oxide/kg-day, respectively; the 5 and 25 ppm commercial
oxide provided doses of 0.332 and 1.74 mg bis(tributyltin)oxide/kg-day; and, the 5 and 25 ppm
salt provided doses of 0.403 and 1.70 mg tributyltin chloride. In terms of tributyltin, alone, these
doses correspond to 0.410 and 1.39 mg/kg-day for the 5 ppm and 25 ppm pure TBTO exposures
respectively, 0.315 and 1.65 mg/kg-day for the commercial TBTO exposures, respectively, and
0.359 and 1.52 mg/kg-day for the 5 ppm and 25 ppm tributyltin chloride exposures, respectively.
A group of control and 25 ppm rats were sacrificed after 1 week of treatment. At necropsy,
major organs were examined, and liver, spleen, thymus, and brain were weighed and processed
for histological examination. Mesenteric lymph nodes were also examined. Rats treated with 25
ppm of pure bis(tributyltin)oxide or tributyltin chloride ate significantly less than controls and
gained weight at a significantly reduced rate. After 1 week of exposure to 25 ppm pure bis(tri-n-
butyltin)oxide, relative, but not absolute liver weight was significantly increased; other organs
were not affected. Thymus weight was not provided for this time-point. Histologically, there
were changes indicative of atrophy and lymphocyte depletion in the thymus; the spleen showed a
decrease in thymus-dependent lymphocytes. No effects were observed in the kidneys. After 4
weeks of treatment, the only effect on organ weights noticed was a significant decrease in
relative and absolute thymus weight at 25 ppm pure bis(tributyltin)oxide and tributyltin chloride.
No histological alterations were noticed in the liver. All rats exposed to tributyltin chloride or to
25 ppm of either pure or commercial oxide had markedly hemorrhagic lymph nodes; this same
effect was seen in half of the rats treated with 5 ppm of either oxide. Partial atrophy was evident
in these nodes, whereas the appearance of the thymus was almost completely normal. No
histopathological alterations were detected in the liver, kidneys, or spleen. This study identifies a
LOAEL of 0.315 for hemorrhage and partial atrophy of lymph nodes in rats There was no
NOAEL.
Barnes and Stoner (1958) investigated the effects of tributyltin acetate in rats. Groups of
12 rats (sex distribution was not specified) were given tributyltin acetate in the diet at levels of 0,
25, 50, or 100 ppm for three months. These levels correspond to approximately 0, 2.5, 5.0, or 10
mg/kg-day tributyltin acetate, respectively, based on a food consumption rate of 0.1. Graphic
data presented in the report show that dose levels >50 ppm greatly decreased body weight gain in
14

-------
3-1-2006
a dose-related manner (-9% decrease in weight gain at 50 ppm and -26% at 25 ppm), but a
statistical analysis of the data was not reported. Food consumption data were not reported.
Without giving further details, the investigators indicated that animals in the 50 ppm group
showed slightly impaired health. Two rats died at 100 ppm during the first three weeks, but the
survivors appeared healthy at sacrifice. At necropsy, 4 out of 11 rats in the 100 ppm group
showed some degree of bile duct injury. No bile duct lesions were seen in other groups. In
addition, the authors stated that the water content in the spinal cord, but not in the brain, was
significantly increased (p<0.01) in rats treated with the 100 ppm level; the toxicological
significance of this finding is unclear. Based on the limited information provided in this study, it
appears that the 50 ppm (5.0 mg/kg-day) level represents a NOAEL for tributyltin acetate. The
LOAEL, which is also a frank effect level, is 100 ppm (10 mg/kg-day) for bile duct injury and
death in rats exposed to tributyltin acetate. The NOAEL and LOAEL/FEL values are equivalent
to 4.2 and 8.4 mg tributyltin/kg-day.
A two-generation reproductive toxicity study was conducted in Wistar rats exposed to
tributyltin chloride (>95% purity) in feed; methods and results for the parental generation and for
female offspring were published by Ogata et al. (2001), whereas methods and results for male
offspring were published by Omura et al. (2001). Groups of male and female Wistar rats were
mated over a 4-day period and females with confirmed vaginal plugs (10-12/group) were given
diets containing 0.03 (control), 5, 25, or 125 ppm of tributyltin chloride from gestational day 0
(GD0) until weaning of the Fj generation (postnatal day 22, PND 22). The authors estimated
doses as 0, 0.4, 2.0, or 10 mg/kg-day. Maternal body weights were recorded on GD 0, 7, and 14,
and food consumption was measured between GD 7 and 8 and GD 14 and 15. During the
lactational period, body weights were recorded on PND 7, 14, and 21, and food consumption was
measured between PND7 and 8 and PND 14 and 14. F0 generation rats were euthanized at
weaning on PND22. On the day of birth of Fj rats (PND 0), the live and dead offspring were
counted, sexed, and examined for gross malformations. On PND 1, litters were culled to 4 males
and 4 females. The body weights of Fj rats were recorded on PND 1, 7, and 21, the anogenital
distance was recorded on PND 1 and 4, and eye opening was examined from PND 14. Fj rats
were weaned on PND 22, at which time they received the same parental diet Body weights and
mean food consumption were recorded weekly for Fj rats. Vaginal opening was examined from
PND30 and the estrous cycle was evaluated during PND71-92. A 14-day cohabitation period of
Fj mating pairs started on PND 92; females that did not mate were euthanized. Procedures for
the Fj generation (10-14 per group) during gestation and lactation were the same as for the F0
generation. On the first day of the estrous stage from PND 148, Fj rats were killed, a blood
sample was taken for measurement of serum estradiol and testosterone, and the uterus and
ovaries were weighed. Procedures for the F2 offspring were identical except that these rats were
not mated.
Ogata et al. (2001) reported that for both the F0 and Fj parental generations, treatment
with tributyltin chloride had no significant adverse effect on maternal survival, food
15

-------
3-1-2006
consumption, the fertility index, the number of litters, or the mean duration of gestation; there
were also no effects in the Fj and F2 pups on the neonatal sex ratio, or at termination on ovarian
or uterine histology or serum levels of 17p-estradiol or testosterone. The following statistically
significant changes compared to controls were observed. In both generations, treatment at 125
ppm significantly reduced maternal weight gain, the total number of pups per litter, the live birth
index, and pup body weights per litter on PND1. The weights of Fj and F2 female pups were
significantly reduced at >25 ppm during lactation and at 125 ppm after weaning up to PND92;
the effect at 25 ppm appears to be minimal and transient (observed only at PND 14 and PND 21)
and therefore, not biologically significant. The day of eye opening was not affected in Fj pups,
but was significantly delayed in F2 pups exposed at 125 ppm. There was a dose-related trend for
increasing anogenital distances (measured on PND 1 and PND 4) in treated female pups, but only
the 125 ppm group showed statistically significant increases (ranging from 10 to 15%) in both
the Fj and F2 generations. The statistically significant increases at 5 ppm and 25 ppm in Fj pups
were judged not to be biologically relevant, as they were minimal (ca. 7%), and because no effect
was noted in the F2 generation. The age of vaginal opening was delayed by 6 days in Fj and F2
pups exposed at 125 ppm. Estrous cycle effects at 125 ppm included a significantly reduced
duration (F2 only) and a significantly reduced percentage of normal cycles in both Fj and F2 rats.
At 125 ppm, uterine weights relative to body weight were significantly elevated only in F2 rats
and ovarian relative weights were significantly reduced only in Fj rats. In this study, the 25 ppm
dietary level of tributyltin chloride, equivalent to 2 mg/kg-day, is a NOAEL and 125 ppm,
equivalent to 10 mg/kg-day is a LOAEL for significant effects noted in both generations:
reductions in maternal weight gain, number of live pups per litter, and pup body weights (at birth,
during lactation and after weaning), and effects on sexual development (delays in vaginal
opening and increases in anogenital distances and the frequency of abnormal estrous cycles).
The NOAEL and LOAEL are equivalent to 1.8 and 8.9 mg tributyltin/kg-day.
Omura et al. (2001) reported the results for Fj and F2 male rats exposed to tributyltin
chloride in the two-generation reproductive toxicity study; see Ogata et al. (2001) for initial
description of parental treatments. Dietary exposure levels were 0, 5, 25, or 125 ppm, reported as
equivalent to daily doses of 0, 0.4, 2, or 10 mg/kg-day. Body weights of Fj pups were recorded
on postnatal days (PND) 1,4, 14, and 21. Anogenital distances for male pups were recorded on
PND 1 and 4, eye opening was examined from PND 14 and the descent of the testes was
examined from PND 20. Beginning at weaning on PND 22, body weights and food consumption
of Fj male were recorded weekly. On PND 91, Fj males cohabited with Fj females for 14 days.
Fj males were sacrificed on PND 119, at which time venous blood was collected for the analysis
of testosterone, 17-p-estradiol and luteinizing hormone (LH). Absolute weights of the testes and
epididymis, and weights of the ventral prostate and seminal vesicles (without fluid) relative to
100 g body weight were recorded; the testes were evaluated for histopathology. Spermatids from
the testes and sperm from the cauda epididymis were counted; mature sperm were evaluated for
motility and structural aberrations. F2 males were treated as the Fj generation except that single
males from each litter underwent terminal sacrifice and analysis on PND 91 and were not mated.
16

-------
3-1-2006
The results of exposure to tributyltin chloride were similar in Fj and F2 male rats (Omura
et al., 2001). Body weight gains were significantly reduced during lactation (PND 1-21) and
weaning (PND 22-91) at 125 ppm in both generations (70-80% for Fj and 65-71% for F2 males
compared to controls); weight gains were also significantly reduced at PND 14 and PND 21 in Fj
males exposed at 25 ppm. Treatment had no significant effect on food consumption (PND 22-
91), anogenital distance (AGD), or on the day of testis descent in either generation, or on fertility
or copulation indices in Fj adults. The opening of the eyes was delayed in Fj males exposed at
>25 ppm and in F2 males exposed at 125 ppm; at 125 ppm, the delay was 0.6 days for Fj rats and
1.2 days for F2 rats. Note that for the following parameters under discussion, Fj males were
exposed up to PND 119, whereas F2 males were exposed up to PND 91; despite the shorter
duration of exposure, compound-related changes in male reproductive parameters tended to be
more severe in the F2 generation than in the Fj generation. Treatment at 125 ppm resulted in
biologically and statistically significant reductions in organ weights; for the Fj and F2
generations, respectively, the absolute weights were reduced in the testes by 16 and 21% and in
the epididymis by 12 and 20%. Relative weights of the ventral prostate were significantly
reduced in Fj males by 16% at 125 ppm and in F2 males by 16% at 25 ppm and 31% at 125 ppm;
treatment had no effect on relative weights of the seminal vesicles. The testicular and epididymal
weight effects at 5 ppm in Fj rats were judged not to be of biological significance because the
effects were minimal (less than 5%) and were not observed in the F2 males. Statistically
significant effects on sperm parameters included lowered testicular spermatid counts (by 20% at
125 ppm in Fj males and by 11% at 25 ppm and 23% at 125 ppm in F2 males), reduced
epididymal sperm counts (by 24% at 125 ppm in F2 males only), and increased incidence of
sperm abnormalities (tailless sperm at 125 ppm in Fj males only); sperm motility was not
affected by treatment. The authors did not regard occasional testicular histopathological findings
(vacuolization of and spermatid retention in seminiferous epithelium and germ cell degeneration)
in Fj males to be abnormal because of the low frequency (data not reported). In F2 rats, the
frequencies of minimal changes were high in some treated rats and thus were categorized as
abnormal by the authors, but the overall number of affected animals was low and not statistically
different from controls. Statistically significant changes in serum levels were observed at 125
ppm: elevations in testosterone and decreases in 17-p-estradiol in both generations and increased
LH in F2 males only. In this study, the lowest dietary level of 5 ppm tributyltin chloride (a dose
0.4 mg/kg-day) was a NOAEL and 25 ppm (a dose of 2 mg/kg-day) was a LOAEL for decreased
relative weight of the ventral prostate and lowered testicular spermatid counts in F2 rats. The
NOAEL and LOAEL are equivalent to 0.36 and 1.8 mg tributyltin/kg-day, respectively.
Harazono et al. (1996) evaluated the effects of tributyltin chloride (96% purity) on early
gestation in rats. Groups of 10-14 inseminated female Wistar rats received tributyltin by gavage
in olive oil at doses of 0, 8.1, 12.2, or 16.3 mg/kg-day on GD 0-7. Maternal body weight, food
consumption and clinical signs of toxicity were recorded daily. At termination on GD 20, live
fetuses were sexed, weighed, and inspected for external and oral malformations; two-thirds of the
live fetuses were examined for skeletal malformations and the rest for visceral malformations.
17

-------
3-1-2006
Treatment had no effect on maternal mortality, but resulted in dose-related increases in the
incidences (not reported) of clinical signs of toxicity (sluggishness, bloody stain around the eyes
and diarrhea). For the reporting period GD 0-8, which included days of dosing, food
consumption was severely reduced compared to control values: by 41, 67, and 73% in the low-to-
high dose groups. Food consumption was normal in all treated groups for the reporting period
after dosing (GD 8-20). Statistically significant reductions in maternal body weight occurred at
> 12.2 mg/kg-day during GD 0-8; maternal body weight gain at 8.1 mg/kg-day was lower than in
controls, but the difference was not statistically significant. Maternal body weight gain excluding
the uterus was not affected by treatment. Treatment with tributyltin chloride resulted in a dose-
related increase in percent pregnancy failure that was statistically significant at > 12.2 mg/kg-day:
0, 18.2, 71.4, and 76.9% for the control and low-to-high dose groups. There were no dose-
related effects on the number of corpora lutea, the number of implantations, preimplantation or
postimplantation losses per litter, the sex ratio or body weights of live fetuses, or the incidences
of malformations. For treatment with tributyltin chloride on GD 0-7, the NOAEL for maternal
toxicity was 8.1 mg/kg-day and the LOAEL was 12.2 mg/kg-day (equivalent to 7.2 and 10.9 mg
tributyltin/kg-day respectively) for reduced feed consumption and absence of implantation. The
highest dose of tributyltin chloride, 16.3 mg/kg-day (equivalent to 14.6 mg tributyltin per kg-
day), was a NOAEL for fetal toxicity in successful pregnancies.
As part of a comparative developmental toxicity study on mono-, di- and tributyltin
compounds, groups of 6-11 pregnant female Wistar rats were given tributyltin chloride at doses
of 0, 40, or 80 mg/kg-day by gavage in olive oil on gestational days (GD) 7 and 8 (Ema et al.,
1995a). Maternal body weight was monitored up to GD 20; results were reported for the periods
GD 0-7, 7-9, 9-20, and 0-20. At termination on GD 20, uteri were examined for the numbers of
resorptions and dead and live fetuses. All live fetuses were sexed, weighed, and examined for
external malformations, including those of the oral cavity; two thirds of the fetuses were
examined for skeletal malformations and the rest for visceral malformations. Treatment with
tributyltin chloride had no effect on maternal survival. Dose-related reductions in maternal body
weight were reported for the period GD 7-9, resulting in significant reductions in body weight
gain in both dosed groups compared to controls at termination. Treatment at >40 mg/kg-day
caused a significant dose-related increase in the percentage of postimplantation loss per litter.
Treatment at 80 mg/kg-day significantly increased the number of litters totally resorbed and
reduced the number of live fetuses per litter. Tributyltin chloride did not significantly increase
the incidences of external, skeletal, or visceral malformations in rats at doses that were
maternally toxic. In this study, 40 mg/kg-day (equivalent to 35.8 mg tributyltin/kg-day) was a
LOAEL for maternal toxicity (reduced body weight gain) and fetal toxicity (increased
postimplantation loss per litter) following exposure to tributyltin chloride.
Ema et al. (1995b) also exposed groups of 11-14 pregnant Wistar rats to tributyltin
chloride (96% purity) by gavage in olive oil at doses of 25 or 50 mg/kg-day on GD 7-9, doses of
50 or 100 mg/kg-day on GD 10-12 or doses of 25, 50, or 100 mg/kg-day on GD 13-15. A control
18

-------
3-1-2006
group received olive oil on GD 7-15. Analysis was as described for Ema et al. (1995a) except
that results for maternal body weight were reported for the periods GD 0-7, GD 7-10, GD 10-13,
GD 13-16, GD 16-20, and GD 0-20. There was no treatment-related maternal mortality.
Maternal weight gain was significantly reduced for the period of dosing for all treated groups and
reduced overall (GD 0-20) for treatment at >25 mg/kg-day on GD 7-9 or at 100 mg/kg-day on
GD 10-12 or GD 13-15. Treatment at >25 mg/kg-day on GD 7-9 resulted in significant
reductions in the numbers of live fetuses per litter and increases in post-implantation losses per
litter and total litter resorptions, but had no effect on the incidence of malformations. Treatment
with tributyltin chloride at >50 mg/kg-day on GD 10-12 resulted in significantly lower body
weight for female fetuses. Treatment at 100 mg/kg-day on GD 10-12 significantly reduced the
numbers of live fetuses per litter and increased post-implantation losses per litter; at this dose,
male and fetal body weights were reduced and the incidence of fetuses or litters with
malformations (cleft palate) was increased. Effects observed following treatment on GD 13-15
included significantly reduced fetal body weights in both sexes at 100 mg/kg-day and increased
external malformations (cleft palate) at >25 mg/kg-day. The lowest dose of 25 mg/kg-day for
tributyltin chloride (equivalent to 22.4 mg tributyltin/kg-day) was a LOAEL for maternal toxicity
(reduced body weight gain following treatment on GD 7-9), fetal toxicity (increased death and
total litter losses after treatment on GD 7-9) and teratogenicity (external malformations, primarily
cleft palate, after treatment on GD 13-15).
In another comparative developmental toxicity study, Ema et al. (1996) exposed groups
of 10-11 pregnant Wistar rats to 0, 50, or 100 mg/kg-day of tributyltin chloride by gavage in
olive oil on gestational days (GD) 13-15. The protocol was the same as for the study by Ema et
al. (1995a) except that maternal body weights were reported for the periods GD 0-13, GD 13-16,
GD 16-20, and GD 0-20. One of the high dose rats died, but the cause of death was not reported.
Maternal body weight gain was significantly reduced in both treated groups. Treatment had no
effect on numbers of litters or implantations, the fetal sex ratio, or the incidences of skeletal or
visceral malformations. Treatment at 100 mg/kg-day significantly reduced body weights of male
and female fetuses and treatment at >50 significantly increased the incidence of fetuses or litters
with external malformations, primarily cleft palate. The low dose of 50 mg/kg-day for tributyltin
chloride (equivalent to 44.7 mg tributyltin/kg-day) was a LOAEL for maternal toxicity (reduced
body weight gain) and teratogenicity.
Other Studies
An acute exposure study conducted by Snoeij et al. (1988) compared the toxicity of
monobutyltin trichloride, dibutyltin dichloride, and tributyltin chloride in rats. Significant
decreases in thymus weight were observed at >5 mg/kg dibutyltin dichloride and at > 10 mg/kg
tributyltin chloride, but no changes in thymus weight were observed at doses of 10-180 mg/kg
monobutyltin trichloride. These results suggest that the dose-responses for immunological
19

-------
3-1-2006
effects observed following exposure to dibutyltin or tributyltin compounds may not apply to
monobutyltin compounds.
Data summarized by Boyer (1989) indicate that alkyltin compounds are metabolized
mainly in the liver by the P-450 monooxygenase system. Tributyltin acetate was metabolized by
isolated rat microsomes to form alpha-, beta-, gamma- and delta-hydroxybutyldibutyltin
derivatives, as well as 1-butanol and 1-butane. Further oxidation of the gamma-hydroxy
compound yielded the corresponding ketone. Tetrabutyltin incubated with the liver microsome
fraction produced tributyltin derivatives; similarly, dibutyltin diacetate produced butyltin
derivatives. Several of the metabolites that were produced in vitro were also detected in the liver
and feces of mice exposed to tributyltin acetate or dibutyltin diacetate by gavage. In rats, an
initial increase in tributyltin observed in the liver after exposure to tributyltin fluoride by gavage
was followed by an increase in dibutyltin, monobutyltin and inorganic tin. Biliary excretion
represents the main route of excretion of butyltin compounds.
In a comparative in vitro toxicity experiment, Seinen et al. (1977b) exposed primary
cultures of human (children), rat, mouse or guinea pig thymocytes to dibutyltin dichloride at
concentrations of 0, 0.5, 0.5, 5 or 50 [ig/mL for up to 24 hours. The compound had no effect on
the cell counts or viability of thymocytes from mice or guinea pigs. Cell counts were reduced for
human thymocytes exposed at >0.5 mg/mL and cell counts and viability of rat thymocytes were
reduced following exposure at >0.05 mg/mL. These data suggest that rats are a more appropriate
animal model than mice or guinea pigs for the immunotoxicity of dibutyltin dichloride in
humans.
Measured and estimated chemical properties of three tributyltin compounds are compared
in Table 2. Tributyltin oxide is different from the acetate or chloride in having a larger molecular
size (because of its bis configuration) and the lowest solubility in water. However, the Log Kow
values suggest that the ability of the three compounds to cross biological membranes may be
similar.
Table 2. Comparison of Chemical Properties of Some Tributyltin Compounds2

Tributyltin Acetate
Tributyltin Chloride
Tributyltin Oxide
CAS No.
56-36-0
1461-22-9
56-35-9
Molecular Weight
349.1
325.51
596.12
Log Kow
3.24 (estimate)
4.76 (experimental)
4.70 (estimated)
3.84 (experimental)
4.05 (estimated)
Water Solubility at 25
degrees C (mg/L)
10.78 (estimated)
0.7478 (estimated)
0.08958 (estimated)
" Based on EPIWIN version 3.11
20

-------
3-1-2006
DERIVATION OF PROVISIONAL SUBCHRONIC AND CHRONIC RfDs FOR
BUTYLTIN COMPOUNDS
Monobutyltin Compounds
No data are available for the oral toxicity of monobutyltin compounds in humans and no
subchronic- or chronic-duration oral studies are available for animals. Three developmental
toxicity studies are available for Wistar rats exposed to monobutyltin chloride by gavage (Noda
et al., 1992a; Ema et al., 1995a; Ema and Harazono, 2001). The results of these studies are
summarized in Table 3. No teratogenicity was reported for monobutyltin trichloride and the
compound appears not to be a specific developmental toxicant, because fetal effects only
occurred at doses that were toxic to dams. These data are insufficient to derive an RfD for
monobutyltin compounds because sensitive targets have not been identified.
Table 3. Developmental Toxicity in Wistar Rats Exposed to Monobutyltin Trichloride by Oral Gavage
Study
Treatment Days
NOAEL/LOAEL
(mg monobutyltin/kg-day)
Effect
Noda et al. (1992a)
GD 7-17
Maternal: 248 / ND
Fetal: 248 / ND
none
Ema et al. (1995a)
GD 7-8
Maternal: ND / 620
Fetal: 620 / 930
reduced body weight gain in dams and
fetuses
Ema and Harazono
(2001)
GD 0-3 or
GD 4-7
Maternal: 140 / 560
Fetal: 140 / 560
reduced feed consumption and body
weight gain in dams and female fetuses
ND = not determined
Dibutyltin Compounds
Table 4 summarizes NOAEL and LOAEL values (expressed as mg dibutyltin per kg body
weight per day) for the toxicity studies most relevant to risk assessment for dibutyltin.
No data are available for the oral toxicity of dibutyltin compounds in humans. The oral
toxicity database includes chronic-duration (primarily carcinogenicity) studies in rats and mice,
subchronic studies in rats, short-term studies in rats and mice, and developmental studies in rats.
Based on the available data, the most sensitive target of dibutyltin compounds is the immune
system. Thymic atrophy, as evidenced by a decrease in thymus weight, was observed in
weanling rats exposed to >5.4 mg dibutyltin/kg-day as dibutyltin dichloride in the diet for two
weeks (Seinen et al., 1977b) and in pregnant rats given gavage doses of 7.7 mg dibutyltin/kg-day
as dibutyltin dichloride on GD 6-15 (Farr et al., 2001) or >3.3 mg dibutyltin/kg-day as dibutyltin
21

-------
3-1-2006
Table 4. Oral Toxicity of Dibutyltin Compounds in Rodents
Reference	Exposure Conditions
(days/vehicle/ compound)
NOAEL / LOAEL (mg
dibutyltin/kg-day)
Effect
Rats- Systemic toxicity studies
Seinen et al.
(1977a)
Seinen et al.
(1977b)
Seinen et al.
(1977b)
Gaunt et al.
(1968)
Barnes and
Stoner (1958)
NCI (1979)
2 weeks/feed/dibutyltin
dichloride
4-6 weeks/feed/dibutyltin
dichloride
9 weeks/arachis oil gavage/
dibutyltin dichloride
90 days/feed/dibutyltin
dichloride
6 months/feed/dibutyltin
dichloride
2 years/feed/dibutyltin
diacetate
Mice- Systemic toxicity studies
Seinen et al.
(1977a,b)
NCI (1979)
4 weeks/feed/dibutyltin
dichloride
2 years/feed/dibutyltin
diacetate
Rats- Developmental toxicity studies
Nodaetal.	GD 7-17/olive oil gavage/
(1992a)	dibutyltin diacetate
Ema et al.	GD 7-15/olive oil gavage/
(1991)	dibutyltin dichloride
Farr et al.	GD 6-15/ olive oil gavage/
(2001)	dibutyltin dichloride
Seinen et al. GD 2-PND39/feed/ dibutyltin
(1977a)	dichloride
ND/5.4
ND/3.8
ND/0.33
3 /ND
1.5/3.0
3.5/7.0
24.5/ND
8.9/17.8
maternal: 1.1/3.3
fetal: 1.1/3.3
maternal: 3.8 / 5.7
fetal: 1.9/3.8
maternal: 3.8 / 7.7
fetal: 3.8/7.7
pups: ND / 3.8
Reduced thymus weight and size; depletion
of thymic lymphocytes
Reduced thymus-dependent humoral
immunity (to sheep RBCs)
Reduced body weight gain and thymus-
dependent cellular immunity (delayed
allograft rejection)
No systemic toxicity
Reduced body weight gain; histopathology
of bile duct
Reduced body weight gain in both sexes;
reduced survival and increased bile calculi
in males
No systemic or immunotoxic effects
Reduced survival and body weight gain in
females
Reduced thymus weight in dams; increased
skeletal variations in fetuses
Increased mortality and decreased body
weight gain in dams; decreased body
weight and increased external and skeletal
malformations in fetuses
Decreased food consumption, body weight
gain, and thymus weight in dams; slight
increased incidence of defects in fetuses
Reduced number of spleen cells, splenic
antibody cells and body weight gain in
pups
ND = not determined.
22

-------
3-1-2006
diacetate on GD 7-17 (Noda et al., 1992a, 1992b). Thymus-dependent immune function was
impaired in weanling rats fed diets supplying 4.5 mg dibutyltin/kg-day as dibutyltin dichloride
for 4-6 weeks (Seinen et al., 1977b). Thymus-dependent cellular immunity (allograft rejection)
was impaired in newborn rats that received dibutyltin dichloride by gavage at 1 mg/kg-day, three
days/week for nine weeks beginning two days after birth (Seinen et al., 1977b); averaged over a
week, the doses were 0.33 mg dibutyltin/kg-day. Seinen et al. (1977b) mentioned anecdotally
(possibly as a result of a range-finding study) that severe atrophy of lymphoid organs was the
only pathological alteration observed in rats following postnatal intubation with 5 mg/kg
dibutyltin dichloride, leading to growth stunting and increased mortality. No immunotoxicity or
any other systemic effect was observed in mice that received <24.5 mg dibutyltin/kg-day as
dibutyltin dichloride via the diet for four weeks (Seinen et al., 1977a, 1977b). No thymus effects
were noted in the two-year dietary studies in rats and mice in which the animals were six weeks
old at the start of exposure to dibutyltin diacetate (NCI, 1979).
The most appropriate basis for the provisional RfD for dibutyltin compounds appears to
be a LOAEL for immunotoxicity (reduced thymus-dependent cellular immunity, as exemplified
by delayed allograft rejection) and reduced body weight in neonatal rats exposed to 1 mg/kg-day
dibutyltin dichloride by gavage 3 days/week (daily average of 0.33 mg dibutyltin/kg-day) for nine
weeks beginning on postnatal day 2 (Seinen et al., 1977b). A NOAEL for dibutyltin has not been
identified. The study did not provide sufficient information to support a benchmark dose
analysis. The LOAEL is divided by an uncertainty factor of 1000 (10 for extrapolation from a
LOAEL, 10 for interspecies extrapolation, 10°5 for intraspecies variability, and 10°5 for database
deficiencies) to yield provisional subchronic and chronic RfD values of 3E-4 (3 x 10"4) mg
dibutyltin/kg-day, as follows:
subchronic p-RfD = LOAEL / UF
= 0.33 mg dibutyltin/kg-day / 1000
= 3E-4 mg dibutyltin/kg-day
p-RfD = LOAEL / UF
= 0.33 mg dibutyltin/kg-day / 1000
= 3E-4 mg dibutyltin/kg-day
A factor of 3 for intraspecies variability was chosen because the neonatal rats were treated
during the postnatal period critical for thymus maturation (Seinen et al., 1977b), and therefore
represent a sensitive population or stage of development. The primary database deficiency is the
lack of a 2-generation reproduction study; the lack of a developmental toxicity study in a second
species is less critical because immunotoxicity is well-established as the most sensitive effect for
butyltins in general, usually occurring at exposures more than an order of magnitude below
developmental toxicity levels. A subchronic-to-chronic uncertainty factor for the provisional
chronic RfD appears to be unjustified considering that no thymus histopathology was noted in
23

-------
3-1-2006
rats treated with dibutyltin diacetate for two years from the age of six weeks at doses as high as
12.3 mg/kg-day (NCI, 1979). Although the short study duration of 9 weeks would generally be
of concern when extrapolating to chronic exposure, it is not an issue in this case because dosing
in this study included the postnatal period critical for thymus maturation. The provisional RfD
for dibutyltin should be protective against developmental toxicity.
Confidence in the principal study, Seinen et al. (1977b), is medium. It examined the
effect of dibutyltin on immune function during a sensitive period of thymus maturation, but the
group sizes were small and limited to males only, and a NOAEL was not identified. The results
of the critical experiment were supported by other functional tests in the same paper in which
older (weanling) rats of one sex were exposed via the diet at slightly higher doses. Confidence in
the database is low. The NCI (1979) study examined the chronic toxicity of dibutyltin diacetate
in rats and mice exposed via the diet, but was designed to assess carcinogenicity and did not
include evaluations of hematology, clinical chemistry, or urinalysis parameters (although the
thymus was examined for histopathology). Immunotoxicity has not been examined in studies of
long-term duration. In addition, there are no data on the developmental toxicity of dibutyltin
compounds in a second species and no data on the potential of dibutyltin compounds to induce
reproductive effects. Reflecting the low confidence in the database, there is low confidence in
the provisional subchronic and chronic RfD values.
This p-RfD is for mg dibutyltin/kg-day. If soil or water concentrations at the site of
concern are expressed in units of dibutyltin compound (such as dibutyltin dichloride), a
molecular weight conversion can be made as follows:
RfD dibutyltin compound
(mg/kg-day) = RfDdlbutyltm (mg/kg-day) x [MWdlbutyltm
compound ^^^^^dibutyltin]
where: MW = molecular weight.
Tributyltin Compounds
Table 5 summarizes the results of oral toxicity studies of tributyltin acetate and tributyltin
chloride (expressed as mg tributyltin per kg body weight per day).
No data are available for the oral toxicity of tributyltin compounds in humans. No
chronic-duration exposure data are available for tributyltin compounds other than tributyltin
oxide. Based on the available data (excluding tributyltin oxide), the most sensitive LOAEL was
for hemorrhagic, partially atrophic lymph nodes in rats that received 0.4 mg/kg-day tributyltin
chloride (equivalent to 0.36 mg tributyltin/kg-day) via the diet for 4 weeks (Bressa et al., 1991); a
NOAEL was not identified. Relative and absolute thymus weights were reduced at higher doses
in this study. Immune system toxicity following dietary exposure to tributyltin chloride was also
24

-------
3-1-2006
Table 5. Oral Toxicity of Tributyltin Compounds in Rats
Reference
Exposure Conditions:
days/vehicle/doses
NOAEL/LOAEL
(mg tributyltin/kg-day)
Effect
Systemic toxicity
Snoeij et al.
(1985)
Bressa et al.
(1991)
Barnes and
Stoner (1958)
2	weeks/diet/tributyltin
chloride (males only)
4 weeks/diet/tributyltin
chloride
3	months/diet/tributyltin
acetate
Reproductive/Developmental Toxicity
Ogata et al.
(2001)
Omura et al.
(2001)
Harazono et
al. (1996)
Ema et al.
(1995a)
Ema et al.
(1995b)
Ema et al.
(1996)
Two generations/diet/
tributyltin chloride
Two generations/diet/
tributyltin chloride
GD 0-7/olive oil gavage/
tributyltin chloride
GD 7-8/olive oil gavage/
tributyltin chloride
GD 7-9, 10-12, or 13-15/
olive oil gavage/tributyltin
chloride
ND / 1.8
ND / 0.315
4 / 8.1
1.8 / 8.9
0.36 / 1.8
maternal: 7.2 / 10.9
fetal: 14.6 / ND
maternal: ND / 35.8
fetal: ND / 35.8
maternal: ND / 22.4
fetal: ND /22.4
GD 13-15/olive oil gavage/ maternal: ND / 44.7
tributyltin chloride	fetal: ND / 44.7
Reduced relative spleen weight
Hemorrhage and partial atrophy of
lymph nodes
Bile duct injury and reduced body
weight gain
Reduced total and live pups/litter and
pup body weights, increased
anogenital distance, abnormal estrous
cycle, delayed vaginal opening
Reduced relative prostate weight and
testicular spermatid counts in F2 males
Reduced feed consumption and
inhibited implantation in dams; no
fetal effects
Reduced body weight gain and
increased postimplantation loss
Reduced body weight gain in dams;
increased fetal deaths / litter losses
and external malformations
Reduced body weight gain in dams;
increased external malformations in
pups
ND = not determined
25

-------
3-1-2006
reported in studies on weanling male rats by Snoeij et al. (1985). In the two-week study, relative
spleen weights were significantly reduced at >1.8 mg tributyltin/kg-day, and relative thymus
weights were reduced and thymus glands were depleted of lymphocytes at higher doses. In the
four-week study, adverse effects on the thymus and body weight gain were observed at 13
mg/kg-day (11.6 mg tributyltin/kg-day), the only dose tested (Snoeij et al., 1985).
After immunotoxicity, the next most sensitive target of tributyltin was the male
reproductive system, as reported in the two-generation reproductive study in rats dietarily
exposed to tributyltin chloride (Omura et al., 2001). F2 males appeared to be more sensitive to
tributyltin chloride than Fj males, although they were treated for 28 fewer days. In this study, the
NOAEL was 0.36 mg tributyltin/kg-day; the relative weight of the ventral prostate and testicular
spermatid counts were reduced in F2 males at > 1.8 mg tributyltin/kg-day. At higher doses,
testicular and epididymal weights were reduced in Fj and F2 males, testicular spermatid counts
were reduced in Fj males, and epididymal sperm counts were reduced in F2 males. In the F0
treated dams, as reported in the companion study by Ogata et al. (2001), maternal toxicity
(reduced body weight gain) and reproductive toxicity (reductions in litter size, pup survival and
pup body weights at birth and weight gains during lactation and weaning) were observed at 8.9
mg tributyltin/kg-day. Female offspring were less sensitive to tributyltin chloride, as shown by
the NOAEL of 1.8 mg tributyltin/kg-day (Ogata et al., 2001). At 8.9 mg tributyltin/kg-day, Fj
and F2 females exhibited an increased anogenital distance (indicative of interference in sexual
development), delayed vaginal opening (possibly related to reductions in body weight gain), and
an increase in the frequency of abnormal estrous cycles.
Adverse effects on the bile duct were noted at 8.2 mg tributyltin/kg-day in rats exposed to
tributyltin acetate in the diet for six months (Barnes and Stoner, 1958). However, as discussed
above in the derivation for dibutyltin compounds, it is not certain that this effect is relevant to
humans. Developmental toxicity studies on tributyltin chloride observed impaired implantation,
post-implantation losses, and external malformations, but only at maternally toxic doses of 22.4
mg tributyltin/kg-day or more (Ema et al., 1995a, 1995b, 1996; Harazono et al, 1996).
The most sensitive endpoint in the literature was identified in the Bressa et al. (1991)
4-week rat dietary exposure study. The LOAEL of 0.332 mg/kg-day for hemorrhagic, partially
atrophic lymph nodes in rats exposed to the commercial grade formulation of TBTO (equivalent
to 0.315 mg tributyltin/kg-day) is selected as the basis for the subchronic RfD. A NOAEL was
not identified. Data from this study were not suitable for benchmark dose analysis. The LOAEL
is divided by an uncertainty factor of 1000 (10 for extrapolation from a LOAEL, 10 for
interspecies extrapolation, 10 for human variability) to yield a provisional subchronic RfD of
3E-4 (3 x 10"4) mg tributyltin/kg-day, as follows:
26

-------
3-1-2006
subchronic p-RfD = LOAEL / UF
= 0.315 mg tributyltin/kg-day / 1000
= 3E-4 mg tributyltin/kg-day
An uncertainty factor for database weakness was not used because the database includes a
multigeneration reproduction study and developmental toxicity studies showing effects only at
high doses that also produced overt toxic effects in the dams. An uncertainty factor for
extrapolation from a less-than-subchronic duration was not used as chronic toxicity data for
TBTO indicate that the LOAEL does not decrease with increased duration of exposure
(Toxicological Review for TBTO on IRIS, U.S. EPA, 2006). The provisional subchronic RfD is
expected to be protective against developmental toxicity.
Confidence in the critical study (Bressa et al., 1991) is medium; it thoroughly examined
sensitive endpoints relative to immunotoxicity, but was limited by relatively small group sizes,
short study duration, lack of testing in females, and failure to identify a NOAEL. Confidence in
the subchronic database is medium. Supporting systemic toxicity data are limited, but adequate
testing for developmental and reproductive effects showed effects only at higher doses.
Confidence in the provisional subchronic RfD for tributyltin compounds other than tributyltin
oxide is medium.
The 4-week study by Bressa et al. (1991) is of insufficient duration to serve as the basis
for a chronic RfD. In the absence of suitable data on other tributyltin compounds, application of
the verified RfD for tributyltin oxide [bis(tri-n-butyltin)oxide] (U.S. EPA, 1997b, 2005) to other
tributyltin compounds was considered. A comparison of Log Kow values indicates that
absorption and distribution of tributyltin oxide would be similar to that of the acetate and
chloride (Table 4). Furthermore, the comparative study by Bressa et al. (1991) reported identical
LOAEL values of 0.4 mg/kg-day for hemorrhagic lymph nodes in rats exposed to tributyltin
oxide or tributyltin chloride for 4 weeks. Similar to the other tributyltin compounds, the
extensive database for tributyltin oxide (U.S. EPA, 2005) confirms that developmental effects of
treatment occur at doses higher than those effective for immunotoxicity.
The verified chronic oral RfD on IRIS (U.S. EPA, 2006) of 3 x 10"4 mg/kg-day for
tributyltin oxide is adjusted for molecular weight (see Table 1) to serve as the basis for the
chronic oral RfD of 3E-4 mg tributyltin/kg-day for tributyltin compounds.
This provisional RfD is for mg tributyltin/kg-day. If soil or water concentrations at the
site of concern are expressed in units of tributyltin compound (such as tributyltin dichloride), a
molecular weight conversion can be made as follows:
27

-------
3-1-2006
tributyltin compound
(mg/kg-day) = RfDtnbutyltm (mg/kg-day) x [MWtnbutyltm
compound
/MWtributyltin]
where: MW= molecular weight.
REFERENCES
ATSDR (Agency for Toxic Substances and Disease Registry). 2003. Toxicological Profile for
Tin. Update Draft for Public Comment. Public Health Service. Atlanta, GA.
Barnes, J.M. and H.B. Stoner. 1958. Toxic properties of some dialkyl and trialkyl tin salts. Brit.
J. Ind. Med. 15:15-22.
Bartalini, E. 1959. Studio sperimentale sulla tossicita' di un composto organico dello stagno
usato come plastificante. Med. Lavoro. 50:338-350. (Italian)
Boyer, I.J. 1989. Toxicity of dibutyltin, tributyltin and other organotin compounds to humans
and to experimental animals. Toxicology. 55:253-298.
Bressa, G., R.H. Hinton, S.C. Price et al. 1991. Immunotoxicity of tri-n-butyltin oxide (TBTO)
and tri-n-butyltin chloride (TBTC) in the rat. J. Appl. Toxicol. 11:397-402.
Bulten, E.J. and H.A. Meinema. 1991. In: Metals and their Compounds in the Environment:
Tin. Merian, E., ed. VCH, New York. p. 1243-1259.
Ema, M. and A. Harazono. 2000. Adverse effects of dibutyltin dichloride on initiation and
maintenance of rat pregnancy. Reprod. Toxicol. 14:451-456.
Ema, M. and A. Harazono. 2001. Toxic effects of butyl tin trichloride during early pregnancy in
rats. Toxicol. Lett. 125:99-106.
Ema, M., T. Itami and H. Kawasaki. 1991. Teratogenicity of di-n-butyltin dichloride in rats.
Toxicol. Lett. 58:347-356.
Ema, M., T. Itami and H. Kawasaki. 1992. Susceptible period for the teratogenicity of di-n-
butyltin dichloride in rats. Toxicol. 73:81-92.
Ema, M., R. Kurosaka, H. Amano and Y. Ogawa. 1995a. Comparative developmental toxicity
of butyltin trichloride, dibutyltin dichloride and tributyltin chloride in rats. J. Appl. Toxicol.
15:297-302.
28

-------
3-1-2006
Ema, M., R. Kurosaka, H. Amano and Y. Ogawa. 1995b. Further evaluation of the
developmental toxicity of tributyltin chloride in rats. Toxicology. 96:195-201.
Ema, M., R. Kurosaka, H. Amano and Y. Ogawa. 1996. Comparative developmental toxicity of
di-, tri- and tetrabutyltin chloride compounds after administration during late organogenesis in
rats. J. Appl. Toxicol. 15:297-302.
Farr, C.H., K. Reinisch, J.F. Holson and D. Neubert. 2001. Potential teratogenicity of di-n-
butyltin dichloride and other dibutyltin compounds. Teratogen. Carcinogen. Mutagen. 21:405-
415.
Gaunt, I.F., J. Colley, P. Grasso et al. 1968. Acute and short-term toxicity studies on di-n-
butyltin dichloride in rats. Fd. Cosmet. Toxicol. 6:599-608.
Harazono, A., M. Ema and Y. Ogawa. 1996. Pre-implantation embryonic loss induced by
tributyltin chloride in rats. Toxicol. Lett. 89:185-190.
IARC (International Agency for Research on Cancer). 2003. IARC Agents and Summary
Evaluations. Online, http://www-cie.iarc.fr/htdig/search.html
Kimbrough, R.D. 1976. Toxicity and health effects of selected organotin compounds: a review.
Environ. Health Perspect. 14:51-56.
Magos, L. 1986. In: Handbook on Toxicology of Metals: Tin, Vol II, 2nd Ed. Friberg, L.,
Nordberg, G.F. and Vouk, V.R., Eds. Elsevier Science, Amsterdam, p. 568-593.
NCI (National Cancer Institute). 1979. Bioassay of Dibutyltin Diacetate for Possible
Carcinogenicity. U.S. Dept of Health, Education and Welfare, Public Health Service, National
Institutes of Health, Bethesda, MD. NCI Technical Report Series 183.
Nicklin, S. and M.W. Robson. 1988. Organotins: toxicology and biological effects. Appl.
Organometallic Chem. 2:487-508.
NIOSH (National Institute for Occupational Safety and Health). 1976. NIOSH Criteria for a
Recommended Standard...Occupational Exposure to Organotin Compounds. DHHS (NIOSH)
Publication Nol. 77-115. NTIS PB-274 766. Online, http://www.cdc.gov/niosh/77-l 15.html
Noda, T., T. Yamano, M. Shimizu et al. 1992a. Comparative teratogenicity of di-n-butyltin
diacetate with n-butyltin trichloride in rats. Arch. Environ. Contam. Toxicol. 23:216-222.
29

-------
3-1-2006
Noda, T., T. Nakamura, M. Shimizu et al. 1992b. Critical gestational day of teratogenesis by di-
n-butyltin diacetate in rats. Bull. Environ. Contam. Toxicol. 49:715-722.
Noda, R., S. Morita and A. Baba. 1993. Teratogenic effects of various di-n-butyltins with
different anions and butyl(3-hydroxybutyl)tin dilaurate in rats. Toxicol. 85:149-160.
NTP (National Toxicology Program). 2003a. Nomination background document for organotin
(methyl and butyl) toxicity. Online.
http://ntp-server.niehs.nih.gov/htdocs/Chem Background/ExSumPdl70rganolins.pdf
NTP (National Toxicology Program). 2003b. Monobutyltin trichloride. Management Status
Report. Online. http://ntp-server.niehs.nih.gov/htdocs/Results Status/Resstatm/M000066.Html
NTP (National Toxicology Program). 2003c. Dibutyltin diacetate. Management Status Report.
Online. http://ntp-server.niehs.nih.gov/htdocs/Results Status/Resstatd/10670-W.Html
NTP (National Toxicology Program). 2003d. Dibutyltin diacetate. Health and Safety Report.
Online. http://ntp-server.niehs.nih.gov/htdocs/CHEM H&S/NTP Cheml/Radianl067-33-
O.html
Ogata, R., M. Omura, Y. Shimisaki et al. 2001. Two-generation reproduction toxicity study of
tributyltin chloride in female rats. J. Toxicol. Environ. Health., Pt. A. 63:127-144.
Omura, M., R. Ogata, K. Kubo et al. 2001. Two-generation reproductive toxicity study of
tributyltin chloride in male rats. Toxicol. Sci. 64:224-232.
Seinen, W., J.G. Vos, I. van Spanje, et al. 1977a. Toxicity of organotin compounds. II.
Comparative in vivo and in vitro studies with various organotin and organolead compounds in
different animal species with special emphasis on lymphocyte cytotoxicity. Toxicol. Appl.
Pharmacol. 42:197-212.
Seinen, W., J.G. Vos, R. Van Krieken et al. 1977b. Toxicity of organotin compounds. III.
Suppression of thymus-dependent immunity in rats by di-n-butyltindichloride and di-n-
octyltindichloride. Toxicol. Appl. Pharmacol. 42:213-224.
Snoeij, N.J., A.A.J, van Iersel, A.H. Penninks and W. Seinen. 1985. Toxicity of triorganotin
compounds: comparative in vivo studies with a series of trialkyltin compounds and triphenyltin
chloride in male rats. Toxicol. Appl. Pharmacol. 81:274-286.
Snoeij, N.J., A.H. Penninks and W. Seinen. 1987. Biological activity of organotin compounds-
an overview. Environ. Res. 44:335-353.
30

-------
3-1-2006
Snoeij, N.J., A.H. Penninks and W. Seinen. 1988. Dibutyltin and tributyltin compounds induce
thymus atrophy in rats due to a selective action on thymic lymphoblasts. Int. J.
Immunopharmacol. 10:891-899.
U.S. EPA. 1988. Recommendations for and Documentation of Biological Values for Use in
Risk Assessment. Environmental Criteria and Assessment Office, Office of Health and
Environmental Assessment, Cincinnati, OH. NTIS PB88-17874. EPA/600/6-87/008.
U.S. EPA. 1991. Chemical Assessments and Related Activities (CARA). Office of Health and
Environmental Assessment, Washington, DC. April.
U.S. EPA. 1994. Chemical Assessments and Related Activities (CARA). Office of Health and
Environmental Assessment, Washington, DC. December.
U.S. EPA. 1997a. Health Effects Assessment Summary Tables. Annual Update. FY-1997
Update. Prepared by the Office of Research and Development, National Center for
Environmental Assessment, Cincinnati, OH, for the Office of Emergency and Remedial
Response, Washington, DC. July. EPA-540-R-97-036. PB97-921199.
U.S. EPA. 1997b. Toxicological Review for Tributyltin Oxide (CAS No. 56-35-9) in Support of
Summary Information on the Integrated Risk Information System. Washington, DC. July.
Online, http://www.epa.gov/iris/toxreviews/0349-tr.pdf
U.S. EPA. 2002. 2002 Edition of the Drinking Water Standards and Health Advisories. Office
of Water, Washington, DC. EPA 822-R-02-038. Washington, DC.
http://www.epa. gov/waterscience/drinking/standards/dwstandards .pdf
U.S. EPA. 2006. Integrated Risk Information System (IRIS). Office of Research and
Development, National Center for Environmental Assessment, Washington, DC. Online.
http://www.epa.gov/iris
Vos, J.G., A. DeKlerk, E.I. Krajnc et al. 1990. Immunotoxicity of bis(tri-n-butyltin)oxide in the
rat: effects on thymus- dependent immunity and on nonspecific resistance following long-term
exposure in young versus aged rats. Toxicol. Appl. Pharmacol. 105:144-155.
WHO (World Health Organization). 1980. Tin and Organotin Compounds. A preliminary
review. Environmental Health Criteria 15. World Health Organization, Geneva.
WHO (World Health Organization). 1990. Tributyltin Compounds. Environmental Health
Criteria 116. Geneva, Switzerland. Online.
http ://www. inchem.org/ documents/ ehc/ ech/ehc 116 .htm
31

-------
3-1-2006
WHO (World Health Organization). 1993. Dibutyltindichloride. Poison Information
Monograph G586. Geneva, Switzerland. Online.
http://www.inchem.org/documents/pims/chemical/pim586.htm
WHO (World Health Organization). 1994. Tributyltin Compounds. Poison Information
Monograph GO 18. Geneva, Switzerland. Online.
http://www.inchem.org/documents/pims/chemical/pimgO 18 .htm
WHO (World Health Organization). 1999. Tributyltin Oxide. Consise International Chemical
Assessment Document 14. Geneva, Switzerland. Online.
http ://www. inchem.org/ documents/ cicads/cicads/ cicad 14 .htm
32

-------
7-6-2006
Provisional Peer Reviewed Toxicity Values for
Mono-, Di- and Tri- Butyltin Compounds
(Various CASRN)
Derivation of Subchronic and Chronic Inhalation RfCs
Superfund Health Risk Technical Support Center
National Center for Environmental Assessment
Office of Research and Development
U.S. Environmental Protection Agency
Cincinnati, OH 45268

-------
Acronyms and Abbreviations
bw	body weight
cc	cubic centimeters
CD	Caesarean Delivered
CERCLA	Comprehensive Environmental Response, Compensation and Liability Act
of 1980
CNS	central nervous system
cu.m	cubic meter
DWEL	Drinking Water Equivalent Level
FEL	frank-effect level
FIFRA	Federal Insecticide, Fungicide, and Rodenticide Act
g	grams
GI	gastrointestinal
HEC	human equivalent concentration
Hgb	hemoglobin
i.m.	intramuscular
i.p.	intraperitoneal
i.v.	intravenous
IRIS	Integrated Risk Information System
IUR	inhalation unit risk
kg	kilogram
L	liter
LEL	lowest-effect level
LOAEL	lowest-observed-adverse-effect level
LOAEL(ADJ)	LOAEL adjusted to continuous exposure duration
LOAEL(HEC)	LOAEL adjusted for dosimetric differences across species to a human
m	meter
MCL	maximum contaminant level
MCLG	maximum contaminant level goal
MF	modifying factor
mg	milligram
mg/kg	milligrams per kilogram
mg/L	milligrams per liter
MRL	minimal risk level
1

-------
MTD
maximum tolerated dose
MTL
median threshold limit
NAAQS
National Ambient Air Quality Standards
NOAEL
no-observed-adverse-effect level
NOAEL(ADJ)
NOAEL adjusted to continuous exposure duration
NOAEL(HEC)
NOAEL adjusted for dosimetric differences across species to a human
NOEL
no-observed-effect level
OSF
oral slope factor
p-IUR
provisional inhalation unit risk
p-OSF
provisional oral slope factor
p-RfC
provisional inhalation reference concentration
p-RfD
provisional oral reference dose
PBPK
physiologically based pharmacokinetic
PPb
parts per billion
ppm
parts per million
PPRTV
Provisional Peer Reviewed Toxicity Value
RBC
red blood cell(s)
RCRA
Resource Conservation and Recovery Act
RDDR
Regional deposited dose ratio (for the indicated lung region)
REL
relative exposure level
RfC
inhalation reference concentration
RfD
oral reference dose
RGDR
Regional gas dose ratio (for the indicated lung region)
s.c.
subcutaneous
SCE
sister chromatid exchange
SDWA
Safe Drinking Water Act
sq.cm.
square centimeters
TSCA
Toxic Substances Control Act
UF
uncertainty factor
Hg
microgram
|j,mol
micromoles
voc
volatile organic compound
11

-------
7-6-2006
PROVISIONAL PEER REVIEWED TOXICITY VALUES FOR
MONO-, DI- AND TRI- BUTYLTIN COMPOUNDS (Various CASRN)
Derivation of a Subchronic and Chronic Inhalation RfC
Background
On December 5, 2003, the U.S. Environmental Protection Agency's (EPA's) Office of
Superfund Remediation and Technology Innovation (OSRTI) revised its hierarchy of human
health toxicity values for Superfund risk assessments, establishing the following three tiers as the
new hierarchy:
1.	EPA's Integrated Risk Information System (IRIS).
2.	Provisional Peer-Reviewed Toxicity Values (PPRTV) used in EPA's Superfund
Program.
3.	Other (peer-reviewed) toxicity values, including:
~	Minimal Risk Levels produced by the Agency for Toxic Substances and Disease
Registry (ATSDR),
~	California Environmental Protection Agency (CalEPA) values, and
~	EPA Health Effects Assessment Summary Table (HEAST) values.
A PPRTV is defined as a toxicity value derived for use in the Superfund Program when
such a value is not available in EPA's Integrated Risk Information System (IRIS). PPRTVs are
developed according to a Standard Operating Procedure (SOP) and are derived after a review of
the relevant scientific literature using the same methods, sources of data, and Agency guidance
for value derivation generally used by the EPA IRIS Program. All provisional toxicity values
receive internal review by two EPA scientists and external peer review by three independently
selected scientific experts. PPRTVs differ from IRIS values in that PPRTVs do not receive the
multi-program consensus review provided for IRIS values. This is because IRIS values are
generally intended to be used in all EPA programs, while PPRTVs are developed specifically for
the Superfund Program.
Because science and available information evolve, PPRTVs are initially derived with a
three-year life-cycle. However, EPA Regions (or the EPA HQ Superfund Program) sometimes
request that a frequently used PPRTV be reassessed. Once an IRIS value for a specific chemical
becomes available for Agency review, the analogous PPRTV for that same chemical is retired. It
should also be noted that some PPRTV manuscripts conclude that a PPRTV cannot be derived
based on inadequate data.
1

-------
7-6-2006
Disclaimers
Users of this document should first check to see if any IRIS values exist for the chemical
of concern before proceeding to use a PPRTV. If no IRIS value is available, staff in the regional
Superfund and RCRA program offices are advised to carefully review the information provided
in this document to ensure that the PPRTVs used are appropriate for the types of exposures and
circumstances at the Superfund site or RCRA facility in question. PPRTVs are periodically
updated; therefore, users should ensure that the values contained in the PPRTV are current at the
time of use.
It is important to remember that a provisional value alone tells very little about the
adverse effects of a chemical or the quality of evidence on which the value is based. Therefore,
users are strongly encouraged to read the entire PPRTV manuscript and understand the strengths
and limitations of the derived provisional values. PPRTVs are developed by the EPA Office of
Research and Development's National Center for Environmental Assessment, Superfund Health
Risk Technical Support Center for OSRTI. Other EPA programs or external parties who may
choose of their own initiative to use these PPRTVs are advised that Superfund resources will not
generally be used to respond to challenges of PPRTVs used in a context outside of the Superfund
Program.
Questions Regarding PPRTVs
Questions regarding the contents of the PPRTVs and their appropriate use (e.g., on
chemicals not covered, or whether chemicals have pending IRIS toxicity values) may be directed
to the EPA Office of Research and Development's National Center for Environmental
Assessment, Superfund Health Risk Technical Support Center (513-569-7300), or OSRTI.
INTRODUCTION
RfCs for monobutyltin, dibutyltin or tributyltin compounds are not listed on IRIS (U.S.
EPA, 2006) or the HEAST (U.S. EPA, 1997a). The CARA list (U.S. EPA, 1991, 1994a) does
not include any documents for butyltins. The ATSDR (2003) updated draft Toxicological Profile
for tin (which includes organotin compounds) does not establish inhalation MRLs for any
butyltin compound because of a lack of suitable data. ACGIH (2001, 2003), NIOSH (2003) and
OSHA (2003) have established occupational exposure limits (8-hour TWA) of 0.1 mg/m3, as Sn,
for organic tin compounds, including tributyltins, to protect against irritation of the eyes, skin and
respiratory tract, as well as potential adverse effects on immune function and the central nervous
system. A NIOSH (1976) Criteria Document for organotin compounds, Poison Information
Monographs on dibutyltin dichloride (WHO, 1993) and tributyltin compounds (WHO, 1994), an
Environmental Health Criteria document on tributyltin compounds (WHO, 1990) and a review of
2

-------
7-6-2006
the biological activity of organotin compounds (Snoeij et al., 1987) were consulted for relevant
information. In addition, the NTP (2003a) background document for testing of methyltin and
butyltin compounds, the management status documents for monobutyltin trichloride (NTP,
2003b) and dibutyltin diacetate (NTP, 2003c), and the health and safety report for dibutyltin
acetate (NTP, 2003d) were also consulted. IARC (2003) has not reviewed butyltin compounds.
In May,1992, literature searches were conducted in the following databases: TOXLINE (1965-
1992), CANCERLINE (1963-1992), CHEM ID, HSDB, RTECS and TSCATS. In March 1995,
update computer literature searches were conducted in: TOXLINE, MEDLINE, CANCERLINE,
TSCATS and RTECS. More recently, update literature searches were conducted on
monobutyltin, dibutyltin and tributyltin compounds for the period from 1994 to August 2003 in
the following databases: TOXLINE (including NTIS and BIOSIS updates), CANCERLIT,
MEDLINE, CCRIS, GENETOX, HSDB, EMIC/EMICBACK, DART/ETICBACK, RTECS and
TSCATS. An additional literature search was conducted through September 2004 which
produced no new data.
Tributyltin oxide is the only butyltin compound for which a toxicity assessment is
available on IRIS (U.S. EPA, 2006), but this did not include an RfC. Documents specific to
tributyltin oxide, a Toxicological Review (U.S. EPA, 1997b) and a Concise International
Chemical Assessment Document (WHO, 1999), were also consulted for this provisional value
assessment.
REVIEW OF THE PERTINENT LITERATURE
Human Studies
No relevant data were located regarding the toxicity of monobutyltins, dibutyltins or
tributyltins to humans following subchronic or chronic inhalation exposure.
Irritation of the upper respiratory tract and eye was reported following acute occupational
exposure to tributyltin oxide (ACGIH, 2001). Portal-of-entry effects following inhalation
exposure in humans would also be expected for other butyltin compounds for which acute eye
irritation or skin irritation has been reported, such as tributyltin chloride and dibutyltin dichloride
(ACGIH, 2001; WHO, 1993).
Animal Studies
The only relevant inhalation study in animals that was located was an unpublished 4-
week inhalation toxicity study of monobutyltin trichloride in rats (Biodynamics, 1988). Groups
of Sprague-Dawley rats (15/sex/concentration) were whole-body exposed to vapor/aerosol
atmospheres of 95% pure monobutyltin trichloride (MW = 282.1) at mean measured
3

-------
7-6-2006
concentrations of 3.4, 18, or 24 mg/m3 (1.3, 7.2, or 9.5 mg Sn/m3) for 6 hours/day, 5 days/week
for 4 weeks. In terms of monobutyltin (MW = 175.74), the exposure levels correspond to 2.1,
11.2 and 15.0 mg/kg-day, respectively. Controls were chamber-exposed to room air. The test
atmospheres were generated from the liquid test material using a bubbler with in-line trap. The
measured concentrations were much lower than the corresponding nominal values calculated
based on amount of material added to the system, and the difference increased dramatically with
concentration, apparently due to problems with formation of the aerosol and chemical reaction of
the test material within the chamber. Measurement of the aerosols within the exposure chamber
showed that a high percentage of test material in the chamber was in the form of aerosol in the
low- and mid- exposure groups; the percentage was considerably lower in the high-exposure
group. The mass median aerodynamic diameter MMAD (and geometric standard deviation
GSD) of the aerosols in the exposure chamber were 0.91 |j,m (1.6), 0.98 |j,m (4.1), 1.7 |j,m (2.3),
and 1.5 |j,m (2.0) in the control, low-, mid-, and high-exposure groups, respectively, indicating
that particles in the chamber were respirable. Rats were evaluated twice daily for mortality and
clinical signs, and received a weekly detailed physical examination for abnormal signs. Body
weights were recorded before testing and weekly thereafter. Ophthalmoscopic examinations
were conducted before testing and at termination. After 4 weeks of treatment, 15 rats/sex/group
were sacrificed; of these, 10 were evaluated for hematology, clinical chemistry, gross pathology,
organ weights (adrenals, brain, kidneys, liver ovaries and testes) and histopathology (more than
30 tissues examined in the control and high-concentration groups; only tissues with gross lesions
examined in the low- and mid-dose groups). Overnight urine samples were collected for
urinalysis from the other 5 rats, which were subsequently used for analysis of tissue tin content.
Mortality was observed in 3 males and 1 female at the highest concentration; deaths
occurred after 13-15 days of exposure (Biodynamics, 1988). Deaths were considered to be
treatment-related by the researchers, although cause of death could not be identified from gross
necropsy. Clinical signs in high-concentration rats during the 4-week exposure period included
mucoid nasal discharge, rales, lacrimation, salivation, rough coat, ano-genital staining,
discoloration of the fur, and (in males only) abdominal distension. Statistically significant
reductions in mean terminal body weights were seen in both males (-7%) and females (-5%) in
the highest exposure group, compared to the controls. Hematological evaluation revealed no
effects in treated males and only slight changes in females (small statistical increases in
hemoglobin and hematocrit in the high-exposure group and erythrocyte counts in all exposed
groups). There were no exposure-related ophthalmoscopic, clinical biochemistry or urinalysis
findings, or effects on organ weights. The primary gross lesion was discoloration of the lungs,
observed in most treated males (77-90% of each group) and females (80-100% of each group),
but not in controls. Histopathological examination revealed amorphous material (hypothesized
by the researchers to be the test material and/or its hydrolysis product monobutyltin dihydroxy
chloride) in the alveolar sacs of treated rats (7/10, 9/10, and 9/9 males and 9/10, 8/10, and 10/10
females in the low-to-high concentration groups, respectively). This was accompanied in many
cases by alveolar edema (6/10, 7/10 and 3/9 males and 3/10, 6/10 and 3/10 females in the low-to-
4

-------
7-6-2006
high concentration groups, respectively) and in some cases by subacute bronchopneumonia (2/10,
2/10 and 1/9 males and 0/10, 1/10 and 0/10 females, respectively). Severity of the pulmonary
lesions was similar in all treated groups. None of these lesions were observed in controls. Other
than the increase above controls, a dose-related pattern was not observed. Lung tin burdens in
the mid- and high-level groups were similar, and considerably higher than the low-level group.
Histological findings in other tissues were generally unremarkable, although there was some
evidence for an effect on the nasal turbinates (purulent exudate observed in the anterior section of
the nasal cavity in 5/9 males and 3/10 females, but not in controls) and the skin (epidermal
acanthosis and hyperkeratosis observed in 8/8 males and 5/8 females, but not controls) in the
high-exposure group (neither of these tissues were examined in the low- or mid-level groups).
The authors of the study (Biodynamics, 1988) described the lesions as the expected response of
lung tissue to the introduction of foreign corrosive material. The EPA agrees with this conclusion
but considers the resulting effects to be adverse. The observed alveolar edema and
bronchopneumonia is most likely a result of localized hypoxia to cells covered by the foreign
material. This effect would also be expected in humans provided the tissue coverage was similar
to that in the rats. However, the effects are likely dominated by the physical processes and less
affected by interspecies or inter-individual physiological differences, consideration of which will
reduce the overall uncertainty factor applied to the RfC (see RfC derivation section following).
The lowest concentration of monobutyltin trichloride, 3.4 mg/m3 (2.1 mg/m3 as monobutyltin), is
determined to be a LOAEL for pulmonary lesions in male and female rats. A NOAEL is not
identified in this study.
DERIVATION OF PROVISIONAL SUBCHRONIC AND CHRONIC RfCs FOR
BUTYLTIN COMPOUNDS
Monobutyltin Compounds
No subchronic or chronic toxicity data are available for humans exposed to butyltins by
inhalation. The only available animal study identified a LOAEL of 2.1 mg/m3 for monobutyltin
in rats exposed intermittently for 4 weeks, based on development of pulmonary lesions
(Biodynamics, 1988). The data from this study were not amenable to benchmark concentration
dose modeling, because incidence of lesions was high in the low-level group, and incidence and
severity of effects were similar at higher exposure levels. Therefore, the LOAEL/NOAEL
approach was used.
A provisional subchronic RfC for monobutyltin can be derived from the Biodynamics
(1988) study using the methodology for aerosols presented in U.S. EPA (1994b). Although the
exposure atmosphere included both vapors and aerosols of monobutyltin trichloride, the
atmosphere at the LOAEL of 2.1 mg/m3 (as monobutyltin) consisted largely of aerosols. In order
to derive the provisional subchronic RfC, the LOAEL of 2.1 mg/m3 is adjusted for continuous
5

-------
7-6-2006
exposure based on the exposure protocol (2.1 mg/m3 x 6/24 hr x 5/7 d = 0.38 mg/m3) to obtain a
duration-adjusted LOAEL (LOAELadj) of 0.38 mg/m3. The LOAELadj is multiplied by the
RDDR (regional deposited dose ratio) to calculate the human equivalent concentration
(LOAELm < ). The RDDR for pulmonary effects in rats (average body weight of 353 g used,
based on males at the LOAEL) for particles with MMAD = 0.98 |im and o = 4.1 (the mean
values at the LOAEL) is 0.335. The LOAE!^^ is 0.38 mg/m3 x 0.335 = 0.13 mg/m3. An
uncertainty factor (UF) of 300 was calculated from factors of 10 for use of a LOAEL, 3 (10°5) to
protect sensitive individuals, and 10 for deficiencies in the database, including the lack of
reproductive, developmental, or supporting systemic studies. An additional 3-fold factor for
interspecies toxicodynamic uncertainty was deemed to be unnecessary because of the
predominantly physical nature of the effect. The human inter-individual uncertainty factor was
reduced by half-an-order of magnitude, because toxicokinetic uncertainty is minimal. Dividing
the LOAELjjgc of 0.13 mg/m3 by the UF of 300 produces a provisional subchronic RfC of 4E-4
(4 x 10"4) mg/m3 for monobutyltin trichloride. A provisional chronic RfC was not derived due to
the short exposure duration of the Biodynamics (1988) study.
Confidence in the key study for the provisional subchronic RfC is medium. The study
included adequate numbers of animals and dose groups, and examined a wide range of endpoints,
including systemic and portal-of-entry effects. However, exposure duration was short, there were
evident problems generating the test atmospheres, reporting of methods and results was unclear
(particularly with regard to exposure concentrations experienced by test animals), and exposure
levels were too high (neither a NOAEL nor a dose-response was identified). Confidence in the
database is low because developmental, reproductive, and supporting systemic studies are
unavailable, yielding a low confidence in the subchronic p-RfC.
Di- and Tri- Butyltin Compounds
Derivation of provisional subchronic or chronic RfCs is not feasible for dibutyltin or
tributyltin compounds due to the absence of relevant data.
REFERENCES
ACGIH (American Conference of Government Industrial Hygienists). 2001. Tin, Organic
Compounds. Documentation of the Threshold Limit Values (TLV) and Biological Exposure
Indices. 7th Edition. Cincinnati, OH.
ACGIH (American Conference of Governmental Industrial Hygienists). 2003. 2003 TLVs and
BEIs Based on the Documentation of the Threshold Limit Values for Chemical Substances and
Physical Agents and Biological Exposure Indices. Cincinnati, OH. p. 56.
6

-------
7-6-2006
ATSDR (Agency for Toxic Substances and Disease Registry). 2003. Toxicological Profile for
Tin. Update Draft for Public Comment. Public Health Service. Atlanta, GA.
Biodynamics. 1988. Four-Week Inhalation Toxicity Study with Monobutyltin Trichloride in the
Rat with a Recovery Period. Produced 1/14/88. Submitted 2/12/88 by M & T Chemical, Inc. to
U.S. EPA under TSCA section 8D. EPA Doc. No. 86-880000133. FicheNo. OTS0514023.
TSCATS 305212.
I ARC (International Agency for Research on Cancer). 2003. IARC Agents and Summary
Evaluations. Online, http://www-cie.iarc.fr/htdig/search.html
NIOSH (National Institute for Occupational Safety and Health). 1976. NIOSH Criteria for a
Recommended Standard...Occupational Exposure to Organotin Compounds. DHHS (NIOSH)
Publication Nol. 77-115. NTIS PB-274 766. Online, http://www.cdc.gov/niosh/77-l 15.html
NIOSH (National Institute for Occupational Safety and Health). 2003. Tin (organic compounds,
as Sn). NIOSH Pocket Guide to Chemical Hazards. Online.
http://www.cdc.gov/niosh/ngp/npgd0614.html
NTP (National Toxicology Program). 2003a. Nomination background document for organotin
(methyl and butyl) toxicity. Online.
http://ntp-server.niehs.nih.gov/htdocs/Chem Background/ExSumPdf/Organotins.pdf
NTP (National Toxicology Program). 2003b. Monobutyltin trichloride. Management Status
Report. Online. http://ntp-server.niehs.nih.gov/htdocs/Results Status/Resstatm/M000066.Html
NTP (National Toxicology Program). 2003c. Dibutyltin diacetate. Management Status Report.
Online. http://ntp-server.niehs.nih.gov/htdocs/Results Status/Resstatd/10670-W.Html
NTP (National Toxicology Program). 2003d. Dibutyltin diacetate. Health and Safety Report.
Online. http://ntp-server.niehs.nih.gov/htdocs/CHEM H&S/NTP Chem 1 /Radian 1067-33-
O.html
OSHA (Occupational Safety and Health Administration). 2003. Chemical sampling
information: Tin, organic compounds (as Sn). Online.
http://www.osha.gov/dts/chemicalsampling/data/CH 271900.html
Snoeij, N.J., A.H. Penninks and W. Seinen. 1987. Biological activity of organotin compounds-
an overview. Environ. Res. 44:335-353.
7

-------
7-6-2006
U.S. EPA. 1988. Recommendations for and Documentation of Biological Values for Use in
Risk Assessment. Environmental Criteria and Assessment Office, Office of Health and
Environmental Assessment, Cincinnati, OH. NTIS PB88-17874. EPA/600/6-87/008.
U.S. EPA. 1991. Chemical Assessments and Related Activities (CARA). Office of Health and
Environmental Assessment, Washington, DC. April.
U.S. EPA. 1994a. Chemical Assessments and Related Activities (CARA). Office of Health and
Environmental Assessment, Washington, DC. December.
U.S. EPA. 1994b. Methods for Derivation of Inhalation Reference Concentrations and
Application of Inhalation Dosimetry. Office of Research and Development, Washington, DC.
October. EPA/600/8-90/066F.
U.S. EPA. 1997a. Health Effects Assessment Summary Tables. Annual Update. FY-1997
Update. Prepared by the Office of Research and Development, National Center for
Environmental Assessment, Cincinnati, OH, for the Office of Emergency and Remedial
Response, Washington, DC. July. EPA-540-R-97-036. PB97-921199.
U.S. EPA. 1997b. Toxicological Review for Tributyltin Oxide (CAS No. 56-35-9) in Support of
Summary Information on the Integrated Risk Information System. Washington, DC. July.
Online, http://www.epa.gov/iris/toxreviews/0349-tr.pdf
U.S. EPA. 2006. Integrated Risk Information System (IRIS). Office of Research and
Development, National Center for Environmental Assessment, Washington, DC. Online.
http://www.epa.gov/iris/
WHO (World Health Organization). 1990. Tributyltin Compounds. Environmental Health
Criteria 116. Geneva, Switzerland. Online.
http ://www. inchem.org/documents/ehc/ech/ehc 116 .htm
WHO (World Health Organization). 1993. Dibutyltindichloride. Poison Information
Monograph G586. Geneva, Switzerland. Online.
http://www.inchem.org/documents/pims/chemical/pim586.htm
WHO (World Health Organization). 1994. Tributyltin Compounds. Poison Information
Monograph GO 18. Geneva, Switzerland. Online.
http://www.inchem.org/documents/pims/chemical/pimgQ18.htm
8

-------
7-6-2006
WHO (World Health Organization). 1999. Tributyltin Oxide. Consise International Chemical
Assessment Document 14. Geneva, Switzerland. Online.
http ://www. inchem.org/documents/cicads/cicads/cicad 14 .htm
9

-------
6-12-2006
Provisional Peer Reviewed Toxicity Values for
Mono-, Di- and Tri- Butyltin Compounds
(Various CASRN)
Derivation of a Carcinogenicity Assessment
Superfund Health Risk Technical Support Center
National Center for Environmental Assessment
Office of Research and Development
U.S. Environmental Protection Agency
Cincinnati, OH 45268

-------
Acronyms and Abbreviations
bw	body weight
cc	cubic centimeters
CD	Caesarean Delivered
CERCLA	Comprehensive Environmental Response, Compensation and Liability Act
of 1980
CNS	central nervous system
cu.m	cubic meter
DWEL	Drinking Water Equivalent Level
FEL	frank-effect level
FIFRA	Federal Insecticide, Fungicide, and Rodenticide Act
g	grams
GI	gastrointestinal
HEC	human equivalent concentration
Hgb	hemoglobin
i.m.	intramuscular
i.p.	intraperitoneal
i.v.	intravenous
IRIS	Integrated Risk Information System
IUR	inhalation unit risk
kg	kilogram
L	liter
LEL	lowest-effect level
LOAEL	lowest-observed-adverse-effect level
LOAEL(ADJ)	LOAEL adjusted to continuous exposure duration
LOAEL(HEC)	LOAEL adjusted for dosimetric differences across species to a human
m	meter
MCL	maximum contaminant level
MCLG	maximum contaminant level goal
MF	modifying factor
mg	milligram
mg/kg	milligrams per kilogram
mg/L	milligrams per liter
MRL	minimal risk level
1

-------
MTD
maximum tolerated dose
MTL
median threshold limit
NAAQS
National Ambient Air Quality Standards
NOAEL
no-observed-adverse-effect level
NOAEL(ADJ)
NOAEL adjusted to continuous exposure duration
NOAEL(HEC)
NOAEL adjusted for dosimetric differences across species to a human
NOEL
no-observed-effect level
OSF
oral slope factor
p-IUR
provisional inhalation unit risk
p-OSF
provisional oral slope factor
p-RfC
provisional inhalation reference concentration
p-RfD
provisional oral reference dose
PBPK
physiologically based pharmacokinetic
PPb
parts per billion
ppm
parts per million
PPRTV
Provisional Peer Reviewed Toxicity Value
RBC
red blood cell(s)
RCRA
Resource Conservation and Recovery Act
RDDR
Regional deposited dose ratio (for the indicated lung region)
REL
relative exposure level
RfC
inhalation reference concentration
RfD
oral reference dose
RGDR
Regional gas dose ratio (for the indicated lung region)
s.c.
subcutaneous
SCE
sister chromatid exchange
SDWA
Safe Drinking Water Act
sq.cm.
square centimeters
TSCA
Toxic Substances Control Act
UF
uncertainty factor
Hg
microgram
|j,mol
micromoles
voc
volatile organic compound
11

-------
6-12-2006
PROVISIONAL PEER REVIEWED TOXICITY VALUES FOR
MONO-, DI- AND TRI- BUTYLTIN COMPOUNDS (Various CASRN)
Derivation of a Carcinogenicity Assessment
Background
On December 5, 2003, the U.S. Environmental Protection Agency's (EPA's) Office of
Superfund Remediation and Technology Innovation (OSRTI) revised its hierarchy of human
health toxicity values for Superfund risk assessments, establishing the following three tiers as the
new hierarchy:
1.	EPA's Integrated Risk Information System (IRIS).
2.	Provisional Peer-Reviewed Toxicity Values (PPRTV) used in EPA's Superfund
Program.
3.	Other (peer-reviewed) toxicity values, including:
~	Minimal Risk Levels produced by the Agency for Toxic Substances and Disease
Registry (ATSDR),
~	California Environmental Protection Agency (CalEPA) values, and
~	EPA Health Effects Assessment Summary Table (HEAST) values.
A PPRTV is defined as a toxicity value derived for use in the Superfund Program when
such a value is not available in EPA's Integrated Risk Information System (IRIS). PPRTVs are
developed according to a Standard Operating Procedure (SOP) and are derived after a review of
the relevant scientific literature using the same methods, sources of data, and Agency guidance
for value derivation generally used by the EPA IRIS Program. All provisional toxicity values
receive internal review by two EPA scientists and external peer review by three independently
selected scientific experts. PPRTVs differ from IRIS values in that PPRTVs do not receive the
multi-program consensus review provided for IRIS values. This is because IRIS values are
generally intended to be used in all EPA programs, while PPRTVs are developed specifically for
the Superfund Program.
Because science and available information evolve, PPRTVs are initially derived with a
three-year life-cycle. However, EPA Regions (or the EPA HQ Superfund Program) sometimes
request that a frequently used PPRTV be reassessed. Once an IRIS value for a specific chemical
becomes available for Agency review, the analogous PPRTV for that same chemical is retired. It
should also be noted that some PPRTV manuscripts conclude that a PPRTV cannot be derived
based on inadequate data.
1

-------
6-12-2006
Disclaimers
Users of this document should first check to see if any IRIS values exist for the chemical
of concern before proceeding to use a PPRTV. If no IRIS value is available, staff in the regional
Superfund and RCRA program offices are advised to carefully review the information provided
in this document to ensure that the PPRTVs used are appropriate for the types of exposures and
circumstances at the Superfund site or RCRA facility in question. PPRTVs are periodically
updated; therefore, users should ensure that the values contained in the PPRTV are current at the
time of use.
It is important to remember that a provisional value alone tells very little about the
adverse effects of a chemical or the quality of evidence on which the value is based. Therefore,
users are strongly encouraged to read the entire PPRTV manuscript and understand the strengths
and limitations of the derived provisional values. PPRTVs are developed by the EPA Office of
Research and Development's National Center for Environmental Assessment, Superfund Health
Risk Technical Support Center for OSRTI. Other EPA programs or external parties who may
choose of their own initiative to use these PPRTVs are advised that Superfund resources will not
generally be used to respond to challenges of PPRTVs used in a context outside of the Superfund
Program.
Questions Regarding PPRTVs
Questions regarding the contents of the PPRTVs and their appropriate use (e.g., on
chemicals not covered, or whether chemicals have pending IRIS toxicity values) may be directed
to the EPA Office of Research and Development's National Center for Environmental
Assessment, Superfund Health Risk Technical Support Center (513-569-7300), or OSRTI.
INTRODUCTION
Tributyltin oxide (CASRN 56-35-9) is the only butyltin compound for which there is a
carcinogenicity assessment on IRIS (U.S. EPA, 1997a, 2006). In this assessment (consensus
review date 07/02/1997), tributyltin oxide is assigned to U.S. EPA (2005) cancer weight-of-
evidence category "Inadequate Information to Assess Carcinogenic Potential. " Tributyltin
oxide is not mutagenic to bacteria, but yields positive results in mammalian systems in vitro and
in vivo. Since an assessment is available on IRIS, tributyltin oxide is not considered further in
this issue paper. However, some documents on this compound were consulted for possible
information on other butyltin compounds: the IRIS summary sheets (U.S. EPA, 2006) and
Toxicological Review (U.S. EPA, 1997a), and a Concise International Chemical Assessment
Document (WHO, 1999).
2

-------
6-12-2006
No carcinogenicity assessments are available for monobutyltin, dibutyltin, or tributyltin
compounds (other than tributyltin oxide) on IRIS (U.S. EPA, 2006), the HEAST (U.S. EPA,
1997b), or the Drinking Water Standards and Health Advisories list (U.S. EPA, 2002). The
CARA database (U.S. EPA, 1991, 1994) does not list any document covering organotin
compounds. A NIOSH (1976) Criteria Document for organotin compounds, a draft of the
ATSDR (2003) updated draft Toxicological Profile for tin (which includes organotin
compounds), Poison Information Monographs on dibutyltin dichloride (WHO, 1993) and tributyl
tin compounds (WHO, 1994), an Environmental Health Criteria document on tributyl tin
compounds (WHO, 1990) and a review of the biological activity of organotin compounds (Snoeij
et al., 1987) were consulted for relevant information. In addition, the NTP (2003a) background
document for testing of methyltin and butyl tin compounds, the management status documents
for monobutyltin trichloride (NTP, 2003b) and dibutyltin diacetate (NTP, 2003c) and the health
and safety report for dibutyltin acetate (NTP, 2003d) were also consulted. IARC (2003) has not
reviewed organotin compounds. In August 1992, computer literature searches of TOXLIT
(1965-1992), TOXLINE (1965-1992), CHEM ID, RTECS (through August, 1992), and TSCATS
databases were conducted for monobutyltin oxide and dibutyltin oxide. In March 1995, update
literature searches of TOXLINE, MEDLINE, EMIC, HSDB, DART, and RTECS were
performed for dibutyltin and tributyltin compounds. More recently, update literature searches
were conducted for the period from 1994 to August 2003 for monobutyltin, dibutyltin and
tributyltin compounds in the following databases: TOXLINE (including NTIS and BIOSIS
updates), CANCERLIT, MEDLINE, CCRIS, GENETOX, HSDB, EMIC/EMICBACK,
DART/ETICBACK, RTECS and TSCATS. An additional literature search was conducted
through September 2004 which produced no new data.
Monosubstituted organotins have had limited application as stabilizers in PVC films.
Dialkylorganotin compounds such as dibutyltin are used in the chemical industry as heat
stabilizers in the production of PVC, curing agents for silicon rubber, and catalysts in the
production of polyurethane. Tributyltin compounds are used mainly for their biocidal properties
as molluscicides, fungicides, insecticides and miticides. Tetrasubstituted organotin compounds
are mainly used as intermediates in the preparation of other organotin compounds (Boyer, 1989;
Bulten and Meinema, 1991; Magos, 1986; Nicklin and Robson, 1988; NIOSH, 1976; WHO,
1980).
REVIEW OF THE PERTINENT LITERATURE
Human Studies
No data were located on the carcinogenicity of mono-, di- or tri- butyltin compounds in
humans.
3

-------
6-12-2006
Animal Studies
Monobutyltin Compounds
No data were located on the carcinogenicity of monobutyltin compounds in animals.
Dibutyltin Compounds
Carcinogenicity data for dibutyltin compounds in animals are limited to oral-exposure
studies for dibutyltin acetate in rats and mice (NCI, 1979).
Fischer 344 rats (50/sex/dose group) received dibutyltin diacetate at time-weighted
average dietary levels of 66.5 or 133 ppm for 78 weeks, and then received control diets for a
period of 26 weeks (NCI, 1979). Twenty animals/sex served as controls. Based on approximate
average body weights in the study of 0.350 kg for males and 0.225 kg for females, and using a
food consumption rate calculated as described in U.S. EPA (1988), it can be estimated that males
ingested 0, 5.3, or 10.6 mg/kg-day and females ingested 0, 6.2, or 12.3 mg/kg-day. Endpoints
examined included body weight and food consumption throughout the study, and gross and
microscopic examination of major tissues and organs at sacrifice and from animals that died
early. Body weights of high-dose male rats were lower than controls throughout the experiment,
but statistical analysis was not reported; body weights in females were not affected by treatment
with dibutyltin diacetate. Survival was significantly reduced in high-dose male rats (apparently
due to pneumonia), but remained adequate for assessment of risk due to late-developing tumors
(26/50 survived to termination). Survival in treated females was not significantly lower than
controls, and was adequate for assessment of late-developing tumors (32/50 survived to
termination in the high-dose group and 42/50 in the low-dose group). However, tissues from 17
out of 50 high-dose females were lost, limiting the adequacy of the evaluation in this group. The
only tumorigenic response of note in rats was an apparent increase in neoplasms of the uterus in
low-dose females, that did not, however, achieve statistical significance. The incidence of
uterine neoplasms was 1/19 (5%), 10/49 (20%), and 2/33 (6%) in the control, low-dose, and
high-dose groups, respectively. Uterine neoplasms included adenocarcinomas, leiomyomas,
endometrial stromal polyps, and hemangiomas. The lack of increase in the high-dose group is
confounded by tissues that were lost from 17/50 high-dose females. Of these 17 animals, 5 were
noted as having undetermined uterine tumors on the basis of gross observation. NCI (1979)
concluded that there was no evidence for carcinogenicity in male rats, and that the study in
female rats was inadequate because tissues from many high-dose rats were not analyzed.
In the mouse study, B6C3F1 mice (50/sex/dose level) were treated with dibutyltin
diacetate at time-weighted average dietary levels of 76 or 152 ppm for 78 weeks, and then
received control diets for an additional 14 weeks (NCI, 1979). A control group was composed of
20 mice/sex. Based on estimated average body weights of 0.037 kg for males and 0.034 kg for
females in this study, and using a food consumption rate calculated as described in U.S. EPA
4

-------
6-12-2006
(1988), the doses were estimated as 0, 13.0, or 26.0 mg/kg-day for male mice and 0, 13.4, or 26.8
mg/kg-day for female mice. The endpoints examined were the same as in the rat study. In mice,
body weight gain did not appear to be significantly affected by treatment with dibutyltin
diacetate. Survival was significantly reduced in high-dose female mice starting at about week 45
of the study, but remained adequate for evaluation of late-developing tumors (29/50 survived to
study termination). The cause of death in high-dose females was not discussed. The study found
marginal evidence for a treatment-related increase in liver tumors (see Table 1). The incidence
of hepatocellular adenomas or carcinomas (combined) appeared to be increased in treated males,
but the differences from control were not statistically significant. Females did not have
carcinomas. There was a significant trend for hepatocellular adenomas in females, but only a
marginal pairwise increase in the high-dose group. NCI (1979) determined that under the
conditions of the study, there was no conclusive evidence for the carcinogenicity of dibutyltin
diacetate in male or female mice.
Tumor
Table 1. The Incidence of Liver Tumors in Mice Studied by NCI (1979)
Dose Group
Control	Low	High
Male
Hepatocellular adenoma	2/19(11%)	9/49(18%)	13/49 (27%)
Hepatocellular carcinoma	0/19(0%)	2/49(4%)	2/49(4%)
Hepatocellular adenoma or carcinoma	2/19 (11%)	11/49 (22%)	15/49 (31%)
Female
Hepatocellular adenoma	1/20 (5%)a	4/47(9%)	12/43 (28%)b
b
' significant trend (p=0.006) by Cochran-Armitage test (reported by NCI, 1979)
marginally significant pairwise increase (p=0.03) by Fisher Exact test that is not significant after Bonferroni
correction for multiple comparisons (reported by NCI, 1979)
5

-------
6-12-2006
Tributyltin Compounds
Aside from the oral studies on tributyltin oxide, the only information on carcinogenicity
of tributyltin compounds in animals comes from a dermal exposure study by Sheldon (1975).
Sheldon (1975) applied tributyltin fluoride to the shaved backs of male Swiss albino mice
(50/dose group) three times per week for 6 months. Two treated groups were included: one that
received 15 mg of a 10% solution of the compound in propylene glycol for 26 weeks and one that
received a 30% solution for the first 3 weeks of the study, but had the concentration lowered
to5% for the remaining 23 weeks due to irritant effects. A control group received the solvent
alone, and a positive control group was treated with a known carcinogen identified as R-911-10
in propylene glycol. Ten animals treated with 30% and then 5% solution showed hyperplastic
skin changes, attributed by the authors to the irritant effects of the 30% solution. No neoplastic
changes were seen in this group. Mice treated with the 10% solution showed no neoplastic or
nonneoplastic skin lesions. The incidence of skin lesions in the positive control group was 56%,
86% of which were neoplastic. Negative controls had no lesions.
Other Studies
Toxicokinetics
Data summarized by Boyer (1989) indicate that alkyltin compounds are metabolized
mainly in the liver by the P-450 monooxygenase system. Tributyltin acetate is metabolized by
isolated rat microsomes to form alpha-, beta-, gamma- and delta-hydroxybutyldibutyltin
derivatives, as well as 1-butanol and 1-butane. Further oxidation of the gamma-hydroxy
compound yields the corresponding ketone. Tetrabutyltin incubated with the liver microsome
fraction produces tributyltin derivatives; similarly, dibutyltin diacetate produces monobutyltin
derivatives. Several of the metabolites that are produced in vitro have also been detected in the
liver and feces of mice exposed to tributyltin acetate or dibutyltin diacetate by gavage. In rats, an
initial increase in tributyltin observed in the liver after exposure to tributyltin fluoride by gavage
was followed by an increase in dibutyltin, monobutyltin and inorganic tin. Biliary excretion
represents the main route of excretion of butyltin compounds.
Ueno et al. (1997) examined the hepatic metabolism of di- and tributyltin chlorides in
male mice exposed by gavage. Three hours after exposure to 0.18 mM/kg tributyltin chloride,
the main hepatic metabolites were dibutyltin dichloride (40%) and dibuty(3-carboxylpropyl)tin
(12%>); by 24 hours the content had changed to 36.4%) and 25.6%, respectively. In contrast, 24
hours after treatment with dibutyltin chloride, most of the hepatic butyltin was the parent
compound (94.8%). Pretreating mice with an inhibitor to cytochrome P-450 significantly
reduced hepatotoxicity caused by exposure to tributyltin chloride, but did not affect the hepatic
toxicity caused by exposure to dibutyltin chloride. The authors suggested that dibutyltin is the
primary agent for hepatotoxicity in mice.
6

-------
6-12-2006
In an in vitro study, Ohhira et al. (2003) compared the metabolism of tributyltin by
hepatic microsomes derived from rats, hamsters, male humans, and female humans. Under
comparable reaction conditions, rat hepatic microsomes exhibited the highest dealkylation and
dearylation activity compared to the other samples. The percentage of total metabolites
(dibutyltin, monobutyltin, and inorganic tin) to parent tributyltin was 214, 55.1, 11.4 and 27.6,
respectively, for rat, hamster, male human, and female human hepatic microsomes. The authors
suggested that the hamster is more appropriate than the rat as a model for evaluating tributyltin
effects in humans.
Tributyltin compounds may reduce cellular levels of glutathione, increasing susceptibility
of cells to oxidative stress. Following a 15 minute incubation, tri-n-butyltin chloride, but not
tetrabutyltin chloride, at concentrations of >3 nM, reduced the cellular content of glutathione in
cultured rat thymocytes (Okada et al., 2000). At a concentration of 300 nM, the glutathione
content was nearly depleted.
Genotoxicity
Mono-, di-, and tri- butyltin compounds are mutagenic in some bacterial systems.
Dibutyltin diacetate was not mutagenic in Salmonella typhimurium strains TA100, TA1535,
TA1537, or TA98 in the presence or absence of metabolic activation from induced rats or
hamsters (Boyer, 1989). Using a modified assay for mutagenicity in S. typhimurium TA100,
significant increases in the number of revertants were observed for mono-n-butyltin oxide, n-
butyltin trichloride, di-n-butyltin dichloride, tri-n-butyltin chloride and bis-(tri-n-butyltin) oxide
(Hamasaki et al., 1993). Only di-n-butyltin dichloride had positive results when the S.
typhimurium TA98 strain was used (Hamasaki et al., 1993). Positive results were found in the
SOS chromotest with Escherichia coli PQ37 for mono-n-butyltin oxide, n-butyltin trichloride
and di-n-butyltin dichloride (Hamasaki et al., 1992); negative results were observed for tri-n-
butyltin chloride. In the rec-assay for mutagenicity in Bacillus subtilis, positive results were
found for di-n-butyltin dichloride and tri-n-butyltin chloride, while negative results were found
for mono-n-butyltin oxide and n-butyltin oxide (Hamasaki et al., 1992).
Dibutyltin compounds gave mixed results for genotoxicity in mammalian cells in vitro.
Dibutyltin dichloride increased the frequency of mutations (HGPRT assay) in cultured Chinese
hamster ovary cells (Li et al., 1982). In tests without metabolic activation, tributyltin chloride
did not induce chromosomal aberrations in cultured Chinese hamster ovary (CHO Kl) cells
(Sasaki et al., 1993). Treatment with dibutyltin chloride or tributyltin chloride did not
significantly increase the frequency of aneuploidy in cultured peripheral lymphocytes taken from
one human donor (non-smoking healthy female) (Jensen et al., 1991). Tributyltin chloride
caused a slight elevation in the frequency of hyperdiploid cells, but the increase was not
statistically significant. Neither n-butyltin trichloride nor di(n-butyltin) dichloride induced
breaks in double-stranded lambda DNA (Hamasaki et al., 1995). Di-n-butyltin dichloride at
concentrations between 100 and 1500 nM promoted morphological transformation in previously
7

-------
6-12-2006
initiated, cultured murine C3H/10T1/2 cells (Parfett et al, 2000). The compound also induced
the expression of mRNA species associated with cell transformation: the proliferin gene (a
growth hormone with angiogenic properties) and members of the fos and jun proto-oncogene
families.
In vivo, dibutyltin diacetate was inactive in Drosophila melanogaster in an assay for sex-
linked recessive lethal mutations in which the male flies were either fed or injected the
compound before mating (Woodruff et al., 1985). Micronucleus formation in bone marrow
erythrocytes of mice exposed orally to dibutyltin dichloride was observed after 48 or 72 hours of
administration, but not after 24 hours, suggesting that biotransformation was needed for the
effect (Life Science Research, 1991).
In summary, there is evidence for the genotoxicity of mono-, di- and tri- butyltin
compounds in various in vitro and in vivo systems.
DERIVATION OF A PROVISIONAL CARCINOGENICITY ASSESSMENT FOR
MONO-, DI-, AND TRI- BUTYLTIN COMPOUNDS
There are no data for the carcinogenicity of mono-, di- or tributyltin compounds in
humans or for the carcinogenicity of monobutyltin compounds in animals. Data for dibutyltin
compounds in animals are limited to the NCI (1979) study, which reported negative or
inconclusive evidence in rats and mice exposed in the diet to dibutyltin diacetate. Data for
tributyltin compounds (excluding tributyltin oxide) are limited to a skin painting assay by
Sheldon (1975) in which no neoplastic changes were noted in mice exposed to tributyltin
fluoride. Short-term studies suggest that the butyltin compounds may produce genotoxic effects.
Under the guidelines for carcinogen risk assessment (U.S. EPA, 2005), there is inadequate
information to assess the carcinogenic potential of butyltin compounds. The lack of positive
cancer data precludes the derivation of provisional quantitative risk estimates for these
compounds.
REFERENCES
ATSDR (Agency for Toxic Substances and Disease Registry). 2003. Toxicological Profile for
Tin. Update Draft for Public Comment. Public Health Service. Atlanta, GA.
Boyer, I.J. 1989. Toxicity of dibutyltin, tributyltin and other organotin compounds to humans
and to experimental animals. Toxicol. 55:253-298.
Bulten, E.J. and H.A. Meinema. 1991. In: Metals and their Compounds in the Environment:
Tin. Merian, E., ed. VCH, New York. p. 1243-1259.
8

-------
6-12-2006
Hamasaki, T., T. Sato, H. Nagase and H. Kito. 1992. The genotoxicity of organotin compounds
in SOS chromotest and rec-assay. Mutat. Res. 280:195-203.
Hamasaki, T., T. Sato, H. Nagase and H. Kito. 1993. The mutagenicity of organotin compounds
as environmental pollutants. Mutat. Res. 300:265-271.
Hamasaki, T., T. Sato, H. Nagase and H. Kito. 1995. Breakage of lambda-DNA by inorganic tin
and organotin compounds as environmental pollutants. Appl. Organometall. Chem. 9:693-697.
[Toxline abstract]
IARC (International Agency for Research on Cancer). 2003. IARC Agents and Summary
Evaluations. Online, http://www-cie.iarc.fr/htdig/search.html
Jensen, K.G., O. Andersen and M. Ronne. 1991. Organotin compounds induce aneuploidy in
human peripheral lymphocytes in vitro. Mut. Res. 246:109-112.
Li, A.P., A.R. Dahl and J.O. Hill. 1982. In vitro cytotoxicity and genotoxicity of dibutyltin
dichloride and dibutylgermanium dichloride. Toxicol. Appl. Pharmacol. 64:482-485.
Life Science Research. 1991. Dibutyl Tin Chloride: Assessment of Clastogenic Action on Bone
Marrow Erythrocytes in the Micronucleus Test (Final Report). Produced 5/01/91. Submitted
5/13/91 by Atochem North America, Inc. to U.S. EPA under TSCA section 8E. EPA Doc. No.
88-910000159. Fiche No. OTS0529932. TSCATS 417298.
Magos, L. 1986. In: Handbook on Toxicology of Metals: Tin, Vol II, 2nd Ed. Friberg, L.,
Nordberg, G.F. and Vouk, V.R., Eds. Elsevier Science, Amsterdam, p. 568-593.
NCI (National Cancer Institute). 1979. Bioassay of Dibutyltin Diacetate for Possible
Carcinogenicity. U.S. Dept of Health, Education and Welfare, Public Health Service, National
Institutes of Health, Bethesda, MD. NCI Technical Report Series 183.
Nicklin, S. and M.W. Robson. 1988. Organotins: toxicology and biological effects. Appl.
Organometallic Chem. 2:487-508.
NIOSH (National Institute for Occupational Safety and Health). 1976. NIOSH Criteria for a
Recommended Standard...Occupational Exposure to Organotin Compounds. DHHS (NIOSH)
Publication Nol. 77-115. NTIS PB-274 766. Online, http://www.cdc.gov/niosh/77-115.html
NTP (National Toxicology Program). 2003a. Nomination background document for organotin
(methyl and butyl) toxicity. Online.
http://ntp-server.niehs.nih.gov/htdocs/Chem Background/ExSumPdf/Organotins.pdf
9

-------
6-12-2006
NTP (National Toxicology Program). 2003b. Monobutyltin trichloride. Management Status
Report. Online. http://ntp-server.niehs.nih.gov/htdocs/Results Status/Resstatm/M000066.Html
NTP (National Toxicology Program). 2003c. Dibutyltin diacetate. Management Status Report.
Online. http://ntp-server.niehs.nih.gov/htdocs/Results Status/Resstatd/10670-W.Html
NTP (National Toxicology Program). 2003d. Dibutyltin diacetate. Health and Safety Report.
Online. http://ntp-server.niehs.nih.gov/htdocs/CHEM H&S/NTP Chem 1 /Radian 1067-33-
O.html
Ohhira, S., M. Watanabe and H. Matsui. 2003. Metabolism of tributyltin and triphenyltin by rat,
hamster and human hepatic microsomes. Arch. Toxicol. 77:138-144.
Okada, Y., Y. Oyama, L. Chikahisa et al. 2000. Tri-n-butyltin-induced change in cellular level
of glutathione in rat thymocytes: a flow cytometry study. Toxicol. Lett. 117:123-128.
Parfett, C.L.J., T. Marquardt and R. Pilon. 2000. Promotion of transformation by di-//-butyl tin
dichloride in C3H/10T1/2 cells prediction by prior expression of tumour promoter-responsive
genes. Food Chem. Toxicol. 38:339-349.
Sasaki, Y.F., H. Yamada, C. Sugiyama and N. Kinae. 1993. Increasing effect of tri-n-butyltins
and triphenyltins on the frequency of chemically induced chromosome aberrations in cultured
Chinese hamster cells. Mut. Res. 300:5-14.
Sheldon, A.W. 1975. Effects of organotin anti-fouling coatings on man and his environment. J.
Paint Technol. 47:54-58.
Snoeij, N.J., A.H. Penninks and W. Seinen. 1987. Biological activity of organotin compounds-
an overview. Environ. Res. 44:335-353.
Ueno, S., T. Suzuki, N. Susa et al. 1997. Effect of SKF-525A on liver metabolism and
hepatotoxicity of tri- and dibutyltin compounds in mice. Arch. Toxicol. 71:513-518.
U.S. EPA. 1988. Recommendations for and Documentation of Biological Values for Use in
Risk Assessment. Environmental Criteria and Assessment Office, Office of Health and
Environmental Assessment, Cincinnati, OH. NTIS PB88-17874. EPA/600/6-87/008.
U.S. EPA. 1991. Chemical Assessments and Related Activities (CARA). Office of Health and
Environmental Assessment, Washington, DC. April.
U.S. EPA. 1994. Chemical Assessments and Related Activities (CARA). Office of Health and
Environmental Assessment, Washington, DC. December.
10

-------
6-12-2006
U.S. EPA. 1997a. Toxicological Review for Tributyltin Oxide (CAS No. 56-35-9) in Support of
Summary Information on the Integrated Risk Information System. Washington, DC. July.
Online, http://www.epa.gov/iris/toxreviews/0349-tr.pdf
U.S. EPA. 1997b. Health Effects Assessment Summary Tables. Annual Update. FY-1997
Update. Prepared by the Office of Research and Development, National Center for
Environmental Assessment, Cincinnati, OH, for the Office of Emergency and Remedial
Response, Washington, DC. July. EPA-540-R-97-036. PB97-921199.
U.S. EPA. 2002. 2002 Edition of the Drinking Water Standards and Health Advisories. Office
of Water, Washington, DC. EPA 822-R-02-038.
http://www.epa. gov/waterscience/drinking/ standards/dwstandards .pdf
U.S. EPA. 2005. Guidelines for Carcinogen Risk Assessment. Office of Research and
Development, National Center for Environmental Assessment, Washington, DC.
EPA/63 0/P-03/001F.
U.S. EPA. 2006. Integrated Risk Information System (IRIS). Office of Research and
Development, National Center for Environmental Assessment, Washington, DC. Online.
http ://www. epa. gov/ iris/
WHO (World Health Organization). 1980. Tin and Organotin Compounds. Environmental
Health Criteria 15. Geneva, Switzerland. Online.
http ://www. inchem.org/ documents/ehc/ehc/ehcO 15 .htm
WHO (World Health Organization). 1990. Tributyltin Compounds. Environmental Health
Criteria 116. Geneva, Switzerland. Online.
http ://www. inchem.org/documents/ehc/ehc/ ehc 116 .htm
WHO (World Health Organization). 1993. Dibutyltindichloride. Poison Information
Monograph G586. Geneva, Switzerland. Online.
http://www.inchem.org/documents/pims/chemical/pim586.htm
WHO (World Health Organization). 1994. Tributyltin Compounds. Poison Information
Monograph GO 18. Geneva, Switzerland. Online.
http://www.inchem.org/documents/pims/chemical/pimgQ18.htm
WHO (World Health Organization). 1999. Tributyltin Oxide. Consise International Chemical
Assessment Document 14. Geneva, Switzerland. Online.
http ://www. inchem.org/documents/cicads/cicads/cicad 14 .htm
11

-------
6-12-2006
Woodruff, R.C., J.M. Mason, R. Valencia and S. Zimmering. 1985. Chemical mutagenesis
testing in Drosophila. V. Results of 53 coded compounds tested for the National Toxicology
Program. Environ. Mutagen. 7:677-702.
12

-------