r/EPA
          United States
          Environmental Protection
          Agency
           Office of Water
           4301
EPA-820-B95-007
March 1995
Great Lakes Water
Quality Initiative
Technical Support
Document for
Human Health
Criteria and Values

-------
                             DISCLAIMER

  This document  has been reviewed by the Health and Ecological
  Criteria Division,  Office of Science and Technology,  U.S.
  Environmental  Protection Agency, and approved for publication
  as a support document for the Great Lakes Water  Quality
  Initiative.  Mention of trade names and commercial products
  does not constitute endorsement of their use.
                        AVAILABILITY NOTICE

  This document  is  available for a fee upon written request or
  telephone  call to:

            National Technical Information Center (NTIS)
                    U.S. Department of Commerce
                        5285 Port Royal Road
                       Springfield, VA 22161
                            (800) 553-6847
                          (703) 487-4650
1                 NTIS Document Number:  PB95187316
N
                                 or

      Education Resources Information  Center/Clearinghouse for
  Science,  Mathematics, and Environmental Education (ERIC/CSMEE)
                    1200 Chambers Road, Room 310
                        Columbus,  OH  43212
                            (800) 276-0462
                            (614) 292-6717
                         ERIC Number:  D051
                                         U.S. Environmental Protection Agency
                                         Region 5, Library (PL-12J)
                                         77 West Jackson Boulevard, 12tn Hoor
                                         Chicago, IL 60604-3590

-------
         GREAT LAKES WATER QUALITY INITIATIVE TECHNICAL
                SUPPORT DOCUMENT FOR HUMAN HEALTH
I.  INTRODUCTION  	   1
     A.  Goal	   1
     B.  Level of Protection  	   1
     C.  Two Tiered Approach	   4
     D.  Technical Background 	   4

II.  MINIMUM DATA REQUIREMENTS	   6
     A.  Carcinogens  	   6
          1. Weight of Evidence	   6
          2. Appropriate Study Design and Data Development  .   8
          3. Borderline Conditions  	  12
     B.  Noncarcinogens	14
          1.  Appropriate Study Design  	  15
               a.  Acute Toxicity	15
               b.  14 Day or 28 Day Repeated Dose Toxicity  .  16
               c.  Subchronic and Chronic Toxicity  	  18
               d.  Reproductive and Developmental Toxicity  .  19
     C.  Tier Designation	21
          1. Carcinogens	21
          2. Noncarcinogens	22

III.  PRINCIPLES FOR CRITERIA DEVELOPMENT	24
     A.  General	24
     B.  Carcinogens	25
          1. Mechanism/Mode of Action	25
          2. Data Review	27
          3. Model	28
               a. Nonthreshold Approach 	  28
               b. Threshold Approach  	  29
          4. Lifespan Adjustment	30
          5. Species Scaling	30
     C.  Noncarcinogens	31
          1. Mechanism	31
          2. Data Review	33
          3. Uncertainty Factors  	  34
               a.  Intraspecies uncertainty factor  	  35
               b.  Interspecies uncertainty factor  	  35
               c.  Subchronic to chronic uncertainty factor .  35
               d4.  Less than subchronic duration uncertainty
                    factor	35
               e.  LOAEL to NOAEL uncertainty factor  ....  35
               f.  Limited database uncertainty factor  ...  36
     D.  Exposure Assumptions	36
          1.  Body Weight	36
          2.  Duration of Exposure	39
               a.  Population Mobility	39
               b.  Life Expectancy	39
          3. Incidental Exposure  .  . •	40

-------
          4.  Drinking Water	41
          5.  Fish Consumption	42
          6.  Relative Source Contribution  	  44

IV.  CRITERIA CALCULATIONS	46
     A.  Standard Exposure Assumptions  	  46
     B.  Carcinogens	46
     C.  Noncarcinogens	47

APPENDIX A	Al
                                11

-------
I.  INTRODUCTION

A.  Goal

     The goal of the human health criteria and values for the
Great Lakes and their tributaries is the protection - of humans
from unacceptable exposure to toxicants from consumption of
contaminated fish, drinking water and water related to
recreational activities.  Emphasis is on protection of the
individual in evaluating toxicity information and its application
in the derivation of criteria and values.  Exposure assumptions
follow trends and activities for the general population as a
region and also attempt to protect sensitive subpopulations.
Based on differences in behavior, there may be some individuals
who receive a greater level of protection or a lesser level of
protection via these procedures.

B.  Level of Protection

     Numeric criteria or interpretations of narrative criteria
developed for human health generally restrict chemical carcinogen
exposure in a target population to levels estimated to result in
a lifetime incremental risk of no greater than 1 in 100,000 of
developing cancer.  The procedure generally used to estimate the
risk level leads to the development of a plausible upper limit of
the risk.  This means that the actual risk is unknown, is
unlikely to exceed 10"5,  and may even be as low as zero.
     The selection of an "acceptable" target risk level does not
turn on scientific analysis, but on more subjective "risk
management" considerations.  Differences in perception of risk,
opinions as to benefit versus risk reduction costs, as well as
distinctions between risks that are considered voluntary or
involuntary, all could play a meaningful role in determining risk
acceptability.  For this initiative, a 10-5 cancer risk level has
been selected as acceptable.  This is consistent with the
existing practice of the eight Great Lakes states, and therefore
is consistent with existing risk management policy in these
states.  It is a.lso a risk level that EPA has found acceptable in
its review of state criteria in the past, and which EPA itself
has used as a basis for certain of its regulations.  EPA notes
that States and Tribes are free to adopt a more stringent
approach than that contained in the final Guidance.  It is also
instructive to note that this level of risk of developing cancer
appears to be roughly comparable to that which exists for death
due to natural phenomena.  Table 1 represents data from a study
of everyday risks of death from several naturally occurring
incidents such as tornados, floods, lightning and animal bites or
stings.  When extrapolated to lifetime risks, we see that these
risks range from 1.4 in 100,000 for animal bites or stings to 4
in 100,000 for floods and tornadoes.  It is acknowledged that
risk of death (as described in Table 1) is not equatable with

-------
risk of cancer since many forms of cancer are now easily curable.
The comparison is made only for the purpose of illustrating the
potential background risk in the region.
     For noncarcinogens,  protection of human health is generally
centered on determining a level of daily exposure that is likely
to be without an appreciable risk of deleterious effects for a
lifetime.  The concept of acceptable exposure incorporates the
potential for long term exposure of sensitive individuals in a
population to an environmental contaminant without any
anticipated adverse health effects.

-------
                     TABLE  1.    EVERYDAY RISKS
4

• *•»*»•*•***••»»•*»•***»••
Motor vehicle accident
Falls
Drowning
Fires
Firearms
Electrocution
Tornadoes
Floods
Lightning
Animal bite or sting
^•^HHHHHHHHHfrtHHHHHHHHHHHHI
GENERAL
Manufacturing
Trade
Service and government
Transport and public utilities
Agriculture
Construction
Mining and quarrying
SPECIFIC
Coal mining (accidents)
Police duty
Railroad employment
Fire fighting
Time to Accumulate a 1 in
100 ,000 Risk of Death

*****
1 5 days
60 days
1 00 days
130 days
360 days
20 months
200 months
200 months
20 years
40 years

45 days
70 days
35 days
10 days
1 50 hours
140 hours
90 hours

.140 hours
15 days
15 days
110 hours
Average Annual
Risk per Capita

in the United State;
2x 10-4
6x 10'B
4 x 10-s
3 x 10'5
1 x 106
5 x 10-'
6x 10'7
6x10'7
5 x 10-7
2 x 10'7

8x10'5
5 x 10'6
1 x TO"4
4x ID"4
6x 10"4
6x 10"4
1 x 10'3

6x 10"4
2x 10"4
2x10-*
8x 10"4
Extrapolated to
Risk/Lifetime*

;
1.4x 10'2
4.2 x 10'3
2.8 x 10-3
2 x 10'3
7 x 10-4
3.5 x 10"4
4 x 10'6
4x 10'5
3.5 x 10'6
1.4x10-6

5.6 x 10'3
3.5 x 10'3
7 x 10'3
3 x 10'2
4x 10'2
4x 102
7 x 102

3 x 10'2
1.4x 10'2
1.4x 10'2
5.4 x 10 2
                        Some One in a Million Cancer Risks
Cosmic rays
Other radiation
Eating and drinking
Smoking
one transcontinental round trip by air
living 1.5 months in Colorado compared to New York
camping at 15,000 feet for 6 days compared to sea level
20 days of sea level natural background radiation
2.5 months in masonry rather than wood  building
1H of a chest x-ray using modern equipment
40 diet sodas (saccharin)
6 pounds of peanut  butter (aflataxia)
180 pints of milk (aflataxia)
200 gallons of drinking water from Miami or New Orleans
90 pounds of broiled steak (cancer risk only)
2 cigarettes
     Adapted from Crouch and Wilson (1962)

     Risk/Lifetime - 1 -(1-s)70

                                       3

-------
C.  Two Tiered Approach

     A two tiered approach is used to derive ambient
concentration levels protective of human health.  Both tiers rely
generally on the same standard procedure for data review and
criteria derivation.  The difference between the two focuses
heavily on the certainty with which one can predict a level of
risk or a level of safety for humans from the data available.
The more adequate the database to estimate actual human risk or
to establish no adverse effect levels, the greater the certainty
in the appropriateness of the criterion or value.  This level of
certainty depends heavily on the weight of experimental evidence
which includes factors such as:  the quantity of studies or size
of the experimental database available for review; the quality of
study design, its conduct and range of effects evaluated; the
potency or range and type of adverse effects observed and, the
appropriateness of this data in predicting human effects, i.e.
evaluation of effects in humans or in animal species biologically
similar to human.
     The greater the level of certainty in the database for
noncancer effects, generally the lower the need for adjustment of
the research findings to assure a level without appreciable risk.
The greater the weight of evidence for carcinogenicity, the
greater the strength in predicting cancer risk to humans.
Chemicals with databases providing a high level of certainty in
predicting a level of risk or safety for humans from adverse
health effects are suitable for Tier I numeric criteria
derivation.  Tier I criteria are conceptionally those criteria
where the probability of change is low.
     Chemicals with less extensive data or where the weight of
evidence toward predicting human health effects is less certain,
are subject to Tier II values.  Under Tier II, the probability of
future change is greater than for Tier I as demonstrated by the
extent, level of quality and/or weight of evidence or
conclusiveness of effects demonstrated by the database.  The
values derived via Tier II are more likely to change based on new
data and/or reinterpretation of effect or potency.

D.  Technical Background

     The process used to evaluate effects and in development of
criteria shall be based on currently acceptable scientific
methods and consider guidance offered by the various USEPA
methods.  Particular attention should be paid to RfD and cancer
risk estimation development contained within the Integrated Risk
Information System  (IRIS).
     To promote consistency with other USEPA guidance for
chemical management, it is important to review and strongly
consider the IRIS values for chemicals undergoing criteria or
values development whenever available.  Although consistency is
important, it is also important that the most current and
complete data should be used when generating criteria or values

-------
whether IRIS has considered this data or not.  Further, since
IRIS values are developed under guidance and through the
judgement of workgroups, the final values may not always be
arrived at consistently, i.e., duration of studies, selected,
uncertainty factors applied, basis for derivation of potency
slopes, etc., may differ between decisions.  If data used in
deriving Tier I criteria or Tier II values have not been
considered by the IRIS RfD or CRAVE workgroups, the appropriate
workgroup should be advised of the data.  In cases where IRIS
RfDs or potency slopes have not been developed consistent with
these procedures, it is suggested that the rationale for RfD or
potency slope development be evaluated and determination made
whether 1) justification is sufficient to support deviating from
these procedures, or 2) justification exists to deviate from IRIS
guidance.   When deviations from IRIS are contemplated, EPA
strongly urges States and Tribes to communicate these potential
changes to EPA, either through a Regional EPA Office or directly
to the EPA Reference Dose (RfD) and/or Cancer Risk Assessment
Verification Endeavor  (CRAVE) workgroups, as soon as possible.
This will help foster consistency between EPA and the States and
Tribes.  Additionally, when deviating from IRIS, States and
Tribes are encouraged to work with the Clearinghouse described in
Section II of the SID, to ensure that other States and Tribes are
aware of the deviations.
     Specific references which should be reviewed and evaluated
for greater details on the basic parameters of the criteria and
values derivation methodology are as follows:

National Cancer Institute (NCI).  1976.  Guidelines for
     Carcinogen Bioassay in Small Rodents, Technical Report
     Series No. 1, U.S. Department of Health, Education and
     Welfare, NCI-CG-TR-1.

Office of Science and Technology Policy  (OSTP).  1985.  Chemical
     Carcinogens; A Review of the Science and Its Associated
     Principles, Federal Register, Vol. 50, No. 50.  March 14,
     1985, 10371-10442.

Organization for' Economic Cooperation and Development (OECD).
     1987. Guidelines for Testing of Chemicals, Paris, France.

U.S. Environmental Protection Agency (EPA).  1989.  Risk
     Assessment Guidance for Superfund, Volume 1, Human Health
     Evaluation Manual (Part A) - Interim Final, Office of
     Emergency and Remedial Response, Washington, D.C.,
     EPA/540/1-89/002.

U.S. Environmental Protection Agency (EPA).  1980.  Water Quality
     Criteria Availability,  Appendix C Guidelines and Methodology
     Used in the Preparation of Health Effects Assessment
     Chapters of the Consent Decree Water Quality Criteria

-------
     Documents, Federal Register, Vol. 45, November 28, 1980,
     79347-79357.

U.S. Environmental Protection Agency  (EPA).  1985.  Toxic
     Substances Control Act Test Guidelines; Final Rules, Federal
     Register, Vol. 50, NO. 188.  September 27, 1985, 39421- .
     39425.

U.S. Environmental Protection Agency  (EPA).  1986.  Guidelines
     for Carcinogen Risk Assessment.  Federal Register, Vol. 51,
     No. 185.  September 24, 1986, 33992-34002.

U.S. Environmental Protection Agency  (EPA).  1986.  Guidelines
     for the Health Assessment of Suspect Developmental
     Toxicants, Federal Register, No. 51, No. 185.  September 24,
     1986  34028-34040.

This is by no means a complete list.  Other sources of
information and guidance may also be considered as appropriate.


II.  MINIMUM DATA REQUIREMENTS

A.  Carcinogens

1. Weight of Evidence

     Evidence of a chemical's possible carcinogenic effects in
humans shall be categorized according to the existing EPA weight
of evidence classification system, which is adapted from the
International Agency for Research on Cancer  (IARC).  The five
categories or groups are as follows:

     Human Carcinogen  (identified as Group A under existing
     classification scheme)
     "sufficient" evidence from epidemiologic studies to support
     a causal association between exposure to the chemical and
     cancer;

     Probable Human Carcinogen  (identified as Group B)
     "limited" evidence from epidemiologic studies with or
     without supporting animal data  (Group Bl); or,  "sufficient"
     evidence of carcinogenicity based on  animal studies, but for
     which there may be "inadequate  evidence" or "no data" from
     epidemiologic studies  (Group B2);

     Possible Human Carcinogen  (identified as Group C)
     "limited" evidence of  carcinogenicity in animals and the
     absence of data for humans;

     Not Classifiable as to Human Carcinogenicity  (identified as
     Group D)

-------
     "inadequate" evidence of carcinogenicity in humans and
     animals, or, for which no data are available; and

     Evidence of Noncarcinogenicity for Humans (identified as
     Group E)
     "no evidence" for carcinogenicity in at least two adequate
     animal tests in different species or in both adequate
     epidemiologic and
     animal studies.

The definitions of the EPA weight of evidence classifications are
as follows:

1.   Humans
     a.   Sufficient evidence - a causal association can be
          inferred between exposure to the chemical and human
          cancer.

     b.   Limited evidence - a causal interpretation is credible,
          but that alternative explanations, such as chance, bias
          or confounding could not adequately be excluded.

     c.   Inadequate evidence - there were few pertinent data,
          or, a causal interpretation is not credible from
          available studies since they did not exclude change,
          bias or confounding.

     d.   No evidence - no association was found between exposure
          and an increased risk of cancer in well-designed and
          well-conducted independent analytical epidemiologic
          studies.

2.   Animals
     a.   Sufficient evidence - an increased incidence of
          malignant or combined malignant and benign tumors:  1)
          in multiple species or strains, 2) in multiple
          experiments using different dosage levels and possible
          different routes of exposure; or 3) in a single
          experiment with a high incidence, unusual site or type
          of tumor, or early onset.

     b.   Limited evidence - data suggest a carcinogenic effect
          but are limited because:  1) the studies involve a
          single species, strain or experiment which does not
          demonstrate a high incidence, unusual site or type of
          tumor, or early onset; 2) the experiments used
          inadequate dosage levels, inadequate exposure duration,
          inadequate follow-up periods, poor survival, too few
          animals, or inadequate reporting; 3) an increase in
          benign tumor incidence only and no response in a
          variety of short-term tests for mutagenicity; or 4)
          tumor responses of marginal statistical significance

-------
          due to inadequate study design or reporting, or, in
          tissue known to have a high or variable background
          rate.

     c.   Inadequate evidence - because of major qualitative or
          quantitative limitations, the studies cannot be
          interpreted as showing either the presence or absence
          of a carcinogenic effect.

     d.   No evidence - no increased tumor incidence in at least
          two well-designed and well conducted animal studies in
          different species.

Further detail regarding this classification system for
categorizing weight of evidence for carcinogenicity may be found
in the EPA Guidelines for Carcinogen Risk Assessment  (EPA, 1986).


2. Appropriate Study Design and Data Development

     The following discussion summarizes the process for
evaluating evidence of carcinogenicity and outlines an approach
study design by which one may measure the quality and adequacy of
data development. When available, human epidemiologic data with
quantifiable exposure levels are preferred for evaluating a
chemical's carcinogenic potential over use of animal data alone.
Epidemiological studies can provide direct evidence of a
chemical's carcinogenicity in humans (OSTP, 1985).  The type of
epidemiologic study conducted indicates whether the study may be
useful in assessing carcinogenic risk to exposed humans
(analytical studies) or if it is merely hypothesis-generating and
inherently incapable of proving a causal association.  Case
reports, descriptive studies and ecological (correlational)
studies generally cannot establish whether risks are associated
with particular exposures.  Analytical studies can assess
carcinogenic risks to exposed humans, and can infer a casual
association (Mausner and Kramer, 1985; OSTP, 1985).  The two
general types of analytical studies are case-control and cohort.
In case-control studies, a group of diseased "case" individuals
is initially identified and matched with nondiseased  "controls".
Information on past exposure to reputed risk factors or causative
agents is then collected for both groups.  If the proportion of
cases with a certain exposure is significantly different than
that of controls., an association between exposure and disease may
be indicated.  A cohort study starts by identifying a group of
individuals with a particular exposure and a similar group of
unexposed persons and follows both groups over time to determine
subsequent health outcomes.  The rates of disease in  the exposed
and unexposed groups are then compared.  Cohort studies may be
based on current exposure and future health outcomes  (prospective
cohort study), or on past exposure information and disease
occurrence  (historical cohort study).  As with case-control

                                8

-------
studies, cohort studies that are well-designed, well-conducted,
and well-evaluated can test hypotheses and provide the basis for
causal inferences (OSTP, 1985; EPA, 1986). Factors such as proper
selection and characterization of exposed and control groups,
adequacy of duration and quality of follow-up, proper
identification and characterization of confounding factors and
bias,'appropriate consideration of latency effects, valid
ascertainment of. causes of morbidity and mortality, and the
ability to detect specific effects are all elements for
determining the adequacy of epidemiologic studies  (EPA, 1986) .
In interpreting a reported causal association, reference may be
made to the following criteria, as described by IARC  (1985), EPA
(1986),  and the Tripartite Working Group  (1985):

          There is no identifiable positive bias which could
          explain the association.

          The possibility of positive confounding factors has
          been considered and ruled out as explaining the
          association.

          The association is unlikely to be due to chance alone.

Although the weight of evidence increases with the number of
adequate studies, in some instances, a single epidemiologic study
may be strongly indicative of a cause-effect relationship  (IARC,
1985; EPA, 1986).  Confidence to infer a causal association is
increased by any of the following:  when several independent
studies are concordant in showing the association; when the
association is strong; when there is a dose-response relationship
when a reduction in exposure is followed by a reduction in the
incidence of cancer; when the effect is biologically plausible;
or when the effect is specific for a particular chemical.  When
epidemiological evidence based on analytical studies appears to
be significantly flawed, the evidence may then be downgraded to
being suggestive of an association based on scientific judgment.
This may still provide evidence that a causal interpretation is
credible, but that alternative explanations, such as chance, bias
or confounding factors, could not adequately be excluded.
     Epidemiological studies are inherently capable of detecting
only comparatively large increases in the relative risk of cancer
(EPA, 1986).  Other limitations of epidemiological studies
include the long latency of cancer, and the difficult task of
exposure assessment, including multiple exposures.  Therefore,
negative results from such studies do not verify that a
particular agent is noncarcinogenic in humans  (IARC, 1985; EPA,
1986; OSTP, 1985).
     Although epidemiologic studies are preferable for assessing
carcinogenic potential for humans, the relative paucity of such
data necessitates the use of animal data as a surrogate for
humans in most situations.  In the absence of adequate data on
humans,  it is biologically plausible and prudent to regard agents

-------
for which there is sufficient evidence of carcinogenicity in
experimental animals as if they present a carcinogenic risk to
humans (IARC, 1991).  The weight of evidence that an agent is
potentially carcinogenic in humans increases with:  a) the
increase in tissue sites affected; b)  the increase in number of
animal species, strains, sexes, doses and experiments showing a
carcinogenic response; c) the occurrence of clear-cut dose-
response relationships as well as a high level of statistical
significance of the increased tumor incidence in treated groups
as compared to controls; d) a dose related shortening of the
time-to-tumor occurrence or time to death with tumor; and e) a
dose-related increase in the proportion of tumors that are
malignant (EPA, 1986).
     The guidelines detailed by EPA (1985), OSTP  (1985) and NCI
(1976) for evaluating long-term carcinogenicity bioassays will be
utilized to determine the adequacy of design and the strength of
evidence provided by the study.  Specific study design elements
of these guidelines are synopsized as follows:

     Species used:  The most widely used and accepted test
     species is the rat.  NCI/NTP bioassays routinely use the
     Fischer inbred  (F344) strain of rat and the B6C3F1 hybrid
     mouse.   Hamsters have also been frequently used.  Other
     animal species and strains may also be appropriate
     surrogates to demonstrate a chemical's carcinogenic
     potential.

     Number of animals:  At least 100 rodents (50 of each sex)
     should be used at each dose level and concurrent control.

     Age at start:  Dosing of rodents should begin as soon as
     possible after weaning to allow for the long latency of
     cancer.  For rats, dosing ideally begins before the age of 6
     weeks and should not begin after 8 weeks of age.

     Survival:  All groups should have at least 50% survival at
     the time of termination.

     Concurrent control groups:  These should be untreated, sham
     treated, or, if a vehicle is used in administering the test
     substance, vehicle control groups.  The use of historical
     control data is desirable for assessing the significance of
     changes observed in exposed animals, but only if the strain
     of animals and laboratory conditions have not changed.  For
     the evaluation of rare tumors, even small tumor responses
     may be significant compared to historical data.  The review
     of tumor data at sites with high spontaneous background
     requires special consideration (OSTP, 1985).  For instance,
     a response that is significant with respect to the
     experimental control group may become questionable if the
     historical control data indicate that the experimental


                                10

-------
control group had an unusually low background incidence
(NTP, 1984).

Dose levels':  At least 3 dose levels are recommended in
addition to the concurrent control group, for the purpose of
risk assessment (OSTP, 1985; EPA, 1985).  For the purpose of
hazard assessment, detection of a carcinogenic response is
possible with one dose level, although 2 dose levels are
preferred and are necessary to demonstrate a dose-response
relationship.  The highest dose level should target the
maximum tolerated dose (MTD).  The MTD is the dose which,
when given for the duration of the chronic study, elicits
signs of minimal toxicity  (e.g., less than or equal to 10%
weight gain decrement) without substantially altering the
normal life span due to effects other than carcinogenicity.
The MTD is intended to provide an adequate statistical power
for the detection of carcinogenic activity.  While not an
ideal solution to the problem of low bioassay sensitivity,
use of the MTD is appropriate if it is properly determined
(OSTP, 1985; EPA,  1986).

Dosing route:  The test substance should be administered via
the oral,, dermal or inhalation route.

Dosing schedule:  The animals should ideally be dosed on a 7
day per week basis.  However, based primarily on practical
considerations, dosing on a 5 day per week basis is
acceptable.  Treatment preferably should be continued for
the major portion of the animal's lifespan.  This is at
least 18 months for mice and hamsters, and 24 months for
rats.

Data collection:  During the study, animals should be
monitored for body weight and food intake, as well as for
the onset and progression of all toxic effects.  Clinical
examinations, including hematology, biochemistry of blood,
urinalysis, and ophthalmological examination, should be
made.  Gross necropsy and histopathology should be performed
on all animals.  Specific requirements are too numerous to
list here, but may be reviewed via the EPA (1985) and NCI
(1976) guidelines.

All observed results should be evaluated by an appropriate
and generally accepted statistical method.  Evidence for
carcinogenic action should be based on the observation of
statistically significant tumor responses in specific organs
or tissues.  Appropriate statistical analysis should be
performed on data from long-term studies to help determine
whether the effects are treatment-related or possibly due to
chance.  These should at least include a statistical test
for trend, including appropriate correction for differences
in survival. . The weight to be given to the level of

                           11

-------
     statistical significance (the p-value)  and. to other
     available pieces of information is a matter of overall
     scientific judgment.  In a review of 25 NTP feeding studies
     as discussed by OSTP (1985),  a simple statistical rule was
     derived by Haseman which appeared to mimic the scientific
     judgment process used in those experiments.  "Regard as
     carcinogenic any chemical that produces a high dose increase
     in a common tumor that is statistically significant at the
     0.01 level or a high-dose increase in an uncommon tumor that
     is statistically significant at the 0.05 level.  The overall
     false positive rate associated with this rule was estimated
     to be no more than 7-8% for the NTP two-sex, two-species
     protocol".  A statistically significant excess of tumors of
     all types in the aggregate, in the absence of a
     statistically significant increase of any individual tumor
     type, should be regarded as minimal evidence of carcinogenic
     action unless there are persuasive reasons to the contrary
     (OSTP, 1985).

     These guidelines represent ideal parameters.  Studies will
not be expected to meet all of these desirable conditions in
order to be further considered for use in the process.  The
adequacy and appropriateness of all animal carcinogenicity
bioassays will be carefully considered.  It is crucial that
judgment of adequate testing be based on sound scientific
principles.  In general, it can be expected that most substances
tested for carcinogenicity have been reviewed by NCI/NTP, IARC,
and/or EPA.  Historically the evaluations by these agencies have
been sufficient for decision-making.  A thorough assessment of
the data should be performed regardless of the findings of those
independent agencies since these reviews might be dated in that
research data available subsequent to the date of review were not
considered by the reviewing group.  The overall  assessment of a
chemical's carcinogenic potential will depend on weight-of-
evidence based upon full consideration of all the evidence.  Also
see Section III. Principles for Criteria Development for a
discussion of Mechanism/Mode of Action and the use of
mutagenicity studies in determining carcinogenicity.


3. Borderline Conditions

     With regard to the overall database used in determining
carcinogenicity, a variety of studies may be encountered which
may be considered flawed or lacking in adequate design or
reporting.  Such studies may only be able to be utilized
anecdotally and only considered suggestive evidence of
carcinogenicity.  Examples of conditions meeting such a criteria
are:

1.   Borderline conditions of:
                                12

-------
     a.   Statistical significance.  A general example would be a
          study in which the MTD was administered and the test
          for positive dose-related trend (e.g., Cochran-Armitage
          Test) determined that the slope of the dose-response
          curve was different from zero; however, comparisons of
          the tumor incidences in treated groups with that in the
          control group (e.g., Fisher-Irwin exact test) were not
          significant at p = 0.05.

     b.   Study design.

     c.   Study reporting.  A general example would be a study
          reporting a tumorigenic response,  but lacking
          statistical analyses to verify that an apparent
          increase in incidence was statistically significant.

     d.   A tumor response in a tissue known to have a high and
          variable background rate.

2.    Tumor responses or lack of response which are more than
     likely attributable to excessive doses that compromise major
     organ systems.  Positive studies at levels above the MTD
     should be carefully reviewed to ensure that the responses
     are not due to factors which do not operate at exposure
     levels at or below the MTD.  Evidence indicating that high
     exposures alter tumor responses by indirect mechanisms that
     may be unrelated to effects at lower exposures should be
     dealt with pn an individual basis.  As noted by the OSTP
     (1985), "Normal metabolic activation of carcinogens may
     possibly also be altered and carcinogenic potential reduced
     as a consequence  [of high-dose testing]."  Negative long-
     term animal studies at exposure levels above the MTD may not
     be acceptable if animal survival is so impaired that the
     sensitivity of the study is significantly reduced below that
     of a conventional chronic animal study at the MTD.

     "The carcinogenic effects of an agent may be influenced by
     non-physiological responses  (such as extensive organ damage,
     radical disruption of hormonal function, saturation of
     metabolic pathways, formation of stones in the urinary
     tract,  saturation of DNA repair with a functional loss of
     the system) induced in the model systems.  Testing regimes
     inducing these responses should be evaluated for their
     relevance to the human response to an agent and evidence
     from such a study, whether positive or negative, must be
     carefully reviewed." (OSTP, 1985).

3.    Tumors at the site of oral, dermal or inhalation
     administration attributable to irritation or frank tissue
     damage.
                                13

-------
4.   Tumor responses following administration by a route other
     than oral, dermal or inhalation.  Such tumors may be at the
     site of administration or removed from it.  Some general
     examples are tumors induced following intraperitoneal,
     intravenous or subcutaneous injection, or bladder
     implantation.

     Solid-state carcinogenesis is the occurrence of tumors
     around an inserted inert object.  It is a phenomenon that is
     dependent primarily on the size and shape of the object,
     rather than the chemical composition of the implanted
     material  (Williams and Weisburger, 1986).   Therefore,
     induction of solid-state tumors generally will not be
     considered in the weight-of-evidence approach.

     Data from all long-term animal studies should be considered
     in evaluating carcinogenicity.  However,  carcinogenic
     responses should be evaluated as to their relevance of
     predicting cancer risks to humans.  Therefore, data from
     species that respond most like humans should be used
     preferentially when such information exists.  Data on tumors
     in organs or as a result of effects on metabolic or
     biochemical pathways that don't exist in humans should be
     evaluated very carefully as to their inference of human
     cancer risk-.  Further, a positive carcinogenic response in
     one species/strain or sex is not generally negated by
     negative results in other species.  Replicate negative
     studies, however, that are essentially identical in all
     other respects to a positive study may cast doubt on the
     validity or reproducibility of a positive study.  A variety
     of other weight of evidence issues may make it difficult to
     interpret the significance of tumor data and therefore
     result in a lower classification of carcinogenicity.
     Examples of such issues include:  increased incidence of
     tumors in the highest dose group only and/or only at the end
     of the study;  no substantial dose-related increase in the
     proportion of tumors that are malignant;  the occurrence of
     tumors that are predominantly benign; no close-related
     shortening of the time to the appearance of tumors; negative
     or inconclusive results from a spectrum of short-term tests
     for mutagenic activity; or, the occurrence of excess tumors
     only in a single sex  (EPA, 1985).


B.  Noncarcinocrens

     The full range of possible adverse health effects shall be
evaluated when establishing an acceptable exposure to
noncarcinogens.  Acute/subacute, subchronic/chronic and
reproductive/developmental effects shall be considered.  The
principles of data selection are similar to those for
carcinogenic effects, a well-conducted epidemiologic study which

                                14

-------
demonstrates a positive association between a quantifiable
exposure to a chemical and human disease is generally preferred
for evaluating adverse health effects.  At present, however,
human data adequate to serve as a basis for quantitative risk
assessment are available for only a few chemicals.  Frequently,
inference of adverse health effects to humans must be drawn from
toxicity information gained through animal experiments with human
data serving qualitatively as supporting evidence.  Under this
condition, health effects data must be available from well
conducted studies in animals relevant to humans based on a
defensible biological rationale, i.e. similar metabolic pathways,
etc.
     The following provides guidance on appropriate study design
for a variety of types of toxicity studies against which one may
evaluate the quality and adequacy of data development.  This
evaluation of adequacy of data coupled with effects information
forms the basis for selection of uncertainty factors and
subsequent acceptable exposure levels.


1.  Appropriate Study Design

a.  Acute Toxicity

     Acute Toxicity Determination of an LD50 or LC50 is often an
initial step in experimental assessment and evaluation of a
chemical's toxic characteristics.  Such studies are used in
establishing a dosage regimen in subchronic and other studies and
may provide initial information on the mode of toxic action of a
substance.  Because LD50 or LC50 studies are of short duration,
inexpensive and easy to conduct, they are commonly used in hazard
classification systems.  Acute lethality studies are of limited
use in this process.  However, the data from such studies do
provide information on health hazards likely to arise from
individual short-term exposures.  Although this process should
never allow exposures which approach such acute levels, such
studies provide high dose effects data from which to evaluate
potential effects from exposures which may temporarily exceed the
acceptable chronic exposure level.  An evaluation of the data
should include the incidence and severity of all abnormalities,
the reversibility of abnormalities observed other than lethality,
gross lesions, body weight changes, effects on mortality, and any
other toxic effects.
     In recent years guidelines have been established to,improve
quality and provide uniformity in test conditions.
Unfortunately, many published LD50 or LC50 tests were not
conducted in accordance with current EPA or OECD guidelines since
they were conducted prior to establishment of guidelines.  For
this reason, it becomes necessary to examine each test or study
to determine if the study was conducted in an adequate manner.
     The following is a list of ideal conditions compiled from
various testing guidelines which may.be used for determination of

                                15

-------
adequacy.  Unfortunately, many published studies do not report
details of test conditions making such determinations difficult.
However, test conditions guidelines that might be considered
ideal may include:
          animal- age and species identified;

          minimum of 5 animals per sex per dose group (Both sexes
          should be used.);
          14 day or longer observation period following dosing;
          minimum of 3 dose levels appropriately spaced.  (Most
          statistical methods require at least 3 dose levels.);
          identification of purity or grade of test material used
          (particularly important in older studies);
          if a vehicle used, the selected vehicle is known to be
          non-toxic;
          gross necropsy results for test animals; or
          acclimation period for test animals before initiating
          study.
     Specific conditions for oral LD50:
          dosing by gavage or capsule;
          total volume of vehicle plus test material remain
          constant for all dose levels; and
          animals were fasted before dosing.
     Specific conditions for dermal LD50:
          exposure on intact, clipped skin and involve
          approximately 10% of body surface; and
          animals prevented from oral access to test material by
          restraining or covering test site.
     Specific conditions for inhalation LC50:
          duration of exposure at least 4 hours; and
          if an aerosol  (mist or particulate) the particle size
          (median diameter and deviation) should be reported.

     Although the above listed conditions would be included in an
ideally conducted study, not all of these conditions need to be
included in an adequately conducted study.  Therefore, some
discretion is required on the part of the individual reviewing
these studies (EPA, 1985, OECD, 1987).


b.  14 Day or 28 Day Repeated Dose Toxicity

     The following guidelines were derived using the OECD
Guideline for Testing of Chemicals  (1987), for determining the
design and quality of a repeated dose short-term toxicity study.
The similarity between the conduct of a 14-day and 28-day study
is sufficient to consider them under the same guideline.  The
main difference is the time period over which the dosing takes
place.  These guidelines represent ideal conditions and studies
will not be expected to meet all standards in order to be
considered.  For example, the National Toxicology Program's
cancer bioassay program has generated a substantial database of

                                16

-------
short-term repeated dose studies.  The study periods for these
range from 14 days to 20 days with 12 to 15 doses administered
generally for 5 dose levels and a control.  Since the quality of
this data is goo'd, it is desirable to consider these study
results even though they do not always identically follow the
protocol.
     The purpose of short-term repeated dose studies is to
promote information on possible adverse health effects from
repeated exposures over a limited time period.  Where subchronic
or chronic data are lacking, short-term repeated dose studies of
28 days or longer, with the application of appropriate
uncertainty factors, may be used by this initiative to estimate
acceptable long-term exposure levels.
     According to OECD Guidelines, short-term repeated dose
studies should include the following:
          minimum of 3 dose levels administered and an adequate
          control group used;
          minimum of 10 animals per sex, per dose group (both
          sexes should be used);
          the highest dose level should ideally elicit some signs
          of toxjLcity without inducing excessive lethality and
          the lowest dose should ideally produce no signs of
          toxicity;
          ideal dosing regimes include 7 days per week for a
          period of 14 days or 28 days;
          all animals should be dosed by the same method during
          the entire experiment period;
          animals should be observed daily for signs of toxicity
          during the treatment period  (i.e. 14 or 28 days).
          Animals which die during the study are necropsied and
          all survivors in the treatment groups are sacrificed
          and necropsied at the end of the study period;
          all observed results, quantitative and incidental,
          should be evaluated by an appropriate statistical
          method;

          clinical examinations should include hematology and
          clinical biochemistry, urinalysis may be required when
          expected to provide an indication of toxicity.
          Pathological examination should include gross necropsy
          and histopathology.

     The findings of short-term repeated dose toxicity studies
should be considered in terms of the observed toxic effects and
the necropsy and histopathological findings.  The evaluation will
include the incidence and severity of abnormalities, gross
lesions, identified target organs, body weight changes, effects
on mortality and' other general or specific toxic effects  (OECD,
1987).
                                17

-------
c.  Subchronic and Chronic Toxicity

     The following guidelines were derived using the EPA Health
Effects Testing Guidelines (1985), for determining the quality of
a subchronic or chronic (long term) study.  Additional detailed
guidance may be found in that document.  These guidelines
represent ideal conditions and studies will not be expected to
meet all standards in order to be considered.  The subchronic and
chronic studies have been designed to permit determination of no-
observed-effect levels (NOEL) and toxic effects associated with
continuous or repeated exposure to a chemical.  Subchronic
studies provide information on health hazards likely to arise
from repeated exposure over a limited period of time.  They
provide information on target organs, the possibilities of
accumulation, and, with the appropriate uncertainty factors, may
be used in establishing safety criteria for human exposure.
Chronic studies provide information on potential effects
following prolonged and repeated exposure.  Such effects might
require a long latency period or are cumulative in nature before
manifesting disease.  The design and conduct of such tests should
allow for detection of general toxic effects including
neurological, physiological,  biochemical and hematological
effects and exposure-related pathological effects.
     According to the EPA Guidelines, high quality
subchronic/chronic studies include the following:
          minimum of 3 dose levels administered and an adequate
          control group used;
          minimum of 10 animals for subchronic, 20 animals for
          chronic studies per sex, per dose group (both sexes
          should be used);
          the highest dose level should ideally elicit some signs
          of toxicity without inducing excessive lethality and
          the lowest dose should ideally produce no signs of
          toxicity;
          ideal dosing regimes include dosing for 5-7 days per
          week for 13 weeks or greater  (90 days or greater) for
          subchronic and at least 12 months or greater for
          chronic studies in.rodents.  For other species,
          repeated dosing should ideally occur over 10% or
          greater of animals lifespan for subchronic studies and
          50% or greater of the animal's lifespan for chronic
          studies;
          all animals should be dosed by the same method during
          the entire experimental period;
          animals should be observed daily during the treatment
          period  (i.e., 90 days or greater);
          animals which die during the study are necropsied and,
          at the conclusion of the study, surviving animals are
          sacrificed and necropsied and appropriate
          histopathological examinations carried out;
                                18

-------
          results should be evaluated by an appropriate
          statistical method selected during experimental design;
          and
          such toxicity tests should evaluate the relationship
          between the dose of the test substance and the
          presence, incidence and severity of abnormalities
          (including behavioral and clinical abnormalities),
          gross lesions, identified target organs, body weight
          changes, effects on mortality and any other toxic
          effects noted (EPA, 1985).


d.  Reproductive and Developmental Toxicity

     Studies considered here can be evaluated for quality by
comparing the study protocol or methods section with accepted
testing guidelines prepared by EPA, OECD or Interagency
Regulatory Liaison Group (IRLG).  The EPA Health Effects Testing
Guidelines (1985) include guidelines for both reproduction and
fertility studies and developmental studies.  These EPA
guidelines can serve as the ideal experimental situation with
which to compare study quality.  Studies being evaluated do not
need to match precisely but rather should be similar enough that
one can be assured that the chemical was adequately tested and
that the results closely reflect the true reproductive or
developmental toxicity of the chemical.
     Developmental toxicity can be evaluated via a relatively
short-term study in which the compound is administered during the
period of organogenesis.  Some of the specific guidelines for
developmental studies are cited below.
          minimum of 20 young, adult, pregnant rats, mice or
          hamsters or 12 young, adult, pregnant rabbits
          recommended per dose group;
          minimum of 3 dose levels with an adequate control group
          used;
          the highest dose should induce some slight maternal
          toxicity but no more than 10% mortality.  The lowest
          dose should not produce grossly observable effects in
          dams or fetuses.   The middle dose level, in an ideal
          situation, will produce minimal observable toxic
          effects;
          dose period should cover the major period of
          organogenesis (days 6 to 15 gestation for rat and
          mouse,  6 to 14 for hamster, and 6 to 18 for rabbit);

          dams should be observed daily; weekly food consumption
          and body weight measurements should be taken;
          necropsy should include both gross and microscopic
          examination of the dams; the uterus should be examined
          so that the number of embryonic or fetal deaths and the
          number of viable fetuses can be counted; fetuses should
          be weighted; and

                                19

-------
          one-third to one-half of each litter should be prepared
          and examined for skeletal anomalies and the remaining
          animals prepared and examined for soft tissue
          anomalies.

     The EPA.Health Effects Testing Guidelines (1985) recommend a
two-generation reproduction study to provide information on the
ability of a chemical to impact gonadal function, conception,
parturition and the growth and development of the offspring.
Additional information concerning the effects of a test compound
on neonatal morbidity, mortality and developmental toxicity may
also be provided.  The recommendations for reproductive testing
are lengthy and quite detailed and may be reviewed further in the
Health Effects Testing Guidelines.  In general, the test compound
is administered to the parental (P) animals (at least 20 males
and enough females to yield 20 pregnant females)  at least 10
weeks before mating, through the resulting pregnancies and
through weaning of their Fl offspring.  The compound is then
administered^ to the Fl generation similarly through the
production of their F2 offspring until weaning.  Recommendations
for numbers of dose groups and dose levels are similar to those
reported for developmental studies.  Details are also provided on
mating procedures, standardization of litter sizes (if possible,
4 males and 4 females from each litter are randomly selected),
observation, gross necropsy and histopathology.  Full
histopathology is recommended on the following organs of all high
dose and control P and Fl animals used in mating:  vagina,
uterus, testes, epididymides,  seminal vesicles, prostate,
pituitary gland and target organs.  Organs of animals from other
dose groups should be examined when pathology has been
demonstrated in high dose animals  (EPA, 1985).
     As with any other type of study, the appropriate statistical
analyses must be performed on the data for a study to qualify as
a good quality study.  In addition, developmental studies are
unique in the sense that they yield two potential experimental
units for statistical analysis, the litter and the individual
fetus.  The EPA testing guidelines do not provide any
recommendation on which unit to use, but the Guidelines for the
Health Assessment of Suspect Development Toxicants (EPA, 1986)
states that "since the litter is generally considered the
experimental unit in most developmental toxicity studies, the
statistical analyses should be designed to analyze the relevant
data based on incidence per litter or on the number of litters
with a particular end point".   Others  (Palmer, 1981 and Madson et
al., 1982) identify the litter as the preferred experimental unit
as well.
     Information on maternal toxicity is very important when
evaluating developmental effects because it helps determine if
differential susceptibility exists for the offspring and mothers.
Since the conceptus relies on its mother for certain
physiological processes, interruption of maternal homeostasis
could result in abnormal prenatal development.  Substances which

                                20

-------
affect prenatal development without compromising the dam are
considered to be a greater developmental hazard than chemicals
which cause developmental effects at maternally toxic doses.
Unfortunately, maternal toxicity information has not been
routinely presented in earlier studies and has become a routine
consideration in, studies only recently.  In an attempt to use
whatever data are available, maternal toxicity information may
not be required if developmental effects are serious enough to
warrant consideration regardless of the presence of maternal
toxicity.


C.  Tier Designation

1.  Carcinogens .

     Adequate weight-of-evidence of potential human carcinogenic
effects sufficient to calculate a Tier I Human Cancer Criterion
(HCC) generally consists of data sufficient to meet the
categorical definition of a Human Carcinogen and Probable Human
Carcinogen.  Certain Possible Human Carcinogens may also be
suitable for Tier I criterion development.  Designation of
Possible Carcinogens should be done on a case-by-case basis.  For
example, where cancer bioassays have been well conducted, yet are
limited because they only involve a single animal species, strain
or experiment and do not demonstrate a high incidence, unusual
site or type of tumor, or early onset of tumorigenesis, such data
may be suitable for Tier I criterion development.  In addition,
mode of action, the potential for the compound to interact
directly with DNA as discussed earlier, should be reviewed in
making a Tier designation.
     As discussed earlier, data used for developing Tier I
criteria are expected to carry a high degree of certainty in
their ability to predict an effect.  In this case, the quality of
data and the weight-of-evidence needs to be sufficient to
ascertain that the chemical holds at least a good potential of
producing carcinogenic effects in humans.
     For chemicals where the weight-of-evidence and quality of
data is not sufficient for Tier I numeric criteria the database
may be adequate to develop Tier II values.  In this case, the
data needs to be sufficient to ascertain that the chemical is at
least a possible4 human carcinogen, i.e. Group C.  As discussed
previously under Weight-of-Evidence and Appropriate Study Design,
data on chemicals in this Group suggest only limited evidence of
carcinogenicity.  Studies may be flawed or lacking adequate
design or reporting yet show strong enough evidence of
carcinogenicity or the potential for carcinogenic effects such
that the data should not be ignored.  Examples of such data may
be studies where statistical analysis may be lacking or tumor
incidence may be only marginally significant; tumor responses or
lack of response* may be attributable to excessive dosing, or
there may be high mortality in the exposed groups also due to

                                21

-------
excessive dosing; increases exist for benign tumors only with no
evidence of mutagenicity, etc.  Further discussion as to how
these data are treated in criteria derivation and what potential
differences may exist in such treatment will be discussed further
in the section on criteria development.
     It is important to note that the Group C category may
contain chemicals with databases of highly variable quality.
Because of this," EPA has decided to allow States and Tribes to
address Group C chemicals on a case-by-case basis.  As the final
GLWQI Guidance is written, States and Tribes have the discretion
to develop Tier I criteria or Tier II values for Group C
chemicals based on the overall toxicological database.   The
final Guidance directs that this case-by-case determination be
made taking into account information on mode of action, including
mutagenicity, genotoxicity, structure activity and metabolism.
Those Group C chemicals  (and all chemicals, in general) which act
via a genotoxic mechanism, that is through direct interaction
with DNA and in which a linear low-dose tumor incidence
relationship is expected, may be most appropriately dealt with
through use of a linearized multistage model (LMS) or other model
which appropriately reflect this type of mechanism of action.
The quality of data, as discussed above, would then determine the
Tier designation.  If the chemical does not interact with DNA and
the dose response is considered nonlinear, it may be best dealt
with as a noncancer agent and an RfD should be developed.   See
section on Mode of Action under Section III. Principles for
Criteria Development, B. Carcinogens.


2. Noncarcinogens

     All available toxicity data should be evaluated considering
the full range of possible effects of a chemical.  Unfortunately,
expansive data exists for a limited number of chemicals.
Although all data are evaluated, a line must be drawn below which
data are not sufficient for criteria development.  Adequate data
necessary to develop a Human Noncancer Criterion  (HNC) for
noncancer effects should ideally incorporate at least one well
conducted epidemiologic study which demonstrates a positive
association between a quantifiable exposure to a chemical and
human disease.  Such data exist for only a few chemicals,
therefore, reliance on animal data in establishing noncancer
criteria and values is usually necessary.  Although a more
extensive effects database is desirable, for this initiative, the
minimum database for a Tier I criterion must contain at least a
well conducted subchronic mammalian study.  The duration of the
study must be at least 90 days in rodents or 10% of the lifespan
of other appropriate species with exposure preferably via the
oral route.  Subchronic toxicity studies utilizing dosing periods
of approximately 10% of the test animal's lifespan  (approx. 90
days in rodents) are sufficient to provide information on target
organ effects and can provide an estimate of a no effect level of

                                22

-------
exposure which can be used to establish human health criteria and
values  (OECD, 1981).
     It has been observed, with up to a 95% degree of certainty,
that as little as a 6 fold difference may exist between chemical
effect levels observed at 90 days exposure and at lifetime  (7
years) in rodents. (Weil,et al.,1969.)  Such a study  (90-day or
otherwise used to develop a HNV) should ideally establish a
frank-effect-level (PEL), a lowest-observed-adverse-effect-level
 (LOAEL) and a no-observed-adverse-effect-level  (NOAEL).  The
study must be conducted  in an animal species relevant to humans
 (for example, birds, reptiles, and fish are not considered
biologically relevant to humans due to incompatible
pharmacokinetics, organ  structure, toxicokinetics, etc.) based on
a defensible biological  rationale and generally follow the study
protocol previously discussed.  To further reduce uncertainty,
data from longer studies approaching the lifetime of the test
animal are preferable.   In some cases, chronic studies of one
year or longer in rodents or 50% of the lifespan or greater in
other appropriate test species may b$ sufficient.  Dose response
must be demonstrated in  these longer term studies, however a
LOAEL involving relatively mild and reversible effects may be
considered an acceptable data point for decision making.  For
example, there are many  studies for which only one dose has been
tested with resulting minimal, reversible effects such as minimal
enzyme changes or slight body weight decreases.  These minimal
changes or effects, on their own, may not be thought of as
adverse but may be indicators or precursors to more severe
effects which result from extended exposure and or higher doses.
In those cases, while it can be argued that such an effect may be
a LOAEL, it may also be very close to the NOAEL and is therefore
suitable for criteria derivation.
     Reproductive/developmental effects data as well as evidence
of effects seen in test animals consistent with human
epidemiologic data are also highly desirable in order to evaluate
the full range of potential adverse effects to humans. When data
are not sufficient to meet the minimum requirements for deriving
Tier I numeric criteria, such data may be considered for
development of Tier II values.  As with Tier I, all available
data should be considered, however, a minimum database suitable
for Tier II must, contain at least a well conducted subacute
mammalian study with an exposure period of at least 28 days,
preferably via the oral route of exposure.  The 28-day study was
chosen as a minimally acceptable test that can yield sufficient
information upon which to derive a Tier II value.  Please refer
to Appendix A for further discussion of the use of less than
chronic data to predict chronic endpoints.  The study should,
ideally, establish a dose-response relationship including a
frank-effect-level (PEL), a lowest-observed-adverse-effect-level
(LOAEL), and a no-observed-adverse-effect-level (NOAEL).
Acceptable protocol for conducting such 28 day studies may be
found in the OECD Testing Guidelines (OECD, 1987)  as discussed
previously.  Although the effects observed from short duration

                                23

-------
studies are usually fewer than normally evaluated in longer
duration studies., such effects should at least include mortality,
clinical observations, body weight changes and necropsy of major
organs with whatever histopathology that may be available.  The
minimum data point for decision making on such short term
exposure data must be a NOAEL.  A NOAEL was chosen over a LOAEL
since it is believed the use of a LOAEL may result in
underprotective Tier II values.  A LOAEL from a 28-day study may
not capture the most critical toxic endpoint or be predicting of
chronic endpoints.  Structure-activity relationship (SAR) review
should also accompany the minimum data evaluation.  SAR compares
a chemical with substances that have structural similarities in
order to predict whether the chemical might cause similar toxic
effects.  Such information may then be used in deciding what
uncertainty factors may be appropriate to apply to such limited
data in order to protect against potential similar effects.
     Studies of longer duration than 28 days and with greater
evaluation of effects are more desirable for use in Tier II and
may allow the use of a LOAEL for decision making, depending on
the quality and duration of the study.  As with Tier I,
reproductive/developmental effects data as well as any supportive
epidemiologic evidence is highly desirable in order to evaluate
the full range of potential adverse effects of the chemical. As
with carcinogens, further discussion as to how these data will be
applied in the derivation of acceptable exposure levels and what
adjustments must be made to account for uncertainty will be
discussed in further detail in the section on criteria
development.


III.  PRINCIPLES FOR CRITERIA DEVELOPMENT

A.  General

     The process to derive Tier I criteria or Tier II values is
generally the same.  The weight of evidence and level of
certainty in the^ data available for calculating acceptable
exposure levels establishes the major difference between the two.
For risk assessment of noncarcinogenic effects, the minimum data
requirements differ between tiers.  Therefore, differences in
adjustments to the data  (i.e., uncertainty factors) may also
occur between tiers.  These differences reflect differing levels
of certainty in the data base and an attempt to estimate a level
without appreciable risk of deleterious effects over a lifetime.
In the case of carcinogens, the same quantitative risk assessment
approach generally followed for Tier I is used as well for Tier
II when the data allow.  When the bioassay data for Tier II
carcinogens are not suitable for quantitative risk assessment and
the chemical does not appear to interact with DNA, yet the
weight-of-evidence supports concern for possible threshold
carcinogenic effects, an additional uncertainty factor may be


                                24

-------
applied to the LOAEL or NOAEL for the chemical in order to
account for carcinogenicity.
     All available appropriate human epidemiologic data and
animal toxicologic data shall be considered.  Data specific to an
environmentally appropriate route of exposure shall be used for
criteria and values development, i.e. oral, dermal or inhalation
versus injection, implantation, etc.  Findings from studies using
less than appropriate routes of exposure may be considered
supportive of data obtained through more appropriate routes.
Although local effects are important, for the purposes of this
initiative oral exposure should be considered preferential to
dermal and inhalation data since ingestion is the primary route
of exposure, i.e. water and fish consumption.  Caution must be
exercised in the use of dermal and inhalation data.  Strong
consideration must be given for pharmacokinetic information on
absorption, distribution and metabolism in establishing
equivalent doses with oral exposure.  Effects produced through
exposure via a non-oral route generally should be as a result of
systemic distribution of a toxicant rather than as local effects
to the skin or the respiratory tract.

     In general, study results shall be converted, as necessary,
     to the standard unit of milligrams of toxicant per kilogram
     of body weight per day (mg/kg/day).

     If a study does not specify water or food consumption rates,
     or body weight of the test animals, standard values may be
     used for the test species, such as may be obtained from the
     National Institute of Occupational Safety and Health,
     Registry of Toxic Effects of Chemical Substances (RTECS) or
     similar appropriate references.

     Study results from multiple exposures shall be adjusted, as
     necessary, to a daily dose exposure as if received daily for
     the duration of the exposure period.  The exposure period
     shall be defined as the interval beginning with
     administration of the first dose through the last dose,
     inclusively.


B.  Carcinogens

1.  Mechanism/Mode of Action

     The mechanism by which chemicals cause cancer is not
completely known4, and may involve a variety of mechanisms
occurring at various stages in the carcinogenic process.  A
chemical may act at a single stage or more than one stage.
Currently,  the dominant theory regarding the process by which a
chemical causes cancer is based on two stages:  initiation and
promotion (Borzsonyi, 1984; OSTP, 1985; Trosko, 1983; Williams,
1986).   The concept of two-stage carcinogenesis has been

                                25

-------
supported by investigations involving skin and liver systems
(Argyris, 1985; Pitot and Sirica, 1980).   This operational theory
allows the classification of carcinogens according to their
apparent biological activity.  Some chemicals are capable, by a
variety of genotoxic mechanisms, of triggering the carcinogenic
process  (initiation).  Other chemicals may only alter the
expression of the initiated genome and enhance tumor development
by a variety of non-genetic mechanisms (promotion).   Complete
carcinogens operate by both processes.  Initiators are capable of
directly altering in an irreversible manner the native structure
of the DNA.  Promotion may be reversible in the early stages,
appears to be highly dose-dependent, and apparently requires
prolonged or repeated exposure  (Pitot, 1981; Slaga,  1984; Thomas,
1986) .
     Calling an agent a promoter does not eliminate the
carcinogenic hazard potential of a chemical.  Indeed, data
indicate that promoting phenomena are largely responsible for the
expression of many human cancer types  (Williams, 1986).  However,
it is very difficult both in principle and in practice to confirm
the assertion that a given chemical acts by promotion alone
(OSTP, 1985) .
     Currently, for most if not all chemicals, data are not
available to determine the exact mechanism by which they cause
cancer.  As a result, significant controversy exists regarding
the existence of thresholds for carcinogens.  Therefore, for the
purpose of routine cancer risk assessment, agents that are
positive in long-term animal experiments should be considered as
complete carcinogens unless there is evidence to the contrary
because, at present, it is difficult to determine whether an
agent is acting only as a promoting or cocarcinogenic agent  (EPA,
1986) .  However, in making all judgements with regard to
mechanism, all data related to mode of action should be
considered
     EPA, in revising its Guidelines for Carcinogenic Risk
Assessment, is suggesting that mode of action information,
reflecting the manner in which an agent causes cancer, be used
more extensively in carcinogen assessments than has been done in
the past.   As a result, the final GLWQI Guidance now includes a
requirement to review all possible evidence including available
information on mode of action including
mutagenicity/genotoxicity, structure activity, and metabolism.
Mode of action should be used in the assessment and
characterization of the potential human carcinogenicity of a
substance, and in the selection of a model for quantifying its
risks, especially at low doses.  This  change in emphasis, while
still draft and in the formative stages,  is being recommended so
that all relevant scientific data can be used to  carry out cancer
risk assessments.  Of particular importance, in determining mode
of action, is distinguishing between carcinogens  that are
mutagenic  (i.e., interact directly with DNA) and  those that are
non-mutagenic.


                                26

-------
     To distinguish between carcinogenic agents that are
mutagenic and non-mutagenic, many test systems can be used.
These include assays for changes in DNA base pairs of a gene such
as gene mutation- tests in bacteria or mammalian cells (see 40 CFR
798:5265, USEPA 1991) and chromosomal aberrations, such as in
vivo cytogenetics tests.  Initial consideration is usually given
to mammalian-bone marrow using either micronucleus assays to
detect damage of chromosomes or mitotic apparatus by agents  (See
40 CFR 798:5398, USEPA 1991; Dearfield et al. 1991) or metaphase
chromosomal analysis for detection of structural aberrations
(also see 40 CFR 798:5385, USEPA 1991).  Mutagenicity assessment
guidelines are provided in USEPA (1991).   Other assays that do
not measure gene- mutations or chromosomal aberrations per se
(e.g., tests for DNA adducts, unscheduled DNA synthesis, sister
chromatid exchange, strand breaks,  repair and recombination) are
not sufficient in and of themselves to make a determination of
mutagenicity;  they only provide supportive evidence of
mutagenicity.
     For the purpose of this initiative, unless adequate
mechanistic data demonstrate otherwise, a nonthreshold mechanism
will be assumed for those chemicals classified as Group A, B and
C carcinogens.


2. Data Review

     If acceptable human epidemiologic data are available, a risk
associated dose shall be set equal to the lifetime exposure which
would produce an incremental increased cancer risk of 1 in
100,000.  If more than one study is judged acceptable, the study
resulting in the' most protective risk associated dose is
generally used to calculate the human cancer criterion.
     In the absence of appropriate human studies, data from a
species that responds most like humans should be used, if
information to this effect exists.   Where several studies are
available, which may involve different animal species, strains,
and sexes at several doses and by different routes of exposure,
the following approach to selecting the data sets is used:

     The tumor incidence data are separated according to organ
     site and tumor type.

     All biologically and statistically acceptable data sets are
     presented.

     The range of the risk estimates is presented with due regard
     to biological relevance (particularly in the case of animal
     studies)  and appropriateness of route of exposure.

     Because is it possible that human sensitivity is as high as
     the most sensitive responding animal species, in the absence
     of evidence to define the most relevant species to humans,

                                27

-------
     the biologically acceptable data set from long-term animal
     studies showing the greatest sensitivity should generally be
     given the greatest emphasis, again with due regard to
     biological and statistical consideration (EPA, 1986).

Exceptions to the above may exist as follows:

     If two or more studies exist which are identical with
     respect to species, strain, sex and tumor type and are of
     equal quality, the geometric mean of the potency from these
     studies may be used (EPA,  1980).  In certain instances where
     there are several studies  in various strains and even
     several species and where  there is no indication of a single
     study or species judged most appropriate, the geometric mean
     estimates from all studies may be used to determine the
     potency.  This ensures that all relevant data are included
     in the derivation  (EPA, 1989d).

     Where two or more significantly elevated tumor sites or
     types are observed in the  same study, extrapolations may be
     conducted on selected sites or types.  These selections will
     be made on biological grounds.  To obtain a total estimate
     of carcinogenic risk,  animals with one or more tumor sites
     or types showing significantly elevated tumor incidence
     should be pooled and used  for extrapolation so long as
     double-counting of tumor-bearing animals is prevented.  The
     pooled estimates will generally be used in preference to
     risk estimates based on single sites or types.  Quantitative
     risk extrapolations will generally not be done on the basis
     of totals that include tumor sites without statistically
     significant elevations  (EPA, 1986).

     Benign tumors should generally be combined with malignant
     tumors for risk estimates  unless the benign tumors are not
     considered to have the potential to progress to the
     associated malignancies of the same histogenic origin.  The
     contribution of the benign tumors, however, to the total
     risk should, be indicated  (EPA, 1986).


3.  Model

a.  Nonthreshold Approach

     When acceptable human epidemiologic data are not available
and a nonthreshold mechanism is assumed, carcinogenesis bioassay
data, as appropriate, are fitted to a linearized multistage
computer model.   (Note:  Other models, such as time-to-tumor,
modifications or variations of the multistage model may be used
which consider the data more appropriately under case-by-case
circumstances.)
                                28

-------
     Since risks at low exposure levels cannot be measured
directly either by animal experiments or by epidemiologic
studies, a number of mathematical models have been developed to
extrapolate from high to low dose.  Different extrapolation
models, however, may fit the observed data reasonably well but
may lead to large differences in the projected risk at low doses
(EPA, 1986).
     "No single mathematical procedure is recognized as the most
appropriate for low-dose extrapolation in carcinogenesis.  When
relevant biological evidence on mechanism of action exists  (e.g.,
pharmacokinetics, target organ dose), the models or procedures
employed should be consistent with the evidence.  When data and
information are limited, however, and when much uncertainty
exists regarding the mechanism of carcinogenic action, models or
procedures which incorporate low-dose linearity are preferred
when compatible with the limited information."   (OSTP, 1985)
     In an attempt to characterize the underlying dose-response
relationship, models which use the nonthreshold assumption of
carcinogenicity are commonly used.  The linearized multistage
(LMS) model calculates an upper bound based on the theory that a
developing tumor goes through several different stages which can
be affected by a chemical carcinogen.  The LMS model is forced to
be linear in the low-dose region, regardless of the shape of the
dose response curve, and therefore LMS-based risk estimates may
be regarded as relatively conservative when used for public
health protection.
     In calculating upper bounds on potency from the LMS model,
the bioassay data are fitted to the LMS model, e.g. Global 86
developed by Howe et al. (1986).  The 95 percent upper bound
estimate on the linear term, q.^*! is used to calculate the upper
confidence bound on risk for a given dose, or the lower
confidence bound on dose for a given risk.  The slope factor
(q-L*) is taken as an upper bound of the potency of the chemical
in inducing cancer at low doses. When pharmacokinetic or
metabolism data are available, or when other substantial evidence
on the mechanistic aspects of the carcinogenesis process exists,
a low-dose extrapolation model other than the linearized
multistage procedure might be considered more appropriate on
biological grounds.  When a different model is chosen, the risk
assessment should clearly discuss the nature and weight-of-
evidence that led to the choice.  Considerable uncertainty will
remain concerning response at low doses; therefore, in most cases
an upper-limit risk estimate using the linearized multistage
procedure should also be presented for comparison  (EPA,  1986).


b. Threshold Approach

     Whenever appropriate human epidemiological data are not
available,  and the preponderance of data suggest that a chemical
causes cancer via a threshold mechanism, the risk associated dose
                                29

-------
may be calculated via a method other than a linearized multistage
model on a case-by-case basis.
     As a default, a safety factor approach may be pursued after
thorough evaluation of. the toxicologic and pharmacologic data on
the compound including, but not limited to:  mechanism of
carcinogenesis, number and type of tumors induced, the
spontaneous incidence of tumors, the number of animal species
tested and affected, metabolic considerations, epidemiologic
data, extent of data supporting a non-genotoxic mechanism of
tumor induction,' i.e. mutagenicity assay data, initiation/
promotion assay data, etc.


4.  Lifespan Adjustment

     If the duration of the study (Le) is significantly less than
the natural lifespan for the species  (L), the slope factor, q *
can be adjusted to account for unobserved tumors due to the snort
study duration. * The assumption is that if the duration of the
study was increased, tumor incidence would continue to increase
as a constant function of the background rate.  EPA believes this
adjustment should be made on a case-by-case basis taking into
consideration factors such as mechanism of action, the type of
tumor and the organ affected.  One option for correcting for less
than lifetime duration is to use the method described by EPA
(1980).   Based on the EPA (1980) method it is assumed that the
cumulative tumor rate would increase at least by the 3rd power of
age since age specific rates for humans increase at least by the
2nd power of age and often considerably higher.
     For mice and rats, the natural lifespan  (L) is defined as 90
weeks and 104 weeks, respectively.  The slope factor adjustment
may be conducted for mice and rat data if the study duration (Le)
is significantly less than natural lifetime such as less than 78
weeks for mice or 90 weeks for rats, by multiplying the slope
factor by the factor (L/Le) .  For other species, this adjustment
factor may also be used whenever appropriate, using species-
specific values for L and the Le trigger level.  The latter may
be determined using the trigger levels for mice and rats as a
guideline.


5. Species Scaling

     Low-dose risk estimates derived from laboratory animal data
extrapolated to humans are complicated by a variety of factors
that differ among species and potentially affect the response to
carcinogens.  Included among these factors are differences
between humans and experimental test animals with respect to life
span, body size, genetic variability, population, homogeneity,
existence of concurrent disease, pharmacokinetic effects such as
metabolism and excretion patterns, and the exposure regimen.


                                30

-------
     The usual approach for making interspecies comparisons has
been to use standardized scaling factors.  Commonly employed
standardized dosage scales include3mg per kg body weight per dayj
ppm in the diet or water, mg per m  body surface area per day,
and mg per kg body weight per lifetime.  In the absence of
comparative toxicological, physiological, metabolic and
pharmacokinetic data for a given chemical, extrapolation on the
basis of surface area is considered to be most appropriate
because certain pharmacological effects commonly scale according
to surface area '(Dedrick, R.L., J. Pharmacokin. Biopharm. 1:435-
461; Freireich et al., Cancer Chemother. Rep. 50:219-244  (1966);
Pinkel, D., Cancer Res., 18:853-856 (1958)).
     The species scaling factor is calculated by dividing the
average weight of a human (Wh) by the weight of the test species
(Wa) and taking the cube root of the resultant value.  This is
based on the premise that a close approximation of the surface
area is 2/3 the power of weight, and that the exposure in mg-2/3
the power of body weight/day is similarly considered to be an
equivalent exposure (EPA, 1980).  The animal slope factor is
multiplied by this factor to obtain the human slope factor.
                 yi^ (human) = q.* (animal) X 3 J  "" kg
                                          \  Wa kg


The weight  (Wa) of the test species should be the average adult
weight from the particular bioassay if possible, or derived from
available data tables or standard assumed weights.
     EPA also believes other scaling factors may be used as long
as there is justification on the basis of species-specific
pharmacokinetic data.


C.  Noncarc inogens

1.  Mechanism

     Noncarcinogens generally are assumed to have a threshold
dose or level below which no adverse effects should be observed
(NOAEL).  For many noncarcinogenic effects,  protective mechanisms
are believed to exist that must be overcome before an adverse
effect is manifested.  For example, where a large number of cells
perform the same or similar function, the cell population may
have to be significantly depleted before the effect is seen.  As
a result, a range of exposures exists from zero to some finite
value that can be tolerated by the organism with essentially no
expression of adverse effects.  In the development of an estimate
without appreciable risk of deleterious effect from exposure to a
chemical, the effort exists to find the upper bound of this
tolerance range (i.e., the maximum subthreshold level).  Because
variability exists in the human population,  attempts are made to


                                31

-------
assure a subthreshold level which would not result in appreciable
risk to sensitive individuals in the population.  For most
chemicals, this level can only be estimated and incorporates the
use of uncertainty factors indicating the degree of extrapolation
used to derive the estimated value (EPA, 1989d).
     Exceptions to this principle exist.  Noncarcinogenic
chemicals may exist with no identifiable threshold.  One example
of this phenomenon appears to be nickel, for which there is no
apparent threshold for subsequent dermal effects of the chemical.
Another example is the effects of lead exposure, where no
discernable threshold has been identified.  Other examples of
this exception may include genotoxic teratogens and germline
mutagens.  These agents have been specifically identified to
differentiate between chemicals thought to produce reproductive
and/or developmental effects via a genetically linked effect from
those chemicals more routinely considered to act via a nongenetic
mechanism.  There are few chemicals,  if any, which currently have
sufficient mechanistic information about their mode of action to
link teratogenic or developmental effects to mutational events
during organogenesis, histogenesis or other stages of
development.  These chemicals may also interact with germ cells
to produce mutations which may be transmitted to the zygote and
also be expressed during one or more of these stages of
development.
     EPA has recognized this potential and discussed this issue
in their 1989 Proposed Amendments to Agency Guidelines for Health
Assessments of Suspect Developmental Toxicants (EPA, 1989c) and
in their 1986 Guidelines for Mutagenicity Risk Assessment  (EPA,
1986) .  Various statements within these guidelines should raise
concern for the potential for future generations inheriting
chemically induced germline mutations or suffering from
mutational events occurring in utero:

     "It is estimated that at least 10% of all human disease is
     related to specific genetic abnormalities...11

     "Life in our technological society results in exposure to
     many natural and synthetic chemicals.  Some have been shown
     to have mutagenic activity in mammalian and sub-mammalian
     test systems, and these may have the potential to increase
     genetic damage in the human population...  The extent to
     which exposure to natural and synthetic environmental agents
     may have increased the frequency of genetic disorders in the
     present human population and contributed to the mutational
     "load" that will be transmitted to future generations is
     unknown at this time.  However,  for the reasons cited above,
     it seems prudent to limit exposure to potential mutagens."

     "Approximately 3% of newborn children are found to have one
     or more significant congenital malformation at birth, and by
     the end of the first postnatal year, about 3% more are found
     to have serious developmental defects.  Of these, it  is

                                32

-------
     estimated that 20% are of known genetic transmission, 10%
     are attributable to known exogenous factors (including
     drugs, infections, radiation and environmental agents) ..."

     An awareness of the potential for such teratogenic/mutagenic
effects should be established in order to deal with such data
should it occur in the future.  However, without adequate data to
support a genetic or mutational basis for developmental or
reproductive effects, the default becomes an uncertainty factor
approach.  This approach follows the procedure identified for
noncarcinogens assumed to have a threshold.  Genotoxic teratogens
and germline mutagens should be considered an exception while the
traditional uncertainty factor approach is the general rule for
calculating criteria or values for chemicals demonstrating
developmental/ reproductive effects.
     A nonthreshpld mechanism shall be assumed for genotoxic
teratogens and germline mutagens.  Since there is no well
established mechanism for calculating criteria protective of
human health from the effects of these agents, criteria will be
established on a case-by-case basis.  For more information on
this phenomenon, it is recommended that the reader refer to the
EPA Drinking Water Criteria Documents for Nickel and Lead.


2.  Data Review .

     All toxicity data on a chemical should be evaluated for
criterion or level of concern development.  Those studies
representing the best quality and most appropriate data as
discussed previously under appropriate study design should be
selected for defining adverse effects and their level of
occurrence.  As previously discussed, adequate human
epidemiologic data should be used in evaluating the adverse
health effects of a chemical whenever available.  When adequate
human data are not available, animal data from species most
relevant to humans should be used.  In the absence of data on the
"most relevant" species or the inability to identify the most
relevant species,  data from the most sensitive animal species
tested, i.e., the species demonstrating an adverse health effect
at the lowest administered dose via a relevant route of exposure,
shall generally be used.
     For guidance, adverse health effects are those deleterious
effects which are or may become debilitating, harmful or toxic to
the normal functions of an organism including reproductive and
developmental effects.  These do not include such effects as
tissue discoloration without other noted effects, or the
induction of enzymes involved in the metabolism of the substance.
Guidelines for defining the severity of adverse effects have been
suggested by Hartung and Durkin  (1985) which proposes a ranking
from slight to severe effects.  Distinguishing slight effects
such as reversible enzyme induction and reversible subcellular
change from more' severe effects is critical in distinguishing

                                33

-------
between a no observed adverse effect level (NOAEL) and a low-
observed-adverse-effect (LOAEL).
     The experimental exposure level representing the highest
dosage level tested at which no-adverse-effects were demonstrated
(NOAEL) shall be. used in the formula for criteria development.
In the absence of such data, the dosage level at which the
lowest-observed adverse-effect-level was demonstrated may be used
in some circumstances for criteria development. .
     Preference should be given to studies involving exposure
over a significant portion of the animal's lifespan since this is
anticipated to reflect the most relevant environmental exposure.
An exception to this is where reproductive and/or developmental
effects may be demonstrated to have a lower NOAEL over a shorter
exposure period..  When two or more studies of equal quality and
relevance exist, the geometric means of the NOAEL or LOAEL may be
used.
3.  Uncertainty Factors

     The choice of appropriate uncertainty and modifying factors
reflects a case-by-case judgement by experts and should account
for each of the applicable areas of uncertainty and any nuances
in the available data that might change the magnitude of any
factor.  Several reports describe the underlying basis of
uncertainty factors  (Zielhuis et al., 1979; Dourson and Stara,
1983) and research into this area (Calabrese, 1985; Hattis et
al., 1987; Hartley and Ohanian, 1988; Lewis et al., 1990; Dourson
et al., 1992).
     The following are examples of where uncertainty exists as a
result of weakness either in the data base or the process which
needs accommodation:

     using dose-response information from effects observed at
     high doses to predict the adverse health effects that may
     occur following exposure to the low levels expected from
     human contact with the agent in the environment;

     using dose-response information from short-term exposure
     studies to predict the effects of long-term exposures, and
     vice-versa;'

     using dose-response information from animal studies to
     predict effects in humans; and

     using dose-response information from homogeneous animal
     populations or healthy human populations to predict the
     effects likely to be observed in the general population
     consisting of individuals with a wide range of
     sensitivities.  (EPA, 1989d)
                                34

-------
For this initiative, accommodation for these uncertainties will
be handled in the following process.  For further detail in the
selection of these uncertainty factors, please see Appendix A..


a.  Intraspecies. uncertainty factor

     An uncertainty factor of 10 shall generally be used when
extrapolating from valid experimental results from studies on
prolonged exposure to average healthy humans.  This 10-fold
factor is used to protect sensitive members of the human
population.


b.  Interspecies. uncertainty factor

     An uncertainty factor of 100 shall generally be used when
extrapolating from valid results of long-term studies on
experimental animals when results of studies of human exposure
are not available or are inadequate.  In comparison to a, above,
this represents an additional 10-fold uncertainty factor in
extrapolating data from the average animal to the average human.


c.  Subchronic to chronic uncertainty factor

     An uncertainty factor of up to 1000 shall generally be used
when extrapolating from animal studies for which the exposure
duration is less than chronic (but greater than subchronic, e.g.,
90 days or more in length) or when other significant deficiencies
in study quality are present, and when useful long-term human
data are not available.  In comparison to b, above, this
represents an additional uncertainty factor of up to 10-fold for
less than chronic (but greater than subchronic)  studies.


d.  Less than subchronic duration uncertainty factor

     An uncertainty factor of up to 3000 shall generally be used
when extrapolating from animal studies for which the exposure
duration is less than subchronic (<90 days, e.g., 28 days).  In
comparison to b,' above, this represents an additional uncertainty
factor of up to 30-fold for less than subchronic studies (<90
days,  e.g., 28-day).  The level of additional uncertainty applied
for less than chronic exposures depends on the duration of the
study used relative to the lifetime of the experimental animal.


e.  LOAEL to NOAEL uncertainty factor

     An additional uncertainty factor of between one and ten may
be used when deriving a criterion from a lowest observable

                               35

-------
adverse effect level (LOAEL).   This uncertainty factor accounts
for the lack of an identifiable no observable adverse effect
level (NOAEL).  The level of additional uncertainty applied may
depend upon the severity and the incidence of the observed
adverse effect.


f.   Limited database uncertainty factor

     An additional uncertainty factor of between one and ten may
be applied when there are limited effects data or incomplete
subacute or chronic toxicity data (e.g.,
reproductive/developmental data).   The level of quality and
quantity of the experimental data available as well as structure-
activity relationships may be used to determine the factor
selected.
     When deriving an uncertainty factor in developing a Tier I
criterion or Tier II value, the total uncertainty, as calculated
following the guidance of a-f, cited above, shall not exceed
10,000 for Tier I criteria and 30,000 for Tier II values.


D.   Exposure Assumptions

     When dealing with site specific and individual specific
exposure, it is more accurate to use actual available exposure
information to estimate an individual's specific risk.
Individual behaviors can be assessed and specific activity
information compiled to address quantity, frequency and duration
of exposure.  When dealing with such diverse populations of
individuals covering as large an area as the Great Lakes Basin,
extreme ranges of behaviors and activities are likely.
Therefore, deriving default assumptions that can estimate
reasonable exposures which address the vast majority of the Basin
population becomes necessary.


1.   Body Weight

     National body weight data has been compiled by the National
Center for Health Statistics from a survey conducted from 1976
through 1980 entitled the second National Health and Nutrition
Examination Survey  (NHANES II).  Approximately 28,000 people aged
6 months to 74 years were surveyed with other 20,000 individuals
actually interviewed and examined.  Weighted mean body weights
have been determined from this data.  Since body weights change
so rapidly during childhood, it is reasonable to use mean adult
body weight to reflect population body weights when assuming a
long exposure duration.  From national survey data, the mean
adult body weight appears to be approximately 72 kg  (EPA, 1989).
If NHANES data are separated out by Great Lakes regional data, it
appears that the mean may even be higher for the Great Lakes

                                36

-------
Basin population.  However, as a matter of convention, 70 kg has
been used for many years in chemical regulatory programs and
still appears appropriate for this initiative.
     EPA believes 70 kg is an appropriate body weight because it
represents a reasonable measurement for the entire population.
If a State believes that use of a lower body weight is
appropriate (which yields a more stringent criterion), the State
or Tribe may adopt such an assumption in calculating their
criteria and values under their authority to establish more
stringent requirements pursuant to section 510 of the Act.
     As to whether lower body weights should be used to protect
women of childbearing age, children and fetuses, EPA believes
that categorically adopting more conservative body weight
assumptions may not be appropriate.  Each chemical must be
addressed separately since some chemicals may be generically
toxic to both adult sexes, while others may be specifically toxic
to one sex more than the other, or children, specifically.  It
therefore would not be appropriate to require generally that all
criteria be based on conservative body weight assumptions.  In
the case of mercury, however,  a fetotoxic chemical, to be
protective of women of child bearing age, EPA has assumed a body
weight of 65 kg (as opposed to 70 kg)  which results in a Tier I
mercury criterion of 1.8 ng/L, which is slightly less than the
proposed criterion of 2 ng/L.   EPA has set a final Tier I
criterion for mercury at 1.8 ng/L.
                               37

-------
Body Weights of Adults  (kilograms)


Age
18
25
35
45
55
65
18




25
35
45
55
65
75
75



Mean
73.7
78.7
80.8
81.0
78.8
74.8
78.1

Men
Std. Error

Women
Men and Women
Std. Error
of Mean Mean
' 0.0035
0.0034
0.0040
0.0041
0.0041
0.0051
0.0016
.
Body Weights


Age

3
6
9
12
15



3
6
9
12
15
18


Mean
11.9
17.6
25.3
35.7
50.5
64.9
Boys
Std. Error
60.6
64.2
67.1
67.9
67.9
66.6
65.4

of Mean
0.0032
0.0037
0.0043
0.0044
0.0045
0.0048
0.0017

Mean
67.2
71.5
74.0
74.5
73.4
70.7
71.8

Std. Error
of Mean
...
—
—
—
—
—
_ _ _
(USEPA, 1989a)
of Children (kilograms)

Girls

Std . Error
of Mean Mean
. 0.0016
0.0014
0.0023
0.0038
0.0051
0.0047
11.2
17.1
24.6
36.1
50.7
57.4
of Mean
0.0011
0.0015
0.0024
0.0043
0.0049
0.0042
Mean
11.6
17.4
25.0
36.0
50.6
61.2
Boys and Girls
Std. Error
of Mean
...
—
—
—
—
_ _ _
                                   (USEPA, 1989a)
                 38

-------
2.  Duration of Exposure

a.  Population Mobility

    The  default assumptions  for mobility  is  to  consider that  an
individual remains in the same residence for a "lifetime".
Movement of individuals from individual residences, communities,
or even regions of the country may influence exposure duration to
contaminants from sources such as drinking water and sport caught
fish dramatically.  If movement occurs within the same community,
the influence by drinking water may not change.  If movement is
still within the region, the influence of contaminated sport fish
may not change.  Be that as it may, mobility may lower or
increase exposure duration and intensity.
    Based on a survey  conducted by  the Oxford Development
Corporation,  a property management company, the average residence
time for an apartment dweller is estimated to range from 18 to 24
months.  A survey conducted by the Bureau of the Census in 1983,
determined that 93% of householders moved into their present home
between 1950 and 1983.  Using this information, the following
time of residence ranges have been determined:

           Years in Current Home         Total % of Householders

                  0 -  1                       7.5
                  1 -  3                      16.9
                '3 -  13                      40.2
                13-18                      11.0
                18 -  23                       7.9
                23 -  33                       9.5
                     33                       7.0

    Based on these  statistics, the  50th percentile of
householders living in their current residence is 9.4 years and
the 90th percentile is 29.8 years.  This data does not, of
course, indicate how far people move or whether they will
increase or decrease their exposure by moving.  Accordingly, it
is only of limited relevance in determining exposure patterns.


b.  Life Expectancy

    Life Expectancy Statistical data on life expectancy is
gathered annually by the U.S. Department of Commerce.  Data
presented by the Bureau of Census for 1985 show that life
expectancy for the total U.S. population is 74.7 years.  The
breakdown of this average is as follows:
                                 Male         Female  Total
           white                 71.8         78.7     75.3
           black and other       67.2         75.2     71.2
           black                 65.3         73.7     69.5


                               39

-------
            total average        71.2         78.2    74.7

                                              (USEPA,  1989a)
Although the average life expectancy now is approximately 75
years, it is probable that over the course of a lifetime there
should be periods of no exposure that add up to at least five
years.  Accordingly, the traditional default value for "lifetime"
exposure of 70 years appears adequate for considering chronic
"lifetime" exposure.


3. Incidental Exposure

   The suggested  0.01 liter/day adjustment for recreational
exposure is based on an assumption of 123 hours of recreational
exposure equivalent to swimming,  and consumption of an average
mouthful (30 mL) of water per hour of such recreational exposure.
Exposure potential, when averaged over a year, equals 0.01
liters/day.  Such exposures could result from an average of one
hour swimming per day during the four month warm weather period
starting in mid May and ending in mid September (i.e., 123 days).
   EPA has recently estimated a national average  frequency of
swimming to be 7 days/year with a 2.6 hour duration (EPA, 1989d).
An earlier EPA publication estimated an average annual frequency
of 9 days/year with a 2 hour duration of exposure.  Other total
body contact recreation such as water skiing was also identified
as having approximately 20 million participants with a total
exposure of 260 million hours per year or an average of 14 hours
exposure per participant.  Partial body contact was identified as
20% body exposure for fishing and 40% body exposure for boating.
This earlier reference listed 68 million people involved
nationally in boating with an average duration of 1600 million
person hours per. year and 54 million people involved nationally
in fishing with 6600 million person hours duration per year.  The
resulting individual participant average exposure duration equals
approximately 24 hours and 122 hours of participation,
respectively  (EPA, 1979).  If each hour of total body contact
equivalent for bathing, water skiing, boating and fishing were
calculated (18, 14, 10 and 24 hours, respectively), the total
equals 66 hours of average body contact exposure.
   Various recreational surveys have been  conducted in Michigan
and may serve as- a typical example of Great Lakes Basin activity.
Estimations similar to EPA's for activities per participant and
hours per participation may be calculated from this information.
If we were to assume an individual were to participate in all
activities for the number of days listed from the 1981 Michigan
Travel and Recreation Survey and for the duration of hours per
participation as identified in the 1976 Recreation survey, and
the percentage adjustment made for total body contact exposure
from the older EPA reference, the following calculations may be
made:
                                40

-------
              Activity Days
                   per
               Participant
               Hours per
              Participati
                  on
   Body
  Contact
Adjustment
Hours of
Exposure
 Swimming
 Fishing
 Power
 Boating
 Water
 Skiing
 Sailing
 Canoeing
13.3
14.3
24.5 (total)
2.1 (ave.)
3.7 (ave.)
3.2
1.0
0.2
0.4
27.9
10.6
31.4
 9.6           1.5


10.4 (total)    3.2

 4.8           3.9
    1.0


    0.4

    0.4

   TOTAL
  14.4


  13.3

   7.5

 105.1
                                                 (Wells,  1990)
Given these comparisons of water recreation activities, the
suggested incidental exposure level appears appropriate, given
the variability in individual behavior.


4.  Drinking Water

   Two liters of water has been the nationwide  conventional
estimate of aduljb human's daily water consumption.  The 2 liters
of water per day is a historical figure set by the U.S. Army in
determining the amount of water needed for each person in the
field.  The National Academy of Sciences  (NAS) estimates that
daily water consumption may vary with physical exercise and
fluctuations in temperature and humidity.  It is reasonable to
assume those living in a more arid, hot climate will consume
higher levels of water.  NAS has calculated the average per
capita water consumption to be 1.64 liters per day.  The National
Cancer Institute. (NCI) in a study, also known as the Cantor
(1987) study, also has looked at this issue with an overall tap
water consumption rate of 1.39 liters of water per day as their
study average.  The NCI study is of particular interest since
data were compiled from Detroit,  Iowa, New Jersey and Connecticut
giving a database of over 3500 respondents with similar weather
conditions to the Great Lakes Basin.  The consumption rate of
less than or equal to 1.96 liters per day is equated to the 100%
cumulative frequency level as seen in the following table:
                                41

-------
     Frequency Distribution of Tap Water Consumption Rates*

      Consumption Rate  (L/day)            Cumulative Frequency
           0.80                                   19.2
        0.81-.-  1.12                                39.6
        1.13  -  1.33                                59.7
        1.45  -  1.95                                79.9
           1.96                                   100.0

    *Represents consumption in a  "typical" week.(Cantor et al.,
   1987)

   Other researchers have discovered average levels both higher and
lower than NCI.   The Food and Drug Administration's  (FDA)  Total
Diet Study estimated rates  for water and water-based  foods for two
groups of adults to  be 1.07 and 1.3 liters per day with an average
of 1.2 liters per day.  The U.S.  Department of Agriculture (USDA)
in the 1977-78  Nationwide Food Consumption Survey identified daily
beverage intakes of  from 1.24 to  1.73  liters  per day.  In a more
recent study specifically characterizing tap water intake by Ershow
and Cantor (1989), 2 liters/day represents approximately the 85th
percentile value of  drinking  water consumption.   After review of
all these studies,  EPA has  judged  the average adult drinking water
consumption  rate  to be 1.4  liters per day  with  a reasonably
conservative assumption of 2  liters  per day  as being  the  90th
percentile value  (USEPA, 1989a).   This  compensates  in  part  for
parts of the population, such as  manual  or migrant laborers,  who
drink much more than 2  liters a day.


5.  Fish Consumption

   Much debate has  occurred  over  the  years as to the  appropriate
regionally caught fish consumption rate for  the Great Lakes Basin.
This is one  area where extreme differences  exist in the region's
consumption behavior.   A large segment of the population consumes
little or no fish caught from the  region, while a small segment of
the population c6nsumes a significant quantity of  regionally caught
fish.
   Several  studies  of fish consumption and sport angler behavior
have been  evaluated to  estimate  an appropriate  fish  consumption
value  for  the region.   Three regional  surveys;  Michigan (West,
1989), Wisconsin (Fiore, 1989) and New  York  (Connelly,  1990) ; have
been selected  for consideration.   In  summary,  the results of the
Michigan survey suggest that  approximately 65%  of  the licensed
anglers consume less than  one meal per week of all fish.  This is
consistent  with' Wisconsin data which estimates  the mean annual
total number of all  fish meals consumed by anglers to be  41.  This
is also consistent  with New York anglers  who consume 45.2 meals
statewide and  approximately  41.6  total meals in the  regions with

                                42

-------
the greatest  number of sport anglers  and greatest sport fishing
effort.  Based on the Michigan and Wisconsin surveys, approximately
43% of the fish meals consumed are sport  caught, or approximately
18-19 meals per year.  .Estimates of meal sizes range up to 8 ounces
(0.5 pounds)  or an approximate total of 9-9.5 pounds per year. This
equates  to  a daily  fish consumption  rate of 11-12  g/day.   The
Michigan survey data indicate a mean annual total fish consumption
rate of  17 gm/day or  (at 43%) approximately 7 gm of  sport caught
fish.  There is poor data on the proportion of the nonsport caught
(commercial)   fish  consumed within the region which  is  actually
caught within the region.  Using the Michigan survey data,  at least
22% of  the fish consumed are species from outside  the region.
Thereby, the maximum proportion of regionally caught  and  consumed
fish in Michigan may be  estimated to  be only 78% or  13 grams per
day.  All those contacted familiar with commercial fishing within
the  region  estimated  the  major amount  of  regionally caught
commercial fish are sold outside of  the  region  and therefore,
generally  not  available to regional  anglers.   If one assumes a
conservative  mean  total of  regionally caught meals  to  equal 24
meals per year at 8 ounces per meal or up  to 48 meals per year at
4 ounces per meal,  the mean daily consumption rate is 15  gm/day.
   A second study conducted by West et al. (1993) for  the State of
Michigan provided results which were very supportive of the use of
15 grams/day.   This  study is a full year (February 1991 to February
1992)  fish consumption  survey of  7000 licensed Michigan  anglers.
The survey  found  that  the  average sport  fish  consumption rate,
adjusted for  non-response bias, was  14.5 grams/day.  The average
total fish (all fish,  not just Great Lakes sport fish)  consumption
rate,  adjusted  for  non-response  bias, was 24.4  grams/day.   This
study indicated that fish consumption rates may differ  according to
race and income level.   The lowest income group (< $14,999/year)
averaged 21 grams/day sport  fish  consumption as compared to 14.7
grams/day for those  making $40,000 or more/year.   The average sport
fish consumption'rate for minorities was 23.2 grams/day as compared
with 16.3 grams/day for non-minority  individuals.   Lower income
($24,999 or less) minorities averaged  the  highest consumption rate
of all groups in the survey: 43.1 grams/day sport caught  fish and
57.9 grams/day total  fish;   Non-minority individuals  of  lower
income averaged 18.6 grams/day sport fish and 25.8 grams/day total
fish.   The study also indicated that minorities eat  less  fish from
the Great Lakes and more fish from the  inland tributaries  than non-
minority individuals.   For greater detail on the West et al. (1993)
study and a statistical analysis of the west study findings refer
to U.S. EPA (1995).
   For  this initiative,  the assumption of 15 g/day of regionally
caught fish should adequately estimate the consumption rate of the
mean angler population and  their  families for all  sport caught
fish.    A much larger segment of  the  sport  angler population is
included if this consumption is attributed totally to species of
fish   more    susceptible   to  persistent   and   bioaccumulative
contaminants,  i ..e.,  the  salmonids.  Based on the Regional Survey
data,  including number  of licenses bought and  used,  members per

                               43

-------
family, and  fish consumption rates  for sport anglers,  15 g/day
approximates  at  least  the  90%  consumption level  of  regionally
caught  fish  for  the  regional  population as  a  whole,  i.e.,
fisherpersons as. well as nonfisherpersons.


6.  Relative Source Contribution

   In  the  final  GLWQI Guidance,  the  Agency assumes an 80 percent
relative source  contribution  (RSC)  from surface  water pathways
(water and fish) for  all  chemicals,  bioaccumulative chemicals of
concern (BCCs) and non-BCCs,  in deriving noncancer criteria/values.
A 100 percent RSC is assumed for all chemicals in deriving cancer
criteria/values.  EPA also recommends that actual data be used in
developing an RSC when available.  As stated in the 1980 National
Guidelines, to  account for  exposures  from  other  sources,  actual
exposure data can be subtracted from the RfD (ADI, as it was called
in 1980) to account for contributions  of the pollutant from diet
and air (ADI  -  (DT + IN) where DT is the estimated non-fish dietary
intake and IN is the estimated daily  intake by  inhalation (U.S.
EPA,  1980).   Therefore, where data  are available, if  States or
Tribes want to use actual data in developing their RSC, they may do
so,  following  the  procedure  outlined  in  the  1980  National
Guidelines.  It  is important to note,  however, that  EPA's policy on
how to use exposure data in  developing  an RSC is now under review.
Once EPA  has finalized its  policy review  on  the  RSC,  EPA will
address the application of the RSC during the triennial review of
Water Quality Standards under section 303 of the Clean Water Act.
Until such time, the Agency has decided to apply an RSC of 80% to
all noncarcinogenic chemicals (both BCCs and non-BCCs).
   With regard to using different RCSs  for BCCs and non-BCCs which
was  presented in  the Proposed  Human  Health TSD,  EPA  does  not
believe there is a clear difference in  RSC development for BCCs as
opposed to non-BCCs.  While it may be true that surface water may
be the major route  of exposure for bioaccumulatives (through fish
consumption),  even though a pollutant is not bioaccumulative, it
does  not  preclude  the  possibility  that  there  may  be  other
significant sources of exposure.
   With  regard  to  the use  of  a  80  percent default  value,  EPA
believes that  the  assumption  helps  to  provide  some  measure of
protection  against the possibility  that  exposures  from other
sources may contribute to the overall exposure of the public to a
particular contaminant.   Available data  indicate  that non-water
sources  contribute varying amounts  to  overall   exposure to  a
particular chemical (U.S.  EPA 1982, U.S. EPA 1983).   Such exposures
can occur through air and  the diet.  Since available data indicate
that such  exposures can and do  occur, but these  data are often
limited in their ability  to predict with  precision the relative
source contribution, EPA believes it  is prudent not  to assume that
all  exposure  to a  pollutant occurs  from one medium.     The 80
percent default  was  chosen  because  it  reflects  the  approximate
contribution from surface  water pathways  (fish consumption) to the

                                44

-------
overall exposure to BCCs such as PCBs  in the Basin.  For PCBs, the
FDA  Total  Diet 'Study  estimates  that consumption  of  pollutant-
bearing fish represents  the most significant exposure.  The average
adult's daily intake of  PCBs  via diet is  estimated to be 560 ng,
versus estimated inhalation  levels  of 100 ng per  day.   Based on
these estimates, diet  contributes approximately 85%  of exposure
(ATSDR,   1987).    .It  appears  likely  that,   for  other  highly
bioaccumulative chemicals, a similar estimate may be made as well.
   For nonbioaccumulatives, 80 percent was also chosen as a default
value to account,for the other possible non-water sources which may
contribute  to the  overall  exposure  of  the chemical.   However,
actual exposure data  may also  be used in the  final  Guidance by
States and Tribes to calculate a relative source  contribution.  EPA
recognizes that the choice of a default value of  80% in these cases
is fundamentally a policy judgment that criteria development should
reflect the fact that  exposures to a pollutant occur through other
media, rather than  an empirically-based calculation of the precise
proportion of exposure via water versus  non-water  sources,  since
such values vary, on a case-by-case  basis.   EPA also acknowledges
that use of a 80% default for non-BCCs is a conservative measure,
however, if other significant exposures are not accounted for, the
criteria could underestimate overall exposure to the chemical and
thus could underestimate the risk of adverse health effects.   In
addition, in the absence of data, it  is prudent and  consistent with
the  health protection goals  of the  CWA  to include a  margin of
safety in  the event that there are  exposures  from other sources.
The  important  fact, EPA believes, is to  take some accounting of
other possible exposure pathways.
   With  regard to  the  concern that point sources  should  not be
expected to compensate for the failure to address other pollutant
sources, EPA does not believe that the  relative source contribution
factor  in  the  final  methodology  unduly  burdens point  source
dischargers.   It is  common  practice  in  EPA  programs  (e.g.,  in
establishing maximum contaminant level goals  under the SDWA)  to
take  into  account  other  routes of  exposure  to a  chemical when
establishing  health-based standards  for a particular  route  of
exposure.  If this step  is not taken, and EPA were always to assume
that  no exposures  occurred  through   other  media  (in  spite  of
evidence to  the  contrary),  then  the  totality of  exposures could
obviously result in adverse health effects,  contrary to EPA's goal
of establishing  standards that  insure  that such  effects  do not
occur.  EPA agrees,  however,  that it is important to take steps to
address all routes of exposure to pollutants  in order to achieve
the greatest overall public health protection at the least cost.
                                45

-------
IV.  CRITERIA CALCULATIONS

A.  Standard Exposure Assumptions

   BW = weight of an average human  (BW = 70 kg).

   WC = per capita water consumption for surface waters
        classified as public water  supplies  (WC, = 2 liters/day)
                             -or-
      average per  capita  incidental daily  water exposure for
        surface waters not classified as drinking water  supplies
        (WCr  =0.01 liters/day)

   FC = per capita daily consumption of regionally caught
        fish  = 0.015 kg/day

   BAF = bioaccumulation factor.

B.  Carcinogens  *

   When  a  linear,  nonthreshold dose-response relationship  is
assumed, the  human cancer value shall be calculated using the
following equation:

HCV  =       RAD x BW
      WC  + [(Peru x  BAF^) +  (PC™ x BAF^) ]
   Where:
                4

   HCV =   Human Cancer Value in milligrams per liter (mg/L).

   RAD =   RAD in milligrams toxicant per kilogram body weight per
           day   (mg/kg/day)  that is associated with a  lifetime
           incremental  cancer risk  equal to 1 in 100,000.

   BW =   Body weight of an average human  (BW = 70kg).

   WC =   average per capita water consumption  (both drinking and
          incidental exposure)  for  surface waters  classified as
          public water  supplies  (WCd = 2  L/day)  and average per
          capita incidental daily water exposure for surface waters
          not used as public water supplies  (WCr =0.01 liters/day)

        =  mean  consumption of  trophic level  3 fish by regional
           sport fishers  = 0.0036 kg/day

        =  mean  consumption of  trophic level  4 fish by regional
           sport fishers  = 0.0114 kg/day

   BAFTL3 « BAF  for trophic level 3  fish

         = BAF  for trophic level 4  fish


                                46

-------
C.  Noncar c inogens

   The human noncancer value shall be  calculated as  follows:

     HNV  =  APE x BW x RSC
             WC + [(FCru x  BAFTLa)  +  (FC^ x BAF^) ]
   Where :

   HNV =    HNV in milligrams per liter (mg/L) .

   ADE =    ADE in milligrams toxicant per kilogram  body weight per
            day (mg/kg/day) .

   RSC =    RCS factor of 0.8  for all chemicals of  concern.  This
            is  used to  allow  for potential exposure via sources
            other than consumption of contaminated water and fish
            recreational exposure.  States may develop an RSC using
            actual exposure data following the procedures specified
            in  the 1980  National Guidelines.

An ADE may be derived directly from the following example methods
depending on the type and quality of the toxicity database:

   1.  a  scientifically valid  reference dose  (RfD) as identified
   through best available information  sources,  such  as IRIS; and

   2.   a scientifically valid acceptable daily intake  (ADI)  as
   identified  from  the  U.S.  Food and  Drug Administration.   Both
   sources should be updated with the most recent data available.

   3 .   a chronic or  subchronic NOAEL for humans  exposed  to the
   toxicant via contaminated drinking  water as  follows:

          ADE  =  NOAEL  (ma/1)  x WC ,
                        U x Wh     ~

          Where : •
          U = Uncertainty factor of 10-100 depending on the quality
              of the data.

   4.  a  chronic  or subacute NOAEL from a mammalian test species
   exposed to  toxicant contaminated drinking water as  follows:
          ADE  =  NOAEL (mg/1) x
                                 wa

                         U

          Where:
          Vw =  Volume  of  water consumed per day  by test animals
          (L/day) .
                                47

-------
       Wa = Weight of test animal  (kg) .
       U = Uncertainty factor of 100-1000  depending on quality
       of data.   An additional uncertainty factor  of  up  to 10
       may be used to account for studies of very  short  term,
       e.g.,  28  days .

5.  a chronic or  subacute NOAEL from a mammalian test species
exposed to toxicant -contaminated food as follows:

       ADE  - NOAEL  (mg/kg food)  x
                          U

        Where :
        f c =  Daily food consumption by test  animal  (kg) .
        Wa =  Weight of  test animal  (kg) .
        U = Uncertainty factor of 100-1000 depending on quality
       of data.  An additional  uncertainty factor of up to 10
       may be used to account  for studies of very short term,
       i.e.,  28  days .

6. a  chronic or subacute NOAEL from a mammalian test species
   exposed to a toxicant by gavage as follows:

       ADE = NOAEL (mq/kq) x Fw
                       U

       Where:
       Fw = Fraction of week dose.
       U = Uncertainty  factor  of 100-1000 depending on quality
       of data.   An additional uncertainty  factor  of  up  to 10
       may be used to account for studies of very  short  term,
       i.e.,  28  days .

7.  A  chronic or  subacute  NOAEL from a mammalian test species
exposed to a toxicant by inhalation:

       ADE
NOAEL (me

T^)
U
x
X
I X
Wa
fw

x

fd

x

r

       Where :                                  3
       I = Inhalation rate for test species (m /day) .
       fw = Fraction of week exposed.
       fd = Fraction of day exposed.
       r = Absorption coefficient.
       Wa = Weight of test animal (kg) .
       U = Uncertainty factor of 100-1000 depending on quality
       of data. An additional uncertainty  factor of up to 10 may
       be used to account for studies of  very short term,
       i.e., 28 days .
                            48

-------
8. Similar approaches shall be followed when data is limited to
a LOAEL with an appropriate increase in uncertainty factor.  For
example, a subacute LOAEL from a mammalian test species exposed
to toxicant contaminated drinking water would be calculated as
follows :
       ADE   =   LOAEL (mg/1)  x
                              wa
                      U

       Where :
       Vw = Volume  of water consumed per  day by test  animal
       (L/day) .
       Wa = Weight  of  the  test animal  (kg) .
       U =  Uncertainty  factor of  1000-30,000  depending  on
       quality of data and severity  of effect.
                            49

-------
                            REFERENCES

Agency for Toxic Substances and Disease Registry  (ATSDR), 1987,
   Toxicology  Profile.for Selected PCBs  (Aroclor-1260,  -1254,  -
   1248, -1242, -1232, -1221,  and -1016), Syracuse Research Corp.,
   Contract  No.  68-03-3228,  Oak Ridge National  Laboratory,  Oak
   Ridge, TN.

Agency for Toxic Substances and Disease Registry  (ATSDR), 1989,
   Toxicological   Profile  for  Toluene,  Clement  Assoc.,  Inc.,
   Contract No. 205-88-0608, Centers for Disease Control, Atlanta,
   GA.

Argyris, T.S., 1985, Regeneration and the Mechanism of Epidermal
   TumorPromotion,  CRC Reviews  in Toxicology,  14:211-258.

Borzsonyi,  M. et al.,  (Ed.), 1984, Models, Mechanisms and Etiology
   of Tumor Promotion, International Agency for Research on Cancer,
   World Health Organization,  IARC Scientific Publications No. 56,
   Lyon, France.

Cantor, K.P., R.'Hoover,  P. Hartge,  et al., 1987, Bladder cancer
   drinking water source,  and  tap water consumption: A case-control
   study, J. National Cancer  Institute, 79 (6)-.1269-1279.

Connelly,  N.A., T.L. Brown and B. A. Knuth, 1990, New York
   Statewide  Angler Survey,  1988,  New York  State  Department of
   Environmental Conservation,  Albany,  NY.

Crouch, A.C.  and R. Wilson,  1982, Risk/Benefit Analysis, Ballinger
   Publishing Co.,  Cambridge, MA.

Dearfield,  K.,A. Auletta, M.C. Cimino, and M.M. Moore. 1991.
   Considerations   in  the  U.S.  EPA's   Testing  Approach  for
   Mutagenicity.   Mutation  Research.   258:259-283.

Dedrick, R.L., 1973, Animal scale-up, J. Pharma. Biopharm., 1:435-
61.

Dedrick, R.L., K.B. Bischoff,  and D.S.  Zaharko, 1970, Interspecies
   correlation  of  plasma  concentration history  of methotrexate
    (NSC-740), Cancer  Chemother.  Rep. Pt. 1,  54:95-101.

Ershow, A.G. and K.P. Cantor, 1989.  Total Water  and Tapwater
   Intake  in  the  United  States:  Population-Based  Estimates of
   Quantities and Sources, National Cancer Institute, Bethesda, MD.

Fiore,B.J.  et al..,  1989, Sport  Fish Consumption and Body Burden
   Levels  of  Chlorinated Hydrocarbons:   A Study  of Wisconsin
   Anglers,  Archives  of  Environmental  Health,  44:82-88.

Freireich,  E.J., E.A. Gehan, D.P. Rail, L.H. Schmidt, and H.E.

                                50

-------
   Skipper, 1966, Quantitative comparison of toxicity of anticancer
   agents  in mouse,  rat,  hamster,  dog,  monkey and man,  Cancer
   Chemother. Rep.,  50:  219-44.

Howe, R.B.  and K.S.  Crump and Landingham,  1986, A Computer Program
   to  Extrapolate  Quantal  Animal  Toxicity Data  at  Low  Doses,
   Prepared for Office of Carcinogen Standards,  Occupational Safety
   and Health Administration,  U.S. Department of Labor, Contract
   41 USC 252C3, Washington, D.C.

International Agency for Research On Cancer  (IARC),  1985, IARC
   Monographs on  the  Evaluation of  the  Carcinogenic Risk of
   Chemicals  to Humans,   Volume  36,   Preamble,   World  Health
   Organization, Lyon,  France.

International Agency for Research on Cancer  (IARC),  1991, IARC
   Monographs on the Evaluation of Carcinogenic Risks to Humans,
   Volume 53, Preamble,  World  Health Organization,  Lyon, France.

Manson,  J.M. et al., 1982, Teratology Test Methods for Laboratory
   Animals, In:  Principles and Methods  of Toxicology, Hayes, A.W.
    (Ed), Raven  Press,  New York,  NY.

Mausner,  J.  and  S. Krammer, 1985, Mausner and Bahn Epidemiology; An
   Introductory Text,  W.B.  Saunders  Company, Philadelphia, PA.

National Cancer Institute (NCI), 1976,  Guidelines for Carcinogen
   Bioassay in  Small Rodents, Technical Report Series No. 1, U.S.
   Department of  Health,  Education and Welfare,  NCI-CG-TR-l.
   National Toxicology Program (NTP),  1984,  Report of the Ad Hoc
   Panels on Chemical Carcinogenesis Testing and Evaluation of the
   National  Toxicology Program, Board  of Scientific Counselors,
   U.S.

Government Printing Office,  Washington,  D.C., 1984-421-1324726.
   Office of  Science and Technology Policy  (OSTP), 1985, Chemical
   Carcinogens;  A  Review  of  the  Science and  Its  Associated
   Principles,  Federal Register, Vol. 50, No. 50.  March 14, 1985,
   10371-10442.

Organization for Economic Cooperation and Development (OECD) , 1987,
   Guidelines fdr Testing of Chemicals,  Paris,  France.

Palmer,  A.K., 1981,  Regulatory Requirements for Reproductive
   Toxicology:  Theory and Practice, In:  Developmental Toxicology,
   Kimmel, C.A.  and J.  Buelke-Sam (Eds), Raven Press,  New York, NY.
   Pinkel, D, 1958,  The use of body surface  area as a criterion of
   drug dosage  in cancer chemotherapy,  Cancer  Res.,18:853-6.

Pitot,  H.,  et al. 1981. The Natural History of
                                51

-------
   Carcinogenesis:  Implications of Experimental Carcinogenesis in
   the Genesis of Human Cancer, Journal of. Supramolecular Structure
   and Cellular  Biochemistry,  17:133-146.

Pitot, H.C. and A.E. Sirica, 1980, The Stages of Initiation and
   Promotion  in  Hepatocarcinogenesis,  Biochimica  et  Biophysica
   Acta, 605:191-215.

Slaga, T.f  1984, 'Can Tumor Promotion be Effectively Inhibited?  In:
   Borzsonyi,  M.  et al.   (Eds),  1984,  Models,   Mechanisms  and
   Etiology of Tumor Promotion, International Agency for Research
   on  Cancer,   World   Health  Organization,   IARC  Scientific
   Publications  No. 56,  Lyon,  France.

Thomas, R.  (Ed), Safe Drinking Water Committee, 1986, Drinking
   Water and  Health,  National Research Council,  National Academy
   of Sciences,  National Academy Press, Washington, D.C., p. 141,
   157.

Tripartite Working  Party,  1985,  Criteria  for   Identifying  and
   Classifying  Carcinogens,  Mutagens  and  Teratogens,  Developed
   jointly by the  Safety  of  Chemicals  Committee  of  CEFIC,  the
   International Affairs Group  of  CMA/SOCMA,  and  the  Canadian
   Chemical   Producers  Association,   Obtained   from  Chemical
   Manufacturers Association,  Washington, D.C.

Trosko, J., C. Jones and C.  Chang, 1983,  The Role  of Tumor
   Promoters  on  Phenotypic Alterations Affecting Intercellular
   Communication and Tumorigenesis, Annals N.Y. Academy of Science,
   407:316-327.

U.S.  Department  of Health and Human Services, 1979, Registry of
   Toxic   Effects  of  Chemicals  Substances  (RTECS),  National
   Institute  for Occupational Safety and Health,  Cincinnati, OH.

U.S.  Environmental Protection Agency (EPA), 1979,  Identification
   and Evaluation of Waterborne Routes of Exposure from Other Than
   Food  and  Drinking Water,  Washington,   D.C.  Office  of  Water
   Planning and  Standards.  EPA  440/4-79-016.

U.S.  Environmental Protection Agency (EPA), 1980,  Water Quality
   Criteria  Availability,  Appendix  C  Guidelines  and Methodology
   Used in the  Preparation  of Health Effects Assessment Chapters
   of the  Consent Decree Water Quality  Criteria Documents, Federal
   Register,  Vol.  45, November 28, 1980, 79347-79357.

U.S.  Environmental Protection Agency (EPA),  1985,  Toxic Substances
   Control Act Test Guidelines; Final Rules, Federal Register, Vol.
   50, No. 188,  September 27,  1985,  39421-39425.

U.S.   Environmental Protection Agency (EPA), 1986,  Guidelines for


                                52

-------
    Carcinogen Risk Assessment, Federal Register, Vol. 51,  No.  185,
    September 24,  1986,  33992-34002.

U.S. Environmental Protection Agency  (EPA), 1986a, Guidelines for
    the  Health  Assessment  of  Suspect  Developmental Toxicants,
    Federal  Register, Vol.  51, No.  185, September 24,  1986,  34028-
    34040.

U.S. Environmental Protection Agency  (EPA), 1986b, Guidelines for
    Mutagenicity Assessment,  Federal Register,  Vol.  51,  No.  185,
    September 24,  1986,  34006-34012.

U.S. Environmental Protection Agency (EPA), 1989a, Exposure Factors
    Handbook.  Washington,  B.C. Office  of Health and  Environmental
    Assessment,  Exposure Assessment Group.   EPA/600/8-89/043.

U.S. Environmental Protection Agency (EPA), 1989b, National Primary
    and Secondary.Drinking Water Regulations, Proposed Rule, Federal
    Register Vol.  54, No. 97, May 22,  1989,  p.  22069.

U.S. Environmental Protection Agency  (EPA), 1989c, Proposed
    Amendments to Agency Guidelines for Health Assessment of Suspect
    Developmental  Toxicants,  Federal  Register,  Vol.  54, No. 9386,
    March 6,  1989.

U.S. Environmental Protection Agency  (EPA),  1989d,  Risk Assessment
    Guidance for Superfund,  Volume 1, Human Health Evaluation  Manual
    (Part  A)  -  Interim Final, Office  of Emergency  and Remedial
    Response,  Washington, D.C., EPA/540/1-89/002.

U.S.EPA. 1991.  Pesticide Assessment Guidelines, Subdivision  F.
    Hazard  Evaluation:  Human and Domestic  Animals,  Series   84,
    Mutagenicity, Addendum 9, EPA-540/09-91, 122, OPPTS, Washington,
    D.C. PB91-158394.

U.S. Environmental Protection Agency.  1995.  Fish  Consumption
    Estimates  Based  on  the 1991-1992 Michigan  Sport Anglers  Fish
    Consumption  Survey.  Final Report. EPA Contract  No.  68-C4-0046,
    SAIC Project No.  01-0813-07-1676-040. February  21, 1995.

Wells, Philip, 1990, Michigan Department of Natural  Resources
    (MDNR),Recreation Division,  Personal  Communication   to  Gary
    Hurlburt,  Surface Water Quality Division, MDNR.

West, P.,  M. Fly', R. Marans,  F. Larkiri and D. Rosenblatt. 1993.
    1991-1992 Michigan Sport Anglers Fish Consumption Study.  Final
    report  to the Michigan Great  Lakes Protection Fund,  Michigan
    Dept. of Natural  Resources.  University of Michigan, School of
    Natural  Resources.  Natural Resources Sociology Research  Lab.
    Technical  Report  #6. May 1993.

West, P.C.  et al., 1989, Michigan Sport Anglers  Fish Consumption

                                53

-------
   Survey:   A  Report  to  the Michigan  Toxic  Substance  Control
   Committee, University of Michigan Natural  Resource Sociology
   Research Lab.   Technical Report #1,  Ann Arbor, Michigan, MDMB
   Contract #87-20141,

Williams, G. and J. Weisburger, 1986, Chemical Carcinogens, In:
   Klaassen, C., M. Amdur and J. Derell  (Eds), 1986, Casarett and
   Doull's Toxicology; The Basic Science of Poisons,  Third Edition,
   MacMillan Publishing Co., New York, NY.
                                54

-------
                            APPENDIX A
                       UNCERTAINTY FACTORS
A. INTRODUCTION

   Uncertainty factors  (also called safety factors) are intended
for  use  in extrapolating  toxic  responses  thought  to have  a
threshold (i.e., noncarcinogenic effects) .  "Uncertainty factor" is
defined  as  a  number  that  reflects,  the  degree  or   amount  of
uncertainty  that  must be  considered  when experimental  data  in
animals  are extrapolated  to  man  (EPA,  1980).     In addition,
uncertainty  factors  are  used  when  extrapolating   from  small
populations of  humans to  the  entire heterogeneous human population
and when  extrapolating from a single  animal  species to wildlife
communities.   The  use  of uncertainty factors in extrapolating
animal toxicity data to acceptable exposure levels for humans has
been the cornerstone of regulatory toxicology  (National Academy of
Sciences, 1980) .. This appendix will provide the risk assessor with
additional  guidelines, rationale  and  information  concerning the
selection of uncertainty factors.
   Because of the high degree of judgment involved  in the selection
of uncertainty factors,  the  risk  assessment justification should
include a detailed discussion of the selection of the uncertainty
factors along with the data to which they are applied.
   This  report  is   organized  with  the  recommended  uncertainty
factors listed in Part B for quick reference,  and a discussion of
those factors and their support in Part C.  Also included in Part
C  is  a  discussion   of the exposure  duration terms   "subacute",
"subchronic", and "chronic".
                               Al

-------
B. RECOMMENDED UNCERTAINTY FACTORS

   1.   An uncertainty factor of 10 shall  generally be used when
extrapolating  from valid  experimental results  from  studies  on
prolonged exposure to average healthy  humans.  This  10-fold factor
is used to protect sensitive members of the human population.
   2.   An uncertainty factor of 100 shall  generally be used when
extrapolating  from   valid   results  of   long-term  studies  on
experimental animals when results of studies of human exposure are
not available or are inadequate.  In comparison to  1, above, this
represents   an   additional    10-fold  uncertainty   factor   in
extrapolating data from the average animal to the average human.
   3.  An uncertainty factor of up  to  1000  shall generally be used
when  extrapolating from  animal studies for  which  the  exposure
duration is less than chronic  (but greater than subchronic, e.g.,
90 days or more in length) or when other significant deficiencies
in study quality are present,  and when useful long-term human data
are not available.  In comparison to 2, above,  this represents an
additional  uncertainty factor  of  up  to  10-fold  for less  than
chronic (but greater than subchronic)  studies.
   4.  An additional uncertainty factor of  between one  and ten may
be used when deriving a criterion from a lowest observable adverse
effect  level  (LOAEL).   This uncertainty factor accounts  for the
lack of an identifiable no observable adverse effect  level (NOAEL).
The level  of  additional uncertainty applied may depend  upon the
severity and the incidence of the observed adverse  effect.
   5.  An uncertainty factor of up  to  3000  shall generally be used
when  extrapolating from  animal studies for  which  the  exposure
duration  is less  than subchronic (<90 days,  e.g.,  28  days).   In
comparison to 2, above, this represents an additional uncertainty
factor of up to 3*0-fold for less than subchronic studies (<90 days,
e.g., 28-day).   The level of  additional uncertainty applied for
less than  chronic  exposures  depends on the duration of the study
used relative to the lifetime of the experimental animal.
   6.  An additional uncertainty factor of  between one  and ten may
be applied when  there are  limited  effects  data  or  incomplete
subacute or chronic toxicity data (e.g., reproductive/developmental
data).  The level of quality  and quantity of the experimental data
available as well as structure-activity relationships may be used
to determine the" factor selected.
   When deriving  an  uncertainty factor in  developing a  Tier  I
criterion or Tier  II  value,  the total uncertainty,  as calculated
following the guidance of  1-6, cited above,  shall not exceed 10,000
for Tier I criteria and 30,000  for Tier II values.
   The  following  discussion  is generalized  for  categories  of
commonly  applied  uncertainty  factors which  are  used  in  the
developing  the  specific  uncertainty  factors  used  in  GLWQI,
described above..
                                A2

-------
C. DISCUSSION

   Dourson  and  Stara  (1983)  reviewed  available literature  on
uncertainty factors  which are used to  estimate  acceptable daily
intakes (ADIs) for toxicants.  They found that the use and choice
of these factors is supported by reasonable qualitative biological
premises and specific biological data.  Therefore, in the absence
of  adequate  chemical-specific  data,  uncertainty  factors  for
criteria  derivation  may  be  selected  according  to  reasonable
assumptions and .approximations rather  than  total arbitrariness.
They presented  a set  of guidelines  for  the use  of uncertainty
factors based on those utilized by  the FDA, WHO, NAS,  and EPA,
indicating  consistency   and  widespread   acceptance  among  the
scientific community.  Those guidelines have been adapted herein
for use in risk assessment under the Great Lakes Initiative.  Their
rationale and experimental  support  are  discussed  below.   The
guidelines should  not be  misconstrued  as being  unalterable and
inflexible.   They are intended to help ensure appropriateness and
consistency of  -risk  assessments.    They should  be regarded  as
general recommendations, with the realization that the data for a
particular chemical may be such that a different uncertainty factor
would be more appropriate.
   A 10-fold  factor is recommended when extrapolating from valid
experimental  results  from human studies of  prolonged exposure.
People of all ages, states of health, and genetic predispositions
may be exposed to environmental contaminants.  The 10-fold factor
is intended to offer  protection  for the sensitive subpopulations
(the  very  young,   the  aged,  medically  indigent,   genetically
predisposed,  etc.),  since the observed no-effect level is generally
based on  average healthy individuals.  Experimental support for
this 10-fold  factor is  provided by log-probit analysis  and the
study of composite human sensitivity  (Dourson and Stara, 1983).
   However,   Calabrese  (1985)  has  presented   data   on  human
variability   in   several   physiological   parameters   and   in
susceptibility  to   several   diseases,  and  concluded that  human
variation may range up to two or three orders  of magnitude.  While
human variation in the metabolism of various xenobiotics may have
a 1000-fold range,  Calabrese  (1985)  noted that the  vast majority of
the  responses addressed fell  clearly  within  a  factor  of  10.
Another study on key human  pharmacokinetic  parameters indicates
that the  10-fold factor to  encompass human  variability may only
capture the variability  among normal  healthy adult humans.  That
report  recommends   further  study   to  determine   the  degree  of
additional susceptibility among  sensitive  subpopulations  (EPA,
1986).
   Given  the  heterogeneous and highly outbred state  of the human
population,    and   the    multifactorial    nature   of   disease
susceptibility,  reliance on the adequacy of the 10-fold factor for
extrapolation to "safe"  levels appears  somewhat  precarious.   But
because of its history  of use and  current widespread acceptance,
this factor may continue to be used until the availability of new
data indicating quantitatively a more acceptable factor.

                               A3

-------
   A 100-fold factor is recommended when extrapolating from valid
results of  long-term studies on  experimental animals with
results of  studies of human exposure  not available or scanty
(e.g..  acute exposure only). This represents the 10-fold factor for
intraspecies extrapolation  (see C.I) and an  additional  10-fold
uncertainty factor for extrapolating data from the average animal
to the average man.
   The 100-fold uncertainty factor has been justified for use with
the risk extrapolation for food additives.  That justification has
been based  on  differences  in  body  size,  differences  in  food
requirements varying  with  age,  sex, muscular expenditure,  and
environmental conditions  within  a species,  differences  in  water
balance of  exchange between the body  and its  environment  among
species, and differences  among  species  in susceptibility to  the
toxic effect of a  given contaminant  (Bigwood,  1973).  The use of
the 100-fold  uncertainty  factor has  also been substantiated by
citing differences in  susceptibility between  animals and humans to
toxicants, variations  in sensitivities in the  human population,  the
fact that the number of animals  tested is small compared with the
size of the human population that may be  exposed, the difficulty in
estimating human intake, and the possibility  of synergistic action
among chemicals (Vettorazzi, 1976).
   On a dose per  unit of  body weight basis,  large animals (e.g.,
man)  are  generally more  sensitive to  toxic effects than  small
animals (e.g., rats, mice).   This  principle  is attributed to  the
relationship between animal  size and pharmacokinetics, whereby the
tissues of a large animal  are exposed to a substance (mg/kg dose)
for a much longer  time than  the  tissues of a small animal.   This
principle   has    been    demonstrated    experimentally.       The
pharmacokinetic processes  underlying this phenomenon include:   in
general, large animals metabolize  compounds  more  slowly  than do
small animals; large animals have many more susceptible cells; in
large animals, substances  are distributed more slowly and tend to
persist longer; the blood volume circulates  much  more rapidly in
small animals.  Thus, for the same mg/kg dose,  human tissues  are
exposed to a substance for a much longer time than rodent tissues
(National Academy of Sciences, 1977).
   Experimental  support   for  the additional  10-fold  uncertainty
factor when extrapolating  from animal data to  humans is provided by
studies  on  body-surface  area  dose  equivalence  and  toxicity
comparisons between humans and  different animal species (Dourson
and Stara,  1983).  On a dose per unit of body-surface area basis,
the effects seen  in man are  generally in the same range as those
seen in experimental  animals.   An  interspecies adjustment factor
accounts for  differences  in mg per  kg  body weight doses  due to
different body-surface areas between  experimental animals and man.
The factor may be  calculated by  dividing the average weight of a
human (70 kg)  by the weight  of the  test  species  (in kg) and taking
the cube root of this value.  Thus on a body weight basis, man is
assumed to  be more  sensitive than  the experimental animals by
factors of approximately 5 and 13 for rats  and mice, respectively.
For most experimental  animal species (i..e., all species larger than

                               A4

-------
mice) ,  the   10-'fold  decrease  in  dose  therefore  appears  to
incorporate  a margin of  safety.   For  mice,  the interspecies
adjustment factor suggests that the additional  10-fold uncertainty
factor for interspecies extrapolation to humans is not large enough
 (Dourson and  Stara,  1983).   Nevertheless, the additional 10-fold
factor is  considered adequate  to adjust  from mice to humans when
chemical-specific data are not available.
    A factor  of up to 1000 is recommended when extrapolating from
animal studies for which  the exposure duration is less than chronic
 (i.e..less than'50% of  the  lifespan)  or  when other significant
deficiencies in study quality are present, with no useful long-term
or  acute  human data.   This represents  the 10-fold factors  for
intraspecies  and  interspecies  extrapolation  (see  C.2),  and an
additional uncertainty factor  of up to 10-fold for extrapolating
from  less  than chronic to chronic  animal  exposures (or when the
data  are  significantly flawed  in some other  way).   Injury from
chronic exposure may occur in at least three ways:  by accumulation
of  the  chemical.to  a  critical concentration  at  sites  of action
sufficient to  induce detectable injury; by accumulation of injury
until physiological reserves can no longer compensate (i.e., repair
is never complete);  or after a long, latent period beginning with
an  exposure  that  has  an unrecognized  biological  effect  and
precipitates the eventual appearance of injury (National Academy of
Sciences,  1977).   Obviously,  sufficient  duration of exposure is
necessary  in  order for  the  effects seen  in chronic toxicity to
become manifest.    Subchronic  toxicology  studies may  not offer
reliable  means .for  assessment  of  long-term  toxic effects  in
animals,   let  along  extrapolation  to  chronic   effects  in  man
 (National Academy  of Sciences,  1977).   However,  it is  often the
case that a good quality, chronic exposure study  for a particular
chemical  is  unavailable.   The  intention  of   this  additional
uncertainty factor  is to enable the use  of  subchronic  or flawed
studies to protect against the  risk of  adverse  effects which might
only appear with chronic dosing.
    Experimental  support  for  the additional uncertainty factor is
given by  literature reviews  which  compare subchronic  NOAELs and
chronic  NOAELs  for many compounds   (McNamara,   1971;  Weil  and
McCollister,  1963).   The studies reviewed by those investigators
employed a variety of rodent  and non-rodent species.  The duration
of the subchronic exposures was usually 90 days, but  ranged from 30
to 210 days.  Wide variations in endpoints and criteria for adverse
effects were  encountered in  these  literature  reviews.   However,
their findings do give a rough indication of the general subchronic
and  chronic  NOAELs  for  other  than  carcinogenic  or reproductive
effects.   For over 50% of the compounds tested, the chronic NOAEL
was less  than the 90-day  NOAEL by a factor of 2  or less.  There was
some indication that chronic dosing may result in the development
of tolerance  toward certain  chemicals, as the chronic  NOAEL was
larger than the 90-day NOAEL  in a few cases.  However, it was also
found that the chronic NOAEL may be less than the 90-day NOAEL by
a  factor  of  10  or more.   The  latter situation appeared  to be
uncommon.  Therefore, these reviews report that the additional ID-

                                AS

-------
fold uncertainty  factor  appears to be adequate  or incorporate a
margin of safety in the majority of cases.
   As  the literature  reviews  by  McNamara  (1971)  and  Weil  .and
McCollister (1963) are limited and  the studies reviewed utilized a
variety of toxicologic endpoints with questionable sensitivities,
one must be cautious  in  interpreting their conclusions.   But for
lack of data to -the contrary,  it appears that  application of the
additional 10-fold uncertainty factor is  appropriate and justified
when extrapolating a NOAEL from a 90-day study to a chronic NOAEL
estimate.  This practice may underestimate the true chronic NOAEL
far more  often than overestimating it,  thus adding a  margin of
safety to the risk calculations.
   One remaining question regarding exposure duration is:  At what
point is the duration considered adequate, such that the additional
uncertainty factor of  up  to 10 is unnecessary? In other words, how
is "chronic" defined for the sake of this guideline?
   At  this  point,  further  discussion  of  the  terms  "chronic"
"subchronic", and  "subacute",  is necessary.  The term "subacute"
has been used to describe a duration less  than subchronic, while it
has also been used as a term analogous to subchronic.  EPA  (1980)
describes  "subacute"   exposures (in  this  case,   analogously  to
"subchronic") as often exceeding 10% of the lifespan,  e.g., 90 days
for the  rat  with  an average lifespan of  30  months.   However,  as
pointed  out  by  the  Organization  for  Economic  Cooperation  and
Development  (OECD,  1981),  the term  "subacute"  is  semantically
incorrect. The OECD prefers to use  the phrase "short-term repeated
dose  studies",  referring  to   14,   21  and  28   day  studies,  to
distinguish from  "subchronic" studies of greater duration.
   "Subchronic" is generally defined as part of  the  lifespan  of the
test species, although opinions differ on the precise definition.
Klaassen  (1986)   defines  "subacute" as   repeated  exposure to  a
chemical  for one  month or  less,  and   "subchronic" as  repeated
exposure for 1-3 months.  Chan et al. (1982) describe "subchronic"
exposure  durations  as generally ranging from 1 to 3  months  in
rodents and one year  in  longer-lived  animals (dogs,  monkeys),  or
for part  (not  exceeding  10%)  of the lifespan.   Stevens and Gallo
(1982) define "long-term toxicity tests"  (encompassing subchronic
and chronic toxicity  studies)  as studies of longer than 3 months
duration, i.e., greater than 10% of the lifespan  in the laboratory
rat.     EPA  (1985)  describes  "subchronic"  toxicity testing  as
involving continuous or repeated exposure for a period of 90 days,
or approximately.  10% of the lifespan for  rats.
   The  various definitions  offered for  "chronic"  are generally
inconsistent.    Klaassen  (1986)  defines  "chronic"  as  repeated
exposure for more than 3 months.  According to the National Academy
of  Sciences   (1977),  chronic  exposure   in  animals  is  generally
considered  to  be  at  least  half  the life  span.   In  estimating
chronic  SNARLs,  the  National Academy of Sciences  (1980)  in most
cases utilized data from studies lasting a "major portion of the
lifetime  of  the  experimental  animal".    According  to  the EPA's
Health  Effects  Testing Guidelines  (EPA,  1985),  chronic toxicity
tests should involve  dosing  over a period of at  least 12 months.

                                A6

-------
The application of their guidelines, they add,  should generate data
on which  to identify the  majority of chronic  effects  and shall
serve to define  long-term  dose-response  relationships.   The OECD
 (1981)  states  that the  division between subchronic  and chronic
dosing regimes is sometimes taken as 10%  of  the  test animal's life
span.  They also state that the duration of the exposure period for
chronic toxicity studies  should be  at  least  12  months.   They
describe "chronic" as prolonged  and repeated exposure capable of
identifying the majority of chronic effects  and  to determine dose-
response relationships.
   Others have investigated the delayed appearance of toxic effects
which might  be missed under  shorter dosing  regimes.   Frederick
 (1986)  conducted  a  pilot  survey  of  new  drug evaluators  for
incidences  of  delayed  (greater  than  12  month)   drug-induced
pathology.    It  was  concluded  that  new  toxic  effects  "not
infrequently" arise after one year of dosing in rodents.   It was
further stated  that  those  findings  formed  the basis  for  the
conclusion of the Bureau of Human Prescription Drugs:  the duration
of the long-term-toxicity tests of drugs that are likely  to be used
in man  for more  than  a  few days  should be at  least  18 months.
Glocklin (1986) reviewed the issues regarding testing requirements
for new drugs,  and concluded that 12 month chronic toxicity studies
seemed to  be an appropriate requirement for characterization of the
dose-response.
   It is  evident that  there are discrepancies in the qualitative
and quantitative characterization of "chronic" animals  studies.  An
appropriate and reasonable working definition for "chronic" would
appear to be at 'least half  the  life span (therefore,  at least 52
weeks for  rats  and at  least 45  weeks  for mice) .  Qualitatively,
"chronic"   means  that  the  exposure duration  was  sufficient  to
represent  a full  lifetime  exposure,  in terms  of  dose-response
relationships.   For example, a  study providing an experimental
NOAEL which approximates a lifetime NOAEL is considered a chronic
study.  It  is recognized that the above quantitative definition (at
least half  the life span)  does  not  demonstrate the  flexibility
inherent in the  above  qualitative  description.   That flexibility
reflects  the  vast  differences   in  the toxicology  of  various
chemicals:  demonstration of a  lifetime  NOAEL for some chemicals
may require dosing for  half the life span, while  the toxicology of
most chemicals may allow demonstration of a  lifetime NOAEL under a
much shorter dosing  regime.   It may  be  argued  that the lifetime
NOAEL  for  noncarcinogenic  effects  of   many   chemicals  can  be
demonstrated in rodent studies of much less than one year.  While
the previously-discussed  works   of  McNamara (1971)  and  Weil  and
McCollister (1963) support  that  view,  they  also demonstrate that
the chronic NOAEL may be less  than  the 90-day NOAEL by a factor of
10 or more, for some chemicals.
   This discussion is necessary in order  to  properly interpret the
uncertainty factor guideline, which recommends that the additional
uncertainty  factor of  up  to  10  be applied  when  the  exposure
duration is less than  "chronic".   The intent  of the uncertainty
factor is to adjust the experimental NOAEL to a lifetime NOAEL in

                               A7

-------
those cases where the  lifetime NOAEL was presumably not adequately
demonstrated.   The key  issues  are summarized  in the  following
points and recommendations:

   a.  An  acceptable  quantitative  definition  of  "chronic"  is
       elusive.   Due  to  differing toxicological  properties, .the
      'necessary  minimum exposure  duration  to  demonstrate  a
       lifetime   NOAEL  differs   widely   among  .chemicals.     A
       qualitative,   philosophical  definition   of   chronic   is:
       "Chronic" is when the exposure duration  is sufficient for
       the identification of the majority of long-term effects and
       their dose-response relationships.   Therefore,  a "chronic"
       study reporting a  NOAEL is  one which  can be  reasonably
       presumed  to predict the lifetime NOAEL.

   b.  The  use  of  scientific  judgment   is  predominant  in  the
       decision  of  when  chronic  exposure  conditions exist,  and
       hence,  when the additional  uncertainty factor  is no longer
       appropriate.

   c.  That scientific judgment should be guided  by a review of all
       available    pertinent    data,    e.g.,    metabolism,
       pharmacokinetics,  bioaccumulation,  mechanism  of  action,
       target organ characteristics, potential for latent effects,
       etc.

   d.  Available'reviews of rodent studies indicate that, for many
       chemicals,  studies of much  less than one  year  duration can
       provide reasonable estimates  of lifetime  NOAELs.   However,
       it is also recognized that the toxicological characteristics
       of  some  • chemicals   will  prevent  the   qualitative   and
       quantitative demonstration  of latent adverse effects and a
       lifetime  NOAEL if the duration is less than one  year.   If
       the lack  of  additional data prevents scientific judgment in
       these cases, 50%  of  the lifespan  (52 weeks for  rats;  45
       weeks for mice)  may be  considered the  minimum necessary
       duration  for  a "chronic"  exposure.   Application  of  the
       additional   uncertainty   factor   for   these   apparently
       "subchronic" studies may later provide  to  be excessively
       conservative in  some  cases.   But,  if the  toxicologic
       database  is inadequate, the  additional uncertainty factor
       should be  included,  both  as a matter  of prudent  public
       policy and  as an incentive  to  others   to  generate  the
       appropriate data.

   e.  Ordinarily,  the additional 10-fold factor may be applied for
       all  rodent  studies  of  90  days  duration, unless  there is
       chemical-specific data  indicating that would be unnecessary
       and overly conservative.

   f.  For  rodent   studies  of  between  90  days and  12  months
       duration, the  use of  the  additional  10-fold uncertainty

                                A3

-------
       factor is best  determined by  professional judgment.   As
       described above,  if data are not available to sufficiently
       guide professional  judgment,  then  such  studies  may. be
       subject  to part.or all of the additional 10-fold factor.  A
       "sliding scale" or between 1 and 10 is a reasonable means of
       selecting a  lesser factor when 10 appears excessive.  Under
       this concept,  the additional uncertainty factor applied may
       vary on  a scale  of  one to ten according to how closely the
       dosing duration approached  50% of the lifespan.  Of course,
       consideration must be given  of  the study  quality  and the
       other pertinent data  mentioned  in 3.c  above.   A 90-day
       rodent  study  would be  subject  to  a  10-fold  additional
       factor,  if  study  quality  is  otherwise nominal  and  other
       chemical-specific  data  are  lacking.   A  nominal-quality
       study, with  exposure  over 50%  of  the  lifespan, would be
       subject   to   a   "1",   i.e.,   no   additional   adjustment.
       Situations where the exposure duration is  between  90 days
       and 50% of  the  lifespan,  and/or study quality  is  flawed,
       must be handled  on a case-by-case basis.  This "sliding
       scale" concept may offer guidance to the scientific judgment
       that will be necessary.

   Dosing  duration is but  one  parameter upon  which to assess the
adequacy of  a  study.  Other  deficiencies  in the study design may
cause increased concern about the validity of the reported NOAEL or
LOAEL.  Therefore,  risk assessors may utilize  part or all of this
additional 10-fold uncertainty  factor to compensate for data which
appears less-than-adequate. Factors which may  affect  the degree of
confidence  in  the data  include  the number of  animals per dose
group, the  sensitivity and appropriateness of  the endpoints, the
quality  of  the  control  group,  the exposure  route,  the  dosing
schedule,  the   age  and  sex  of  the  exposed   animals,  and  the
appropriateness  of the surrogate  species tested, among  others.
EPA's  Health  Effects  Testing Guidelines (EPA,  1985)  provide
specific information on the desirable qualities of subchronic and
chronic toxicity' tests.

An additional uncertainty factor of between 1 and 10 is  recommended
depending  on the severity and sensitivity of the adverse effect
when extrapolating from a LOAEL rather than a  NOAEL.        This
uncertainty  factor  reduces the LOAEL into the  range of a NOAEL,
according  to  comparisons of  LOAELs  and NOAELs   for  specific
chemicals.   There  is evidence  available which  indicates,  for a
select set of chemicals,  96%  have  LOAEL/NOAEL ratios  of 5 or less,
and that all are'10 or less (Dourson and Stara, 1983) .  In practice
the value for this variable uncertainty factor has been chosen by
the U.S. EPA from values among 1 through 10 based on the severity
and sensitivity of the adverse effect of the LOAEL.  For example,
if the  LOAEL represents liver cell  necrosis,  a  higher value is
suggested for this uncertainty factor  (perhaps 10).  If the LOAEL
is fatty  infiltration  of  the liver  (less  severe  than liver cell
necrosis),  then  a  lower value is suggested (perhaps  3;  see the

                               A9

-------
following discussion).   The hypothesized NOAEL should be closer to
the LOAEL showing less severe effects (Dourson and Stara,  1983) .
   In some cases the data do not completely fulfill the conditions
for  one  category  of .the  above  guidelines,  and  appear to  be
intermediate  between  two  categories.    Although  one  order  of
magnitude  is  generally  the smallest  unit  of  accuracy  that  is
reasonable for uncertainty  factors,  an  intermediate value may be
used if felt necessary  (Dourson,  1987).  According to EPA (1980),
such an intermediate uncertainty factor may be developed based on
a logarithmic scale rather  than a  linear  scale.   Calculating the
mean logarithmically may  be the more appropriate option,  because
the precision of all uncertainty factor estimates is poor,  and a
logarithmic scale  is  the best  way to  estimate  the mean  of two
imprecise estimates (Dourson, 1987).   Halfway between l and 10 is
approximately 3.16 on a logarithmic scale.  However, so as not to
imply excessive accuracy in  the estimate, that mean value should be
rounded-off to 3 (Dourson, 1987).
   An additional uncertainty factor of up to  10 may be applied when
there are limited or incomplete subacute or chronic toxicity data.
such as  with short-term repeated dose animal studies  where the
exposure regime involves a limited period that is markedly short-
term    relative to  the lifespan of the  test species  (e.g.. 28-
day rodent NOAEL).  As previously noted  (see  C.3)  the OECD (1981)
distinguishes between  14,  21 or 28 day studies  and "subchronic"
studies of greater duration, by referring  to the former as "short-
term repeated dose studies".  The short-term studies are commonly
conducted  by  the  NTP   to enable  appropriate  dose  selection  in
subchronic studies (NCI,  1976).  When a limited database exists,
short-term animal studies of 28 days or longer may be of sufficient
quality to support risk assessment of potential chronic exposure.
Because the duration of  exposure is substantially less than the 90-
day period discussed under C.3, the risk assessment may require an
additional uncertainty  factor in conjunction with  the  1000-fold
factor recommended under C.3.  As guidance, an additional factor of
up to 10 is recommended when extrapolating from a  short-term NOAEL
(e.g., 28 days) to subchronic duration  (e.g., 90 days).
   Although  the. extrapolation  from oral LDcns   to  chronic  oral
NOAELs  has been reported by several  investigators  (Venman and
Flaga, 1985; Layton et al., 1987;  McNamara,  1971), there has been
relatively little  investigation of  the extrapolation from short-
term NOAELs (much less  than 90 days in rodents) to chronic NOAELs.
EPA  (1989) states  that  when experimental  data  are available only
for shorter durations than desired for subchronic  RfD derivation an
additional uncertainty factor is applied.  However, further details
on the selection of an  adequate and appropriate uncertainty factor
for those "shorter durations" are not provided.  Weil et al. (1969)
evaluated the relationship between 7-day, 90-day and  2-year minimum
effect levels (MiE) for 20 materials via feed exposure.  They found
that the median value for a  90-day MiE was  obtained by dividing the
7-day MiE by a factor of 3.  The 95th percentile for  the 90-day MiE
was obtained by dividing the 7-day  MiE by  6.2.  Also noteworthy is


                               A10

-------
the  finding that  the  95th  percentile  for the  2-year MiE  was
obtained by dividing the 7-day MiE by a factor of 35.3.
   These data, Albeit limited, support the general principle that
as  exposure duration,  decreases,  the  ability  of  the  data  to
demonstrate chronic  dose-response relationships  also decreases.
While an additional 10-fold uncertainty factor may reasonably and
appropriately convert a 90-day NOAEL  to a surrogate chronic NOAEL,
an   additional   uncertainty   factor  may   be ,  necessary   when
extrapolating from short-term  exposures.   Applying an additional
uncertainty factor  of up  to  10 will help  ensure that  the risk
assessment  for   potential   chronic  exposures   is  adequately
conservative,  i.e., the true  chronic NOAEL  will generally not be
overestimated.
                               All

-------
                            REFERENCES

Bigwood, E.   1973.  The Acceptable Daily Intake of Food Additivies.
   CRC  Grit. Rev. Toxicol. June 41-93.  As cited in:  Dourson, M.
   and  J.  Stara.    1983.   Regulatory History and Experimental
   Support of Uncertainty (Safety)  Factors.  Regulatory Toxicology
   and  Pharmacology.  3:224-238.

Calabrese,  E.  1985.  Uncertainty Factors and Interindividual
   Variation.  Regulatory Toxicology and Pharmacology.  5:190-196.

Chan, P., O'Hara, G. and A. Hayes.  1982.   Principles and Methods
   for  Acute and Subchronic Toxicity.   In:  Hayes, A. (Ed.) 1982.
   Principles and Methods of Toxicology.  1982.  Raven Press.  New
   York, NY.

Dourson, M.   1987.  U.S. EPA.  Environmental Criteria and
   Assessment Office.   Personal  communication  with Robert Sills,
   Michigan Department  of Natural  Resources.

Dourson, M.  and J. Stara.  1983.   Regulatory History and
   Experimental   Support   of   Uncertainty    (Safety)   Factors.
   Regulatory Toxicology and  Pharmacology.  3:224-238.

Frederick,  G.  1986.  The Evidence Supporting 18 Month Animal
   Studies. In:  Walker, S. and A.  Dayan.  1986.  Long-Term Animal
   Studies;  Their Predictive Value for Man:   Proceedings of the
   Centre  for  Medicines  Research  Workshop  held  at  the  Ciba
   Foundation, London,  2 October  1984.

Glocklin, V.  1986.  Justification for 12 Month Animal Studies.
   In:  Walker, S. and A. Dayan.  1986.  Long-Term Animal Studies;
   Their Predictive  Value for Man:  Proceedings of the Centre for
   Medicines Research Workshop held at the Ciba Foundation, London,
   2 October 1984.

Klaassen, C.  1986.  Principles of Toxicology.   In Doull, J.,
   Klaassen, C. and  M.  Amdur  (Eds.).   1986.  Casarett and Doull's
   Toxicology  -  The Basin  Science  of Poisons.    3rd  Edition.
   Macmillan Publishing Co.,  Inc.  New  York, NY.

Layton, D.W., et al.  1987.  Deriving allowable daily intakes for
   systemic  toxicants  lacking chronic toxicity data.  Regulatory
   Toxicology and Pharmacology.   7:96-112.

McNamara, B., 1971.  Concepts in Health Evaluation of Commercial
   and  Industrial Chemicals.  In:  Mehlman, M.,  Shapiro, R. and H.
   Blumenthal  (Eds.).   1971.   Advances  in Modern Toxicology.
   Volume 1, Part  1:  New Concepts in  Safety Evaluation.  Chapter
   4.   John Wiley and  Sons. New  York,  NY.

National Academy of  Sciences.  1980.  Drinking Water and Health.

                               A12

-------
   Vol. 2.  National Academy  Press. Washington, D.C.

National Academy of Sciences.  1977.  Drinking Water and Health.
   Vol. 1.  National Academy  Press. Washington, D.C.

National Cancer Institute.  1976.  Guidelines for Carcinogen
   Bioassay in Small Rodents.  U.S.  DHEW.  Technical Report Series
   No.  1.

Organization for Economic Cooperation and Development  (OECD).
   1981.  OECD Guidelines for  Testing of Chemicals.   Section 4:
   Health Effects.  Paris, France.

Stevens, K. and M. Gallo.  1982.  Practical Considerations in the
   Conduct of Chronic Toxicity Studies.  In:  Hayes, A.  (Ed.) 1982.
   Principles and Methods of Toxicology.  1982.  Raven Press. New
   York, NY.

U.S.  Environmental Protection Agency  (EPA).  1986.  Human
   Variability   in   Susceptibility  to  Toxic  Chemicals   -   I.
   Noncarcinogens.  EPA/600/8-86/033.

U.S.  Environmental Protection Agency (EPA).   1985.  Health Effects
   Testing Guidelines.  40 CFR Part 798.  Federal Register, v. 50,
   n. 188, September 27,  1985.

-------
   Vol. 2.  National Academy  Press. Washington,  D.C.

National Academy of Sciences.  1977.  Drinking Water and Health.
   Vol. 1.  National Academy  Press. Washington,  D.C.

National Cancer Institute.  1976.  Guidelines for Carcinogen
   Bioassay in Small Rodents.  U.S.  DHEW.  Technical Report Series
   No. 1.

Organization for Economic Cooperation and Development  (OECD).
   1981.  OECD Guidelines for  Testing of Chemicals.   Section 4:
   Health Effects.  Paris,  France.

Stevens,  K. and M. Gallo.  1982.  Practical Considerations in the
   Conduct of Chronic Toxicity Studies.  In:  Hayes, A.  (Ed.) 1982.
   Principles and Methods of Toxicology.  1982.  Raven Press. New
   York, NY.

U.S. Environmental Protection Agency  (EPA) .  1986.  Human
   Variability   in   Susceptibility  to   Toxic   Chemicals   -   I.
   Noncarcinogens.  EPA/600/8-86/033.

U.S. Environmental Protection Agency (EPA).   1985.  Health Effects
   Testing Guidelines.  40 CFR Part 798.   Federal Register, v. 50,
   n. 188, September 27, 1985.

U.S. Environmental Protection Agency  (EPA).  1980.  Water Quality
   Criteria Documents:  Availability.   Appendix C - Guidelines and
   Methodology Used in the Preparation of  Health Effect Assessment
   Chapters  of  the  Consent  Decree  Water Criteria  Documents.
   Federal Register,  v. 45,  n.  231,  November  38, 1980.   79347-
   79357.

Venman,  B. and C-. Flaga.  1985.  Development of an Acceptable
   Factor to Estimate Chronic End Points  from Acute Toxicity Data.
   Toxicology and Industrial  Health.  1(4):  261-269.

Vettorazzi, G.  1976.   Safety Factors  and  their Application in the
   Toxicological Evaluation.  In:  The Evaluation of Toxicological
   Data for  the Protection of Public health.   Permagon,  Oxford.
   As  cited  in:   Douson,  M.  and J. Stara.    1983.   Regulatory
   History  and  Experimental  Support  of  Uncertainty  (Safety)
   Factors.  Regulatory Toxicology and Pharmacology.  3:  224-238.

Weil, C.  and D. McCollister.  1963.  Relationship Between Short-
   and Long-Term Feeding Studies  in Designing an Effective Toxicity
   Test.  Agricultural  and  Food  Chemistry.   11(6):  486-491.
                               A13
                                                                   Agency

-------