2                                                                       EPA/635/R-11/012C
 3                                                                         www.epa.gov/iris
 4
 5
 6
 7
 8
 9
10
11
12          Toxicological review of 1,2,4- and 1,3,5-Trimethylbenzene
13
14                              (CAS No.  95-63-6 and 108-67-8)
15
16                         In Support of Summary Information on the
17                         Integrated Risk Information System (IRIS)
18
19
20                                      JANUARY 2012
21
22
23
24                                        NOTICE
25
26   This document is an Interagency Science Consultation draft.  This information is distributed
27   solely for the purpose of pre-dissemination peer review under applicable information quality
28   guidelines.  It has not been formally disseminated by EPA. It does not represent and should not
29   be construed to represent any Agency determination or policy.  It is being circulated for review
30   of its technical accuracy and science policy implications.
31
32
33
34
35
36                          National Center for Environmental Assessment
37                              Office of Research and Development
38                             U.S. Environmental Protection Agency
39                                      Washington, DC

40
     DRAFT - DO NOT CITE OR QUOTE

-------
 2                                      DISCLAIMER
 o
 3
 4
 5    This document is a preliminary draft for review purposes only. This information is
 6    distributed solely for the purpose of pre-dissemination peer review under applicable
 7    information quality guidelines. It has not been formally disseminated by EPA. It does not
 8    represent and should not be construed to represent any Agency determination or policy.
 9    Mention of trade names or commercial products does not constitute endorsement of
10    recommendation for use.
     DRAFT - DO NOT CITE OR QUOTE

-------
 2
 3   CONTENTS	

 4
 5   CONTENTS	4
 6   LIST OF TABLES AND FIGURES	5
 7   ABBREVIATIONS AND ACRONYMS	6
 8   PREAMBLE	8
 9   AUTHORS | CONTRIBUTORS | REVIEWERS	18
10   PREFACE	20
11   EXECUTIVE SUMMARY	24
12   LITERATURE SEARCH STRATEGY AND STUDY EVALUATION	31
13          1.1. Synthesis of Major Toxicological Effects	34
14          1.2. Selection of Candidate Principal Studies and Critical Effects for Derivation of Reference
15          Values	61
16          1.3. Carcinogenicity Analysis	68
17   2.  DOSE-RESPONSE ANALYSIS	69
18          2.1. Inhalation Reference Concentration for Effects other than Cancer	69
19          2.2. Oral Reference Dose for Effects other than Cancer	86
20          2.3. Cancer Assessment	89
21   3.  REFERENCES	91
22   APPENDICES	97
23          Appendix A: Toxicological Information in Support of Hazard Identification and Dose-Response
24          Analysis for 1,2,4- and 1,3,5-Trimethylbenzene	97
25          Appendix B: Benchmark Dose Modeling Results for the Derivation of Reference Values for
26          1,2,4- and 1,3,5-Trimethylbenzene	97
27          Appendix C: Summary of External Peer Review and Public Comments and Disposition	97
28
29
     DRAFT - DO NOT CITE OR QUOTE

-------
 1
 2   LIST OF TABLES AND FIGURES

 3   Table I. Other Agency and International Assessments	22
 4   Table II. Reference concentration (RfC) for 1,2,4-TMB	25
 5   Table III. Reference dose (RfD) for 1,2,4-TMB	28
 6   Table IV: Details of the search strategy employed for 1,2,4-TMB and 1,3,5-TMB	31
 7   Table 1-1: Summary of observed in vivo neurotoxicity in subchronic and short-term studies
 8   of male Wistar rats following inhalation exposure to 1,2,4-TMB or 1,3,5-TMB	40
 9   Tables 1-2: Summary of observed in vivo neurotoxicity for 1,2,4-TMB and 1,3,5-TMB — oral
10   exposures	43
11   Tables 1-3: Summary of observed in vivo respiratory toxicity for 1,2,4-TMB and 1,3,5-TMB —
12   inhalation exposures	50
13   Table 1-4: Summary of observed developmental toxicity for 1,2,4-TMB and 1,3,5-TMB —
14   inhalation exposures	54
15   Tables 1-5: Summary of observed in vivo hematological toxicity and clinical  chemistry
16   effects for 1,2,4-TMB and 1,3,5-TMB — inhalation exposures	56
17   Table 1-6: Non-cancer endpoints resulting from subchronic inhalation exposure to
18   1,2,4-TMB considered for the derivation of the RfC	62
19   Figure 1-1. Exposure response array for inhalation exposure to 1,2,4-TMB	64
20   Table 1-7: Non-cancer endpoints resulting from gestational inhalation exposure (GD 6-20) to
21   1,3,5-TMB considered for the derivation of the RfC	66
22   Figure 1-2. Exposure response array for inhalation exposure to 1,3,5-TMB	67
23   Table 2-1: Summary of dose-response analysis and point of departure estimation for
24   endpoints resulting from subchronic inhalation exposure to 1,2,4-TMB	70
25   Table 2-2: Candidate PODADj values, human equivalent concentrations (HECs), and applied
26   uncertainty factors used in the derivation of RfCs for 1,2,4-TMB	71
27   Figure 2-1: Array of candidate PODHEc values with applied UFs and candidate RfCs for CNS,
28   hematological, and pulmonary effects  resulting from inhalation exposure to 1,2,4-TMB	75
29   Table 2-4: Duration adjusted point of departure (PODADj) estimates from short-term and
30   gestational inhalation exposures to 1,3,5-TMB	78
31   Table 2-5: Candidate PODADj values, human equivalent concentrations (HECs), and applied
32   uncertainty factors used in the derivation of RfCs for 1,3,5-TMB	80
33
     DRAFT - DO NOT CITE OR QUOTE

-------
ABBREVIATIONS AND ACRONYMS
1,2,4-TMB  1,2,4-trimethylbenzene                    n
1,3,5-TMB  1,3,5-trimethylbenzene                    NCEA
AAQC      Ambient air quality criterion
ACGIH     American Conference of                   NIOSH
           Governmental Industrial Hygienists
ADME     Absorption, distribution, metabolism        NLM
           and excretion                            NOAEL
AEGL      Acute exposure guideline limit              OMOE
AIC        Akaike Information Criterion               OSHA
BAL       bronchoalveolar lavage
BMD       benchmark dose                          p
BMDL     lower confidence limit on the               p-RfC
           benchmark dose                          PBPK
BMDS     benchmark dose software
BMR       benchmark response                      PEL
BW        body weight                             POD
C          Celsius                                  PODADj
CAS        Chemical Abstracts Service                 POI
CASRN     Chemical Abstracts Service Registry         ppm
           Number                                 RBC
CI         confidence interval                       RDso
CNS        central nervous system                    REL
CYP450    cytochrome P450                         RfC
DAF       dosimetric adjustment factor               RfD
DMBA     dimethylbenzoic acid                      RGDR
DMHA     dimethylhippuric acid                     ROS
DNA       deoxyribonucleic acid                     SCE
ECso       half maximal effective concentration         SD
EEC        Electroencephalogram                     SOA
EPA       U.S. Environmental Protection              t
           Agency                                  TLV
g          gram                                    TMB
GD        gestational day                           TSCA
Hb/g-A    animal blood:gas partition coefficient        TWA
Hb/g-H    human blood:gas partition coefficient        UF
HEC       human equivalent concentration            UFA
HEK       Human epidermal keratinocytes            UFn
HERO      Health Effects Research Online              UFS
HEV       human epithelial keratinocytes
HSDB      Hazardous Substance Database              UFt
IL-8        interleukin-8                             UFD
i.p.        intraperitoneal
IRIS       Integrated Risk Information System         jig
JP-8       jet propulsion fuel  8                       jil
k          kilogram                                (imol
Km        Michaelis-Menten constant                 UV
L          liter                                     V
LDH       lactate dehydrogenase                     VOC
LOAEL     lowest-observed-adverse-effect level        W
m3        meter cubed                             WBC
mg        milligram                                WS
nanogram
National Center for Environmental
Assessment
National Institute for Occupational
Safety and Health
National Library of Medicine
No-observed-adverse-effect level
Ontario Ministry of the Environment
Occupational Safety and Health
Administration
probability value
Provisional RfC
physiologically based
pharmacokinetic (model)
permissible exposure limit
point of departure
duration adjusted POD
Point of impingement
parts per million
red blood cell
50% respiratory rate decrease
Recommended exposure limit
reference concentration
reference dose
regional gas dose ratio
reactive oxygen species
sister chromatid exchange
standard deviation
Secondary organic aerosol
time
threshold limit value
trimethylbenzene
Toxic Substances Control Act
time-weighted average
uncertainty factor
interspecies uncertainty factor
intraspecies uncertainty factor
subchronic-to-chronic uncertainty
factor
LOAEL-to-NOAEL uncertainty factor
database deficiency uncertainty
factor
microgram
microliter
micromol
ultraviolet
volt
volatile organic compound
watt
white blood cell
white spirit
DRAFT - DO NOT CITE OR QUOTE

-------
          chi-squared
DRAFT - DO NOT CITE OR QUOTE

-------
PREAMBLE
1.  Scope of the IRIS Program

    Soon after EPA was established in 1970, it was
at the forefront of developing risk assessment as a
science  and applying  it in decisions to protect
human health and the environment. The Clean Air
Act, for  example, mandates that EPA provide  "an
ample margin of safety to  protect public health";
the Safe Drinking Water  Act,  that "no  adverse
effects on the health of persons may reasonably be
anticipated to occur, allowing an adequate margin
of safety." Accordingly,  EPA  relies  on health
assessments  to  identify  adverse effects  and
exposure levels below  which these effects are  not
anticipated to occur.
    IRIS assessments critically review the publicly
available studies to identify adverse health effects
of  chemicals  and  to  characterize   exposure-
response relationships. Exceptions are chemicals
currently used exclusively as pesticides,  ionizing
and   non-ionizing  radiation,   and criteria  air
pollutants listed  under section 108 of the Clean
Air Act  (carbon monoxide, lead, nitrogen oxides,
ozone, particulate matter, and sulfur oxides; EPA
evaluates    these    in   Integrated    Science
Assessments). An assessment may cover  a single
chemical, a group of structurally or lexicologically
related chemicals, or a complex mixture.
    Once  a  year, the  IRIS Program  asks  EPA
programs and regions,  other  federal agencies,
state  governments,  and  the  general  public  to
nominate  chemicals   and mixtures  for future
assessment or reassessment. These agents may be
found in air, water, soil, or sediment. Selection is
based on  program and regional office priorities
and on  availability  of adequate information  to
evaluate the potential for adverse effects.  IRIS  can
assess other  agents as an urgent public health
need  arises.   IRIS  also  reassesses  agents   as
significant new data are published.

2.  Process for developing and peer-
    reviewing IRIS assessments

    The process for developing IRIS assessments
(revised in May 2009)  involves  systematic review
of the pertinent studies, opportunities for public
input, and multiple levels of scientific review. EPA
revises  draft assessments after each review, and
external drafts and comments become part of the
public record (EPA 2009).

Step 1. Development of a draft Toxicological
    Review  (usually  about  11-1/2   months
    duration). The draft assessment considers all
    pertinent  publicly  available  studies  and
    applies  consistent  criteria to  evaluate the
    studies,  identify health  effects,  weigh the
    evidence of causation for each effect, identify
    mechanistic events and pathways, and derive
    toxicity values.

Step 2. Internal review by scientists in  EPA
    programs and regions (2 months). The draft
    assessment is revised to address comments
    from within EPA.

Step 3. Interagency science consultation with
    other federal agencies and White House
    offices (1-1/2 months). The draft assessment
    is  revised  to  address  the  interagency
    comments.  The  science consultation draft,
    interagency comments, and EPA's response to
    major comments become part of the public
    record.

Step 4.  External  peer  review, after public
    review  and  comment  (3-1/2 months or
    more, depending on the review process). EPA
    releases the  draft  assessment  for  public
    review and comment, followed by external
    peer review. The peer review  meeting is open
    to the public and includes time for oral public
    comments. The peer  reviewers also receive
    the written  public  comments.  The  peer
    reviewers assess whether  the evidence has
    been assembled and  evaluated according to
    guidelines  and whether  the  conclusions are
    justified by the  evidence.  The peer review
    draft, peer review report, and written public
    comments become part of the public record.

Step 5. Revision of draft Toxicological Review
    and development of draft  IRIS summary
    (2 months). The draft assessment is revised to
    reflect  the  peer review comments,  public
    comments, and  newly available studies. The
    disposition  of peer review  comments  and
    public comments becomes part of the public
    record.
DRAFT - DO NOT CITE OR QUOTE

-------
Step  6.  Final EPA  review  and  interagency
    science  discussion  with  other  federal
    agencies and White  House offices [1-1/2
    months). The draft assessment and summary
    are revised to address EPA  and interagency
    comments.  The  science  discussion  draft,
    written  interagency comments, and  EPA's
    response to major comments become part of
    the public record.

Step 7. Completion and posting (1 month). The
    Toxicological  Review and IRIS summary are
    posted   on   the   IRIS   website   (http://
    www.epa.gov/iris/).


    The remainder of this Preamble addresses
step 1, the development of a draft Toxicological
Review.   IRIS  assessments   follow  standard
practices of evidence evaluation and peer review,
many of which are discussed  in EPA guidelines
(EPA 1986a, 1986b,  1991, 1996,  1998, 2000a,
2005a, 2005b) and other descriptions  of  "best
practices" (EPA 1994, 2000b, 2002, 2006, 2011).
Transparent application of scientific judgment is
of  paramount   importance.   To   provide  a
harmonized approach across  IRIS  assessments,
this  Preamble summarizes concepts from these
guidelines and emphasizes principles of general
applicability.

3.  Identifying and selecting pertinent
    studies

3.1 Identifying studies

    Before   beginning  an   assessment,   EPA
conducts a comprehensive search of the primary
scientific literature. The literature search follows
standard practices and includes the PubMed and
ToxNet databases  of the  National Library of
Medicine  and other  databases  listed  in  EPA's
HERO   system   (Health   and   Environmental
Research  Online,  http://hero.epa.gov/).   Each
assessment   specifies   the  search  strategies,
keywords,  and  cut-off dates  of  its  literature
searches. EPA posts the results  of the  literature
search  on   the   IRIS  website  and   requests
information from  the public on additional studies
and ongoing research.
    Each  assessment  also  considers  studies
received  through  the  IRIS Submission  Desk and
studies (typically  unpublished) submitted to EPA
under the Toxic Substances Control Act. If a study
that may be critical  to the conclusions of the
assessment has not been peer-reviewed, EPA will
have it peer-reviewed.
    EPA also examines  the toxicokinetics of the
agent to  identify  other chemicals  (for example,
major metabolites of the agent) to include in the
assessment if adequate information is available, in
order to more fully explain the toxicity  of the
agent and to suggest dose metrics for subsequent
modeling.
    In assessments of chemical mixtures, mixture
studies  are preferred for their ability to  reflect
interactions among components  (EPA  1986a,
2000a). The literature search seeks, in decreasing
order of preference:

    Studies of the mixture being assessed.

    Studies of a sufficiently similar mixture. In
    evaluating   similarity,    the    assessment
    considers  the alteration of mixtures  in the
    environment   through   partitioning   and
    transformation.

    Studies of individual chemical components of
    the mixture, if there are not adequate studies
    of sufficiently similar mixtures.

3.2 Selecting pertinent epidemiologic studies

    Study design is the key  consideration  for
selecting pertinent epidemiologic studies from the
results of the literature search.

    Cohort  studies   and  case-control   studies
    provide the strongest epidemiologic evidence,
    as they collect  information about individual
    exposures and disease.

    Cross-sectional    studies    provide    useful
    evidence if they relate exposures and disease
    at the  individual level and it  is  clear  that
    exposure preceded the onset of disease.

    Ecologic   studies  (geographic  correlation
    studies) relate  exposures  and disease  by
    geographic area.  They can provide  strong
    evidence if there are large exposure contrasts
    between  geographic  areas, relatively little
    exposure variation within study areas,  and
    population migration is limited.

    Case reports of high or  accidental exposure
    lack definition of the population at risk and
    the expected number  of  cases.  They  can
    provide information about a rare disease or
    about the  relevance of analogous results in
    animals.

    The  assessment  briefly  reviews  ecologic
studies and case reports but includes details only
if  they  suggest  effects not  identified by other
epidemiologic studies.

3.3 Selecting pertinent experimental studies

    Exposure route  is a key design consideration
for selecting pertinent experimental studies from
the results of the literature search.
DRAFT - DO NOT CITE OR QUOTE

-------
    Studies of oral, inhalation, or dermal exposure
    involve   passage  through  an   absorption
    barrier and are considered most pertinent to
    human environmental exposure.

    Injection or implantation studies  are often
    considered less  pertinent but may  provide
    valuable    toxicokinetic    or    mechanistic
    information. They also may be useful  for
    identifying effects in  animals if deposition or
    absorption is problematic  (for  example, for
    particles and fibers).

    Exposure  duration  is  also  a  key  design
consideration for selecting pertinent experimental
studies.

    Studies of effects from chronic exposure are
    most pertinent to lifetime human exposure.

    Studies of effects from subchronic exposure
    are pertinent but less preferred than studies
    of chronic exposure.

    Short-term  and  acute   studies  are  less
    pertinent  but   are  useful  for  obtaining
    toxicokinetic or mechanistic information. The
    assessment reviews  short-term  and  acute
    studies if they suggest distribution or effects
    at a site not identified by longer-term studies.

    For developmental toxicity and reproductive
    toxicity, irreversible effects may result from a
    brief  exposure  during a  critical  period  of
    development. Accordingly,  specialized  study
    designs are used for  these effects (EPA 1991,
    1996,1998).

4.  Evaluating the quality of individual
    studies

4.1 Evaluating the quality of epidemiologic
    studies

    The   assessment  evaluates   design  and
methodologic  aspects   that  can  increase   or
decrease the  weight given to each epidemiologic
study in the overall  evaluation (EPA  1991, 1994,
1996,1998, 2005a):

    Documentation  of study design,  methods,
    population characteristics, and results.

    Definition and  selection  of the  study and
    comparison populations.

    Ascertainment of exposure and the potential
    for misclassification.

    Ascertainment of disease or effect and  the
    potential for misclassification.
    Duration  of  exposure  and  follow-up  and
    adequacy  for  assessing  the occurrence  of
    effects, including latent effects.

    Characterization of exposure during critical
    periods for the development of effects.

    Sample size and statistical power to detect
    anticipated effects.

    Participation rates and the resulting potential
    for selection bias.

    Potential confounding and other sources  of
    bias are identified and addressed in the study
    design or in the analysis of results. The basis
    for   consideration  of  confounding  is   a
    reasonable expectation that the confounder is
    prevalent in the population and is related to
    both exposure and outcome.

    For  developmental   toxicity,  reproductive
toxicity, neurotoxicity, and cancer there is further
guidance   on  the  nuances   of   evaluating
epidemiologic  studies of these effects  (EPA 1991,
1996,1998, 2005a).

4.2 Evaluating the quality of experimental
    studies

    The   assessment   evaluates   design   and
methodologic  aspects  that  can  increase   or
decrease the weight given to  each experimental
study in  the overall evaluation (EPA 1991, 1994,
1996,1998, 2005a):

    Documentation of  study design,  animals  or
    study population, methods,  basic data, and
    results.

    Relevance  of  the  animal  model or study
    population and the experimental methods.

    Characterization of the nature and extent of
    impurities   and    contaminants   of  the
    administered chemical or mixture.

    Characterization of dose and dosing regimen
    (including  age  at  exposure)   and  their
    adequacy  to elicit  adverse effects, including
    latent effects.

    Sample sizes and statistical power to detect
    dose-related differences or trends.

    Ascertainment of survival, vital signs, disease
    or effects, and cause of death.

    Control of other variables that could influence
    the occurrence of effects.

    The  assessment  uses  statistical tests  to
evaluate  whether the observations may be due to
chance. The standard for  determining statistical
significance of a  response  is a trend  test  or
DRAFT - DO NOT CITE OR QUOTE
                                              10

-------
comparison of outcomes in the exposed groups
against those of concurrent controls.  In  some
situations, examination of historical control data
from the same laboratory within a few years of the
study  may   improve  the  analysis.  For  an
uncommon   effect   that   is   not  statistically
significant  compared with  concurrent  controls,
historical  controls may show that the  effect is
unlikely to be due to chance. For a response that
appears significant against a concurrent control
response that is unusual, historical controls may
offer a different interpretation (EPA 2005a).
    For  developmental  toxicity,   reproductive
toxicity, neurotoxicity, and cancer there is further
guidance   on   the   nuances  of   evaluating
experimental studies of these effects (EPA 1991,
1996, 1998, 2005a).  In multi-generation studies,
agents that  produce developmental effects  at
doses that are not toxic to the maternal animal are
of special concern. Effects  that occur  at  doses
associated  with mild maternal  toxicity are not
assumed to result only from maternal toxicity.
Moreover,  maternal  effects may  be  reversible,
while effects on the offspring may  be  permanent
(EPA 1991,1998).

4.3 Reporting study results

    The assessment uses evidence tables  to report
details of the design  and key results of pertinent
studies. There may be separate tables for each site
of toxicity or type of study.
    If a large number of studies observe the same
effect,  the   assessment  considers   the   study
characteristics  in  this  section  to identify the
strongest  studies or types  of study.  The tables
report  details  from  these studies,   and the
assessment explains the reasons for not reporting
details of other studies or groups of studies that
do  not  add new  information.  Supplemental
material   provides   references  to  all  studies
considered, including those not summarized in the
tables.
    The  assessment  discusses strengths  and
limitations that affect the interpretation of each
study.  If the  interpretation of a  study  in the
assessment differs from that of the  study authors,
the  assessment  discusses  the basis  for the
difference.
    As a check on the selection and evaluation of
pertinent  studies,  EPA  asks peer reviewers  to
identify  studies  that  were   not   adequately
considered.
5.  Weighing the overall evidence of
    each effect

5.1 Weighing epidemiologic evidence

    For each effect, the assessment evaluates the
evidence from  the  epidemiologic studies  as  a
whole  to determine the extent to which  any
observed associations may be  causal.  Positive,
negative,  and  null  results  are  given  weight
according  to  study quality.  This  evaluation
considers aspects of an association that suggest
causality, discussed by Hill (1965) and elaborated
by Rothman and Greenland  (1998)  (EPA  1994,
2002, 2005a;DHHS 2004).

Strength of association: The finding of a large
    relative risk with narrow confidence intervals
    strongly suggests that  an association is  not
    due to chance, bias, or other factors. Modest
    relative risks, however, may reflect  a  small
    range of exposures,  an  agent of low potency,
    an increase in  a disease that is common,
    exposure misclassification, or other sources of
    bias.

Consistency of association: An  inference  of
    causality is strengthened if elevated risks are
    observed in independent studies of different
    populations    and    exposure   scenarios.
    Reproducibility of findings constitutes one of
    the  strongest   arguments   for   causality.
    Discordant    results   sometimes   reflect
    differences  in  exposure  or  in confounding
    factors.

Specificity of association: As originally intended,
    this refers to one cause associated with  one
    disease.  Current understanding that  many
    agents   cause  multiple diseases  and  many
    diseases have multiple causes make this a less
    informative aspect  of  causality,  unless  the
    effect is rare  or unlikely to  have  multiple
    causes.

Temporal  relationship: A causal interpretation
    requires that exposure precede development
    of the disease.

Biologic     gradient     (exposure-response
    relationship):           Exposure-response
    relationships  strongly  suggest causality. A
    monotonic  increase is  not the only  pattern
    consistent with causality. The presence of an
    exposure-response   gradient  also   weighs
    against bias and confounding as the source of
    an association.

Biologic plausibility: An inference of causality is
    strengthened by data demonstrating plausible
    biologic mechanisms, if available.
DRAFT - DO NOT CITE OR QUOTE
                                              11

-------
Coherence:  An  inference  of   causality   is
    strengthened  by  supportive  results  from
    animal experiments, toxicokinetic studies, and
    short-term  tests.  Coherence  may  also  be
    found  in other  lines of evidence,  such as
    changing disease patterns in the population.

"Natural experiments": A  change in exposure
    that  brings  about  a  change  in   disease
    frequency  provides  strong   evidence  of
    causality.

Analogy: Information on structural analogues or
    on chemicals that induce similar mechanistic
    events can provide insight into causality.

    These  considerations  are   consistent with
contemporary guidelines that evaluate the quality
and weight of evidence. Confidence is increased if
the magnitude of effect is large, if there is evidence
of an  exposure-response relationship, or  if an
association was observed and the plausible biases
would tend to  decrease the magnitude  of the
reported effect.  Confidence is decreased for study
limitations, inconsistency of results, indirectness
of evidence, imprecision, or reporting bias (Guyatt
eta!2008a,b).
    To make clear how much the epidemiologic
evidence contributes to the overall weight of the
evidence, the assessment may choose a descriptor
such  as  sufficient evidence, suggestive evidence,
inadequate evidence,  or  evidence suggestive of no
causal   relationship   to    characterize   the
epidemiologic  evidence  of each  effect  (DHHS
2004).

5.2 Weighing experimental evidence

    For each effect, the assessment evaluates the
evidence from the animal experiments as a whole
to determine the extent to which they indicate a
potential for effects in humans. Consistent results
across  various   species  and  strains  increase
confidence  that similar results  would occur in
humans.  Although causality is  not at issue in
controlled   experiments,   several   concepts
discussed by Hill (1965)  affect the weight of
experimental results:  consistency  of response,
dose-response relationships, strength of response,
biologic plausibility,  and coherence (EPA  1994,
2002,2005a).
    In   weighing   evidence    from   multiple
experiments, EPA (2005a) distinguishes

Conflicting evidence (that is, mixed positive and
    negative results  in the same sex and strain
    using a similar study protocol) from

Differing results  (that is, positive results and
    negative  results  are  in  different sexes or
    strains or use different study protocols).
Negative or null results do not invalidate positive
results in a different experimental system.  EPA
regards all as valid observations and looks to
mechanistic information, if available, to reconcile
differing results.
    It is well established  that there are critical
periods for some developmental and reproductive
effects.  Accordingly,  the assessment determines
whether critical periods have been adequately
investigated  (EPA  1991,   1996,  1998,  2005a,
2005b).  Similarly,  the  assessment determines
whether the  database is  adequate  to  evaluate
other critical sites and effects.

5.3 Characterizing modes of action

    For each effect, the assessment discusses the
available information on its modes of action and
associated key events (key events being empirically
observable, necessary precursor steps or biologic
markers of such steps;  mode  of action being a
series  of key events involving interaction with
cells,  operational  and  anatomic  changes,  and
resulting in disease). Pertinent information  may
also come from  studies  of   metabolites  or of
compounds that are structurally similar or that act
through  similar mechanisms. The  assessment
addresses   several   questions   about   each
hypothesized mode of action (EPA 2005a).

(a) Is   the   hypothesized mode  of  action
    sufficiently  supported in  test  animals?
    Strong support for  a   key  event  being
    necessary to a mode of action can come from
    experimental challenge to the hypothesized
    mode of action, where suppressing  a  key
    event suppresses the  disease. Support for a
    mode of action is meaningfully strengthened
    by consistent results in different experimental
    models, but not  by replicate  experiments in
    the  same  model.  The   assessment   may
    consider  various  aspects of  causality in
    addressing this question.

(b) Is   the   hypothesized mode  of  action
    relevant  to  humans?   The  assessment
    reviews  the key events to  identify critical
    similarities and differences between the test
    animals and humans. Site concordance is not
    assumed  between  animals   and   humans,
    though it may  hold  for  certain modes of
    action. Information suggesting  quantitative
    differences  is  considered  in dose-response
    analyses  but  is not  used  to  determine
    relevance.  Similarly,  anticipated  levels of
    human exposure are not used to determine
    relevance.

(c) Which populations  or life-stages can be
    particularly     susceptible    to     the
DRAFT - DO NOT CITE OR QUOTE
                                              12

-------
    hypothesized   mode   of  action?   The
    assessment reviews the key events to identify
    populations  and  life-stages that  might be
    susceptible to their occurrence. Quantitative
    differences may result  in  separate  toxicity
    values  for susceptible  populations  or  life-
    stages.

    The assessment discusses the likelihood that
an  agent  operates through  multiple  modes  of
action. An uneven level of support for different
modes  of  action can  reflect  disproportionate
resources spent investigating them (EPA 2005a).
It should be noted that in  clinical reviews, the
quality of evidence may be reduced if evidence is
limited to studies funded by one interested sector
(Guyattetal2008b).
    Studies of genetic toxicity are often available,
and the assessment  evaluates the  evidence  of a
mutagenic mode of action.

    Demonstration     of     gene     mutations,
    chromosome  aberrations, or aneuploidy  in
    humans or experimental mammals  (in vivo)
    provides the strongest evidence.

    This is followed  by positive results in lower
    organisms or in cultured cells (in vitro) or for
    other genetic events.

    Negative results  carry  less  weight, partly
    because they cannot exclude the possibility of
    effects in other tissues (IARC 2006).

    For germ-cell mutagenicity, EPA has defined
categories  of  evidence,  ranging from  positive
results  of  human  germ-cell  mutagenicity  to
negative results for  all effects  of  concern (EPA
1986b).

5.4 Characterizing the overall weight of the
    evidence

    After   weighing   the  epidemiologic   and
experimental studies pertinent to each effect, the
assessment may select a  standard descriptor  to
characterize the overall weight of the evidence.
For example, the following standard descriptors
combine   epidemiologic,   experimental,   and
mechanistic evidence  of carcinogenicity  (EPA
2005a).

Carcinogenic  to  humans: There  is  convincing
    epidemiologic evidence of a  causal association
    (that is, there is reasonable confidence  that
    the association cannot be fully explained by
    chance,  bias, or  confounding);  or there  is
    strong  human  evidence of cancer  or  its
    precursors,   extensive   animal   evidence,
    identification of  key precursor  events  in
    animals, and strong evidence  that they are
    anticipated to occur in humans.
Likely to  be  carcinogenic  to  humans:  The
    evidence demonstrates a potential hazard to
    humans but does  not meet the criteria for
    carcinogenic.  There  may be  a  plausible
    association  in  humans,  multiple positive
    results in animals, or a combination of human,
    animal, or other experimental data.

Suggestive evidence of carcinogenic  potential:
    The data raise concern for effects in humans
    but  are  not  sufficient  for  a  stronger
    conclusion. This descriptor covers  a range of
    evidence, from  a  positive result in the  only
    available study to a single positive result in an
    extensive  database that  includes negative
    results in other species.

Inadequate information to assess carcinogenic
    potential:  No  other   descriptors   apply.
    Conflicting  evidence  can be  classified  as
    inadequate information if all positive results
    are opposed by  negative studies of equal
    quality in the same sex and strain. Differing
    results,   however,  can   be   classified   as
    suggestive  evidence  or  as   likely  to   be
    carcinogenic.

Not likely to be carcinogenic to humans: There
    are robust data for concluding that there is no
    basis for concern. There may be no effects in
    both sexes of at least two appropriate animal
    species; positive animal results  and strong,
    consistent evidence that each mode of action
    in animals  does not operate in humans;  or
    convincing evidence that effects are not likely
    by a particular exposure route  or below a
    defined dose.

6.  Selecting studies for derivation of
    toxicity values

    For each effect associated with an agent, the
assessment derives toxicity values if there  are
suitable epidemiologic or experimental data. The
derivation of toxicity values may be linked to the
weight-of-evidence descriptor. For example,  EPA
typically  derives  toxicity  values   for  agents
classified as carcinogenic to humans or likely to be
carcinogenic, but not for agents with  inadequate
information or that are not likely to be carcinogenic
(EPA2005a).
    Dose-response analysis requires quantitative
measures  of dose  and response.  Then, other
factors being equal (EPA 1994, 2005a):

    Epidemiologic  studies  are  preferred   over
    animal studies, if quantitative measures  of
    exposure are available and effects  can  be
    attributed to the agent.
DRAFT - DO NOT CITE OR QUOTE
                                              13

-------
    Among  experimental  animal models,  those
    that respond most like humans are preferred,
    if  the  comparability  of  response  can be
    determined.

    Studies by  a route of human environmental
    exposure are preferred,  although  a validated
    toxicokinetic   model    can   be   used  to
    extrapolate across exposure routes.

    Studies  of longer  exposure  duration and
    follow-up   are   preferred,    to   minimize
    uncertainty  about  whether  effects   are
    representative of lifetime exposure.

    Studies  with   multiple  exposure  levels  are
    preferred   for  their   ability  to   provide
    information about the shape  of the exposure-
    response curve.

    Studies  that   show  an exposure-response
    gradient are preferred,  as long as lack of  a
    monotonic  relationship  at higher exposure
    levels  can be satisfactorily  explained by
    factors such as competing toxicity, saturation
    of absorption or metabolism,  misclassification
    bias, or selection bias.

    Among  studies  that  show  an  exposure-
    response  gradient,  those  with  adequate
    power  to  detect  effects at  lower exposure
    levels are preferred, to minimize the extent of
    extrapolation   to   levels   found   in   the
    environment.

    If a large number of studies are suitable for
dose-response analysis, the assessment considers
the study characteristics in this section to focus on
the  most  informative  data. The  assessment
explains  the reasons  for  not  analyzing  other
groups of studies.  As a check on the selection of
studies for dose-response analysis, EPA asks peer
reviewers  to  identify  studies  that  were  not
adequately considered.

7.  Deriving toxicity values

7.1 General framework for dose-response
    analysis

    EPA   uses  a  two-step   approach   that
distinguishes  analysis of  the  observed  dose-
response data from inferences about lower  doses
(EPA2005a).
    Within the observed range,  the  preferred
approach is to use modeling to incorporate a wide
range of data  into the analysis. The modeling
yields a point of departure (an exposure level near
the lower end of the observed  range,  without
significant extrapolation to lower doses) (sections
7.2-7.3).
    Extrapolation to lower doses considers what
is known about the modes of action for each effect
(sections  7.4-7.5).  An  alternative  to low-dose
extrapolation is  derivation  of reference values,
which  are calculated  by  adjusting the point of
departure  by factors  that  account for  several
sources of uncertainty and  variability (section
7.6).
    Increasingly, EPA is  making use of multiple
data sets or combining  multiple responses  in
deriving   toxicity  values.  EPA   also  considers
multiple  dose-response  approaches when  they
can be supported by robust data.

7.2 Modeling dose

    The preferred approach for analysis of dose is
toxicokinetic  modeling because  of its ability to
incorporate a wide range of data. The preferred
dose metric would refer to the active agent at the
site of its  biologic  effect or  to  a close, reliable
surrogate measure. The active agent may be the
administered chemical or a metabolite. Confidence
in the use of a toxicokinetic model depends on the
robustness of its validation process and on the
results of sensitivity analyses (EPA 1994, 2005a,
2006).
    Because  toxicokinetic modeling  can require
many parameters and more data than are typically
available, EPA has developed standard approaches
that can be applied to typical data sets. These
standard  approaches  also facilitate comparison
across exposure patterns and species.

    Intermittent    study    exposures     are
    standardized  to a  daily average over  the
    duration  of exposure. For  chronic  effects,
    daily  exposures   are averaged  over  the
    lifespan.  Exposures during  a critical period,
    however, are  not averaged over a longer
    duration (EPA 1991,1996,1998, 2005a).

    Doses are standardized to equivalent human
    terms to facilitate comparison of results from
    different species.

        Oral doses are scaled allometrically using
        mg/kg3/4-d as the equivalent dose metric
        across species. As allometric  scaling is
        typically based on adult body weight, it is
        not used for  early-life  exposure or for
        developmental effects (EPA 2005a, 2011).

        Inhalation exposures are  scaled  using
        dosimetry models that  apply species-
        specific physiologic and  anatomic factors
        and consider whether the effect occurs at
        the site of first contact or after systemic
        circulation (EPA 1994).
DRAFT - DO NOT CITE OR QUOTE
                                              14

-------
    It can be informative to convert doses across
exposure routes. If this is done, the assessment
describes  the underlying data,  algorithms, and
assumptions (EPA2005a).

7.3 Modeling response in the range of
    observation

    Toxicodynamic     ("biologically     based")
modeling  can  incorporate  data  on   biologic
processes leading  to  a disease.  Such  models
require sufficient data  to ascertain a mode  of
action   and   to  quantitatively  support  model
parameters   associated  with  its  key  events.
Because different models may provide equivalent
fits to the observed  data but diverge substantially
at lower doses, critical biologic parameters should
be  measured from  laboratory  studies,  not by
model   fitting.  Confidence  in  the  use  of  a
toxicodynamic model depends on the robustness
of its validation process and on the results  of
sensitivity analyses. Peer review of the scientific
basis  and performance of a model is essential
(EPA2005a).
    Because toxicodynamic modeling can require
many parameters and more knowledge and data
than are typically available, EPA has developed a
standard set of empirical ("curve-fitting") models
(http://www.epa.gov/ncea/bmds/)  that  can be
applied to typical data sets,  including those that
are nonlinear. EPA  has  also developed guidance
on modeling dose-response data, assessing model
fit,  selecting  suitable  models,  and  reporting
modeling   results   (EPA  2000b).   Additional
judgment or  alternative analyses are used  when
the procedure fails to yield reliable results, for
example, if  the  fit  is poor, modeling may be
restricted to the lower doses, especially if there is
competing toxicity at higher doses (EPA 2005a).
    Modeling is  used  to  derive   a point  of
departure (EPA 2000b,  2005a).  (See  section 7.6
for  alternatives if a  point of departure cannot be
derived by modeling.)

    For dichotomous  responses,  the point  of
    departure is the  95% lower bound on the
    dose associated with a  small increase of a
    biologically significant effect.

        If linear extrapolation to lower doses will
        be used, a  standard value near  the low
        end of the observable range is used (10%
        response   for   animal  data, 1% for
        epidemiologic  data,  depending  on the
        observed response rates).

        If nonlinear extrapolation will be  used,
        both statistical  and  biologic factors are
        considered  (10% response for minimally
        adverse  effects, 5% or  lower for  more
        severe   effects  or  for  developmental
        toxicity data on individual offspring).

    For  continuous  responses,  the  point  of
    departure is ideally a level where the effect is
    considered minimally adverse. In the absence
    of such definition, both statistical and biologic
    factors are considered in selecting a response
    level.

7.4 Extrapolating to lower doses

    The purpose of extrapolating to lower doses is
to estimate responses at exposures below the
observed data. Low-dose extrapolation is typically
used for known and likely carcinogens. Low-dose
extrapolation considers what  is known  about
modes of action (EPA 2005a).

(1) If  a  biologically  based  model has  been
    developed  and  validated  for  the  agent,
    extrapolation  may  use  the  fitted  model
    beyond the  observed  range  if significant
    model  uncertainty can  be  ruled out with
    reasonable   confidence.   Below   the  range
    where  confidence bounds on the predictions
    are reasonably  precise,  extrapolation may
    continue using a linear model.

(2) Linear  extrapolation is  used if the  dose-
    response curve is expected to have a linear
    component below the point of departure. This
    includes:

        Agents or their metabolites that are DNA-
        reactive  and have   direct  mutagenic
        activity.

        Agents  or  their  metabolites for  which
        human  exposures or body  burdens are
        near doses associated with key  events
        leading to an effect.

    Linear  extrapolation  is  also  used  if the
    evidence is insufficient to establish a mode of
    action.

    The result of linear extrapolation is described
    by an oral slope factor or an inhalation unit
    risk, which is the slope of the dose-response
    curve at lower doses.

(3) Nonlinear extrapolation  is used  if there are
    sufficient data to ascertain the mode of action
    and to  conclude that it is not linear at lower
    doses,  and  the  agent does not demonstrate
    mutagenic or other activity consistent with
    linearity  at  lower  doses.  If  nonlinear
    extrapolation is appropriate but  no model is
    developed, a default is to calculate reference
    values.
DRAFT - DO NOT CITE OR QUOTE
                                              15

-------
    If linear extrapolation is used, the assessment
develops a candidate slope factor or unit risk for
each suitable data set.  These results are arrayed,
using  common  dose  metrics,  to  show  the
distribution of relative  potency across various
effects and experimental systems. The assessment
then derives an overall slope factor and an overall
unit risk for the agent,  considering the various
dose-response  analyses,  the  study preferences
discussed in section  6,  and the  possibility of
basing a more robust result on multiple data sets.

7.5 Considering susceptible populations and
    life-stages

    The   assessment  analyzes  the  available
information on  populations  and life-stages  that
may be particularly susceptible to  each effect.  A
tiered approach is used (EPA 2005a).

(1) If  an epidemiologic  or  experimental  study
    reports quantitative results for a susceptible
    population   or life-stage,  these  data  are
    analyzed to derive  separate toxicity values for
    susceptible individuals.

(2) If  data  on risk-related  parameters  allow
    comparison  of the general population  and
    susceptible individuals, these data are used to
    adjust the general-population toxicity values
    for application to susceptible individuals.

(3) In the  absence  of  chemical-specific  data,
    application  of  age-dependent  adjustment
    factors   is   recommended  for   early-life
    exposure to suspected carcinogens. There  is
    evidence of early-life susceptibility to various
    carcinogenic agents,  but most  epidemiologic
    studies and  cancer bioassays do  not include
    early-life exposure. To address the potential
    for early-life susceptibility, EPA recommends:

        10-fold adjustment for exposures before
        age 2 years.

        3-fold adjustment for exposures between
        ages 2 and 16 years.

    These adjustments are generally applied only
    for a mutagenic mode of action, though early-
    life susceptibility  has been   observed for
    several carcinogens  that are not mutagenic
    (EPA2005b).

7.6 Reference values and uncertainty factors

    An oral reference  dose  or  an  inhalation
reference  concentration  is an  estimate  of an
exposure  (including in  susceptible  subgroups)
that is likely to be without an appreciable risk of
adverse health effects over a lifetime (EPA 2002).
Reference  values  are typically calculated for
effects  other  than  cancer  and for  suspected
carcinogens if a well characterized mode of action
indicates  that  a  threshold can  be  based  on
prevention of an early key event. Reference values
provide no information about risks at exposures
above the reference value.
    The  assessment  characterizes  effects  that
form the basis for reference values  as adverse,
considered to be adverse, or  a precursor to  an
adverse effect.  For developmental,  reproductive,
and neurotoxicity there is guidance  on  adverse
effects and their  biologic markers (EPA 1991,
1996,1998).
    To account for uncertainty and variability in
the derivation of a lifetime human exposure where
effects are not anticipated to  occur, reference
values are calculated by adjusting the point of
departure by a series of uncertainty factors. If a
point of departure cannot be derived by modeling,
a no-observed-adverse-effect level  or a  lowest-
observed-adverse-effect level is substituted. The
assessment discusses  scientific considerations
involving   several  areas  of  variability  or
uncertainty.

Human variation. A factor of 10  is  applied to
    account for variation in susceptibility across
    the human population and the possibility that
    the available data may not be representative
    of individuals who are most susceptible to the
    effect. This factor is reduced only if the  point
    of  departure  is   derived  specifically for
    susceptible  individuals (not for  a   general
    population that includes both susceptible and
    non-susceptible individuals) (EPA 1991,1994,
    1996,1998,2002).

Animal-to-human extrapolation. A factor  of 10
    is applied if animal results are  used to make
    inferences about humans. This factor is  often
    regarded as  comprising toxicokinetics and
    toxicodynamics in equal parts. Accordingly, if
    the  point   of   departure  is  based  on
    toxicokinetic  modeling, dosimetry modeling,
    or allometric scaling across  species, a factor of
    101/2 (rounded to 3) is applied to account for
    the    remaining    uncertainty   involving
    toxicodynamic  differences.  An   animal-to-
    human factor is not applied if  a biologically
    based  model adjusts fully  for toxicokinetic
    and toxicodynamic differences  and residual
    uncertainty across species  (EPA 1991, 1994,
    1996,1998,2002).

Adverse-effect level to  no-observed-adverse-
    effect level. If a point of departure is based on
    a lowest-observed-adverse-effect  level, the
    assessment must infer a  dose where  such
    effects are not expected. This can be a matter
    of great uncertainty, especially  if there  is  no
DRAFT - DO NOT CITE OR QUOTE
                                              16

-------
    evidence available at lower doses. A factor of
    10 is applied to account for the uncertainty in
    making this inference. A factor other than 10
    may be used,  depending on  the magnitude
    and nature of the response  and the shape of
    the dose-response curve (EPA  1991, 1994,
    1996,1998,2002).

Subchronic-to-chronic  exposure. If a point of
    departure  is based on  subchronic studies, the
    assessment   considers   whether   lifetime
    exposure would have effects at lower levels. A
    factor of  10  is applied to  account for the
    uncertainty in using  subchronic studies to
    make inferences about lifetime exposure. This
    factor may also be applied for developmental
    or reproductive effects  if exposure covered
    less than the full critical period. A factor other
    than 10 may be  used,  depending on  the
    duration of the studies and  the nature of the
    response (EPA 1994,1998, 2002).

Incomplete database. If an  incomplete database
    raises  concern that  further  studies  might
    identify a more sensitive effect, organ system,
    or life-stage,  the assessment may apply a
    database uncertainty factor (EPA 1991, 1994,
    1996, 1998, 2002). EPA  typically follows the
    suggestion that a factor of 10 be  applied if
    both a  prenatal  toxicity study and a two-
    generation reproduction  study  are missing,
    and a factor of 1Q1/2 if either is missing (EPA
    2002).

    In this way, the assessment derives  candidate
reference values for  each suitable data set  and
effect that is plausibly associated  with the agent.
These  results  are  arrayed, using common dose
metrics,  to  show  where effects occur  across a
range of exposures (EPA 1994). The assessment
then selects an  overall reference dose and  an
overall reference  concentration for  the agent to
represent lifetime  human  exposure  levels  where
effects are not anticipated to occur.
    The  assessment  may  also  report  reference
values  for  each  effect.  This  would   facilitate
subsequent cumulative risk assessments, where it
may be important to consider the combined effect
of chemicals acting at a common site or  operating
through common mechanisms (EPA 2002).
7.7 Confidence and uncertainty in the
    reference values

    The assessment selects a standard descriptor
to characterize the level  of confidence in each
reference value, based on  the likelihood that the
value  would  change  with   further   testing.
Confidence in reference values is based on quality
of the studies  used and  completeness of the
database, with more weight given to the latter.
The level of confidence is increased for reference
values based on human data supported by animal
data (EPA 1994).

High confidence: The reference value is not likely
    to change with  further testing, except for
    mechanistic  studies  that  might  affect the
    interpretation of prior test results.

Medium  confidence:  This   is  a  matter  of
    judgment, between high and low confidence.

Low confidence: The reference value is especially
    vulnerable to change with further testing.

    These    criteria   are   consistent   with
contemporary guidelines that evaluate the quality
of evidence. These also focus on whether further
research would be likely to change confidence in
the estimate of effect (Guyatt et al 2008a).

    All  assessments  discuss   the   significant
uncertainties  encountered  in the analysis. EPA
provides   guidance   on   characterization  of
uncertainty  (EPA  2005a).  For  example,  the
discussion  distinguishes model  uncertainty (lack
of  knowledge   about  the  most  appropriate
experimental   or  analytic  model), parameter
uncertainty  (lack  of  knowledge   about  the
parameters of a  model),  and  human  variation
(interpersonal     differences     in     biologic
susceptibility or  in  exposures  that  modify the
effects of the agent).

    For other  general  information  about  this
assessment or other questions relating to IRIS, the
reader is referred to EPA's IRIS Hotline at (202)
566-1676   (phone),  (202) 566-1749  (fax),  or
hotline.iris@epa.gov       (email       address).
DRAFT - DO NOT CITE OR QUOTE
                                             17

-------
AUTHORS  |  CONTRIBUTORS  | REVIEWERS
Authors
J. Allen Davis, M.S.P.H. (Chemical Manager)
Eva McLanahan, Ph.D. (LCDR, USPHS)
Paul Schlosser, Ph.D.
John Cowden, Ph.D.
Gary Foureman, Ph.D. (Currently ICF Int.)
Andrew Kraft, Ph.D
U.S. Environmental Protection Agency
Office of Research and Development
National Center for Environmental Assessment
Research Triangle Park, NC

U.S. Environmental Protection Agency
Office of Research and Development
National Center for Environmental Assessment
Washington, DC
Ray Antonelli, B.S.
Oak Ridge Institute for Scientific Education
Research Triangle Park, NC
Contributors
Reeder Sams, Ph.D.
John Stanek, Ph.D.
Rob Dewoskin, Ph.D.
George Woodall, Ph.D.
Geniece Lehmann, Ph.D.
Connie Meacham, M.S.
U.S. Environmental Protection Agency
Office of Research and Development
National Center for Environmental Assessment
Research Triangle Park, NC
Technical Support

Ellen Lorang, M.S.
Deborah Wales
Gerald Gurevich
U.S. Environmental Protection Agency
Office of Research and Development
National Center for Environmental Assessment
Research Triangle Park, NC
Contractor Support
Battelle Memorial Institute, Pacific Northwest Division, Richmond, WA
Karla D. Thrall, Ph.D.
Battelle Memorial Institute, Columbus, OH
Jessica D. Sanford, Ph.D.
Maureen A. Wooton
Robert A. Lordo, Ph.D.
Anthony Fristachi
DRAFT - DO NOT CITE OR QUOTE
                                                   18

-------
Toxicology Excellence for Risk Assessment (TERA)
Under Battelle Memorial Institute Contract EP-C-09-006
Lisa M. Sweeney, Ph.D., DABT
Melissa J. Kohrman-Vincent, B.A.
Executive Direction

Reeder Sams Ph D                        U'S' Environmental Protection Agency
,,.,,'    _, _.                    Office of Research and Development
John Vandenberg, Ph.D.                           I^^FC-       ,. i A
'  ,    ... ,  ,  .°                         National Center for Environmental Assessment
Debra Walsh, M.S.
                                       Research Triangle Park, NC

Vincent Cogliano, Ph.D.                    U.S. Environmental Protection Agency
Samantha Jones, Ph.D.                     Office of Research and Development
Lynn Flowers, Ph.D.                      National Center for Environmental Assessment
Jamie Strong Ph.D.                        Washington, DC
Reviewers

This document has been provided for review to EPA scientists.
DRAFT - DO NOT CITE OR QUOTE                                                             19

-------
PREFACE
Background of Current Toxicological Review

   There is currently no entry on the IRIS Database for either 1,2,4-TMB or 1,3,5-TMB.
The current assessment has undergone draft development in which an RfC and RfD were
derived for both 1,2,4-TMB and 1,3,5-TMB. No cancer values are derived for either isomer.
Chemical and Physical Information

   The commercially available substance known as trimethylbenzene (TMB}, CAS No.
25551-13-7, is a mixture of three isomers in various proportions, namely CAS No. 526-73-8
(1,2,3-trimethylbenzene or hemimellitene], CAS No. 108-67-8 (1,3,5-trimethylbenzene or
mesitylene}, and CAS No. 95-63-6 (1,2,4-trimethylbenzene or pseudocumene}. The focus of
this EPA review is two of these isomers: 1,2,4-trimethylbenzene (1,2,4-TMB} and 1,3,5-
trimethylbenzene (1,3,5-TMB}.
   The TMBs are aromatic hydrocarbons with three methyl substituents attached to a
benzene ring and the chemical formula C9H12. The chemical and physical properties of the
TMB isomers are similar to one another. TMB is a colorless, flammable liquid with a strong
aromatic odor; an odor threshold of 0.4 parts per million (ppm} of air has been reported
(U.S. EPA. 1994}. It is insoluble in water but miscible with organic solvents such as ethyl
alcohol, benzene, and ethyl ether [OSHA. 1996}.
   Vehicle emissions are a major anthropogenic source of 1,2,4-TMB and 1,3,5-TMB, due to
the widespread use of the C9 fraction as a gasoline additive (U.S. EPA. 1994}. Other uses of
1,2,4-TMB and 1,3,5-TMB include solvents in research and industry, uses as a dyestuff
intermediate, paint thinner, and as a UV oxidation stabilizer for plastics (HSDB. 2011a. b}.
Production and use of 1,2,4-TMB and 1,3,5-TMB may result in their release to the
environment through various waste  streams. If released to the atmosphere, 1,2,4-TMB and
1,3,5-TMB will exist solely in the vapor phase in the ambient atmosphere, based  on
measured vapor pressures of 2.10 and 2.48 mm Hg at 25QC, respectively (HSDB. 2011a. b}.
Both isomers are expected to have limited mobility through  soil based on their Log KOC -
values, but are expected to volatilize from both moist and dry soil surfaces and surface

DRAFT - DO NOT CITE OR QUOTE                                                     20

-------
waters based on their respective Henry's Law constants and vapor pressures (see Table A.l-
1}.  Degradation of both isomers in the atmosphere occurs by reaction with hydroxyl
radicals, the half-life of which is 11-12 hours (HSDB. 2011a. bj.  Non-volatilized 1,2,4-TMB
and 1,3,5-TMB may be subject to biodegraelation under aerobic conditions (HSDB. 2011a. bj.
The estimated bio-concentration factors (439 and 234} and high volatility of 1,2,4-TMB and
1,3,5-TMB suggest that bioaccumulation of these chemicals will not be significant (U.S. EPA.
19871
   Additional information on the chemical identities and physicochemical properties of
1,2,4-TMB and 1,3,5-TMB are listed in Table A. 1-1.

Table 1. Physical properties and chemical identity of 1,2,4-TMB and 1,3,5-TMB
CAS Registry Number
Synonym(s)
Molecular formula
Molecular weight
Chemical structure
Melting point, °C
Boiling point, °C @ 760 mm Hg
Vapor pressure, mm Hg @ 25°C
Density, g/mL at 20 °C relative to
the density of H2O at 4 °C
Flashpoint, °C
Water solubility, mg/L at 25 °C
Other solubilities
Henry's Law Constant, atm-
m3/mole
Log KOW
Log KOC
Bioconcentration Factor
95-63-6
1,2,4-
Trimethylbenzene,
pseudocumene,
asymmetrical
trimethylbenzene
108-67-8
1,3,5-Trimethyl benzene,
mesitylene, symmetrical
trimethylbenzene
C9H12
120.19
CH3
^^./CH3
k^^
CH3
-43.8
168.9
2.10
0.8758
44
57
Miscible with ethanol,
benzene, ethyl ether,
acetone, carbon
tetrachloride, petroleum
ether
6.16 x 10"3
3.78
2.73
439
CH3
H3C CH3
-44.8
164.7
2.48
0.8637
50
48.2
Miscible with alcohol,
ether, benzene, acetone,
and oxygenated and
aromatic solvents
8.77 x 10"3
3.42
2.70-3.13
234
DRAFT - DO NOT CITE OR QUOTE
21

-------
CAS Registry Number
Synonym(s)
Conversion factors
95-63-6
1,2,4-
Trimethylbenzene,
pseudocumene,
asymmetrical
trimethylbenzene
108-67-8
1,3,5-Trimethyl benzene,
mesitylene, symmetrical
trimethylbenzene
1 ppm = 4.92 mg/m3
1 mg/m3 = 0.2 ppm
 Source: [HSDB. 2011a. b; U.S. EPA. 1987]


Programmatic Interest

   1,2,4-trimethylbenzene (1,2,4-TMB) and 1,3,5-trimethybenzene (1,3,5-TMB) are
industrial solvents found at Superfund sites. This IRIS assessment is being developed due
to the potential for human environmental exposure to these compounds. The related
isomer, 1,2,3-trimethylbenzene, is not included in this Toxicological Review.
Other Agency and International Assessments
Table 2. Other Agency and International Assessments
Agency
National Institue of
Occupational Safety and
Health (NIOSH. 1992)
American Conference of
Governmental Industrial
Hygienists (ACGIH. 2002)
National Advisory Committee
for Acute Exposure Guideline
Levels for Hazardous
Substances (U.S. EPA, 2007)

Ontario Ministry of the
Environment (MOE. 2006)
Inhalation value
Recommended Exposure Limit (REL) for TMBs- 25 ppm (123 mg/m3)
time weighted average for up to a 10 hour work day and a 40 hour
work week, based on the risk of skin irritation, central nervous system
depression, and respiratory failure (reference not provided)
Threshold Limit Value (TLV) for VOC mixture containing 1,2,4-TMB and
1,3,5-TMB- 25 ppm (123 mg/m3) time weighted average for a normal
8-hour work day and a 40-hour work week, based on the risk of
irritation and central nervous system effects (Battig et al., 1956)

For TMBs: AEGL-1 (nondisabling) - 180 ppm (890 mg/m3) to 45 ppm
(220 mg/m3) (10 minutes to 8 hours, respectively) (Korsak and
Rydzynski, 1996)
AEGL-2 (disabling) - 460 ppm (2300 mg/m3) to 150 ppm (740 mg/m3)
(10 minutes to 8 hours, respectively) (Gage, 1970)
For TMBs: 24 hour Ambient Air Quality Criterion (AAQC) - 0.3 mg/m3
based on CNS effects; half-hour Point of Impingement (POI) - 0.9
mg/m3 based on CNS effects (Wiaderna et al., 2002; Gralewicz and
Wiaderna, 2001; Gralewicz et al., 1997a; Korsak and Rydzynski, 1996)

DRAFT - DO NOT CITE OR QUOTE
22

-------
EXECUTIVE SUMMARY
Effects other than cancer observed following inhalation exposure to
1,2,4-TMB

      The relationship between exposure to 1,2,4-TMB and health effects has been
evaluated in studies of (1] exposed human adults, (2} animals exposed via inhalation for
acute, short-term, and subchronic durations, and (3} animals exposed gestationally via
inhalation. Human studies included occupational exposure to various solvent mixtures and
controlled human exposures to 1,2,4-TMB or solvent mixtures containing 1,2,4-TMB.
Health effects noted in these studies were limited to irritative (eye irritation] and
neurological effects (hand tremble, abnormal fatigue, lack of coordination] (Lammers et al..
2007: Chenetal.. 1999: Norseth et al.. 1991: Battigetal.. 19561  No human studies were
found for 1,3,5-TMB.
   Animal inhalation studies included acute and short-term studies for both isomers that
reported respiratory irritative (decreased respiration rates] and neurological effects
(decreased pain sensitivity and decreased neuromuscular function and coordination] that
supported effects seen in human studies (Wiaderna et al.. 2002:  Gralewicz and Wiaderna.
2001: Gralewicz etal.. 1997a: Gralewicz etal.. 1997b: Korsaketal.. 1995]. Three
subchronic inhalation studies were found for 1,2,4-TMB that observed exposure-response
effects in multiple organ systems, including the nervous, hematological, and pulmonary
systems fKorsaketal.. 2000: Korsaketal.. 1997: Korsakand Rydzynski. 19961 In these
studies, disturbances in CNS function, including decreased pain sensitivity and decreased
neuromuscular function and coordination appear to be the most sensitive endpoints
following exposure to 1,2,4-TMB. No subchronic studies were found that investigated
exposure to 1,3,5-TMB. One developmental study that was found (Saillenfait et al.. 2005]
that observed similar levels of maternal and fetal toxicity (i.e., decreased maternal weight
gain and fetal weight] following exposure to either isomer; other indices of fetal toxicity
(i.e., fetal death and malformations] were not affected by exposure.

Inhalation Reference Concentration (RfC) for 1,2,4-TMB

   The chronic RfC of 2 x 10-2 mg/m3 for 1,2,4-TMB was calculated from a BMDL of 84.0
mg/m3 (resulting in an PODADJ of 0.085 mg/L blood] for decreased pain sensitivity in
DRAFT - DO NOT CITE OR QUOTE                                                     24

-------
male rats exposed to 1,2,4-TMB via inhalation for 90 days (6 hours/day, 5 days/week]
[Korsakand Rydzynski. 1996). A PBPK model was then used to estimate a human
equivalent concentration of 15.6 mg/m3, which was used as the POD to derive the RfC. A
total UF of 1000 was used: 3 to account for uncertainty in extrapolating from laboratory
animals to humans (interspecies variability], 10 to account for variation in susceptibility
among members of the human population (interindividual variability], 10 to account for
subchronic-to-chronic extrapolation due the lack of a suitable chronic study, and 3 to
account for deficiencies in the database (no chronic study, no two-generation
reproductive/developmental toxicity study].

Table II. Reference concentration (RfC) for 1,2,4-TMB
Critical Effect
Decreased pain sensitivity

90 day rat study

Korsak and Rydzynski (1996)


Point of Departure (mg/m3)
PODHEc = 15.6
A PBPK model was used to
calculate an internal blood dose
from the rat inhalation study and
then a human equivalent
concentration was calculated. This
HEC served as the POD.
UF
1,000






Chronic RfC (mg/m3)
2 x 10"2






Confidence Levels for the Derivation of the RfC 1,2,4-TMB

   A confidence level of high, medium, or low is assigned to the study used to derive the
RfC, the overall database, and the RfC itself, as described in Section 4.3.9.2 of EPA's
Methods for Derivation of Inhalation Reference Concentrations and Application of
Inhalation Dosimetry (U.S. EPA. 1994]. Confidence in the study from which the  critical
effect was identified, Korsak and Rydzynski [1996] is medium. The study is a well-
conducted peer-reviewed study that utilized three dose groups plus untreated controls, an
appropriate number of animals per dose group, and performed statistical analyses.  The
critical effect on which the RfC is based is well-supported as the weight of evidence for
1,2,4-TMB-induced neurotoxicity is coherent across multiple animals species (i.e., human,
mouse, and rat] and consistent across multiple exposure durations (i.e., acute, short-term,
and sub-chronic] (Gralewicz and Wiaderna. 2001: Chen etal.. 1999: Wiaderna etal.. 1998:
Gralewicz etal.. 1997a: Gralewicz etal.. 1997b: Korsak and Rydzynski. 1996: Norseth et al..
1991]. Confidence in the database for 1,2,4-TMB is low to medium as the database includes
acute, short-term, subchronic, and developmental toxicity studies in rats and mice. The
database lacks a chronic and multigenerational reproductive study, and the studies
DRAFT - DO NOT CITE OR QUOTE                                                      25

-------
supporting the critical effect predominately come from the same research institute.  Overall
confidence in the RfC for 1,2,4-TMB is low to medium.

Inhalation Reference Concentration (RfC) for 1,3,5-TMB

   No chronic or subchronic studies exist that would support the derivation of an RfC for
1,3,5-TMB, however two short-term neurotoxicity studies and one developmental toxicity
study were identified as potential studies from which to identify a critical effect for RfC
derivation. Ultimately, the two short-term neurotoxicity studies were inappropriate for the
derivation of an RfC due to the magnitude of uncertainty associated with those data sets: in
order to use the endpoints from these studies, a total uncertainty factor of 10,000 would be
necessary: 3  to account for uncertainty in extrapolating from laboratory animals to humans
(interspecies variability], 10 to account for variation in susceptibility among members of
the human population (interindividual variability], 10 to account for extrapolation from a
LOAEL to a NOAEL, 10 to account for subchronic-to-chronic extrapolation due the lack of a
suitable chronic study, and 3 to account for deficiencies in the database (no chronic study,
no two-generation reproductive/developmental toxicity study].  Using the Saillenfait et al.
[2005] study and the most sensitive endpoint in that study, decreased maternal weight
gain, would result in an RfC 15-fold greater than that derived for 1,2,4-TMB (3 x 10-1 vs. 2
x 10-2 mg/m3]. This was deemed to not be scientifically justified as the toxicological
database for 1,2,4-TMB and 1,3,5-TMB, demonstrates that the two isomers are similar to
one another regarding respiratory, neurological, and developmental toxicity in acute and
developmental studies (Saillenfait et al.. 2005: Korsak and Rydzynski. 1996: Korsak et al..
1995], although 1,3,5-TMB was observed to induce neurotoxicity at a slightly greater
magnitude and earlier onset of effect compared to 1,2,4-TMB at the same exposure
concentration (Wiaderna etal.. 2002: Gralewicz and Wiaderna. 2001]. Additionally,
available toxicokinetic data regarding blood:air partition coefficients, respiratory uptake,
and absorption into the bloodstream in humans and rats do not suggest any appreciable
differences can be expected between the two isomers (Meulenberg and Vijverberg. 2000:
larnberg et al.. 1996: Dahletal.. 1988]. Therefore, given  the apparent similarities between
the two isomers (see Section 2.1.6], it was determined that the scientific database
supported adopting the RfC for 1,2,4-TMB for the RfC for 1,3,5-TMB. Thus, the chronic
RfC of 2 x 10-2 mg/m3 derived for 1,2,4-TMB was adopted as the RfC for 1,3,5-TMB
based on the determination of sufficient similarity regarding chemical properties,
kinetics, and toxicity between the two isomers.


DRAFT - DO NOT CITE OR QUOTE                                                      26

-------
Confidence Levels for the Derivation of the RfCfor 1,3,5-TMB

    As noted previously, a confidence level of high, medium, or low is assigned to the study
used to derive the RfC, the overall database, and the RfC itself, as described in EPA (1994}.
Section 4.3.9.2. The chronic RfC of 2 x 10-2 mg/m3 derived for 1,2,4-TMB was adopted as
the RfC for 1,3,5-TMB based on the conclusion that the two isomers were sufficiently
similar regarding chemical properties, kinetics, and toxicity. Thus, confidence in the study
from which the critical effect was identified, Korsakand Rydzynski (1996) is medium (see
above}.  Confidence in the database is low to medium as the database includes acute, short-
term, and developmental toxicity studies in rats and mice. The database lacks a chronic,
subchronic, and multigenerational reproductive study. Additionally, the studies supporting
the critical effect predominately come from the same research institute Overall confidence
in the RfC for 1,3,5-TMB is low due to uncertainties surrounding the adoption of the RfC
derived for 1,2,4-TMB as the RfC for 1,3,5-TMB.

Effects other than cancer observed following oral exposure

   No chronic, subchronic, or short-term studies were identified that examined the
noncancer effects of oral exposure to 1,2,4-TMB or 1,3,5-TMB. A series of oral or i.p.
injection studies were identified that investigated the acute neurotoxic effects of 1,2,4-TMB
and 1,3,5-TMB exposure fTomas et al.. 1999a: Tomasetal.. 1999b: Tomasetal.. 1999cj. In
these studies exposed rats demonstrated changes in electrocortical arousal, altered EEC
activity in the cortical and hippocampal regions of the brain, and increase locomotor
activity (possible due to difficulty maintain balance due to motor ataxia}. As these effects
were only observed in studies investigating acute exposures, they were not deemed
sufficient for derivation  of human health values.

Oral Reference Dose (RfD) for 1,2,4-TMB

   A PBPK model (Hissink et al.. 2007}. modified by EPA to include an oral compartment
was available for estimating the oral dose that would yield a blood concentration equal to
the blood concentration at the POD used in the derivation of the RfC for 1,2,4-TMB. Under
the assumption of constant oral ingestion and 100% absorption of 1,2,4-TMB via constant
infusion rate into the liver, a PODHED of 6.2 mg/kg-day was derived. Hepatic first pass
metabolism was also evaluated in humans using the modified PBPK model; at low daily
doses, inhalation doses were estimated to result in steady state venous blood

DRAFT - DO NOT CITE OR QUOTE                                                     27

-------
concentrations 4-fold higher than blood concentrations resulting from equivalent oral
doses, due to hepatic first pass metabolism.
   The same total UF of 1,000 as was used for the RfC derivation was applied: 3 to account
for uncertainty in extrapolating from laboratory animals to humans (interspecies
variability], 10 to account for variation in susceptibility among members of the human
population (interindividual variability], 10 to account for subchronic-to-chronic
extrapolation due the lack of a suitable chronic study, and 3 to account for deficiencies in
the database (no multigeneration reproductive/developmental toxicity study].

Table 3. Reference dose (RfD) for 1,2,4-TMB
Critical Effect
Decreased pain sensitivity

90 day rat study

Korsak and Rydzyriski (1996)
Point of Departure (mg/kg-
day)
Route to route
extrapolation using Korsak
and Rydzyriski (1996)
subchronic inhalation study
in Wistar rats
UF
1000




Chronic RfD (mg/kg-
day)
6 x 10"3




Confidence Levels for the Derivation of the RfD for 1,2,4-TMB

   A PBPK model was utilized to perform a route-to-route extrapolation to determine a
POD for the derivation of the RfD from the Korsak and Rydzynski [1996] inhalation study
and corresponding critical effect. The confidence in the study from which the critical effect
was identified, Korsak and Rydzynski [1996] is medium (see above]. Confidence in the
database for 1,2,4-TMB is low to medium as the database includes acute, short-term,
subchronic, and developmental toxicity studies in rats and mice. The database lacks a
multigenerational reproductive study, and the studies supporting the critical effect
predominately come from the same research institute. Overall confidence in the RfD for
1,2,4-TMB is low due to uncertainties surrounding the application of the available PBPK
model for the purposes of a route-to-route extrapolation.
Oral Reference Dose (RfD) for 1,3,5-TMB

   The oral database for 1,3,5-TMB includes no chronic, subchronic, or short-term oral
exposure studies.  However, as determined for the RfC derivation for 1,3,5-TMB, the
toxicokinetic and toxicological similarities between 1,3,5-TMB and 1,2,4-TMB demonstrate
sufficient similarity between the two isomers to support adopting the RfD for 1,2,4-TMB for
DRAFT - DO NOT CITE OR QUOTE                                                       28

-------
the RfD for 1,3,5-TMB. In addition to the previously discussed similarities in toxicokinetics,
the qualitative metabolic profiles for the two isomers are similar to such a degree that first-
pass metabolism through the liver is not expected to differ greatly between 1,2,4-TMB and
1,3,5-TMB. Therefore, the chronic RfD of 6 x 10-3 mg/kg-day derived for 1,2,4-TMB
was adopted as the RfD for 1,3,5-TMB based on the determination of sufficient
similarity regarding toxicokinetics and toxicity between the two isomers.

Confidence Levels for the Derivation of the RfD for 1,3,5-TMB

    As noted previously, a confidence level of high, medium, or low is assigned to the study
used to derive the RfD, the overall database, and the RfD itself, as described in EPA (1994}.
Section 4.3.9.2.  The chronic RfD of 6 x 10-3 mg/kg-day derived for 1,2,4-TMB was adopted
as the RfD for 1,3,5-TMB based on the conclusion that the two isomers were sufficiently
similar regarding chemical properties, kinetics, and toxicity. Thus, confidence in the study
from which the critical effect was identified, Korsakand Rydzynski (1996} is medium (see
above}.  Confidence in the database is low to medium as the database includes acute, short-
term, and developmental toxicity studies in rats and mice. The database lacks a
multigenerational reproductive study, and the studies supporting the critical effect
predominately come from the same research institute. Overall confidence in the RfD for
1,3,5-TMB is low due to uncertainties surrounding the adoption  of the RfD derived for
1,2,4-TMB as the RfD for 1,3,5-TMB.

Evidence for human carcinogenicity

   No chronic inhalation studies that investigated cancer outcomes were identified in the
literature for 1,2,4-TMB or 1,3,5-TMB. One oral cancer study was found in which rats were
exposed via oral gavage to one experimental dose of 800 mg/kg-day (Maltoni etal.. 1997}.
This  study observed marginal increases in total malignant tumors and head tumors (e.g.,
neuroesthesioepithelioma} and provided no statistical analyses of results. A number of
methodological  issues limit the utility of this study (only one dose group, no discussion of
histopathological analyses}. When Fisher's exact test was performed by EPA on the
incidences calculated from the reported percentages of animals bearing tumors in the
control and 800 mg/kg dose groups no statistically significant associations were observed.
Therefore, in accordance with the Guidelines for Carcinogen Risk Assessment (U.S. EPA.
2005aj. the database for 1,2,4-TMB was deemed to provide inadequate information to
assess carcinogenic potential, and thus, no cancer values for 1,2,4-TMB are derived in this
document. No studies of carcinogenicity were available for 1,3,5-TMB and thus the
DRAFT - DO NOT CITE OR QUOTE                                                      29

-------
database was deemed inadequate to assess carcinogenic potential; no cancer values were
derived for 1,3,5-TMB.
DRAFT - DO NOT CITE OR QUOTE                                                     30

-------
LITERATURE SEARCH STRATEGY AND  STUDY
EVALUATION
Literature Search Strategy and Study Selection

   The literature search strategy employed for 1,2,4-TMB and 1,3,5-TMB was conducted
with the keywords listed in Table VI. Primary, peer-reviewed literature identified through
December 2011 was included where that literature was determined to be relevant to the
assessment. Potentially relevant publications on 1,2,4-TMB and 1,3,5-TMB were identified
through a literature search conducted with the EBSCO Discovery Service feature of Health
and Environmental Research On-Line (HERO], a meta-search engine with access to
numerous databases including the Science Citation Index (SCI], Toxicology Literature
Online (TOXLINE), The National Library of Medicine (NLM, PubMed/Medline], and Web of
Science (WOS}. Other peer-reviewed information, including health assessments developed
by other organizations, review articles, literature necessary for the interpretation of TMB-
induced health effects, and independent analyses of the health effects data was retrieved
and included in the assessment where appropriate. A data call-in was announced by EPA in
April,  2008 and any pertinent scientific information submitted by the public to the IRIS
Submission Desk was also considered in the development of this document.

Table IV: Details of the search strategy employed for 1,2,4-TMB and 1,3,5-TMB
Databases
Keywords
Limits
EBSCO DISCOVERY
SERVICE:
HERO
SCI
NLM
TOXLINE
WOS
1,2,4-trimethylbenzene OR pseudocumene OR 95-63-6
1,3,5-trimethylbenzene OR mesitylene OR 108-67-8
Additional search terms: neurotoxicity, genotoxicity,
developmental toxicity inflammation, irritation, toxicokinetics,
pbpk, mode of action, white spirit, C9, C9 fraction, JP-8
Also, specific searches were performed on specific
metabolites: 2,4-dimethylbenzoic acid OR 611-01-8; 2,4-
dimethylhippuric acid OR 41859-41-0; 2,5-dimethylbenzoic acid
OR 610-72-0; 2,5-dimethylhippuric acid OR 41859-40-9; 3,4-
dimethylbenzoic acid OR 619-04-5; 3,4-dimethylhippuric acid OR
23082-12-4; 2,4,5-trimethylphenol OR 496-78-6; 2,3,5-
Search
constraints:
none

Last search:
December
2011
DRAFT - DO NOT CITE OR QUOTE
                                                              31

-------

trimethylphenol OR 697-82-5; 2,3,6-trimethylphenol OR 2416-
94-6; 2,4,6-trimethylphenol OR 527-60-6; 3,5-dimethylbenzoic
acid OR 499-06-9; 3,5-dimethylhippuric acid OR 23082-14-6

   The first step in the review of the available literature is the identification of studies
pertinent to the development of the document. These references include, but are not
limited to, studies related to: toxicokinetics, toxicity, carcinogenicity, mode of action, and
physiologically-based pharmacokinetic modeling.  The pertinent studies are then identified
for inclusion in the hazard identification based on a preliminary review of the overall study
design, with particular attention to the exposure route and duration.  The available
epidemiological and toxicological studies are further evaluated and identified for
consideration for quantitative analysis based on a more specific evaluation of the study
design, methods, and data quality.
   Approximately 2500 references were obtained from the literature searches for
1,2,4-TMB and 1,3,5-TMB including references retrieved from specific literature searches
necessary for the interpretation of TMB-induced health  effects (e.g., literature on specific
modes of action, PBPK analysis}. The comprehensive, unedited list of studies captured in
the literature search can be found on the HERO website. From this full list of references,
there are 583 references that were considered for inclusion in the Toxicological Review.
From this list of "considered" references, 112 full text publications were identified as
providing relevant information for use in the development of this document.
   The references that are cited in the document, as well as those that were considered but
not included in the Toxicological Review of 1,2,4-TMB and 1,3,5-TMB, can be found within
the HERO website (http(s]:hero.epa.gov/tmb}. This site contains HERO links to lists of
references, including bibliographic information and abstracts, which were considered for
inclusion in the Toxicological Review of 1,2,4-TMB and 1,3,5-TMB.  This document is not
intended to be a comprehensive treatise on the chemical or toxicological nature of 1,2,4-
TMB and 1,3,5-TMB.

Study Evaluation for Hazard Identification

   In general, the quality and relevance of health effects studies were evaluated as outlined
in the Preamble to this assessment. In addition, A Review of the Reference Dose and
Reference Concentration Processes [U.S. EPA. 2002) and Methods for Derivation of
Inhalation Reference Concentrations and Application of Inhalation Dosimetry [U.S. EPA.
DRAFT - DO NOT CITE OR QUOTE                                                       32

-------
1994) were consulted for guidance in evaluating scientific quality of the available studies.
All studies that were considered to be of acceptable quality, whether yielding positive,
negative, or null results, were considered in assessing the totality of the evidence for health
effects in humans. The hazard identification analyses for each health endpoint in Section 1
discuses the breadth of the available literature and the extent to which the studies
informed the conclusions concerning hazard. The available studies examining health
effects of 1,2,4-TMB and 1,3,5-TMB exposure in humans (three occupational exposure
studies, one cross-section residential study, and two controlled experiments of acute
exposures] are discussed and evaluated in the hazard identification sections of the
assessment (Section 1], with specific limitations of individual studies and of the collection
of studies noted. The evaluation of the effects seen in the experimental animal studies
focuses  on the available acute, short-term, subchronic, and developmental toxicity studies,
as no chronic inhalation exposure studies were  found and the only identified chronic oral
exposure study did not include data on effects other than cancer.
DRAFT - DO NOT CITE OR QUOTE                                                        3 3

-------
   1.  HAZARD IDENTIFICATION
1.1. Synthesis of Major Toxicological Effects

1.1.1. Neurotoxic Effects

   There is evidence in humans and animals that inhalation exposure to TMBs induces
neurotoxic effects.  Occupational exposure studies in humans provide evidence of
neurotoxicity following inhalation exposure to complex volatile organic compound (VOC)
mixtures containing 1,2,4-TMB and 1,3,5-TMB. Prevalence rates of neuropsychological
symptoms increased with exposure duration in dockyard painters [Chen etal.. 1999):
similarly, a significant positive association between 1,2,4-TMB exposure and exposure
symptoms (e.g., abnormal fatigue] was reported in asphalt workers (Norseth etal.. 1991}.
Nervousness, tension, headaches, vertigo, and anxiety were reported in paint shop workers
exposed to 49-295 mg/m3 of a solvent mixture containing 50% 1,2,4-TMB and 30%
1,3,5-TMB (Battig et al. (1956). as reviewed by [MOE. 2006)).  Increased fatigue, decreased
vigor, and increased reaction time were noted in controlled, acute volunteer studies in
which humans were exposed to mixtures containing 1,2,4-TMB (Lammers etal..  2007).
although it is unclear whether 1,2,4-TMB or other constituents within the mixtures were
responsible for the observed effects. Uptake of 1,2,4-TMB and 1,3,5-TMB was reported in
volunteers exposed for 2 hours to 11 or 123 mg/m31,2,4-TMB, or 123 mg/ m31,3,5-TMB;
however, effects on the CNS, based on measures of overt CNS depression (heart rate during
exposure and pulmonary ventilation) and a subjective rating of CNS symptoms (data not
reported), were not observed (Jarnbergetal.. 1996). The Jarnberg et al. (1997a: 1996]
studies are limited for evaluating neurotoxicity to TMBs due to a lack of methods to
adequately assess CNS function and lack of no-exposure controls, short exposure duration,
and exposure of individual subjects to multiple, different concentrations of TMB isomers
and/or mixtures containing TMBs.
   In animals, there is consistent evidence of neurotoxicity following inhalation exposure
to either 1,2,4-TMB or 1,3,5-TMB. Decreased pain sensitivity has been observed following
inhalation exposure to 1,2,4-TMB and 1,3,5-TMB in multiple studies conducted in male
Wistar rats. To test pain responses following TMB exposure, animal studies have employed
the hot plate test. In this test, a thermal stimulus is applied to determine pain sensitivity, as
indicated by the animals' latency to paw-lick following introduction of the stimulus.
DRAFT - DO NOT CITE OR QUOTE                                                     34

-------
Decreases in pain sensitivity have been observed at concentrations > 492 mg/m3 following
subchronic and short-term exposure to 1,2,4-TMB (Wiaderna etal.. 2002: Gralewicz and
Wiaderna. 2001: Korsak and Rydzynski. 1996} and short-term exposure to 1,3,5-TMB
(Wiaderna et al.. 2002: Gralewicz and Wiaderna. 2001}. In the subchronic study (Korsak
and Rydzynski. 1996}. 1,2,4-TMB inhalation resulted in reduced pain sensitivity which
occurred in a concentration-dependent manner. In short-term studies that examined a
range of concentrations [Wiaderna etal.. 2002: Gralewicz etal.. 1997aj these decreases in
pain sensitivity following exposure to 1,2,4-TMB or 1,3,5-TMB were non-monotonic.
Differences in experimental design may account for the lack of monotonicity in these short-
term studies, in contrast to the observations in Korsak and Rydzynsky, (1996}.  Similar to
the subchronic study, acute exposures to 1,2,4-TMB or 1,3,5-TMB induced concentration-
dependent decreases in pain sensitivity, with ECso values of 5,682 and 5,963 mg/m3,
respectively (Korsak and Rydzynski. 1996: Korsak etal.. 1995}.
   A second, somewhat different measure of pain sensitivity was reported in studies
evaluating performance in the hot plate test (before and after footshock} several weeks
following short-term, inhalation exposure to 1,2,4-TMB or 1,3,5-TMB (Wiaderna et al..
2002: Gralewicz and Wiaderna. 2001: Gralewicz etal.. 1997aj. In these studies, treatment-
related, statistically significant changes in pain sensitivity at > 492 mg/m31,2,4-TMB or
1,3,5-TMB were observed 24 hours after rats were given a footshock; no statistically
significant effects at any exposure concentration were observed prior to or immediately
following footshock. These findings indicate that inhalation exposure to TMBs may prolong
footshock-induced reductions in pain sensitivity.  It is also plausible that an amplification of
responses associated with classically conditioned analgesia (i.e., decreased pain sensitivity}
occurs following 1,2,4-TMB and 1,3,5-TMB exposure. Specifically, footshock can cause
contextual cues (e.g., hot plate test} to become associated with the noxious stimulus
(footshock}, inducing stress or fear-related responses in the shocked animal such that,
subsequently, both footshock itself as well as the contextual cues associated with
footshock, can reduce sensitivity to pain (possibly via the release of endogenous opiods}.
Thus, exposure to the hot plate apparatus immediately following footshock may associate
this test environment with the footshock, such that subsequent re-exposure to the hot plate
apparatus can, itself, produce analgesia. From the data available, the relative contribution
of these behaviors to the observed effects cannot be easily distinguished.
   The decreases in pain sensitivity measured in the subchronic and acute studies were
observed immediately after exposure, with no significant effects persisting two  weeks after
exposures were terminated (Korsak and Rydzynski. 1996: Korsak et al.. 1995}.  In contrast,
performance in the hot plate test was significantly impaired following short-term exposure
DRAFT - DO NOT CITE OR QUOTE                                                       3 5

-------
to either 1,2,4-TMB or 1,3,5-TMB when tested 50-51 days after exposure (Wiaderna et al..
2002: Gralewicz and Wiaderna. 2001: Gralewicz etal.. 1997a). indicating a persistence of
these effects. The ability of male Wistar rats to respond to a thermal stimulus in the hot
plate test was consistently impaired following inhalation exposure to TMBs. In these
studies, 1,2,4-TMB and 1,3,5-TMB similar in their capacity to decrease pain sensitivity
(Table 1-1}. Pain sensitivity was not examined following oral exposure.
   Human exposures to solvent mixtures containing 1,2,4-TMB (Lammers etal.. 2007) or
1,2,4-TMB and 1,3,5-TMB (Battig etal.. 1956). as reviewed by MOE (2006) suggest possible
effects on the neuromuscular system, as effects reported included increased reaction time
and vertigo, respectively. Animal studies using rotarod performance, which tests motor
coordination, balance, and overall neuromuscular function, indicate that inhalation of 1,2,4-
TMB or 1,3,5-TMB can affect neuromuscular system function. Significant decreases in
rotarod performance were observed at 1230 mg/m31,2,4-TMB when tested immediately
after exposure for either 8 or 13 weeks (Korsakand Rydzynski. 1996). This impaired
function was still evident at 2 weeks post-exposure and, while not statistically significant,
may indicate long-lasting neuromuscular effects of subchronic exposures to 1,2,4-TMB.
Acute inhalation exposure studies support this observation. Effects such as loss of reflexes
and righting responses have been observed following acute inhalation exposure to 1250-
45,000 mg/m3 of 1,2,4-TMB (MOE. 2006: Henderson. 2001). Similarly, acute exposure to
1,2,4-TMB or 1,3,5-TMB resulted in decreased performance in rotarod tests immediately
following exposure, with ECso values of 4693 mg/m3 and 4738 mg/m3, respectively
(Korsak and Rydzynski. 1996: Korsaketal.. 1995): these results indicate the 1,2,4-TMB and
1,3,5-TMB may be similar in their ability to impair neuromuscular function, balance, and
coordination following acute inhalation exposure. No studies evaluating oral exposure to
TMB isomers address this endpoint.
   The neurobehavioral tests administered (i.e., hot plate and rotarod) in the subchronic
and acute studies by Korsak and Rydzynsky, (1996) and Korsak et al. (1995) appear to
have been administered on the same days; however, it is unclear whether the tests were
performed sequentially in the same cohorts of animals.  Performing the hot plate test
immediately following the rotarod test could introduce a potential confounder, as shock
alone (such as that used as negative reinforcement following rotarod failure) can cause
reductions in pain sensitivity. Thus, if the tests were performed sequentially in the same
animals, TMB-exposed animals failing more often in the rotarod test may exhibit increases
in paw-lick latency unrelated to treatment, as compared to controls receiving less shock
reinforcement. However, the observations by Korsak and Rydzynsky, (1996) and  Korsak et
al. (1995) are supported by 2.8 and 2.9-fold increases in latency to paw-lick that, although
DRAFT - DO NOT CITE OR QUOTE                                                      36

-------
not statistically significant, were observed weeks subsequent to short-term exposures to
492 mg/m31,2,4-TMB or 1,3,5-TMB fGralewicz and Wiaderna. 20011
   Effects in open field testing have been consistently reported in oral and inhalation
studies of exposure to 1,2,4-TMB and 1,3,5-TMB in male rats. Altered behaviors in open
field tests can involve contributions not only from elevated anxiety due to open spaces and
bright light but also from changes in motor function. Decreased anxiety and/or increased
motor function at > 492 mg/m31,2,4-TMB or 1,3,5-TMB has been reported in short-term
studies, as evidenced by increases in horizontal locomotion or grooming activities
(Gralewicz and Wiaderna. 2001: Gralewicz etal.. 1997a}. Statistically significant increases
in horizontal locomotion were observed in short-term studies assessing open field
behavior following inhalation exposure to either isomer [Gralewicz and Wiaderna. 2001).
Non-monotonic increases in grooming were reported following short-term exposure to
1,2,4-TMB (with a statistically significant increase only in the mid-exposure group};
changes in horizontal locomotion were not statistically significant (increases of 3-35%
were non-monotonic) (Gralewicz etal.. 1997a). As open field testing was conducted 25
days after termination of exposure in these studies, the results suggest latency for the
effects on anxiety and/or motor function.
   Slight increases in locomotor activity were also observed in open field tests
immediately following acute, oral exposure to 1,2,4-TMB or 1,3,5-TMB.  Significant
increases were observed at 3850 mg/kg for 1,2,4-TMB and at > 1,920 mg/kg for 1,3,5-TMB,
with minimal dose-effect or time-effect relationships and negligible differences in the
magnitude of the change in activity between isomers (Tomas et al., 1999b). The study
authors attributed these changes to discomfort arising from difficulty in breathing and/or
maintaining balance due to the onset of motor ataxia; notably, by 90 minutes following
exposure, the rats were reported to be completely inactive and several rats in the high dose
group died within 24 hours (Tomas etal.. 1999b). Open field tests cannot easily distinguish
between anxiety-related responses and changes in motor activity. However, effects on
motor activity were observed following inhalation exposure to elevated concentrations of
TMBs in several acute studies, although the results are somewhat inconsistent with
observations in open field tests.  Decreased motor activity was observed in male rats
immediately after exposure at 5000 mg/m31,2,4-TMB (McKee etal.. 2010). Decreased
motor activity was also reported in rats acutely exposed to a mixture containing TMB
isomers (Lammers etal.. 2007). but the use of a mixture precludes a determination of the
toxicity specifically associated with 1,2,4-TMB or 1,3,5-TMB.  As biphasic changes in
activity are frequently observed following exposures to solvents, it is likely that the timing
of the evaluations conducted in the short-term versus acute studies may influence the
DRAFT - DO NOT CITE OR QUOTE                                                       37

-------
consistency of these results. Overall, exposure to 1,2,4-TMB and 1,3,5-TMB affects anxiety
and/or motor function at concentrations above 492 mg/m3, although the exact, potentially
biphasic, concentration-response relationship remains unclear.
   Cognitive function following exposure to 1,2,4-TMB or 1,3,5-TMB has not been
evaluated in humans or following oral exposure in animals; exposure of human volunteers
to mixtures containing TMBs did not indicate any effects on short-term learning and
memory tests (Lammers etal.. 2007). Similarly, short-term spatial memory (radial maze
performance] was unaffected by exposure to either TMB isomer in animal studies
(Wiaderna etal.. 2002: Gralewicz and Wiaderna. 2001: Gralewicz etal.. 1997a}. In contrast,
effects on cognitive function in different neurobehavioral tests, observed as altered
conditioning behaviors, were  consistently observed in multiple studies in male rats weeks
following short-term inhalation exposure to 1,2,4-TMB or 1,3,5-TMB, although clear
concentration-effect relationships were not observed. Comparing the results of the
behavioral tests reveals that there are differences in these neurological effects reported for
1,2,4-TMB and 1,3,5-TMB, as well as differences in the exposure concentrations at which
the cognitive effects were observed. Decreased step-down latency in passive avoidance
tests 35-45 days after short-term inhalation exposure to > 492 mg/m31,2,4-TMB or > 123
mg/m31,3,5-TMB was observed in treated male rats (Wiaderna etal.. 2002: Gralewicz and
Wiaderna. 2001: Gralewicz etal.. 1997a}: decreases may be attributed to a reduced ability
to inhibit motor reactions  (or a lowered motor threshold] in response to stress. These
responses were consistently observed and similar in magnitude across all studies at 7 days
post footshock. Statistically significant changes were not observed < 24 hours following
footshock and were not consistently observed 3 days following footshock, suggesting that
1,2,4-TMB and 1,3,5-TMB exposure may affect adaptive behaviors associated with the
persistence of stress or fear-related responses. Reduced active avoidance learning was also
observed in male rats following short-term inhalation exposure to 492 mg/m31,2,4-TMB
(Wiaderna etal.. 2002: Gralewicz and Wiaderna. 2001]: however, these changes were not
observed in the other 1,2,4-TMB short-term study (Gralewicz etal.. 1997a]. Decreased
performance in active avoidance tests was consistently observed following short-term
exposure to > 492 mg/m31.3.5-TMB(Wiaderna etal.. 2002: Gralewicz and Wiaderna.
20011 Similar to  1,2,4-TMB fWiaderna etal.. 2002: Gralewicz and Wiaderna. 20011 the
effects of 1,3,5-TMB were particular to the learning component of the test (acquisition
training], rather than the memory component (retention session 7 days later].  It is unclear
whether potential TMB-induced alterations in locomotor activity would affect performance
in these tests. Acute inhalation exposure studies provide some support for the observed
effects of TMBs on learned behaviors. Significant increases in response latency in
DRAFT - DO NOT CITE OR QUOTE                                                      3 8

-------
psychomotor tasks, observed immediately after exposure (effects did not persist to 24
hours later], were reported in male rats following acute exposure to 5000 mg/m31,2,4-
TMB (McKee et al., (2010}} or to 4800 mg/m3 of a mixture containing TMBs (Lammers et
al.. 2007}. The effects on active and passive avoidance behaviors indicate that learning
and/or long-term memory processes are affected by exposure to 1,2,4-TMB or 1,3,5-TMB.
The data suggest that 1,3,5-TMB may be a more potent inducer of toxic effects on cognitive
function than 1,2,4-TMB, as the effects following exposure to 1,3,5-TMB occurred at lower
concentrations (123 mg/m3 vs. 492 mg/m3, were of greater intensity and occurred earlier
than those reported following exposure to 1,2,4-TMB.
   Controlled human exposure studies suggest that exposures of < 123 mg/m31,2,4-TMB
or 1,3,5-TMB do not cause overt CNS depression (larnberg et al.. 1996}. although
symptoms related to this effect (e.g., lightheadedness, fatigue} have been reported in
workers occupationally exposed to mixtures containing TMBs. In animals, CNS depression
has been observed following acute inhalation exposure to 25,000-44,000 mg/m31,3,5-TMB
(ACGIH. 2002}. Neurophysiological evidence from short-term inhalation studies, as well as
supportive evidence from acute oral and injection studies, suggests that exposures to 1,2,4-
TMB or 1,3,5-TMB at lower concentrations (at least for 1,2,4-TMB} may affect parameters
associated with brain arousal. Concentration-dependent decreases (statistically significant
at 1230 mg/m3} in electrocortical arousal (i.e., spike-wave discharge activity in recordings
from cortical-hippocampal electroencephalograms, EEGs}  were observed in male rats 120
days after short-term exposure to > 492 mg/m31,2,4-TMB, suggesting persistent functional
changes in the rat CNS (Gralewicz etal.. 1997b}. In recordings from rats that were awake,
but immobile (not exhibiting pronounced exploratory activity, as determined by EEC
morphology}, statistically significant decreases in spike wave discharge activity were
observed at 24 hours following short-term exposure to 492 mg/m31,2,4-TMB (Gralewicz
etal.. 1997b}. Dose-related decreases in spike wave discharges were observed at > 240
mg/kg 1,2,4-TMB or 1,3,5-TMB subsequent to acute oral exposure (Tomas etal.. 1999a}:
stronger and more persistent effects on electrocortical arousal were observed following
oral exposure to 1,3,5-TMB compared with 1,2,4-TMB (Tomas etal.. 1999a}.  Similar effects
were observed followingi.p. injection of 1,2,4-TMB and 1,3,5-TMB (Tomas etal.. 1999c}.
The observed EEC abnormalities following inhalation and  oral exposure to 1,2,4-TMB or
1,3,5-TMB provide supportive evidence of the CNS depressant effects of these compounds
(Gralewicz etal..  1997b}.
   A summary of the observed neurotoxicity for 1,2,4-TMB and 1,3,5-TMB is shown in
Tables 1-1 and 1-2 for inhalation and oral exposures, respectively.

DRAFT - DO NOT CITE OR QUOTE                                                     3 9

-------
Table 1-1: Summary of observed in vivo neurotoxicity in subchronic and short-term
studies of male Wistar rats following inhalation exposure to 1,2,4-TMB or 1,3,5-TMB
   Health Effect
 Study Design and Reference
                 Results
Lowest Level
  at which
 Significant
Effects were
  observed
  (mg/m3)3
                                              1,2,4-TMB
  Decreased Pain
    Sensitivity
                  0, 123, 492, 1,230 mg/m
                  recovery (1,230 mg/ m3 at 2
                  weeks post-exposure)
                  90 days; 10/group
                  Korsak & Rydzynsky (1996)
0, 492 mg/m3
4 weeks; 11/group
Gralewicz & Wiaderna (2001)
                  0, 123, 492, 1,230 mg/m3
                  4 weeks; 15/group
                  Gralewicz et al. (1997a)
                            Exposure-dependent increases in paw-lick
                            latency which recover by two weeks post-
                            exposure.

                            Response relative to control: 0,18, 79*, 95*%
                            (recovery= 12%)
Increased paw-lick latency 24 hours after
intermittent footshockb.
                                               Response relative to control: 0,191*%
                            Non-monotonic increases in paw-lick latency
                            24 hours after intermittent footshockb.
                                               Response relative to control: 0, 9, 61*, 46*%
                                               492
                                                                                             492
                                                                           492
     Impaired
  Neuromuscular
   Function and
   Coordination
0, 123, 492, 1,230 mg/m
recovery (1,230 mg/m3 at 2
weeks post-exposure)
90 days; 10/group
Korsak & Rydzynsky (1996)
Exposure-dependent increases in rotarod
failures which do not recover by two weeks
post exposure.

Response relative to control: 0,10, 20, 40*%
(recovery= 30%)
   1,230
 Decreased Anxiety
 and/or Increased
  Motor Function
                  0, 492 mg/m
                  4 weeks; 11/group
                  Gralewicz & Wiaderna (2001)
                            Increased horizontal locomotion in open field
                            activity tests.

                            Response relative to control: 0, 62*%
                  0, 123, 492, 1,230 mg/m
                  4 weeks; 15/group
                  Gralewicz et al. (1997a)
                            Non-monotonic increases in grooming in open
                            field activity tests at middle concentration; no
                            change in horizontal locomotion.

                            Response relative to control: 0, 82,147*, 76%
                                               492
                                               492
 Altered Cognitive
     Function
                  0, 492 mg/m
                  4 weeks; 11/group
                  Gralewicz & Wiaderna (2001)
                            Decreased step down latency in passive
                            avoidance tests and decreased performance in
                            active avoidance tests.

                            Response relative to control: 0, 43*%c; 0,
                            60*%d
                  0,123, 492, or 1,230 mg/m
                  4 weeks; 15/group
                  Gralewicz et al. (1997a)
                            Non-monotonic decreases in step down latency
                            in passive avoidance tests.

                            Response relative to control: 0, 21, 81*, 49*%c;
                                               492
                                               492
DRAFT - DO NOT CITE OR QUOTE
                                                                                40

-------

Decreased Cortico-
Hippocampal
Activity

0, 123, 492, 1,230 mg/m3
4 weeks; 9/group
Gralewicz et al. (1997b)
0,30, 27, 34% e
Non-monotonic decreases in spike-wave
discharges at 24 hours post-exposure (EEG) at
middle concentration.
Response relative to control: 0, 31, 64*, 19%

492
DRAFT - DO NOT CITE OR QUOTE
41

-------
  Decreased Pain
    Sensitivity
                   0, 492 mg/m
                   4 weeks; 11/group
                   Gralewicz & Wiaderna (2001)
                             1,3,5-TMB
                              Increased paw-lick latency 24 hours after
                              intermittent footshockb.

                              Response relative to control: 0, 250*%
                   0, 123, 492, 1,230 mg/m3
                   4 weeks; 12/group
                   Wiaderna et al. (2002)
                              Non-monotonic increases in paw-lick latency
                              24 hours after intermittent footshock at middle
                              concentration11.

                              Response relative to control: 0, 4, 70*, 17%
                                                 492
                                                 492
 Decreased Anxiety
 and/or Increased
  Motor Function
0, 492 mg/m
4 weeks; 11/group
Gralewicz & Wiaderna (2001)
Increased horizontal locomotion in open field
activity tests.

Response relative to control: 0, 70*%
492
                   0, 123, 492, 1,230 mg/m
                   4 weeks; 12/group
                   Wiaderna et al. (2002)
 Altered Cognitive
     Function
                              Non-monotonic decreases in step down latency
                              in passive avoidance tests and in performance
                              in active avoidance tests.

                              Response relative to control: 0, 48*, 55*, 46*%c
                              ; 0, 40*, 35*, 50*%d
                   0, 492 mg/m3
                   4 weeks; 11/group
                   Gralewicz & Wiaderna (2001)
                              Decreased step down latency in passive
                              avoidance tests and decreased performance in
                              active avoidance tests.

                              Response relative to control: 0, 57*%c;
                              0, 70*%d
                                                 123
                                                 492
* Significantly different from controls
For studies other than Korsak and Rydzynsky, 1996, % change from control calculated from digitized data using Grablt
software
a Lowest effect level at which statistically significant changes were observed
b This effect was only observed 24 hours following intermittent foot shock; no significant effects at any
exposure were observed prior to or immediately following foot shock
c Decreased step down latency in passive avoidance tests at 7 days post footshock
 Increased number of trials to reach avoidance criteria
e Decreased avoidance response % in trials 25-30 of training
DRAFT - DO NOT CITE OR QUOTE
                                                                                    42

-------
Tables 1-2: Summary of observed in vivo neurotoxicity for 1,2,4-TMB and 1,3,5-TMB
— oral exposures
Health Effect
Study Design and Reference
Results
Lowest Level
at which
Significant
Effects were
observed
(mg/kg)a
1,2,4-TMB
Decreased Cortico-
Hippocampal
Activity
Decreased Anxiety
and/or Increased
Motor Function
Decreased Cortico-
Hippocampal
Activity
240, 960, 3,850 mg/kg, single
oral gavage
Rat, Wag/Rij, male, 18/group
Tomas et al. (1999a)
960, 1920, 3850 mg/kg single
oral gavage
Rat, Wag/Rij, male, 10/group
Tomas et al. (1999b)
790 mg/kg, single i.p injection
Rat, Wistar, Male, 4
Tomas etal. (1999c)
Inhibition of the number and duration of high
voltage spindle activity in the hippocampus and
cortex.
Response relative to control: All doses
produced differences from control during at
least one of the measured time points
Slight increase in spontaneous locomotor
activity in open field test.
Response relative to control: Significant
difference from control reported at 3,850
mg/kg when data were considered by time
points (i.e., dose x time interaction)
Significant differences in hippocampal and
cortical brain wave amplitude following
injection.
Response relative to control: cortical wave
amplitude decreased up to 6.5%; hippocampal
wave amplitude decreased up to 59.6%
240
3,850
790
1,3,5-TMB
Decreased Cortico-
Hippocampal
Activity
Decreased Anxiety
and/or Increased
Motor Function
Decreased Cortico-
Hippocampal
Activity
240, 960, 3,850 mg/kg, single
oral gavage
Rat, Wag/Rij, male, 18/group
Tomas et al. (1999a)
960, 1920, 3850 mg/kg single
oral gavage
Rat, Wag/Rij, male, 10/group
Tomas et al. (1999b)
790 mg/kg, single i.p injection
Rat, Wistar, Male, 4
Tomas et al. (1999c)
Inhibition of high voltage spindle activity in the
hippocampus and cortex.
Response relative to control: All doses
produced differences from control during at
least one of the measured time points
Slight increase in spontaneous locomotor
activity in open field test.
Response relative to control: Significant
difference from control reported at 1,920 and
3,850 mg/kg
Significant differences in hippocampal and
cortical brain wave amplitude following
injection.
Response relative to control: cortical wave
amplitude decreased up to 13.9%;
hippocampal wave amplitude decreased up to
38%
240
1920
790
* Significantly different from controls
a Lowest effect level at which statistically significant changes were observed
DRAFT - DO NOT CITE OR QUOTE
43

-------
   > Mode of Action Analysis for neurotoxicity

   The observation of neurotoxicity following acute-, short-term-, and subchronic-
duration exposure to TMB (Lammers etal.. 2007: Wiaderna etal.. 2002: Gralewicz and
Wiaderna. 2001: Wiaderna et al.. 1998: Gralewicz et al.. 1997a: Gralewicz et al.. 1997b:
Korsak and Rydzynski. 1996: Korsaketal.. 1995} may indicate that TMB perturbs normal
neurotransmission in exposed animals (see Table 1-3], although the specific key events
necessary for TMB-induced neurotoxicity are not established. Although limited
mechanistic data for TMBs exists, structurally similar compounds like toluene and xylene
have been more thoroughly characterized and it is hypothesized that TMBs would operate
through a similar mechanism in producing the resultant neurotoxicological effect.
Aromatic hydrocarbons are known to interact with catecholaminergic systems [Kyrklund.
1992). Inhalation exposures to toluene and xylene have been shown to significantly change
concentration and turnover rate of both dopamine and norepinephrine in various regions
of the rat brain (Rea etal.. 1984: Andersson etal.. 1983: Andersson etal.. 1981: Andersson
etal.. 1980). These changes have been hypothesized to be due to potential metabolites
with affinity to catecholamine receptors that would, in turn, influence the uptake and
release of neurotransmitters [Andersson etal.. 1983: Andersson etal.. 1981: Andersson et
al.. 1980).
   Catecholaminergic changes with toluene have been reported and are similar to that
observed with TMBs which would therefore increase the plausibility that the mechanisms
of neurotoxicity are similar between the two compounds.  For example, subchronic
inhalation exposures of rats to low concentrations of toluene (as low as 80 ppm [300
mg/m3]) have been shown to decrease spatial learning and memory, increase dopamine-
mediated locomotor activity, increase in the number of dopamine D2 receptors, and
increase dopamine D2 agonist receptor binding (Hillefors-Berglund et al.. 1995: von Euler
etal.. 1994: von Euler et al.. 1993). These effects were observed to persist up to four weeks
after the termination of the toluene exposure. Activation of the dopaminergic system may
also result in an inability to inhibit locomotor responses normally suppressed by
punishment (Jackson and Westlind-Danielsson. 1994). Direct application of dopamine to
the nucleus accumbens of rats has been observed to result in retardation of the acquisition
of passive avoidance learning at concentrations that also stimulated locomotor activity
(Braes etal.. 1984). Increases in catecholaminergic neurotransmission (through exposure
to norepinephrine or dopamine agonists) result in dose-dependent reductions in the
duration of spike wave discharges in rats (Snead. 1995: Warter etal.. 1988). These
observations and findings are in concordance with those resulting from exposure to

DRAFT - DO NOT CITE OR QUOTE                                                     44

-------
1,2,4-TMB and 1,3,5-TMB fWiaderna etal.. 2002: Gralewicz and Wiaderna. 2001:
Gralewiczetal.. 1997a: Gralewicz etal.. 1997b1 fTomas etal.. 1999a: Tomas etal.. 1999d.
Additionally, with regards to toluene and related aromatic hydrocarbons, it is known that
there is direct interaction with these compounds on various ion channels (ligand and
voltage gated] that are present in the central nervous system (Bowen etal.. 2006: Balster.
1998). There is not enough information to ascertain the specific molecular sites and how
the changes correlate to the observed neurotoxicological effects. However, it is widely
believed that the interactions with the neuronal receptors in the brain (e.g. ion channels,
catecholaminergic systems] may influence these changes.
   Aromatic hydrocarbons may also affect the phospholipids in the nerve cell membrane
[Andersson etal.. 1981]. Pertubation of the phospholipids on the cell membrane could
indirectly affect the binding of neurotransmitters to the catecholamine receptors and
potentially lead to alterations in receptor activity or uptake-release mechanisms. Uneven
distribution of metabolites within differing regions of the brain, or spatial variations in
phospholipid composition of nerve cell membranes may explain the differential effects
seen in regard to catecholamine levels and turnover. Based on effect levels with other
related solvents (e.g., toluene - see  Balster (1998]]. it is overall hypothesized that with
TMBs there may be an initial interaction with the neuronal receptors (e.g.,
catecholaminergic systems, ion channels]  followed by, at much higher exposures,
interaction with the lipid membrane when the available sites on the neuronal receptors are
completely occupied.
   Additional mechanisms that may play a role in TMB neurotoxicity include production of
reactive oxygen species (ROS]. Myhre et al. (2000] found increased respiratory burst in
neutrophils after 1,2,4-TMB exposure demonstrated by fluorescence spectroscopy,
hydroxylation of 4-hydroxybenzoic acid, and electron paramagnetic resonance
spectroscopy. The authors suggest the observation of solvent-induced ROS production may
relevant to brain injury as microglia cells have a respiratory burst similar to neutrophils.
Stronger evidence of potential ROS-related mechanisms of neurotoxicity was observed in a
related study by Myhre and Fonnum (2001] in which rat neural synaptosomes exposed to
1,2,4-TMB produced a dose-dependent increase in reactive oxygen and nitrogen species
demonstrated by the formation of the fluorescence of 2'7'-dichlorofluorescein (DCF]. This
observation of ROS production in rat synaptosomes may explain the observed TMB-
induced neurotoxicity in acute, short-term, and subchronic inhalation studies.
DRAFT - DO NOT CITE OR QUOTE                                                      45

-------
   7s the hypothesized mode of action sufficiently supported in test animals?

   The hypothesis that TMB exposure results in abnormal neurotransmission in animals is
supported by the available literature, including the observation that related methylated
aromatic compounds (i.e., toluene and xylene} perturb the catecholaminergic system and
elicit similar neurological effects as TMB isomers.

   7s the hypothesized mode of action relevant to humans?

The observed neurotoxic effects in animals are relevant to humans, especially given the
observation of similar neuropsychological effects in humans exposed to complex solvent
mixtures containing TMB isomers.

   In summary, neurotoxicity is associated with exposure to 1,2,4-TMB and 1,3,5-TMB
based on evidence in humans and animals. The information regarding neurotoxicity in
humans is limited and most of the available studies evaluating these effects involve
exposure to mixtures containing TMB isomers and not specific exposure to the individual
isomers. Additionally, none of the available studies have addressed the potential for latent
neurological effects or effects in sensitive populations. However, the available information
shows uptake of 1,2,4-TMB and 1,3,5-TMB in humans and suggests an association between
TMB exposure in humans and neurotoxic effects. The observation of neurotoxicity in
1,2,4-TMB-exposed male Wistar rats was consistent across multiple exposure
concentrations, including subchronic, short-term, and acute exposures. Similar indices of
neurotoxicity were observed in male rats exposed to 1,3,5-TMB for short-term and acute
durations. Although the oral database is limited, similar effects were observed (e.g., EEC;
open field] across inhalation and oral study paradigms.
   All of the available animal studies were conducted in male rats (Wistar or Wag/Rij} and
by the same research group (The Nofer Institute of Occupational Medicine, Lodz Poland}.
No chronic studies are available, although there is consistent evidence of neurotoxicity
following inhalation and oral exposure to 1,2,4-TMB and 1,3,5-TMB. Most of the
neurotoxicity tests incorporated the application of footshock which could involve multiple
neurological functions. The spectrum of effects suggests that TMBs affect multiple, possibly
overlapping, CNS systems rather than a single brain region or neuronal nuclei (suggested
by the solvent activity of the compounds}. Almost all tests (other than pain} involve a
contributing component of motor system function. Some endpoints exhibited clear
exposure-response relationships (e.g., pain sensitivity and rotarod, although the former
was not consistent across studies with different experimental design (i.e., varying exposure
DRAFT - DO NOT CITE OR QUOTE                                                      46

-------
durations and timing of endpoint analyses}. Other endpoints did not show a clear
concentration-effect relationship, suggesting either that exposures below a threshold value
were not tested or do not exist, or that the presence of TMBs alone was sufficient to elicit a
response in these tests, possibly via irritation or stress-related phenomena.  However,
irritation is highly unlikely given the latency between the exposures and the effects.
      Multiple neurotoxic effects were observed weeks to months after cessation of
inhalation exposure despite rapid clearance of these chemicals from blood and CNS tissues
(see Appendix A], indicating that these effects are persistent. Although the reported human
symptoms do not directly parallel the animal data, some similar effects were observed in
both humans and rats. The majority of the neurotoxicity evidence available for TMBs was
observed in laboratory animals as neurobehavioral effects including decreased pain
sensitivity, impaired neuromuscular function and coordination,  altered cognitive function,
and decreased anxiety and/or increased motor function; and neurophysiological effects
including decreased cortico-hippocampal activity. These effects are recognized in the U.S.
EPA's Guidelines for Neurotoxicity Risk Assessment (U.S. EPA, 1998} as possible indicators of
neurotoxicity. EPA considered the neurotoxic effects to be biologically plausible and
analogous to effects that could occur in humans; and concluded that the available evidence
for 1,2,4-TMB and 1,3,5-TMB identified neurotoxicity as a toxicity hazard.


1.1.2. Respiratory Effects

   There is evidence in humans and animals that inhalation exposure to TMBs induces
respiratory toxicity. Occupational and residential exposure studies in humans provide
evidence of respiratory toxicity following inhalation exposure to complex VOC mixtures
containing 1,2,4-TMB and 1,3,5-TMB. While controlled human exposures (lones et al..
2006: Jarnbergetal.. 1997a: Jarnberg et al.. 1996} have failed to observe substantial
irritative symptoms following acute (less than 4 hours} inhalation exposures of up to 25
ppm (123 mg/m3} 1,2,4-TMB or 1,3,5-TMB, occupational exposures have been shown to be
associated with increased measures of sensory irritation, such as laryngeal and/or
pharyngeal irritation (Norseth etal.. 1991} and asthmatic bronchitis (Battig et al.. 1956}.
as reviewed in (MOE. 2006}.  Residential exposures have demonstrated significant
associations between 1,2,4-TMB and asthma (Billionnet et al.. 2011}. However, these
studies evaluated TMB exposures occurring as exposures to complex solvent or VOC
mixtures, thereby precluding a determination of the ultimate etiological agent.
DRAFT - DO NOT CITE OR QUOTE                                                       47

-------
   In animals, there is consistent evidence of respiratory toxicity following inhalation
exposure of rodents to 1,2,4-TMB (Table 1-3}.  Markers of pulmonary inflammation and
irritation in the lungs of rats have been observed following subchronic inhalation
exposures of Wistar rats to 1,2,4-TMB. Increases in populations of immune and
inflammatory cells in bronchoalveolar lavage (BAL) fluid have been observed at
concentrations > 123 mg/m3 following subchronic exposures of male Wistar rats to 1,2,4-
TMB [Korsak et al..  1997). Specifically, the amount of cells in the BAL fluid of exposed rats
was increased for total cells (> 123 mg/m3, increased 2.3-3.0 fold] and macrophages (> 492
mg/m3, increased 2.1-2.7 fold}. However, some attenuation of these effects was observed
at high concentrations (i.e., 1230 mg/m3} compared to lower doses. For example, the
number of macrophages was increased 2.7-fold relative to control at 492 mg/m3, but only
2.2-fold at 1,230 mg/m3. This may indicate either adaptation to the respiratory irritation
effects  of 1,2,4-TMB during exposure, saturation of metabolic pathways, or immune
suppression at higher doses. Subchronic exposure of male Wistar rats also significantly
increased the BAL numbers of polymorphonuclear leukocytes and lymphocytes;  however
the specific exposure concentrations eliciting these significant increases were not reported
by study authors. A small, but not significant, decrease in cell viability was observed at >
123 mg/m3 following subchronic exposure to  1,2,4-TMB (Korsak et al.. 1997}.
   In addition to increased populations of immune and inflammatory cells,
histopathological alterations described as peribronchial,  lung parenchymal, and
perivascular lymphocytic infiltration in the lower respiratory tract have also been observed
following subchronic exposures of 1,2,4-TMB to male and female Wistar rats (Korsak et al..
2000).  Significant proliferation of peribronchial lymphatic tissue and interstitial
lymphocytic infiltrations were observed in male rats exposed to 492 mg/m3, although
trend-analysis demonstrated these increases were not concentration-dependent. The
bronchial epithelium lost its cuboidal shape in some rats with peribronchial lymphatic
proliferation, and was observed to form lymphoepithelium. Interstitial lymphocytic
infiltrations were also observed in female rats exposed to 1230  mg/m3. Although unlike
male rats, this increase in females was concentration-dependent as determined by a trend
analysis. Alveolar macrophages were increased in both sexes at 1,230 mg/m3 with trend
analysis demonstrating concentration-dependence across the entire concentration range.
However, when the incidences of all pulmonary lesions were analyzed in aggregate, trend-
analysis demonstrated significant increases in pulmonary lesions in both sexes across the
entire concentration range. Pulmonary lesions were significantly increased in males at >
492 mg/m3, but not at any exposure concentration in females. Studies on the respiratory
effects  of subchronic exposures to 1,3,5-TMB were not available.
DRAFT - DO NOT CITE OR QUOTE                                                      48

-------
   Additional effects on clinical chemistry including increased total protein (37% increase
at both 123 and 492 mg/m3), decreased mucoprotein (13% decrease, 123 mg/m3),
increased lactate dehydrogenase (170% and 79% increase at 123 and 492 mg/m3,
respectively] and increased acid phosphatase activity (47-75% increase at > 123 mg/m3}
were observed in 1,2,4-TMB-exposure animals; suggesting pulmonary irritation or
inflammation. All of these effects also exhibited either some attenuation of effect at high
concentrations compared to lower concentrations, or no increase in effect as exposure
concentration increased. Therefore, some adaptation to the respiratory irritation effects of
1,2,4-TMB may be occurring.
DRAFT - DO NOT CITE OR QUOTE                                                      49

-------
Tables 1-3: Summary of observed in vivo respiratory toxicity for 1,2,4-TMB and
1,3,5-TMB — inhalation exposures
Health Effect
Study Design and Reference
Results
Lowest Level
at which
Significant
Effects were
observed
(mg/m3)a
1,2,4-TMB
Pulmonary
inflammation/
irritation
Clinical Chemistry
Effect
Sensory Irritation
(Decreased
respiration)
123-1,230 mg/m3, 90 days
(6h/day, 5 days/week)
Rat, Wistar, male, 6-7
Korsaketal. (1997)
123-1,230 mg/m3, 90 days
(6h/day, 5 days/week)
Rat, Wistar, male, 6-7
Korsaketal. [1997]
123-1,230 mg/m3, 90 days
(6h/day, 5 days/week)
Rat, Wistar, male and female,
6-7
Korsaketal. [2000]
123-1,230 mg/m3, 90 days
(6h/day, 5 days/week)
Rat, Wistar, male, 6-7
Korsaketal. [1997]
1,245-9,486 mg/m3, single
exposure, 6 minutes
Mouse, BALB/C, male, 8-10
Korsaketal. [1997]: Korsak
etal. [1995]
Increased total bronchoalveolar cell count
with evidence of attenuation at high
exposure
Response relative to control: 0, 202*, 208*,
231*%
Increased macrophage count with evidence
of attenuation at high exposure
Response relative to control:0, 107, 170*,
116*%
Increase in number of pulmonary lesions
Response relative to control: Incidences not
reported, calculation of response relative to
control not possible; authors report
statistically significant increases at 492 and
1,230 mg/m3
Increased acid phosphatase activity with
evidence of attenuation at high exposure
Response relative to control:0, 47*, 74*, 45*%
Decreased respiratory rate as measured
during first minute of exposure
Response relative to contro/;RDso = 2,844
123
492
492
123
2,844
1,3,5-TMB
Sensory Irritation
(Decreased
respiration)
1,245-9,486 mg/m3, single
exposure, 6 minutes
Mouse, BALB/C, male, 8-10
Korsaketal. (1997)
Decreased respiratory rate as measured
during first minute of exposure
Response relative to control: RDso = 2,553
2,553
* Significantly different from controls
a Lowest effect level at which statistically significant changes were observed
   Decreased respiration, a symptom of sensory irritation, has been observed in male
BALB/C mice following acute inhalation exposures either 1,2,4-TMB or 1,3,5-TMB for six
minutes. These acute exposures were observed to result in dose-dependent depression of
DRAFT - DO NOT CITE OR QUOTE
50

-------
respiratory rates, with the maximum decrease in respiration occurring in the first one or
two minutes of exposure [Korsak et al.. 1997: Korsaketal.. 1995). The concentration of
1,2,4-TMB and 1,3,5-TMB that was observed to result in a 50% depression in the
respiratory rate (RDso) was similar between the two isomers: 578 and 519 ppm (2,844 and
2,553 mg/m3}, respectively.

   > Mode of Action Analysis for respiratory toxicity

   Data regarding the potential mode  of action for the respiratory effects resulting from
TMB exposure are limited and the key  events for TMB-induced respiratory toxicity are not
established. However, the available toxicological data suggest that TMB isomers act as
potent acute respiratory irritants and induce inflammatory responses following longer
exposures (i.e., subchronic) in animals. The study authors (Korsaketal.. 1997: Korsak et
al.. 1995) suggested that the decreased respiratory rate is indicative of irritation, and
proposed that respiratory irritants such as TMB may activate a "sensory irritant receptor"
on the transgeminal nerve ending in the nasal mucosa leading to an inflammatory
response.  Korsak et al. (1997:1995) further suggest that activation of this irritant receptor
follows either adsorption of the agonist, or adsorption and chemical reaction with the
receptor. The authors reference a proposed model for the receptor protein that includes
two main binding sites for benzene moieties and a thiol group. Further, the study authors
suggest that in the case of organic solvents (i.e., toluene, xylene, TMB) a correlation
between the potency of the irritating effect and the number of methyl groups is likely given
the observation that RDso values for depressed respiratory rates following exposure to
TMB isomers is approximately 8-fold lower than toluene and 4-fold lower than xylene.
   Following subchronic exposure of rats to 1,2,4-TMB, inflammatory cell (macrophages,
polymorphonuclear leukocytes, and lymphocytes) numbers were increased along with
markers of their activation (total lactate dehydrogenase and acid phosphatase activity in
BAL) (Korsaketal.. 1997). further indicating the inflammatory nature of responses in the
respiratory tract of TMB-exposed animals. Inflammatory pulmonary lesions were also
observed following subchronic exposures in rats. However, many of these effects were not
observed to be concentration-dependent in repeated exposure studies (i.e., no progression
of effect over an order of magnitude of doses), suggesting that there may be adaptation to
respiratory irritation that occurs following extended exposure to TMB. The processes
responsible for the respiratory inflammatory responses in sub chronically exposed animal
are unknown. However, a major inflammatory mediator, interleukin 8 (IL-8), was
increases following exposure of porcine and human macrophages to secondary organic

DRAFT - DO NOT CITE OR QUOTE                                                      51

-------
aerosol (SOA} particles derived from 1,3,5-TMB [Gaschen etal.. 2010). The observation
that IL-8 levels increase following exposure to 1,3,5-TMB-derived SOA is noteworthy as a
major function of IL-8 is to recruit immune cells to sites of inflammation. Therefore, the
observation of inflammatory lesions involving immune cells (i.e., macrophages, leukocytes]
may be partially explained by increases in inflammatory cytokines following TMB
exposures. Additionally, ROS-generation has been observed in cultured neutrophil
granulocytes and rat neural synaptosomes exposed to TMB [Myhre and Fonnum. 2001:
Myhre etal.. 2000}. and the related compounds benzene and toluene have been shown to
induce oxidative stress in cultured lung cells (Mogel etal.. 2011}. Although pulmonary
ROS-generation has not been observed following in vivo or  in vitro TMB exposures, there is
suggestive evidence that it could play a role in the irritative and inflammatory responses
seen in exposed animals.
   In a study investigating jet fuel-induced cytotoxicity in human epidermal keratinocytes
(HEK}, aromatic hydrocarbons were more potent inducers of cell death than aliphatic
constituents, even though the aromatic compounds only accounted for less than one-fourth
of aliphatic constituents (Chouetal.. 2003}. Of the single aromatic ring hydrocarbons,
1,2,4-TMB and xylene were the most lethal to HEK.  Increased cytotoxicity may explain the
small, but insignificant, decrease in BAL cell viability observed in Korsak et al. (1997}.

   7s the hypothesized mode of action sufficiently supported in test animals?

   Data that would allow for the determination of a mode of action for respiratory effects
due to TMB exposure are limited. However, the observation of ROS generation in cultured
neutrophils and ROS generation in lung cells exposed to related aromatic compounds (i.e.,
benzene and toluene}, suggests that oxidative stress may play a role in the observed TMB-
induced respiratory effects in exposed animals.

   7s the hypothesized mode of action relevant to humans?

   The respiratory effects in animals are relevant to humans, especially given the
observation of irritative and inflammatory respiratory effects (e.g., asthma} in humans
exposed to complex solvent mixtures containing TMB isomers.
   In summary, respiratory toxicity is associated with exposure to 1,2,4-TMB and 1,3,5-
TMB based on evidence in humans and animals. The information in humans is limited for a
number of reasons including: all studies investigating exposure to 1,2,4-TMB and 1,3,5-
TMB were conducted using a complex VOC mixture. However, the available information

DRAFT - DO NOT CITE OR QUOTE                                                      52

-------
demonstrates uptake of 1,2,4-TMB and 1,3,5-TMB by humans, and suggests an association
between TMB exposure in humans and respiratory toxicity. The observation of respiratory
irritation and inflammation in Wistar rats and BALB/C mice following exposure to 1,2,4-
TMB was consistent across multiple exposure concentrations, and subchronic and acute
exposure durations. All of the available animal studies were conducted in rodents (Wistar
rats or BALB/C mice] and by the same research group (The Nofer Institute of Occupational
Medicine, Lodz Poland}. No chronic studies are available. Although some endpoints (BAL
macrophages, alkaline phosphatase] showed dose-dependence at low and mid exposure
concentrations, all effects were observed to exhibit some attenuation of effect at high
doses, potentially indicating either adaptation to the respiratory irritation effects,
saturation of metabolic and/or toxicity pathways, or immune suppression at higher doses.
   Although the reported human symptoms (laryngeal and/or pharyngeal irritation,
asthmatic bronchitis, asthma] do not directly parallel the effects observed in animal
studies, the observation of irritative and/or inflammatory responses in multiple species
(including humans] demonstrates a consistency in TMB-induced respiratory toxicity. EPA
considered the observed respiratory effects in animals to be biologically plausible and
analogous to effects that could occur in humans; and concluded that the available evidence
for 1,2,4-TMB identified respiratory toxicity as a toxicity hazard.


1.1.3. Reproductive and Developmental Toxicity
   There are no studies in humans that investigated the reproductive or maternal toxicity
of either 1,2,4-TMB or 1,3,5-TMB. Maternal toxicity in the form of decreased corrected
body weight (i.e., maternal body weight minus the weight of the gravid uterus] was
observed in dams following exposure during gestational exposure to 1,2,4-TMB or 1,3,5-
TMB (Saillenfait et al., 2005]. Dams exposed to 2,952 mg/m3 1,2,4-TMB gained only 50%
of the weight gained by control animals, whereas dams exposed to 2,952 mg/m3 1,3,5-TMB
gained only 25% of the weight gained by controls. Decreased maternal food consumption
(across GD 6-21] was also observed at 2,952 mg/m3 1,2,4-TMB and 1,476 mg/m3
1,3,5-TMB, although the difference compared to controls (9-13%] was modest compared to
the observed decreases in maternal weight gain.  The decrease in food consumption at
1,476 mg/m3 1,3,5-TMB was determined to not be a marker of adversity given no
accompanying decrease in maternal weight gain at that concentration.
   There are no studies in humans that investigated the developmental toxicity of either
1,2,4-TMB or 1,3,5-TMB.  Developmental toxicity (reported as decreased fetal body weight]
has been observed in male and female rat fetuses following gestational exposure to 1,2,4-
DRAFT - DO NOT CITE OR QUOTE                                                     53

-------
TMB and 1,3,5-TMB on gestational days 6 through 20 via inhalation for 6 hours a day
(Saillenfait et al., 2005} (Table 1-4}. Fetal body weights were decreased (statistically
significant} by 5-13% at concentrations of > 2,952 mg/m3 of 1,2,4-TMB and 1,3,5-TMB. No
adverse effects were noted on embryo/fetal viability and no increase in skeletal, visceral, or
external morphology (i.e., teratogenesis} was observed up to the highest concentrations for
either isomer.
 Table 1-4: Summary of observed developmental toxicity for 1,2,4-TMB and
1,3,5-TMB — inhalation exposures
Health Effect


Developmental
Toxicity

Maternal Toxicity


Developmental
Toxicity

Maternal Toxicity
Study Design and Reference

0, 492, 1,476, 2,952, 4,428
mg/m3, GD 6-20 [6h/day]
Rat, Sprague-Dawley, female
&male, 24-25 dams
Saillenfait et al. [2005]
0, 492, 1,476, 2,952, 4,428
mg/m3, GD 6-20 [6h/day]
Rat, Sprague-Dawley, female
&male, 24-25 dams
Saillenfait et al. [2005]

0, 492, 1,476, 2,952, 4,428
mg/m3, GD 6-20 [6h/day]
Rat, Sprague-Dawley, female
&male, 24-25 dams
Saillenfait et al. [2005]
0, 492, 1,476, 2,952, 4,428
mg/m3, GD 6-20 [6h/day]
Rat, Sprague-Dawley, female
&male, 24-25 dams
Saillenfait et al. [2005]
Results
1,2,4-TMB
Decreased fetal body weight of male and
female fetuses
Response relative to control:
Male: 0, 1, 2, 5*, 11*%
Female: 0, 1, 3, 5*, 12*%

Decreased corrected maternal weight gain
Response relative to control: 0, +7, 7, 51*,
100*% [weight gain = 0 g]
1,3,5-TMB
Decreased fetal body weight of male and
female
Response relative to control:
Male: 0, 1, 5, 7*, 12*%
Female: 0, 1, 4, 6, 13*%

Decreased corrected maternal weight gain
Response relative to control: 0, +3, 31, 76*,
159*% [weight gain = -12 g]
Lowest Level
at which
Significant
Effects were
observed
(mg/m3)a


2,952

2,952


2,952

2,952
* Significantly different from controls
a Lowest effect level at which statistically significant changes were observed
DRAFT - DO NOT CITE OR QUOTE
54

-------
   > Mode of Action Analysis for developmental toxicity

   The mode of action for 1,2,4-TMB- and 1,3,5-TMB-induced developmental toxicity is
unknown. The database for developmental toxicity following exposure to 1,2,4-TMB is
limited to one animal study; no studies in humans are available. Although there is only one
study available, 1,2,4-TMB and 1,3,5-TMB demonstrated effects on fetal and maternal body
weights.  The developmental and maternal toxicity in animals was considered by the
Agency to be biologically plausible and potentially analogous to effects that could occur in
humans.  EPA concluded that the available evidence for 1,2,4-TMB and 1,3,5-TMB identified
maternal and developmental toxicity as a toxicity hazard.

1.1.4. Hematological Toxicity and Clinical Chemistry Effects

   There is limited evidence in humans, and stronger evidence in animals, that exposure to
TMBs induces hematological toxicity. Alterations in blood clotting and anemia in workers
exposed to a paint solvent containing 1,2,4-TMB and 1,3,5-TMB was reported by Battig et
al. (1956). as reviewed by MOE (2006). A LOAEL of 295 mg/m3 was identified from this
study. However, as workers were exposed to a solvent mixture containing TMB isomers, it
is impossible to ascertain the ultimate contribution of either isomer to the observed health
effects.
   In animals, there is evidence of hematological toxicity following subchronic exposure to
1,2,4-TMB and short-term exposure to 1,3,5-TMB (Table 1-5).  Subchronic exposures to
1,2,4-TMB have been shown to result in hematological effects and changes in serum
chemistry in exposed rats (Korsak et al.. 2000). In male rats at termination of exposure,
RBC  counts were decreased by 23% and WBC counts increased 180% at 1,230 mg/m3; the
observed alteration in blood cell counts were exposure concentration-dependent as
determined by trend analysis. A concentration-dependent increase in WBC count was also
observed in female rats, pair-wise comparisons of individual doses failed to reach
statistical significance at any concentration. WBC counts were observed to be slightly
decreased (18%) relative to controls two weeks after the termination of exposure, whereas
RBC  counts were still decreased by 24% relative to control (although this decreased failed
to reach statistical significance). Significant decreases in reticulocytes (71% of controls)
and clotting time (37% of controls) were observed in female rats exposed to 1,230 mg/m3
and 492 mg/m3 1,2,4-TMB, respectively. Both of these effects were concentration-
dependent when analyzed over the entire range of exposure concentrations with values
60-65% greater than controls after end of exposure; animals fully recovered within two
weeks after termination of exposure. The only clinical chemistry parameter significantly
DRAFT - DO NOT CITE OR QUOTE                                                      55

-------
altered was an increase in sorbitol dehydrogenase at > 123 mg/m3 in exposed male rats,
although these increases were not exposure-dependent. Sorbitol dehydrogenase activity
was also higher in exposed female rats, but the increases in activity were not significantly
higher when compared to controls.

    An increase in aspartate aminotransferase, but no other substantial hematological
effects, was observed in rats 14 days following short-term exposure (6 hours/day, 6
days/week for five weeks] [Wiglusz etal.. 1975a: Wiglusz etal.. 1975b]. The adversity of
aspartate aminotransferase is unclear given the lack of a clear pattern in temporality
(effects at some days post-exposure, but not others] and the lack of accompanying liver
histopathology.
Tables 1-5: Summary of observed in vivo hematological toxicity and clinical
chemistry effects for 1,2,4-TMB and 1,3,5-TMB — inhalation exposures
  Health Effect
    Study Design and
       Reference
               Results
  Lowest
  Level at
  which
Significant
  Effects
   were
 observed
 (mg/m3)3
                                         1,2,4-TMB
  Hematological
     toxicity
                123-1,230 mg/m3,90 days
                (6h/day, 5days/week)
                Rat, Wistar, female & male,
                6-7
                Korsaketal. (2000)
123-1,230 mg/m3,90 days
(6h/day, 5days/week)
Rat, Wistar, female & male,
6-7
Korsaketal. [2000]
123-1,230 mg/m3,90 days
(6h/day, 5days/week)
Rat, Wistar, female & male,
6-7
Korsaketal. f20001
                123-1,230 mg/m3,90 days
                (6h/day, 5days/week)
                Rat, Wistar, female & male,
                6-7
                Korsaketal. [2000]
                         Decreased red blood cells in males only.

                         Response relative to control:0,1,15, 23*%
                         (recovery = 24%)
                                          Increased white blood cells in males only.

                                          Response relative to control:0, 2, 4, 80*%
                                          (recovery = 18% decrease)
                                          Decreased reticulocytes in females only.

                                          Response relative to control:0, 51, 49,71*%
                                          (recovery = 65% increase)
                         Non-monotonic decreases in clotting time in
                         females only.

                         Response relative to control:0, 23,37*,27*%
                         (recovery = 60% increase)
                                         1,230
                                         1,230
                                         1,230
                                          492
 Clinical Chemistry
     Effects
123-1,230 mg/m3,90 days
(6h/day, 5days/week)
Rat, Wistar, female & male,
6-7
Non-monotonic increases in sorbitol
dehydrogenase in males only.

Response relative to control:0, 73*, 74* 73*
   123
DRAFT - DO NOT CITE OR QUOTE
                                                                       56

-------
                Korsaketal. f20001
                                        1,3,5-TMB
  Hematological
     Effect
1,500-6,000 mg/m3, single
exposure, 6 hours
Samples collected 0,1, 7,14,
and 28 days post exposure
Rat, Wistar, male, 6
Wiglusz et al. [Wiglusz et al..
1975a)
Increased segmented neutrophilic
granulocytes (1-28 days post exposure).

Response relative to control: Increased across
all days of exposure.
3,000
 Clinical Chemistry
     Effects
3,000 mg/m3, 5 weeks (6
h/day, 6days/week)
Samples collected 1, 3, 7,14,
and 28 days during exposure
Rat, Wistar, male, 6
Wiglusz et al. [1975b]
                                         Increased aspartate aminotransferase on day
                                         14

                                         Response relative to control (day 14):12*%
                                        3,000
300-3,000 mg/m3, single
exposure, 6 hours
Samples collected 0, 2, 7,14
and 28 days post exposure
Rat, Wistar, male, 6
Wiglusz et al. fl975bj
                                         Increased alkaline phosphatase on day 7 post-
                                         exposure

                                         Response relative to control (on day 7):Q, -0.1,
                                         0.03, 84*%
                                        3,000
* Significantly different from controls
a Lowest effect level at which statistically significant changes were observed

   Acute exposures of male Wistar rats to 1,500-6,000 mg/m3 1,3,5-TMB for six hours did
not result in substantial effects on hemoglobin or RBC or WBC count [Wiglusz et al..
1975a}.  However, the number of segmented neutrophilic granulocytes was increased in
1,3,5-TMB-exposed rats up to 28 days following exposure (statistics not reported}. The
greatest increase in granulocyte numbers (100%} was observed the day of exposure and
one day following in rats exposed to 6,000 mg/m3, although attenuation was seen 7-28
days following exposure, possibly indicating induction of metabolizing enzymes or
saturation of toxicity pathways. Investigation of clinical chemistry parameters in rats
acutely exposed to 300- 3,000 mg/m3 for six hours did not reveal any consistent pattern in
the levels of aspartate or alanine aminotransferases, although alkaline phosphatase was
statistically increased 84% in rats seven days following exposure to 3,000 mg/m3 (Wiglusz
etal.. 1975bj. NOAELs and LOAELs determined from these acute exposure studies are
provided in Table 1-5.


    >  Mode of Action Analysis for hematological toxicity and clinical chemistry
       effects

   The mode of action for 1,2,4-TMB-induced hematological and clinical chemistry effects
is currently unknown. Increased sorbitol dehydrogenase activity is a marker for hepatic
DRAFT - DO NOT CITE OR QUOTE
                                                                       57

-------
injury [Ramaiah. 2007) and therefore, underlying hepatotoxicity could explain its increase
in exposed males. However, absolute and relative liver weights were not observed to
increase with exposure, and microscopic histopathological analysis of the liver did not
demonstrate any observable changes. The increases in WBC counts in exposed animals
could be secondary to the observed respiratory irritative and inflammatory effects of
1,2,4-TMB exposure (2000; 1997}.
   In summary, hematological toxicity was observed with exposure to 1,2,4-TMB and
1,3,5-TMB in humans and animals. The information regarding hematological toxicity in
humans is limited to one study involving exposure to a complex VOC mixture containing
both 1,2,4-TMB and 1,3,5-TMB, but it did report hematological effects (alterations in
clotting an anemia] that are roughly analogous to those effects (decreased RBCs and
decreased clotting time] observed in rats following subchronic exposure to 1,2,4-TMB.
Although both databases are limited (there are no chronic studies in animals], there is
some consistency in effects across species (i.e., rats and humans]. EPA considered the
hematological effects to be biological plausible and analogous to effects that could occur in
humans; and concluded that the available evidence for 1,2,4-TMB identified hematological
toxicity as a toxicity hazard.

1.1.5. Other lexicological Effects

   One animal study was identified that investigated the association of chronic oral
exposure (via gavage] to 1,2,4-TMB and cancer endpoints (Maltoni etal.. 1997]. Male and
female Sprague-Dawley rats were exposed to a single dose of 800 mg/kg-day of 1,2,4-TMB
in olive oil by stomach tube for four days/week starting at 7 weeks of age. Exposures were
terminated at the end of 104 weeks (i.e. at 111 weeks of age] and the animals were kept
under observation until natural death. The authors report that chronic oral exposure to
1,2,4-TMB resulted in an "intermediate" reduction of survival in male rats and a "slight"
reduction in females (no quantitative information on survival was reported]. A slight
increase in total malignant tumors in both sexes of rats was observed, with the incidence of
head cancers being specifically increased in male rats. The predominant type of head
cancer identified was neuroesthesioepithelioma, which arises from the olfactory
neuroepithelium and is normally rare in Sprague-Dawley rats. Other head cancers
observed included those in the Zymbal gland, ear duct, and nasal and oral cavities. No tests
of statistical significance were reported for these data. When Fisher's exact test was
performed (by EPA] on the incidences calculated from the reported percentages of animals


DRAFT - DO NOT CITE OR QUOTE                                                       58

-------
bearing tumors in the control and exposed animals, no statistically significant associations
were observed.
   Janik-Spiechowicz et al. (1998} investigated the genotoxicity of the trimethylbenzene
isomers 1,2,4-TMB, and 1,3,5-TMB by measuring three genotoxic endpoints: mutation
frequency in bacteria, micronucleus formation in mice, and sister chromatid exchanges in
mice.  Neither isomer induced gene mutations in any Salmonella typhimurium strain tested
(TA102, TA100, TA98, and TA97a}. Both isomers were also negative for the formation of
micronuclei in Imp:BALB/c mice following i.p. injection. Males in the high dose groups for
1,2,4-TMB and 1,3,5-TMB exposures had a statistically significant reduction in the ratio of
PCEs to NCEs indicating bone marrow cytotoxicity. However, both isomers significantly
increased the frequency of sister chromatid exchanges (SCEs} in Imp:BALB/c mice
following i.p. injection, with 1,2,4-TMB having the more significant response. These results
appeared to have occurred at doses that did not induce significant bone marrow
cytotoxicity except at the highest dose.
   In summary, although very little genotoxicity data are available on  1,2,4-TMB and
1,3,5-TMB, Janik-Spiechowicz et al. (1998) observed negative results in several key
mutagenicity assays, including the Ames mutation assay in Salmonella and in vivo assays
for micronucleus formation in mouse bone marrow cells. However, Janik-Spiechowicz et al.
(1998} did observe increased incidence of SCE in mice exposed to both TMB isomers.
Increased frequency of SCEs indicates that DNA damage has occurred as a result of
exposure to these isomers, but it does not provide a specific indication of mutagenic
potential, as there is no known mechanistic association between SCE induction and a
transmissible genotoxic effect. With only one positive SCE result, and all other data
showing negative results for gene and chromosomal mutation in vitro and in vivo, there is
not enough evidence to conclude that either isomer is directly genotoxic.

1.1.6. Similarities between 1,2,4-TMB and 1,3,5-TMB regarding observed inhalation
and oral toxicity

   In the existing toxicological database for 1,2,4-TMB and 1,3,5-TMB, important
similarities have been observed in the potency and magnitude of effect resulting from
exposure to the two isomers, although some important differences also exist. In acute
studies investigating respiratory irritative effects, the RDso of the two isomers were very
similar: 2,844 mg/m3 for 1,2,4-TMB and 2,553 mg/m3 for 1,3,5-TMB fKorsak et al.. 19971
The similarity regarding toxicity was also observed in acute inhalation neurotoxicity
studies: the ECso for decreased coordination, balance, and neuromuscular function was
DRAFT - DO NOT CITE OR QUOTE                                                      59

-------
4,694 mg/m3 for 1,2,4-TMB and 4,738 mg/m3 for 1,3,5-TMB. The EC5o for decreased pain
sensitivity were also similar for both isomers: 5,683 mg/m3 for 1,2,4-TMB and 5,963
mg/m3 for 1,3,5-TMB (Korsakand Rydzynski. 1996}.  Neurotoxic endpoints were also
similarly affected by oral exposure to individual isomers: increased electrocortical arousal
was observed in rats exposed to 240 mg/kg 1,2,4-TMB or 1,3,5-TMB, and altered brain
EEGs were observed in rats exposed to 790 mg/kg 1,2,4-TMB or 1,3,5-TMB [Tomas et al..
1999a: Tomas etal.. 1999c}. Although these effects were seen at the same dose levels in
animals  exposed to either isomer, these doses were LOAELs, and it is unclear whether this
similar potency would be observed at lower doses. Additionally, there were differences in
the magnitude of effect between the isomers, with 1,2,4-TMB inducing larger abnormalities
in brain  EEGs and 1,3,5-TMB affecting electrocortical arousal to a greater degree than
1,2,4-TMB.
   In short-term neurotoxicity studies, a similar pattern of effects (inability to learn
passive and/or active avoidance and decreased pain sensitivity] indicating altered
neurobehavioral function were observed for both isomers (Wiaderna etal.. 2002:
Gralewicz and Wiaderna. 2001: Gralewicz etal..  1997a}. In these studies, 1,3,5-TMB was
shown be more toxic than 1,2,4-TMB: rats exposed to 123 mg/m3 1,3,5-TMB displayed
significantly decreased abilities to learn passive or active avoidance (Wiaderna et al..
2002}. whereas 1,2,4-TMB elicited the inability to learn passive avoidance in rats exposed
to 492 mg/m3 1,2,4-TMB and did not affect active avoidance at any exposure concentration
[Gralewicz etal.. 1997a}. Additionally, in animals exposed to either isomer at 492 mg/m3,
exposure to 1,3,5-TMB decreased the ability to learn passive avoidance to a greater degree
than 1,2,4-TMB (approximately 50% decrease vs. 40%}, and the effect of 1,3,5-TMB
manifested at earlier time points than 1,2,4-TMB (three vs. seven days} (Gralewicz and
Wiaderna. 20011
   Lastly, similarities were observed in 1,2,4-TMB- and 1,3,5-TMB-induced developmental
and maternally toxic effects (Saillenfait et al.. 2005}. Male fetal weights were significantly
reduced in animals exposed gestationally to 2,952 mg/m3 1,2,4-TMB (5% decrease} or
1,3,5-TMB (7% decrease}.  1,2,4-TMB also significantly decreased female fetal weights by
approximately 5% in animals exposed to the same concentration. Although, 1,3,5-TMB
significantly reduced female fetal weights by 13% in animals exposed to 5,904 mg/m3,
female weights were decreased at 2,952 mg/m3 to a similar magnitude (6%} as animals
exposed to the same concentration of 1,2,4-TMB. Maternal toxicity, measured as decreased
corrected maternal weight gain, was significantly decreased in animals exposed to 2,952
mg/m3 1,2,4-TMB or 1,3,5-TMB. However, 1,3,5-TMB exposure resulted in a 75%

DRAFT - DO NOT CITE OR QUOTE                                                      60

-------
reduction of maternal weight gain compared to controls, whereas 1,2,4-TMB exposure
reduced maternal weight gain by 50%.

1.1.7. Susceptible Populations orLifestages

   Although there are no chemical-specific data that would allow for the definitive
identification of susceptible subpopulations, the reduced metabolic and elimination
capacities in children relative to adults may be a source of susceptibility [Ginsberg etal..
2004). TMB isomers are metabolized via side-chain oxidation to form alcohols and
aromatic carboxylic/mercapturic acids or by hydroxylation to form phenols, which are
then conjugated with glucuronic acid, glycine, or sulfates for urinary excretion. The
activities of multiple cytochrome P450 (GYP P450) mono-oxygenase isozymes have been
shown to be reduced in children up to 1 year of age compared to adult activities [Ginsberg
etal.. 2004). Additionally, the rate of glucuronidation and sulfation is decreased in
children. Therefore, as both GYP P450 mono-oxygenase activities and the rate of
glucuronidation and sulfation appear to be decreased in early life, newborns and young
infants may experience higher and more persistent blood concentrations of 1,2,4-TMB,
1,3,5-TMB, and/or their respective metabolites compared with adults at similar exposure
levels.  Reduced renal clearance in children may be another important source of potential
susceptibility. TMB isomers and their metabolites are excreted in the urine of exposed
laboratory animals and occupationally exposed humans. Data indicating reduced renal
clearance for infants up to 2 months of age [Ginsberg et al.. 2004) may suggest a potential
to affect TMB excretion, thus possibly prolonging its toxic effects. Additionally, those with
pre-existing respiratory diseases [e.g., asthma) may be more sensitive to the respiratory
irritative and inflammatory effects of TMB isomers.


1.2. Selection of Candidate Principal Studies and Critical Effects for
   Derivation of Reference Values

1.2.1. Inhalation Exposure - Effects Other Than Cancer - 1,2,4-TMB

   While literature exists on the noncancer effects of 1,2,4-TMB  exposure in humans,
including neurological, respiratory, and hematological toxicities, no human studies are
available that would allow for the  quantification of subchronic or chronic noncancer
effects. The available human studies evaluated TMB exposures occurring as complex
solvent or VOC mixtures, and this consideration along with other uncertainties including

DRAFT - DO NOT CITE OR QUOTE                                                      61

-------
high imprecision in effect measures due to low statistical power, lack of quantitative
exposure assessment, and lack of control for co-exposures, limit their utility in derivation
of quantitative human health toxicity values.  However, these studies provide supportive
evidence for the neurological, respiratory, and hematological toxicity of TMB isomers in
humans and determination of coherency of effect in both humans and laboratory animals
for deriving toxicity values.
   Studies investigating 1,2,4-TMB noncancer effects in experimental animal models were
identified in the literature. Acute inhalation studies observing neurotoxicity and
respiratory toxicity in exposed rodents were identified, but the high exposure
concentrations used and acute exposure duration of these studies limit their applicability
for quantitation of chronic human health effects. However, as with the human mixture
studies, these studies do provide qualitative information regarding consistency and
coherence of effect that is supportive of the development of quantitative human health risk
values.
   1,2,4-TMB-induced toxicity across several organ systems was observed in three
subchronic studies by Korsak et al., [2000:1997) and Korsak and Rydzynski (1996). Data
from these studies were considered as candidate critical effects for the purpose of
determining the point of departure (POD] for derivation of the inhalation RfC for
1,2,4-TMB. These studies were determined to be adequate as evaluated using study quality
characteristics related to study populations (studies used rats as an appropriate laboratory
animal species and utilized appropriate sham-exposed controls], exposure (the purity of
1,2,4-TMB was reported as > 97% pure (impurities not reported], the studies utilized an
appropriate route [inhaled air] and duration [subchronic] of exposure, and the studies  used
a reasonable range of appropriately-spaced dose levels to facilitate dose-response
analysis], and data (appropriate latency between exposure and development of
toxicological outcomes was used, the persistence of some outcomes after termination of
exposure was investigated, adequate numbers of animals per dose group were used, and
appropriate statistical tests including pair-wise and trend analyses were performed].
When considered together, these subchronic studies examined 1,2,4-TMB-induced toxicity
in multiple organ systems including CNS, hematological, and pulmonary effects (Table 1-6].

Table 1-6: Non-cancer endpoints resulting from subchronic inhalation exposure to
1,2,4-TMB considered for the derivation of the RfC
Endpoint
Species/
Sex
Exposure Concentration (mg/m3)
0
123
Neurotoxicological Endpoints
492
1,230

DRAFT - DO NOT CITE OR QUOTE                                                       62

-------
Decreased pain sensitivity (measured
as latency to paw-lick in seconds)
Impaired neuromuscular function
and coordination (% failures on
rotarod)
Rat, male
15.4±5.8a
(n = 9)
0
(n = 10)
18.2 ±5.7
(n = 10)
10
(n = 10)
27.6±3.2d
(n = 9)
20
(n = 10)
30.1 ± 7.9d
(n = 10)
40e
(n = 10)
Hematological Endpoints
Decreased RBCs (106/cm3)
Increased WBCs (106/cm3)
Decreased reticulocytes (%)
Decreased clotting time (seconds)
Rat, male
Rat, female
9.98 ±1.68
(n = 10)
8.68 ±2.89
(n = 10)
3.5 ±2. 6
(n = 10)
30 ±10
(n = 10)
9.84 ±1.82
(n = 10)
8.92 ± 3.44
(n = 10)
1.7 ±2.0
(n = 10)
23 ±4
(n = 10)
8.50 ±1.11
(n = 10)
8.30 ±1.84
(n = 10)
1.8 ±0.9
(n = 10)
19±5d
(n = 10)
7.70 + 1.38d
(n = 10)
15.89 ± 5.74d
(n = 10)
1.0±0.6C
(n = 20)
22±7C
(n = 20)
Pulmonary Endpoints
Increased BAL macrophages
(106/cm3)
Increased BAL total cells (106/cm3)
Increased inflammatory lung lesions
Rat, male
1.83 ± 0.03
(n = 6)
1.93 ± 0.79
(n = 6)
b
(n = 10)
3.78 ±0.8
(n = 6)
5.82 ± 1.32f
(n = 6)
b
(n =10)
4.95 ± 0.2d
(n = 7)
5.96±2.80d
(n = 7)
b
(n = 10)
3.96 ± 0.3d
(n = 6)
4.45 ± 1.58C
(n = 7)
b
(n = 20)
a Values are expressed as mean ± one standard deviation
b Incidences for individual dose groups not reported in the study. NOAEL for males identified by EPA was
123mg/m3
cp < 0.05; dp < 0.01;e p < 0.005; fp < 0.001
Adapted from Korsak et al., (2000:1997) and Korsak and Rydzynski (1996)

   Endpoints from these studies that demonstrated statistically significant pair-wise
increases or decreases relative to control were considered for the derivation of the RfC for
1,2,4-TMB.  These endpoints included decreased neuromuscular function and coordination,
and decreased pain sensitivity in male rats (Korsak and Rydzynski. 1996}. increased BAL
total cells and increased BAL macrophages in male rats (Korsak et al.. 1997}. and decreased
RBCs, increased WBCs, and increased pulmonary inflammatory lesions in male rats and
decreased reticulocytes and clotting time in female rats [Korsak et al.. 2000}. Increases in
BAL polymorphonuclear leukocytes and lymphocytes were not considered for RfC
derivation due to a lack of reporting at which doses statistically significant increases
occurred. Changes in BAL protein and enzyme activity level were not considered due to
non-monotonically increasing dose-responses, and increases in sorbitol dehydrogenase
were not further considered due to the lack of accompanying hepatocellular
histopathological alterations in exposed animals. Endpoints carried forward for derivation
of an RfC for 1,2,4-TMB, along with their NOAEL and LOAEL values, are graphically
represented in Figure 1-1.
DRAFT - DO NOT CITE OR QUOTE
63

-------




:
c
4

Doses
ONOAEL
DLOAEL
1 4
> [
> C
I • • 4
1 [
!> C
I 4
) C
> [
: c
> n n n <
] c
3 4
) C
I 4
) C
) 4
) [
> d
<
{
(
•
] <
3
» 1
] [
) (
» 1
>
]
)
>
neuro- pain BAL BAL pulmonary RBCs WBCs reticulo- clotting maternal fetal
muscular sensitivity macro- total cells lesions (c) (c) cytes time weight gain weight
function (a) (a) phages (b) (b) (c) (c) (d) (d)
(b)
Solid lines represent range of exposure concentrations, (a) Korsak and Rydzynski [1996]: (b) Korsak et al.
(1997); (c) Korsak et al. (2000): (d) Saillenfait et al. (2005)

Figure 1-1. Exposure response array for inhalation exposure to 1,2,4-TMB.
    Although the Saillenfait et al. (2005) study was a well conducted developmental
toxicity study, data from this study were not considered for identification of candidate
critical effects for 1,2,4-TMB due to the fact that maternal and developmental toxicities
were observed at doses 6- to 24-fold higher than the doses that resulted neurological,
hematological, and pulmonary effects observed in the subchronic Korsak studies.

1.2.2. Inhalation Exposure - Effects Other Than Cancer - 1,3,5-TMB

   No human studies are available that would allow for the quantification of subchronic or
chronic noncancer effects resulting from inhalation exposure to 1,3,5-TMB. The available
human studies evaluated TMB exposures occurring as complex solvent or VOC mixtures,
and this consideration along with similar uncertainties as discussed for 1,2,4-TMB limit
their utility in derivation of quantitative human health toxicity values. As for 1,2,4-TMB,
the human studies  do provide supportive evidence for the neurological toxicity of 1,3,5-
TMB in humans and strengthen the determination of consistency and coherency of effect in
humans and laboratory animals.
DRAFT - DO NOT CITE OR QUOTE
64

-------
   No suitable chronic or subchronic inhalation studies investigating 1,3,5-TMB
noncancer effects in experimental animal models were identified in the literature that
would support the derivation of the RfC. Two short-term inhalation studies (Wiaderna et
al.. 2002: Gralewicz and Wiaderna. 2001} investigating neurotoxicity outcomes were
identified in the literature and data from these studies were considered as candidate
critical effects for the purpose of derivation of an RfC for 1,3,5-TMB in the absence of a
suitable chronic or subchronic study. Additionally, one developmental toxicity study was
identified in the literature; Saillenfait et al. (2005}. Data from these studies were
considered as candidate critical effects for the purpose of determining the POD for
derivation of the inhalation RfC for 1,3,5-TMB. Based on the noncancer database for 1,3,5-
TMB, these studies were determined to be adequate as evaluated using study quality
characteristics related to study populations (studies used rats as an appropriate laboratory
animal species and utilized appropriate sham-exposed controls}, exposure (the purity of
1,3,5-TMB was reported as 99% pure, the studies utilized an appropriate route [inhaled
air] duration [short-term and gestational] of exposure (although the duration for short-
term studies was not optimal}, and the studies used a reasonable range of appropriately-
spaced dose levels to facilitate dose-response analysis}, and data (appropriate latency
between exposure and development of toxicological outcomes was used, the persistence of
some outcomes after termination of exposure was investigated, adequate numbers of
animals per dose group were used, and appropriate pair-wise statistical tests were
performed}.
   When considered together, these short-term and developmental studies examined
1,3,5-TMB-induced toxicity in multiple organ systems in adult, pregnant, and developing
organism. Endpoints from these studies that demonstrated statistically significant pair-
wise increases or decreases relative to control were considered for the derivation of the
RfC for 1,3,5-TMB. Gralewicz and Wiaderna (2001} and Wiaderna et al. (2002} both
indicate altered cognitive function, decreased pain sensitivity, and decreased anxiety
and/or increased motor function following inhalation exposure to 1,3,5-TMB (see Table 1-
1}. Wiaderna et al. (2002} reported that 123 mg/m3 was the  LOAEL for altered cognitive
function the NOAEL  for decreased pain sensitivity. As altered cognitive function was
observed at a lower  exposure concentration than decreased pain sensitivity, only altered
cognitive function was further considered for derivation of an RfC for 1,3,5-TMB from the
Wiaderna et al. (2002} study. All three neurotoxic effects (altered cognitive function,
decreased pain sensitivity, and decreased anxiety and/or increased motor function} were
observed at the only exposure concentration utilized in the Gralewicz and Wiaderna (2001}
(i.e., 492 mg/m3}; these LOAELs were further considered for derivation of an RfC for 1,3,5-
DRAFT - DO NOT CITE OR QUOTE                                                       65

-------
TMB. From the Saillenfait et al. (2005) study, decreased male and female fetal weights and
decreased corrected maternal weight gain were considered for derivation of the RfC (Table
1-7}. Changes in serum chemistry parameters in rats exposed sub chronically to 1,3,5-TMB
were not considered for derivation of the RfC due to inconsistent temporal patterns of
effect and the lack of accompanying histopathology.

Table 1-7: Non-cancer endpoints resulting from gestational inhalation exposure (GD
6-20) to 1,3,5-TMB considered for the derivation of the RfC
Endpoint
Species/
Sex
Exposure Concentration (mg/m3)
0
fn = 211"
492
fn = 221
1,476
fn = 211
2,952
fn = 171
5,904
fn = 181
Developmental Endpoints
Decreased fetal weight
GO
Rat, male
Rat, female
5.80 ±
0.41b
5.50 ±0.32
5.76 ±0.27
5.74 ±0.21
5.50 ±0.31
5.27 ±0.47
5.39 ±
0.55C
5.18 ±0.68
5.10 ±
0.57d
4.81 ±
0.45d
Maternal Endpoints
Decreased maternal
weight gain (g)
Rat, female
29 ±14
30 ±9
20 ±12
7±20C
-12±19d
a Number of dams with live litters, numbers of live fetuses not explicitly reported
b Values are expressed as mean ± one standard deviation
cp < 0.05; dp < 0.01
Adapted from Saillenfait et al. [2005]
DRAFT - DO NOT CITE OR QUOTE
66

-------
10000

1000
100







Doses
ONOAEL
DLOAEL

D

Altered
cognitive
function (a)
t ?
I 1
<
I
C
T T
o
i
> D D • •
:
Altered 4. Pain 4. Anxiety f f
cognitive sensitivity (a) and/or T" maternal fetal
function (b) motor activity (a) weight gain... weight...
Solid lines represent range of exposure concentrations, (a) Gralewicz and Wiaderna [2001]: (b) Gralewicz et
al. (19973); (c] Saillenfait et al. [2005]

Figure 1-2. Exposure response array for inhalation exposure to 1,3,5-TMB.

1.2.3. Oral Exposure - Effects Other Than Cancer - 1,2,4-TMB and 1,3,5-TMB

   No human studies are available that would allow for the quantification of subchronic or
chronic noncancer effects resulting from oral exposure to either 1,2,4-TMB or 1,3,5-TMB.
Additionally, no suitable chronic or subchronic oral studies investigating 1,2,4-TMB or
1,3,5-TMB noncancer effects in experimental animal models were identified in the
literature that would support the derivation of an RfD. Although the oral database for
1,2,4-TMB and 1,3,5-TMB are inadequate to support the derivation of an RfD, a PBPK model
is available to perform a route-to-route extrapolation  [Hissink et al.. 2007). The Hissink
model was chosen as an appropriate model because it was the only published 1,2,4-TMB
model that included parameterization for both rats  and humans, the model code was
available, and the model adequately predicted experimental data in the dose range of
concern. The use of inhalation toxicity data to derive an oral RfD is supported by the
1,2,4-TMB and 1,3,5-TMB database: sufficient evidence exists that demonstrates similar
qualitative profiles of metabolism (i.e., observation  of dimethylbenzoic and hippuric acid
metabolites] and patterns of parent compound distribution across exposure routes.
DRAFT - DO NOT CITE OR QUOTE                                                      67

-------
Further, no evidence exists that would suggest toxicity profiles would differ to a substantial
degree between oral and inhalation exposures.


1.3. Carcinogenicity Analysis

Synthesis and Overall Weight of Evidence

   Under the Guidelines for Carcinogen Risk Assessment (2005a}. the database for
1,2,4-TMB and 1,3,5- TMB provides "inadequate information to assess the carcinogenic
potential" of these isomers. This characterization is based on the fact that there is no
information regarding the carcinogenicity of TMB in humans and that the only animal
study on the carcinogenicity of 1,2,4-TMB observed no statistically significant carcinogenic
effects. No studies regarding the carcinogenicity of 1,3,5-TMB were identified in the
available scientific literature.
   Only one animal carcinogenicity study was identified [Maltoni etal.. 1997). involving
exposure to 1,2,4-TMB by oral gavage. Although an increased incidence of total malignant
tumors in both sexes and head cancers (predominately neuroethesioepithelioma] in males
was observed in exposed rats, no statistical analyses were reported.  When EPA
independently performed the Fisher's exact test on the reported data, no statistically
significant effects were observed.
DRAFT - DO NOT CITE OR QUOTE                                                       68

-------
2.  DOSE-RESPONSE ANALYSIS
2.1. Inhalation Reference Concentration for Effects other than Cancer

2.1.1. Dose-Response Assessment for RfC derivation for 1,2,4-TMB

   As discussed in Section 1.2.1, endpoints observed in Korsak et al., (2000:1997} and
Korsak and Rydzynski (1996} that demonstrated statistically significant (at p < 0.05 or
greater} pair-wise increases or decreases relative to control for at least one dose group
were considered for the derivation of the RfC for 1,2,4-TMB; these effects are listed in Table
1-6 above. This assessment used the benchmark dose (BMD} approach, when possible, to
estimate a point of departure (POD} for the derivation of an RfC for 1,2,4-TMB (Table 2-1;
see Section C.I of Appendix B (U.S. EPA. 2011cj for detailed methodology}. The BMD
approach involves fitting a suite of mathematical models to the observed dose-response
data using EPA's Benchmark Dose Software (BMDS, version 2.2}. Each fitted model
estimates aBMD and its associated 95% lower confidence limit (BMDL} corresponding to a
selected benchmark response (BMR}. For dichotomous data (i.e., impaired neuromuscular
function and coordination, measured as % failure on rotarod} from Korsak and Rydzynski
(1996}, no information is available regarding the change in this response that would be
considered biologically significant, and thus a BMR of 10% extra risk was used to model
this endpoint, consistent with the  Benchmark Dose Technical Guidance (U.S. EPA. 2000aj.
For continuous data (i.e., decreased pain sensitivity, increased BAL macrophages,
decreased RBCs, decreased reticulocytes, and decreased clotting time} from the Korsak and
Rydzynski (1996} and Korsak et al. (2000:1997} studies, no information is available
regarding the change in these responses that would be considered biologically significance,
and thus a BMR equal to a change  in the mean equal to 1 standard deviation of the model
estimated control mean was used  in modeling the endpoints, consistent with the
Benchmark Dose Technical Guidance (U.S. EPA. 2000aj. The estimated BMDL is then used
as the POD for deriving the RfC.
   The suitability of the above methods to determine a POD is dependent on the nature of
the toxicity database for a specific chemical. Some endpoints for 1,2,4-TMB were not
amenable to BMD modeling for a variety of reasons, including equal responses at all
exposure groups (e.g., increased BAL total cells}, responses only in the high dose group
with no significant changes in responses in lower dose groups (e.g., increased WBCs}, and
DRAFT - DO NOT CITE OR QUOTE                                                    69

-------
absence of incidence data (e.g., increased inflammatory lung lesions}. Additionally, some
datasets were deemed adequate for BMD modeling, but no model provided an adequate fit
to the data (e.g., increased BAL macrophages}, estimated BMDs were greater than the
highest exposure concentration (e.g., decreased reticulocytes}, or estimated BMDLs were
determined to be biologically implausibly low (e.g., decreased clotting time}. In these
cases, the NOAEL/LOAEL approach was used to identify a POD. Detailed modeling results
are provided in Section C.2 of Appendix B (U.S. EPA. 2011c) (detailed modeling results for
maternal and fetal endpoints observed in Saillenfait et al. (2005} are provided in Appendix
B (U.S. EPA. 2011cj for comparison to endpoints observed in the Korsak et al., (2000: 1997}
and Korsak and Rydzynski (1996} studies}.

Table 2-1: Summary of dose-response analysis and point of departure estimation for
endpoints resulting from subchronic inhalation exposure to 1,2,4-TMB
Reference
Endpoint
Sex/
Species
POD
Basis
Best-fit
Model; BMR
Candidate
POD
(mg/m3)
BW
(kg)
PODAD,b
(mg/L)
Neurotoxicological Endpoints
Korsak and
Rydzynski
(1996)
Decreased pain
sensitivity
Impaired
neuromuscular
function and
coordination
Male, rat
Male, rat
BMDL
BMDL
Exponential
4;1SD
Log-logistic;
10% ER
84.0
93.9
0.387
0.387
0.085
0.096
Hematological Endpoints
Korsak et
al. (2000)
Decreased RBCs
Increased WBCs
Decreased
reticulocytes
Decreased clotting
time
Male, rat
Male, rat
Female, rat
Female, rat
BMDL
NOAEL
NOAEL
NOAEL
Exponential
4;1SD
n/a
n/a
n/a
174.1
492
492
123
0.390
0.399
0.230
0.243
0.187
0.867
0.890
0.127
Pulmonary Endpoints
Korsak et
al. (1997)
Korsak et
al. (2000)
Increased BAL
macrophages
Increased BAL total
cells
Increased
inflammatory lung
lesions
Male, rat
Male, rat
Male and
female, rat
NOAEL
LOAEL
NOAEL
n/a
n/a
n/a
123
123
123
0.383
0.383
0.390
0.127
0.127
0.127
DRAFT - DO NOT CITE OR QUOTE
70

-------
aGroup specific mean body weight [BW] reported in Korsak et al. (2000: Korsak et al.. 19971. For endpoints
from these studies using a NOAEL or LOAEL for the POD, the reported group specific mean BW for that dose
group was used in PBPK PODADj calculations. For decreased RBCs from Korsak et al. [2000]. the group
specific mean BW for the dose group closest to the BMDL was used. For decreased pain sensitivity and
coordination, balance, and neuromuscular function from Korsak and Rydzynski [1996]. the average of the
group specific mean BWs from Korsak et al. [2000: Korsak etal.. 1997] for the dose group closest to the
BMDL was used.
HVeekly average venous blood TMB concentration [mg/L] estimated for a rat exposed to the corresponding
candidate POD for 6 h/day, 5 d/wk. See Appendix A [U.S. EPA, 2011d] for details on PBPK modeling.
    Because an RfC is a toxicity value that assumes continuous human inhalation exposure
over a lifetime, data derived from inhalation studies in animals need to be adjusted to
account for the noncontinuous exposures used in these studies.  For 1,2,4-TMB, a PBPK
model (Hissink et al.. 2007} was employed to make this adjustment. This PBPK model
(described in Appendix A (U.S. EPA. 2011d}: Section A.I} was used to estimate the steady-
state weekly average venous blood concentration (mg/L} of 1,2,4-TMB for rats exposed to
1,2,4-TMB for 6 h/d, 5 d/wk. For each exposure concentration, once the model reached
steady-state, the resulting weekly average venous blood concentration (mg/L} of 1,2,4-
TMB was employed as the dose metric for these endpoints. This dose metric was
considered the duration-adjusted POD (PODADj} for each candidate critical effect (Table 2-
1).
2.1.2. RfC Derivation for 1,2,4-TMB

   For the derivation of an RfC based upon an animal study, the calculated PODADj values
are further adjusted to reflect the human equivalent concentration (HEC} (Table 2-2}.

Table 2-2: Candidate PODADJ values, human equivalent concentrations (HECs), and
applied uncertainty factors used in the derivation of RfCs for 1,2,4-TMB
Reference
Endpoint
PODADJ
(mg/L)
HEC
(mg/m3
)a
Uncertainty Factors (UF)
UFA
UFH
UFL
UFS
UFD
UFTOTAL
Candidate
RfC
(mg/m3)b
Neurotoxicological Endpoints
Korsak and
Rydzynski
[1996]
Decreased pain
sensitivity
Impaired
neuromuscular
function and
coordination
0.085
0.096
15.6
17.6
3
3
10
10
1
1
10
10
3
3
1,000
1,000
1.56xlO-2
1.76 x ID'2
Hematological Endpoints
Korsak et
al. [2000]
Decreased RBCs
Increased WBCs
Decreased
reticulocytes
0.187
0.867
0.890
33.7
131.5
134.0
3
3
3
10
10
10
1
1
1
10
10
10
3
3
3
1,000
1,000
1,000
3.37x10-2
1.31 x 10-1
1.34x10-1
DRAFT - DO NOT CITE OR QUOTE
71

-------

Decreased
clotting time
0.127
23.2
3
10
1
10
3
1,000
2.32 x lO-2
Pulmonary Endpoints
Korsak et
al. [1997]
Korsak et
al. (2000)
Increased BAL
macrophages
Increased BAL
total cells
Increased
inflammatory
lung lesions
0.127
0.127
0.127
23.2
23.2
23.2
3
3
3
10
10
10
1
10
1
10
10
10
3
3
3
1,000
10,000
1,000
2.32 x lO-2
n/ac
2.32 x lO-2
a Human equivalent concentration
b As calculated by application of uncertainty factors, not rounded.
c Endpoint excluded for further consideration due to a UP-TOTAL of 10,000. The 2002 report, "A Review of the
Reference Dose and Reference Concentration Processes"  [U.S. EPA, 20021 recommends a maximum total UF
of 3000 for derivation of an RfC.
   The HEC was derived using a human PBPK model (Hissinketal.. 2007} to account for
interspecies differences in toxicokinetics. The human PBPK model was run (as described in
Appendix A [U.S. EPA. 2011d}}. assuming a continuous (24 h/day, 7 day/week] exposure,
to estimate a human PODHEC that would result from the same weekly average venous
blood concentration reflected in the PODADJ in animals (Table 2-2}.
   Neurotoxicity is the most consistently observed endpoint in the toxicological database
for 1,2,4-TMB. According to EPA's Guidelines for Neurotoxicity Risk Assessment (1998},
many neurobehavioral changes are regarded as adverse, and the observation of correlated
and replicated measures of neurotoxicity strengthen the evidence for a hazard. Decreased
pain sensitivity, as measured as latency to paw-lick, is a measure of nociception (i.e.,
decreased pain sensitivity} and therefore this endpoint represents an alteration in
neurobehavioral function (U.S. EPA. 1998}. The observation of decreased pain sensitivity
was observed in multiple studies across multiple exposure durations (Gralewicz and
Wiaderna. 2001: Gralewicz etal.. 1997a: Korsak and Rydzynski. 1996: Korsak etal.. 1995}.
and in the presence  of other metrics of altered neurobehavior, including impaired
neuromuscular function and coordination and altered cognitive function. Additionally,
neurotoxicological endpoints (hand tremble, weakness} are observed in human worker
populations exposed to complex VOC mixtures containing 1,2,4-TMB. In consideration of
the recommendations in the U.S. EPA's Guidelines for Neurotoxicity Risk Assessment (1998}
and given the consistency of effect across independent studies, multiple durations of
exposure in animal studies,  the consistency of observed neurotoxicity in animals and
humans, the EPA concluded that neurotoxicity represents strong evidence of toxicity
hazard and that decreased pain sensitivity is an adverse effect, and the most appropriate
effect on which to base the RfC. Therefore, decreased pain sensitivity was selected as the
DRAFT - DO NOT CITE OR QUOTE
72

-------
critical effect and Korsak and Rydzynski (1996) as the principal study for the 1,2,4-TMB
RfC.
   A PODHEC of 15.6 mg/m3 for decreased pain sensitivity (Korsak and Rydzynski. 1996}
was used as the POD to derive the chronic RfC for 1,2,4-TMB.  The uncertainty factors,
selected based on EPA's A Review of the Reference Dose and Reference Concentration
Processes (2002) (Section 4.4.5 of the report], address five areas of uncertainty resulting in
a total UF of 1,000.  This composite uncertainty factor was applied to the selected POD to
derive an RfC.
   An interspecies uncertainty factor, UFA, of 3 (W1/2 = 3.16, rounded to 3} was applied to
account for uncertainty in characterizing the toxicokinetic and toxicodynamic differences
between rats and humans following inhalation exposure to 1,2,4-TMB. In this assessment,
the use of a PBPK model to convert internal doses in rats to administered doses in humans
reduces toxicokinetic uncertainty in extrapolating from the rat to humans, but does not
account for interspecies differences due to toxicodynamics. A default UFA of 3 was thus
applied to account for this remaining toxicodynamic uncertainty.
   An intraspecies uncertainty factor, UFn, of 10 was applied to account for potentially
susceptible individuals in the absence of data evaluating variability of response in the
human population following inhalation of 1,2,4-TMB. No information is currently available
to predict potential variability in human susceptibility, including variability in the
expression  of enzymes involved in 1,2,4-TMB metabolism.  Due to this lack of data on
variability within the human population, a default 10-fold UFn was applied.
   A LOAEL to NOAEL uncertainty factor, UFi of 1 was applied because the current
approach is to address this factor as one of the considerations in selecting a BMR for BMD
modeling. In this case, a BMR equal to a change in the mean equal to 1 standard deviation
of the model estimated control mean for decreased pain sensitivity was selected under the
assumption that this BMR represents a minimally, biologically significant change for this
endpoint.
   A subchronic to chronic uncertainty factor, UFs, of 10 was applied to account for
extrapolation from a subchronic exposure duration study to derive a chronic RfC.
   A database uncertainty factor, UFo, of 3 (W1/2 = 3.16, rounded to 3} was applied to
account for database deficiencies due to the lack of a multi-generation reproductive toxicity
study. The  database contains three subchronic studies that are well-designed and observe
exposure-response effects in multiple organ systems in rats exposed to  1,2,4-TMB via
inhalation (nervous, hematological, and pulmonary systems}.  The database additionally

DRAFT - DO NOT CITE OR QUOTE                                                       73

-------
contains a well-designed developmental toxicity study that investigated standard measures
of maternal and fetal toxicity in a different strain of rat. Although there is no information
regarding the potential for developmental neurotoxicity and the critical effect for the RfC is
altered CNS function, this raises concern regarding possible neurodevelopmental effects of
1,2,4-TMB exposure. However, in the absence of information regarding the magnitude of
transfer of 1,2,4-TMB or its metabolites across the placenta, and given the observation that
other developmental effects occur at concentrations 6- to 24-fold greater than those
eliciting neurotoxicity in adult animals, it may be unlikely that neurodevelopmental data
would result in a lower RfC.
   Application of this  1000-fold composite UF to the PODHEC yields the following chronic
RfC for 1,2,4-TMB:

RfC = PODHEC * UF = 15.6 mg/m3 * 1,000 = 0.02  mg/m3 = 2 x 10 2 mg/m3 (rounded to
one significant digit)

   A confidence level of high, medium, or low is assigned to the study used to derive the
RfC, the overall database, and the RfC itself, as described in Section 4.3.9.2 of EPA's Methods
for Derivation of Inhalation Reference Concentrations and Application of Inhalation
Dosimetry [U.S. EPA. 1994). Confidence in the study from which the critical effect was
identified, Korsak and Rydzynski (1996} is medium. The study is a well-conducted peer-
reviewed study that utilized three dose groups plus untreated controls, an appropriate
number of animals per dose group, and performed statistical analyses. The critical effect
on which the RfC is based is well-supported as the weight of evidence for 1,2,4-TMB-
induced neurotoxicity is coherent across multiple animals species (i.e., human, mouse, and
rat] and consistent across multiple exposure durations (i.e., acute, short-term, and sub-
chronic] (Gralewicz and Wiaderna. 2001: Chenetal.. 1999: Wiaderna etal.. 1998:
Gralewicz etal.. 1997a: Gralewicz etal.. 1997b: Korsak and Rydzynski. 1996: Norseth et al..
1991). Confidence in the database for 1,2,4-TMB is low to medium as the database includes
acute, short-term, subchronic, and developmental  toxicity studies in rats and mice.  The
database lacks a chronic and multigenerational reproductive study, and the studies
supporting the critical effect predominately come from the same research institute. Overall
confidence in the RfC for 1,2,4-TMB is low to medium.

2.1.3. Comparison of Candidate RfCs for 1,2,4-TMB

   The predominant noncancer effect observed following acute, short-term, and
subchronic inhalation  exposures to 1,2,4-TMB is neurotoxicity, although respiratory
DRAFT - DO NOT CITE OR QUOTE                                                      74

-------
toxicity is also observed following acute and subchronic exposures, while hematological
effects are observed after subchronic exposures. Figure 2-1 provides a graphical display of
all of the candidate PODs and RfCs derived from the three subchronic studies considered in
the selection of the final POD for the inhalation RfC.
Decreased
  pain
sensitivity
                  Decreased  Increased BAL  Increased   Decreased
                   neuro-   macrophages inflammatory   RBCs
                  muscular             lung lesions
                  function
          Neurotoxicological
             Endpoints
                    Pulmonary Endpoints
ncreased   Decreased   Decreased
 WBCs    reticulocytes  clotting time
                                                        Respiratory Endpoints
      D Data base Uncertainty • Subchronic to Chronic  Q Intraspecies Variability  Qlnterspecies Extrapolation  ORfC  O POD
Figure 2-1: Array of candidate PODHEC values with applied UFs and candidate RfCs for
CNS, hematological, and pulmonary effects resulting from inhalation exposure to
1,2,4-TMB

2.1.4. Uncertainties in the Derivation of the RfC for 1,2,4-TMB

   As presented above, the UF approach following EPA practices and RfC guidance (U.S.
EPA. 2002.19941 was applied to the PODHEC in order to derive the chronic RfC for
1,2,4-TMB. Factors accounting for uncertainties associated with a number of steps in the
analyses were adopted to account for extrapolation from animals to humans, a diverse
human population of varying susceptibilities, POD determination methodologies (NOAEL,
LOAEL, or BMDL), and database deficiencies.
   The critical effect selected, decreased pain sensitivity, does not introduce substantial
uncertainty into the RfC calculation as selection of alternative CNS, hematological, or
pulmonary effects would result in an equivalent RfCs (i.e., 2 x 10-2 mg/m3,  see Figure 2-1}.
Some uncertainty does exist regarding the selection of the BMRs for use in BMD modeling,
but selection of 10% extra risk for dichotomous endpoints and 1 SD for continuous
endpoints is supported by current EPA guidance (U.S. EPA. 2000a).  Uncertainty regarding
DRAFT - DO NOT CITE OR QUOTE
                                                                             75

-------
the selection of particular models for individual endpoints does exist as selection of
alternative models could decrease or increase the RfC. However, the best-fit model is the
most appropriate for RfC derivation based on current EPA guidance (U.S. EPA. 2000a}.
Uncertainty may exist in the PBPK model estimates of internal blood dose metrics for the
rat, and subsequent HEC calculations for the human, including parameter uncertainly, but
such uncertainties would apply equally to all endpoints. Lastly, the extent of inter-
individual variation of 1,2,4-TMB metabolism in humans and potential susceptible
subpopulations have not been well characterized, and thus these two considerations
remain sources of some uncertainty in the current assessment.

2.1.5. Dose-Response Assessment for RfC derivation for 1,3,5-TMB

   As discussed above in Section 1.2.2, endpoints observed in Saillenfait et al. (2005) that
demonstrated statistically significant (at p < 0.05 or greater] pair-wise increases or
decreases relative to control for at least one dose group were considered for the derivation
of the RfC for 1,3,5-TMB; these effects are listed in Table 1-7. Additionally, altered
cognitive function, decreased pain sensitivity, and decreased anxiety and/or increased
motor function observed in Gralewicz et al. (2001) and Wiaderna et al. (2002) were also
considered as the basis for the derivation of the RfC for 1,3,5-TMB. This assessment used
the BMD approach, when possible, to estimate a POD for the derivation of an RfC for
1,3,5-TMB (Table 2-4; see Section C.I of Appendix B (U.S. EPA. 2011c} for detailed
methodology}. The BMD approach involves fitting a suite of mathematical models to the
observed dose-response data using EPA's BMDS (version 2.2}. Each fitted model estimates
a BMD and its associated BMDL corresponding to a selected BMR. For continuous data (i.e.,
decreased male and female fetal weight} from the Saillenfait et al. (2005} study, a BMR
equal to 5% relative deviance from the control mean was used. A decrease in body weight
of 10% is generally assumed to be a minimally biologically significant response in adult
animals.  Because the developing organism may be more sensitive to decreases in body
weight, a 5% decrease in fetal body weight relative to control was assumed to be a
minimally biologically significant response for the fetuses in the Saillenfait et al. (2005}
study. As recommended by EPA's Benchmark Dose Technical Guidance (2000aj. a BMR
equal to a change in the mean of 1 standard deviation of the model estimated control mean
was also used in modeling the fetal endpoints for comparison purposes. No information is
available regarding the magnitude of response that would be considered biologically
significant for decreased maternal weight gain. Thus, a BMR equal to a change in the mean
equal to 1 standard deviation of the model estimated control mean was used in modeling
this endpoint. The estimated BMDL is then used as the POD  for deriving the RfC.
DRAFT - DO NOT CITE OR QUOTE                                                       76

-------
   The suitability of the above methods to determine a POD is dependent on the nature of
the toxicity database for a specific chemical. The data for neurotoxicity (i.e., altered
cognitive function, decreased pain sensitivity, and decreased anxiety and/or increased
motor function] for 1,3,5-TMB were not amenable to BMD modeling. Gralewicz and
Wiaderna (2001} only employed one exposure concentration when investigating the
neurotoxic effects of 1,3,5-TMB following short-term inhalation exposures.  Thus, the
observed neurotoxic effects in this study were not amenable to modeling according to EPA
guidance (2000a}. For altered cognitive function (as measured as decreased passive and
active avoidance] reported in Wiaderna et al. (2002], responses were observed to be equal
in all exposure groups. Therefore, for the neurotoxic effects observed in Gralewicz and
Wiaderna (2001] and Wiaderna et al. (2002] the NOAEL/LOAEL approach was used to
determine a POD. In the Saillenfait et al. (2005] study, although decreased fetal body
weight in females was considered appropriate for BMD modeling, BMDS was unable to
adequately model the variance in response for this endpoint. Therefore, the
NOAEL/LOAEL approach was also used in this case to identify a POD. Detailed modeling
results are provided in Section B.2 of Appendix B (U.S. EPA. 2011c].
   Because an RfC is a toxicity value that assumes continuous human inhalation exposure
over a lifetime, data derived from inhalation studies in animals need to be adjusted to
account for the noncontinuous exposures used in these studies. In the Gralewicz and
Wiaderna (2001] and Wiaderna et al. (2002] studies, rats were exposed to 1,3,5-TMB for 6
hours/day, 5 days/week for 4 weeks. Because no PBPK model exists for 1,3,5-TMB, the
duration-adjusted PODs for neurobehavioral effects in rats were calculated as follows:
   PODADJ (mg/m3) = POD (mg/m3) x hours exposed per day/24 hours x days
   exposed per week/7 days
   Therefore, for altered cognitive function from Gralewicz and Wiaderna (2001], the
PODADJ would be calculated as follows:
   PODADJ (mg/m3) = 492 mg/m3x 6 hours/24 hours x 5 days/7 days
   PODADJ (mg/m3) = 87.9 mg/m3
   In the Saillenfait et al. (2005] study, rats were exposed to 1,3,5-TMB for 6 hours/day for
15 consecutive days (CDs 6-20]. Therefore, the duration-adjusted PODs for
developmental/ maternal effects were calculated as follows:
   PODADJ (mg/m3) = POD (mg/m3) x hours exposed per day/24 hours
   For example, for decreased fetal weight in males, the PODADJ would be calculated as
follows:
DRAFT - DO NOT CITE OR QUOTE                                                    77

-------
          (mg/m3) = 1649 mg/m3 x 6 hours/24 hours
   PODADj (mg/m3) =412.0 mg/m3
   The calculated PODADJ (mg/m3} values for all neurotoxicity and developmental
endpoints considered for RfC derivation are presented in Table 2-4.

Table 2-4: Duration adjusted point of departure (PODADj) estimates from short-term
and gestational inhalation exposures to 1,3,5-TMB
Reference
Endpoint
Sex/
Species
POD
Basis
Best-fit
Model; BMR
Candidate
POD
(mg/m3)
PODADJ
(mg/L)»
Neurotoxicological Endpoints
Gralewicz and
Wiaderna
[2001]
Wiaderna et al.
[20021
Altered cognitive
function
Decreased pain
sensitivity
Decreased anxiety
and/or increased
motor function
Altered cognitive
function
Male, rat
Male, rat
Male, rat
Male, rat
LOAEL
LOAEL
LOAEL
LOAEL
n/a
n/a
n/a
n/a
492
492
492
123
87.9
87.9
87.9
22.0
Developmental Endpoints
Saillenfait et al.
[2005]
Decreased fetal
body weight
Male, rat
Female, rat
BMDLC
NOAELC
Exponential
2; 5% RD
n/a
1,649
2,952
412.0
738.0
Maternal Endpoints
Saillenfait et al.
[2005]
Decreased
maternal weight
body gain
Female, rat
BMDL
Power, 1 SD
1,303
326.0
a Duration adjusted PODADj [mg/m3] = POD x [6 hours/24 hours] for developmental/maternal endpoints, or
POD x [6 hours/24 hours] x [5 days/week] in accordance with EPA policy [U.S. EPA. 2002]
2.1.6. RfC Derivation for 1,3,5-TMB

   Because the selected endpoints for consideration as the critical effect (altered cognitive
function, decreased pain sensitivity, decreased anxiety and/or increased motor function,
decreased fetal body weight, and maternal body weight gain] are assumed to result
primarily from systemic distribution of 1,3,5-TMB, and no available PBPK model exists for
1,3,5-TMB, the human equivalent concentration for 1,3,5-TMB was calculated by the
application of the appropriate dosimetric adjustment factor (DAF) for systemically acting
gases (i.e., Category 3 gases], in accordance with the U.S. EPA RfC Methodology (U.S. EPA.
DRAFT - DO NOT CITE OR QUOTE
78

-------
1994). DAFs are ratios of animal and human physiologic parameters, and are dependent on
the nature of the contaminant (particle or gas] and the target site (e.g., respiratory tract or
remote to the portal-of-entry) (U.S. EPA. 1994}. For gases with systemic effects, the DAF is
expressed as the ratio between the animal and human blood:air partition coefficients:
   DAF = (Hb/g)A/(Hb/b)H
   DAF = 55.7/43
   DAF = 1.3
   where:
   (Hb/g)A  = the animal blood:air partition coefficient
   (Hb/g)H  = the human blood:air partition coefficient
   In cases where the animal blood:air partition coefficient is higher than the human value
(Meulenberg and Vijverberg. 2000: larnbergandlohanson. 1995). resulting in a DAF > 1, a
default value of 1 is substituted (U.S. EPA. 1994). For example, the HEC for altered CNS
function (reported in Wiaderna et al. (2002) is calculated as follows:
   PODHEc  = PODADj (mg/m3) x DAF
   PODHEc  = PODADj (mg/m3) x 1.0
   PODHEC  = 22 mg/m3 x 1.0
   PODHEC  = 22 mg/m3
   Table 2-5 presents the derivation of candidate RfCs from the selected short-term and
developmental toxicity studies (Saillenfait et al.. 2005: Wiaderna etal.. 2002: Gralewicz and
Wiaderna. 20011
DRAFT - DO NOT CITE OR QUOTE                                                      79

-------
Table 2-5: Candidate PODADJ values, human equivalent concentrations (HECs), and
applied uncertainty factors used in the derivation of RfCs for 1,3,5-TMB
Referenc
e
Endpoint
PODADJ
(mg/L)
HEC
(mg/m3
)a
Uncertainty Factors (UF)
UFA
UFH
UFL
UFS
UFD
UFTOTAL
Candidate
RfC
(mg/m3)b
Neurotoxicological Endpoints
Gralewicz
and
Wiaderna
C20Q1)
Wiaderna
etal.
[2002]
Altered cognitive
function
Decreased pain
sensitivity
Decreased anxiety
and/or increased
motor function
Altered cognitive
function
87.9
87.9
87.9
22.0
87.9
87.9
87.9
22.0
3
3
3
3
10
10
10
10
10
10
10
10
10
10
10
10
3
3
3
3
10,000
10,000
10,000
10,000
n/ac
n/ac
n/ac
n/ac
Developmental Endpoints
Saillenfait
etal.
[2005]
Decreased fetal
body weight, male
Decreased fetal
body weight,
female
412.0
738.0
412.0
738.0
3
3
10
10
1
1
1
1
3
3
100
100
4.12
7.38
Maternal Endpoints
Saillenfait
etal.
T20051
Decreased
maternal weight
body gain
326.0
326.0
3
10
1
10
3
1,000
3.26x10-1
a Human equivalent concentration
b As calculated by application of uncertainty factors, not rounded.
c Endpoint excluded for further consideration due to a UFTOTAL of 10,000. The 2002 report "A Review of the
Reference Dose and Reference Concentration Processes" [U.S. EPA. 2002] recommends a maximum total UF
of 3000 for derivation of an RfC.
   The magnitude of the total uncertainty factors associated with the neurotoxicological
endpoints from Gralewicz and Wiaderna (2001} and Wiaderna et al. (2002} indicate that
these endpoints cannot support the derivation of an RfC for 1,3,5-TMB. The composite UF
for 1,3,5-TMB for the neurotoxicological endpoints from Gralewicz and Wiaderna (2001}
and Wiaderna et al. (2002} would be 10,000. In the report, A Review of the Reference Dose
and Reference Concentration Processes fU.S. EPA. 20021 the RfD/RfC  Technical Panel
concluded that, in cases where maximum uncertainly exists in four or more areas of
uncertainly, or when the total uncertainly factor is 10,000 or more, it is unlikely that the
database is sufficient to derive a reference value. Therefore, consistent with the
recommendations in U.S. EPA (2002J. the available neurotoxicity data following short-term
DRAFT - DO NOT CITE OR QUOTE
80

-------
inhalation exposure to 1,3,5-TMB were considered insufficient to support reference value
derivation and an RfC for 1,3,5-TMB was not derived based on these data.
   Of the remaining effects considered for derivation of the RfC, decreased maternal
weight gain (PODHEC = 326.0 mg/m3} was identified as the most sensitive endpoint. A
PODHEC of 326.0 mg/m3 for decreased maternal weight gain by Saillenfait et al. (2005}
was used as the PODHEC to derive a candidate chronic RfC for 1,3,5-TMB as shown in Table
2-5. The uncertainty factors, selected based on EPA's A Review of the Reference Dose and
Reference Concentration Processes (2002}. address five areas of uncertainty resulting in a
total UF of 1000. This composite uncertainty factor was applied to the selected POD to
derive an RfC.
   An interspecies uncertainty factor, UFA, of 3 (W1/2 = 3.16, rounded to 3} was applied to
account for uncertainty in characterizing the toxicokinetic and toxicodynamic differences
between rats and humans following inhalation exposure to 1,3,5-TMB. In this assessment,
the use of a DAF to  extrapolate external exposure concentrations from rats to humans
reduces toxicokinetic uncertainty in extrapolating from the rat data, but does not account
for the possibility that humans may be more sensitive to 1,3,5-TMB than rats due to
toxicodynamic differences. A default UFA of 3 was thus applied to account for this
remaining toxicodynamic uncertainty.
   An intraspecies uncertainty factor, UFn, of 10 was applied to account for potentially
susceptible individuals in the absence of data evaluating variability of response in the
human population following inhalation of 1,3,5-TMB. No information is currently available
to predict potential variability in human susceptibility, including variability in the
expression of enzymes involved in 1,3,5-TMB metabolism. Due to this lack of data on
variability within the human  population, a default 10-fold UFn is applied.
   A LOAEL to NOAEL uncertainty factor, UFi of 1 was applied because the current
approach is to address this factor as one of the considerations in selecting a BMR for BMD
modeling. In this case, a BMR equal to a change in the mean of 1 standard deviation of the
model estimated control mean for decreased maternal body weight gain was selected
under an assumption that this BMR level represents a minimally, biologically significant
change for this endpoint.
   A subchronic to chronic uncertainty factor, UFs, of 10 was applied  to account for
extrapolation from a subchronic exposure duration study to derive a chronic RfC.
   A database uncertainty factor, UFo, of 3 (101/2 = 3.16, rounded to 3} was applied to
account for database deficiencies due to the lack of a multi-generation reproductive toxicity

DRAFT - DO NOT CITE OR QUOTE                                                       81

-------
study. The database contains two short-term studies that are well-designed and observe
exposure-response effects in the central nervous system of exposed rats. The database
additionally contains a well-designed developmental toxicity study that investigated
standard measures of maternal and fetal toxicity in a different strain of rat. A limitation of
the database is the lack of any information regarding the potential for developmental
neurotoxicity. As altered neurobehavioral function is observed in rats exposed to
1,3,5-TMB (manifested as decreased ability to learn passive and active avoidance and
decreased pain sensitivity], this raises concern for neurodevelopmental effects of
1,3,5-TMB exposure. However, in the absence of information regarding the magnitude of
transfer of 1,3,5-TMB or its metabolites across the placenta, it may be unlikely that
neurodevelopmental data would result in a lower RfC.
   Application of this 1000-fold composite UF yields the calculation of the chronic RfC for
1,3,5-TMB as follows:

RfC = PODHEc * UF = 326 mg/m3 * 1000 = 0.326 mg/m3 = 3 x 10 1 mg/m3 (rounded to
one significant digit)

   While Saillenfait et al. (2005) is a well-conducted developmental toxicity study that
utilizes appropriate study design, group  sizes, and statistics, and investigates a wide range
of fetal and maternal endpoints resulting from 1,3,5-TMB inhalation exposure, a number of
additional factors lessens its suitability with which to derive the RfC for 1,3,5-TMB.  First,
although maternal and fetal toxicities were observed in this study, it is important to note
that the candidate RfC for 1,3,5-TMB derived based on the critical effect of decreased
corrected (for gravid uterine weight] maternal body weight gain is  15-fold higher than the
RfC derived for 1,2,4-TMB (based on altered CNS function measured as decreased pain
sensitivity]. As discussed in Section 1.1,  the available toxicological database for 1,2,4-TMB
and 1,3,5-TMB, across all exposure durations, indicates there are important similarities in
the two isomer's toxicity that are supportive of not deriving an RfC for 1,3,5-TMB that is
substantially greater than the RfC value derived for 1,2,4-TMB.
   In acute studies investigating the respiratory irritative effects of the two isomers, the
RD50 of 1,2,4-TMB and 1,3,5-TMB were  observed to  be very similar: 2,844 and 2,553
mg/m3, respectively (Korsak et al.. 1997]. This similarity regarding toxicity was also
observed in acute neurotoxicity studies:  the EC50 for decreased coordination, balance, and
neuromuscular function (i.e., performance on the rotarod] was 4,694 mg/m3 for 1,2,4-TMB
and 4,738 mg/m3 for 1,3,5-TMB.  The ECso for decreased pain sensitivity (i.e., latency to
paw-lick measured on the hot plate apparatus] were also similar for both isomers: 5,683

DRAFT - DO NOT CITE OR QUOTE                                                       82

-------
mg/m3 for 1,2,4-TMB and 5,963 mg/m3 for 1,3,5-TMB fKorsakand Rydzynski. 19961
Other neurotoxic endpoints similarly affected by either isomer (albeit from oral exposures
or i.p. injections] included increased electrocortical arousal and altered EEC function
(Tomas etal.. 1999a: Tomas etal.. 1999c}. However, the doses eliciting these effects were
LOAELs, and therefore it is unclear whether this represents true similarity in toxic potency
or whether testing at lower doses would reveal differences between the two isomers.
Additionally, the magnitude of effect differed between isomers, with 1,2,4-TMB and
1,3,5-TMB inducing greater changes in brain EEGs and electrocortical arousal, respectively.
   In short-term neurotoxicity studies, a similar pattern of effects (inability to learn
passive or active avoidance, decreased pain sensitivity, increased spontaneous motor
activity] indicating altered neurobehavioral function was observed in rats exposed to
either isomer (Wiaderna etal.. 2002: Gralewicz and Wiaderna. 2001: Gralewicz et al..
1997a].  In these studies, 1,3,5-TMB was shown to be more toxic that 1,2,4-TMB, with
neurobehavioral effects occurring at lower exposures (123 vs. 492 mg/m3] in animals
exposed to 1,3,5-TMB vs. those exposed to 1,2,4-TMB. When comparing the magnitude of
TMB isomer-induced neurotoxicity, exposure to 492 mg/m3 1,3,5-TMB induced greater
decrements in avoidance acquisition (50% vs. 40%] compared to exposure to the same
concentration of 1,2,4-TMB. Lastly, manifestation of neurotoxicity occurred at earlier time
points (three vs. seven days] in rats exposed to 1,3,5-TMB compared to those exposed to
1,2,4-TMB.
   Finally, the observed developmental effects observed in Saillenfait et al. (2005] were
shown to be similar between isomers. Exposure to 1,2,4-TMB and 1,3,5-TMB significantly
decreased male  fetal body weights to a similar degree (5% and 7%, respectively] at 2952
mg/m3. 1,2,4-TMB and 1,3,5-TMB also decreased female body weights to a similar degree
(5% and 6%, respectively] at the same exposure concentration. This body weight decrease
was significant in animals exposed to 1,2,4-TMB, but was not significant in those females
exposed to 1,3,5-TMB. 1,3,5-TMB was observed to be more toxic with regard to maternal
toxicity, inducing a 75% reduction in maternal weight gain at 2952 mg/m3 compared to a
50% reduction in animals exposed to the same concentration of 1,2,4-TMB.
   The two isomers are similar to one another in their chemical and toxicokinetic
properties, although important differences do also exist. Both isomers have very similar
Log KOW values, and blood:air partition coefficients reported for humans and rats in the
literature are similar between isomers: 43.0 for 1,2,4-TMB and 59.1 for 1,3,5-TMB. This
gives the strong indication that the two isomers would partition into the blood in a similar
fashion.  Supporting this is the observation that 1,2,4-TMB and 1,3,5-TMB absorb equally

DRAFT - DO NOT CITE OR QUOTE                                                      83

-------
into the bloodstream of exposed humans (6.5 and 6.2 |iM, respectively] Qarnberg et al..
1996). Also, the net respiratory uptake of 1,2,4-TMB and 1,3,5-TMB was similar in humans,
and the respiratory uptake for 1,2,4-TMB was similar between humans and rats Qarnberg
etal.. 1996: Dahl etal.. 1988}. Distribution of the two isomers throughout the body is
qualitatively similar, although it appears that liver and kidney concentrations for
1,2,4-TMB were greater than those for 1,3,5-TMB in both acute and short-term exposures
fSwiercz etal.. 2006: Swiercz etal.. 2003: Swiercz etal.. 20021 Although 1,2,4-TMB was
observed to distribute to the brain (Swiercz et al.. 2003: Eide and Zahlsen. 1996}.
distribution of 1,3,5-TMB to the brain was not experimentally measured in any study.
However, the predicted brain:air partition coefficient was similar between 1,2,4-TMB and
1,3,5-TMB for both humans (206 vs. 199} and rats (552 vs. 535} fMeulenberg and
Viiverberg. 20001 This strongly suggests that 1,2,4-TMB and 1,3,5-TMB can be expected to
distribute similarly to the brain in both humans and rats. Both isomers were observed to
primarily metabolize to benzoic and hippuric acids in humans and rats Qarnberg etal..
1996: Huoetal.. 1989: Mikulski and Wiglusz. 19751 although the amount of inhaled TMB
recovered as hippuric acid metabolites following exposure to 1,2,4-TMB or 1,3,5-TMB was
somewhat dissimilar in humans (22% vs. 3%, respectively} and rats (24-38% vs. 59%,
respectively} Qarnberg et al.. 1996: Mikulski and Wiglusz. 1975}. Other terminal
metabolites included mercapturic acids (-14-19% total dose}, phenols (-12% total dose},
and glucuronides and sulphuric acid conjugates (4-9% total dose} for 1,2,4-TMB and
phenols (-4-8% total dose} and glucuronides and sulphuric acid conjugates (-5-9% total
dose} for 1,3,5-TMB fTsuiimoto etal.. 2005: Tsuiimoto etal.. 2000: Huoetal.. 1989:
Wiglusz. 1979: Mikulski and Wiglusz.  1975}. In humans, the half-lives of elimination from
blood were observed to be greater for 1,3,5-TMB (1.7 minutes, 29 minutes, 4.9 hours, and
120 hours} than for 1,2,4-TMB (1.3 minutes, 21 minutes, 3.6 hours, and 87 hours}
Qarnberg etal.. 1997a: Tarnbergetal.. 1997b: Tarnbergetal.. 19961 although this
difference may be due to small sample sizes and difficulties in measuring slow elimination
phases rather than a true difference in half-lives.  At low exposure concentrations, half-lives
in elimination from the blood were somewhat similar for 1,2,4-TMB and 1,3,5-TMB (3.6 vs.
2.7 hours}, but this difference became much greater with increasing doses (17.3 hours for
1,2,4-TMB and 4 hours for 1,3,5-TMB following exposure to 1230 mg/m3 for six hours}
fSwiercz etal.. 2003: Swiercz etal.. 20021.
   Given the above information regarding the observed toxicity following 1,2,4-TMB and
1,3,5-TMB exposures across acute, short-term, and developmental studies, the use of
1,3,5-TMB-specific data for derivation of an RfC was not considered by EPA to be
scientifically supported. Derivation of an RfC for 1,3,5-TMB using the only adequate
DRAFT - DO NOT CITE OR QUOTE                                                     84

-------
toxicity data available (i.e., Saillenfait et al. (2005)) would result in an RfC 15-fold higher
than the RfC derived for 1,2,4-TMB based on altered CNS function (i.e., decreased pain
sensitivity}. The available toxicity data indicates that 1,2,4-TMB and 1,3,5-TMB are similar
in acute respiratory and neurological toxicity and developmental toxicity, but that
1,3,5-TMB appears to be more potent in eliciting neurotoxicity and maternal toxicity
following short-term exposures. 1,3,5-TMB is observed to elicit neurotoxic effects in rats in
acute and short-term studies, and therefore the selected critical effect for 1,2,4-TMB,
altered CNS function, is relevant to observed 1,3,5-TMB-induced toxicity.  Similarities in
blood:air partition coefficients, respiratory uptake, and absorption into the bloodstream
between the two isomers support the conclusion that internal blood dose metrics for
1,3,5-TMB would be similar as those calculated for 1,2,4-TMB using the available PBPK
model.
   Thus, the chronic RfC of 2 x 10-2 mg/m3 derived for 1,2,4-TMB was adopted as
the RfC for 1,3,5-TMB based on the conclusion that the two isomers were sufficiently
similar regarding chemical properties, kinetics, and toxicity.
      As noted previously, a confidence level of high, medium, or low is assigned to the
study used to derive the RfC, the overall database, and the RfC itself, as described in EPA
(1994). Section 4.3.9.2.  The chronic RfC of 2 x 10-2 mg/m3 derived for 1,2,4-TMB was
adopted as the RfC for 1,3,5-TMB based on the conclusion that the two isomers were
sufficiently similar regarding chemical properties, kinetics, and toxicity. Thus, confidence
in the study from which the critical effect was identified, Korsak and Rydzynski (1996) is
medium (see above). Confidence in the database is low to medium as the database includes
acute, short-term, and developmental toxicity studies in rats and mice. The database lacks
a chronic, subchronic, and multigenerational reproductive study. Additionally, the studies
supporting the critical effect predominately come from the same research institute.
Overall confidence in the RfC for 1,3,5-TMB is low due to uncertainties surrounding the
adoption of the RfC derived for 1,2,4-TMB as the RfC for 1,3,5-TMB.
   2.1.7. Uncertainties in the Derivation of the RfC for 1,3,5-TMB
   Uncertainties exist in adopting the RfC derived for 1,2,4-TMB based on altered CNS
function (i.e., decreased pain sensitivity) as the RfC for 1,3,5-TMB. The available database
for 1,3,5-TMB was considered insufficient with which to derive an RfC. If the most
sensitive endpoint from the only adequate study in the 1,3,5-TMB database (i.e., decreased
maternal weight gain; Saillenfait et al. (2005)). was used for the RfC derivation, an RfC 15-
fold higher would be derived for 1,3,5-TMB vs. that derived for 1,2,4-TMB (3 x 10-1 vs. 2 x
10-2 mg/m3, respectively). Although uncertainly exists in adopting the 1,2,4-TMB RfC
DRAFT - DO NOT CITE OR QUOTE                                                      85

-------
value for 1,3,5-TMB, both isomers share multiple commonalities and similarities regarding
their toxicokinetic and toxicological properties that support the adoption of the value of
one isomer for the other. The majority of uncertainty regarding 1,3,5-TMB's database
involves the lack of a chronic, subchronic, or multi-generational reproductive study for this
isomer. Given the similarities in toxicity from the available developmental toxicity study,
and neurotoxicity and respiratory toxicity observed in the available acute and short-term
studies, there is strong evidence that the two isomer's toxicity resulting from subchronic
exposure can be expected to be similar. More so, 1,3,5-TMB may actually be expected to be
slightly more toxic than 1,2,4-TMB following subchronic exposures than 1,2,4-TMB given
the observation of greater magnitude and earlier onset of effect following 1,3,5-TMB
exposures in short-term studies. Therefore, while uncertainty does exist in the derivation
of 1,3,5-TMB's RfC, the available information regarding sufficient toxicokinetic and
toxicological similarity between the two isomers indicates this uncertainly does not
preclude adopting the RfC for 1,2,4-TMB as the RfC for 1,3,5-TMB.


2.2. Oral Reference Dose for Effects other than Cancer

2.2.1. Methods of analysis for RfD derivation for 1,2,4-TMB

   No  chronic or subchronic studies were identified for 1,2,4-TMB that utilized the oral
route of exposure. Therefore, the available oral database for 1,2,4-TMB is minimal as
defined by EPA guidance (i.e., there is no human data available nor any adequate oral
animal data] (U.S. EPA. 2002}. and this database is inadequate for the derivation of an RfD.
   Even though the available oral database for 1,2,4-TMB is inadequate to derive an RfD, a
route-to-route extrapolation from inhalation to oral for the purposes of deriving an RfD is
possible using the existing inhalation data and the available 1,2,4-TMB PBPK model
(Hissink et al.. 2007}. Using route-to-route extrapolation via application of PBPK models is
supported by EPA guidance (U.S. EPA. 2002.1994} given enough data and ability to
interpret that data regarding differential metabolism and toxicity between different routes
of exposure. The available database for 1,2,4-TMB supports the use of route-to-route
extrapolation: sufficient evidence exists that demonstrates similar qualitative profiles of
metabolism (i.e., observation of dimethylbenzoic and hippuric acid metabolites} and
patterns of parent compound distribution across exposure  routes (Appendix A (U.S. EPA.
2011djj. Further, no evidence exists that would suggest toxicity profiles would differ to a
substantial degree between oral and inhalation exposures.

DRAFT  - DO NOT CITE OR QUOTE                                                       86

-------
   Therefore, assuming oral exposure would result in the same systemic effect as
inhalation exposure (altered CNS function, measured as decreased pain sensitivity [Korsak
and Rydzynski. 1996}. an oral exposure component was added to the Hissink et al. (2007}
PBPK model by EPA (Appendix A (U.S. EPA. 2011dj: Section A.1.4}, assuming continuous
oral ingestion and 100% of the ingested 1,2,4-TMB is absorbed by constant infusion of the
oral dose into the liver. This is a common assumption when information about the oral
absorption of the compound is unknown. The contribution of the first-pass metabolism in
the liver for oral dosing was evaluated by simulating steady state venous blood levels (at
the end of 50 days continuous exposure}  for a standard human at rest (70 kg} for a range of
concentrations and doses; at low daily doses (0.1-10 mg/kg-day}, equivalent inhalation
concentrations result in steady state blood concentrations 4-fold higher than those
resulting from oral doses, indicating the presence of first-pass metabolism following oral
exposure. This difference became insignificant for daily doses exceeding 50 mg/kg-day
(Appendix A (U.S. EPA. 2011dj: Section A.1.4}.
   The human PBPK model inhalation dose metric (weekly average blood concentration,
mg/L}  for the PODADJ (0.085 mg/L} was used as the target for the oral dose metric. The
human PBPK model was run to determine what oral exposure would yield an equivalent
weekly average blood concentration and the resulting value of 6.2 mg/kg-day was used as
the human equivalent dose POD (PODHED} for the RfD derivation.

2.2.2. RfD Derivation for 1,2,4-TMB

   A PODHED of 6.2 mg/kg-day was derived for the oral database using route-to-route
extrapolation based on the neurotoxic effects observed by Korsak and Rydzynski (1996}
following inhalation exposure (decreased pain sensitivity}. Thus, the same uncertainty
factors applied to derive the RfC (see Section 2.1.2} were also applied to derive the RfD.
The uncertainty factors, selected based on EPA's report, A Review of the Reference Dose and
Reference Concentration Processes (2002J. address five areas of uncertainty resulting in a
composite UF of 1,000.
   Application of this 1,000-fold composite UF yields the calculation of the chronic RfD for
1,2,4-TMB as follows:

RfD = PODHED •*• UF = 6.2 mg/kg-day •*• 1,000 = 0.006 mg/kg-day = 6 x 10 3 mg/kg-day
(rounded to one significant digit)

   A PBPK model was utilized to perform a route-to-route extrapolation to determine a
POD for the derivation of the RfD from the Korsak and Rydzynski (1996} inhalation study
DRAFT  - DO NOT CITE OR QUOTE                                                      87

-------
and corresponding critical effect.  The confidence in the study from which the critical effect
was identified, Korsak and Rydzynski (1996) is medium (see above}.  Confidence in the
database for 1,2,4-TMB is low to medium as the database includes acute, short-term,
subchronic, and developmental toxicity studies in rats and mice.  The database lacks a
multigenerational reproductive study, and the studies supporting the critical effect
predominately come from the same research institute.  Overall confidence in the RfD  for
1,2,4-TMB is low due to uncertainties surrounding the application of the available PBPK
model for the purposes of a route-to-route extrapolation.

2.2.3. Uncertainties in the Derivation of the RfD for 1,2,4-TMB

   As the oral RfD for 1,2,4-TMB was based on a route-to-route extrapolation in order to
determine the oral dose that would result in the same effect as inhalation  exposure
(decreased pain sensitivity; Korsak and Rydzynski (1996)). the uncertainties regarding this
derivation are the same as for the RfC for 1,2,4-TMB (see Section  2.1.4), with the exception
of the uncertainty surrounding the route-to-route extrapolation.  The model used to
perform this route-to-route extrapolation is a well-characterized model deemed
appropriate for the purposes of the Toxicological Review. One source of uncertainty
regarding the route-to-route extrapolation is the assumption of 100% bioavailability, that
is,100% of the ingested 1,2,4-TMB would be absorbed and pass through the liver.  If not all
of the compound is bioavailable, a lower blood concentration would be expected compared
to the current estimate, and thus,  a higher RfD would be calculated.

2.2.4. Methods of analysis for RfD derivation for 1,3,5-TMB

   The available oral database is  inadequate to derive an RfD for 1,3,5-TMB. No chronic,
subchronic, or short-term oral exposure studies were found in the literature. However, as
outlined in RfC Derivation for 1,3,5-TMB, the toxicokinetic and toxicological similarities
between 1,3,5-TMB and 1,2,4-TMB support adopting the  RfC for 1,2,4-TMB as the RfC
1,3,5-TMB. These considerations  also apply to the oral reference value, thus the RfD for
1,2,4-TMB was adopted for 1,3,5-TMB.  1,3,5-TMB is observed to  elicit neurotoxic effects in
rats in acute and short-term studies, and therefore the selected critical effect for 1,2,4-TMB,
altered CNS function, is relevant to observed 1,3,5-TMB-induced toxicity.  Similarities in
blood:air and tissue:air partition coefficients and absorption into the bloodstream between
the two isomers support the conclusion that internal blood dose metrics for 1,3,5-TMB
would be similar as those calculated for 1,2,4-TMB using the available PBPK model. Also,
the qualitative metabolic profiles  for the two isomers are similar, with dimethylbenzyl
DRAFT - DO NOT CITE OR QUOTE                                                       i

-------
hippuric acids being the major terminal metabolite for both isomers, so that first-pass
metabolism through the liver is not expected to differ greatly between 1,2,4-TMB and
1,3,5-TMB.
   Therefore, given the above similarities in toxicokinetics and toxicity, the RfD derived for
1,2,4-TMB: 6 x 10-3 mg/kg-day was adopted for the RfD for 1,3,5-TMB.
    As noted previously, a confidence level of high, medium, or low is assigned to the study
used to derive the RfD, the overall database, and the RfD itself, as described in EPA (1994).
Section 4.3.9.2. The chronic RfD of 6 x 10-3 mg/kg-day derived for 1,2,4-TMB was adopted
as the RfD for 1,3,5-TMB based on the conclusion that the two isomers were sufficiently
similar regarding chemical properties, kinetics, and toxicity.  Thus, confidence in the study
from which the critical effect was identified, Korsak and Rydzynski (1996) is medium (see
above}.  Confidence in the database is low to medium as the database includes acute, short-
term, and developmental toxicity studies in rats and mice. The database lacks a
multigenerational reproductive study, and the studies supporting the critical effect
predominately come from the same research institute. Overall confidence in the RfD for
1,3,5-TMB is low due to uncertainties surrounding the adoption of the RfD derived for
1,2,4-TMB as the RfD for 1,3,5-TMB.

2.2.5. Uncertainties in the Derivation of the RfD for 1,3,5-TMB

   The uncertainties regarding adopting the RfD for 1,2,4-TMB as the RfD for 1,3,5-TMB
encompass previous areas of uncertainty  involved in the derivation of the RfC for 1,3,5-
TMB and the RfD for 1,2,4-TMB (see Sections 2.1.7 and 2.2.4}. There does exist uncertainty
regarding this adoption. However, as discussed above in Section 2.1.7, both isomers share
multiple commonalities and similarities regarding their toxicokinetic and toxicological
properties that support adopting one isomer's value for the other. Additionally, as the RfD
derivation for 1,2,4-TMB was based on a route-to-route extrapolation, the uncertainties in
that toxicity value's derivation (see Section 2.2.4} apply to the derivation of the RfD for
1,3,5-TMB.


2.3. Cancer Assessment

   Under the U.S. EPA Guidelines for Carcinogen Risk Assessment (2005aj. the database for
1,2,4-TMB and 1,3,5-TMB  provides "inadequate information to assess carcinogenic
potential". This characterization is based on the limited and equivocal genotoxicity
findings, and the lack of data indicating carcinogenicity in experimental animal species.
DRAFT - DO NOT CITE OR QUOTE                                                       89

-------
Information available on which to base a cancer assessment is lacking, and thus, no cancer
risk value is derived.
DRAFT - DO NOT CITE OR QUOTE                                                       90

-------
3.  REFERENCES
ACGIH. (American Conference of Governmental Industrial Hygienists). (2002). Trimethyl benzene isomers. In
Documentation of the threshold limit values and biological exposure indices (7 ed.). Cincinnati, OH.

Andersson. K: Fuxe. K: Toftgard. R: Nilsen. OG: Eneroth. P: Gustafsson. JA. (1980). Toluene-induced activation of
certain hypothalamic and median eminence catecholamine nerve terminal systems of the male rat and its effects on
anterior pituitary hormone secretion. ToxicolLett 5: 393-398. http://dx.doi.org/10.1016/0378-4274(80)90021-1.

Andersson. K: Fuxe. K: Nilsen. OG: Toftgard. R: Eneroth. P: Gustafsson. JA. (1981). Production of discrete
changes in dopamine and noradrenaline levels and turnover in various parts of the rat brain following exposure to
xylene, ortho-, meta-, and para-xylene, and ethylbenzene. Toxicol Appl Pharmacol 60: 535-548.

Andersson. K: Nilsen. OG: Toftgard. R: Eneroth. P: Gustafsson. JA: Battistini. N: Agnati. LF. (1983). Increased
amine turnover in several hypothalamic noradrenaline nerve exposure to various concentrations of toluene.
Neurotoxicology 4: 43-55.

Balster. RL. (1998). Neural basis of inhalant abuse. Drug Alcohol Depend 51: 207-214.

Battig. K: Grandjeaa E: Turrian. V. (1956). Health damage after continuous exposure to trimethyl benzene in a
painting workshop. Soz Praventivmed 1: 389-403. http://dx.doi.org/10.1007/BF02031676.

Billionnet C: Gay. E: Kirchner. S: Leynaert B: Annesi-Maesano. I. (2011). Quantitative assessments of indoor air
pollution and respiratory health in a population-based sample of French dwellings. Environ Res 111: 425-434.
http://dx.doi.0rg/10.1016/j.envres.2011.02.008.

Bowen. SE: Batis. JC: Paez-Martinez. N: Cruz. SL. (2006). The last decade of solvent research in animal models of
abuse: Mechanistic and behavioral studies. Neurotoxicol Teratol 28: 636-647.
http://dx.doi.0rg/10.1016/j.ntt.2006.09.005.

Braes. PU: Gregory. P: Jackson. DM. (1984). Passive avoidance in rats: Disruption by dopamine applied to the
nucleus accumbens. Psychopharmacology 83:  70-75.

CDC. (Centers for Disease Control and Prevention). (2004). The health consequences of smoking: A report of the
Surgeon General. Washington, DC: U.S. Department of Health and Human Services.
http://www.surgeongeneral.gov/library/smokingconsequences/.

Chen. R: Dick. F: Seaton. A. (1999). Health effects of solvent exposure among dockyard painters: Mortality and
neuropsychological symptoms. Occup EnvironMed 56: 383-387. http://dx.doi.0rg/10.1136/oem.56.6.383.

Chou. CC: Riviere. JE: Monteiro-Riviere. NA. (2003). The cytotoxicity of jet fuel aromatic hydrocarbons and dose-
related interleukin-8 release from human epidermal keratinocytes. Arch Toxicol 77: 384-391.
http://dx.doi.org/10.1007/s00204-003-0461-z.

Dahl AR: Damon. EG: Mauderly. JL: Rothenberg. SJ: Seiler. FA: McClellan. RO.  (1988). Uptake  of 19
hydrocarbon vapors inhaled by F344 rats. Fundam Appl Toxicol 10: 262-269. http://dx.doi.org/10.1016/0272-
0590(88)90310-7.

Eide. I: Zahlsen. K. (1996). Inhalation experiments with mixtures of hydrocarbons. Experimental design, statistics
and interpretation of kinetics and possible interactions. Arch Toxicol 70: 397-404.
http://dx.doi.org/10.1007/s002040050291.


DRAFT - DO NOT CITE OR QUOTE                                                                    91

-------
Gage. JC. (1970). The subacute inhalation toxicity of 109 industrial chemicals. Br J Ind Med 27: 1-18.
http://dx.doi.org/10.1136/oem.27.Ll.

Gaschen. A: Lang. D: Kalberer. M: Savi M: Geiser. T: Gazdhar. A: Lehr. CM: Bur. M: Dommen. J: Baltensperger.
U: Geiser. M. (2010). Cellular responses after exposure of lung cell cultures to secondary organic aerosol particles.
Environ Sci Technol 44: 1424-1430. http://dx.doi.org/10.1021/es902261m.

Ginsberg. G: W Jr. S: Bruckner. J: Sonawane. B. (2004). Incorporating children's toxicokinetics into a risk
framework. Environ Health Perspect 112: 272-283. http://dx.doi.org/10.1289/ehp.6013.

Gralewicz.  S: Wiaderna. D: Tomas. T: Rydzyriski. K. (1997a). Behavioral changes following 4-week inhalation
exposure to pseudocumene (1,2,4-trimethylbenzene) in the rat. Neurotoxicol Teratol 19: 327-333.
http://dx.doi.org/10.1016/S0892-0362(97)00001-9.

Gralewicz.  S: Wiaderna. D: Tomas. T. (1997b). Retardation of the age-related increase in spontaneous cortical
spike-wave discharges (SWD) in rats after a 28-day inhalation (SWD) in rats after a 28-day inhalation exposure to
an industrial solvent, pseudocumene (1,2,4-trimethylbenzene). Int J Occup Med Environ Health 10: 213-222.

Gralewicz.  S: Wiaderna. D. (2001). Behavioral effects following subacute inhalation exposure to m-xylene or
trimethylbenzene in the rat: A comparative study. Neurotoxicology 22: 79-89. http://dx.doi.org/10.1016/S0161-
813X(00)00003-6.

Guvatt GH: Oxman. AD: Vist GE: Kunz. R: Falck-Ytter. Y: Alonso-Coello. P: Schiinemann. HJ. (2008a).
GRADE: An emerging consensus on rating quality of evidence and strength of recommendations. BMJ 336: 924-
926. http://dx.doi.org/10.1136/bmj.39489.470347.AD.

Guvatt. GH: Oxman. AD: Kunz. R: Vist. GE: Falck-Ytter. Y: Schiinemann. HJ. (2008b). GRADE: What is "quality
of evidence" and why is it important to clinicians. BMJ 336: 995-998.
http://dx.doi.org/10.1136/bmj.39490.551019.BE.

Henderson. RF. (2001). Aromatic hydrocarbons: Benzene  and other alkylbenzenes. 4: 231-301.

Hill. AB. (1965). The environment and disease: Association or causation? Proc R Soc Med 58: 295-300.

Hillefors-Berglund. M: Liu. Y: von Euler. G. (1995). Persistent, specific and dose-dependent effects of toluene
exposure on dopamine D2 agonist binding in the rat caudate-putamen. Toxicology 100: 185-194.
http://dx.doi.org/10.1016/0300-483X(95)03084-S.

Hissink. AM: Kriise. J: Kulig. BM: Verwei M: Muijser. H: Salmon. F: Leenheers. LH: Owen. DE: Lammers. JH:
Freidig. AP: McKee. RH. (2007). Model studies for evaluating the neurobehavioral effects of complex hydrocarbon
solvents III. PBPK modeling of white spirit constituents as a tool for integrating animal and human test data.
Neurotoxicology 28: 751-760. http://dx.doi.0rg/10.1016/j.neuro.2007.03.005.

Huo. JZ: Aldous. S: Campbell. K: Davies. N. (1989). Distribution and metabolism of 1,2,4-trimethylbenzene
(pseudocumene) in the rat. Xenobiotica 19: 161-170. http://dx.doi.org/10.3109/00498258909034688.

IARC. (International Agency  for Research on Cancer). (2006). Preamble to the IARC monographs. Lyon, France.
http://monographs.iarc.fr/ENG/Preamble/.

Jacksoa DM: Westlind-Danielssoa A. (1994). Dopamine receptors: Molecular biology, biochemistry and
behavioural aspects. Pharmacol Ther64: 291-370. http://dx.doi.org/10.1016/0163-7258(94)90041-8.

Janik-Spiechowicz. E: Wvszynska. K: Dziubaltowska. E. (1998). Genotoxicity evaluation of trimethylbenzenes.
Mutat Res Genet Toxicol Environ Mutagen 412: 299-305. http://dx.doi.org/10.1016/81383-5718(97)00202-7.

Jarnberg. J: Johanson. G. (1995). Liquid/air partition coefficients of the trimethylbenzenes. Toxicol Ind Health 11:
81-88. http://dx.doi.org/10.1177/074823379501100107.

DRAFT - DO NOT CITE OR QUOTE                                                                     92

-------
Jarnberg. J: Johanson. G: Lof. A. (1996). Toxicokinetics of inhaled trimethylbenzenes in man. Toxicol Appl
Pharmacol 140: 281-288. http://dx.doi.org/10.1006/taap.1996.0223.

Jarnberg. J: Johanson. G: Lof. A: Stahlbom. B. (1997a). Inhalation toxicokinetics of 1,2,4-trimethylbenzene in
volunteers: Comparison between exposure to white spirit and 1,2,4-trimethylbenzene alone. Sci Total Environ 199:
65-71. http://dx.doi.org/10.1016/S0048-9697(97)05482-X.

Jarnberg. J: Stahlbon. B: Johanson. G: Lof. A. (1997b). Urinary excretion of dimethylhippuric acids in humans after
exposure to trimethylbenzenes. Int Arch Occup Environ Health 69: 491-497.
http://dx.doi.org/10.1007/s004200050179.

Jones. K: Meldrum M: Baird. E: Cottrell. S: Kaur. P: Plant. N: Dyne. D: Cocker. J. (2006). Biological monitoring
for trimethylbenzene exposure: A human volunteer study and a practical example in the workplace. Ann Occup Hyg
50: 593-598. http://dx.doi.org/10.1093/annhyg/mel016.

Korsak. Z: Swiercz. R: Rydzyfiski. K. (1995). Toxic effects of acute inhalation exposure to 1,2,4-trimethylbenzene
(pseudocumene) in experimental animals. Int J Occup Med Environ Health 8:331-337.

Korsak. Z: Rydzyfiski. K. (1996). Neurotoxic effects of acute and subchronic inhalation exposure to
trimethylbenzene isomers (pseudocumene, mesitylene,  hemimellitene) in rats. Int J Occup Med Environ Health 9:
341-349.

Korsak. Z: Rydzynski. K: Jajte. J. (1997). Respiratory irritative effects of trimethylbenzenes: An experimental
animal study. Int J Occup Med Environ Health 10: 303-311.

Korsak. Z: Stetkiewicz. J: Majcherek. W:  Stetkiewicz. I: Jaite. J: Rydzynski. K. (2000). Sub-chronic inhalation
toxicity of 1,2,4-trimethylbenzene (pseudocumene) in rats. Int J Occup Med Environ Health 13: 155-164.

Kyrklund. T. (1992). The use of experimental studies to reveal suspected neurotoxic chemicals as occupational
hazards: Acute and chronic exposures to organic solvents. Am J Ind Med 21: 15-24.
http://dx.doi.org/10.1002/ajim.4700210105.

Lammers. JH: Emmea HH: Muijser. H: Hoogendijk. EM: McKee. RH: Owen. DE: Kulig. BM. (2007). Model
studies for evaluating the neurobehavioral effects of complex hydrocarbon solvents II. Neurobehavioral effects of
white spirit in rat and human. Neurotoxicology 28: 736-750. http://dx.doi.0rg/10.1016/j.neuro.2007.03.003.

Maltoni. C: Ciliberti. A: Pinto. C: Soffritti M: Belpoggi F: Menarini. L. (1997). Results of long-term experimental
carcinogenicity studies of the effects of gasoline, correlated fuels, and major gasoline aromatics on rats. Ann N Y
Acad Sci 837: 15-52. http://dx.doi.0rg/10.llll/j.1749-6632.1997.tb56863.x.

McKee. RH: Lammers. JH: Muijser. H: Owen. DE: Kulig. BM. (2010). Neurobehavioral effects of acute exposure
to aromatic hydrocarbons. Int J Toxicol 29: 277-290. http://dx.doi.org/10.1177/1091581810365089.

Meulenberg. C: Vijverberg. H. (2000). Empirical relations predicting human and rat tissue: Air partition coefficients
of volatile organic compounds. Toxicol Appl Pharmacol 165: 206-216. http://dx.doi.org/10.1006/taap.2000.8929.

Mikulski. PI:  Wiglusz. R. (1975). The comparative metabolism of mesitylene, pseudocumene, and hemimellitene in
rats.  Toxicol Appl Pharmacol 31: 21-31. http://dx.doi.org/10.1016/0041-008X(75)90048-4.

MOE. (Ontario Ministry of the Environment). (2006). Rationale for the development of Ontario air standards for
trimethylbenzenes: 1,2,3-Trimethylbenzene. Ontario, Canada.

Mogel. I: Baumann. S: Bohme. A: Kohaida. T: von Bergen. M: Simon. JC: Lehmann. I. (2011). The aromatic
volatile organic compounds toluene, benzene and styrene induce COX-2 and prostaglandins in human lung epithelial
cells via oxidative stress and p38 MAPK activation. Toxicology 289: 28-37.
http://dx.doi.0rg/10.1016/j.tox.2011.07.006.


DRAFT - DO NOT CITE OR QUOTE                                                                     93

-------
Myhre. O: Vestad. TA: Sagstuen. E: Aarnes. H: Fonnum. F. (2000). The effects of aliphatic (n-nonane), naphtenic
(1,2,4-trimethylcyclohexane), and aromatic (1,2,4-trimethylbenzene) hydrocarbons on respiratory burst in human
neutrophil granulocytes. Toxicol Appl Pharmacol 167: 222-230. http://dx.doi.org/10.1006/taap.2000.9008.

Myhre. O: Fonnum F. (2001). The effect of aliphatic, naphthenic, and aromatic hydrocarbons on production of
reactive oxygen species and reactive nitrogen species in rat brain synaptosome fraction: the involvement of calcium,
nitric oxide synthase, mitochondria, and phospholipase A. Biochem Pharmacol 62: 119-128.
http://dx.doi.org/10.1016/80006-2952(01)00652-9.

NIOSH. (National Institute for Occupational Safety and Health). (1992). NIOSH recommendations for occupational
safety and health: Compendium of policy documents and statements. (92-100). Cincinnati, OH: U.S. Department of
Health and Human Services, Public Health Service, Centers for Disease Control, National Institute for Occupational
Safety and Health. http://www.cdc.gov/Niosh/92-100.html.

Norseth. T: Waage. J: Dale. I. (1991). Acute effects and exposure to organic compounds in road maintenance
workers exposed to asphalt. Am J Ind Med 20: 737-744. http://dx.doi.org/10.1002/ajim.4700200604.

Ramaiah. SK. (2007). A lexicologist guide to the diagnostic interpretation of hepatic biochemical parameters. Food
Chem Toxicol 45: 1551-1557. http://dx.doi.0rg/10.1016/j.fct.2007.06.007.

Rea. TM: Nash. JF: Zabik. JE: Born. GS: Kessler. WV. (1984). Effects of toluene inhalation on brain biogenic
amines in the rat. Toxicology 31: 143-150. http://dx.doi.org/10.1016/0300-483X(84)90006-4.

Rothman. KJ: Greenland.  S. (1998). Modern epidemiology (2nd ed.). Philadelphia, PA: Lippincott, Williams, &
Wilkins.

Saillenfait AM: Gallissot F: Sabate. JP: Morel  G. (2005). Developmental toxicity of two trimethylbenzene
isomers, mesitylene and pseudocumene, in rats following inhalation exposure. Food Chem Toxicol 43: 1055-1063.
http://dx.doi.0rg/10.1016/j.fct.2005.02.008.

Snead. OC. III. (1995). Basic mechanisms of generalized absence seizures. AnnNeurol 37: 146-157.
http://dx.doi.org/10.1002/ana.410370204.

Swiercz. R: Rydzyfiski. K: Wasowicz. W: Maicherek. W: Wesolowski. W. (2002). Toxicokinetics and metabolism
of pseudocumene (1,2,4-trimethylbenzene) after inhalation exposure in rats. Int J Occup Med Environ Health 15:
37-42.

Swiercz. R: Wiaderna. D:  Wasowicz. W: Rydzynskj K. (2003). Pseudocumene in brain, liver,  lung and blood of rats
after single and repeated inhalation exposure. Int J Occup Med Environ Health 16: 61-66.

Swiercz. R: Wasowicz. W: Maicherek. W. (2006). Mesitylene (1,3,5-trimethylbenzene) in the liver, lung, kidney,
and blood and 3,5-dimethylbenzoic acid in the liver, lung, kidney and urine of rats after single and repeated
inhalation exposure to mesitylene. Pol J Environ Stud 15: 485-492.

Tomas. T: Lutz. P: Wiaderna. D. (1999a). Changes in electrocortical arousal following acute trimethylbenzene
administration in rats. Int J Occup Med Environ Health 12: 67-78.

Tomas. T: Swiercz. R: Wiaderna. D: Gralewicz.  S. (1999b). Effects of acute exposure to aromatic hydrocarbons C 9
on locomotor activity in rats. Trimethylbenzene isomers. Int J Occup Med Environ Health 12: 331-343.

Tomas. T: Wiaderna. D: Swiercz. R. (1999c). Neurotoxicity assessment of selected organic solvents based on
spontaneous and evoked cortical and hippocampal activity in rats. Int J Occup Med Environ Health 12: 73-84.

Tsuiimoto. Y: Noda. T: Shimizu. M: Moriwakj H: Tanaka. M. (2000). Identification of the dimethylbenzyl
mercapturic acid in urine of rats administered with 1,2,4-trimethylbenzene. Chemosphere 40: 893-896.
http://dx.doi.org/10.1016/S0045-6535(99)00467-l.


DRAFT - DO NOT CITE  OR QUOTE                                                                     94

-------
Tsuiimoto. Y: Warashina. M: Nam. VD: Noda. T: Shimizu. M: Yamaguchi. Y: Moriwaki. H: Morimoto. T:
Kakiuchi. K: Maeda. Y: Tanaka. M. (2005). Determination of urinary phenolic metabolites from rats treated with
1,2,3-and 1,3,5-trimethylbenzenes. JOccup Health 47: 337-339.

U.S. EPA. (U.S. Environmental Protection Agency). (1986a). Guidelines for mutagenicity risk assessment [EPA
Report]. (EPA/630/R-98/003). Washington, DC. http://www.epa.gov/iris/backgrd.html.

U.S. EPA. (U.S. Environmental Protection Agency). (1986b). Guidelines for the health risk assessment of chemical
mixtures [EPA Report]. (EPA/630/R-98/002). Washington, DC.

U.S. EPA. (U.S. Environmental Protection Agency). (1991). Guidelines for developmental toxicity risk assessment
[EPA Report]. (EPA/600/FR-91/001). Washington, DC: U.S. Environmental Protection Agency, Risk Assessment
Forum, http://www.epa.gov/iris/backgrd.html.

U.S. EPA. (U.S. Environmental Protection Agency). (1994). Methods for derivation of inhalation reference
concentrations and application of inhalation dosimetry. (EPA/600/8-90/066F). Research Triangle Park, NC: U.S.
Environmental Protection Agency, Office of Research and Development, Office of Health and Environmental
Assessment, Environmental Criteria and Assessment Office.
http://cfpub.epa. gov/ncea/cfm/recordisplay.cfm?deid=71993.

U.S. EPA. (U.S. Environmental Protection Agency). (1996). Guidelines for reproductive toxicity risk assessment
[EPA Report]. (EPA/630/R-96/009). Washington, DC: U.S. Environmental Protection Agency, Risk Assessment
Forum. http://www.epa.gov/raf/publications/pdfs/REPRO51 .PDF.

U.S. EPA. (U.S. Environmental Protection Agency). (1998). Guidelines for neurotoxicity risk assessment.
(EPA/630/R-95/001F). Washington, DC: U.S. Environmental Protection Agency, Risk Assessment Forum.
http://www.epa.gov/raf/publications/pdfs/NEUROTOX.PDF.

U.S. EPA. (U.S. Environmental Protection Agency). (2000a). Benchmark dose technical guidance document
[external review draft]. (EPA/630/R-00/001). Washington, DC: U.S. Environmental Protection Agency, Risk
Assessment Forum, http://www.epa.gov/raf/publications^enchmark-dose-doc-draft.htm.

U.S. EPA. (U.S. Environmental Protection Agency). (2000b). Supplementary guidance for conducting health risk
assessment of chemical mixtures. (EPA/630/R-00/002). Washington, DC: U.S. Environmental Protection Agency,
Risk Assessment Forum. http://cfpub.epa.gov/ncea/cfm/recordisplay.cfm?deid=20533.

U.S. EPA. (U.S. Environmental Protection Agency). (2002). A review of the reference dose and reference
concentration processes. (EPA/630/P-02/002F). Washington, DC.
http://cfpub.epa.gov/ncea/cfm/recordisplay.cfm?deid=51717.

U.S. EPA. (U.S. Environmental Protection Agency). (2005a). Guidelines for carcinogen risk assessment.
(EPA/630/P-03/001F). Washington, DC. http://www.epa.gov/cancerguidelines/.

U.S. EPA. (U.S. Environmental Protection Agency). (2005b). Supplemental guidance for assessing susceptibility
from early-life exposure to carcinogens. (EPA/630/R-03/003F). Washington, DC: U.S. Environmental Protection
Agency, Risk Assessment Forum, http://www.epa.gov/cancerguidelines/guidelines-carcinogen-supplement.htm.

U.S. EPA. (U.S. Environmental Protection Agency). (2006). Approaches for the application of physiologically
based pharmacokinetic (PBPK) models and supporting data in risk assessment (final report). (EPA/600/R-05/043F).
Washington, DC: U.S. Environmental Protection Agency, Office of Research and Development.

U.S. EPA. (U.S. Environmental Protection Agency). (2007). Acute exposure guideline levels (AEGLs) for 1,3,5-
trimethylbenzene (CAS reg. no. 108-67-8), 1,2,4-trimethylbenzene (CAS reg. no. 95-63-6), 1,2,3-trimethylbenzene
(CAS reg. no. 526-73-8). Washington, DC.
http://www.epa.gov/opptintr/aegl/pubs/123_%20124_%20135_trimethylbenzenes_%20interim_ll_2007.vl.pdf.
DRAFT - DO NOT CITE OR QUOTE                                                                    95

-------
U.S. EPA. (U.S. Environmental Protection Agency). (201 la). IRIS Process Retrieved August 24, 2011, from
http://epa.gov/iris/process.htm

U.S. EPA. (U.S. Environmental Protection Agency). (201 Ib). Recommended use of body weight 3/4 as the default
method in derivation of the oral reference dose. (EPA/100/R11/0001). Washington, DC.
http://www.epa.gov/raf/publications/interspecies-extrapolation.htm.

U.S. EPA. (U.S. Environmental Protection Agency). (201 Ic). Supplemental Material: Benchmark Dose Modeling
Results for the Derivation of Reference Values for 1,2,4- and 1,3,5-Trimethylbenzene (Science Consultation Draft
[Appendix C]). Washington, DC.

U.S. EPA. (U.S. Environmental Protection Agency). (20lid). Supplemental Material: Toxicological Information
(PBPK and TK) in Support of Hazard Identification and Dose-Response Analysis for 1,2,4- and 1,3,5-
Trimethylbenzene (Science Consulation Draft [Appendix A]). Washington, DC.

von Euler. G: Ogrea SO: Li. XM: Fuxe. K: Gustafsson. JA. (1993). Persistent effects of subchronic toluene
exposure on spatial learning and memory, dopamine-mediated locomotor activity and dopamine D2 agonist binding
in the rat. Toxicology 77: 223-232. http://dx.doi.org/10.1016/0300-483X(93)90162-L.

von Euler. G: Ogren. SO: Eneroth. P: Fuxe. K: Gustafsson. JA. (1994). Persistent effects of 80 ppm toluene on
dopamine-regulated locomotor activity and prolactin secretion in the male rat. Neurotoxicology 15: 621-624.

Warter. JM: Vergnes. M: Depaulis. A: Tranchant. C: Rumbach. L: Micheletti. G: Marescaux. C. (1988). Effects of
drugs affecting dopaminergic neurotransmission in rats with spontaneous petit mal-like seizures.
Neuropharmacology 27: 269-274. http://dx.doi.org/10.1016/0028-3908(88)90043-3.

Wiaderna. D: Gralewicz. S: Tomas. T. (1998). Behavioral changes following a four-week inhalation exposure to
hemimellitene (1,2,3-trimethylbenzene) in rats. Int J Occup Med Environ Health 11: 319-334.

Wiaderna. D: Gralewicz. S: Tomas. T. (2002). Assessment of long-term neurotoxic effects of exposure to
mesitylene (1,3,5-trimethylbenzene) based on the analysis of selected behavioral responses. Int J Occup Med
Environ Health 15: 385-392.

Wiglusz. R: Kienitz. M: Delag.  G: Galuszko. E: Mikulski. P. (1975a).  Peripheral blood of mesitylene vapour treated
rats. Bull Inst Marit Trop Med Gdynia 26: 315-321.

Wiglusz. R: Delag. G: Mikulski. P. (1975b). Serum enzymes activity of mesitylene vapour treated rats. Bull Inst
Marit Trop Med Gdynia 26: 303-313.

Wiglusz. R. (1979). The effect of 1, 3, 5-trimethylbenzene inhalation exposure on the glucuronic acid pathway and
activity of some xenobiotic-metabolizing enzymes. Bull Inst Marit Trop Med Gdynia 30: 189-196.
DRAFT - DO NOT CITE OR QUOTE                                                                     96

-------
APPENDICES
Appendix A: Toxicological Information in Support of Hazard Identification
   and Dose-Response Analysis for 1,2,4- and 1,3,5-Trimethylbenzene

Appendix B: Benchmark Dose Modeling Results for the Derivation of
   Reference Values for 1,2,4- and 1,3,5-Trimethylbenzene

Appendix C: Summary of External Peer Review and Public Comments and
   Disposition
DRAFT - DO NOT CITE OR QUOTE                                            97

-------