ORNL
Oak Ridge
National
Laboratory
Operated by
Union Carbide Corporation
for the Department of Energy
Oak Ridge, Tennessee 37830
ORNL/EIS-151
&EPA
United States
Environmental Protection
Agency
Office of Pesticides and
Toxic Substances
Washington, DC 20460
EPA-560/1 -80-001
December 1980
            Pesticides and Toxic Substances
   SCIENTIFIC RATIONALE FOR THE
   SELECTION OF TOXICITY TESTING
   METHODS: HUMAN HEALTH ASSESSMENT

-------
                                                    ORNL/EIS-151
                                                    EPA-560/1-80-001
                   Contract No.  W-7405-eng-26
                      Information Division
       SCIENTIFIC RATIONALE FOR THE SELECTION OF TOXICITY
            TESTING METHODS:  HUMAN HEALTH ASSESSMENT
       Robert H. Ross, Michael G. Ryon, Mary W.  Daugherty,
     John S. Drury, John T. Ensminger, and M. Virginia Cone

            Health and Environmental Studies Program
                   Information Center Complex
Work sponsored by the Office of Pesticides and Toxic Substances,
U.S. Environmental Protection Agency, Washington, D.C., under
Interagency Agreement No. 80-D-X0856.
                        Project Officers

                      Norbert Page, D.V.M.
                  Daljit Sawhney, D.V.M., Ph.D.
                 Date Published - December 1980
                  OAK RIDGE NATIONAL LABORATORY
                   Oak Ridge, Tennessee 37830
                           operated by
                    UNION CARBIDE CORPORATION
                             for the
                      DEPARTMENT OF ENERGY

-------
     This report was prepared as an account of work sponsored by an agency
of the United States Government.  Neither the United States Government nor
any agency thereof, nor any of their employees, contractors, subcontrac-
tors, or their employees, makes any warranty, express or implied, nor
assumes any legal liability or responsibility for any third party's use
or the results of such use of any information, apparatus, product or
process disclosed in this report, nor represents that its use by such
third party would not infringe privately owned rights.

     This report has been reviewed by the Office of Pesticides and Toxic
Substances, U.S. Environmental Protection Agency, and approved for publi-
cation.  Approval does not signify that the contents necessarily reflect
the views and policies of the U.S. Environmental Protection Agency, nor
does mention of trade names or commercial products constitute endorsement
or recommendation of use.
                                  ii

-------
                                CONTENTS
Figures	    vii
Tables	     ix
Foreword	xiii
Acknowledgments 	     xv
Abstract	xvii
1.  Summary	      1
    1.1  Basic Experimental Considerations  	      1
    1.2  Acute Toxicity Testing 	      2
    1.3  Subchronic Toxicity Testing  .....  	      2
    1.4  Chronic Toxicity and Carcinogenicity Testing 	      4
2.  Basic Experimental Considerations 	      7
    2.1  Introduction 	      7
    2.2  Test Materials	      7
         2.2.1  Introduction  	      7
         2.2.2  Chemical Stability  	      8
         2.2.3  Chemical Impurities 	      8
         2.2.4  Vehicles	      9
         2.2.5  Homogeneity in Vehicle	     10
         2.2.6  Conclusions	     10
    2.3  Husbandry	     10
         2.3.1  Introduction	     10
         2.3.2  Animal Selection	     11
         2.3.3  Transportation, Quarantine, and Disease
                  Control	     12
         2.3.4  Design of Testing Facility  	     13
         2.3.5  Caging	     14
         2.3.6  Bedding	     15
         2.3.7  Temperature and Humidity	     18
         2.3.8  Light	     19
         2.3.9  Ventilation	     19
         2.3.10 Noise and Handling  	     21
         2.3.11 Personnel 	     22
         2.3.12 Conclusions 	     22
    2.4  Diet	     23
         2.4.1  Introduction	     23
         2.4.2  Dietary Requirements for Laboratory Animals ...     24
         2.4.3  Types of Diets for Laboratory Animals 	     26
         2.4.4  Analysis for Nutrients and Contaminants 	     31
         2.4.5  Effects of Diet on Toxicity Test Results  ....     33
         2.4.6  Conclusions	     35
    2.5  Pathology	     36
         2.5.1  Introduction	    36
         2.5.2  Gross Examination 	     36
         2.5.3  Tissue Preservation and Storage 	    40
         2.5.4  Trimming, Staining, and Embedding of Tissue ...    41
         2.5.5  Microscopic Examination 	    42
         2.5.6  Conclusions	    45
                                   iii

-------
    2.6  Pharmacokinetics 	     45
         2.6.1  Introduction	     45
         2.6.2  Basic Concepts	     46
         2.6.3  Utility of Pharmacokinetics 	     48
         2.6.4  Conclusions	     50
3.  Acute Toxicity	     59
    3.1  Introduction	     59
    3.2  LD5Q Determinations	     59
         3.2.1  Introduction	     59
         3.2.2  Difference Between Species  	     63
         3.2.3  Difference Between Administration Routes  ....     67
         3.2.4  Sex Differences in the Laboratory Rat	     70
         3.2.5  Test-Limiting Criteria  	     71
         3.2.6  Conclusions	     71
    3.3  Human vs Animal Response	     72
         3.3.1  Introduction	     72
         3.3.2  Comparison of Lowest Published Lethal Doses
                  0V	     72
         3.3.3  Comparison of Acute Toxicity Response 	     75
         3.3.4  Conclusions	     76
    3.4  Pathology	     77
         3.4.1  Introduction	     77
         3.4.2  Histopathology	     77
         3.4.3  Conclusions	     80
    3.5  Observation Period 	     80
         3.5.1  Introduction	     80
         3.5.2  Purpose of Observation Period 	     84
         3.5.3  Length of Observation Period  	     84
         3.5.4  Conclusions	     86
4.  Subchronic Test Design	     93
    4.1  Introduction	     93
    4.2  Species	     95
         4.2.1  Introduction	     95
         4.2.2  Discussions in Literature Reviews Concerning
                  Species Suitability 	     95
         4.2.3  Species Comparison Studies  	    101
         4.2.4  Conclusions	    121
    4.3  Duration	    123
         4.3.1  Introduction	    123
         4.3.2  Duration Reviews  	    124
         4.3.3  Comparison Summaries  	    130
         4.3.4  Conclusions	    150
    4.4  Route of Exposure	    151
         4.4.1  Introduction	    151
         4.4.2  Route  Discussions  	    152
         4.4.3  Route  Comparisons  	    157
         4.4.4  Conclusions	    168
    4.5  Pathology	    168
         4.5.1  Introduction	    168
         4.5.2  Use  of Pathology	    170
         4.5.3  Basis  for a  Minimum Pathology Screen	    180
         4.5.4  Conclusions	    182
                                   iv

-------
    4.6  Clinical Laboratory Tests   	    184
         4.6.1  Introduction	    184
         4.6.2  General Clinical  Testing   	    185
         4.6.3  Hematology	    188
         4.6.4  Biochemical and Organ Function   	    192
         4.6.5  Urinalysis	    198
         4.6.6  Conclusions	    202
         Appendix A	    217
         Appendix B	    229
         Appendix C	    237
5.  Chronic Toxicity and Carcinogenicity  Testing  	    265
    5.1  Introduction	    265
    5.2  Test Animals	    266
         5.2.1  Species	    266
         5.2.2  Strain	    288
         5.2.3  Spontaneous Tumors   	    294
         5.2.4  Number	    300
         5.2.5  Controls	    306
         5.2.6  Age	    308
         5.2.7  Sex	    311
         5.2.8  Conclusions	    320
    5.3  Routes of Administration	    323
         5.3.1  Exposure Routes	    324
         5.3.2  Carcinogen Exposure by Different Routes 	    329
         5.3.3  Conclusions	    340
    5.4  Dose and Duration	    340
    5.5  Interim Sacrifice	    349
    5.6  Data Collection and Evaluation	    350
         5.6.1  Food Consumption and Body Weight	    350
         5.6.2  Clinical and Laboratory Examinations  	    351
         5.6.3  Pathological Examinations 	    352
         5.6.4  Conclusions	    354
    5.7  Short-Term Tests for Carcinogenicity 	    356
         5.7.1  Embryo Homograft  	    356
         5.7.2  Site Transfer	    357
         5.7.3  Partial Hepatectomy 	    358
         5.7.4  Alkaline Elution  	    360
         5.7.5  a-Fetoprotein in Serum	    361
         5.7.6  Strain Susceptibility 	    362
         5.7.7  The Sebaceous Gland Test	    364
         5.7.8  Host-Mediated In Vivo-In Vitro Assay  	    364
         5.7.9  Conclusions	    365

-------
                                FIGURES
2.1  Hydraulic analogy of a three-compartment body model ....   47

4.1  Changes in the mean body weight of rats fed Sumithion in
       the diet for 90 days	133

4.2  Changes in cholinesterase activity in the kidney, liver,
       red blood cells, and brain cortex of rats fed Sumithion
       for 90 days	135
                                  vii

-------
TABLES
2.1
2.2
2.3

2.4
2.5
2.6
2.7
2.8
2.9

2.10

3.1

3.2
3.3

3.4

3.5
3.6

3.7
4.1

4.2

4.3

4.4
4.5

4.6

4.7
4.8
Space recommendations for laboratory animals 	
Cage space requirements for rodents . . 	 	
Requirements for essential amino acids expressed as
percentage of protein 	 	 	 	
NIH-7 open formula rat and mouse ration 	
AIM-76® purified diet (for rats and mice) 	
AIH-7^8 vitamin mixture 	
MH-76® mineral mixture 	
Tissue to be included in a gross examination 	
Correlation between gross and microscopic lesions in
carcinogenic studies in mice .... 	
Organs and tissues to be examined in routine toxic ity
tests ... 	
Compounds examined for LD50 differences between species
and between routes of administration 	
Percent LDsg differences between species 	
Percent U>5o differences between routes of
administration 	 . 	
Compounds selected for human vs animal LD
comparisons . . 	
LD_ differences between humans and animals 	
Lo
Examples of histopathological changes from acute exposures
to chemicals ..... 	
LDso observation periods 	
Experimental design for recently proposed subchronic oral
toxic ity tests 	
Ranking of various animal models as predictors of
metabolic fate in man 	
Occurrence of 39 physical signs from six drugs in three
species 	
Organ system toxic it ies 	
Comparison of significant variations in the response of
four species to an oral analgesic 	
Predictive abilities of the dog, monkey, and
combination 	
Summary of dioxin oral administration data 	
Summary of biological effects of TCDD 	
16
17

25
28
29
30
31
38

39

43

61
64

68

73
74

78
81

96

98

103
104

108

110
114
116
  ix

-------
 4.9   Summary of treatment-related effects in hamsters,  rats,
         and rabbits repeatedly exposed to acrolein (ppm) for
         13 weeks	120

 4.10  Relationship of dosage levels of short-term and 2-year
         feeding of materials in the diets of rats	125

 4.11  Parameters of ratios of acute peroral LDsgs, 7- or
         90-day and 2-year minimum effect (MiE) dosage levels .  .  .   126

 4.12  Prediction formulas  	   127

 4.13  Approximate duration of drug administration required to
         define toxicity in animals 	   128

 4.14  Prediction of long-term no-effect doses  	   129

 4.15  Urinary ascorbic acid excretion and relative liver and
         adrenal weights of rats fed butylated hydroxyanisole
         (BHA) or butylated hydroxytoluene (BHT)  at 0.1%  of
         the diet for 16 weeks	131
 4.16  Mean values of body weights, food consumption,  and
         dibutyl(diethylene glycol bisphthalate)  (DDGB)  intake
         of rats fed DDGB at 0% to 2.5% of the diet for  11
         weeks	132

 4.17  Mean testes and brain weights,  as percentage of body
         weight,  of control rats and rats fed a dietary  level
         of 500 ppm of Sumithion for various periods of  time  .  .  .   134

 4.18  Cumulative toxicity of epichlorohydrin:   body weight
         gain in grams (mean ± statistical error)  	   137

 4.19  Subacute toxicity of epichlorohydrin:   body weight gain
         in grams (mean ± statistical  error)   	   138

 4.20  Cumulative toxicity of epichlorohydrin:   percent organ
         weight to body weight of rats (mean ± statistical
         error)	139

 4.21  Subacute toxicity of epichlorohydrin:   percent  organ
         weight to body weight of rats (mean ± statistical
         error)	139

 4.22  Comparison of 13-week and 2-year 2,3,7,8-tetrachloro-
         dibenzo-p-dioxin (TCDD)  studies  	   140

 4.23   Weight  gain of  female rats receiving 31  daily doses of
         2,3,7,8-tetrachlorodibenzo-p-dioxin  (TCDD) 	   141

 4.24   Liver and  thymus weights  of  female rats  receiving  daily
         doses  of  2,3,7,8-tetrachlorodibenzo-p-dioxin  (TCDD)   .  .  .   142

 4.25  Mean body weights and water  intake of  rats  fed  diets
         containing  0%  to 2.0% di-(2-ethylhexyl)phthalate (DEHP)
         for up to 17 weeks	147
4.26  Mean hematological values  for rats fed diets containing
         0% to 2.0%  di-(2-ethylhexyl)phthalate  (DEHP)  for 2,  6,
        or 17 weeks	148

-------
4.27  Relative organ weights of rats fed diets containing
        0% to 2.0% di-(2-ethylhexyl)phthalate (DEHP)  for 2,
        6, or 17 weeks	149
4.28  Summary of chemical examples tested for toxicity by
        various routes of administration 	   158
4.29  Main toxic effects produced by oral or intravenous
        administration of A9-tetrahydrocannabinol  	   166
4.30  Postmortem studies 	   169
4.31  Range-finding data on subacute oral toxicity	171
4.32  Summary of observations of effects detected in short-
        term and two-year oral studies	173
4.33  Incidence (in percent) of drug-induced pathological
        changes encountered over a ten-year period in
        approximately 14 000 animals	176
4.34  Summary of histological quantification methods and
        their applications for chemically induced lesions  ....   178
4.35  Comparison of the usefulness of individual organs to
        indicate toxicity when pathologically examined in a
        subchronic study 	   181
4.36  Hematological studies during toxicity tests  	   190
4.37  Relative sensitivity of liver enzyme tests 	   197
5.1   Some factors in the experiments with MH 101 upon a
        variety of species	272
5.2   Induction of skin carcinomas in mice, hamsters, and
        rats with weekly applications of 9,10-dimethy1-
        1,2-benzanthracene 	   274
5.3   Tumor induction in various species with aromatic
        amines	277
5.4   Compounds, not previously listed, which have produced
        bladder cancer in the dog	280
5.5   The carcinogenic action of diethylnitrosamine  in
        different animal species  	  281
5.6   Tumor induction of various species with diethylnitro-
        samine and dimethylnitrosamine	,	283
5.7   Carcinogenicity of different forms of asbestos in
        various species  	  287
5.8  Chemicals or industrial processes  associated with  cancer
        induction in humans:  comparison of  target organs and
        main routes of exposure in animals and  humans	289
5.9   Spontaneous tumor incidence in  animals  used in the
        National Cancer Institute Bioassay Program  	  296
                                   xi

-------
5.10  Sex and age incidence of characteristic tumors of inbred
        strains	297

5.11  Spontaneous tumors in laboratory animals  	  301

5.12  Incidence of  tumors  in  treated groups required for
        significance  (P =  0.05) depending on experimental
        group size  and spontaneous tumors in controls	307

5.13  Systemic tumor  induction in mice with 3-methylcho-
        lanthrene by  the intragastric route  	  313

5.14  Influence of  sex on  response to carcinogens	314

5.15  Comparison of continuous versus intermittent  adminis-
        tration of  BZ»2HC1 on development of liver, Harderian
        gland, and  lung	319

5.16  Induction of  tumors  in  the Syrian hamster with diethyl-
        nitrosamine	330
5.17  Local tumor induction in mice with 3-methylcholanthrene
        by various  routes  of  administration  	  332
5.18  Incidence of  pulmonary  tumors in strain A mice after
        intravenous injection of methylcholanthrene 	  335

5.19  Incidence of  pulmonary  tumors in strain A mice after
        intravenous injection of 0.5 mg of methylcholanthrene
        or 0.5 mg of  dibenzanthracene	336

5.20  Incidence of  pulmonary  tumors in strain A mice given
        subcutaneously 0.5 mg of methylcholanthrene or 0.5 mg
        of dibenzanthracene dispersed in 0.5 cc of  horse serum
        and cholesterol	336

5.21  Tumor induction with aromatic amines by various routes
        of exposure	338

5.22  Summary of recommendations on dose selection	342

5.23  Time relationships among drug exposure, life  span, and
        time equivalents in man	347
                                  xii

-------
                                FOREWORD

     This report was prepared by the Health and Environmental Studies
Program (HESP),  Information Center Complex, Information Division,  Oak
Ridge National Laboratory, under an interagency agreement between the
U.S. Department of Energy and the U.S. Environmental Protection Agency
(EPA).   Under this agreement HESP is providing a variety of technical
information support services to the EPA Office of Testing and Evaluation,
including:  performing technical review and follow-up on chemical studies,
arranging workshops, writing reviews on health effects of specific
chemicals, and conducting structure-activity oriented reviews of chemicals
associated with mutagenic or teratogenic effects.
     This document is part one of a two-part literature analysis of
parameters associated with the various toxicity testing methods.  The
information contained herein has been compiled for the purpose of assisting
and supporting EPA in its efforts to develop guidelines for more efficient
and more economical toxicity testing procedures.
                                  xiii

-------
                            ACKNOWLEDGMENTS

     The authors would like to thank Halgo Cerstner, manager of the
Information Center Complex (ICC), Information Division, Oak Ridge National
Laboratory (ORNL), for her support during the preparation of this docu-
ment.  The advice and support of Drs.  Norbert Page and Daljit Sawhney,
U.S. Environmental Protection Agency (EPA) project officers, and the
assistance of Helen Warren and other members of the Toxicology Information
Response Center of the ICC are gratefully acknowledged.  Special thanks
are extended to Dr. Peter Witschi of the ORNL Biology Division, to Dr.
Guy Griffin of the ORNL Health and Safety Research Division, to Drs.
Harold Grlce and Clifford Chappel, FDC Consultants, Inc., and to Drs.
David Anderson, Salvatore Biacardi, William Butler, Larry Chltlik,
William Dykstra, Richard Fournier, Wayne Galbraith, Stan Gross, Robert
Jaeger, Steve Johnson, Raymond Landolf, Carl Morris, and James Murphy of
EPA for their technical review and helpful suggestions.  The authors are
also greatly indebted to Judy Crutcher, Sherry Hawthorne, Pat Hartman,
and Donna Stokes, of the ICC Publications Office, and to Carolyn Seaborn
for her assistance in the collection and organisation of data.
                                   xv

-------
                               ABSTRACT
     This document is the first of a two-part literature analysis of
parameters associated with the various toxicity testing methods (test
animal selection, pathology requirements, etc.)-  Acute, subchronic,
chronic, and carcinogenic testing methods are covered; a discussion of
some basic experimental considerations is also included.  This report
was prepared for the purpose of assisting and supporting the U.S. Environ-
mental Protection Agency in its efforts to develop guidelines for more
efficient and economical testing procedures.
                                  xvii

-------
                               1.   SUMMARY

1.1  BASIC EXPERIMENTAL CONSIDERATIONS

     To assist in the development  of  test standards,  this  section
addresses certain aspects of test  materials,  husbandry,  diet, patho-
logical examination,  and pharmacokinetics.
     The chemical and physical properties of  the test chemical  should  be
known prior to conducting a toxicity  test of  a chemical.   The chemical
and physical properties will influence  laboratory storage  practices
and will permit the researcher to  determine the stability  of the chemical
under the conditions of the experiment.   In addition, the  chemical
purity of the test material and possible interactions with the  vehicle
may be investigated.
     Husbandry conditions associated with toxicity testing can  affect
the test results and should be controlled or standardized.  Included
among the husbandry factors are animal  selection, transportation,
quarantine, disease control, cage design,  test facility design, regula-
tion of macroclimate and microclimate variables, and experienced
technical personnel.
     Nutrients required by laboratory animals are supplied through
various types of diets.  Variations in the concentration of essential
nutrients and the presence of contaminants in the diet may affect the
response of test animals to toxic substances.  Periodic testing of diets
for nutrient concentrations and contaminants has been proposed.
     Pathology is a very essential part of the evaluation of a chemical's
toxicity, providing information on the nature of the lesions and  indicat-
ing dose-effect relationships.  A successful pathology evaluation should
include the following:   (1) the pathologist should perform  or  supervise
all steps in the protocol;  (2) the gross necropsy should be extremely
thorough and include every animal in the study;  (3)  the tissues  should
be properly preserved  in an accepted fixative  immediately after  the
necropsy; (4) the staining, embedding, and sectioning of  the tissues
must be well planned and coordinated, especially if  special stains are
utilized; and  (5) the  microscopic examination should be as  thorough as
practical limitations  will  allow.

-------
     The pharmacokineti.es of a chemical, which is influenced by four
major rate processes — absorption, distribution, metabolism, and elimin-
ation — can  provide valuable information to the researcher.  The results
of  pharmacokinetic studies  can help decide the future testing protocol
for a chemical  as well as explain why a chemical can be teratogenic when
administered during one  part of  the gestation period but not teratogenic
when administered in another part of the gestation period.

1.2 ACUTE TOXICITY TESTING

     Acute toxicity studies are  very useful because they provide base-
line information such  as the LDsg  (the most frequently determined index
of  toxicity), the relative  sensitivity of the various species, and the
comparative  toxicity of  various  chemicals.  They also identify the
nature  of the toxicity or the mode of the toxic action and provide
guidance on  doses to be  used in  subsequent experiments.  Administration
may be  a single oral or  single parenteral injection or may be given in
food or drinking water over a 24-h period.  When the laboratory rat is
used as the  test animal, both sexes should be tested because of the
possibility  of  sex-related  differences in LDsg values.
     The time following  dosing,  during which deaths are recorded for the
purpose of determining an LD5Q,  is the observation period.  This period
must be of sufficient  duration for manifestation of delayed effects but
not so  long  that there is doubt  whether the animal died as a direct
result  of the test substance.  The 7- and 14-day observation periods,
particularly the 14-day  period,  seem to be the most common lengths for
observation  periods among researchers in the United States.  Researchers
in  other countries use observation periods of various lengths.  Close
clinical observation of  the animal is necessary to assist in the assess-
ment of the  toxic effects.

1.3  SUBCHRONIC  TOXICITY TESTING

     Subchronic  tests  are designed to provide two types of toxicity data.
As a precursor to a chronic study, subchronic tests should identify target

-------
organs and define appropriate dose ranges for long-term exposures.   When
used as the primary evaluation of the effects of chronic exposure,
subchronic tests can provide detailed information on toxicity and the
potential hazard of the chemical to man.   The parameters to be employed
to assess toxicity vary with the purpose  of the test;  they are usually
more complex when the subchronic test is  to be the primary evaluation of
long-term effects.
     The number of species employed in a  subchronic test is an area
under discussion in the protocol design.   Most information suggests that
in a subchronic test both a rodent and nonrodent species should be used.
The overlap provided by the use of two mammals increases the efficiency
of hazard assessment for human exposure.   In general the rat and dog
appear to be the species most often employed in these hazard assessments.
However, whenever possible the choice of  species should be governed by
information on pharmacokinetics and metabolism of the compound.  Under
such circumstances other species might also be considered.
     The duration of exposure for a subchronic test is another area under
discussion.  The literature suggests that a 90-day exposure can provide
essentially the same toxicity data as would be provided by a long-term
test, excluding carcinogenicity and teratogenicity.  This information
could be the basis for regulatory decisions.  However, there is addi-
tional information in the literature suggesting that a 30-day duration
might be acceptable.  The comparisons available indicate that in many
cases the effects can be predicted qualitatively but not quantitatively.
This question needs more data for analysis.  Comparison studies or
access to unpublished information in industry files is needed to fully
resolve the debate over the minimum duration necessary  for a subchronic
test.
     The routes of exposure for a subchronic test  should  ideally represent
the expected routes of human exposure.  Most often this is by  ingestion
or inhalation, which are the principal exposure routes  utilized  in test
designs.  Oral exposure can be achieved  relatively easily, but  inhalation
exposure is more  complicated, requiring  special test  facilities.   Extrap-
olation of data obtained from one route  to  evaluate the hazard with
exposure to another route could  provide  a solution to the problems of

-------
 inhalation  tests.   However,  there  is  little information on the criteria
 necessary for  or  the  hazards involved in such an extrapolation.  Much
 more research  is  needed to  assess  the predictability and feasibility of
 route-to-route extrapolations.
      In subchronic testing  much  use is made of biochemical tests and
 pathology as parameters to  evaluate toxicity.  Both biochemical tests
 and  pathology  can provide data to  assess toxicity.
      Pathological evaluations should  include a gross examination of all
 organs  of all  test animals  at all  dose levels.  Organ weights should
 also be recorded  for  the major tissues.  Microscopic examination, because
 of its  higher  costs,  may be limited to the evaluation of selected
 tissues at  high and control dose levels.  Additionally, any  lesion-
 bearing organs, detected in the  gross examination, should be examined
 microscopically.  Further microscopic  examination at other dose levels
 should  be based on the pathology results from the high dose  level
 examination.
      Biochemical  tests should be employed to provide information on
 subtle  changes and to fully assess the extent of toxic effects.  A
 well-designed  subchronic study should include a basic hematology examina-
 tion (evaluating  cellular damage and  hemorrhaging), a set of enzyme and
 organ function tests  (primarily  evaluating liver and kidney  damage), and
 a urinalysis examination (primarily for kidney damage).  Changes indi-
 cated by these tests  can be investigated further by more complex bio-
 chemical tests.

 1.4   CHRONIC TOXICITY AND CARCINOGENICITY TESTING

      Many factors  influence the  results of chronic toxicity  and carcino-
 genicity tests  in  animals,  including  route of administration, dosage and
 frequency of exposure,  species,  strain, sex, age of the animal when the
 test  is  initiated,  dietary  constituents, immunologic status  of the animal,
duration of the experiment,  and  other factors.
      The principal  objective for conducting chronic toxicity tests is to
determine the toxic potential of a chemical when it is administered for
the greater part of the  life span  of  the animal.  In testing of chemicals

-------
for their carcinogenic potential the major objective may be  simply  to
determine whether or not the chemical is capable of inducing tumors in a
particular strain or species of animals.
     Because most chronic toxicity and carcinogenicity tests are con-
ducted over the major portion of the life span of the  animal,  short-
lived rodents are the choice of most investigators.  However,  the dog
is occasionally used, particularly in tests for chronic toxicity.
     Test duration is an issue under discussion.  Attempts to standard-
ize this parameter have been unsuccessful.  Generally, when mice and
rats are used for chronic toxicity and carcinogenicity tests,  24 to
27 months and approximately 24 months are recommended, respectively.
Typically, dogs are treated for 1 to 2 years in chronic toxicity studies.
     New strains of animals that are especially susceptible to tumor
induction and genetic factors that influence the spontaneous tumor
incidences in laboratory animals have been exploited  in the development
of certain short-term assays.  These and other in vivo short-term bio-
assays could be considered as intermediate between in vitro short-
term studies and lifetime studies and may become valuable tools in
screening potential carcinogens.
     The number of test animals used is a compromise between requirements
for good statistical precision and reasonable costs and work loads.
Most authorities now recommend 50 rodents or four to eight nonrodents
per test group.  Concurrent control groups comprising animals of the
same species, age, sex, weight, and number are also required.
     In tests for chronic toxicity at least three dose levels should be
used, and both treatment and observation of animals should be conducted
7 days per week.  In tests for carcinogenicity  in which induction  of
tumors is the primary end point, two or three doses of the  test chemical
may be employed.  If information on dose-response relationships is re-
quired, then several doses are necessary  to provide data  for appropriate
statistical analysis.
     Interim sacrifices may be performed  to provide information on the
progression of toxic events.  Macroscopic examination of  animals and
microscopic examination of tissues are  necessary in evaluating  the hazard

-------
of chemical exposure.  As a minimum, all major organs and tissues of
high dose and control animals are recommended for histological examina-
tion.  This study should also include any other tissues exhibiting gross
lesions.

-------
                  2.   BASIC EXPERIMENTAL  CONSIDERATIONS

2.1  INTRODUCTION

     This section will briefly discuss  some basic  experimental  parameters,
such as the purity and chemical stability of the test  chemical;  the
importance of selecting the appropriate vehicle; the diet,  husbandry,
and pathological examination of the test  animal; and the  importance  of
pharmacokinetics in the toxicological evaluation of chemicals.   The
discussions will illustrate why careful attention  to each of these is
essential to accurate and reliable experimental results.

2.2  TEST MATERIALS

2.2.1  Introduction
     The chemical and physical properties of the  test  material  must  be
known prior to the initiation of a toxicity experiment.   This is essen-
tial because the toxicologist must know that the  results  obtained from
his experiment are caused by the compound under investigation and not
by a degradation or contamination product.  The scientific committee of
the Food Safety Council in agreement with this states  that the "lack or
disregard of information on the chemical nature of the material to be
tested not only limits the usefulness of toxicological data for regula-
tory purposes but can, in some instances, provide an erroneous impres-
sion of its toxicity"  (Food Safety Council, 1978).  A document published
by the authority of the Ministry of Health and Welfare Canada  (1975)
further states that, before a long-term study is initiated, batch-
to-batch variations of the compound, chemical synthetic processes,
packaging and handling procedures, storage requirements, and inter-
actions with other chemicals must be considered.   The following subsec-
tions will briefly describe some aspects of the pretesting examination
of the test material.

-------
 2.2.2  Chemical Stability
      As soon as a substance arrives in the toxicology laboratory,  the
 stability of the chemical at various pH values and its photochemical
 properties must be determined (World Health Organization,  1978).   These
 parameters may determine the manner in which the chemical  should be
 stored prior to use.  The stability of the test material under the same
 conditions in which it will be administered must also be determined
 (Sontag, Page, and Saffiotti, 1976).  If the chemical nature of the
 test substance changes during an experiment as a result of the experi-
 mental conditions, the results will not be reliable.   In a worst-case
 situation, the chemical stability of the test material could be altered
 without the researcher's knowledge, thus permitting incorrect conclusions
 concerning the action of the test substance.  Therefore, prior to  the
 start of the toxicity test the stability of the test  chemical in the
 feed and/or vehicle, at the temperature and/or pH to  be used in the test,
 must be investigated to ensure that the chemical stability is not  altered
 by the experimental conditions.   The stability of the test material
 under its conditions of storage and/or administration will influence the
 frequency at which fresh treatment mixtures are prepared (National
 Academy of Sciences, 1975; Sontag, Page, and Saffiotti, 1976).
      When the test material is a chemical used in food processing, the
 possibility exists that under conditions of use the chemical may be
 transformed or degraded in foods,  with the resulting  products being
 either more or less toxic than the original chemical  (Food Safety  Council,
 1978).   The knowledge of potential chemical transformation would be
 essential for the  safety assessments of chemicals in  foods.

 2.2.3   Chemical Impurities
     The  chemical  purity of the  test material should  be established
prior  to  toxicity  testing (Sontag,  Page,  and Saffiotti,  1976).  This is
important because  of  the possiblity that trace contaminants  could be
responsible for, or  at  least modify,  the observed biological effects
attributed to the  test material  under investigation.   As pointed out by

-------
                                   9
the World Health Organization (1978),  if  the  contaminants  are unknown  or
their biological activity unsuspected,  toxicity  tests may  lead  to
erroneous conclusions concerning the test material.
     A related issue that confronts the toxicologist and is  one of the
earliest and most difficult decisions to  be made is  the selection  of the
desired purity of the sample to be studied  (technical grade, highly
purified, etc.) (World Health Organization, 1978).   If  the test material
is a commercial product that is known to  contain impurities, toxicity
testing of a highly purified sample could result in  erroneous conclu-
sions.  If the impurities are standardized, then testing of  the commer-
cial product would be the answer; however,  for those commercial products,
chemicals used in manufacturing, or chemicals incidentally released into
the environment for which the impurities  are  not standardized,  the
toxicologist must make a decision concerning  test sample purity.

2.2.4  Vehicles
     The choice of the appropriate vehicle is important because the
toxic activity of the test chemical can be altered,  especially if the
test chemical reacts chemically with the vehicle (also mentioned in
Sect. 2.2.2).  However, the toxic activity of the test chemical can be
altered without a change in chemical structure.  One example is that of
cottonseed oil, which, unless refined by superheated-steam processing,
may contain cyclopropenoid fatty acids, which can enhance the  activity
of some carcinogens  (Lee et al., 1968, as  cited by National Academy of
Sciences, 1975).  Other examples of vehicle interference in tests  for
carcinogenicity are  described by Pott, Brockhaus, and Huth  (1973),
who observed that when a subcutaneous dose of benzo(a)pyrene was  kept
constant, increasing the dose of the vehicle, tricoprylin, increased  the
rate of tumor production, and by Mori  (1965), who reported an  increase
in organs affected for tumor induction when  4-nitroquinoline-l-oxide
was administered in  olive oil and  lecithin instead  of  in  a  solvent
mixture containing cholesterol.

-------
                                   10
2.2.5  Homogeneity in Vehicle
     If a  text mixture  is prepared in sufficient quantity for the dosing
of several animals that are to receive the same concentration of the
test material, the homogeneity in the vehicle is essential.  Sontag,
Page, and  Saffiotti  (1976) state that in a long-term study where the test
material is  incorporated into the feed, the homogeneity and concentra-
tion in the  diet mix should be determined before the start of the study
and periodic analysis of random samples from freshly mixed batches
should be  performed  to  ensure that proper mixing and formulation pro-
cedures are  being used.

2.2.6  Conclusions
     Prior to the initiation of an experiment designed to evaluate the
toxicity of  a chemical, consideration should be given to the character-
istics of  the test chemical and the vehicle.  Specifically, the stability
and the purity of the test chemical should be known, and the possibility
of the vehicle altering the test results must be considered.

2.3  HUSBANDRY

2.3.1  Introduction
     In all  types of toxicity tests, the results can be influenced by
the husbandry conditions under which the testing is performed.  Husbandry
factors that  should  be managed include selection, transportation, and
quarantine of the test animals; control of diseases; proper design of
the cages  and testing facility; regulation of the temperature, humidity,
light, ventilation,  noise, and handling; and certification of personnel.
Unless these  factors are standardized or controlled, the data obtained
cannot be  used with  any certainty to evaluate the potential harmful
effects of the chemical to man.  The following subsections will briefly
discuss these factors.

-------
                                   11
2.3.2  Animal Selection
     In addition to the consideration  of species  and human metabolic
similarity discussed elsewhere in the  text,  several important factors
should be considered or controlled when selecting animals for use in
toxicity studies.  First, all animals  should have standardized life
cycle variables, such as age, sexual maturity,  and mating status, as
these factors can affect toxicity (Hurni, 1970).   Next,  the genetic
composition should be uniform, because this ensures consistency of
response and reproducibility of results (Food Safety Council, 1978;
Zbinden, 1963).  The current breeding  methods for maintaining a constant
genotype include inbred, outbred, and  hybrid strains.  Inbred animals
are crosses of brother and sister or parent and offspring that have
been maintained for at least 20 generations (National Academy of Sciences,
1969).  This results in a specific homozygous genotype where all animals
in the colony are as similar as identical twins (Hurni, 1970).  Outbred
or random bred strains maintain an unaltered, heterozygous pool of
genetic material on a population level  (Hurni, 1970) but are rarely used
in toxicity studies (National Academy of Sciences, 1969).  Because both
of these options have problems, hybrids from crosses of inbred strains
are often utilized.  These animals are  genetically uniform but are not
homozygous  (Food Safety Council, 1978;  Hurni, 1970).  Festing  (1979)
reviews, in more detail, the pros and cons of inbred, outbred, and
hybrid animals in toxicity testing.
     The need for reduction of animal health as a  factor  stimulated  the
development and use of specific pathogen-free  (SPF)  strains.   These  are
specially bred animals  (best obtained by aseptic hysterectomy  and arti-
ficial rearing of germ-free derived animals) that  have  accepted  levels
of health and are known  to be  free of certain  standard  diseases  and
parasites (Hurni, 1970; Meister, Hobik, and Metzger,  1967).   However,
these animals require  special  isolated  colonies with "clean zone"
barriers and  can be difficult  to maintain  (Hurni,  1970).   Therefore,
as with other  factors,  the nature  of  the experiment and the risks
involved will  decide the animal  type  and quality to be used (Page,
1977).

-------
                                    12
 2.3.3  Transportation,  Quarantine,  and Disease Control
      After selection of the appropriate test animals, transportation to
 the testing facility or animal room is necessary.   This  can  affect the
 response of the animals by subjecting  them to unusual stresses,  and
 they should be given time to recover (National Academy of  Sciences,
 1969).   Heinecke (1967) has shown that even transport between buildings
 (e.g.,  from an in-house breeding colony to the testing facility) has
 affected the blood status of mice for  3 to 4 days  afterwards.  More
 extensive transportation can disrupt the normal state for  up to  6 weeks
 (Hurni, 1970).  Therefore, it is necessary to allow the  animals  to
 readjust and acclimate  before initiating testing.
      While the animals  are acclimating, they can be kept isolated from
 the rest of the facility for quarantine purposes (Page,  1977).   The
 quarantine is necessary to prevent the introduction of diseases  into the
 existing facility population.  The duration of quarantine  varies with
 the species, its source, and the testing purpose (U.S. Department of
 Health, Education, and  Welfare,  1974), but should  be at  least 2  weeks
 (National Academy of Sciences, 1969).   As part of  the quarantine process,
 it  is a good practice to include:   (1) a physical  examination upon
 arrival;  (2) veterinary care to  check  for parasites and  disease  and to
 allow for immunization; (3) general grooming; and  (4) a  pathological
 examination of a small  number of the test animals  (Sontag, Page, and
 Saffiotti,  1976;  U.S. Department of Health, Education, and Welfare,
 1974).   If  any animals  in quarantine are found to  have a communicable
 disease,  the whole group should  be destroyed (Sontag, Page,  and  Saffiotti,
 1976).
      The  quarantine of  animals upon arrival is just part of  a general
 need  to control disease in the test facility.  The presence  of disease
 in  the  test  facility can invalidate the test results, especially if the
 disease produces  subtle,  nonspecific,  or long-term effects (van  der
Waaij and van Bekkum, 1967).   To control disease,  several  steps  are
necessary:   (1) acquire and include (after quarantine) only  healthy
animals;  (2)  prevent  introduction of pathogens into test facility;
 (3) maintain sanitary conditions; and  (4)  utilize  a well-planned disease

-------
                                   13
control program (Flynn, 1967;  National Academy of  Sciences,  1969).   The
prevention of pathogen introduction should include monitoring and/or
control of water supply, air supply, feed and bedding quality,  and
personnel contact (National Academy of Sciences, 1969).   The use of
filters and filter caps is particularly helpful to control airborne
diseases (Clough, 1976).  Sanitary control should  include facility  and
personnel cleanliness, proper waste disposal practices,  and vermin
control (U.S. Department of Health, Education, and Welfare,  1974).   The
disease control program can be the most vital part of the process and
should (1) be specific for that situation; (2) be  aimed  at the most
common or expected diseases; and (3) include frequent observation by
appropriately trained personnel (Flynn, 1967; U.S. Department of Health,
Education, and Welfare, 1974).  A detailed example of a  disease control
program is discussed by Flynn (1967).   Fox (1977)  reviewed some of the
most common diseases, for each organ system, that  occur  in testing
facilities.

2.3.4  Design of Testing Facility
     The design, scope, and size of a toxicity testing facility is the
product of many compromises, depending on the nature of the research,
the number of test animals to be housed, and the types of species used
(U.S. Department of Health, Education, and Welfare,  1974).  The facility
should be designed to provide maximum comfort and safety  for the test
animals and personnel,  to minimize  disease factors, and to minimize  costs
(National Academy of Sciences, 1969).  It is  essential that  a  facility
design include a building, wing, floor,  or room for  the animals  that is
separate from human work areas; specialized  laboratories  for maintenance
and assessment of the  animals; a supply-receiving area; a quarantine
area; and an incinerator  (U.S. Department of Health,  Education,  and
Welfare, 1974).  Specific  construction details are discussed in several
reviews  (Grange, 1976;  National Academy  of  Sciences,  1969;  U.S.  Department
of Health, Education,  and  Welfare,  1974).
     In  general  the  rooms  should be arranged to provide barriers between
individual rooms and the  outside  environment (National  Academy of Sciences,

-------
                                   14
1969;  Sontag, Page,  and  Saffiotti, 1976).  There are three common types
of  control  systems used  in  test  facilities:  open, closed, and isolated
(Shaw,  1976).   The open  system does not have hermetically sealed rooms,
and the entering of  materials  and personnel is not controlled.  The
closed system is hermetically  sealed with  sterilization controls over
entering materials and personnel.  An  isolated system is similar to a
closed one,  but does not allow personnel to enter.  One special type of
control system  is the "clean-dirty" system, which utilizes a unidirec-
tional corridor flow of  materials and  personnel  (Sontag, Page, and
Saffiotti,  1976).  Each  room has a separate entrance and exit door,
allowing personnel and materials to be decontaminated before entering
and to leave only through the  exit door  (Page, 1977).  This system is
especially  useful for minimizing disease contamination (Sontag, Page,
and Saffiotti,  1976).  If possible, there  should also be individual
rooms  for each  species and  each  test treatment.  A separate room for
controls, to evaluate the influence of various factors of the test
facility, has also been  suggested  (Food Safety Council, 1978).  The
overall goal of the  facility design is to  reduce or standardize the
environmental factors affecting  test results in the most efficient
manner.

2.3.5   Caging
     The cage is the single most important element of the physical envi-
ronment for  laboratory animals since it represents the immediate barrier
between the  microenvironment and macroenvironment of the testing facility
(Clough, 1976;  U.S.  Department of Health,  Education, and Welfare, 1974).
The  criteria to evaluate a  good  cage design include:  (1) it should meet
the  investigator's research requirements;  (2) it should be designed with
the  animal's physical comfort  as a prime consideration and; (3) it
should be compatible with proper maintenance requirements, especially
health and sanitary  procedures (U.S. Department of Health, Education,
and Welfare, 1974).   Some important features that a good cage design
incorporates are:  (1) it should be stackable or fit on racks;  (2) it
should have  smooth solid  walls and be  accessible for cleaning;  (3) it

-------
                                   15

should be constructed of stainless steel or plastic;  (4)  it should have
a perforated bottom (depending on species or experiment requirements);
and (5) it should have accessible feed and water sources (usually built
into a removable lid) (National Academy of Sciences,  1969).  Stainless
steel and especially hard plastics are used for cage  designs, because
they are lighter and resistant to corrosion (Clough,  1976).  A solid
cage design offers more control over the microclimate (usually increasing
humidity, NH3, C02, and temperature levels) and affords more protection
from microbial agents than wire mesh designs (Fox, 1977; Hurni, 1970).
However, animals in solid cages require transfer to a clean cage at
least every week, while animals in wire mesh cages can last 2 weeks
(Sontag, Page, and Saffiotti, 1976).  Also Winter and Flataker (1962)
found that rats in solid cages developed more resistance to chemicals
than those in wire mesh.  Therefore, the choice of exact cage design is
dependent on the goals and circumstances of the specific study.
     The number of animals per cage and the amount of space for each are
also determined by practical and experimental considerations.  Hatch et
al. (1965) found that a single mouse per cage was more susceptible to
certain cancers than groups of 1 to 5 mice.  Hurni (1970)  discussed
several studies in which isolated animals showed evidence  of an increase
in toxicity.  However, grouping of animals  in cages can indirectly
affect toxicity, for example, by increasing food competition or fecal/
urinary contamination  (Food Safety Council, 1978).  Table  2.1  gives some
recommended space standards for routine housing of laboratory  animals
(U.S. Department of Health, Education, and  Welfare, 1974).  Table 2.2
lists the minimum number of square inches necessary per rodent and the
maximum number of animals per  cage  (National Academy  of Sciences, 1969).
These guidelines address practical  considerations, but the nature of the
experiment often dictates the  actual  numbers of animals per cage.

2.3.6  Bedding
     Bedding material  is  frequently used in housing  test  animals, espe-
cially rodents, and  has been shown to affect toxicity.  In general,
bedding  serves several purposes:   (1) it provides thermal insulation;

-------
                                         16
              Table 2.1.   Space recommendations  for laboratory animals
Species
Mouse



Rat



Hamster



Guinea pig


Rabbit


Cat

Dog*





Primates0' d
Group 1
Group 2
Group 3
Group 4
Group 5
Weight
Up to 10 g
10 to 15 g
16 to 25 g
Over 25 g
Up to 100 g
100 to 200 g
201 to 300 g
Over 300 g
Up to 60 g
60 to 80 g
81 to 100 g
Over 100 g
Up to 250 g
250 to 350 g
Over 350 g
Up to 2 kg
2 to 4 kg
Over 4 kg
Up to 4 kg
Over 4 kg
Up to 15 kg
15 to 30 kg
Over 30 kg
Up to 15 kg
15 to 30 kg
Over 30 kg

Up to 1 kg
Up to 3 kg
Up to 15 kg
Over 15 kg
Over 25 kg
Type of
housing
Cage
Cage
Cage
Cage
Cage
Cage
Cage
Cage
Cage
Cage
Cage
Cage
Cage
Cage
Cage
Cage
Cage
Cage
Cage
Cage
Pen or run
Pen or run
Pen or run
Cage
Cage
Cage

Cage
Cage
Cage
Cage
Cage
Floor area per
animal (squared)
39 cm (6 in.)
52 cm (8 in.)
77 cm (12 in.)
97 cm (15 in.)
110 cm (17 in.)
148 cm (23 in.)
187 cm (29 in.)
258 cm (40 in.)
64.5 cm (10.0 in.)
83.9 cm (13.0 in.)
103.2 cm (16.0 in.)
122.6 cm (19.0 in.)
277 cm (43 in.)
374 cm (58 in.)
652 cm (101 in.)
0.14 m (1.5 ft)
0.28 m (3.0 ft)
0.37 m (4.0 ft)
0.28 m (3.0 ft)
0.37 m (4.0 ft)
0.74 m (8.0 ft)
1.12 m (12.0 ft)
2.23 m (24.0 ft)
0.74 m (8.0 ft)
1.12 m (12.0 ft)
6

0.15 m (1.6 ft)
0.28 m (3.0 ft)
0.40 m (4.3 ft)
0.74 m (8.0 ft)
2.33 m (25.0 ft)
Heighta
12.7 cm (5 in.)
12.7 cm (5 in.)
12.7 cm (5 in.)
12.7 cm (5 in.)
17.8 cm (7 in.)
17.8 cm (7 in.)
17.8 cm (7 in.)
17.8 cm (7 in.)
15.2 cm (6 in.)
15.2 cm (6 in.)
15.2 cm (6 in.)
15.2 cm (6 in.)
17.8 cm (7 in.)
17.8 cm (7 in.)
17.8 cm (7 in.)
35.6 cm (14 in.)
35.6 cm (14 in.)
35.6 cm (14 in.)
61.0 cm (24 in.)
61.0 cm (24 in.)



81.3 cm (32 in.)
91.4 cm (36 in.)
b

50.8 cm (20 in.)
76.2 cm (30 in.)
76.2 cm (30 in.)
91.4 cm (36 in.)
213.4 cm (84 in.)
     Height means  from the  resting  floor  to the cage top.

      These recommendations  may require modifications according to the body con-
 formations  of particular breeds.  As a further general guide, the height of a dog
 cage should be  equal  to the  height of the  dog over the shoulders (at the withers),
 plus at least 6 inches,  and  the width and  depth of the cage should be equal to
 the length  of the dog from the  tip of the  nose to the base of the tail, plus at
 least 6 inches.
     °The primates  are grouped  according to approximate size with examples of
 species that may be included in each group:  Group 1 — marmosets, tupaias, and
 infants of  various  species;  Group 2  — cebus and similar species; Group 3 —
macaques and large  African species;  Group  4 — baboons, monkeys larger than 15 kg,
and adult members of  brachiating species such as gibbons, spider monkeys and
woolly monkeys;  Group 5 —  great apes.
     'where primates  are housed in groups  in pens, only compatible animals should
be kept.  Minimum height of  pens should be 6 feet.  Resting perches, nesting
boxes, and  escape barriers necessary for the well-being of the particular animals
should also be provided.
     Source:  Adapted from U.S. Department of Health, Education, and Welfare, 1974.

-------
                                   17
           Table 2.2.  Cage space requirements for rodents
                             Minimum square        Maximum number
                           inches per animala    of animals per cage
Mice
Weaning to 5 weeks
5 to 8 weeks
8 to 12 weeks
Over 12 weeks
Hamsters
Weaning to 5 weeks
5 to 10 weeks
Over 10 weeks
Rats
Up to 50 g
50 to 100 g
100 to 150 g
150 to 200 g
200 to 300 g
Over 300 g
Guinea pigs
Weaning to 350 g
350 g and over
Breeders

6
8
12
15

10
12.5
15

15
17
19
23
29
40

60
90
180

40
30
20
20

20
16
13

50
50
40
40
30
25

15
10
5
         In many circumstances, more space per animal may be needed.
        Source:  Adapted from National Academy of Sciences, 1969.
(2) it absorbs fecal and urinary wastes and water spillage;  (3)  it is
used to build nests; and (4)  it reduces stress by providing a protective,
isolating cover (National Academy of Sciences, 1969).  The material
selected should therefore be absorbant, nonedible, and innocuous but not
dusty or highly resinous (Clough, 1976; Hurni, 1970; National Academy of
Sciences, 1969).  Some of the bedding materials rated as acceptable by
the National Academy of Sciences (1969) include:  coarse pine sawdust;
pine, cedar, basswood, or poplar shavings; crushed corn cobs; and
hardwood chips.  However, other researchers have found problems with
some of these materials.  Porter (1967) found that sawdust often  contains
many possible contaminants, and he preferred sterilized pine shavings.
In contrast, Clough  (1976) and Fox  (1977) warned that pine, cedar, and
other softwoods, if  fresh, can induce biological variations in the

-------
                                    18
 microsomal metabolizing enzymes and alter  chemical  toxicity.  Hurni
 (1970)  warned that hardwood shavings, which are high in tannic acid
 content,  can lead to constipation in rodents.  However,  Sontag, Page,
 and Saffiotti (1976) preferred hardwood shavings  for long-term carcino-
 genicity  studies.  In any case, the material must be sterilized, pref-
 erably  by autoclaving, and be changed frequently.

 2.3.7  Temperature and Humidity
      Temperature and humidity are two environmental factors  that should
 be controlled by the experimenter, since they  can influence  the test
 results.   The effect of changes in these factors  will be influenced by
 the magnitude, frequency, and duration of  the  changes, by whether  or not
 the test  animals have behavioral modifications to adapt to these,  and
 by the  current physical and health status  of the  animals (Weihe, 1976a).
 The easiest solution is to control temperature and  humidity  by air
 conditioning each animal room (U.S. Department of Health, Education, and
 Welfare,  1974).
      Weihe (1964, as cited by Heinecke,  1967), by varying the environ-
 mental  temperature,  was able to affect the LDsgS  of chemicals.  Heinecke
 (1967)  found that temperature changes also affected blood values.  Hurni
 (1970)  reviewed several studies in which cold  temperatures or increases
 in ambient temperature affected both the resistance of test  animals and
 the toxic effects of chemicals.  Changes in ambient temperature can
 affect  the metabolism of chemicals, and the effects produced can mimic
 chemical  effects, making diagnosis difficult (World Health Organization,
 1978).  Humidity levels can also affect toxicity  and were found to be a
 principal factor of ringtail in young rats (Clough, 1976; Flynn, 1967).
 Thus  it is very important to control these factors.
      The  exact limits of temperature and humidity beyond which signifi-
 cant  metabolic adaptation will occur are not known, but if the changes
 are moderate (±5°K or 20% to 30% humidity)  the animals should be able to
 adapt behaviorally without damage (Weihe,  1976a).  The National Academy
 of Sciences  (1969) recommends a fluctuation of no more than  ±2°F in a
 range of  70°  to  80°F and 40% to 70% humidity for  rats and mice.  The
U.S. Department  of Health,  Education,  and  Welfare (1974)  suggests  levels

-------
                                   19
of 65° to 85°F and 30% to 70%,  depending on the test species.   Sontag,
Page, and Saffiotti (1976) recommend more precise levels of 74°F ± 2°
and 40% ± 5% for rats and mice.  In any case,  the investigator should
define the levels used, along with the population size and cage type, so
that the study can be properly  evaluated.

2.3.8  Light
     Light is another environmental variable that should be controlled.
The damaging effects of light are dependent on the intensity and duration
(Weihe, 19762?)-  All common laboratory animals are sensitive to light,
and by using low-level, artificial light sources, with exclusion from
alternate sources such as windows, the proper control can be maintained
(Weihe, 19762?).
     Strong light has proved to decrease the reproductive ability of
mice (Porter, 1967) and to damage the retina and indirectly the endocrine
system (Fox, 1977; Weihe, 19762?).  Therefore, the light should be
diffused through the animal room, with minimum levels of 100 lux at  the
level of the cage racks  (U.S. Department of Health, Education, and
Welfare, 1974).  Hurni (1970) suggests room levels of 300 to 500 lux for
rodents.  In the computation of light levels, the effect of opaque or
translucent cage construction  should be  considered, since  this  can
greatly affect  the level of light reaching  the test animal  (Clough,
1976).
     The duration of  the photoperiod  should also be controlled,  prefer-
ably in a constant pattern  (U.S. Department of Health,  Education,  and
Welfare, 1974;  Weihe,  19762?).  The  daylight cycle  should be designed to
suit the test  species, for  example,  10  h for  rats  and mice, but 14 h for
cats  (Hurni,  1970).   In  addition  to controlling  the diurnal pattern, it
is  necessary  to standardize the  time of "day" when blood samples and
sacrifices are made,  in  order  to  reduce variation (Fouts, 1976).

2.3.9  Ventilation
     Proper ventilation  of  the testing facility and particularly the
animal room is necessary to maintain low concentrations of atmospheric

-------
                                    20
 contaminants (e.g., particulates, C02, NHs, moisture,  and microorganisms),
 reduce odors, regulate temperatures, and promote comfort (Clough,  1976;
 U.S. Department of Health, Education, and Welfare,  197A).  The ability
 to limit odor depends on the species used and the population densities,
 as well as the proper design of the facility (U.S.  Department of Health,
 Education, and Welfare, 1974).  The precise pattern of air movements  in
 each room results from the interaction of convection currents arising
 from the test animals, from the air input by the ventilation system,
 and from the deflections caused by the cages, racks, and equipment
 (Clough, 1976).  Nevins (1971) discusses the design criteria needed to
 optimize the ventilation patterns.  Rooms used by humans and animals
 should each be ventilated separately (National Academy of Sciences, 1969;
 U.S. Department of Health, Education, and Welfare,  1974).  The air
 should be changed frequently without causing drafts, and incoming  or
 recirculated air must be filtered (National Academy of Sciences, 1969;
 Hurni, 1970; Sontag, Page, and Saffiotti, 1976). The recommended  rates
 of air changes per hour include:  6 to 15 (National Academy of Sciences,
 1969); 15 to 23 (Hurni, 1970); and 10 to 15 (Shaw,  1976; Sontag, Page,
 and Saffiotti, 1976; U.S.  Department of Health, Education, and Welfare,
 1974).  The exact rate will depend on the population density, refuse
 removal schedule,  test design, and degree of desired comfort (Hurni,
 1970;  National Academy of  Sciences,  1969).   Hurni (1970) also suggests
 that the air should be negatively charged (about 2000  negative ions per
 cubic  centimeter)  to increase the oxidation of odors and microorganisms.
     The use of special ventilation systems for testing facilities
 requires some specific precautions.   If a "clean-dirty" corridor system
 is  used,  the air pressure  in each room should be positive to the "dirty"
 corridor and negative to the "clean" one (Sontag, Page, and Saffiotti,
 1976).   This will  help reduce the possibility of backflow contamination.
 For  a  laminar flow system  where air  is introduced at slow rates of speed
 from large wall-sized vents,  the room design and equipment placement
must be  carefully  considered,  because turbulence and eddy effects  could
disrupt  the  flow (Shaw,  1976).   Other design considerations affecting
air movements and  filtration are reviewed by Shaw (1976) and Dymet
 (1976).

-------
                                   21
2.3.10  Noise and Handling
     Noise can be undesirable because of  its  effects,  often  underrated,
on test animals and personnel.  Because there is  always  some unavoidable
background noise, it should be considered in  designing test  facilities
and toxicity studies (Hurni, 1970;  U.S. Department  of  Health, Education,
and Welfare, 1974).  The sources of noise are derived  from the feeding
and cleaning operations, ventilation equipment, animal vocalizations
(especially dogs and monkeys), and  animal/cage contacts  (Fletcher,
1976).  The damaging effects of noise include the expected auditory
(damage to ear structures and tissues) and nonauditory (stress) effects,
which have been demonstrated in special acute studies  (Fletcher, 1976).
However, extrapolation of these effects to chronic laboratory situations
is uncertain because many critical  factors such as species auditory
levels and frequency, intensity, and temporal patterns of common noises
are unknown.  In general, effects of noise should be minimized, especially
where the exposure could be lengthy or where studies are very delicate
(Fletcher, 1976).  Noise can be minimized by the  separation  of animal
and human occupancy zones, use of soundproofing materials, padding of
equipment, proper personnel training, transferring cleaning  chores
outside the housing area, and acclimating the animals to unavoidable
background noises  (Fletcher,  1976; Hurni, 1970; U.S. Department of
Health, Education, and Welfare, 1974).   If these procedures  are followed,
the effect of noise on  the  test results  should be  insignificant.
     Handling,  or  contact between  the laboratory personnel  and the test
animals, can be a detrimental factor  that reduces  the standardization of
environmental factors (Hurni, 1970).  Improper handling may produce
unnecessary stress or injury  to the  test animal or personnel (Short,
1967).  Porter  (1967) found that increased handling of  mice affected
their reproduction and  lactation and caused  some reduction  in weight
gain.  On the other hand, proper handling produces tameness, which is
healthy for the animal  and  expedient for the researcher.  However, even
with increased  training of  personnel and the use of the proper techniques,
such as those discussed by  Short (1967), the chance for differential
treatment exists and should be  considered, especially for comparative
studies (Hurni, 1970).

-------
                                    22
 2.3.11  Personnel
      One of the basic factors in the design of  a  successful husbandry
 program is the selection and training of staff  personnel.  The numbers
 and types of personnel utilized and the chain of  command developed depend
 on the goals and role of each toxicity testing  facility  (U.S. Department
 of Health, Education, and Welfare,  1974).   Lane-Petter  (1967) outlines
 an example of one such staff setup.  However, there  are certain standards
 that all staffs and personnel must  meet.  Basically  the staff must
 include animal technicians (usually at least junior  and  senior tech-
 nicians; the number of levels depends on the size of facility) and
 veterinary personnel (as supervisors or consultants) (National Academy
 of Sciences, 1969).  The personnel  caring for and monitoring health of
 the animals must be trained in the  theoretical  and practical aspects of
 their jobs (Fox, 1977).  Usually the theoretical  or  course work requires
 certification by such organizations as the American  Association of
 Laboratory Animal Science (Fox, 1977) or the Canadian Association for
 Laboratory Science Program (Arnold  et al., 1977). Typically these
 organizations certify personnel on  three levels:   assistant animal
 technician, animal technician, and  animal technologist.  The individual
 facility can then provide the practical onsite  training, which ensures
 the necessary training flexibility  (Fox, 1977).  To  assist the veter-
 inarian or toxicologist in the treatment,  test  procedures, and assess-
 ment roles, veterinary technologists or toxicology technologists are
 often employed (Fox,* 1977).   By the use of such certified  and special
 personnel, many of the problems associated with husbandry  requirements
 can be minimized so that the test results are not affected.

 2.3.12  Conclusions
     Many husbandry factors  contribute to a scientifically proper
 toxicity evaluation.   The selection of test animals  should result in
 subjects with standardized life cycle variables,  uniform genetic compo-
 sition,  and,  in some  cases,  controlled levels of  health  (e.g., SPF
 strains).   After selection,  the animals are usually  transported and
should be  given time  to acclimate before testing.  They also should be
quarantined  to  prevent  introduction of disease  into  the testing facility.

-------
                                   23
The quarantine is part of a larger disease-control program based on proper
planning and thorough sanitary maintenance procedures.   The design of
the testing facility is another variable that can contribute to the
success of a toxicity evaluation.   A proper testing facility should
include separate human and animal areas, specialized laboratories for
maintenance and assessment of the animals, a supply-receiving area,
a quarantine area, and an incinerator.  The design should also provide
for proper environmental controls.  The cage design, especially solid
plastic models, affects the microclimate and thereby the toxicity
potential of the test chemical.  Bedding material can be another source
of nontreatment toxicity and should not be composed of fresh softwoods
or hardwoods with high tannic acid content.  The temperature and humidity
of the testing environment should be controlled within a range of 65° to
85°F and 30% to 70%, depending on the species, with only gradual changes.
The available light should be regulated to provide diffuse levels with a
standard photoperiod duration.  Ventilation and filtration of the housing
atmosphere can control many other environmental factors and is often
designed to reduce contamination factors  (e.g., "clean-dirty" corridor
system).  Noise and handling variables  can increase animal stress and
may invalidate the test results.  The correct combination of well-
trained personnel likewise minimizes  test interference.  Only by control
of all these husbandry variables will the test results accurately reflect
the toxicity of the test chemical.

2.4  DIET

2.4.1  Introduction
     The major issues necessary to  the  discussion of diets  of  laboratory
animals include general nutritional adequacy of  diets,  types of diets
available  (commercial, open-formula,  semisynthetic),  and the presence in
the diet of toxic contaminants.   Also important,  in the context of this
document,  are the effects  of  diet on the response of experimental animals
to toxic substances.

-------
                                    24
 2.4.2  Dietary Requirements for Laboratory Animals
      Basic dietary requirements are similar for various common laboratory
 animal species (Clarke et al., 1977).  All require proteins, carbohydrates,
 fats, minerals, micronutrients, and energy.
      Dietary proteins are the source of the amino acids that an animal
 requires to build its own proteins.  The protein content of the diet
 should ideally contain, in the correct proportions, all the "essential"
 amino acids (those not formed at all in the animal's own tissues or not
 formed at a rate fast enough to satisfy demand) (Clarke et al., 1977).
 Table 2.3 is a summary of the attempts of various investigators to
 determine the relative proportions in which the essential amino acids
 are required by the growing rat (Coates et al., 1969).  The composition
 of two proteins, hen's egg and human milk, are also presented.  These
 are both known to be 100 percent utilizable by the rat and are assumed
 to have practically ideal proportions of the essential amino acids.
 Since the amino acid requirements for mice and rats seem to be similar
 a single "target pattern," given in the last column of Table 2.3, is
 proposed for both species.  Clarke et al.  (1977) suggested that the same
 target pattern would be satisfactory for all common laboratory animals.
      Carbohydrates are the major energy source in most diets for labora-
 tory animals.   It has been shown that energy requirements for maintenance
 are closely related to basic metabolic rate; therefore, the requirement
 for energy is  higher during pregnancy, lactation,  and periods of rapid
 growth.
      Lipids in the diets of laboratory animals are also a source of
 energy and are necessary for utilization of fat-soluble vitamins.
 Essential fatty acids are required for synthesis of tissue and cell
 components.
      Other general dietary requirements for laboratory animals include
 vitamins  A,  D,  E,  K,  B,  and C and choline.  Essential inorganic elements
 or minerals  include electrolytes (sodium,  potassium, calcium, magnesium,
 chloride,  and  phosphate),  which contribute to ionic and osmotic balance
 between cells,  tissue fluids,  and plasma.   Minor and trace elements
 include iron,  copper,  zinc, magnesium,  iodine, cobalt, manganese, and
 selenium, which take  part  in  intracellular metabolic processes as
metalloprotein  complexes and  coenzymes.

-------
                 Table 2.3.  Requirements for essential amino acids expressed as percentage of protein
Rat
Mouse
Human
milk
Whole
egg
Reference0

Arginlne
Histidine
Lysine
Tyrosine
Tryptophan
Phenylalanine
Methionine -f cystine
Threonine
Leucine
Isoleucine
Valine
1

4.0
10.1

1.0
4.6
7.5
5.0
8.0
5.0
7.0
2
5.0
2.5
9.0
3.0
1.1
4.2
5.0
5.0
7.0
5.5
5.5
3
9.0
2.9
11.8

1.0
7.7
5,4
3.5
7.3
4.8
5.4
4

1.8
5.2

1.0
4.9
4.7
4.1
7.8
4.3
5.0
567
4.6
2.0 2.0
3.7 6.5
4.7
1.5 1.3
6.3
4.4 4.6
2.6 4.2
9.5 8.2
5.1 5.0
6.2

1
2
7

1
7
5
4
6
4
5
8 9
.7
.5
.5

.3
.5
.0 3.0
.2
.7
.2
.8
10
1.9
3.4
9.0
4.8
1.9
14.0
8.0
6.8
12.5
8.7
9.7
11
4.0
2.2
6.4
5.7
1.7
4.8
4.4
4.6
9.2
6.6
6.7
11
6.6
2.4
6.4
4.2
1.6
5.8
5.5
5.1
8.8
6.6
7.3
"Target
pattern"
5.0
2.5
6.0
4.0
1.5
5.0
4.5
4.0
8.0
5.0
5.5
     aHef.  1 — Rose,  W.  C.,  M.  J.  Oesterling,  and M.  Womach.   1948.   Comparative  Growth  on Diets Containing
Ten and Nineteen Amino Acids with  Further Observations upon the Role of Glutamlc  and Aspartic Acids.  J. Biol.
Chem. 176:753.   Ref.  2 — Rama Rao, P.  B., V.  C.  Metta, and B.  C.  Johnson.   1959.   The Amino Acid Composition
and the Nutritive Value  of Proteins.   I.   Essential Amino Acid Requirements of  the Growing Rat.  J. Nutr.
69:387.  Ref.  3 — Rogers,  Q.  R., and A. E. Harper.  1964.   Amino  Acid Diet  for  Rats.  Fed. Proc. Fed. Am. Soc.
Exp. Biol.  23:186.  Ref.  4 - Bender, A. E.  1961.  Determination  of  the Nutritive Value  of Proteins by
Chemical Analysis.  In:   Progress  in Meeting Protein  Needs of  Infants and Preschool Children.  Publ. Natl.
Res. Coun., Wash. 843:407.   Ref. 5 — Fisher,  R.  B.  1954.   Protein Metabolism.  London:  Methuen.  Ref. 6 —
HcLaughlin, J.  M., and W.  I.  Illman.   1967.   Use of Free Plasma Amino Acid  Levels for Estimating Amino Acid
Requirements for the  Growing Rats. J. Nutr.  93:21.  Ref.  7 —  Hartsook, W.  E.,  and H. H. Mitchell.  1956.  The
Effect of Age  on the  Protein and Methionine Requirement of the Rat.   J. Nutr. 60:173.  Ref. 8 — National
Research Council.  1962.   Nutritional  Requirements  of Domestic Animals, Number  10:  Nutritional Requirements
of Laboratory Animals.  National Research Council,  Washington.  990.   Ref.  9 —  Leveille, G. A.,
H. E. Sauberllch, and J. W.  Shockley.  1961.   Sulfur  Amino Acid Requirements for  Growth  of Mice Fed Two Levels
of Nitrogen.  J. Nutr. 75:455.  Ref. 10 — Bauer,  C. D.,  and C.  P.  Berg. 1943.  The Amino Acids Required for
Growth in Mice  and the Availability of the Optical  Isomers.  J. Nutr.  26:51.  Ref.  11 —  FAO.  1965.  Protein
Requirements.   FAO Nutr. Mtg. Rep.  Ser. 37.

     Source:  Coates  et al.,  1969.
                                                                                                                            CO
                                                                                                                            Ui

-------
                                    26
      In selecting the proper diet the  investigator must bear  in mind
 that basal dietary requirements,  as well as specific nutritional require-
 ments of animals, may vary with species or strain and with age, sex,
 gestation, lactation, disease, season of the year,  temperature, relative
 humidity, ventilation, unknown internal physiological mechanisms,  and
 stress (of experimentation) (Hughes and Lang,  1978).  Specific require-
 ments for the various species of laboratory animals are discussed  in
 detail by Clarke et al. (1977) and in  the National Academy of Sciences —
 National Research Council (1972)  document Nutrient Requirements of
 Laboratory Animals.

 2.4.3  Types of Diets for Laboratory Animals
      In choosing a feed one should consider:  (1) constancy of its major
 ingredients and their sources; (2) timely delivery;  (3) moisture content;
 (4)  freshness; (5) storage characteristics (Sontag, Page,  and Saffiotti,
 1976); and (6) purity.
      Many commerical diets are available for laboratory animals.   The
 cost of a diet is a small part of the  overall cost of bioassays, and the
 investigator should buy the best diet  to ensure long-term survival under
 optimal nutritional conditions (Sontag, Page,  and Saffiotti,  1976;
 Weisburger, 1976).
      The goal in testing must be to standardize the diet  in and among
 animal studies so that data can accurately be compared on a worldwide
 level.   Therefore, in the formulation  of a standard diet  international
 availability of the ingredients should be considered.
      The Food Safety Council (1978) recommends the use of standardized
 diets in accordance with guidelines of the National Academy of Sciences'
 1972  Subcommittee on Laboratory Animal Nutrition and 1978 Committee on
 Laboratory Animal Diets.
      Laboratory animal diets have been characterized by the American
 Institute of  Nutrition (1977)  as:
Cereal-based  diet         Diet  formulations composed predominantly  of
Unrefined diet            unrefined plant and animal materials that may
Nonpurified diet          contain  added vitamins or minerals — stock
                          diet,  laboratory chow,  etc.

-------
                                   27
Open-formula             Diet in which precise percentage composition
                         of each ingredient is available either in the
                         published scientific literature or in available
                         commercial information.   No changes are per-
                         mitted in type or amount of ingredients.
Closed-formula           Diet in which exact composition by type and
                         amount of each ingredient is not disclosed by
                         the manufacturer.  Combination of ingredients
                         may change with market conditions.
Purified diet            Diets composed primarily of refined ingredients
                         such as commercially refined proteins, carbo-
                         hydrates, and fats with vitamins and minerals
                         added.  (Semisynthetic.)
Chemically defined       Diets characterized by the nitrogen source being
  diet                   provided by pure amino acids, carbohydrates from
                         refined mono- or disaccharides, and fats from
                         purified fatty acids or triglycerides.  Minerals
                         are reagent grade, and vitamins are of high
                         purity.
     Use of open-formula diets is encouraged by the Food Safety Council
(1978) and the American Institute of Nutrition (1977) and in Guidelines
for Carcinogen Bioassay in Small Rodents  (Sontag, Page, and Saffiotti,
1976).
     The open-formula, cereal-based diet  (NIH-07) proposed by the
American Institute of Nutrition  (1978) was developed by the National
Institute of Health in 1972 and  is shown  in Table 2.4.  The NIH-07  open-
formula rat and mouse diet has been found  to be  satisfactory  for repro-
duction, lactation, and maintenance of the animals.  It is produced by
several feed manufacturers and is priced  competitively with commercial
diets  (American Institute of Nutrition, 1977).   In  spite  of high cost,
the semisynthetic diet may be preferred for  some studies  (Page,  1977).
For example, practical experience has  shown  that semisynthetic diets
could  be of use in subchronic  studies, although  they have not been
particularly satisfactory in chronic reproduction studies (Food Safety
Council, 1978).
     The purified diet AIN-76  and  the vitamin and mineral mixtures used
in this diet are shown in Tables 2.5,  2.6, and 2.7.  This diet supports
growth, lactation, and reproduction in rats  and mice comparable to the
NIH-07 diet  (American Institute  of  Nutrition,  1977).

-------
                                    28

               Table 2.4.  NIH-7 open-formula rat and
                            mouse ration0-

                       Ingredient                           %

         Dried skim milk                                   5.00
         Fish meal (60% protein)                          10.00
         Soybean meal (49% protein)                       12.00
         Alfalfa meal (dehydrated, 17% protein)            4.00
         Corn gluten meal (60% protein)                    3.00
         Ground No. 2 yellow shelled corn                 24.50
         Ground hard winter wheat                         23.00
         Wheat middlings                                  10.00
         Brewer's dried yeast                              2.00
         Dry molasses                                      1.50
         Soybean oil                                       2.50
         Sodium chloride                                   0.50
         Dicalcium phosphate                               1.25
         Ground limestone                                  0.50
                                                           0.25
              Total                                      100.00

               Calculated proximate composition — crude
         protein, 23.5%; crude fat, 5.0%; crude fiber, 4.5%;
         ash, 7.0%.

               Vitamin and mineral premixes shall provide per
         kilogram of diet:  vitamin A (stabilized), 6050 IU;
         vitamin Da, 5060 IU; vitamin K, 3.1 mg; o-tocopheryl
         acetate, 22 IU; choline 0.6 g; folic acid, 2.4 mg;
         niacin, 33 mg; d-pantothenic acid, 20 mg; riboflavin,
         3.7 mg; thiamin, 11 mg; vitamin B-12, 4.4 yg;
         pyridoxine, 1.9 mg; biotin, 0.15 mg; cobalt, 0.44 mg;
         copper, 4.4|mg; iron, 132 mg; manganese, 66 mg; zinc,
         18 mg; iodine, 1.5 mg.

              Source:  American Institute of Nutrition, 1977.
         Reprinted with permission of the publisher.
     For special circumstances,  controlled variation  of  experimental
diets to simulate the variety of human  diets has been suggested by  the
World Health Organization  (1978) and  the National Academy  of  Sciences

(1975).  For example, animal diets could be shifted to a higher fat
content to allow for the expression of  some cocarcinogenicity of  fats or
to a lower protein content to simulate  human deficiency  states.

     Regardless of the type of diet chosen, all test  animals  and  their
corresponding controls should be maintained on identical diets, and when

-------
                             29


  Table 2.5.  AIN-76® purified d±eta»b (for rats and mice)

    Ingredient                                          %


Casein0                                                 20.0
DL-Methionine                                            0.3
Cornstarch                                              15.0
Sucrose                                                 50.0
Fiber**                                                   5.0
Corn oile                                                5.0
AIM mineral mix/                                         3.5
AIN vitamin mix                                          1.0
Choline bitartrate                                       0.2

     Total                                             100.0

      Trademark pending.

      This diet is intended for growth and maintenance dur-
ing the first year of life.  Investigators should be aware
that diets high in sucrose can be cariogenic and that some
strains of rats fed such diets may develop kidney lesions
after extended periods.  The diet has been found to be sat-
isfactory for reproduction and lactation in both rats and
mice.  If used for deficiency studies, modifications will be
necessary.  If used in ultraclean environment, several trace
elements should be added (Federation Proc. 33, 1748-1773,
1974).  The diet can be pelleted satisfactorily, if desired,
by addition of water (no binder).

      Feed-grade casein having at least 85% protein.

      Cellulose-type fiber.
     a
      Some commercial corn oils contain antioxidants  (maximum
0.02%) and a surfactant (dimethyl silicone).  These additives
should be innocuous for most nutritional studies, but inves-
tigators should be aware of their presence.  It is recom-
mended that an oil with added antioxidant be used to prevent
rancidity.  Diet should be stored at 4°C or colder, and  it
is recommended that the diet not be kept longer than
4 months.
     f
     •"The total dietary content of some minerals, due to
their presence in casein, will be slightly higher and will
vary according to the casein used.

     Source:  American Institute of Nutrition,  1977.
Reprinted with permission of the publisher.

-------
                                   30
                  Table 2.6.   AIN-76  vitamin mixture0
                      „.„  .                       Per kilogram
                      Vitamin                       ,.  .   e
                                                   of mixture
           Thiamin-HCl                             600 mg
           Riboflavin                              600 mg
           Pyridoxine-HCl                          700 mg
           Nicotinic acid*                         3 g
           D-Calcium pantothenate                  1.6 g
           Folic acid                              200 mg
           D-Biotin                                20 mg
           Cyanocobalamin (vitamin B-12)           1 mg
           Retinyl palmitate or acetate            *°
             (vitamin A)
           dl-a-Tocopheryl acetate                 4**
             (vitamin E)
           Cholecalciferol (vitamin D3)            2.5 mge
           Menaquinone (vitamin K)J                5.0 mg
           Sucrose, finely powdered                To make
                                                     1000.0 g

                 Based on the National Academy of Sciences-
           National Research Council recommended levels for
           rats (National Academy of Sciences-National
           Research Council, 1972).  To be used at 1% of diet.

                 Nicotinamide is equivalent.

                CAs stabilized powder to provide 400 000 IU
           vitamin A activity or 120 000 retinol equivalents.

                 As stabilized powder to provide 5000 IU
           vitamin E activity.

                e!00 000 IU.  May be in powder form.
                f
                •'Menadione.

                Source:   American Institute of Nutrition, 1977.
           Reprinted with permission of the publisher.
 a  change in diet manufacture occurs,  all animals should be changed to
 the new  diet at  the  same  time (Sontag,  Page,  and Saffiotti,  1976;
Weisburger, 1976).   The diets should  be well  tried  and  readily  available

 to the animals  (Magee, 1970).
     In  addition, feed must  be protected from air,  light,  heat,  chemical
fumigants, radiation, and contamination by microorganisms  — all of which

may damage the nutrient quality of a  diet (Clarke et  al.,  1977).   Sontag,

-------
                                   31

                  Table 2.7.   AIN-76® mineral mixture*2
                       ,.                        Grams  per kilogram
                  Ingredient                        of mixture
      Calcium phosphate,  dibasic (CaHPO^)              500.0
      Sodium chloride (NaCl)                            74.0
      Potassium citrate,  monohydrate                  220.0
         (K3C6H507.H20)
      Potassium sulfate  ^SO^)                         52.0
      Magnesium oxide (MgO)                             24.0
      Manganous carbonate (43% to 48% Mn)                3.5
      Ferric citrate (16% to  17% Fe)                    6.0
      Zinc carbonate (70% ZnO)                          1.6
      Cupric carbonate (53% to 55% Cu)                  0.3
      Potassium iodate (KI03)                           0.01
      Sodium selenite (Na2Se03-5H20)                    0.01
      Chromium potassium sulfate                        0.55
      Sucrose, finely powdered                          To make
                                                          1000.0

            Based on the National Academy of Sciences-National
      Research Council requirements for rats (National Academy of
      Sciences-National Research Council, 1972) .   To be used at
      3.5% of the diet.
          Source:  American Institute of Nutrition, 1977.
      Reprinted with permission of the publisher.
Page, and Saffiotti (1976) recommend sterilization of feed (without

degrading nutrients) when practical and consistent with the disease
control program of the laboratory.


2.4.4  Analysis for Nutrients and Contaminants

     Contaminants in the diet and variations in the concentration of
essential nutrients can influence the response of animals in toxicity

tests and thus alter the interpretation of experimental data (Food
Safety Council, 1978; Fox, 1977; Newberne, 1975).

     Variations in the concentration of essential nutrients can  occur

for many reasons:  batch or lot differences, regional and seasonal
differences, manufacturer variations, and processing.  Most diets con-

tain nutrients in quantities sufficient for the growth, maintenance,

-------
                                   32
 and reproduction of a particular species.   However,  concentrations of
 essential ingredients may vary from batch  to batch of  a formulation made
 with different lots of natural ingredients, and at the same  time  the
 guaranteed analysis shown on the label can remain correct  (Newberne,
 1975).
      Differences in manufacturing procedures were pointed  out by  Porter,
 Lane-Fetter,  and Home (1963), who compared growth rates of  groups of
 young mice fed diets produced in the same  region by  three  different
 manufacturers and found considerable differences (cited by Hurni, 1970).
 Also, nutritional quality may be seriously impaired  during processing,
 particularly  by overheating, which can destroy amino acids or lead to
 the formation of indigestible complexes (Clarke et al., 1977).
      These examples help to illustrate the need for  periodic analysis of
 diets for nutrient content, as proposed by the National Research  Council
 (1977).
      Because  of the presence of a variety  of contaminants  in animal
 diets it is also advisable to test periodically for  pesticides, myco-
 toxins,  trace minerals, and industrial contaminants  (such  as polychlorin-
 ated biphenyls, lead,  and mercury) (Food Safety Council, 1978; National
 Research Council,  1977; Newberne, 1975; Page, 1977).  Low  levels  of
 tf-nitrosamines have been detected in the diets of experimental animals
 in  Germany and in the United States (Edwards et al., 1979; Walker,
 Castegnaro, and Griciute, 1979).   The source of the  nitrosamines  is
 thought  to be fishmeal, and Knapka (1979)  suggested  that the ^-nitrosamines
 are present only in diets purchased in meal form.
      There is a growing interest  in obtaining detailed open-formula infor-
 mation for assurance that diets are free from pesticide residues  and
 other contaminants  (Food Safety Council, 1978);  if prospective analysis
 for contaminants cannot be arranged with feed suppliers, retrospective
 analysis  could be conducted.
      The  ultimate solution to the problems of uniform  constituents and
 toxic contaminants  in  animal diets may be  provided by  the  use of
rigorously purified  semisynthetic diets or entirely  synthetic diets
 (Clarke et al.,  1977;  Food Safety Council,  1978).

-------
                                   33
2.4.5  Effects of Diet on Toxicity Test  Results
     Nutritional imbalances may influence toxicity  through  physiological,
immunological, and/or biochemical mechanisms  (Campbell  and  Hayes,  1974).
The main effects, however, can be attributed  to  the effects of  nutritional
state on the drug-metabolizing enzymes located in the endoplasmic
reticulum of the hepatocyte.  This enzyme complex,  classified as a mixed-
function oxidase, is composed of cytochrome P-450,  phosphatidycholine,
and a flavoprotein reductase.  A wide variety of drugs  and  foreign
compounds are metabolized by the complex to products of greater or
lesser toxicity.  The response of the mixed-function oxidase system to
toxic substances can be modified by deficiency or excess of dietary
constituents such as protein, carbohydrates,  fats,  lipotropes,  vitamins,
and minerals.  Examples of these effects in animals will be reviewed in
the following sections.
     2.4.5.1  Protein — The dietary constituent  most studied is protein.
Qualitative and quantitative changes in  protein  content can alter  mixed-
function oxidase activities.  McLean and McLean  (1969)  pointed  out oppo-
site effects of protein deficiency on the toxicity  of compounds that are
detoxified and the toxicity of those that are rendered toxic by biotrans-
formation.  For example, protein-deficient diets protect against acute
poisoning from carbon tetrachloride and dimethylnitrosamine in rats
(McLean and McLean, 1966; McLean and Verschuuren, 1969), whereas the
toxicities of most pesticides are increased during protein deficiency
(Boyd, 1969).  Protein-deficient rats have become more susceptible to
the toxicity of aflatoxin when administered alone and of chloroform
following enzyme induction with DDT; but chloroform toxicity in a single
oral dose is not altered by protein deficiency  (Madhavan and Gopalan,
1965; McLean and McLean, 1969).  Severe protein depletion has been
implicated in the production of increased  resistance to mercuric  chloride
poisoning in rats (Surthin  and Yagi,  1958).
     Metabolism of a number of carcinogens is affected by  dietary protein
content.  The carcinogenic  action of  dimethylnitrosamine has been enhanced
in rats by protein deficiency  leading to  suppression of microsomal
hydroxylation (Swan and McLean, 1968).

-------
                                    34
      Silverstone and Tannenbaum (1951)  demonstrated that the formation
 of spontaneous hepatomas was substantially retarded by a decrease (from
 18% to 9%) in dietary protein, while increased protein intake (from 18%
 to 45%) had no effect on tumor formation.   The inhibition was found to
 be due to a deficiency in sulfur-containing amino acids in the diet, not
 to the difference in the proportion of  total protein.   Kawachi,  Hirata,
 and Sugimura (1968) demonstrated that addition of 1% tryptophan to the
 diets of rats receiving a low level of  #-nitrosodiethylamine increased
 the liver cancer incidence almost fourfold.  Bryan, Brown, and Price
 (1964) reported that L-tryptophan enhanced the induction of bladder
 tumors by 2-acetamidofluorene.
      In other studies, deficiencies of  protein and individual amino
 acids have been related to congenital malformations in rats.  Early
 experiments in reproduction and embryogenesis were reviewed by Kalter
 and Warkany (1959).
      2.4.5.2  Lipids — Lipids provide energy and essential fatty acids
 for synthesis of tissue and cell components.  High fat diets sensitize
 animals to the toxic effects of chloroform (Goldschmidt, Vars, and
 Raudin, 1939).
      Marshall and McLean (1969) showed  that dietary addition of either
 herring oil, linoleic acid, or 0.1% oxidized sitosterol was required for
 maximum induction of cytochrome P-450 synthesis.
      Rogers et  al.  (1974)  and Rogers (1975) found that liver tumor
 induction with  various carcinogens was  enhanced if the diet contained a
 high fat content and was deficient in choline,  methionine, and folic
 acid.   Carroll  and  Khor (1970)  reported that Sprague-Dawley rats on a
 high fat diet developed more mammary tumors after administration of
 7,12-dimethylbenzanthracene than did rats  on a low fat diet.
      2.4.5.3 Lipotropes — Lipotropes are  required for the synthesis of
 phospholipids,  which transport triglycerides.   Lipotrope deficiency is
 characterized by accumulation of triglycerides  in the  liver.   Aflatoxin
has  an  increased hepatotoxic effect in  rats on  a low lipotrope diet
 (Newberne, Rogers,  and  Wogan,  1968).  Newberne  and Rogers (1976)  reported
a higher  incidence  of #-2-fluorenylacetamide-  and dimethylbenzanthracene-

-------
                                   35
induced mammary tumors in Sprague-Dawley rats on a low lipotrope,  high
fat diet than in animals on a regular diet.   Lipotrope-deficient Fischer
rats, on the other hand, which were resistant to mammary tumor induction
with tf-2-fluorenylacetamide, developed a significantly higher number of
hepatic carcinomas than did controls.
     2.4.5.4  Vitamins — The role of vitamin deficiencies in the mixed-
function oxidase system has been reviewed comprehensively by Campbell
and Hayes (1974).  The effects do not appear to be as serious as the
effects of protein deficiencies, although activities are affected to
some degree if the vitamin deficiencies are severe enough.  The influence
of vitamins on the production of congenital malformations has been
reviewed by Kalter and Warkany (1959).
     Reports of the effects of vitamin A on experimental tumor induction
are conflicting.  Saffiotti et al. (1967) and Cone and Nettesheim (1973)
demonstrated inhibition by vitamin A of the induction of squamous
metaplasia and squamous cell tumors of the respiratory tract of rats and
hamsters with polycyclic hydrocarbons.  However, Smith et al. (1975) and
Smith, Rogers, and Newberne (1975) reported enhancement by vitamin A of
the induction of respiratory tract tumors by benzo(a)pyrene.
     2.4.5.5  Minerals — Deficiencies in dietary trace minerals, such as
iron, iodine, zinc, selenium, and copper, and their effects on the
mixed-function oxidase system have been reviewed by Campbell and Hayes
(1974).  Moffitt and Murphy (1974) have shown that the trace mineral
concentration can influence the metabolism, distribution, and toxic
action of chemicals.  The interactions between nutritional minerals and
toxic substances may be of a deleterious or protective nature  (Brinkman
and Miller, 1961; Farkas, 1978).  For example, Brinkman  and Miller
observed that toxicity to molybdnenum was increased  in rats whose diets
contained higher amounts of zinc  than usual.  Farkas, however,  suggested
that selenium may play a role in  decreasing the  toxicity of  cadmium and
mercury through  its interactions  with those metals.

2.4.6  Conclusions
     Diets of experimental  animals  should  contain proteins,  carbohydrates,
fats, minerals,  micronutrients, and energy in the proportions suitable

-------
                                    36
 for a particular species or strain.  Various types of diets available
 include nonpurified or cereal based, open-formula, closed-formula,  puri-
 fied or semisynthetic, and chemically defined.
      There is growing concern regarding contaminants in the diet and
 variations in the concentrations of essential nutrients and their con-
 sequent effects on the response of test animals to toxic substances.
 Periodic testing of diets for nutrient concentration and contaminants
 has been proposed, but the ultimate solution to the problem may be
 provided by the use of synthetic or semisynthetic diets.

 2.5  PATHOLOGY

 2.5.1  Introduction
      The importance of proper pathology examination and reporting tech-
 niques cannot be overemphasized (Prieur et al.,  1973).   They must be
 planned before the start of the experiment because they influence the
 test design and the selection of the animal models (Page,  1977).   Quality
 control must be emphasized at all times,  because important lesions  may
 be  lost at any of several steps:  necropsy, trimming,  fixation,  paraffin
 blocking,  or slide preparation (Page,  1977).  With the proper techniques
 and quality control,  a pathology workup gives information on the morphol-
 ogy of the chemically induced lesions present at that time and some
 indication of a dose-effect relationship  (Zbinden, 1976).   However, the
 limitations of pathology must be recognized,  including a lack of informa-
 tion on early lesions that disappear before necropsy,  the sequence  of
 lesion appearance,  the reversibility of lesions, and the developmental
 history (morphogenesis)  of each lesion (Zbinden, 1976).   The following
 subsections will discuss the procedures that should be used to obtain
 the desired pathology information.

 2.5.2   Gross  Examination
     The gross examination or necropsy is the first step in a pathology
 evaluation  and the most  influential factor in determining the toxic
 effects of  a  chemical.  Unless it is  done properly,  much valuable infor-
mation will be lost.   Therefore  the necropsy  should either be performed

-------
                                   37
directly by the pathologist (or the scientist who is to do the micro-
scopic examination) or by a trained technician with consultation from
the pathologist (Food Safety Council,  1978;  Magee, 1970;  Prieur et al.,
1973; Sontag, Page, and Saffiotti,  1976;  U.S. Food and Drug Administra-
tion, 1971; World Health Organization, 1978).  An extensive necropsy
(including blood counts, bone marrow smears, and organ weights) is a
desirable objective, but usually the design must be varied for each
experiment (Roe, 1965).
     To ensure that the maximum information is obtained from the test
animals, a daily observation should be included to reduce loss from
postmortem degeneration  (Magee, 1970; Roe, 1965).  The necropsy should
be done as soon as possible after death and, if not, the animal should
be placed in a refrigerator for storage  (Prieur et al., 1973).  Even if
some tissue degeneration has occurred, a necropsy should be performed,
because it can indicate  the general lesions  to be expected  (Magee, 1970;
World Health Organization, 1978).  If an animal is moribund,  sacrifice
is often required  and  should be determined based  on a  standard set of
criteria or on the experience  of the  observer  (Magee,  1970).   However,
the number of animals  sacrificed should not  exceed  the short-term capac-
ity of  the examiners  (World Health Organization,  1978).
     The organs examined should all be identified regardless  of  the
results  (Roe, 1965).   It is often  easiest and  most  thorough to use a
checklist  system  (U.S.  Food and Drug  Administration,  1971;  World Health
Organization, 1978).   The  examination of  each  organ should be performed
in a standard procedure, with  the  same slices  and angles  examined for
every test animal  (Roe,  1965).  During the  examination,  all gross
lesions observed  should be recorded and  described,  including  information
on the  size, location, number, shape, color, and texture (Sontag, Page,
and  Saffiotti,  1976;  World Health  Organization,  1978).  The weight of
the  organ  can  also be determined during  a gross examination,  but the
results are  of  doubtful significance  for lifetime studies (Magee, 1970).
If weight  is to be determined, it  should be done as soon as possible to
reduce  drying  effects (World Health Organization, 1978).
     The necropsy should start with an external examination including
orifices  (Sontag,  Page, and Saffiotti,  1976; World Health Organization,

-------
                                   38

1978).  Next, examine all the internal cavities and organs in situ and
do not separate the connective tissue until it also has been examined
(Prieur et al., 1973).  Table 2.8 lists some tissues that have been sug-
gested for examination.  Special procedures for individual organs have
also been recommended:  (1) parenchymal and endocrine glands should be
thoroughly examined, including multiple cut slices, since these tissues
often contain deep-seated lesions (Sontag, Page, and Saffiotti, 1976;
Food Safety Council, 1978); (2) all hollow organs, including the urinary
bladder, gastrointestinal tract, and respiratory tract, should be cut
open and extensive examination of the mucosal surfaces performed (Food
Safety Council, 1978; Sontag, Page, and Saffiotti, 1976; World Health
Organization, 1978); (3) the skull and brain should be cut open and
examined, including examination of the pituitary (Magee, 1970); and  (4)
                Table 2.8.  Tissues to be included in a
                           gross examination

        Gross lesions                          Heart
        Tissue masses or suspect tumors        Thyroids
          and regional lymph nodes             Parathyroids
        Skin                                   Esophagus
        Mandibular lymph node                  Stomach
        Mammary gland                          Duodenum
        Salivary gland                         Jejunum
        Larynx                                 Ileum
        Trachea                                Spleen
        Cecum                                  Kidneys
        Colon                                  Adrenals
        Rectum                                 Bladder
        Mesenteric lymph node                  Seminal vesicles
        Liver                                  Prostate
        Thigh muscle                           Testes
        Sciatic nerve                          Ovaries
        Sternebrae, vertebrae, or femur        Uterus
          (plus marrow)                        Nasal cavity
        Costochondral junction, rib            Brain
        Thymus                                 Pituitary
        Gallbladder                            Eyes
        Pancreas                               Spinal cord
        Lungs and bronchi

             Source:   Adapted from Sontag, Page, and Saffiotti,
        1976.

-------
                                  39
the spinal cord in rodents is best examined in situ (World Health
Organization, 1978) and often is not routinely examined (Magee, 1970).
     In carcinogenicity studies, the gross examination is even more
important than usual because much of the lesion incidence data are gen-
erated during this procedure (Food Safety Council, 1978).  To evaluate
the effectiveness of the necropsy to detect lesions, Frith et al. (1979)
and Kulwich et al. (1980) correlated observations of the gross and
microscopic examinations in carcinogenicity studies.  Table 2.9 shows
how frequently the gross examination detected lesions also seen in
microscopic examinations in mice.  Frith et al. (1979) concluded that
the size of the organ, the stage and nature of the lesion, and the
number of slices examined all affect the efficiency of the gross examin-
ation.  Kulwich et al. (1980) found correlation rates between  gross and
microscopic examinations of 70% and 76% for two chemicals in rats.  They
concluded that gross examination is less successful for small  organs,
because a single histological slice represents a  larger portion of the
organ and can often find lesions not seen grossly.  However, for tissues
like the skin and mammary gland, the gross examination is much more
successful.  They also found that the size and nature of  the lesions
affected the gross detection.
    Table 2.9.  Correlation between gross and microscopic lesions in
                      carcinogenic studies in mice
Organ
Liver
Thymus
Spleen
Mammary tissue
Uterus (adenocarcinomas)
Uterus (polyps)
Lung
Adrenal gland
Harderian gland
Testes
Pituitary gland
Lesions
seen in
gross exam
5 550
725
6 589
6 503
50
2 423
4 503
711
1 438
330
486
Lesions seen
microscopically
5 968
1 480
8 340
6 993
81
6 922
10 007
1 776
4 637
402
836
Percentage
found in
gross
93
49
79
93
62
35
45
40
31
82
58
     Source:  Adapted from Frith et al., 1979.

-------
                                    40
 2.5.3  Tissue Preservation and Storage
      After the gross examination,  tissues  should  be  excised and preserved
 to prevent further degeneration.   This should be  done  as  quickly as
 possible,  since degeneration can  occur quite rapidly,  especially with
 mice (Magee,  1970).   Several techniques for preservation  are  available,
 with immersion being the most common (World Health Organization, 1978).
 However,  for  some tissues (lung and central nervous  system) and for
 electron  microscopy, perfusion is preferred.   This involves draining the
 blood,  followed by injection of a proper fixative (World  Health Organi-
 zation, 1978).  Perfusion cannot  be used if organ weights are needed
 (World  Health Organization, 1978).
      The  fixative most recommended is neutral buffered formalin at
 concentrations of 10% (Prieur et  al., 1973; Sontag,  Page, and Saffiotti,
 1976; World Health Organization,  1978) or  4% (Zbinden, 1976). Special
 preservatives are often required  (e.g., Bouin's solution  for  paraffin-
 embedded  material) depending on the stain  to be used and  the  tissue
 involved  (Prieur et  al., 1973; Zbinden, 1976).  The  tissues should be
 preserved at  a maximum thickness  of 0.5 cm (Prieur et  al., 1973; Sontag,
 Page, and Saffiotti, 1976;  World  Health Organization,  1978).  Some tis-
 sues, like the testicles, should  first be  immersed intact and then
 sliced  in 0.5-cm sections for further preservation (Zbinden,  1976).  The
 tissue/preservative  ratio on a volume basis should be  greater than 1:10
 (World  Health Organization, 1978)  and preferably  1:15  to  1:20 (Prieur
 et  al., 1973).   The  tissues should be left in the preservative for at
 least 24 h, and up to 72 h  has been suggested (Prieur  et  al., 1973;
 Sontag, Page,  and Saffiotti,  1976; World Health Organization, 1978).
 Often special procedures are suggested, including:   covering  tissues
 that  float with absorbant material (e.g.,  cheesecloth) for uniform
 preservation  (Prieur et  al.,  1973);  skin,  stomach mucosa, and nerves
 need  to be straightened  out and often attached to a  card  or filter paper
 to prevent crumpling (Prieur et al.,  1973;  World  Health Organization,
 1978; Zbinden,  1976);  several thoracic-lumbar vertebrae should be fixed
 in situ with  the  spinal  cord (Sontag,  Page,  and Saffiotti, 1976); urinary-
bladder should  be filled with fixative (Zbinden,  1976); and multiple
representative  samples of variable tissue  masses  should be fixed (Sontag,

-------
                                   41
Page, and Saffiotti,  1976).   After preservation,  the material should be
stored in plastic bags or jars and kept at least  until  the final con-
clusions are made for the study (Food Safety Council, 1978).

2.5.4  Trimming, Staining, and Embedding of Tissue
     The preparation of tissues for microscopic evaluation includes
techniques for trimming, staining, and embedding.   The  tissues should be
sampled from the same specific sites for each organ (Prieur et al.,
1973), and the slices should be made so that the  cut surfaces represent
the maximum possible area for examination (World  Health Organization,
1978).  Often a set trimming schedule should be used.  The trimming
should be performed by or in the presence of the  pathologist, with con-
sideration of the gross examination observations  (Sontag,  Page, and
Saffiotti, 1976).  The tissues should be trimmed  to a thickness of 2 to
3 mm for histologic processing (Sontag, Page, and Saffiotti, 1976; World
Health Organization, 1978).   Specific recommendations for trimming of
certain organs have been made by the National Cancer Institute  (Sontag,
Page, and Saffiotti, 1976) and the World Health Organization  (1978).
These recommendations include:  (1) multiple portions of large masses or
tumors should be submitted with some of the normal surrounding  tissue;
(2) parenchymal organs  (e.g., liver) should be sliced to give the
maximum observable area;  (3) the kidneys should be sampled through the
cortex and medulla, one by a midlongitudinal section and the  other by  a
midtransverse section;  (4) the lungs should be sectioned transversely
(parallel to the body axis) including  the bronchi and carina;  (5) at
least three brain cross sections are needed, with one through the
frontal cortex and basal  ganglia, one  through the parietal  cortex and
thalamus, and one through the cerebellum with the pons;  (6)  the hollow
organs should be trimmed  to include a  cross-section slide  from mucosa  to
serosa; (7) if  the larnyx is  to be examined,  the section  should also
include the pharnyx; and  (8)  if the nasal  cavity is to  be  examined,
three transverse sections are needed.
     Routinely,  the  trimmed slices  should  be stained with hematoxylin
and eosin and embedded  in paraffin  (Zbinden,  1976).  These are sliced to
4 to 6 ym for microscopic slides  (Sontag,  Page,  and Saffiotti, 1976;

-------
                                    42
 World Health Organization, 1978).   The paraffin embedding can be  per-
 formed using automation techniques.  Often semithin slices (1 ym)  are
 needed for such organs as the bone marrow, kidney,  or endocrine glands
 (World Health Organization,  1978).   Special stains  are often  employed,
 for example in evaluations for the presence of  fats,  carbohydrates,
 or special structures (World Health Organization, 1978;  Zbinden,  1976).
 These can in turn necessitate special preservation  techniques.  Obviously,
 to obtain the maximum information  from the ensuing  microscopic evaluation,
 proper planning prior to the testing is necessary,  so that the special
 techniques for tissue preservation, staining, and embedding can be
 coordinated.

 2.5.5  Microscopic Examination
      The microscopic examination of properly prepared tissue  slides is
 usually the final step in a pathology evaluation.   Although its importance
 is overwhelmingly accepted,  the details of the  design are quite variable,
 depending on the type of toxicity  test employed.  The main questions are
 which tissues to examine,  for how  many animals  and  levels, and which
 evaluation techniques to use.
      Many recommendations have been made concerning the  organs that
 should be examined.   Section 4.5 and Sect. 5.6.3 review  some  of the
 specific literature  discussions for individual  toxicity  tests.  In
 general,  the choice  depends  on the scope of the test  and is decided by
 the pathologist  (Magee,  1970).   The primary problem is a conflict  between
 the desire to obtain the maximum amount of scientific information, and
 the limits imposed by practical economic and time constraints.  Therefore,
many  discussions recommend only vague guidelines, although others  are
extremely  specific and encompassing (Page, 1977).   As an attempt  to
reach  a  compromise solution,  Zbinden (1976)  recommended  a priority
system based on  the  frequency  of morphological  changes that are likely
to occur  (Table  2.10).   This  system addresses both  the specific organs
to be  examined and the test animals from each dose  level from which the
organs are  to be taken.   It is  obvious that, with the number  of chemicals
to be  evaluated,  the  microscopic examination will have to be  based on
some similar  type  of  ranking system.

-------
                                 43
         Table 2.10.  Organs and tissues to be examined in
                      routine toxicity tests
First Priority:  All animals including controls
Liver (f)a
Kidney (f)
Adrenal glands (£)
Heart (left ventricle) (f)
Spleen (f)
Thymus
Testis
Epididymis
Lung
Bone marrow
Mesenterial lymph node
All organs showing gross changes
  of shape, weight, color, or
  structure
Second Priority:  High dose animals and part of controls.  Medium
and low dose animals only when significant changes observed in
high dose group
Thyroid
Parathyroid
Pituitary gland
Salivary glands
Stomach
Duodenum
Small intestine
Large intestine
Pancreas
Ovary
Uterus
Cervix uteri
Vagina
Seminal vesicles
Prostate
Coagulating gland
Tonsils
Brain
Spinal cord
Eye
Optic nerve
Peripheral nerve
Urinary bladder
Skin
Mammary gland
Bone-cartilage
Skeletal muscle
Gallbladder
Third Priority:  Organs and tissues not examined in routine
experiments, unless indicated by clinical observations or motivated
by scientific interest
Heart valves
Purkinje fibers
Aorta and other blood vessels
Thoracic duct
Tongue
Teeth
Lips
Gingivae
Hard palate
Nasopharynx
Larynx
Trachea
Esophagus
Vermiform appendix
Rectum
Anus
Ureter
Urethra
Oviduct
Vulva
Penis
Paraurethral and preputial  glands
Pineal gland
Spinal ganglions and  roots
Sympathetic ganglions and trunk
Nerve fiber endings
Meninges
External  ear
Middle ear
Inner ear
Olfactory organ
Subcutaneous lymph nodes
Tendons
Adipose  tissue
Intervertebral disc
Synovial  membrane
Lacrimal  glands
       (f) — frozen sections  stained  for  fat  obligatory.
     Source:  Adapted from Zbinden,  1976.  Reprinted by  permission
of the publisher.

-------
                                    44

      Another approach to reducing the amount of microscopic examination
 has been proposed by Fears and Douglas (1977; 1978).   On the basis of  a
 statistically valid sampling method,  they suggest  examination of  all of
 the treatment groups first and then limited examination  of  the  controls.
 The reduction occurs by limiting examination of the control groups to
 tissues that were found to be affected in the treatment  groups.   They
 also suggest several further modifications of this system,  which  result
 in even less pathology.  One involves random sampling of the treatment
 groups based on gross observations to reduce pathology in those groups.
 Another incorporates techniques to allow for "blind"  evaluations  where
 the potential biases of the investigator are controlled.  These approaches
 can reduce the microscopic examination by 10% to 50%.  However, Kulwich
 et al. (1980) and Frith et al. (1979), in their evaluations of  the
 correlation between the gross and microscopic examinations, found that
 one cannot rely totally on the gross  examination to select  affected
 tissues for microscopic study.  They  stressed that a  thorough microscopic
 examination is still necessary.  Therefore, the approach suggested by
 Fears and Douglas still needs more refinement, but represents a potential
 solution for reducing the amount of microscopic examination.
      No matter what organs are examined microscopically,  the information
 will have to be evaluated and presented in a uniform  manner. It  is
 often necessary to use a checklist to ensure that  all intended  organs
 were examined (World Health Organization, 1978).   Also,  standard  criteria
 should be developed and reported for  the descriptions and classifications
 of the lesions,  especially if these are done semiquantitatively (World
 Health Organization,  1978).   The formation of tumor tables  incorporating
 information on the group, organ, and  sex variables for each lesion can
 be useful for comparing chemical effects (World Health Organization,
 1978).
      The  manner  in which the slides are examined is also an issue of
 debate.   It  has  been  suggested that the slides should be examined by the
 pathologist  without knowledge of the  treatment or  control group repre-
 sented  (U.S.  Food  and Drug Administration,  1971).  This  "blind" evaluation
would reduce possible biases introduced by the pathologist  and  produce
more objective conclusions (Fears  and Schneiderman, 1974, as cited in
Page, 1977).  However,  many  other  reviewers stress that  the danger of

-------
                                  45
missing lesions or incorrectly interpreting the results far outweighs
bias problems, so that the pathologist should be aware of the sample
group (Roe, 1965; World Health Organization, 1978;  Zbinden, 1976).   The
Food Safety Council (1978) suggests a compromise situation where the
control groups are evaluated first and the treatment groups are done
"blindly."  In any case, maximum use should be made of the results of
prior clinical studies and gross examinations (Food Safety Council,
1978; World Health Organization, 1978; Zbinden, 1976).

2.5.6  Conclusions
     Pathology is extremely useful in toxicity testing and can provide
information on the chemically induced lesions present at the time of
sacrifice and any dose-effect relationship.  To obtain a useful pathology
evaluation several requirements must be met:  (1) the pathologist should
perform or supervise all steps in the protocol; (2) the gross necropsy
should be extremely thorough and include every animal in the study;  (3)
the tissues should be properly preserved in an accepted fixative immedi-
ately after the necropsy;  (4) the staining, embedding, and sectioning of
the tissues must be well planned and coordinated, particularly  if
special stains are utilized; and (5) the microscopic  examination should
be as thorough as practical limitations will allow.   Only  if these
requirements are met will  the pathology contribute meaningful data  for
evaluations of toxicity.

2.6  PHARMACOKINETICS

2.6.1  Introduction
     Pharmacokinetics is  the quantitative  and  qualitative  description
of the time course of absorption, distribution, metabolism,  and elimina-
tion of an agent in an  intact animal  (Young and Holson,  1978).   The
following sections will examine some  of the basic  concepts of  pharmaco-
kinetics and show how they can be used to  supplement the routine
toxicological data and  aid in the hazard  evaluation of chemicals.

-------
                                   46
2.6.2  Basic Concepts
     Withey  (1978) describes absorption, distribution, metabolism, and
elimination  as  the four major rate processes that are involved when a
substance  is administered  to a human or animal subject.  He further
describes  these as follows:
 1.  absorption — to the bloodstream from the site of administration;
 2.  distribution — to organs, tissues, storage depots, and receptor
     sites;
 3.  metabolism — usually  in the  liver but  in other organs also; and
 4.  elimination  — mainly  via the urine or  feces but also via the
     lung, from sweat and  skin, and  into hair.
     Gehring, Watanabe, and Blau  (1976) present a modification of a
graphical  illustration published  by  Garrett in 1971 of a hydraulic
system used  as  an analogy  of a three-compartment body model portraying
the  general  concepts of pharmacokinetics  (Fig. 2.1).  The amount of
chemical corresponding to  the original dosage in the blood before any
distribution to other tissue compartments is represented by D , whereas
the  amount of chemical at  any given  time in the vascular compartment is
represented  by  A  .  In both the a and b parts of Fig. 2.1, the amount of
chemical in  the vascular compartment (A ) has equilibrated with the
amount of  the chemical in  the shallow or rapidly equilibrating tissue
compartment  (T) but not with the  amount of  chemical in the deep-seated
or slowly  equilibrating tissue compartment  (2"); 2"    is the maximum
                                                   in£i,x>
amount of  chemical that will exist in the deep-seated tissue compartment.
The  amount of chemical eliminated by the lungs and urine or metabolized
is represented  by E and is equal  to  D  -  (A + T + 2").  As time
approaches infinity, E -+ E<» = D .
     Gehring, Watanabe, and Blau  (1976) define a compartment in the body
as those organs,  tissues,  and cells  for which the rates of uptake and
clearance of a  chemical are so similar as to not permit resolution.  The
rapidly equilibrating compartment can be compared to those tissues with
a profuse compartment blood supply,  whereas the slowly equilibrating
compartment can be compared to those tissues such as fat and bone that

-------
                                  47
                               ORNL-DWG  80-18285




1
\
*
E°









r





^^
•-
>


















i









|DO
1
1
I Av
1

T
? kF
— e 	 fq
o
0
0
o
o
o
0
o
u







=c
















t
















T'
'MAX
T'
1











                                 (a)


Av


J
T'
'MAX
T'
                          .00
>
 0
 TB-
 O
 O
                                (b)

     Fig.  2.1.   Hydraulic analogy  of  three-compartment body model.  Do
is the original dose in the vascular  compartment prior to any equilibra-
tion at any time.   AV is the amount in the vascular  compartment at any
time.  In both (a)  and (b), Av has equilibrated with the shallow or
rapidly equilibrating compartment  T.   3"  represents  the level in the
deep or slowly equilibrating compartment, and  T'mayi  is the maximum level
achieved in the deep compartment.   E  is the  amount of chemical eliminated
or metabolized, and E<*> corresponds to the amount eliminated or metab-
olized at infinite time.  ke represents the  overall  rate constant for
the elimination of the chemical from  the body. Modified from Garrett.
(1971).  Source:  Gehring, Watanabe,  and Blau, 1976. Reprinted with
permission of the publisher.

-------
                                    48
 have a more limited blood supply.   Phannacokinetic  rates may be
 influenced by such factors as the  physical characteristics of the sub-
 stance (e.g., solubility, molecular weight,  and  surface area or solid
 particles), pharmacologic properties (e.g.,  peristalsis and blood flow),
 gastrointestinal functions (e.g.,  gastric emptying  and intestinal
 transit and motility),  permeability of membrane  barriers at the site of
 absorption, local blood flow, metabolism, and specialized transport
 mechanisms (Withey, 1978).

 2.6.3  Utility of Pharmacokinetics
      One example of the use of pharmacokinetics  as  an aid in the hazard
 evaluation of chemicals is provided by a study of the absorption of
 methylcellulose (Braun, Ramsey, and Gehring, 1974).   Administration of
 ll*C-labeled methylcellulose (viscosity of 3300 centipoise and molecular
 weight of 77 000) in both single and repeated oral  daily doses of
 500 mg/kg to rats showed that virtually none of  the methylcellulose was
 absorbed,  with essentially all being excreted in the  feces.  Therefore,
 if  it can be demonstrated that no  absorption occurs there is no need
 for further ingestion studies to ascertain systemic toxicity.
      Another example of the utility of pharmacokinetics in the toxico-
 logical evaluation of chemicals is the oral toxicity  study of 6-chloro-
 picolinic acid (6-CPA), a degradation  product of 2-chloro-6-(trichloro-
 methyl)pyridine (CTP)  (Gehring, Watanabe, and Blau, 1976).  Analysis of
 the urine of rats given single doses of 10 mg/kg of llfC-labeled 6-CPA
 revealed that 30% of the 1UC activity  present was attributable to 6-CPA
 and 62% to the glycine  conjugate.   The rapid (half-life, 1.1 ± 0.2. h) and
 complete elimination of 6-CPA and  its  glycine conjugate from the rat
 suggested  that ingestion of trace  amounts of 6-CPA  does not constitute a
hazard.  Gehring,  Watanabe,  and Blau state that, as expected from the
pharmacological  evaluation,  a 90-day toxicological  study in which rats
were given  daily doses  of 6-CPA gave no indication  of cumulative toxicity.
     An  example  of the  utility of  pharmacokinetics  to the pharmaceutical
industry is  given by Nelson (1976).  A hypothetical situation is described
of an antibiotic  that is  being developed for deep-seated tissue infection.

-------
                                   49
Kinetic analysis of this antibiotic in animals  or  human  volunteers  shows
the drug to have a minimal distribution with a  rapid elimination half-
life; furthermore, renal excretion kinetics  indicate that  it  is  elimin-
ated by both glomerular filtration and tubular  secretion.   It is
therefore doubtful that high serum and tissue levels of  this  antibiotic
could ever be realistically achieved,  and thus  the drug  would not be
useful for deep-seated tissue infections. The  renal excretion kinetics,
however, suggest that the antibiotic could be useful in  treating urinary
infection.  Nelson states that by performing such  pharmacokinetic studies
many costly and needless clinical studies would be circumvented.
     One other example of the value of pharmacokinetics  in toxlcological
studies is shown by the work of Piper et al. (1973).  The  herbicide
2,4,5-trichlorophenoxyacetic acid (2,4,5-T)  was administered to  rats and
dogs in single oral doses of 5 mg/kg with the resultant  half-life
clearance values being 4.7 and 77.0 h, respectively.  It was demonstrated
by Piper et al. that these rates of elimination can be correlated with
the toxicity of 2,4,5-T in rats and dogs.  The slower rate of elimination
by dogs than by rats correlated with the higher toxicity in dogs for
which the single oral LDso is 10° mg/kg compared to 300 mg/kg for rats
(National Institute for Occupational Safety and Health,  1977).  These
correlations support the statement of Gehring and Young (1978)  that there
is an inverse relationship between the  toxicity of  an agent  to  an
individual or species and the ability of the individual or species  to
eliminate the chemical and a direct relationship between toxicity  and
the concentrations in the plasma.
     In the testing of a chemical for teratogenicity, a knowledge  of
its pharmacokinetics would be beneficial in explaining  the teratogenic
effects or lack of such effects.  Teratogenic drugs that are dissolved,
absorbed, metabolized, and excreted rather  quickly  and  that  are
administered during the critical  period of  organogenesis will cause
malformations because the action  of the drug will occur shortly after
administration  (Schardein, 1976).  An  example  is  8-aminopropionitrile,
which is metabolized so quickly to its  inactive metabolite,  cyanoacetic
acid, that if day 15  (the critical period for  induction of cleft palate)

-------
                                  50

is not included in the dosing regime, very few malformations are
produced in the rat (King, Horigan, and Wilk, 1972).  However, there are
some teratogenic drugs that are absorbed or metabolized so slowly or
incompletely that their teratogenicity would not be manifested if
administered during the critical period.  An example of this type of
action is provided by the antihypercholesterolemic drug triparanol, now
withdrawn from use, which has been shown to be an active teratogen in
rats when administered as early as day 4 prior to implantation (Roux,
1964, as cited by Schardein, 1976).

2.6.4  Conclusions
     The four rate processes of pharmacokinetics are absorption, distri-
bution, metabolism, and elimination.  Each of these is important in
determining the fate of a chemical in the body and can be influenced by
such factors as the physical characteristics of the test substance,
pharmacologic properties, and gastrointestinal functions.  Knowledge of
the pharmacokinetics of a chemical can aid in the hazard assessment
process by demonstrating in some cases the lack of potential toxicity
because of lack of absorption into the body or a fast elimination from
the body and in other cases by explaining why a chemical is more toxic
to one species of laboratory animal than to another.

-------
                                   51

                       REFERENCES FOR SECTION 2
American Institute of Nutrition.   1977.   Ad Hoc Committee Report on
  Standards for Nutritional Studies.   J.  Nutr.  107:1340-1348.

Arnold, D. L., S. M. Charbonneau, Z.  Z.  Zawidzka,  and H. C. Grice.  1977.
  Monitoring Animal Health During Chronic Toxicity Studies.  J. Environ.
  Pathol. Toxicol. 1:227-239.

Boyd, E. M.  1969.  Dietary Protein and Pesticide Toxicity in Male Wean-
  ling Rats.  Bull. WHO 40:801-805.

Braun, W. H., J. C. Ramsey, and P. J. Gehring.   1974.  The Lack of
  Significant Absorption of Methylcellulose, Viscosity 3300 cps from
  the Gastrointestinal Tract Following Single and Multiple Doses to
  the Rat.  Food Cosmet. Toxicol. 12:373-376.

Brinkman, G. L., and R. F. Miller.  1961.  Influence of Cage Type and
  Dietary Zinc Oxide upon Molybdenum Toxicity.   Science 134:1531-1532.

Bryan, G. T., R. R. Brown, J. M.  Price.  1964.   Incidence of Mouse Blad-
  der Tumors Following Implantation of Paraffin Pellets Containing Certain
  Tryptophan Metabolites.  Cancer Res. 24:582.

Campbell, T., and J. R. Hayes.  1974.  Role of Nutrition in the Drug-
  Metabolizing Enzyme System.  Pharmacol. Rev.  26:171-197.

Carroll, K. K., and H. T. Khor.  1970.  Effects of Dietary Fat and Dose
  Level of 7,12-Dimethylbenz(a)anthracene on Mammary Tumor Incidence in
  Rats.  Cancer Res. 30:2260-2264.

Clarke, H. E., M. E. Coates, J. K. Eva, D. J. Ford, C.  K. Milner,
  P. N. O'Donoghue, P. P. Scott, and R. J. Ward.  1977.  Dietary  Standards
  for Laboratory Animals:  Report  of the Laboratory Animals Centre Diets
  Advisory Committee.  Lab. Anim.  11(1):1-28.

Clough, G.   1976.  The Immediate Environment of the Laboratory Animal.
  In:  Laboratory Animal Handbooks 7:  Control of the Animal  House Envi-
  ronment, T. McSheehy, ed.  Laboratory Animals, Ltd.,  Huntingdon, United
  Kingdom,   pp. 77-94.

Coates, M. E.,  P. N. O'Donoghue, P. R. Payne, and R. J. Ward, eds.   1969.
  Dietary  Standards for Laboratory Rats  and Mice.   In:   Laboratory Animal
  Handbooks  2.  Laboratory Animals, Ltd.,  London,   pp.  1-31.

Cone, M. V., and P. Nettesheim.   1973.   Effects of  Vitamin A on 3-Methyl-
  cholanthrene-Induced Squamous  Metaplasias  and Early  Tumors  in the
  Respiratory  Tract of Rats.   J.  Natl. Cancer  Inst.  50:1599-1606.

-------
                                   52

Dymet, J.   1976.  Air Filtration.  In:  Laboratory Animal Handbooks 7:
  Control  of  the Animal House Environment, T. McSheehy, ed.  Laboratory
  Animals,  Ltd., Huntingdon, United Kingdom,  pp. 209-246.

Edwards, G. S., J.  G. Fox, P. Policastro, U. Goff, M. H. Wolf, and
  D. H. Fine.  1979.  Volatile Nitrosamine Contamination of Laboratory
  Animal Diets.  Cancer Res. 39:1857-1858.

Farkas, C.  S.  1978.  Importance  of Interactions Between Nutrients and
  Environmental Contaminants as a Factor  in Experimental Design in Toxi-
  cological Research, with Emphasis on  Selenium and Ascorbic Acid.  Sci.
  Total Environ. 9(2):148-160.

Fears, T.  R.,  and J. F. Douglas.   1977.   Suggested Procedures for Reducing
  the Pathology Workload  in a Carcinogen  Bioassay Program, Part I.  J.
  Environ.  Pathol.  Toxicol. 1:125-137.

Fears, T.  R.,  and J. F. Douglas.   1978.   Suggested Procedures for Reducing
  the Pathology Workload  in a Carcinogen  Bioassay Program, Part II:
  Incorporating Blind Pathology Techniques and Analysis for Animals with
  Tumors.   J.  Environ. Pathol. Toxicol. 1:211-222.

Testing, M.F.W.  1979.  Properties of Inbred Strains  and Outbred Stocks,
  with Special Reference  to Toxicity Testing.  J. Toxicol. Environ. Health
  5:53-68.

Fletcher,  J. L.  1976.  Influence of Noise on Animals.  In:  Laboratory
  Animal Handbooks  7:  Control of the Animal House Environment,
  T. McSheehy, ed.  Laboratory Animals, Ltd., Huntingdon, United Kingdom.
  pp. 51-62.

Flynn, R.  J.   1967.  The  Control  of Disease in Laboratory Animals.  In:
  Husbandry of Laboratory Animals, Proceedings of the Third International
  Committee on Laboratory Animals, M. L.  Conalty, ed.  Academic Press,
  New York.  pp. 361-372.

Food Safety Council.  1978.  Proposed System for Food Safety Assessment.
  Columbia, Maryland.  136 pp.

Fouts, J. R.   1976.  Overview of  the Field:  Environmental Factors
  Affecting Chemical or Drug Effects in Animals.  Fed. Proc. Fed. Am.
  Soc. Exp. Biol. 35(5):1162-1165.

Fox, J.  G.  1977.  Clinical Assessment  of Laboratory  Rodents on Long Term
  Bioassay  Studies.  J. Environ.  Pathol. Toxicol. 1:199-226.

Frith, C. H., A.  D. Boothe, D. L.  Greenman, and J. H. Farmer.  1979.
  Correlations Between Gross and  Microscopic Lesions  in Carcinogenic
  Studies in Mice.   J. Environ. Pathol. Toxicol. 3:139-153.

-------
                                  53

Gehring, P. J., P- G.  Watanabe,  and G.  E.  Blau.   1976.   Pharmacokinetic
  Studies in Evaluation of the Toxicological and  Environmental Hazard
  of Chemicals.  In:   Advances in Modern Toxicology,  Vol.  1,  Part  1:
  New Concepts in Safety Evaluation, M.  A.  Mehlman,  R.  W.  Shapiro, and
  H. Blumenthal, eds.   John Wiley and Sons, New York.  pp.  195-270.

Gehring, P. J., and J. D. Young.  1978.  Application of Pharmacokinetic
  Principles in Practice.  In:  Proceedings of the First International
  Congress on Toxicology, G. L.  Plaa and W.A.M. Duncan, eds.   Academic
  Press, New York.  pp. 119-141.

Goldschmidt, S., H. M. Vars, and I. S.  Raudin. 1939.  Influence of the
  Foodstuffs upon the Susceptibility of the Liver to Injury by Chloroform,
  and the Probable Mechanism of Their Action.  J. Clin. Invest. 18:277-289.

Grange, P. H.  1976.   Design Considerations Relevant to Controlled Envi-
  ronment Buildings.   In:  Laboratory Animal Handbooks 7:   Control of the
  Animal House Environment, T. McSeehy, ed.  Laboratory Animals, Ltd.,
  Huntingdon, United Kingdom,  pp. 291-299.

Hatch, A., T. Balazs, G. S. Wiberg, and H. C. Grice.  1965-  The Impor-
  tance of Avoiding Mental Suffering in Laboratory Animals.  Animal
  Welfare Inst. Rep.  14 No. 3.

Heinecke, H.  1967.  The Experimental Environment and Its Influence on
  the Blood Picture of Laboratory Animals.  In:  Husbandry of Laboratory
  Animals, Proceedings of the Third International Committee on Laboratory
  Animals, M. L. Conalty, ed.  Academic Press, New York.  pp. 447-457.

Hughes, H. C., Jr., and C. M. Lang.  1978.  Basic Principles  in Selecting
  Animal Species for Research Projects.  Clin. Toxicol. 13(5):611-621.

Hurni, H.  1970.  The Provision of Laboratory Animals.  In:   Methods  in
  Toxicology, G. E. Paget, ed.  F. A. Davis Company, Philadelphia.
  pp. 11-48.

Kalter, H., and J. Warkany.   1959.  Experimental Production  of Congenital
  Malformations in Mammals by Metabolic Procedure.   Physiol.  Rev.
  39:69-115.

Kawachi, T., Y. Hirata,  and T.  Sugimura.   1968.  Enhancement of #-Nitros-
  odiethylamine Hepatocarcinogenesis by L-Tryptophan in Rats.  Gann 59:
  523-525.

King, C.T.G.,  E. Horigan, and A.  L. Wilk.   1972.  Fetal Outcome from
  Prolonged Versus Acute Drug Administration in  the Pregnant Rat.  In:
  Drugs and Fetal Development,  Proceedings of an International Symposium
  on the Effect of Prolonged  Drug Usage on Fetal Development,
  M. A. Klingberg, A.  Abramovici, and  J.  Chemke, eds.   Plenum Press,
  New York.  pp.  61-75.

-------
                                    54

Knapka,  J.  J.   1979.  Laboratory Animal Feed.  Science 204:1367.

Kulwich,  B. A.,  J.  F. Hardisty, C.  E. Gilmore, and J. M. Ward.  1980.
   Correlation  Between Gross  Observations of Tumors and Neoplasms Diag-
   nosed  Microscopically  in Carcinogenesis Bioassays in Rats.  J. Environ.
   Pathol. Toxicol.  3:281-287.

Lane-Petter, W.   1967.   Selection,  Training, and Control of Staff.  In:
   Husbandry of Laboratory Animals,  Proceedings of the Third International
   Committee on Laboratory Animals,  M. L. Conalty, ed.  Academic Press,
   New York.  pp.  61-74.

Madhavan, T. V.,  and C.  Gopalan.   1965.  Effect of Dietary Protein on
   Aflatoxin Liver Injury in  Weanling Rats.  Arch. Pathol. 80:123-126.

Magee, P.   1970.   Tests  for  Carcinogenic Potential.  In:  Methods in
   Toxicology,  G.  Paget,  ed.  F. A.  Davis Company, Philadelphia,  pp.
   158-196.

Marshall, W. J.,  and A.E.M.  McLean.  1969.  A Requirement for Dietary
   Lipids for Induction of Cytochrome P-450 by Phenobarbitone in Rat Liver
   Microsomal Fraction.   Biochem. J.  122:569-573.

McLean,  A.E.M.,  and E. K.  McLean.   1966.  Effect of Diet and 1,1,1-
   Trichloro-2,2-bis(p-chlorophenyl)ethane  (DDT) on Microsomal Hydrox-
   ylating Enzymes and on Sensitivity of Rats to Carbon Tetrachloride
   Poisoning.   Biochem. J.  100:564-571.

McLean,  A.E.M.,  and E. K.  McLean.   1969.  Diet and Toxicity.  Br. Med.
   Bull.  25:278-281.

McLean,  A.E.M.,  and H. G.  Verschuuren.  1969.  Effects of Diet and
   Microsomal Enzyme Induction  on the Toxicity of Dimethyl Nitrosamine.
   Br. J.  Exp.  Pathol. 50:22-25.

Meister, G., H. P.  Hobik,  and  K.-G.  Metzger.  1967.  Comparative Bac-
   teriological and  Pathological Investigations on Commercially Available
   SPF-Animals.   In:  Husbandry of  Laboratory Animals, Proceedings of the
   Third  International Committee on Laboratory Animals, M. L. Conalty,  ed.
   Academic Press, New York.  pp. 387-391.

Ministry of Health  and Welfare Canada.  1975.  The Testing of Chemicals
   for Carcinogenicity, Mutagenicity, and Teratogenicity.  Ministry of
   of Health and Welfare,  Ottawa.   183 pp.

Moffitt, A. E., and S. D.  Murphy.   1974.  Effect of Excess and Deficient
  Copper Intake on Hepatic Microsomal Metabolism and Toxicity of Foreign
  Chemicals.   In:  Trace  Substances  in Environmental Health, Vol. VII,
  D. D.  Hemphill, ed.  University of Missouri Press, Columbia,  pp. 205-210.

-------
                                  55

Mori, K.  1965.  Induction of Pulmonary and Uterine Cancers and Leukemia
  in Mice by Injection of 4-Nitroquinoline-l-oxide.  Gann 56:513.

National Academy of Sciences.  1969.   Rodents.   Standards and Guidelines
  for the Breeding, Care, and Management of Laboratory Animals.  A Report
  of the Subcommittee on Rodent Standards,  Institute of Laboratory Animal
  Resources.  National Academy of Sciences, Washington, D.C.  52 pp.

National Academy of Sciences.  1975.   Chemical Carcinogenesis.  A Report
  of the Committee for the Working Conference on Principles for Evaluat-
  ing Chemicals in the Environment.   National Academy of Sciences,
  Washington, D.C.  pp. 134-155.

National Academy of Sciences — National Research Council.  1972.  Commit-
  tee on Animal Nutrition, Subcommittee on Laboratory Animal Nutrition.
  Nutrient Requirements of Laboratory Animals, 2nd rev. ed.  Nutrient
  Requirements of Domestic Animals,  No. 10.  Washington, D.C.

National Institute for Occupational Safety and Health.  1977.  2,4,5-
  Trichlorophenoxyacetic Acid.  In:   Registry of Toxic Effects of
  Chemical Substances, Vol. II, E. J. Fairchild, R. J. Lewis, Sr., and
  R. L. Tatken, eds.  Government Printing Office, Washington, D.C.
  pp. 33-34.

National Research Council.  1977.  Chronic Toxicity/Carcinogenicity Tests.
  In:  Principles and Procedures for Evaluation of Toxicity and House-
  hold Substances.  National Research Council, Washington, D.C.   pp. 74-85.

Nelson, R. L.  1976.  The Utility of Pharmacokinetics  fo the  Pharmaceutical
  Industry.  J. Clin. Pharmacol. 16:565-569.

Nevins, R. G.  1971.  Design Criteria for Ventilation  Systems.  In:
  Environmental Requirements for Laboratory Animals.   Kansas  State
  University, Manhattan,  pp. 28-43.

Newberne, P. M.   1975.   Influence on Pharmacological Experiments  of
  Chemicals  and Other Factors in Diets  of Laboratory Animals.  Fed.
  Proc. 34(2):209-218.

Newberne, P. M.,  and A.  E. Rogers.  1976.  Nutritional Modulation of
  Carcinogenesis.   In:   Fundamentals in Cancer  Prevention, P. N.  Magee
  et al.t eds.  University of Tokyo Press, Tokyo/University Park  Press,
  Baltimore,   pp.  15-40.

Newberne, P. M.,  A. E. Rogers,  and G. N. Wogan.   1968.  Hepatorenal
  Lesions in Rats Fed  a  Low  Lipotrope Diet and  Exposed to Aflatoxin.
  J. Nutr.  94:331-343.

Page, N. D.  1977.  Concepts of a Bioassay Program in Environmental
  Carcinogenesis.   In:   Advances  in Modern Toxicology, Vol. 3:  Envi-
  ronmental  Cancer, H. F. Kraybill and M.  A.  Mehlman,  eds.  John Wiley
  and Sons,  New York.  pp. 87-171.

-------
                                   56

 Piper,  W.  N.,  J.  Q.  Rose,  M.  L.  Leng,  and P.  J.  Gehring.   1973.  The
   Fate  of  2,4,5-Trichlorophenoxyacetic Acid (2.4.5-T)  Following Oral
   Administration  to  Rats and  Dogs.   Toxicol.  Appl.  Pharmacol.  26:339-351.

 Porter, G.  1967. Assessment of Environmental Influence  on  the Biolog-
   ical  Responses  of  the Animal.   In:  Husbandry of  Laboratory  Animals,
   Proceedings  of  the Third International Committee  on  Laboratory Animals,
   M.  L. Conalty,  ed.  Academic Press,  New York.   pp. 29-42.

 Porter, G. ,  W. Lane-Petter, and N. Home.  1963. Assessment of Diets
   for Mice.   Z. Versuchstierkd.   2:75-91.

 Pott, F.,  A.  Brockhaus, and F. Huth. 1973.  Tests on the  Production of
   Tumors in  Animal Experiments with  Polycyclic Aromatic Hydrocarbons.
   Zbl.  Bakt. Hyg., I.  Abt. Orig. B 157:34-43.

 Prieur, D. J., D. M. Young, R. D. Davis, D. A. Cooney,  E.  R. Homan,
   R.  L. Dixon, and A.  M. Guarino.  1973.  Procedures for  Preclinical
   Toxicologic  Evaluation of Cancer Chemotherapeutic Agents:  Protocols
   of  the Laboratory  of Toxicology.   Cancer Chemother.  Rep. Part 3
   4(1):1-30.

 Roe,  F.J.C.  1965.  Spontaneous Tumors in Rats and  Mice.   Food Cosmet.
   Toxicol. 3:717-720.

 Rogers, A. E.   1975.  Variable Effects of a Lipotrope-Deficient High-
   Fat Diet on  Chemical Carcinogenesis  in Rats.  Cancer Res.  35:2469-2474.

 Rogers, A. E., 0. Sanchez, F.  M. Feinsod, and P. M. Newberne.  1974.
   Dietary  Enhancement  of Nitrosamine Carcinogenesis.   Cancer Res.
   34:96-99.

 Saffiotti, U., R. Montesano,  A.  R. Sellakumar, and  S.  A.  Borg.  1967.
   Experimental Cancer  of the  Lung:   Inhibition by Vitamin A  of the
   Induction of Tracheobronchial  Squamous Metaplasia and Squamous Cell
   Tumors.  Cancer 20:857-864.

 Schardein, J.  L.   1976.  Drugs as Teratogens.   The  Chemical  Rubber
   Company, Cleveland,  Ohio.   291 pp.

 Shaw, B. H.  1976.   Air Movements Within Animal Houses.   In:   Laboratory
   Animal Handbooks 7:   Control of the  Animal  House  Environment, T. McSheehy,
   ed.   Laboratory Animals,  Ltd., Huntingdon,  United Kingdom,   pp. 185-208.

Short,  D.  J.    1967.  Handling,  Sexing  and Palpating Laboratory Animals.
   In:   Husbandry  of  Laboratory Animals,  Proceedings of  the Third Inter-
  national Symposium of  the International Committee on  Laboratory Animals,
  M. L.  Conalty,  ed.   Academic Press,  New York.   pp. 3-15.

Silverstone,  H.,  and A.  Tannenbaum.  1951.  Proportion  of  Dietary Protein
  and the Formation of Spontaneous Hepatomas  in  the Mouse.   Cancer Res.
  11:442-446.

-------
                                  57

Smith, D.  M.,  A.  E.  Rogers,  B.  J.  Herndon,  and  P. M. Newberne.   1975.
  Vitamin A (Retinyl Acetate)  and  Benzo(a)pyrene-Induced  Respiratory
  Tract Carcinogenesis in Hamsters Fed  a  Commercial Diet.   Cancer  Res.
  35:11-16.

Smith, D.  M.,  A.  E.  Rogers,  and P. M. Newberne.  1975.  Vitamin A  and
  Benzo(a)pyrene Carcinogenesis in the  Respiratory Tract  of Hamsters Fed
  a Semisynthetic Diet.  Cancer Res.  35:1485-1488.

Sontag, J. M., N. P. Page, and U.  Saffiotti.   1976.  Guidelines for
  Carcinogen Bioassay in Small Rodents.   U.S.  Department  of Health,
  Education, and Welfare Publ. No. (NIH)76-801.  65 pp.

Surthin, A., and K.  Yagi.  1958.  Distribution in Renal Cell Fractions of
  Sulfhydryl Groups in Rats on Normal and Sucrose Diets and Its Relation
  to Renal Mercury Distribution After Mercuric Chloride Injection.  Am.
  J. Physiol.  192:405-409.

Swann, P. F.,  and A.E.M. McLean.  1968.   The Effect  of Diet on the Toxic
  and Carcinogenic Action of Dimethylnitrosamine.   Biochem. J. 107:14-15.

U.S. Department of Health, Education, and Welfare.   1974.  Guide for the
  Care and Use of Laboratory Animals, 4th ed.   National  Institutes of
  Health.  DHEW Publ. No. (NIH)74-23, Washington,  D.C.  56 pp.

U.S. Food and Drug Administration.  1971.  Panel on Carcinogenesis Report
  on Cancer Testing in the Safety Evaluation of Food Additives and
  Pesticides.   Toxicol. Appl. Pharmacol. 20:419-438.

van der Waaij, D., and D. W. van  Bekkum.  1967.  Resident  Infection in
  Laboratory Animal Colonies and  Their Influence on Experiments.   In:
  Husbandry of Laboratory Animals, Proceedings of the Third International
  Committee on Laboratory Animals, M. L. Conalty, ed.  Academic Press,
  New York.  pp.  373-386.

Walker, E. A., M. Castegnaro, and L. Griciute.  1979.  tf-nitrosamines
  in the Diet of  Experimental Animals.  Cancer Lett. 6(3):175-178.

Weihe, W. H.  1976a.   The Effects on Animals of Changes  in Ambient Tem-
  perature  and Humidity.  In:   Laboratory Animal Handbooks 7:   Control
  of the Animal House  Environment, T. McSheehy, ed.  Laboratory Animals,
  Ltd., Huntingdon, United Kingdom,  pp. 41-50.

Weihe, W. H.  1976fc.   Influence of Light on Animals.  In:   Laboratory
  Animal Handbook 7:   Control of  the Animal House Environment, T. McSheehy,
  ed.  Laboratory Animals, Ltd.,  Huntingdon, United Kingdom,   pp. 63-76.

Weisburger, J. H.   1976.  Bioassays  and  Tests  for Chemical Carcinogens.
  In:  Chemical  Carcinogens,  C. E.  Searle,  ed.  American Chemical Society
  Monograph 173.  pp.  1-23.

-------
                                  58

Winter, C. A., and L. Flataker.  1962.  Cage Design as a Factor Influencing
  Acute Toxicity of Respiratory Depressant Drugs in Rats.  Toxicol.  Appl.
  Pharmacol. 4:650-655.

Withey, J. R.  1978.  Pharmacokinetic Principles.  In:  Proceedings  of
  the First International Congress on Toxicology, G. L. Plaa and
  W.A.M. Duncan, eds.  Academic Press, New York.  pp. 97-118.

World Health Organization.  1978.  Principles and Methods for Evaluating
  the Toxicity of Chemicals, Part I:  Environmental Health Criteria  6.
  Geneva.  273 pp.

Young, J. F., and J. F. Holson.  1978.  Utility of Pharmacokinetics  in
  Designing Toxicological Protocols and Improving Interspecies Extrapola-
  tion.  J. Environ. Pathol. Toxicol. 2:169-186.

Zbinden, G.  1963.  Experimental and Clinical Aspects of Drug Toxicity.
  Adv. Pharmacol. 2:1-112.

Zbinden, G.  1976.  The Role of Pathology in Toxicity Testing.  In:
  Progress in Toxicology, Special Topics, Vol. 2.  Springer-Verlag,  New
  York.  pp. 8-18.

-------
                           3.   ACUTE TOXIC ITY

3.1  INTRODUCTION

     In toxicology the term acute is defined as a single dose or exposure,
or fractions of a dose, administered over a short period (National
Academy of Sciences, 1977).  The toxic effects of such a dosing regime
may be observed within a few minutes or up to several days after dosing.
The effects may vary greatly with sex and age of the animal as well as
within and between different strains and species.
     In acute oral LDso studies the test substance is commonly given by
gavage in a single dose or divided doses within 1 day, and the animals
are observed for 2 weeks; however, when possible, the route of exposure
should be that which is likely to be encountered in humans.  The number
of deaths, time of death, clinical signs, and necropsy are parameters
used in assessing the acute toxicity of the chemical.
     The Food Safety Council (1978) lists the following three purposes
for conducting acute toxicity tests of chemicals:
 1.  to give a quantitative measure of acute toxicity (LDso) for
     comparison with other substances,
 2.  to identify the clinical manifestations of acute toxicity, and
 3.  to give dose-ranging guidance for other tests.
     This section will discuss aspects of acute toxicity such as the
LDso, necropsy as a part of the protocol of an acute toxicity test, and
the length of the observation period following acute administration of
the test chemical.

3.2  LD50 DETERMINATIONS

3.2.1  Introduction
     The most frequently determined index of toxicity is  the LD50,  the
dose that is lethal to one-half of a group of treated animals  (National
Academy of Sciences, 1977).  In addition, the LDso can  be used  as  a
basis for determining doses to be used  in subsequent  subchronic and
                                    59

-------
                                   60
 chronic  toxicity  studies.   It  can also aid the researcher in determining
 which  species  and route of  administration would be the most suitable for
 his  or her  experiments.  The following sections will show how the choice
 of species,  route of  administration, and sex of the animal may affect
 the  LDso and acute toxicity response.
     The information  presented on acute LDs0s is available in the National
 Institute for  Occupational  Safety and Health (NIOSH) Registry of Toxic
 Substances  (1977),  which has entries in the following format:
 BR09500  Ammonium, (2-hydroxyethyl)diisopropylmethyl-, bromide, xanthene-
  9-carboxylate
 TXDS:  Oral-rat LD50:  370  mg/kg
       Intraperitoneal-rat  LDso:  25 mg/kg
       Subcutaneous-rat LD5Q:   298 mg/kg
       Intraduodenal-rat LDso:  125 mg/kg
       Oral-mouse LDso:  445 mg/kg
       Intraperitoneal-mouse LDso:  78 mg/kg
       Oral-rabbit  LDso:  750  mg/kg
       Intravenous-guinea pig  LDT :  51 mg/kg
                                LjO
     This format  provides information on the route of administration,
 the  species, and  the  toxic  LDso dose or LDT  dose  (lowest published
                                          LiO
 lethal dose  reported).  Also included in the NIOSH document are synonyms,
 Chemical Abstracts  Service  registry numbers, and the reference for each
 item of  toxicity  data.  Table  3.1 is a list of 96 compounds, arbitrarily
 chosen,  for which the LDso  values were examined and compared for species
 differences and for differences between routes of administration.
Although not discussed, it  is  recognized that the differences in reported
LDso values may have also been influenced by the age and strain of the
 test animals, duration of the  starvation period prior to dosing, ambient
temperature, concentration  of  the test substance, speed of injection,
and  type  of solvent or suspension medium (Zbinden, 1973), as well as by
interlaboratory variations  in  experimental procedures.

-------
                                              61
                  Table 3.1.   Compounds examined  for  LDso  differences  between
                          species and between routes  of  administration
Ammonium (2-hydroxyethyl)dlisopropylmethyl-,  bromide,  xanthene-9-carboxylate
Ammonium [(p-hydroxy-2-isobutyl)phenyl]trlmethyl-,  iodide,  methylcarbamate
Arsine, dichloro(2-chlorovinyl)-
Benzaldehyde, p-isopropyl-
Benzamide, 4-amino-ff-cyclopropyl-3,5-dichloro-
Benzimidazole, 2-(4-thiazolyl)-, hydrochloride
2-Benzimidazolinone, 5-chloro-l-[3-(diraethylamino)propyl]-3-phenyl-
Benzoic acid, benzyl ester
Benzole acid, 2,3,6-trlchloro-
2-H-Benzo(a)quinolizin-2-one, 1,3,4,6,7,llb-hexahydro-3-isobuty1-9,10-dimethoxy-
Benzoxazole, 2-amino-5-chloro-
Benzoxazoline, 3-methyl-2-(methylamino)-
2-Benzoxazollnone, 5-chloro-
Benzyl alcohol, alpha-(l-aminoethyl)-, hydrochloride,  (+ -)-
Carbamic acid, methyl-, m-tolyl ester
Carbamic acid, methyl-, 3,4,5-trimethylphenyl ester
Carbanilic acid, m-chloro-, 4-chloro-2-butynyl ester
Carbon tetrachlorlde
2-Carboxymethylmercaptobenzenestrlbonic acid
Cerium chloride
Ethylamine, W-methy1-2-[(o-methyl-alpha-phenyIbenzyl)oxy]-,hydrochloride
Ethylene glycol
Guanidine, l-cyano-3-tert-pentyl-
3-Heptanone, 6-(dimethylamino)-4,4-diphenyl-, hydrochloride
1,6-Hexanediamine, N,N,Nl,N^-tetramethyl-, polymer with 1,3-dibromopropane
Hydrazine, methyl-, hydrochloride
Hydroquinone
Imidazole
3-3-Imidazoline,  4-amino-2,2,5,5-tetrakis(trifluoromethy1)-
1,3-Indandione, 2-[(p-chlorophenyl)phenylacetyl]-
IH-Indazole, l-benzyl-3-[3-(dimethylamino)propoxy]-,  monohydrochloride
Indene-1-ethylamine, Af^ff-dimethyl-l-phenyl-, hydrochloride
Indole-3-acetic acid,  l-(p-chlorobenzoyl)-5-methoxy-2-methyl-
Indole, 3-(2-aminoethyl)-5-methoxy-,  hydrochloride
Isonicotinic acid,  2-[2-(benzylcarbamoyl)ethyl]hydrazide
Isonicotinic acid,  2-isopropylhydrazide
Isonipecotamide,  4-(p-chloropheny!)-!-[3-(p-fluorobenzoyl)propyl]-W,W-dimethyl-
Isonipecotic acid,  l-(p-aminophenethyl)-4-phenyl-,  ethyl  ester
Meglumine hydroxamate
Mercury,  [3-(alpha-carboxy-o-anisamide)-2-(2-hydroxyethoxy)propyl]hydroxy-,  monosodium salt
Mercury,  (3-cyanoguanidino)methyl-
Mercury  (11)  iodide
4-Metathiazanone,  2-(3,4-dichlorophenyl)-3-methyl-, 1,  1-dioxide
Methanesulfoanilide, 4'-[l-hydroxy-2-(methylamino)propyl]-, hydrochloride
Methanesulfonic acid,  iodo-,  sodium salt
2,6-Methano-3-benzazocln-8-ol,  1,2,3,4,5,6-hexahydro-3-ally1-6-ethyl-ll-methyl-
4,7-Methanoindene,  l,4,5,6,7,8,8-heptachloro-3a,4,7,7a-tetrahydro-

-------
                                               62
                                       Table 3.1 (continued)
 Monomethylhydrazine nitrate
 Morphinan-3, 14-diol, 17-(cyclopropylmethyl)-
 Morphinan-3,6-alpha-diol, 7,8-didehydro-4,5-alpha-epoxy-17-methy1-
 Morphinan, 17-(p-nitrophenethyl)-, hydrochloride (+ -)-
 Morphinan-6-alpha-ol, 7,8-didehydro-4,5-alpha-epoxy-3-methoxy-17-methyl-
 2-NaphChalenemethanol, alpha-[(isopropylamino)methyl]-
 Naphthalimide, ff-hydroxy-, diethyl phosphate
 2-Naphthol, 1,2,3,4-tetrahydro
 Neomycin
 Nickel (11) acetate(l:2)
 Nicotinamide, tf,tf-diethyl-
 2-Norbornanamine, ff-ethyl-3-phenyl-, hydrochloride
 2-Norbornanamine, tf-3,3-trimethyl-
 2-Norbornanamine, /Y,2,3,3-tetramethyl-, hydrochloride
 l-Oxa-3,9-diazaspiro(5.5)undecan-2-one, 5-ethyl-9-[3-(p-fluorobenzoyl)propyl]-
 1.2,4-Oxadiazole, 5-[2-(diethylamino)ethyl]-3-phenyl-
 l,2,4-Oxadiaxolidine-3,5-dione,  2-(3,4-dichlorophenyl)-4-methyl-
 2H-l,3-Oxazine-2,4(3H)-dione, 5,5-diethyldihydro-
 Pactomycin
 Penlcillanic acid, 6-phenoxyacetamido-
 2,4-Pentanediol, 2-(p-chlorophenyl)-4-methyl-
 4-Pentenoic acid, 2-[(2-diethylamino)ethyl]-2-phenyl-, ethyl ester
 l-Penten-4-yn-3-ol,  l-chloro-3-ethyl
 Phenazine, 3-amino-7-(dimethylamino)-2-methyl-, hydrochloride
 Phenethyl alcohol
 Phenol, 2,4-dinitro-
 Phenol, 2,4,5-trichloro-
 Phenothiazine, 2-chloro-10-[3-(A'-cyclopentyl-W-methyl)amlnopropyl]-
 Phenothiazine, 10-[2-(diethylamino)propyl]-
 Phosphonothioic acid,  phenyl-,  0-ethyl 0-(p-nitrophenyl) ester
 Phosphoramidothioic  acid,  0,5-dimethyl ester
 Phosphoric acid, 2-chloro-l-(2,4-dichlorophenyl)vinyl diethyl ester
 1,3-Propanediol, 2-(hydroxymethyl)-2-nitro-
 12,-Propanediol, 3-(0-methoxyphenoxy)-, 1-carbaoate
 Pyrrolidinium, 3-hydroxy-l,-dimethyl-,  bromide,alpha-cyclopentylamandelate
 3-Pyrrolidinol,  l,2-dimethyl-3-phenyl-, propionate
 Quinine,  hexylbromide
 Sodium  pentafluorostannite
 Streptomycin
 Succlnic  acid,  cadmium salt  (1:1)
 Sulfanilamde
 Sulfanilamide, JV1-2-thiazolyl-
 5H-Tetrazoloazepine, 6,7,8,9-tetrahydro-
 Theophy11ine,  8-benzy1-7-[2-(echy1(2-hydroxyethyl)amino)ethyl]-,hydrochloride
 2H-l,3,5-Thiadiazine-2-thione, tetrahydro-3,5-dimethyl-
 a-Triazine, 2-azido-4-(isopropylamino-6-(methylthio)-
 8-Triazine,2,4-dichloro-6-(o-chloroanilino)-
Urea, 3-(hexahydro-4,7-methanoindan-5-yl)-l,l-dimethyl-
Urea, 1,1,3,3-tetramethyl-

-------
                                  63
3.2.2  Difference Between Species
     Table 3.2 uses the LD50 data for each compound  listed in Table 3.1
to show the LD50 differences (expressed as percentage)  between different
species categorized by route of administration.   For example, the first
entry in Table 3.2, cat, is compared with the pigeon and rat for the
oral route of administration.  The comparison of the cat with the rat
indicates that for one compound the cat had an LDso  greater than the rat
LD50 by 1% to 25%, for three compounds the rat LD50  was greater than
the cat LD50 by 51% to 75%, and for one other compound the rat LD50
was greater by 76% to 100%.  Therefore, for four of  the five chemicals
compared, the rat showed a higher tolerance to the acute dose.  Collec-
tive examination of data in Table 3.2 reveals that 5.8% of the species
compared showed no difference in LDso amounts, 23.3% of the species had
LD50 values differing by 1% to 25%, 29-4% had differing LD50 values of
26% to 50%, 27.7% of the species had LD50 values that differed by 51% to
75%, and 13.9% of the species had LD50 differences between 76% and  100%.
Therefore, less than one-half of the 374 species comparisons  (41.7%)
showed LDso values that differed by more than 50%.  A more detailed
examination of the data in Table 3.2 reveals the following:
 1.  The rat and mouse appear to be  less sensitive  indicators of  toxicity
     than the dog.
 2.  The rabbit is a less  sensitive  toxicity indicator  than  the  rat but
     slightly more sensitive than  the  mouse.
 3.  The mouse is  a less  sensitive toxicity  indicator  than the  hamster.
 4.  The differences between LDso  values  for the mouse  and rat  were less
     than 50% for  56% of  the comparisons.
 5.  The mouse has a larger LD50  (less sensitivity) than the rat in the
     majority of  comparisons (60%).
     The principal observations  from these data are that the mouse and
the  rat represent  the most commonly used test  species (84.3% of all
comparisons involved one  or both)  and that for slightly more than qne-
half  (58%) of all  the  species  comparisons the LDso  values differed by
less than 50%.

-------
Table 3.2.  Percent
differences between species
Species
Cat
Pigeon
Rat
Chicken
Guinea pig
Hamster
Mouse
Pigeon

Quail
Rabbit
Dog
Cat
Guinea pig

Hamster
Monkey

Pigeon
Rabbit



Rat



Duck
Chicken
Dog
Mouse
Pigeon

Quail
Route of
administration n . „,
0 1 to 25

Oral
Oral Cat

Oral Guinea pig
Oral
Subcutaneous Mouse
Oral Mouse (2)b
Oral
Intravenous
Oral
Oral

Oral Cat
Subcutaneous
Oral
Oral
Subcutaneous
Oral
Oral
Subcutaneous 1° Dog
Skin
Intravenous
Oral 1°
Intraperitoneal
Oral
Subcutaneous Rat
Intravenous Rat
Intraperitoneal 1°
Skin

Intravenous Chicken
Oral
Intravenous
Oral Pigeon
Intravenous
Oral
Percent
26 to 50



Guinea pig
Hamster



Quail



Dog(2)b

Dog
Monkey

Pigeon


Dog
Rabbit
Rat, dog(2)b
Dog
Rat
Rat(2)6
Rat






Quail
differences
51 to 75

Rat(3)b



Mouse
Mouse
Pigeon
Pigeon



Dog

Guinea pig





Dog
Dog

Rabbit ,
Rat, dog(2)C

Rat, dog


Chicken




Quail

76 to 100

Pigeon0
Rat







Rabbit



Dog


Monkey





Rat, dog

Dog



Dog
Duck
Pigeon
Pigeon


-------
Table 3.2 (continued)
Species
Guinea pig
Cat
Hamster

Monkey
Mouse


Pigeon
Rabbit



Rat




Mouse
Cat
Dog



Hamster

Monkey

Pigeon
Rabbit

Quail
Dog
Pigeon
Rat
Route of
administration

Oral
Intraperitoneal
Oral
Subcutaneous
Oral
Intraperitoneal
Subcutaneous
Intravenous
Oral
Subcutaneous
Skin
Oral

Oral
Subcutaneous
Skin
Intraperitoneal
Intravenous

Oral
Intraperitoneal
Oral
Subcutaneous
Intraperitoneal
Intravenous
Oral
Intraperitoneal
Subcutaneous
Oral
Intravenous
Oral
Subcutaneous
Intravenous

Oral
Oral
Oral
Percent differences
0 1 to 25


1°
Guinea pig
lc Mouse (2)*
Guinea pig
1°



Guinea pig

1° Guinea pig(2),fc
rat
1° Rat

Guinea pig



Dog, mouse


Mouse
1° Mouse(2)ft
Mouse

Mouse
Mouse ,
Rabbit (2)D



Pigeon

26 to 50




Mouse (A), guinea
Mouse, guinea pig
Guinea pig(2)"

Guinea pig
Rabbit
Guinea pig
Rabbit, guinea
Pig .
Guinea pig(2),fi
rat(2)Z)

Guinea pig



Cat
Mouse
Mouse(3)fc

Mouse
Mouse

Mouse


Rabbit (2),^
mouse ( 3) °
Mouse, rabbit (2)h



Rat
51 to 75

Cat, guinea pig


Monkey
Guinea pig
Guinea pig



t
Rabbit (2), fi
guinea pig
Guinea pig (4),
rat (3)"
Guinea pig, rat


Rat
1.
Mouse (3)°
Mouse
Dog
Mouse
Mouse

Hamster
Monkey

Mouse(2),b
rabbit
Mouse (2)°
Mouse (2)fc

Dog
Pigeon
Rat
76 to 100

Guinea pig


Mouse (2)fc




Guinea pig


Rat
Guinea pig




Mouse
Mouse (2)fc


Mouse




Rabbit (2)fc
Mouse(2)fc




                                                                                    Ui

-------
                                            Table  3.2  (continued)
Species
Rabbit
Cat
Duck
Monkey

Rat
Duck

Hamster
Rat

Monkey

Mouse







Pigeon

Rabbit




Route of
administration _

Oral
Oral
Subcutaneous
Oral

Oral ; .•;•>'>
Intravenous
Oral
Intraperitoneal
Skin
Subcutaneous
Oral
Oral t*°
Intraperitoneal 2°

Intravenous 2°

Subcutaneous

Intramuscular
Oral
Intravenous
Oral 2°
Subcutaneous
Intraperitoneal 2°
Intravenous 2°
Skin
Intramuscular
Percent differences
1 to 25

Cat





Duck
Rat, hamster
Hanmter(3)k
Hamster

i
Mouse (9).°
rat(8)\
Mouse (5),
rat(4)6
Mouse (5), °
rat(2)£
Mouse(5)D


Rat(3)fc

Rabbit (2)


Rat, rabbit
Rabbit

26 to SO


Duck
Monkey




Hamster, rat


Monkey
Monkey
Mouse(7).fc
rat (6)\
Mouse (4), b
rat (3)*
Mouse(3),6
rat(4)6
Mouse(2),fe
rat(2)£



Rabbit
Rat, rabbit
Rat, rabbit
Rat(2),b rabbit
Rat
51 to 75 76 to 100




Monkey

Rat Rat, duck


Hamster (2)b


L t
Mouse (7), b Mouse (5), b
rat (10)* rat(3)6
Mouse (7),* Mouse
rat(2)6
Mouse(3),0 rat Mouse

Mouse (2). * Mouse
rat(2)0
Mouse

Pigeon
Rabbit (3),° rat Rabbit (3),° rat


Rat, rabbit Rat, rabbit (2)b

     "indicates which animal of the comparison had the greatest LD50.  Position of species indicates the per-
cent differences in the LD50s of the two species being compared.
      Represents number of chemicals compared where LDso of species indicated was greater than species being
compared.  If there is no number then only one chemical was compared.
°The number indicates how many times the
                                                   values were identical for the species compared.

-------
                                  67
3.2.3  Difference Between Administration Routes
     Table 3.3 shows the percentage  difference between LDso  values  for
various pairs of routes of administration categorized by species.
Abbreviations in the appropriate columns indicate which route of the two
being compared is the greater.   For  example,  the "inp" (intraperitoneal)
notation in the first entry for the  mouse at  76% to 100% indicates  that
the LDso for the intraperitoneal route of administration was 76% to 100%
greater than the LDso for the intracerebral route for the same compound.
Column by column examination of the  data in Table 3.3 reveals that  2.4%
of the entries fall in the 0% (no difference  in  LD50) column, 20.4% in
the 1% to 25% column, 20.4% in the 26% to 50% column, 26.2% in the  51%
to 75% column, and 29.8% in the 76%  to 100% column.  These data indicate
that the LDso can vary significantly depending on the administration
route.
     A more detailed examination of  the data in Table 3.3 shows the
following:
 1.  For all compounds, the subcutaneous route of administration was
     always associated with a higher LDso than the intravenous route.
 2.  Of the 328 entries, 116 (35%) compared the subcutaneous route with
     other routes, 140  (42.6%) compared intravenous administration with
     other routes, and  199  (60.6%) compared the oral route of administra-
     tion with other routes.  Thus the predominant use of the oral
     administration route is shown.
 3.  When the intravenous route was compared  to the  oral and the intra-
     peritoneal routes, respectively, the LDso values were  greater  for
     the oral and  intraperitoneal routes for  most  of the compounds.   In
     fact, in only four of  56 comparisons  (two being equal)  was  the
     intravenous LDso  greater than the  oral LDso  [those  four compounds
     were methylhydrazine hydrochloride, 2-isopropylhydrazide isonico-
     tinic acid, pactamycin, and  2-chloro-l-(2,4-dichlorophenyl)vinyl
     phosphoric  acid diethyl ester],  and in only five of 35 comparisons
     was the  intravenous LDso greater than the  intraperitoneal route
      [those  four compounds  (two  comparisons  involved the same compound)
     were methylhydrazine hydrochloride, 4-amino-2,2,5,5-tetrakis

-------
Table 3.3.  Percent LD5o differences between routes of administration
Routes of
administration
Intracerebral and
intraperitoneal
Intracerebral and
Intravenous
Intracerebral and
subcutaneous
Intraperitoneal and
intramuscular

Intraperitoneal and
skin


Intraperitoneal and
subcutaneous
Intravenous and
intramuscular


Intravenous and
intraperitoneal
Intravenous and oral



Intravenous and skin

Intravenous and
subcutaneous


Species 	
0

Mouse

Mouse

Mouse 	 -

Mouse
Rat

Rat
Hamster
Guinea pig
Rat 1°
Mouse

Rabbit
Rat
Mouse

Rat
Mouse
Rat
Mouse 1°
Dog
Guinea pig
Rabbit 1°
Rabbit
Rat

Rabbit
Mouse
Rat
Guinea pig
Percent differences
1 to 25







Inm, Inpa





Sub (5),* Inp(3)*


Inm
Inv, Inm

Inv , Inp
Inv(3),& Inp (3)*
Inv
0(4),* Inv






Sub (2)?
Sub (2)*

26 to 50








Inm




Inp, Sub
Sub (7)*




*
Inp (4)*
Inp (7)*
06 ft
0(2)*
0





Sub (6)*
Sub

51 to 75










Skin


Sub (3)*
Sub (3)*





Inp (4)*
Inp (8),° Inv
0(7),* Inv(2)*






Sub
Sub (8)°
Sub (3)

76 to 100

Inp

Inv

Sub



•L
Skin (2)*
Skin
Skin
Sub (2)*
Sub

Inm
Inm


Inp (2)
Inp
0(11)*
0(18)*

0

Skin
Skin
•L
Sub (2)?
Sub (7)*
Sub (7)*
Sub
                                                                                                   00

-------
                                                 Table 3.3 (continued)
Routes of
administration
Oral and
intracerebral
Oral and
intramuscular

Oral and
intraperitoneal




Oral and
subcutaneous

Skin and oral




Subcutaneous and
intramuscular

Subcutaneous and
skin




0 1 to 25

Mouse

Rat
Mouse 1°
Rat 2° 0(5)i.fc Inp(2)&
Mouse 0(7)&
Hamster
Cat Inp
Rabbit Inp
Guinea pig 1°

Rat 0(9)\
Mouse 0(6), Sub (2)*>
Guinea pig lc
Rat
Mouse
Rabbit 0
Hamster
Guinea pig

Rabbit lira
Mouse Sub

Rat
Dog
Rabbit
Guinea pig
Percent differences
26 to 50 51 to 75



Inm
0
0(6) ,b Inp 0(10)fc
0(7), b Inp 0(11), b Inp(2)6
0 0(3)fc



t
0(3), £ Sub Sub .
0(7),° Sub 0(4),° Sub(3)°
i.
Skin 0, Skin(3)D

Skin




Sub

Skin

Skin


76 to 100

0

0

0(12),b Inp
0(9)&





0
t,
Skin(5)fi
0

Skin
Skin




Skin
Skin

Skin
     alnm — intramuscular; Inp — intraperitoneal; Inv — intravenous; 0 — oral; Sub — subcutaneous.
      Represents number of comparisons when route indicated was greater than the route being compared.  If no
number is present then only one  chemical was compared.
The number indicates how many times the
                                                   values were identical in a comparison.

-------
                                   70

      (trifluoromethyl)-3-imidazoline,  2-isopropylhydrazide isonicotinic
      acid,  and N1-2-thiazolylsulfanilamide].
  4.   The oral LD50  was greater than the subcutaneous LDso ^or 82.5% of
      the comparisons;  however, the difference was  generally  less than 50%.
  5.   A comparison between the oral LDso an<*  intraperitoneal  LDso showed
      that for 89% of the entries the oral route had a  greater LDso than
      the intraperitoneal route [those compounds for which the intraperi-
      toneal route was  greater for at least one  species were  2,3,6-tri-
      chlorobenzoic  acid, alpha-(1-aminoethyl)benzyl alcohol  hydrochloride,
      l-cyano-3-tert-pentylguanidine, l-(p-chlorobenzoyl)-5-methoxy-2-
      methylindole-3-acetic acid, pactamycin, tetrahydro-3,5-dimethy1-
      2H-l,3,5-thiadiazine-2-thione, 2,4-dichloro-6-(0-chloroanilino)-s-
      triazine, 4-amino-#-cyclopropyl-3,5-dichlorobenzamide,  and
      2-(3,4-dichlorophenyl)-3-methyl-4-metathiazanone-l,l-dioxide].
      The principal  conclusion from these data is that  the choice of
 route of administration is an important part of the experimental design
 because it  can result  in significant differences in toxicity response.
 This  emphasizes the importance of selecting  the route  of exposure  that
 will  be the likely  exposure route in man when conducting safety assess-
 ments of chemicals.

 3.2.4  Sex  Differences in the Laboratory Rat
      Male and female rats show differing susceptibilities to the acute
 doses of some chemicals.   Hayes (1975)  states that the rat,  which  is
 used  more than any  other species for studies in toxicology,  apparently
 shows more  variation between the sexes  in its response to chemicals than
 does  any other species.   One example is provided by the research of Kast
 et al.  (1975i)  with  the  amino-halogen-substituted  benzylamine, fominoben-
 HC1.   The oral LDso  f°r  males was 6450  mg/kg and for females, 4500 rag/kg.
 Other  examples of sex  differences in LDso  response are seen  in the
 research  of Gaines  (1960,  1969).   He reports that, in  acute  oral toxicity
 tests with 98  pesticides,  most compounds were more toxic to  female than
 to male rats.   This  is demonstrated with the organophosphorus pesticides
azodrin  (male LD50,  126 mg/kg and female LD50,  112 mg/kg), Bayer 37289

-------
                                  71

(male LD50, 180 mg/kg and female LD50,  64 mg/kg),  and ethion (male LD50,
245 mg/kg and female LD50, 62 mg/kg).   Hayes (1975)  used the oral toxicity
LD50 data of Gaines (1960, 1969) on the rat for 69 pesticides and calcul-
ated the ratio of female to male oral LD5QS.  His  calculations showed
that the ratio ranged from 0.21 (indicating greater susceptibility of
the female) to 4.62 (indicating greater susceptibility of the male) and
averaged 0.94.  Therefore, these studies indicate  that both males and
females should be used as test animals when the rat is the animal of
choice for the acute toxicity testing of a chemical.

3.2.5  Test-Limiting Criteria
     Some chemicals are relatively innocuous when given as single doses.
If this relative lack of acute toxicity can be determined at the begin-
ning of the acute toxicity test, there is no need for continuing the
acute testing.  The National Academy of Sciences  (1977) states that,
for most purposes, if animals survive single oral doses of 5 or 10 g/kg,
an adequate estimate of hazard is obtained.  The  Interagency Regulatory
Liaison Group's guidelines indicate that no further  acute oral toxicity
testing of a  chemical is  necessary if no mortality  is seen at a dose  of
5 g/kg.  Similarly, the guidelines for the  Organization for  Economic
Cooperation and Development  state  that,  if  5 of 10  animals  survive a
dose of 5  g/kg of a chemical for 14 days, no further acute  toxicity
testing is necessary.   If these  criteria are met  in one sex but  not  the
other acute toxicity  testing must  proceed with the  sex  that  did  not meet
the criteria  for limiting the acute  test.

3.2.6  Conclusions
     The information  in Sect.  3.2.2  and  Sect.  3.2.3 clearly indicates
the importance of  the selection of proper species and route of adminis-
tration.   Specifically,  42%  of all species comparisons (Table 3.2)
indicated  differences in LD50 values of  more than 50%,  and 56% of all
comparisons  involving administration routes (Table 3.3) showed differ-
ences between LD50  values of more  than 50%.  When the laboratory rat is
the test animal in acute toxicity  studies, both male and female animals

-------
                                   72
 should be  used because  of  the variation  in response between the sexes.
 If  a  chemical does not  cause more  than 50% mortality when administered
 in  acute doses of 5  g/kg,  further  acute  toxicity testing is not required.
 For safety assessment of chemicals with  potential human exposure, it is
 desirable  to select  a test animal  that will reflect what is anticipated
 to  occur in man.  In practice, however,  the rat or mouse is usually the
 species of first choice because  they  are easy to work with and relatively
 inexpensive and there is a vast  amount of information available on which
 to  base comparative  assessments.   The difference in LDsg values as a
 result of  using different  exposure routes indicates the necessity of
 selecting  the exposure  route that  will be the likely exposure route in
 man when chemicals are  tested that have  a potential for human exposure.

 3.3  HUMAN VS ANIMAL RESPONSE

 3.3.1 Introduction
      This  section will  compare the responses of animals and humans to
 acute exposures of chemical substances.  For the most part, information
 on  dermal  and ocular toxicity will not be presented, as this will be
 covered in detail in a  companion document.

 3.3.2  Comparison of Lowest Published Lethal Doses (LD  )

      The source of the  information in this section is the NIOSH Registry
of Toxic Substances  (1977)  in which the  LDLo is defined as the lowest
published  lethal dose.  Twenty-seven  compounds  (Table 3.4) were chosen
at random  and the human and animal LD   data compared in Table 3.5.  The
                                      LiO
following  observations are  evident:
 1.   With  the exception of  two compounds, all comparisons involved the
     oral route of administration.
 2.   In only one instance was the  human  and animal LIL  equal, whereas
     8.6% of the comparisons had LD   differences between 1% and 25%,
     17.3%  between 26% and  50%,  15.2% between 51% and 75%, and 60.8%
     between 76% and 100%.

-------
                               73

      Table 3.4.  Compounds selected for human vs animal
                       LD   comparisons
                         .LiO
Oxalic acid                                               0.071
Quinine, monohydrochloride                                0.230
Hydrocyanic acid                                          0.570
Phenol, 2,4-dinitro                                       4.3
Barbituric acid, 5-ethyl-5-hexyl- ,  sodium salt            5
Ouabain                                                   5
Phosphoramidocyanidic acid, dimethyl-, ethyl ester       23
Mercury (11) chloride                                    29
Phenol, pentachloro-                                     29
Strychinine                                              30
Carbon tetrachloride                                     43
Chloral hydrate                                          50
Veriloid                                                 50
Zinc, bis(dimethyldithiocarbamato)-                      50
Quinine                                                  50
Benzamide, p-amino-tf-(2-diethylamino)ethyl)0-            50
Ammonium, [4-(bis(p-(dimethylamino)phenyl)
  methylene)-2,5-cyclohexadien-l-xylidene]
  dimethyl-, chloride                                    50
Acetanilide, 4'-hydroxy                                  50
Methane , iodo-                                           50
2-Naphthol                                               50
Quinoline, 8[ (4-(diethylamino)-l-methylbutyl)
  amino ] -6-methoxy-                                      50
Sodium fluoride                                          75
Benzoic acid                                             500
Ethyl alcohol                                            500
Sulfamic acid                                            500
2-Pentanone, 4-methyl-                                   500
1 , 4-Naphthoquinone , 2-methyl-                            500
      Compounds listed in  order  of decreasing toxicity.

-------
                           Table 3.5.
LD   differences between humans and animals
  Lo
Species
compared
Rat


Rabbit

Pigeon
Guinea pig
Chicken
Cat
Mouse
Dog


Route of
administration
Oral
Skin
Intravenous
Oral
Intravenous
Oral
Oral
Oral
Oral
Oral
Oral
Skin
Intravenous
Percent differences
0 1 to 25 26 to 50 51 to 75 76 to 100
Rata Human, rat (2r
Human
Rat
Rabbit Rabbit(2)Z? Human, rabbit(3)fc Rabbit(5),fc human
Rabbit
Pigeon Pigeon
Guinea pig(2)i> Guinea pig
Chicken
Cat
Mouse
1° Human Dog (7), human (2)
Dog
Dog
     alndicates which animal of the comparison had the greatest LDLo.  Position of species indicates the
percent differences in the LD^o of the human and animal species being compared.
      Represents the number of chemicals compared where the LDLO of the species indicated was greater than
the species being compared.  If there is no number then only one chemical was compared.
     CFor one chemical, the human and the dog had identical LD^O values when administration was oral.

-------
                                  75

 3.  Only 19.5% of the comparisons showed the human LDLQ value to be
     greater than the animal LD   value.
On the basis of these data,  it can be concluded that there is a signifi-
cant difference between the  human and animal acute LD,   response and
that, in the majority of instances,  the human showed less tolerance
(reflected by lower LDT  values) to chemical insult than did the animal
                      LiO
model.
3.3.3  Comparison of Acute Toxicity Response
     The studies of Goyer (1971) and Lock and Ishmael (1978) show the
effect of the acute administration of chemicals on the rat and human
kidney to be similar.  In his review of the effects of lead on the
kidney, Goyer found that morphological and functional reactions of the
kidneys of rat and man to acute lead exposure have comparable features:
(1) the proximal tubular lining cells are affected, (2) intranuclear
inclusion bodies are formed, and (3) there is an associated aminoaciduria.
He also noted that although the impairment of mitochondrial function has
not been demonstrated in man as it has in rats, the impairment may be
inferred from morphological changes in human biopsy material.  Lock and
Ishmael in studying the acute effects of paraquat and diquat  found that
an oral dose of 680 micromoles/kg of either chemical would  result in
marked diuresis, proteinuria, and glucosuria 6 to 24 h after  dosing of
the male rat.  In addition, histopathological kidney examination showed
mild hydropic change in the proximal convoluted tubules,  and  renal
clearance of insulin, p-aminohippuric acid  (PAH), and /l/'-methylnicotamide
(NMN) was found to be markedly  reduced 2 h  after  dosing.   Lock and
Ishmael reported that these changes  indicate that renal  impairment  is
similar in rat and man.
     Hexachlorophene and  ethchlorvynol have also  been shown to produce
similar symptoms in humans  and  animals after acute  exposure.   Martinez,
Boehm, and Hadfield  (1974)  reported  the  accidental  oral  ingestion of
approximately 45 mL of hexachlorophene by  a seven-year-old boy.   Toxic
reactions included nausea,  vomiting,  anorexia,  diarrhea, decrease in
visual acuity, blurred vision,  blindness,  somnolence,  and disorientation,

-------
                                   76

with  respiratory and  cardiac  arrest  occurring  61 h after hospital
admission.   Further examination revealed  the following:   (1) interstitial
myocarditis,  (2) pneumonitis  and acute bronchiolitis,  (3) edematous
brain tissue,  (4)  occasional  neuronal  degeneration, and  (5) myelin
sheath disintegration and other neuronal  changes.  Martinez, Boehm, and
Hadfield noted that these changes have been produced in experimental
animals exposed to several chemicals including hexachlorophene.
      In a case report of an intravenous injection of ethchlorvynol pre-
sented by Payne, Kerr, and Diaconis  (1977), the individual developed
acute, severe  pulmonary edema accompanied by hypoxemia and acidosis.
Similar toxic  effects on the  pulmonary vasculature were observed in rats
by Payne and coworkers after  administering intravenous injections of
80 mg/kg ethchlorvynol, resulting in death from acute  respiratory distress.

3.3.4  Conclusions
      The information  in Sect.  3.3.2  concerning lowest  lethal doses
clearly indicates a difference between humans  and animals with  respect
to the size  of the dose that  will cause death; in most cases a  smaller
dose  was required to  cause death in  humans than in animals.  Because  of
this  difference, an examination of the pathology and functional changes
induced by the acute  exposure of humans and animals to chemicals  (Sect.
3.3.3)  might be expected to show corresponding differences.  However,
the available  literature showed that,  for those chemicals compared, man
and animal acute toxicity was  similar.  This does not  imply that there
are no chemicals for  which the human and  animal response would  be dif-
ferent,  but  rather indicates  the scarcity of information  comparing human
and animal acute toxicity studies.   Two possible reasons  for the apparent
paucity of comparable data are that  (1) many acute toxicity studies
performed on animals  are done  primarily to establish the  LDsg and pro-
vide  reference toxicity dose  information  for subchronic and chronic
testing  and  (2)  most  acute human exposures, with the possible exception
of  dermal testing,  occur only  from accidental  intake or suicides and,
unless the person  is  admitted  to a hospital soon after intake,  much
important data  go  unrecorded.   Although it is  not possible to satisfactorily

-------
                                   77
appraise the comparative susceptibilities  between man and  animal with
only the few chemicals used as examples in Sect.  3.3.3,  the examples  do
support one of the major premises of toxicity research — that effects
observed in animals serve as an indicator  of what a probable response
would be in man.

3.4  PATHOLOGY

3.4.1  Introduction
     Evidence of tissue damage from the acute exposure of a test animal
to a chemical would be useful information  for the hazard evaluation of
most chemicals, especially for those chemicals for which there is known
or potential acute human exposure.  The following sections consider some
aspects of histopathology in acute toxicity testing of chemicals and
offer some concluding remarks concerning the degree of necropsy required
when evaluating the acute toxicity of a chemical.

3.4.2  Histopathology
     Histopathological changes from acute exposures can be induced by a
variety of compounds  (some examples are given in Table 3.6), a  fact not
surprising since the experimental doses given are usually of such mag-
nitude that they kill at least one-half of  the test animals.  The stress
to the animal's system would in many cases  produce lesions of various
organs, if the animal lives long enough for histologic damage to occur.
On the other hand, there are compounds that, when administered  at doses
high enough to  cause death, do not  show any histopathology related to
the chemical.  An  example of this  type of action was  described  by Robens
(1979) where several  strains of  rats were given  high  doses (2000 mg/kg)
of a short-chain chlorinated hydrocarbon.   Several  animals died as a
result of the exposure, but in no  animals,  living or dead, was  there any
detectable histopathology related  to the  test  chemical.   Another example
is shown by the action of tf-2-fluorenylacetamide in mice  where  LDs0  doses
resulted in essentially normal tissues, as  revealed by  histopathological
examination  (Haley, Dooley, and  Harmon, 1973).

-------
Maytansine
Mirex


Ochratoxin A

Aflatoxin BI
                         Table 3.6.  Examples of histopathological  changes from acute exposures  to chemicals
Test agent
3',4'-Dichloro-
proplonanilide
Cadmium chloride
Dichlorvos
Species
Rat
Rat
Dog
Route
Oral
Intravenous
Intravenous
and oral
Dose
50-700 mg/kg
Not given
2.2-22.0 mg/kg
Histopathology
Congestive hepatitis, gastroenteritis,
and patchy emphysematous lung changes
Lesions in endothelial clefts of small
vessels in target organs such as testis
and gasserian ganglion
Pulmonary changes with generalized con-
gestion and hyperemia and cardiovas-
Reference
Chand, 1973
Gabbiani et al. ,
1974
Snow, 1973
Rat
Subcutaneous
0.38-1.0 mg/kg
Rat and    Intraperitoneal
  mouse
Rat
Oral
Monkey     Oral
330-700 ppm


15-50 mg/kg

1-3 mg/kg
  cular changes
Lesions in gastrointestinal tract mucosa,
  thymus, spleen, bone marrow, and tes-
  tis; arrest of mitotic cell division;
  hemorrhagic lesions in parenchymatous
  organs and brain; and chromatolysis and
  vacuolation of dorsal root ganglion
  cells
Patchy lesions on surface of livers that
  extended into the liver interior sev-
  eral millimeters
Severe catarrhal or erosive enteritis in
  the duodenum and jejunum
Liver damage characterized by hemorrhage,
  necrosis, and massive accumulation of
  lipid
Mugera and Ward,
  1977
                                                                                                                                        00
                                                                                                                   Kendall,  1974
Kanisawa et al.,
  1977
Rao and Gehring,
  1971

-------
                                  79
     What information do acute toxicity tests,  in particular  the
histopathological data, provide?  Robens (1979)  writes that in the
development of a drug, acute toxicity studies can:  (1)  establish some
measure of toxicity that can aid in choosing doses for studies of longer
duration, (2) help to assess whether the drug has efficacy in the non-
toxic range, (3) enable certain conclusions to be made regarding
mechanism of action, if the animals are carefully observed following
treatment and then necropsied, and (4) answer the question of how much
difference exists between dosages that seem to be safe and those that
are known to be highly toxic.  Histopathological information  derived
from acute toxicity tests can also aid physicians confronted  with cases
of accidental poisoning or attempted suicides by providing information
on chemical effects parameters such as target organs.
     Histologic evidence of damage to various organ systems as a result
of acute exposure often provides evidence that permits prediction as  to
whether the organ system will likely be similarly affected after chronic
dosing, but this is not always  the case.  Three  studies that  illustrate
that the results from acute toxicity testing cannot always be used
to predict  the results of chronic toxicity  testing are reported  by
Kanisawa et al.  (1977), Diamond and  Sleight (1972), and Pennarola,
Balletta, and DiPaolo (1969).   Kanisawa et  al.  (1977) found  that oral
administration  of near-lethal single doses  of ochratoxin  A (a mycotoxin)
to rats did not  result in renal damage, but several  consecutive daily
doses  produced  massive acidophilic  degeneration with necrosis and
desquamation  of epithelium  in the proximal tubules.   Second, in a study
of the acute  and subchronic effects of methylmercury dicyandiamide in
the  rat, Diamond and Sleight (1972)  reported that atrophy of the granular
cells  of  the  cerebellum and hepatocytes was present  only  in  those animals
subjected  to  repeated exposures.  Third,  in the research  of  Pennarola,
Balletta,  and DiPaolo (1969), acute exposure of the rabbit to parathion
resulted primarily in hyperemia of the internal organs with  pulmonary
emphysema and edema, whereas chronic exposure produced lesions of a
moderate nature; these lesions were more pronounced in the excretory
organs because of the longer contact of these organs with parathion  and
its  metabolites.

-------
                                   80
 3.4.3  Conclusions
      Many acute toxicity studies are designed to  give  a  quantitative
 measure of acute toxicity (LDsg) for comparison with other  substances
 and/or dose-ranging guidance for subchronic or chronic tests;  thus, the
 necessity of having detailed histological data is minimal.   Those  toxicity
 studies, however, that seek to completely characterize the  acute toxicity
 of a chemical would of course necessitate necropsy.  Because histopatho-
 logical changes that occur from acute exposures to  chemicals may not be
 the same as those changes that result from longer exposures and because
 many acute exposures cause death relatively quickly (especially at LDsg
 levels — see Table 3.7) and thus may not cause tissue  damage,  the
 necessity of performing histopathological examinations for  each chemical
 tested for acute toxicity can be questioned.   However, because some
 chemicals do have the potential for acute exposure  to  humans and because
 some knowledge of soft tissue changes would be useful  (e.g., for com-
 parison with other chemicals tested for acute toxicity) , gross necropsy
 seems warranted for all animals when significant  signs of toxicity are
 observed,  with consideration of histopathology for  organs showing  evidence
 of gross pathology in animals surviving 24 h or more.  This view is
 supported by Zbinden (1973) , who states that histopathological examina-
 tion of the organs is not recommended as a routine  procedure because
 (1)  the organ changes of animals dying from acute overdosage are so
 ambiguous  that an intelligent assessment would be difficult and  (2) the
 information likely to be gained over that furnished by gross necropsy
 would only occasionally justify the additional technical effort.

 3.5   OBSERVATION PERIOD

 3.5.1  Introduction
     The LDso is the dose that will  result  in  the death  of  one-half  of
the test animals, to which it  is administered.   Inherent  in  this  defini-
tion is an observation period of some finite length; however,  after
reviewing several papers that describe the  acute toxicities of various
chemicals, it is obvious that the observation  periods are not  always the

-------
Table 3.7.  LD5o observation periods
Chemical
Methylmercury
dicyandiamlde
Ochratoxin A

Aflatoxin Bj

Maytansine

Proximp'ham

Guanethidine

0-Methyl-ff-(l-
napthyl) f luoro-
acetamide
Probocol

Phenoxarsine
oxide
Pentachlorophenol
Nefopam
Phenazine-5ff-oxide
Cyclopiazonic acid
Aroclor 1242
Budralazine
Species
Rat

Rat

Monkey

Rat

Mice

Rat

Mice


Rat

Guinea pig

Mice
Dog
Rat
Rat
Rat
Rat
Route
Intraperitoneal

Oral

Oral

Subcutaneous

Oral

Oral

Oral


Oral

Oral

Oral
Intravenous
Oral
Oral
Oral
Oral
(mg/kg)
13.35

28

2.2

0.48

1 300

1 050

250 to 370


5 280

24

74
20
6 600
36 (male)
63 (female)
4 250
620 (male)
730 (female)
Observation
period
(days)
7

3

14

14

21

14

14


7

21

7
14
7
10
14
7
Time deaths
occurred Reference
(days)
Diamond and Sleight,
1972a
Kanisawa et al. ,
1977*
4 Rao and Gehring,
1971°
4 Mugera and Ward,
1977a
Lewerenz, Lewerenz,
and Plass, 1970*
Hartnagel et al. ,
1976a
5 Hashimoto et al. ,
1968*

Molello, Gerbig, and
Robinson, 1973a
8 Ballantyne, 1978fc

Ahlborg and Lars son,
1978*
2 h Case, Smith, and
Nelson, 1975a
Koeda et al., 1976*
6 Purchase, 1971b
Bruckner, Khanna,
and Cornish, 197 3a
3 Onodera et al. ,
1978*
                                                                                    00

-------
                               Table 3.7  (continued)
Chemical
2,3-Dihydro-9H-
isoxasolo-(3,2-
b)quinazolin-
9-one
Oxepinac
Fhotomirex
Ethylidene
gyromitrin
Sulfotep
Benzene
Cadmium
Di-2-ethylhexyl
phthalate
Dichlorodiphenyl-
trichloroethane
Ethanol
Clindamycin
hydrochloride
Dipterex
Dimethyl
terephthalate
Peroxyacetyl
nitrate
Chloropyrifoa
Species
Dog
Rat
Rat
Rabbit
Mice
Mice
Rat
Rat
Mice
Mice
Rat
Mice
Mice
Rat
Rat
Rat
Rat
Route
Oral
Oral
Oral
Oral
Oral
Intraperitoneal
Intraperitoneal
Oral
Intraperitoneal
Oral
Oral
Intravenous
Oral
Oral
Intraperitoneal
Inhalation
Oral
. _ Observation
£«•> SJ3
700 ± 110
136
150 to 200 (male)
70
29.4
15.2
2 940
225
37 770
237
10 600
6.08
2 618
649
3 900
95 ppm (LC50)
118 to 245
14
7
28
3.5
14
14
14
7
10
1
7
7
7
14
14

Time deaths
occurred Reference
(days)
1 Banerjee et al. ,
1977a
6 Nomura et al., 1978fc
Hallett et al. ,
1978^
3.5 Makinen, Kreula. and
Krauppi, 19776
1 Kimmerle and
Klimmer, 19 7 V3
<1 Drew and Fouts, 1974a
Kotsonis and Klassen,
1977a
Lawrence et al. ,
1975a
Tomatis et al. , 1972b
Wiberg, Trenholm. and
Coldwell, 19706
Beliles, 1972°
Gray et al., 1972a
5 Edson and Noakes,
I9606
2 Krasavage, Yanno,
and Terhaar, 197 3a
1 Kruysse et al. ,
1977°
McCollister et al. ,
                                                                                                              ts>
(various
strains)
                                                                                   1974°

-------
                                                   Table 3.7  (continued)
Chemical
trans- 2-Hexenal
Thiabendazole
hydrochloride
Cefazolin
SMA 1440-H resin
Alromine RU 100
Fenoterol HBr
Ethylene dibromide
Hexachlorophene
A204
Cadmium ions
1,1,3,3-Tetra-
ethoxypropane
2-Methylaniline
Species
Rat (male)
Rat
Mice
Rat (female)
Rat
Rat (male)
Rat (male)
Rat (male)
Rat
Rat
Rat
Rat
Route
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
._ Observation
(mg/kg) P«£J
780
3 100
>10 000 (no
deaths at 10 000)
>20 but <22
>3 200
2 300
146
69.2
7.62
130 to 180
1 610
900
14
10
7
14
10
7
14
5
7-14
14
14
14
Time deaths
occurred Reference
(days)
2 Gaunt et al., 1971a
Robinson, Stoerk,
and Graessle,
1965a
Birkhead, Briggs,
and Saunders, 1973a
Within Winek and Burgun,
3 days 1977a
Hunter and Stevenson,
1 Kast et al., 1975a*
Rowe et al., 1952a
3 Nakaue, Dost, and
Buhler, 197 3a
3 Worth et al. , 1970a
Lorke, 1978fc
4 Crawford et al.,
1965a
Jacobson, 1972a
Vs.
      paper.
                                                                                                                               00
 Foreign paper.

-------
                                    84
 same.  This section primarily discusses the length of the observation
 period of test animals following acute exposure to a chemical.

 3.5.2  Purpose of Observation Period
      The purpose of the observation period is to allow time for manifes-
 tation of the toxic effect, which in determination of the LDso  is  either
 survival or death.  The observation period thus needs to be of  sufficient
 duration that the metabolism and elimination of the test substance from
 the test animal is near completion when the LD5Q is determined. The
 importance of this is revealed by the fact that many compounds  have dif-
 ferent rates of elimination.  An example is that of three tetrachlorophenol
 compounds:  2,3,5,6-, 2,3,4,6-, and 2,3,4,5-tetrachlorophenol.   Ahlborg
 and Larsson (1978) observed that following intraperitoneal injection to
 rats, 2,3,5,6-tetrachlorophenol was eliminated within 24 h and  2,3,4,6-
 tetrachlorophenol within 48 h, but only 60% of 2,3,4,5-tetrachlorophenol
 was eliminated and subsequently recovered in the urine within 72 h.
      The observation period following acute exposure must also  be  of
 sufficient length to allow for the fact that different routes of admin-
 istration can result in the toxic effect of a chemical being manifested
 at different times.  In a study by Nomura et al. (1978), mice that
 received a lethal dose of oxepinac, an antiinflammatory drug, either
 intraperitoneally or subcutaneously, exhibited convulsion and dyspnea,
 with most deaths occurring within 24 h, whereas mice that received an
 oral lethal dose showed anorexia, emaciation, and bloody feces  with
 deaths 2 to 6 days postadministration.

 3.5.3  Length of Observation Period
      Table 3.7 summarizes reported data from 43 studies.  Examination of
 this information shows that 12 studies had an observation period of
 7  days and 18  studies had an observation period of 14 days, with the
 remaining 13  studies  having observation periods ranging from 3  to  28 days.
     The  Guidelines for CaroinoQen Bioassay in Small Rodents (Sontag,
Page,  and Saffiotti,  1976)  and guidelines proposed by the U.S.  Environ-
mental Protection Agency  (1978)  for registering pesticides in the  United

-------
                                  85

States recommend at least a 14-day observation  period.  This  14-day
period is thus the choice of some U.S.  regulatory  and  chemical  testing
agencies.  It is not surprising that many toxicological researchers  in
the United States, many of whom contribute to the  formulation of  these
agencies' guidelines, also adhere to this 14-day period.   Table 3.7
shows that 14 of 24 studies by U.S. researchers used the  14-day observa-
tion period.
     In addition to showing variation in observation periods, the informa-
tion in Table 3.7 also raises the question of which length is the most
desirable.  As previously mentioned (Sect. 3.5.2), the observation
period must be of sufficient duration to allow  for manifestation  of the
toxic effect; however, the time period cannot be so long  that the
animal's death could be attributed to factors  other than  the primary
effect of the chemical being tested.  Further  examination of some of the
papers cited in Table 3.7 yields some interesting facts with regard to
time of death following acute administration.   Rao and Gehring (1971)
subjected male cynomolgus monkeys  (Macaco, ivus) to acute oral doses of
aflatoxin BI and  found that all deaths occurred within the first 4 days
of the 14-day observation period.  Similar results were seen when
2,3-dihydro-9H-isoxazolo(2,3-b-quinazolin-9-one was administered orally
to dogs  (Banerjee et al., 1977).  Within  24 h  after dosing,  4  out of
4 dogs died at at dose of 1150 mg/kg and  2 out of 4 dogs  died  at a dose
of 750 mg/kg.  The LDsgs of mice  and rats were also determined in this
study, but no indication is provided as  to when the animals  died within
the 2-week observation period.  Research with  budralazine (Onodera  et
al.,  1978) and nefopam  (Case,  Smith, and Nelson,  1975) also  indicated
that  death from  acute exposure may occur within a few hours  or days.
The LDso  studies with budralazine showed that  most  deaths occurred
within 48 h, with all survivors  appearing normal  72 h after  dosing,
whereas  death of mice,  rats,  and dogs  occurred even more quickly with
nefopam — 1  to  10 min after intravenous injection,  15 to 30 min  after
intramuscular injection,  and 30  to 120 min after  oral dosage.
      Still  further evidence of short survival  time following acute
administration  is provided by the works of Kimmerle and  Klimmer (1974)

-------
                                   86

and Diamond and Sleight (1972).  Kimmerle and Klimmer observed that
mice, rats, rabbits, cats, dogs, and hens exposed to sulfotep, either
orally or intraperitoneally, died within 24 h and that survivors
recovered within 1 to 4 days.  Diamond and Sleight (1972) discovered
that, if rats acutely exposed to methylmercury dicyandiamide were going
to die, death would occur within 72 to 78 h.

3.5.4  Conclusions
     Although only a relatively small number of papers were reviewed, it
seems evident that many U.S. researchers employ the 14-day observation
period following acute administration of a chemical, whereas many non—U.S.
researchers appear to use observation periods of various lengths.  It is
also evident that in several instances test animals died well within the
specified observation period.  Therefore, the 14-day observation period
suggested by most regulatory agencies seems sufficient for the determina-
tion of the LD   for most chemicals.

-------
                                  87

                       REFERENCES FOR  SECTION  3
Ahlborg, U. G., and K. Larsson.   1978.   Metabolism of  Tetrachlorophenols
  in the Rat.  Arch. Toxicol.  40:63-74.

Ballantyne, B.  1978.  The Comparative  Short-Term Mammalian Toxicology of
  Phenarsazine Oxide and Phenoxarsine Oxide.   Toxicology 10:341-361.

Banerjee, B. N., R. D. Sofia,  N. J.  Ivins,  and B. J. Ludwig.   1977.   Tox-
  icological Investigation of  2,3-Dihydro-9H-isoxazolo[3,2-b]quinazolin-
  9-one (W-2429):  Acute and Subacute Toxicity in Mice,  Rats,  and Dogs.
  Drug Res. 27(1):793-801.

Beliles, R. P.  1972.  The Influence of Pregnancy on the Acute Toxicity
  of Various Compounds in Mice.   Toxicol. Appl. Phannacol.  23:537-540.

Birkhead, H. A., G. B. Briggs, and L. Z. Saunders.  1973.  Toxicity of
  Cefazolin in Animals.  J. Infect.  Dis. 128 (Suppl.):S379-S381.

Bruckner, J. V., K. L. Khanna, and H. H. Cornish.  1973.  Biological
  Responses of the Rat to Polychlorinated Biphenyls.  Toxicol. Appl.
  Pharmacol. 24:434-448.

Case, M. T., J. K.  Smith, and R. A.  Nelson.  1975.  Reproductive, Acute,
  and Subacute Toxicity Studies with Nefopam in Laboratory Animals.
  Toxicol. Appl. Phannacol. 33:46-51.

Chand, N.  1973.  Acute Toxicity of 3',4'-Dichloropropionanilide  in Rats.
  Indian Vet. J. 50(11):1122-1125.

Crawford, D. L., R.  0. Sinnhuber, F. M.  Stout, J.  E. Oldfield,  and J.
  Kaufmes.   1965.   Acute  Toxicity of Malonaldehyde.  Toxicol. Appl.
  Pharmacol.  7:826-832.

Diamond, S.  S.,  and S. D.  Sleight.   1972.  Acute and  Subchronic Methyl-
  mercury Toxicosis in the  Rat.  Toxicol.  Appl.  Pharmacol.  23:197-207.

Drew, R. T.,  and J.  R. Fouts.   1974.   The  Lack of Effects  of  Pretreatment
  with  Phenobarbital and  Chlorpromazine on the Acute  Toxicity of Benzene
  in Rats.   Toxicol.  Appl.  Pharmacol.  27:183-193.

Edson,  E.  F.,  and  D.  N. Noakes.   1960.   The  Comparative Toxicity of Six
  Organophosphorus Insecticides in  the Rat.   Toxicol.  Appl. Pharmacol.
  2:523-539.

Food Safety Council.  1978.  Proposed  System for Food Safety  Assessment.
  pp. 5, 29-33.

Gabbiani,  G.,  M. C. Badonnel, S. M. Mathewson, and G. B. Ryan.  1974.
  Acute Cadmium Intoxication:  Early Selective Lesions of Endothelial
  Clefts.   Lab.  Invest.  30(6):686-695.

-------
                                    88

 Gaines,  T.  B.   1960.   The Acute Toxicity of Pesticides  to  Rats.  Toxicol.
   Appl.  Pharmacol.  2:88-99.

 Gaines,  T.  B.   1969.   Acute Toxicity of Pesticides.   Toxicol. Appl.
   Pharmacol.  14:515-534.

 Gaunt, I.  F.,  J.  Colley,  M.  Wright, M.  Creasey,  P. Grasso,  and  S.  D.
   Gangolli.  1971.   Acute and Short-Term Toxicity Studies  on Trans-2-
   Hexenal.   Food  Cosmet.  Toxicol.  9:775-786.

 Goyer, R.  A.   1971.   Lead and the  Kidney.  Curr.  Top. Pathol. 55:147-176.

 Gray,  J.  E.,  R. N.  Weaver, J. A. Bollert, and E.  S.  Feenstra.   1972.   The
   Oral Toxicity of  Clindamycin in  Laboratory Animals.   Toxicol.  Appl.
   Pharmacol.  21:516-531.

 Haley, T.  J.,  K.  L.  Dooley,  and J. R.  Harmon.  1973.  Acute Oral Toxicity
   of tf-2-Fluorenylacetamide (2-FAA) in Several Strains  of  Mice  (abstract).
   Proc.  Soc.  Exp. Biol. Med. 143(4):1117-1119.

 Hallett,  D. J., K.  S. Khera, D. R. Stoltz, I. Chu, D. C. Villeneuve, and
   G. Trivett.   1978.   Photomirex:   Synthesis and Assessment of  Acute Tox-
   icity,  Tissue Distribution, and  Mutagenicity.   J.  Agric.  Food Chem.
   26(2):388-391.

 Hartnagel,  R.  E., B.  M. Phillips,  E. H. Fonseca,  and R. L.  Kowalski.   1976.
   The  Acute and Target Organ Toxicity of l-Methyl-3-keto-4-phenylquinucli-
   dinium Bromide  (MA 540) and Guanethidine in the Rat and  Dog.   Drug Res.
   26(9):1671-1672.

 Hashimoto,  Y.,  T. Makita, H. Miyata, T. Noguchi,  and G. Ohta.   1968.   Acute
   and  Subchronic  Toxicity of a New Fluorine Pesticide,  #-Methy!-#-(!-
   naphthyl) Fluoracetamide.   Toxicol.  Appl. Pharmacol.  12(3):536-547.

 Haynes, W.  J.   1975.   General Principles:  Dosage and Other Factors
   Influencing Toxicity.   In:  Toxicity of Pesticides.   Williams and
   Wilkins Company,  Baltimore,  pp. 37-100.

 Hunter, C.  G.,  and D.  E.  Stevenson.  1967.  Acute and Subacute  Oral Tox-
   icity of  Alromine RU 100 in Rats.  Food Cosmet.  Toxicol.  5:491-496.

 Jacobson, K. H.   1972.  Acute Oral Toxicity of Mono- and Di-Alkyl  Ring-
   Substituted Derivatives of Aniline.   Toxicol.  Appl. Pharmacol.
   22:153-154.

 Kanisawa, M., S.  Suzuki,  Y.  Kozuka, and M. Yamazaki.  1977. Histopatholog-
   ical Studies on the Toxicity of  Ochratoxin A in Rats: I. Acute Oral
  Toxicity.  Toxicol. Appl.  Pharmacol.  42(l):55-64.

Kast, A., Y. Tsunenari, M. Honma,  J. Nishikawa,  T. Shibata, and M. Torii.
  1975a.  Acute,  Subacute and Chronic Toxicity Studies  of  the Beta-
  Sympathomimetic, Fenoterol HBr on Rats, Mice and Rabbits. Oyo Yakuri

-------
                                  89

Kast, A., Y. Tsunenari, M.  Honma,  J. Nishikawa,  T.  Shibata,  and M.  Torii.
  19752?.  Acute, Subacute and Chronic Toxicity Studies of an Amino-Halogen-
  Substituted Benzylamine (Fominoben) in Rats and Mice.   Oyo Yakuri
  10(1):31-43.

Kendall, M. W.  1974.  Acute Histopathologic Alterations Induced in Livers
  of Rat, Mouse, and Quail by the Fire-Ant Poison,  Mirex.  Anat. Rec. 178:
  388.

Kinmerle, G., and 0. R. Klimmer.  1974.  Acute and  Subchronic Toxicity of
  Sulfotep.  Arch. Toxicol. 33:1-16.

Koeda, T., M. Odaki, H. Sasaki, M. Yokota, T. Niizato, H. Watanabe, H.
  Kawaoto, and T. Watanuki.  1976.  Toxicological Studies on Phenazine-
  5tf-0xide in Rats.  Oyo Yakuri 12(3):483-499.

Kotsonis, F. N., and C. D. Klaassen.  1977.  Toxicity and Distribution of
  Cadmium Administered to Rats at Sublethal Doses.   Toxicol. Appl. Pharmacol.
  41:667-680.

Krasavage, W. J., F. J. Yanno, and  C. J. Terharr.  1973.  Dimethyl Tere-
  phthalate (DMT):  Acute Toxicity,  Subacute Feeding, and Inhalation
  Studies in Male Rats.  Amer. Ind.  Hyg. Assoc. J.  34(1):455-462.

Kruysse, A., V.  J. Feron, H. R. Immel, B. J. Spit,  and G. J. Von Esch.
  1977.  Short-Term  Inhalation Toxicity Studies with Peroxyacetyl Nitrate
  in Rats.  Toxicology 8:231-249.

Lawrence, W. H., M. Malik, J. E.  Turner, A. R.  Singh, and J. Autian.   1975.
  A  Toxicological Investigation of  Some Acute,  Short-Term,  and Chronic
  Effects of Administering Di-2-Ethylhexyl  Phthalate  (DEHP)  and other
  Phthalate Esters.  Environ. Res.  9:1-11.

Lewerenz, H.  J., G.  Lewerenz,  and R. Plass.   1970.  Acute  (Mouse  and Rat)
  and Subacute  (Rat) Toxicity  of  the Herbicide,  Proximpham.  Food Cosmet.
  Toxicol.  8(5):517-526.

Lock, E. A.,  and J.  Ishmael.   1978.  The Effects of Paraquat and  Diquat on
  Rat Kidney  (abstract).   Toxicol.  Appl.  Pharmacol. 45(1):227.

Lorke, D.   1978. New  Studies  on  Cadmium Toxicology.   Proc. First Int.
  Cadmium  Conf.  pp.  175-180.

Makinen, S. M.,  M.  Kreula,  and M. Kauppi.   1977.  Acute Oral Toxicity of
  Ethylene Gyromitrin  in Rabbits, Rats and Chickens.   Food Cosmet. Toxicol.
  15(6):575-578.

Martinez,  A.  J., R.  Boehm, and M. G. Hadfield.  1974.  Acute Hexachloro-
  phen Encephalopathy:  Clinico-Neuropathological  Correlation.  Acta.
  Neuropathol.  28(2):93-104.

-------
                                   90

McCollister,  S.  B.,  R. J. Kociba,  C. G. Humiston, D. D. McCollister, and
  P.  J.  Gehring.   1974.   Studies of  the Acute and Long-Term Oral Toxicity
  of  Chlorpyrifos  (0,0-Diethy1-0-(3,5,6-trichloro-2-pyridyl)phosphorothi-
  oate).   Food  Cosmet. Toxicol. 12:45-61.

Molello,  J. A.,  C. G. Gerbig,  and  V. B. Robinson.  1973.  Toxicity of
   [4,4'-(Isopropylidenedithio)bis(2,6-di-t-butylphenol)], Probucol, in
  Mice,  Rats, Dogs and Monkeys:  Demonstration of a Species-Specific
  Phenomenon.   Toxicol.  Appl.  Pharmacol.  24:590-593.

Mugera,  G. M.,  and J. M. Ward.  1977.  Acute Toxicity of Maytansine in
  F344 Rats.  Cancer Treat.  Rep.  61(7):1333-1338.

Nakaue,  H. S.,  F.  N. Dost,  and D.  R. Buhler.  1973.  Studies on the
  Toxicity of Hexachlorophene  in  the Rat.   Toxicol. Appl. Pharmacol.
  24:239-249.

National Academy of  Sciences.  1977.  Principles and Procedures for Evalu-
  ating  the Toxicity of  Household  Substances.  Printing and Publishing
  Office, Washington, D.C.   130 pp.

National Institute for Occupational  Safety and Health.   1977.  Registry
  of  Toxic Effects of Chemical Substances, Vol. 2.  E. J. Fairchild,
  R.  J.  Lewis,  and R. L. Tatken,  eds.  987 pp.

Nomura, M., T.  Onodera,  M.  Kato, A.  Yamada, H. Ogawa, and T. Akimoto.
  1978.   Acute,  Subacute, and  Chronic Toxicity of Oxepinac.  Arzneim.-
  Forsch. 28(3):445-451.

Onodera,  T.,  S.  Takayama, A. Yamada, Y. Ono, and T. Akimoto.   1978.
  Toxicological  Studies  of  l-[2-(l,3-Dimethyl-2-butenylidene)hydrazino]-
  phthalazine,  a New Anti-hypertensitive  Drug, in Mice and Rats.  Toxicol.
  Appl. Pharmacol. 44(3):431-439.

Payne, C.  B., Jr., H. D. Kerr, and J. N.  Diaconis.  1977.  Pathophysiologic
  Effects of  Intravenous Ethchlorvynol (Placidyl) in Man Following Acute
  Pulmonary Edema.   Md.  State  Med. J. 26(6):68-70.

Pennarola, R., Balletta, and R. DiPaolo.   1969.  Histopathological Findings
  in Acute and Chronic Experimental  Intoxication with Parathion  (abstract).
  Folia Med.  52(2):118-129.

Purchase,  I.F.H.  1971.   The Acute Toxicity of the Mycotoxin Cyclopiazonic
  Acid to Rats.  Toxicol. Appl. Pharmacol.  18:114-123.

Rao, K. S., and P. J. Gehring.  1971.  Acute Toxicity of Aflatoxin Bj in
  Monkeys.  Toxicol. Appl. Pharmacol. 19:169-175.

Robens, J. F.  1979.  Assessment of  the Safety of Drugs for Animals.  Vet.
  Hum. Toxicol. 21(l):12-23.

-------
                                  91

Robinson, H. J., H. C. Stoerk, and 0.  E. Graessle.  1965.  Studies on the
  Toxicologic and Pharmacologic Properties of Thiabendazole.  Toxicol.
  Appl. Phannacol. 7:53-63.

Rowe, V. K., H. C. Spencer, D. D. McCollister, R. L. Hollingsworth, and
  E. M. Adams.  1952.  Toxicity of Ethylene Dibromide Determined on
  Experimental Animals.  Arch. Ind.  Hyg. Occup. Med. 6:158-173.

Snow, D. H.  1973.  The Acute Toxicity of Dichlorvos in the Dog:  2.  Path-
  ology.  Aust. Vet. J. 49(3) .-120-125.

Sontag, J. M., N. P. Page, and U. Saffiotti.  1976.  Guidelines for Carcin-
  ogen Bioassay in Small Rodents.  National Cancer Institute, NC1-CG-TR-1.
  65 pp.

Tomatis, L., Y. Turusov, N. Day, and R. T. Charles.  1972.  The Effect of
  Long-Term Exposure to DDT on CF-1 Mice.  Int. J. Cancer 10(3):489-506.

U.S. Environmental Protection Agency.   1978.  CFR 40 No. 163.81.

Wiberg, G. S., H. L. Trenholm, and B.  B. Coldwell.  1970.  Increased
  Ethanol Toxicity in Old Rats:  Changes in LDso, In Vivo and In Vitro
  Metabolism, and Liver Dehydrogenase Activity.  Toxicol. Appl. Pharmacol.
  16:718-727.

Winek, C. L. , and J. J. Burgun.  1977.  Acute and Subacute Toxicology and
  Safety Evaluation of SMA 1440-H Resin.  Clin. Toxicol. 10(2):255-260.

Worth, H. M., D. B. Meyers, W. R. Gibson, and G. C. Todd.  1970.  Acute
  and Subacute Toxicity of A204.  In:  Antimicrobial Agents and Chemo-
  therapy - 1970.  pp. 357-360.

Zbinden, G.  1973.  Acute Toxicity.   In:  Progress  in Toxicology,  Springer-
  Verlag,  Inc., New York.  pp. 23-27.

-------
                       4.   SUBCHRONIC  TEST DESIGN

4.1  INTRODUCTION

     The standard protocol for toxicity testing  of  chemicals  generally
proceeds from single-dose tests of short duration  (acute)  to  repeated-
dose tests of long duration (chronic).  Part  of  this  procedure is  a
repeated-dose study of intermediate duration,  referred to  as  a sub-
chronic or subacute test.   The subchronic test provides information on
the toxic effects of chemicals that are likely to  occur from  repeated
exposures over a limited time period.   Currently 14-, 28-, and 90-day
studies are commonly used; however, a  review  of  the literature indicates
the duration of the study may vary widely.
     The first recorded definition of  a subchronic test was as follows:
a repeated exposure test with a duration of one-tenth the  expected life
span of the experimental animal used (Food  and Agriculture Organization/
World Health Organization Technical Report, 1958).   Boyd (1961) thought
that for the rat this meant a duration of approximately 100 days.   A 13-
week test with four dosage levels was  the definition given for subacute
tests by Abrams, Zbinden, and Bagdon  (1965).   Loomis (1974) described
the repeated exposure test as a "prolonged test1' with daily exposure for
about 3 months.  Guarino  (1979) stated that traditionally three terms
(short-term chronic, subchronic3 or prolonged) are used interchangeably
to designate a 90-day test.  Many similar definitions, based upon a
repeated dose given over  a period of a few days to 3 months, have
appeared in the literature, including some by expert committees (National
Academy of Sciences, 1975; 1977).  The cumulation of these efforts was
the definition and guidelines  of the U.S. Environmental Protection
Agency  (EPA) pesticide programs  (Federal Register, 1978).  This guide-
line set standards for subchronic  tests  covering dosage levels, routes
of exposure, number of species, number  of test  animals, type and  degree
of pathology, hematology, biochemical and function evaluations, etc.
The choice of subchronic  as  the  term  of  preference  is  followed in this
text and replaces all other  terms.  "However, the  important  thing is not
the choice of definition  of  a particular fraction  of the  life span but
                                    93

-------
                                    94
 the selection of a testing interval that is  as  short  as  practicable and
 yet will give meaningful information about the  effect of absorbing the
 toxicant during an entire lifetime" (Hayes,  1975).  In addition  to
 summarizing the definition of a subchronic test,  this quotation  describes
 the purpose of this chapter,  which is to evaluate the currently  employed
 subchronic tests to determine whether they provide sufficient  informa-
 tion that makes longer-term tests unnecessary.  The section does not
 consider a comparison of time versus effect  for dermal,  ocular,  or
 inhalation toxicity and does  not consider carcinogenic effect.
      The two primary functions of subchronic tests, as they are  currently
 used, are (1) to assess toxic effects from repeated exposure over a
 relatively short time so as to give maximum  and minimum  effect levels
 with safety margins plus a prediction of possible effects from longer
 exposures and (2) to determine the appropriate  dose levels to  be used if
 longer exposures are planned  (Benitz, 1970;  Feck, 1968).  The  purpose of
 this chapter is to evaluate the first of these  two functions.  To fulfill
 this first function, a battery of biochemical,  hematological,  and patho-
 logical tests is performed.  These tests provide  data upon which more
 selective evaluations can be  based.  The target organ data are especially
 useful.   The value of such data has been recognized for  several  decades,
 and,  as stated by Barnes and  Denz (1954), "It is  in fact doubtful
 whether it is worth proceeding beyond the subacute test."  The second
 function is an elaboration of the acute evaluation of effective  doses
 and should give maximum and minimum effect levels with safety  margins.
 The role of the subchronic test as a preliminary  evaluation of appro-
 priate dose levels for use in longer test designs (such  as a carcino-
 genicity bioassay)  is evaluated in a separate review  by  Prejean  (Appendix
 A).   However,  as the time and cost requirements of toxicity testing
 increase,  so does the value of subchronic tests as reasonable  evalua-
 tions  of chronic effects.   If a more efficient  subchronic test design
 can be  constructed,  the need  for longer, more costly  chronic studies may
be reduced.
     The standard or typical  experimental design  for  a subchronic test
has often been  discussed in the literature.   Recent examples include a

-------
                                  95
discussion of procedures for the preclinical toxicologic  evaluation of
cancer chemotherapy agents (Prieur et  al.,  1973);  a proposed system for
food safety assessments (Food Safety Council,  1978);  a review of
principles and methods for evaluating  chemical toxicities (World Health
Organization, 1978); and proposed designs  being developed by the Inter-
agency Regulatory Liaison Group (IRGL) and Organization for Economic
Cooperation and Development (OECD).  A representative sample of these
test designs is included in Table 4.1.  Overall these systems are quite
similar.  Selected areas of these standard designs will be reviewed in
this document to determine if modifications are possible.  Thus, the
designs in Table 4.1 represent the baseline system for which changes
will be suggested.

4.2  SPECIES

4.2.1  Introduction
     The debate over which and how many species to employ in subchronic
toxicity tests has been a part of toxicology for many years.  Discussions
generally center on:   (1) what species best represents human responses,
 (2) what is  the most  sensitive species for  a given test  or  effect,  and
 (3) what is  the value  of rodent  and nonrodent  combinations.  To evaluate
these  problems, this  review will examine literature  reviews and discus-
sions  concerning species applicability and  data from species  comparison
studies.

4.2.2   Discussions in Literature Reviews  Concerning Species Suitability
      Barnes  and Denz  (1954)  in their  review of chronic tests discussed
 the choice of test animals.   They ranked  the rat  as the "first choice"
because of economic and scientific factors.  The  choice of a second,
nonrodent  species  should be based on  the  similarity to human responses.
 Both the monkey  and dog have disadvantages, particularly if the monkeys
 are imported or  the dogs are mongrel  breeds.   Further, Barnes and Denz
 questioned how often these two species added to the information gained
 by the rat.   Only if inhalation studies (monkey)  or large  tissue/blood

-------
                             Table 4.1.  Experimental design for recently proposed subchronic oral toxicity tests
Agency
Federal Insecticide,
Fungicide, and
Rodenticide Act
(Toxic Substances
Control Act) guide-
lines, 1978-1979a
Interagency Regulatory
Liaison Group, 1979a
Species
Two (rat and
nonrodent)
Two (rat and
dog)
Sex Age at start
Both Rats - 6 weeks
Dogs — 4 to
6 months
Both Rats - 6 weeks
Dogs — 4 to
6 months
Number of animals
per group
(sex per dose)
Rats - 20
Dogs — 6
Rats - 20 (30 days
to 6 months) , 10
(<30 days)
Number
Duration of
doses
Rodents - 90 days 3
Dogs — 6 months
Generally 90 days; 3r
shorter for spe-
cial situations
Dose levels
Highest - effect level,
but not more than
10Z mortality
Lowest — no evidence
of toxicity
Highest — toxic level,
but not excess
mortality
World Health Organi-
  zation, 1979
Food Safety Council,
  1978
Two (rodent and   Both   Rats - just after   10
  nonrodent)               weaning
Two (rodent and
  nonrodent)
Both   (Not indicated)
Rodents - 20
Nonrodents — 3 to
                                                10% life span or 3
                                                  months
Rodents - 90 days
  to 1 year
Nonrodents — 1 year
Lowest — no evidence
  of toxicity

Highest — distinct
  toxic level
Lowest — no detectable
  toxic reaction

Highest — clearly
  toxic, but not lethal
Lowest — no observable
  adverse effect, but a
  reasonably large mul-
  tiple of estimated
  human daily intake
                                                                                                                                                    vo
      Proposed  or  unofficial guidelines.
      Must have control groups also.
     Source:  Adapted from Page,  1979.

-------
                                  97
samples (dog)  are needed,  do these species  add  significant  data.   They
concluded that the use of  a second species  should  be  flexible, with the
toxicologist choosing the  best species,  depending  on  the  chemical  or
test design requirements.
     In later discussions  more emphasis  is  placed  on  the  relationship
between the test species and the human response rather than on economic
or logistic variables.  A good example of this  change is  evident  in a
discussion of inhalation tests (Roe,  1968), in  which  specific compari-
sons of the respiratory systems of several  laboratory species are made.
By examining particle deposition patterns and mucous  secretion mechanisms,
Roe dismissed the mouse, guinea pig,  rabbit, and cat  as suitable  test
animals.  On the basis of anatomical considerations,  monkeys and  dogs
resemble man more closely than do other  species, and  Roe recommended
these for "small-scale, short-term experiments."  The value of the rat
apparently lies between these two groups, because it  is more similar to
man in anatomy and response than the first  group and of a more manageable
size and more certain genetic makeup than the dog or monkey.  Thus, the
importance of similarity to the human response in species selection is
elevated in the present discussion to at least the same level as  economic
and other practical  considerations.
     In a later review of  chronic toxicity, Benitz (1970) documented the
frequency of use  for various  species and the reasons behind  the choices.
He gave the following percentages for the use of  each  species based  on
134 studies of 1  month or  longer  duration  (these  studies appeared in
Toxicology and Applied Pharmacology between 1959  and  1966):  rat, 43.3%;
dog, 38.1%; monkey,  6.7%;  mouse,  3.7%;  and  rabbit, 3.0%.   The emphasis
on the rat and dog is supported in a report by  Bushby, Lechat, and
Santarato  (1966)  in  which 71.7% of all  toxicity testing  laboratories
used the dog and  rat and  only 16% used  the rat, dog,  and monkey.   Benitz
attributed  this  to investigator  familiarity with  the rat and dog as test
species rather than  to  these  species' superiority in sensitivity or
comparability  to  human  response  patterns.
     Following the method of  Pinkel  (1956), several  authors have evalu-
ated species  suitability  in anticancer  drug studies  by using doses based
on milligrams  per square  meter (surface area)  instead of milligrams per

-------
                                    98

kilogram (body weight).   This technique is useful because surface  area
correlates  better with metabolic rate than body weight does.  Therefore,
by  giving doses on a surface area basis the researcher obtains  tighter
control  of  the dosage variable, allowing a truer evaluation of  species
differences.   The results obtained when dosage sensitivity was  compared
in  this  manner suggested that in most cases the monkey, the rat, and  the
dog all  give  similar results (Freireich et al., 1966).  However, in such
comparisons between the dog and monkey using the maximum tolerated dose,
Roman  (1972)  found that the dog was generally more sensitive  and should
be  preferred  over the monkey.
     In  an  evaluation of the predictive value of test species for  the
metabolism  of drugs, Smith (1979) compared the dog, monkey, and man.  In
contrast to Roman (1972), Smith preferred the monkey.  By discussing  and
rating metabolic fate in 34 compounds for these species  (Table  4.2),
Smith  concluded that "(1) for a large group of relatively dissimilar
compounds the rhesus monkey is a significantly better predictor of
metabolic fate in man than either the dog or the rat and (2)  that  there
is  little to  choose between the latter two species, in both cases  the
correspondence is poor for over half the compounds regardless of their
chemical classification."
     Zbinden  (1963)  discussed species selection in evaluations  of  drug
toxicities.   He favored the use of the dog and the rat, with  additional
use of the  monkey if confronted by unexplained toxic effects.  However,
              Table 4.2. Ranking of various animal models as predictors of
                             metabolic fate In man
Species
Rating

Good
Fair
Poor
No data
Total
Rat
Number
of compounds
5
7
19
3
34

Percent
14
21
53
12
100
Dog
Number
of compounds
6
8
20

34

Percent
18
23
59

100
Monkey
Number
of compounds
22
9
3

34

Percent
65
26
9

100
   Source:  Adapted from Smith, 1979.  Reprinted with permission of the publisher.

-------
                                   99
the key factor is metabolic similarity to man.   Since  absorption,
penetration, and distribution processes are similar in most  species,  the
primary differences between species are due to  variations in biotrans-
formation pathways and in rates of inactivation.  The  final  selection of
test species should reflect the species' similarity to man in metabolizing
the chemical under study.
     In another review of drug safety evaluations, Peck (1968) stated
that the basis for experimental species selection should be  a metabolic
similarity to man.  However, as he pointed out, the human metabolic pat-
tern is generally unknown for new drugs.  His final recommendation was
to include several species in drug tests in order to protect against
unknown metabolic variations.
     However, Boyd (1968), in discussing species selection in his review
of drug testing, did not base the choice on metabolic similarity alone.
He felt another factor of equal importance is the ability to vomit (which
parallels the human protective reflex).  Boyd suggested the use of a
species that has vomiting capabilities  (recommending the dog) and one
nonvomiting species that is metabolically similar to man.  Although Boyd
discussed only the vomiting ability of  the test species, this factor is
representative of many such physiological traits.  His conclusions can
be expanded to include the recommendation that  the test species should
have a physiological  functional pattern that is as similar as possible
to the human pattern  for the specific  test chemical.   If such informa-
tion is not known, then "use as many species as may be possible."
     This reliance on several  species  is recommended  again by McNamara
(1976), although he did point  out  that many researchers  feel comfortable
using only  the rat.   McNamara  cited Hebold  (1972)  regarding  the  regula-
tions of six  countries and  three  international policy-setting bodies re-
quiring the use  of the rat,  dog,  and  rabbit, or monkey as test  species.
Hayes  (1967a) also feels  the need exists to  use several species  in
studies of  pesticide  toxicity.  This  is based  on both the general
variability between animal and human responses and the importance of the
species selected in determining toxic results  (second only to dose
levels employed).  Guarino (1979), in his discussion of drug development
programs,  saw a definite value in using many species.  He recommended

-------
                                    100
 the use of "(1) mice and rats for experimental therapeutic data;  (2)
 dogs, monkeys, and mice for toxicologic studies; and (3)  dogs,  monkeys,
 rats, and mice for pharmacologic information."  This scheme is  suggested
 in contrast to the "self-perpetuating choice," based on economic  or
 convenience factors, of the rat and dog as test species.
      Balazs (1976) discussed the rat and dog combination for testing
 chemicals.  He favored the rat because large numbers can be used, and it
 has a relatively short life span.  The choice of the dog allows for
 "in-depth studies of organ systems" and provides greater test sensitivity.
 The use of a third species is variable but "the omission of these species
 [rat and dog]  is seldom justified."  This policy is also recommended  by
 the Food Safety Council (1978),  and in the EPA Proposed Guidelines for
 Pesticides (Federal Register,  1978).   The EPA position is stated  as:
 "Testing shall be performed in at least two mammalian species.  One
 species shall  be a generally recognized strain of laboratory rat.   The
 second species shall be a nonrodent.   The nonrodent species should
 usually be the dog.   Selection of a nonrodent species other than  the  dog
 will require full and adequate justification which should consider such
 factors as the comparative metabolism of the chemical and species
 sensitivity to the toxic effects of the test substance,  as evidenced  by
 the results of other studies."   Thus,  it is assumed that  for most
 substances the combination of  the dog and rat is preferred, although  the
 scientific basis of this conclusion is  not stated.
      However,  in contrast to many discussions concerning  the dog  and  rat
 as  test species,  Aviado (1978)  saw little advantage to including  the
 dog.   In a two-part  literature  review,  Aviado evaluated 110 studies in
 which the dog  and rat were used  as test species during the period 1966
 to  1978.   In most studies the data obtained from the rat  predicted
 cardiovascular,  bronchopulmonary,  hepatorenal,  digestive,  nervous, and
 somatic  system toxicities as well  as  did data obtained from the dog.
 Further,  the rat  studies  were better  than the dog studies for prediction
 of pancreatic  hypertrophy,  hepatic porphyria,  and nephropathy.  He
 concluded  that  the use  of the dog  or  the rat in chronic studies is
sufficient and  that using both species  results  in unnecessary duplica-
tion of scientific manpower  and research effort.

-------
                                  101
     The review by the World Health  Organization (1978)  on toxicity
evaluations summarizes most of the primary factors  discussed  in litera-
ture reviews for the selection of test species.   Of paramount importance
is the similarity of metabolism between the test species and  man.   If
sufficient data are available to allow a choice  on  this  basis, the
species showing the greatest metabolic similarity to man should be used.
If such data are unavailable, then qualitative and  quantitative response
patterns should be obtained for several test species to  increase the
predictive value for the occurrence  of similar toxic effects  in man.  In
most cases this necessitates the use of the dog and rat  as a  minimum in
subchronic studies.  However, despite such reviews  as the World Health
Organization document and Aviado's literature review, the conclusion of
Fancher (1978) in his review of animal models for toxicological studies
is in essence the current general practice in species selection:  "In
spite of all that has been done, choice of species  continues  to be
based, in large part, on precedence, convenience, and economics."

4.2.3  Species Comparison Studies
     Ansbacher, Corwin, and Thomas  (1942) used  the dog, cat,  rabbit, and
monkey to test menadione and menadiol for toxicity.  After subcutaneous
dosing for 4 to 11 days, all species except the monkey developed  a  weak
anemia, with the dog  also showing hemoglobinuria.  The dog and cat
showed some mortality at the higher doses.  In  the discussion of  the
results, Ansbacher et al. felt  that the hematologic  changes  could be
attributed to normal  compensatory mechanisms.   Thus, the  mortality
exhibited by the cat  and dog  implies  a  greater  sensitivity for these two
species than for the  rabbit and monkey.
      In an inhalation study  of  beryllium  toxicity,  Stokinger et al.
 (1950) used 11  species,  examining mortality,  growth retardation,  hema-
tology, and urinalysis  factors.  The  animals  were  exposed 6  h per day
for  14 days at  100 mg/m3,  51 days at  47 mg/m3,  95  days  at 10 mg/m3, and
100  days  at 0.95 mg/m3.  For mortality the rat  and cat  were  the most
sensitive species  at  intermediate and high doses,  with the dog showing
equal mortality rates at the high dose only.   In growth response,  the
monkey was  the most  affected species at the intermediate and high  doses.

-------
                                    102
 The dog showed the most toxic effects in hematologic  and urine analyses
 In summation,  the dog appeared to be the most  sensitive species, but the
 use of only five animals per test dose  may limit  the  validity of this
 study.  Stokinger and Stroud (1951)  tested the dog, rat, and rabbit once
 again for hematological effects from beryllium inhalation.  The exposures
 were 6 h per day, 5 days a week for 6 weeks (continued for  23 weeks in
 the rabbit) at 2.2 to 4.0 mg/m3.   The result was  a decrease in red blood
 cells and an increase in mean corpuscle volume.   Their comparison showed
 that the degree of anemia varied among  species, with  the dog being the
 most susceptible and the cat the least  susceptible.
      Rowe et al. (1952) compared the sensitivity  of the rat, guinea pig,
 rabbit, and monkey to ethylene dibromide using repeated 7-h per day
 inhalation exposures with concentrations of 100,  50,  and 25 ppm.  Sub-
 chronic exposures of less than 91 days  showed  toxic effects only at the
 higher doses.   All species showed some  weight  loss, but only changes in
 the guinea pig were significant.   Mortality levels were statistically
 significant at the high dose in rats, as were  increases in  liver and
 kidney weights.   This increase in organ weight was also shown by the
 guinea pig. Although Rowe et al. concluded that  no species did partic-
 ularly well at high dose, it is apparent that  the rat and guinea pig
 showed the greatest sensitivity to ethylene dibromide.
      Litchfield (1961)  designed a study to compare experimental animal
 response and human response to toxic chemicals.   By comparing the occur-
 rence of toxic effects  from tests performed on the rat and  dog, and from
 known human epidemiological studies, Litchfield was able to evaluate the
 relationships  for six chemicals.   The tests included  acute  studies,
 subchronic studies  (1 to 6 months),  and a 1-year  rat  study. The appear-
 ance  of  toxic  effects in each species and in each species combination
was the  basis  for his evaluation.   The  results are shown in Table 4.3.
The data support  his  conclusion that the dog better represented man's
response  than  did the rat.   However, in summation, Litchfield noted that
the small number  of chemicals  examined  reduced the statistical value of
this comparison,  and  he  urged  that more species be examined for use in
toxicity studies.

-------
                                    103
Table 4.3.  Occurrence of 39 physical signs  from six drugs  in  three species


Physical sign

Weight loss
Weight gain
Muscle atrophy
Myositis
Lymphocytopenia
Neutropenia
Leukopenia
Anemia
Leukocytosis
Hyperglycemia
Liver damage
Jaundice
Fatty liver
Polydipsia
Polyuria
Oliguria
Hematuria
Crystalluria or renal
concretions
Renal damage
Gastroduodenal ulcer
Diarrhea
Salivation
Ataxia
Impaired reflexes
Decreased activity
Tremors
Ptosis
Catatonia
Priapism
Lacrimation
Urinary incontinence
Bacterial invasion
Parasitic invasion
Decreased thyroid function
Genital hypoplasia
Decreased adrenal function
(cortical)
Hypotension
Lung edema
Tachypnea
Total
Source: Adapted from


Absent Rat


3 1
4
5
4
5
3
1
3
5
2 1
5
5 1
5
2
4
3

5
3 1
4
1
5
2
3
3
3
5 1
5 1
5 1
5
4 1
5
4
4
4 2

5
2 1
5
5
146 11
Litchfield, 1961
Association. Reprinted with permission of

Rat
Dog and Man
dog
2
2
1

1
1
1 1
2 1
1 2

2


1
3
1
1


1
1
1 4
1
111
1 1
2
2



1
1

1
1



2

1
16 8 23
. Copyright 1961,
the publisher.
Rat,
Rat Dog d '
and and and'
"*" man man
4

1
1
1

1
2

1
1
1


1
1
2

1
1
1


1
1
1
1





1
1
1


1
1
1

1 12 17
American Medical


-------
                                   104

     In a similar study of anticancer drug toxicities, Owens (1962)  com-
pared the response of the rat, dog, and monkey to man in terms of bone
marrow, gastrointestinal, hepatic, renal, and neural toxicities.   Table
4.4 summarizes the results of 4-week exposure tests.  Owens concluded
that except for neurotoxicity all three species were suitable, and in
particular he saw no advantage in the use of the monkey rather than the
dog.

     Weil and McCollister (1963) evaluated several factors in toxicity
tests, including species sensitivity.  Although their study concentrated
on diet exposure studies in the rat, they also included some data on dog


                  Table  4.4.  Organ  system toxicities

             Compound                 Rodent    Dog    Monkey    Human

                         Bone marrow toxicity

 Mechlorethamine                         +       +        +        +
 Cyclophosphamide                        +       +        +        +
 Myleran                                 +       +        +        +
 1-Sarcolysin                            +       +        +        +
 2-Chloroethylmethanesulfonate                   +        +        ?
 Methotrexate                            +       +        +        +
 6-Mercaptopurine                        +       +                 +
 4-Aminopyrazolo (3,4-D)pyrimidine       +       +                 +
 Vincaleukoblastine                      +       +                 +
 Carzolamide                             -                         +
 Actinomycin P2                          +       +                 +a
 Mithramycin                             -                         +
 Roseolic acid                           -       +                 +

                   !   Gastrointestinal toxicity

 Mechlorethamine                         +       +        +        +
 Cyclophosphamide                        +       +        +        +
 Myleran                                 +       +        +        +
 1-Sarcolysin                            +       +        +        +
 2-Chloroethylmethanesulfonate                   +        +        +
 Methotrexate                            +       +        +        +
 6-Mercaptopurine                        +       +                 +
 4-Aminopyrazolo (3,4-D)pyrimidine       +       +                 +
 Carzolamide                                     +                 +
 6-Azauracil                                     i        ~        "*"
 Vincaleukoblastine                      +       +                 +
 Actinomycin P2                          +       +                 +
 Mithramycin                             -       ~                 +

-------
                                   105

                          Table 4.4 (continued)
             Compound                  Rodent    Dog    Monkey    Human


                        Nervous system toxicity
 Mechlorethamine                         +       +        +        +
 Chloroquine mustard                             +        +        +
 Carzolamide                             -       -                 +
 6-Azauracil                             +       -        -        +,
 Vincaleukoblastine                      -       -                 +•,
 Nitrofurazone                           -       -                 4-
 NSC 38280                               -       -                 +°

                      Skin and appendages toxicity
                       (dermatitis and alopecia)
 Cyclophosphamide                                                  +
 Methotrexate                            -       -                 +
 Vincaleukoblastine                      -       -                 +
 8-Azaguanine                            -       -                 +
 Actinomycin P2                                                    +
 Methylglyoxal-bis-guanylhydrazone       -       -                 +

                            Hepatic toxicity
 2-Chloroethylmethanesulfonate            +       +                 +
 6-Mercaptopurine                         +       +                 +
 4-Aminopyrazolo (3,4-D)pyrimidine        +       +                 +
 Carzolamide                              +       +                 +
 Mithramycin                              +       +                 +
 Roseolic acid                            -       +                 +

                             Renal toxicity

 2-Chloroethylmethanesulfonate            +       +                 +
 Aminoiminomethanesulfinic acid           +       +                 +
 Puromycin nucleoside                     +       +                 +

      Predominantly thrombocytopenia.

       Peripheral neuropathy.

       Extraocular palsies.

      Source:  Adapted from Owens, 1962.  Reprinted with permission
 of the publisher.
studies, for durations up to 2 years.  They found 21 studies in which
the rat and dog were tested together.  In none of these tests was  the
dog more sensitive, and in seven tests it was less sensitive than  the

rat.  They concluded that the use of the dog should be limited to  90-day

-------
                                   106
 studies,  and only when it was  indicated  as  a more  sensitive species
 should it replace or augment the rat  in  long-term  studies.
      The  use of the pig as a test species was  discussed by Earl et al.
 (1964).   By injecting 1, 2,  and 4 mg/kg  of  amphotericin B intravenously
 for 3 days per week over 13  weeks, they  were able  to  test the similarity
 of the pig's response to that  of man.  Unlike  previous rat and dog
 studies on amphotericin B, the toxic  effects appearing in the pig  (renal
 damage and anemia) were the same as human responses.  They concluded
 that the  pig may be a useful test species because  of  these similarities
 to man and because of the ease with which blood  and organ samples are
 taken.
      In a study of the pesticide zenophos,  Kohn, Kay, and Calandra
 (1965) orally dosed the rat  and dog for  90  days  with  diet levels of 0.5,
 2, 8, and 25 ppm.  At the high and intermediate  doses both species
 exhibited lowered food consumption, depressed  growth  rates, and depressed
 cholinesterase activity.  Additionally,  the dog  showed changes in its
 hematologic pattern.   The effects shown  were attributed  (except for
 cholinesterase activity) to  poor diet palatability and not to species
 differences.
      McNerney and MacEwen (1965) studied the inhalation toxicities of
 ozone, carbon tetrachloride, and nitrous oxide in  five test species
 after continuous exposure for  2 weeks.   The effects were generally the
 same for  all  five species, except for the greater  susceptibility to
 nitrous oxide and a resistance to ozone  exhibited  by  the monkey.  In
 contrast,  the dog showed the opposite pattern  of effects.  Also, a
 strain difference in the rats  appeared significant, as Wistar rats were
 extremely  susceptible to carbon tetrachloride  while Sprague-Dawley rats
were not.
      A study  using the rat and dog to test  the oral toxicity of EX5004,
a  sympatholytic  drug,  showed that after  a 3-month  exposure the dog was
more affected than the rat (Yeary, Brahm, and  Miller, 1965).  The
exposure levels  were  10, 100,  500, or 1000  mg/kg in the diet for the  rat
and  for the dog  10,  100, or  500 mg/kg given by gelatin capsule.  The  dog
showed tachycardia and elevation of alkaline phosphatase activity  at  the
high  and intermediate  doses  while the rat did  not.  This response was

-------
                                  107

closer to the expected human response  pattern,  indicating that  for these
variables the dog was the most sensitive test species.
     Atkinson et al. (1966) studied the effects of cephaloridine,  an
antibiotic, by using daily intramuscular or subcutaneous injections into
four species (cat, dog, rat, and rabbit) for 56 to 84 days.   The major
significant effect was an increase in kidney weight in the cat  (at 50
and 150 mg/kg), rabbit (at 30 and 45 mg/kg), and rat (at 50 and
150 mg/kg).  However, the rat was the only species showing kidney
damage.  There was also a slight, transient leukocytopenia in the rat
and dog and some enlargement of the liver (without histopathologic
damage) in the rabbit and cat.  In this study it appeared that the rat
would have been sufficient as a test species.
     Newberne, Gibson, and Newberne (1967) studied an analgesic drug in
the dog, rat, and monkey using oral dosing for 1 to 4 months.  As shown
in Table 4.5, the dog was overwhelmingly more sensitive than the rat or
monkey, especially  for histopathologic and clinical chemistry effects.
The only significant effect that  appeared in the monkey and not in  the
dog was the demyelination of the  cerebral gyri.  Thus, overall the  dog
was the most sensitive species in this study.
     Hagan et al.  (1967) compared the  toxicities of  food  flavorings
using  oral exposure by diet or gavage  for  the  dog  (beagle)  and  the  rat
 (Osborne-Mendel).   For 6-methylcoumarin, they  found  at  the  intermediate
dose  (10 000 ppm  for  rat and  150  mg/kg for dog)  that 20  rats showed no
effect after 14 weeks while the  dog tested became  moribund  after  5  weeks,
exhibiting liver  damage,  skeletal muscle damage,  and general weakness.
For methyl salicylate the  rat  showed  growth retardation after  17  weeks
at the intermediate dose (10  000 ppm).  The dog showed weight  loss and
 slight microscopic liver damage after no more than 29 days at  800 mg/kg.
One dog  died after 4 days of  dosing.   With safrole ingested at inter-
mediate  doses  (5000 ppm),  the rat exhibited growth retardation, liver
pathology, slight leucocytosis,  and anemia after dosing for 2  years.
The dog, however, showed moderate kidney and liver damage and  general
weakness after 96 to 116 days at 40 mg/kg.  In these studies,  the dog
generally  appeared to be more sensitive to the additives than the  rat,
 even  with  shorter exposure periods.

-------
 Table  4.5.   Comparison of significant variations in the response of four species  to  an  oral  analgesic12
Parameter Dog
Clinical observations
Depressed weight gain or
weight loss +a
Depression or
unconsciousness +
Ernes is +
Icterus + —
Death +
Clinical chemistry
Elevated serum glutamic +
pyruvic transaminase
Elevated serum alkaline
phosphatase +
Bilirubinuria +
Necropsy observations
Discolored, yellow liver +
Histopathologic observations
Hepatic necrosis +
Myelin figures in
hepatocytes +
Excess bilirubin — liver
and kidney +
Hemosiderosis +
Depressed spermatogenesis +
Demyelination of cerebral
gyri
Dosage*
(mg/kg/day)


30

30
30
	 30
30

10


10
60

30

30

30

30
30
30

60
»» i Dosage
Monkey , /, / . »
(mg/kg/day)


+ 25

+ 30
+ 25
60
+ 30

60


60
60

60

60

+ 30

60
60
60

+ 30
Dosage*
(mg/kg/day)


+ 30

100
100
100
100

100


100
NTe NT

100

100

100

100
100
100

100
Rabbit


_

-
-
-
-

NT


NT
NT

—

NT

NT

NT
NT
NT

NT
Dosage*
(mg/kg/day)


80

80
80
80
80

NT


NT
NT

80

NT

NT

NT
NT
NT

NT
al-2-(l-Methyl-2-piperidyl)-l,l-diphenylethyl propionate hydrochloride.
 Minimum dose at which an effect occurred or the highest dose given.
^Indicates a response in one or more animals at the indicated dose.
 Indicates no abnormal response.
eNo test.
Source:  Adapted from Newberne, Gibson, and Newberne, 1967.  Reprinted with permission of the publisher.
                                                                                                                     o
                                                                                                                     oo

-------
                                   109
     In a toxicological study of aldrin and dieldrin, Hodge,  Boyce,
Deichmanne, and Kraybill (1967)  noted the subchronic response of  several
species by comparing histopathologic data, body weight  changes, mortality,
and organ weight changes.  Using these criteria, they found wide  vari-
ability in species response.  They found the monkey was the most  sensitive
for body weight and mortality criteria, and both the dog and  rat  were
sensitive to histopathology and liver weight criteria.   In this study it
was the sensitivity of the species that was evaluated and not the
similarity to human responses.  The significance of this study lies in
the selection of the rat, dog, and monkey as the best  indicators  of
toxicity from among a list of ten species.
     Benitz, Roberts, and Yusa (1967) studied the morphological changes
in the rat, dog, monkey, and mouse after dosing for 27  to 38  days with
minocycline, a tetracycline antibiotic.  In the rat, minocycline produced
a black discoloration of the thyroid  (pigment deposition) with oral
doses of 8, 25, and 75 mg/kg per day.  Hyperplastic changes were asso-
ciated with this discoloration.  In the dog, the discoloration and
hyperplasia occurred with intravenous doses of 5, 10,  20, and 40 mg/kg
per day.  Additionally, various degrees of hemolytic anemia  occurred  in
the dose range 10 to 40 mg/kg per day.  In the monkey, the discoloration
occurred (pigmentation less pronounced) at an oral level of  30 mg/kg  per
day, but no hyperplastic changes were observed.  However, in the mouse,
no changes of any kind were noted with 250 mg/kg per day of  oral dosing.
The results in this limited evaluation of toxicity indicated that  the
rat and dog were the most sensitive  species, followed  by  the monkey.
The mouse was the least  sensitive  species tested.
     Schein et al.  (1970) evaluated  the monkey (four species) and  dog
(mongrel and beagle) for their  potential  value in  predicting qualitative
toxicities in man using  25  anticancer drugs.   They used data from their
own tests  (3 to 90  days  of  intravenous or peroral  administration), data
found  in the literature, and data from clinical studies in man.   They
evaluated  approximately  170 parameters to determine toxicities and, as
shown  in Table  4.6,  found  good predictive value by both animals for most
organ  systems.  The dog  was generally a better predictor for the occur-
rence  of a toxic  effect  in man, while the monkey was  better at predicting

-------
                                  110
 Table 4.6.   Predictive abilities  of  the dog, monkey, and combination
   Organ system
TP*2     FP
(%)     (%)   (%)     CO    TP + FN
Number of
compounds
          Dog as a predictor for  organ-specific  toxicity  in man
 Injection site      16      36     40        8        33
 Integument          12      32     40       16        57
 Cardiovascular      28      24     36       12        30
 Respiratory         16      64     16        4        20
 Bone marrow         80      12      0        8        9
 Lymphoid             4      72     24        0        0
 Gastrointestinal    92       8      0        0        0
 Liver               52      44      4        0        0
 Renal               32      56      4        8        20
 Neuromuscular       24      60     12        4        14
                                           25
                                           25
                                           25
                                           25
                                           25
                                           25
                                           25
                                           25
                                           25
                                           25
        Monkey as  a predictor for organ-specific  toxicity  in man

 Injection site      13      26     52        9       40          23
 Integument          13      17     57       13       50          23
 Cardiovascular      22      26     30       22       50          23
 Respiratory          13      48     30        9       40          23
 Bone  marrow          83      13      0        4         5          23
 Lymphoid              0      31     65        4       100          23
 Gastrointestinal     74       9      0       17       19          23
 Liver               52      35     13        0         0          23
 Renal               35      48     13        4       11          23
 Neuromuscular       22      30     39        9       28          23

        The  combination of  dog  and monkey  as  a predictor  for
                    organ-specific toxicity in man
Injection site
Integument
Cardiovascular
Respiratory
Bone marrow
Lymphoid
Gastrointestinal
Liver
Renal
Neuromuscular
16
24
36
16
88
4
92
52
36
24
36
36
32
76
12
76
8
48
56
60
40
36
28
4
0
20
0
0
4
12
8
4
4
4
0
0
0
0
4
4
33
14
10
20
0
0
0
0
10
14
25
25
25
25
25
25
25
25
25
25
      True positive,  toxicity was  observed  in both  the  animals  and
in man.
      False positive,  toxicity was observed in  the  animals  but  not
in man.
      True negative,  no  toxicity was  observed in  the  animals  and man.

     HFalse negative,  toxicity was not observed in  the  animals  but
was recorded in man.
     ^Corrected false  negatives —  an  index  of false negative  predic-
tion which analyzes for  only those compounds which  produced the
specific toxicity in man.
     Source:  Adapted  from Schein  et  al., 1970.   Reprinted  with
permission of the publisher.

-------
                                  Ill
the absence of a toxic effect in man.  This  complementary  pattern is
demonstrated by the correct prediction of toxicity in nine out  of the
ten organ systems by the combination of data from the dog  and monkey
studies.  However, this predictive ability was achieved only with a high
percentage of false positives.
     Worth et al. (1970) tested the antibiotic A204 by oral exposure  for
90 days in the rat (0.06, 0.025, 1.0 mg/kg per day) and dog (0.5, 0.8,
1.25 mg/kg per day).  They found no significant species differences
measured by tests of hematology, clinical chemistry, or histopathology.
However, the mortality rate was slightly greater for the rat, particularly
at the intermediate dose, and so was the depression of the growth rate.
Therefore, in this study, the rat appeared to be more sensitive than the
dog.
     Vogin et al. (1970) used the rat  (T-DKL) and the dog (beagle) to
study azotrek, a  tetracycline phosphate complex-sulfamethizole formula-
tion, in tests for oral  toxicity with dosing for up to 33 weeks.  The
rat showed no effects at any dose level  (125, 250, 500 mg/kg per day).
However, the dog  could not tolerate  the high dose, exhibiting decreases
in hematocrit and hemoglobin levels, increased thyroid weight, and one
fatality.  The dog appeared to  be the most  sensitive  species in  this
study.
     Lyon  et al.  (1970)  tested  acrolein  for inhalation  toxicity  in the
rat, dog,  guinea  pig, and monkey using repeated  exposure  studies (8  h
per day, 5 days  per week,  for 6 weeks, with concentration levels of
0.7 and 3.7 ppm)  and  continuous exposure studies (24  h  per  day for
90 days, with  concentrations  of 0.22,  1.0,  and 1.8 ppm).   In the repeated
exposure test,  the  only effect  at  the  0.7-ppm concentration was  a chronic
inflammation  in the lungs  of  the dog and monkey.  At the  3.7-ppm level,
the effects included  a  significant decrease in weight gain  for the rat,
mortality  in the  monkey, and nonspecific inflammatory changes  in the
lung, liver, and  kidney  (most severe in  the dog  and monkey).   The dog
also developed bronchopneumonia.   In the continuous exposure test at
0.22 ppm,  the dog and monkey  showed  specific inflammatory changes in the
lung, while the  monkey,  guinea  pig,  and  dog had nonspecific inflammation
in the  liver, kidney, and  heart.   At 1.0 ppm,  the dog and monkey showed

-------
                                    112
 ocular and nasal irritation and histopathologic damage to the trachea.
 Also, the rat suffered growth depression.  At 1.8 ppm, the irritation
 and tracheal damage were again noticed only in the dog and monkey.   The
 rat again showed a decrease in weight gain, and all species showed
 nonspecific inflammation in the lung, liver, and kidney.   The authors
 concluded that the dog and monkey were the most sensitive species.
      A comparison of the miniature pig and the dog (beagle) was made by
 Earl et al. (1971) using subchronic tests of 1 to 148 days for the  dog
 and 1 to 69 days for the pig.  The exposure levels were 5, 10, 25,  50,
 100, 300, and 500 mg/kg per day for the pig and 5, 10, 25, 50, 100, 300,
 400, and 500 mg/kg per day for the dog.  Both species suffered mortality,
 with the pig first affected at 5-mg/kg levels while the dog was not
 affected until the 25-mg/kg level.  Neither species showed a change in
 hematological values, but the dog did exhibit erratic increases at  the
 high dose level in alkaline phosphatase, serum glutamic oxaloacetic
 transaminase, lactate dehydrogenase, ornithine carbamyl transferase, and
 amylase values.  Histopathological examination revealed lesions of  the
 small intestine in the dog, and the pig had hemorrhaging of the heart
 and fat deposits in the pancreas, in addition to the intestinal lesions.
 These results indicated that the two species reacted comparably to  the
 pesticide diazon.  The dog showed greater biochemical sensitivity;
 the pig was more sensitive to lower doses and showed more lesion damage.
      Knapp, Busey, and Kundzins (1971) compared the sensitivity of  the
 rat and dog to monochlorobenzene (MCB) for 93 or 99 days of oral dosing.
 The dog was more sensitive to MCB, showing mortality, increases in  the
 activity of alkaline phosphatase and serum glutamic pyruvic transaminase,
 increased numbers of immature leucocytes, and gross or microscopic
 pathology for  the liver,  kidney,  gastrointestinal mucosa,  and hematopoietic
 tissues.   The majority  of the effects were at the high dose (272.5  mg/kg
 per day).   The  rat exhibited  some growth depression and increased liver
 and kidney  weights at the high (250  mg/kg per day)  and intermediate
 (50.0 mg/kg per day) doses.   The  effects found in the rat  were not  as
 severe as  those found in  the  dog.
     Jones, Strickland, and Siegel  (1972)  used the  rat (Sprague-Dawley),
guinea pig  (Hartley), monkey  (squirrel),  and dog  (beagle)  to test the

-------
                                  113
inhalation toxicity of propylene glycol 1,2-dinitrate  for  90  days with
daily exposure levels of 67, 108, and 236 mg/m3-   The  effects detected
were similar among all four species  and included  liver pathology and
changes in hematological-biochemical parameters.   The  appearance of
fatty deposits in the liver first occurred at the low  dose for the
guinea pig and rat, but not until the intermediate-dose level for  the
dog and monkey.  Increased methemoglobin levels were found at the high
dose for all species but were more severe for the dog  and  monkey.   The
dog also showed decreased hemoglobin and hematocrit levels at the high
dose.  The monkey had decreased alkaline phosphatase and increased  blood
urea nitrogen levels at the intermediate and high doses, and some
mortality at the high dose.  The dog and monkey appeared to be the more
sensitive species for effects in hematological and biochemical parameters,
and the rat was more sensitive for liver and kidney changes.
     Verschuuren, Kroes, and Tonkelaar  (1973) compared variability in
species response to oral doses of an acaricide, tetrasul, over a 90-day
test period using diet  levels of 50, 200, 1000, and 3000 ppm.  Growth
retardation was evident for all  species  at the high dose, and for all
species except the rat  and mouse at  the  higher intermediate  dose.   In-
creased liver weights were  found at  the  lower intermediate dose for  all
species except the rat, and for  all  species  at higher doses.  Histopatho-
logical damage of  the liver was  found  in all species  at and  above  the
lowest dose.   In conclusion, Verschuuren and his  associates  ranked the
six species based  on nine  criteria with a rating  of 1 to  6  for  each
criteria.  Based on  this system,  five  species were in a range of ±2.5
points, with  the mouse  separated from this  group  by +10 points.  In the
narrower context of the evaluation of  chlorinated hydrocarbons, the
sensitivity ranks were  as  follows:   the miniature pig and rat were the
most sensitive; the rabbit, guinea pig,  and chicken were  next;  and the
mouse was  the least  sensitive.
     King, Shefner,  and Bates  (1973) studied the effects  of chlorinated
dibenzodioxins in  the rat  and  mouse  after oral dosing in the diet  for
42 weeks.  As shown  in Table  4.7,  the rat generally suffered higher
mortality  rates and  more  liver or lung damage than the mouse.  Using
these  criteria,  the  rat was a more sensitive species than the mouse.

-------
                 Table 4.7.  Summary of dioxin oral administration data
Rat (35 per group)
Compound
Controls

1% Dioxane

0.5% Dioxane

1% Unsubstituted
dibenzodloxin
0.5% Unsubstituted
dibenzodioxin
1% Dichloro-
dibenzodioxin
0.5% Dichloro-
dibenzodioxin
1% Octachloro-
dibenzodioxin
0.5% Octachloro-
dibenzodioxin
0.25% Octachloro-
dibenzodioxin
0.125% Octachloro-
dibenzodioxin
Sex
M
F —
M
F
M
F
M
F
M
F
M
F
M
F
M
F
M
F
M
F
M
F
Week
of
test
34
- 34
42
42
42
42
42
42
42
42
17
17
17
17
32
22
37
25
17
17

17
Number of
Survivors
35
35
24
20
26
32
33
32
31
35
35
35
35
35
0
0
0
0
28
15

30
Significant
pathology
Lung


8/11
3/5
6/6
3/3
2/2
1/2
3/4





1/5
°/5«-
1/5*

1/2
0/3


Liver


1/11
2/5
1/6
1/3
2/2
2/2
1/4





5/5
5/5
5/5

2/2
3/3


Mouse (50 per group)
Week
of
test
31
31
40
43
40
43
39
39
34
39
17
17
17
17
10
37
8
37
17
15
9

Number of
Survivors
50
50
50
49
49
49
48
29
50
49
49
48
50
49
0
5
0
45
1
0
0

Significant
pathology
Lung







3/14


0/1
1/2


0/5
0/5
0/5


2/2


Liver







7/14


0/1
1/2


5/5
6/6
5/5


1/2


Source:  Adapted from King, Shefner, and Bates, 1973.

-------
                                  115
     Harris et al. (1973) compared the guinea pig (Hartley),  rat  (C-D),
and mouse (albino CD-I) for sensitivity to 2,3,7,8-tetrachlorodibenzo-
p-dioxin (TCDD).  They exposed each species to TCDD in the diet for 30
consecutive days and/or once a week for 4 to 8 weeks.   As shown in Table
4.8 the guinea pig appeared to be the most sensitive species, followed
by the rat.
     Kast et al.  (1975a) administered fentoterol-HBr to the mouse (ICR-
JCL) and rat (Sprague Dawley-JCL) by gavage daily for 30 days. At the
highest dose (1500 rag/kg), the rat showed growth depression; a high mor-
tality rate; increased weight of the salivary glands,  heart, and  liver;
and histopathological effects in the heart and salivary glands.  At the
highest dose (150 mg/kg), the mouse showed all the above effects  except
growth depression, liver weight increase, and heart histopathology.
However, the mice did exhibit histopathological damage to the liver.
Both species showed dose-dependent changes in hematology and biochemical
factors.  For  the rat, this included increased blood urea nitrogen
level, glutamic oxaloacetic transaminase activity, and glutamic pyruvic
transaminase activity and decreased platelet and glucose levels.   In the
mouse, hemoglobin, red blood cells, and packed cell volume all increased
in a dose-dependent manner.  The  rat appeared to be slightly more sensi-
tive to fentoterol-HBr  than the mouse.
     In another study by Kast et  al.  (1975Z?) , the  toxicity  of  fominoben-
HC1 was studied by dosing the mouse  (ICR-JCL) and  the  rat  (Sprague
Dawley-JCL) for one month by gavage.   The  daily  exposure levels were
250, 500,  1000, and 2000 mg/kg  for the rat and 500, 10DO,  2000,  and
4000 mg/kg for the mouse.  Mortality occurred only at  the high dose for
both species.  Pathological findings were  limited  to a significant  dose-
dependent  increase in  liver weight.  At  the  highest dose,  this increase
occurred with  a fatty  degeneration in  the  mouse  liver  and with a  focal
necrosis followed by  a widening of interstitial  connective tissue in the
rat liver.  An increase of  enzyme activity paralleled the lesion forma-
tions.  In the rat, the enzymes and biochemical  parameters affected
included bilirubin, cholesterol,  alkaline phosphatase, glutamic  oxalo-
acetic  transaminase,  and glutamic pyruvic transaminase.   The parameters
affected in the mouse were not listed.

-------
                    Table 4.8.   Summary of biological effects  of  TCDDa
Frequency of dose
and criteria
affected
Lethal dose
Single
Weekly
Daily
Body weight
Lowest dose effect
Single
Weekly
Daily
No effect
Single
Weekly
Daily
Thymus weight
Lowest dose effect
Single
Weekly
Daily
Rat
Dose MTD
(yg/kg) (days)

100 18
4 x 25 28
10 22


25
6x5
30 x 1

5
6x1
30 x o.l


5
6x5
30 x 0.1
Guinea pig Mouse
Mortality0 , D°*% ^ \ Mortality0 *»* ,
(yg/kg) (days) J (yg/kg)

6/14 3 18 9/10 >50
2/10 5xl 28 10/10 >4 x 25
15/16


1
8 x 0.2 4 x 25


50
8 x 0.04 4x5



10
8 x 0.04 4x5

a2,3,7,8-Tetrachlorodibenzo-p-dioxin.
 Mean time to death after first exposure.
CNumber of animals dying per number of animals treated.
Source:  Adapted from Harris et al., 1973.

-------
                                   117
     Villeneuve and Newsome (1975)  tested hexachlorobenzene  (HCB)  in the
rat (Wistar) and guinea pig by administering a daily peroral dose  of
500 mg/kg body weight for 16 days.   Both species  showed high mortality,
but the guinea pig was affected most.   The rat showed high tissue  con-
centrations indicating a greater resistance to HCB.   Both species
experienced weight loss before death.   No histopathology, hematology, or
biochemical tests were performed.  On the basis of a greater mortality
rate and lower tissue concentrations,  the guinea pig appeared to be the
more sensitive species to HCB.
     The rat (Charles-River) and dog (beagle) were used to test TR2379,
an antihypertensive agent, for peroral toxicity over a 13-week period
(Hartnagel et al., 1975).  For the rat the dose levels were 50, 200, and
820 mg/kg per day, and the dog was dosed at 30, 70, and 160 mg/kg per
day.  Growth retardation occurred for male rats at the intermediate and
high doses and for female dogs at the high dose.  Increased kidney,
adrenal, and thyroid weights were recorded for male rats at the high
dose, and while both sexes had increased liver and heart weights.    In
the dog only males had organ weight increases, and these were limited to
the liver and kidney at all doses.  Both species suffered mortality at
the high dose.  The sensitivity  of the two species was about equal,
with male dogs showing organ weight increases  at low  doses while male
rats showed more  growth depression at lower  doses.
     Koeferl et al.  (1976) reported the  toxicity of  cyclohexanone  to the
rat, dog, and monkey after intravenous  injection of  142  and 284 mg/kg
per day 5 days per week for 21 days.  The  rat  exhibited  the highest
mortality rate at an additional  high  dose  (568 mg/kg).   However,  the dog
showed erythroid  hyperplasia with  increased  hematocrit counts,  a  myeloid-
erythroid ratio below  unity,  and nucleated red blood cells  in  the
peripheral  blood.  This was termed the  "most significant" finding,
indicating  the authors'  opinion concerning the value of the dog in this
toxicity evaluation.
      In a  study  of the antibiotic  sisomicin, Robbins and Tettenborn
 (1976) tested  intramuscular toxicity in the rat (Wistar and Sprague-
Dawley) and dog  (beagle)  for  3 to  5 weeks and 13 weeks.  For the shorter
duration  the daily dose levels were 2 to 100 mg/kg for the rat and 1 to

-------
                                    118
 60  mg/kg for  the  dog.   During  the  longer duration, the daily levels were
 2,  4,  and 8 mg/kg (rat)  and  2  to 16 mg/kg  (dog).  At the higher doses,
 the rat  experienced a  33% mortality rate,  glycosuria, reduced body
 weight,  increased blood urea nitrogen  and  serum creatinine levels, and
 renal  histopathological effects in the shorter test.  The 13-week exposure
 showed a dose-dependent response for renal damage.  In the 5-week dog
 studies, renal damage  and mortality were the primary effects, and this
 trend  continued for the 13-week exposure.   The renal pathology was again
 dose dependent and occurred  to a greater degree at lower doses in the
 dog than in the rat.   The wider response spectrum shown by the rat
 offsets  the greater sensitivity at low doses shown by the dog, so neither
 species  showed a  definite advantage in this study.
     In  a study of drug metabolism, Litterst et al.  (1976) compared the
 monkey (squirrel), rat (Sprague-Dawley), miniature pig  (Hanford), and
 common tree shrew (Tupaia glis) as replacements for the rhesus monkey.
 Each species  was  evaluated for its enzymatic activity pattern by an
 examination of 14 parameters of drug metabolism in hepatic microsomal
 and soluble fractions, including liver weight to  body weight ratio,
 cytochrome P-450, glutathione  s-aryltransferase activity, reduced
 nicotinamide-adenine dinucleotide  phosphate-cytochrome C reductase
 activity,  mixed-function oxidation, aminopyrine tf-demethylase activity,
 ethylmorphine ff-demethylase  activity,  biphenyl hydroxylase activity,
 aniline  hydroxylase activity,  aryl hydrocarbon hydroxylase activity,
 UDP-glucuronyItransferase activity using both p-nitrophenol and
 0-aminophenol acceptor substrates, and tf-acetyltransferase activity
 using  both p-aminobenzoic and  sulfadiazine acceptor substrates.  All
 four species  demonstrated activity in  13 of the 14 parameters, but no
 one species was similar to the rhesus  in all quantitative aspects.
 Based  on an arbitrary  scale  of favorable response, the similarity to the
 rhesus is  as  follows:   the miniature pig is closest, followed by the
 rat, the squirrel monkey, and  the  tree shrew.  In conclusion, the
 authors  felt  that the  miniature pig would  be suitable if housing and
handling problems could  be solved  and  that in any case a primate replace-
ment for the  rhesus might not  be the best  choice.

-------
                                  119
     Sulfolane, a manufacturing solvent,  was  tested  for  inhalation
toxicity by repeated (8 h per day,  5 days per week,  for  27  exposures)
and continuous (23 h per day for 85 to 110 days)  exposures  of the rat
(Sprague-Dawley), guinea pig (Hartley), monkey  (squirrel),  and dog
(beagle) (Anderson et al., 1977).  In the repeated exposure test a dose
concentration of 495 mg/m3 was used, and  in the continuous  exposure test
the levels were 2.8, 4.0, 20, 159,  and 200 mg/m3-  All animals except
the monkey survived the repeated-dose test.  Of nine monkeys exposed to
the high dose, three died and five were sacrificed in a moribund state
after 17 days.  All had pale livers and hearts, and five showed fatty
metamorphosis of the liver.  No significant hematological,  body weight,
or biochemical changes were noted in the other species.   All species
showed chronic lung inflammation, with the rat also showing liver
inflammation.  In the continuous study, the monkey again exhibited high
mortality rates at the high dose level.  The dog, which was exposed to
the high dose, exhibited highly aggressive behavior, seizures and
pneumonia.  No body weight changes were noted in the rat or the  guinea
pig although  the histopathology  examination revealed liver damage.  The
guinea pig also  showed  some  leukopenia and increased plasma transaminase
activity at the high dose.  All  species again showed chronically in-
flamed and hemorrhagic  lung  tissue  at  the  two higher doses.   In their
discussion of the results, the authors rated the susceptibility of the
species to sulfolane in a monkey-dog-rat  progression.
     Cavender et al. (1977)  compared  the  sensitivity of the  rat (male
Charles River) and  guinea pig  (female  Hartley) to sulfuric  acid mist
(5.0 and 10.0 mg/m3),  ozone  (1.0 and  2.0  ppm), and  a  combination of
both.  Both species were exposed via  inhalation  for 2 and  7 days.  The
guinea pig, which proved to  be more sensitive  to sulfuric  acid mist than
the rat, exhibited  lung damage at  low doses  and  mortality  at higher
doses.  With  ozone, both species showed  lung damage and increased liver
weight  to  body weight  ratios,  but  the rat adapted to this  stress more
quickly  than  did the  guinea pig.  In conclusion, the authors pointed out
that  the differences  in sensitivity between the rat and the guinea pig
illustrate the importance of using "at least two species in inhalation
toxicity studies."

-------
                                     120
       In a test of the  inhalation toxicity of acrolein,  a  cigarette smoke
 component, Feron et al.  (1978) exposed  the rat (Wistar),  rabbit (Dutch),
 and hamster (Syrian Golden) to the chemical for 6 h per day,  5 days per
 week,  for 13 weeks.  As  shown in Table  4.9, all three species were
 affected.  The rat was the most sensitive.  It suffered mortality and
 showed toxic effects at  the lowest dose,  unlike the hamster and rabbit.
       One aspect of species selection  that has not been  considered by
 other reports is the opposition of the  general public to  the use of some
 nonrodent species as test animals because of their status as domestic
 pets.   As a result of  such a consideration, Cummings et al. (1979)
     Table 4.9.  Summary of treatment-related effects  in hamsters, rats, and rabbits
                  repeatedly exposed to acrolein (ppm) for 13 weeks
Criteria affected
Symptomatology
Mortality
Growth
Food intake
Hematology
Urinary amorphous
material
Urinary crystals
Organ weights
Lungs
Heart
Kidneys
Adrenals
Gross pathology
Lungs
Histopathology
Nasal cavity
Larynx
Trachea
Bronchi and lungs
Hamster
0.4
Oa
0
0
NE*
0

0
0

0
0
0
0

0

0
0
0
0
1.4
X
0
0
NE
0

0
0

0
0
0
0

0

X
0
0
0
4.9
XXX
0
**
NE
X

+
*

-H-
+
+
0

0

XXX
X
XX
0
0.4
0
0
*
0
0

0
0

0
0
0
0

0

X
0
0
0
Rat
1.4 4.9
X XX
0 +f+
** ***
* **
0 0

0 +
0 *

0 4+
0 +
0 +
0 +++

0 x

XX XXX
0 xx
0 xxx
0 xxx

0.4
0
0
0
0
0

0
0

0
0
0
0

0

0
NE
0
0
Rabbit
1.4
X
0
*
*
0

0
0

0
0
0
0

0

0
NE
0
0

4.9
xxx
0
**
**
0

+
0

-H-
0
0
0

0

XX
NE
X
XX
fi 	 u^t- •*ffe*r*+-t*A' v — C14»Vi#-1w **ffaf>t-aA' w — MnA&Trat-olv aff&rt'aA* vw —
      \J   HUk OL ± ^\- WCU 9 A   *J ^ JL.*jlt ^ ±.J Bt^»-ww b w-w y  r*n  * •vHb*»tab^y V»~W*.WM j «»»•>
severely affected; * — Slightly decreased; ** - Moderately decreased; *** — Markedly
decreased; + — Slightly increased; -H- — Moderately increased; +++ — Markedly
increased.
      Not examined.
     Source:  Adapted from Feron et al., 1978.   Reprinted with permission of the
publisher.

-------
                                  121
evaluated the rat as a replacement for "pet"  species  (the  dog,  cat,
etc.)-  They specifically tested some physiological and behavioral
techniques, primarily used in larger animals, and evaluated their poten-
tial as toxicity indicators in the rat.  Included in  this  evaluation
were techniques for detecting changes in blood pressure, heart  rate,
electrocardiogram (EGG) patterns, respiratory measurements, treadmill
performance, various reflex and body regulatory mechanisms, body tem-
perature, and behavior (retaining learned behavior, learning new behaviors,
and maintaining unconditioned behaviors).  Changes in these physiological
and behavioral traits can indicate toxic effects in the respiratory,
cardiovascular, neuromuscular, central nervous, and autonomic nervous
systems.  The techniques were all performed using restrained but
unanesthesized rats to avoid interfering effects from the  anesthetic.
The results indicated that the rat could be analyzed  for toxic  effects
using these techniques and would be a suitable replacement for  larger
test species.

4.2.4  Conclusions
     In evaluating the suitability of species for toxicity tests, one
constantly finds conflicting data.  Depending upon the chemical used,
the rat, dog, monkey, and guinea pig have each been selected in various
studies as the most sensitive species.  This trend indicates why  there
is such reluctance to change from the standard dog-rat  combination.   In
papers discussing the value of rodents  (mouse, guinea pig, rabbit,  rat,
and hamster), the guinea pig and rat usually prove to be  the most
sensitive.  The studies by Kast  et al.  (1975a, 1975b),  Benitz,  Roberts,
and Yusa  (1967), and King, Shefner, and Bates  (1973)  eliminate  the  mouse
as a potential test species for  subchronic toxicity  tests, while studies
by Stokinger et al.  (1950) and Feron  et al.  (1978) eliminate the other
rodent species.  In direct comparisons of  the  guinea pig  and rat,
evidence  can be found  to support either species  as the most  sensitive.
In their  studies, Jones, Strickland,  and Siegel (1972)  and Roe (1968)
ranked the rat as more sensitive than the guinea pig, but Harris et al.
(1973) and Villeneuve  and Newsome (1975) found just  the opposite.

-------
                                    122
 Additionally, Rowe et al. (1952) and Cavander et al.  (1977)  all con-
 sidered the sensitivity of both species to be equal.   The selection of  a
 preferred species in this case may have to be based on economic factors
 or on individual species response to certain classes of chemicals.
      The value of including a nonrodent species in the test  protocols is
 also a source of considerable contradiction and disagreement.   Some
 authors feel that the use of the dog-rat-monkey combination  should  be
 encouraged (Anderson et al., 1977; Freireich et al.,  1966; Guarino, 1979;
 Hayes, 1967&; Hodge, Boyce,  Diechmanne, and Kraybill, 1967;  Jones,
 Strickland, and Siegel, 1972; McNerney and MacEwen, 1965; Owens, 1962;
 Zbinden, 1963) at least for some chemicals.  Others feel that  the rat
 and dog are sufficient to detect most toxic effects (Balazs, 1976;
 Hartnagel et  al.,  1975;  Kohn, Kay, and  Calandra,  1965;  Robbins  and
 Tettenborn, 1976).   If no information on the metabolism and  pharmaco-
 kinetics of the chemical in  question is available,  the literature cited
 would support the recommendation that the studies be  done using the dog
 and the rat.   However, as discussed in  Sect. 4.2.2, if data  on  metabolism
 in animals are available that can be related to man,  this information
 should be of  paramount importance in choice of species.   Before embarking
 on lengthy, expensive animal studies, efforts should  be made to ensure
 that the animals chosen for  the studies are not unique in the way they
 metabolize the test  chemical.  The same effort should be made  in regard
 to the physiological functional pattern of the test species, since
 differences in this  area can be just as important as  metabolic  differences.
 In direct comparisons, the dog is usually more sensitive than other
 nonrodent species, such as the monkey or cat, although Smith (1979) did
 find the monkey to be metabolically more similar to man than was the
 dog.   The dog has also been  selected as the most sensitive species  in
 more studies  (Ansbacher,  Corwin,  and Thomas, 1942;  Hagan et  al., 1967;
 Knapp,  Busey,  and Kundzins,  1971;  Koeferl et al., 1976;  Litchfield,
 1961;  Newberne,  Gibson,  and  Newberne, 1967; Stokinger et al.,  1950;
 Stokinger  and Stroud,  1951;  Vogin et al., 1970; Yeary, Braham,  and  Miller,
1965)  than the  rat (Atkinson et al.,  1966;  Verschuuren,  Kroes,  and
Tonkelaar, 1973; Weil  and McCollister,  1963; Worth et al., 1970).  The
review by Aviado  (1978)  suggested  that  the use of both the dog  and  rat

-------
                                   123
is superfluous and that the rat alone may be sufficient.   This  trend is
also supported by the work of Cummings et al.  (1979)  in which physio-
logical and behavioral assessment techniques,  usually restricted to use
with larger animals, were found to be applicable in rat studies.  To
further complicate the issue, Earl et al. (1964, 1971) gave good evidence
supporting the use of the miniature pig in certain instances.  When the
scientific data are conflicting, economic and species availability
factors should also be considered in the task of species selection.
     It is obvious that, in the ideal situation of maximum detection of
toxic effects, a dog-rat combination should be used with selected incor-
poration of alternative species such as the guinea pig, monkey, or
miniature pig.  The exact combination would depend on the chemical or
effect to be evaluated.  The analysis by Gehring, Rowe, and McCollister
(1973) indicated that cost and time parameters could be very high for a
subchronic test using two species.  Therefore one could infer that the
ideal situation would be to select a single test species to reduce these
costs.  However, if by using two species, such as the dog and rat, a
more efficient determination of toxicity is achieved, then a savings in
both time and cost could be managed by reducing or eliminating  the need
for costlier chronic or lifetime studies.  This aspect of the problem
will be discussed further  in the section on subchronic test  durations.

4.3  DURATION

4.3.1  Introduction
     The duration of  a subchronic  test has  always  been a poorly defined
parameter.  As  discussed  in the introduction,  even the definition of a
subchronic  test, which is  based in large part on duration,  has not
specified a standard exposure duration.   The closest to a standard
exposure duration is 90 days.   Therefore,  for this discussion the 90-day
subchronic  test will be considered the standard duration.  However,
tests  of  shorter durations, especially 28- or 30-day tests, in the liter-
ature  are  also discussed.   Durations of 14, 28, and 90 days are commonly
used and  guidelines indicate that these are acceptable.  The 14-  to 28-
day tests  are generally used to provide information on the nature of  the

-------
                                    124
 toxic effect, and likely "no effect" and "effect" doses.   They do not
 usually provide information on maturation or aging effects.   The 90-day
 duration can provide information on the effects of chemicals on the
 maturation process, in addition to evaluating the nature  of  toxic effects
 at lower prolonged doses.  The purpose of this section is to evaluate
 the effectiveness of 90-day and shorter duration tests by presenting
 duration reviews from the literature and summarizing data from duration
 comparison studies.

 4.3.2  Duration Reviews
      One of the first discussions  of the length of exposure  necessary
 for a repeated-dose study was  that of Smith  (1950).   As a result  of  the
 need for antimalarial drugs during World War II,  a short  test  of  11  to
 14  days was designed (Wiselogle, 1946)  to rapidly evaluate the toxicity
 of  these drugs.   Smith later evaluated the effectiveness  of  this  test,
 including increasing the duration  to 21 days,  by comparing growth rate
 curves and particularly  the size of the standard deviations.   He  assigned
 an  arbitrary value of 100% for the sensitivity of the method at 21 days
 and calculated  what percent of this sensitivity was achieved at shorter
 durations.   The results  were 15% to 30% sensitivity at 7  days,  68% to
 85% sensitivity at 11 days, and 75% to 95% sensitivity in 14 days.   He
 concluded that  for absolute toxicity evaluations the gain was  significant
 when the test was  increased from 11 to 14 days but not when  increased to
 21  days.   Thus,  Smith recommended  a subchronic test duration of 14 days.
      In a discussion of  the relationship between short- and  long-term
 oral toxicity studies, Weil and McCollister  (1963) evaluated the  effec-
 tiveness of a 90-day diet test.  Their evaluation was based  on 33 chemicals
 using  the criteria of body weight  gain,  relative weight changes of the
 liver  and kidney,  and liver and kidney pathology studies  in  the rat.
They compared both the minimum effect and maximum no-effect  levels for
short-term  (approximately 90 days)  and  long-term (2-year)  studies,
arriving  at  a 90-day:2-year ratio  for each level (Table 4.10).   These
ratios indicated the degree to which these values changed with increased
test duration.  In  95% of the  studies the ratio was 6.0 or less,  meaning
that the  safe levels  found  for  a chemical tested for 90 days could be

-------
                                       125
            Table 4.10.  Relationship of dosage levels of short-term and 2-year
                       feeding of materials in the diet of rats
Percentage of materials in diet
Material
number
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
25
26
27
28
29
30
31
32
33
Duration
of short-
term test
105
90
90
120
90
90
97
90
130
30
30
90
90
90
130
50
98
90
29
210
90
130
90
90
30
90
90
90
93
91
90
90
142
Short-term
Minimum
effect
0.015
4.0
1.0
3.0
0.25
0.01
0.1
8.0
1.0
0.05
25.0
0.75
10.0
0.03
3.0
0.3
0.01
16.0
0.25
0.25
0.225
0.1
0.5
0.009
0.3
8.0
16.0
M°
M*
M*
M*
M*
M*
Maximum
no-effect
0.005
2.0
0.3
1.0
0.0625
0.003
0.03
4.0
0.3
0.012
10.0
0.375
3.0
0.01
1.0
0.1
0.003
8.0
0.06
0.05
0.15
0.03
0.25
0.003
0.1
4.0
8.0
3.0
5.0
0.18
1.0
2.5
25.0
2-years
Minimum
effect
0.03
8.0
2.0
5.0
0.256
0.01
0.1
8.0
1.0
0.04
20.0
0.40
5.0
0.0125
1.0
0.1
0.003
4.0
0.06
0.05
0.04
0.005
M°
M*
Vf1
M3
M3
3.0
5.0
0.06
MO
M*
M*
Maximum
no-effect
0.01
4.0
0.2
0.5
0.064
0.003
0.03
4.0
0.2
0.01
5.0
0.13
1.0
0.0062
0.2
0.03
0.001
2.0
0.02
0.01
0.02
0.0025
0.5
0.004
0.1
2.0
4.0
1.0
1.0
0.02
0.3
0.5
5.0
Ratio of short-term
to 2-year test
Minimum
effect
0.5
0.5
0.5
0.6
1.0
1.0
1.0
1.0
1.0
1.2
1.2
1.9
2.0
2.4
3.0
3.0
3.3
4.0
4.2
5.0
5.6
20.0











Maximum
no-effect
0.5
0.5
1.5
2.0
1.0
1.0
1.0
1.0
1.5
1.2
2.0
2.9
3.0
1.6
5.0
3.3
3.0
4.0
3.0
5.0
7.5
12.0
0.5*
0.8*
1.0*
2. Of
2.0fc
3.0C
5.0e
9.0*
3.3^
5.0^
5.0d
      Indicates that the maximum no-effect level was the highest dosage level fed.
      As the M level was on the 2-year test, the ratios are a maximum.
      As the M level was on the short-term test, the ratios are a minimum.
      As the M levels were on both the short-term and 2-year tests, the ratios are indicative
 only of which levels were used.
     Source:  Adapted from Weil and McCollister, 1963.  Reprinted with permission of the
 Journal of Agricultural and Pood Chemistry.  Copyright by the American Chemical Society.
used  to predict results for a 2-year study,  with a  significant degree of
accuracy, by dividing the  short-term value by 6.0.   Since  it is  common
to apply a hundredfold decrease, as  a safety factor, to the maximum no-
effect level determined with a 2-year study, the sixfold difference
requiting from the 90-day  study would appear to be  a reasonable  estimate
of chronic effects.   However, Weil and McCollister  did not recommend
that  the 2-year study be abandoned,  stating only  that in  certain cases a

-------
                                   126
 90-day study would be sufficient.  The determination of when the shorter
 study would suffice is left up to the toxicologist based on his
 experience with the chemical and its metabolic pattern.
      In a later paper, Weil et al. (1969) examined the 90-day test in
 the same manner to see if a shorter test could predict the results it
 gives.  Their evaluation included the prediction of 90-day repeated
 exposure results from 7-day repeated exposure and acute LDso data.  They
 tested the oral toxicity of 20 chemicals in the diet of the rat using
 acute LD50, 7-day durations, and 90-day durations and evaluating the same
 criteria used by Weil and McCollister in 1963.  By constructing minimum
 effect level ratios between each duration group (acute, 7-, and 90-day)
 as they had done with 90-day:2-year minimum effect levels, Weil et al.
 (1969) were able to evaluate the predictive relationships (Table 4.11).
 The relationship between the LDso data and the 90-day test was poor,
 needing a ratio of 40 to achieve a predictive accuracy of 95%.  The
 relationship between LDso data and 7-day test data was not much better,
 with a ratio of 20 for 95%.  However, extrapolation from 7- to 90-day
 tests was much better, with a ratio of only 6.2 for 95% of the results.
   Table 4.11.  Parameters of ratios  of  acute peroral LDsoS,  7- or  90-day
              and 2-year minimum effect (MiE) dosage levels
Ratio
Percentile
25th
50th
75th
95th
Semiinterquartile
range
Coefficient of
rank correlation
Formula T „ _ _
. , LDcn to LDso to
or symbol -^ __D"
* 7-day 90-day
MiE MiE
Ql = P2s 1.2
P50 = median 2.3
$3 = P75 6.0
P95 20.0
(#3 - Qi)/2 2.4

P 0.831
2.
10.
25.
40.
11.

0.
3
0
0
0
4

782
7-day
MiE to
90-day
MiE
2
3
5
6
1

0
.2
.0
.2
.2
.5

.943
90-day
MiE to
2-year
MiE
1
1
3
5
1

0
.1
.8
.8
.7
.4

.946
     Source:   Adapted from Weil et al., 1969.  Reprinted with permission
of the publisher.

-------
                                   127
This compares favorably with the extrapolation results of the 2-year
studies done in 1963.  As the authors stated:   "Therefore,  one can
predict 90-day results from a 7-day test with the same confidence as one
can the two-year results from a 90-day test."  The formulas necessary to
predict the minimum effect dose for 50% to 95% of the studies are given
in Table 4.12.  These two studies were among the first to present suf-
ficient significant data indicating that subchronic test durations could
be used to predict chronic effect levels without a substantial loss in
sensitivity.


                   Table 4.12.  Prediction formulas
                            Predicted upper limit to achieve a
                                minimum  effect  level  (MiE)
             Value          	
                             For 90-day          For 2-year
                            feeding study        feeding study
       Median              7-day MiE/3.0a    90-day MiE/1.8fl  or
                                                7-day MiE/5.4
       95th percentile     7-day MiE/6.2     90-day MiE/5.7 or
                                                7-day MiE/35.3
             aThe denominators  for  the 7-day MiE/2-year MiE
       relationships were obtained by multiplying those for  7
       and 90 days [e.g.,. 5.4  = 3.0 (1.8)].
             Source:   Adapted from  Weil et  al.,  1969.   Reprinted
       with permission of the  publisher.
     Peck  (1968) discussed the value of shorter tests in drug evaluation
studies.   He cited  a World Health Organization Technical Report  (1966)
and Davey  (1964) as recommending that  a 3-  or 6-month test  is adequate
to detect  long-term toxicity.  This 3- to 6-month  duration  is also
recommended by  Boyd (1968),  Bein  (1963), Barnes and Denz  (1954), and
Zbinden  (1963).  Peck  gave data from  studies done  from  the  previous
15 years to support the use  of a duration of 3 months.  As  shown in
Table  4.13, only one study showed  additional toxicity after 3 months,
and only four  showed added toxicity after  2 months.  This again supports

-------
                                   128

       Table 4.13.  Approximate duration of drug administration
                required to define toxicitya in animals
Compound
Indomethacin
Hydrochlorothiazide
Amiloride HCl
Cyproheptadine
Amitriptyline
Methyl-DOPA
Penicillamine
Ethacrynic acid
Protriptyline HCl
Thiabendazole
Dexamethasone
Duration
(months)
1/2
J>
X
X
X
X
X
X
X
X
X
X
1
X
X
X
X
X
X
X
X
0
X
X
2
Oc
0
0
0
0
X
X
X
0
X
X
3
0
0
0
0
0
0
X
X
0
X
0
6
0
0
0
0
0
0
0
0
0
0
0
12 18 24
000

0 0

0
xd

0 0
0 0
000

         By physical examination or hematologic, biochemical,  and/or
   anatomical studies.
         x — Toxicity demonstrated.
        ^
         0 — Continuing duration of study, no additional toxicity.
         Additional finding of precipitate in kidneys.   No earlier
   sacrifice, so time of onset not known.  Found in rats but not in
   dogs or monkeys.
        Source:   Adapted from Peck, 1968 — An Appraisal of Drug
   Safety Evaluation in Animals and the Extrapolation of Results to
   Man.  In:   Importance of Fundamental Principles in Drug Evalua-
   tion, D. H. Tedeschi and R. E. Tedeschi, eds.  Raven Press, New
   York.  pp. 450-471.  Reprinted with permission of the publisher.
the idea that shorter tests can provide reasonable indications of long-

term toxicity.
     In contrast to the papers by Weil, Hayes  (1972) stated that although
the 90-day or 70-exposure test is sufficient to determine most long-
term effects, reduction to lesser durations is not adequate.  He recom-
mended this after consideration of some data on chemosterilants.

-------
                                  129

Initially Hayes (19672?) felt that some drugs could be  tested  in  30  expo-

sures, but on reexamination he preferred the 90-day  exposure  test.   Thus,

there is definitely some opposition to reduction of  test  duration.

     McNamara (1976) compiled data similar to Weil and McCollister

(1963) from the literature using 82 studies evaluating 122 compounds

with a variety of test species.  Despite the variability  due  to  differ-

ences in species and experimental design, McNamara's data agree  with

that of Weil and McCollister in recommending that 2-year  studies need

not always be performed.  The results are summarized in Table 4.14  in

which formulas are given designating the safe long-term dose  levels
                  Table 4.14.   Prediction  of  long-term
                            no-effect  doses
                                          Time  in which
                      Dose                repeated  dosage
                                           will produce
                                            no  effect
                LDso/lOO                     3 months

                LD5o/1000                    Lifetime

                3-month no-effect
                  dose/10                    Lifetime

                     Source:  Adapted from McNamara,
                1976.  Reprinted with permission of
                the publisher.
predicted by short-term tests.  As with the other authors, McNamara
qualified this predictive value to exclude carcinogenetic and terato-

genetic effects.   In  conclusion he stated:

          If any  toxic effect  occurs  in 3 months, additional (and
     perhaps more serious)  effects may appear with  continued dosing.
     If no  effect occurs in 3  months,  there  is  a low likelihood that
     any effect will  occur  on  continued dosing  for  1 year.   Thus,
     these  dose relationships  can be  of great value in decreasing
     the time  involved in LT toxicological testing.  Experimental
     evidence  strongly supports  the view  that LT no-effect doses
     can be reliably  predicted from ST studies.

-------
                                   130
 4.3.3  Comparison Summaries
      Gaunt et al. (1965)  studied selected aspects  of the  oral  toxicity
 of butylated hydroxytoluene (BHT) and butylated hydroxyanisole (BHA)  in
 the rat with dose levels  of 0.1% for 16 weeks.   Based on  previous  studies,
 the authors concentrated  on liver effects, although other toxicity
 parameters were monitored.   As shown in Table 4.15, the principal  effects
 were increases in ascorbic acid excretion, liver weight,  and adrenal
 weight.   For BHT, the females showed significant changes  in  ascorbic
 acid excretion and liver  weight by the 4th week, but the  adrenal weight
 effects did not appear at a significant level until the 12th week.  In
 the male rat, a trend of  increasing ascorbic acid  excretion  for BHT was
 noted from the 4th week on, but not always at a significant  level.  The
 same was true for increases in liver and adrenal weights  and in general
 for all parameters of the BHA studies in both sexes.  In  this  study,  the
 12-week duration definitely predicted the longer-term effects  of 16 weeks.
 The 4-week duration did provide good indications of the toxicity trends
 but not always at a significant level.
      In a 90-day study of dibutyl(diethylene glycol bisphthalate)  (DDGB),
 a plasticizer, Hall, Austin, and Fairweather (1966) tested the oral
 toxicity in rats.  As shown in Table 4.16, the effect on  body  weight
 gain was evident by the fourth week when a significant decrease was
 noted for males at the high (2.5% of diet) and intermediate  (1.0%  of
 diet)  doses,  and for females at the high dose.   The only  other toxic
 effect noted was an increase in relative liver, heart, and brain weights
 after 90 days.   The authors felt that these changes were  due to the body
 weight loss since the absolute organ weights did not change.  Therefore,
 in  this  case,  the only toxic effect attributable to DDGB  would have been
 detected by a 4- or 8-week  test.
     Misu et  al.  (1966) tested Sumithion,  an insecticide, for  90 days
 given  in the  diet of the  rat at concentrations of  32, 63, 125, 250, and
 500 ppm.   Toxic effects began to appear at the high dose  during the
 first week of  testing:  the mean body weight was decreased (Fig. 4.1),
 the testes  and  brain weights increased  (Table 4.17), and  the cholinestrase
activity  decreased (Fig.  4.2).   The  growth rate decrease  was most  notice-
able after  the  first  week,  but  began to parallel the control values as

-------
 Table 4.15.   Urinary ascorbic acid excretion and relative liver and adrenal weights of rats fed butylated hydroxyanisole (BHA)
                                or butylated hydroxytoluene (BHT) at 0.1% of the diet for 16 weeks0
Duration
Compound of feeding
Ascorbic acid*
(mg/kg/day)
Male
Arachis oil
BHT
BHA
Arachis oil
BHT
BHA
Arachis oil
BHT
BHA
Arachis oil
BHT
BHA
, control 4
, control 6
, control 12
, control 16
2.20 ±
4.74 ±
1.84 +
4.39 ±
6.90 ±
4.89 ±
2.10 ±
3.41 i
1.87 ±
2.50 ±
3.13 ±
2.29 ±
0.39
0.95
0.24
1.08
1.76
1.56
0.22
0.41
0.37
0.34
0.43
0.24
Female
3.25 ± 0.35
7.83 ± 1.27
4.16 ± 0.38
1.28 i 0.38
3.39 ± 0.51
1.67 ± 0.51
3.12 ± 0.57
4.83 ± 0.63
2.32 ± 0.36
0.98 * 0.14
2.37 ± 0.37
0.92 ± 0.17
aEach result represents the mean ± statistical error for six
Ascorbic acid is the mean of four days' excretion.
°P < 0.05.
dp < o.oi.


Organ
weight Co body weight ratio
Liver
Male
, 4.31
d 4.68
0 4.36
4.13
4.18
4.24
3.73
3.93
3.58
; 3.50
3.73
3.72
animals .
± Q.lf>d
± 0.15
± 0.05
± 0.06
± 0.12
i 0.07
± 0.07
t 0.14
+ 0.10
± 0.08
+ 0.09

Female
3.94
4.45
4.50
3.71
4.00
3.82
3.15
3.51
3.60
3.21
3.63
3.40

± 0.08
± 0.08*;
* 0.12d
+ 0.11,
± 0.09
± 0.12
t 0.10
i 0.08 ,
i 0.06
± 0.10
± o.ioe'
± 0.17

(2)

Adrenal
Male
0.010 i 0.0008
0.013 ± 0.0010°
0.012 ± 0.0007
0.010 ± 0.0005
0.010 ± 0.0004
0.010 ± 0.0011
0.009 i 0.0037
0.010 ± 0.0026
0.011 ± 0.0008
0.009 + 0.0081
0.010 ± 0.0061
0.011 ± 0.0005

Female
0.016
0.018
0.018
0.018
0.015
0.017
0.017
0.022
0.020
0.016
0.019
0.020

± 0.0005
± 0.0008
± 0.0011
± 0.0011
± 0.0009
± 0.0006
± 0.0008^
± 0.0006"
± 0.0009"'
* 0.0014
± 0.0009
± 0.0004G

 P < 0.001.

Source:  Adapted from Gaunt et al.
                                                                                                                                       CO
1965.  Reprinted with permission of the publisher.

-------
                                     132
             Table 4.16.  Mean values of body weights, food consumption, and
             dibutyl(diethylene glycol bisphthalate) (DDGB) intake of rats
                    fed DDGB at 0% to 2.5% of the diet for 11 weeksa
Dietarv Body wei8ht in grams
JeveF at end of week
(%:

0.0
) 0°

96
0.25 100
1.0 91
2.5 98


0.0 82
0.25 82
1.0
2.5
87
83
4

241
226,
2l4d
noe

169
156
160
U5e
8

317
301
280e
232e

205,
185d
196
me
11

352
335
315*
262e

220
199*
211"
190e
Food consumption in
grams per rat per
day at end of week
0

11
11
9
5

9
9
9
5
c

.9
.6
.5
.8

.5
.7
.5
.6
values are the means for groups of 10
DDGB intakes are calculated from data

Day one of
dP < 0.05.
feeding
•



4
Male
23.4
20.5
19.3
16.3
Female
15.7
14.5
14.6
14.2
animals
on body

8

18.7
20.2
17.1
15.5

13.9
13.1
13.5
13.6
weight

11

18.0
19.4
16.7
14.9

14.5
11.3
13.5
11.4
and

DDGB intake" in grams per
kg per day at end of week
0° 4 8


0.29 0.23 0.17
1.04 0.90 0.61
1.48 2.40 1.68

0.30 0.23 0.18
1.09 0.91 0.69
1.67 2.44 1.93
food consumption.

11


0.14
0.53
1.41

0.14
0.64
1.52


     eP < 0.001.
     Source:  Adapted from Hall, Austin, and Fairweather, 1966.  Reprinted with permission
 of the publisher.
 the exposure period progressed.   However, the final body weight  was
 still significantly below  control values.  Testes weight was  significantly
 lower by the llth day and  remained so until the 90th day.  The same
 pattern was found for the  chclinestrase activity in the liver, kidney,
 brain, and red blood cells.   In  each case the initial stress  was the
 greatest, producing the most  effect.  All toxic effects showed recovery
 or  stabilization at the 90th  day.   Some minor effects of the  same type
 as  occurred at the high dose  were noted with a dose of 250 ppm,  and the
no-effect level was 125 ppm.  Thus,  it appears that all of the toxic
 effects could have been detected by an 11- or 30-exposure test,  and
only  the stabilization trend  would have been missed.
      In a study of the oral toxicity of diuron, a herbicide,  Hodge,
Downs,  Fanner, Smith, Maynard, Clayton, and Rhodes (1967) utilized rat
tests  with durations of 1 month,  3 months, or 2 years.  The dose levels
employed were:  200, 400, 2000,  4000,  and 8000 ppm (1 month); 50, 250,
500, 2500,  and 5000 ppm (3 months);  and 25, 125, 250, and 2500 ppm

-------
                                  133
                                          ORNL-DWG 79-43668R
               400 -
               350 ~
               300 -
            U)
            o
            o
            CD
               250 -
               200  ~
                450  -
                400  -
                                I  I   I   I  I   I   I  I   I
             —•—CONTROL
                  500 ppm
                     0  7
24     35    49    63
 EXPERIMENT DAY
      Fig. 4.1.  Changes in  the mean body weight of  rats  fed Sumithion
 in the diet for 90 days.  Source:  Adapted  from Misu  et  al.,  1966.
 Reprinted with permission of  the publisher.
(2 years).  Depression of the growth rate occurred at 1 month  (2000,
4000, and 8000 ppm), at 3 months  (2500 and 5000 ppm), and at 2 years
(2500 ppm).  Mortality was evident at 1 month  (6 of 10 rats at 8000 ppm)
and at 2 years (44 of 70 rats at  2500 ppm), but no mortality was  seen at
3 months.  However, the mortality was affected by disease vectors,
and the authors concluded that  the death  trend in the 2-year  study  was
not due to diuron levels.  The  red blood  cell  counts  were decreased at

-------
    Table A. 17.   Mean testes and brain weights, as percentage of body weight, of control rats and rats  fed  a  dietary level
                                      of 500 ppm of Sumithion for various periods of time
Organ weight after days of feeding indicated
Organ
Left testis
Right testis
Brain stem
Brain cortex
11
Control
(%)
0.41 ± 0.02
0.42 ± 0.02
0.21 ± 0.00
0.58 ± 0.02
500 ppm
00
0.53a ± 0.02
0.52a ± 0.01
0.25* ± 0.01
0.79a ± 0.01
30
Control
(Z)
0.40 ± 0.02
0.40 ± 0.02
0.15 ± 0.04
0.46 i 0.01
500 ppm
(Z)
0.60a ± 0.40
0.59a ± 0.01
0.23a ± 0.01
0.54d ± 0.12
60
Control
(Z)
0.29 ± 0.02
0.29 ± 0.02
0.14 ± 0.01
0.32 ± 0.01
500 ppm
(Z)
0.38fc ± 0.03
0.39^ ± 0.03
0.14d ± 0.01
0.40^ i 0.02
90
Control
(Z)
0.27 ± 0.02
0.26 ± 0.02
0.13 ± 0.01
0.30 ± 0.01
500 ppm
(Z)
0.35° ± 0.02
0.34° ± 0.03
0.15° ± 0.01
0.35° ± 0.01

t-1
CO
4s-


       differs from controls, P < 0.01.
 Value differs from controls, 0.01 < P < 0.05.
°Value differs from controls, 0.05 < P < 0.5.
'Slot significant, P > 0.5.
Source:  Adapted from Misu et al., 1966.  Reprinted with permission of the publisher.

-------
                                                                           ORNL-DWG 79-13665
UJ
CO
<
tr.
UJ
l-
co
UJ
z

-J
o
X
o
CO
UJ
X
o

^-
z
UJ
o
IT
UJ
CL
                                                 -   ~   (OLIVER
    100  -
50  -
      0
n
R
i

         ^CONTROL
                      500ppm
         250 ppm   fl 125 ppm     t 1 63 ppm
         (rfRED BLOOD CELL
    100  -ci
50  -
                                        i
                   -   (
-------
                                   136
 1  month (4000 and 8000 ppm),  at 3 months (2500 and 5000  ppm),  and  at
 2  years (2500 ppm).   The hemoglobin  concentrations were  also depressed
 at 1 month (4000 and 8000 ppm), at 3 months  (5000 ppm),  and at 2 years
 (2500 ppm).   The 1-month study indicated a decrease in hematocrit  that
 was not evident in the longer studies.   Organ weights were also affected.
 After 1 month at 8000 ppm, the liver and kidney weights  were decreased,
 and the spleen weight was increased. At 3 months the spleen weight was
 increased (5000 ppm), but the liver  and kidney weights were unaffected.
 In the 2-year study the spleen weight was increased (2500 ppm) at  9 and
 17 months, but not at 24 months.  Histopathologic data were not signifi-
 cantly different from the controls for  all tests regardless of duration.
 In conclusion, it appears that the major toxic effects were sufficiently
 indicated by the 1-month duration.  Extension to 3 months and  2 years
 provided little additional information.
      Ambrose et al.  (1972) studied 3'-4'-dichloropropionanilide in rats
 after 13 weeks and 2 years of oral exposure.   In the 13-week study, dose
 levels of 100, 330,  1000, 3300, 10 000,  and  50 000 ppm were used,  and
 the 2-year study used levels  of 100, 400, and 1600 ppm.   The significant
 effects recorded after 13 weeks of dosing included growth depression
 (high and intermediate doses), a decrease in hemoglobin  levels (high and
 intermediate doses),  and increases in weight  for the heart (high dose),
 spleen and liver (females at  high and intermediate doses), and testes
 (high dose).   After  the 2-year exposure period,  the effects at the high
 dose included growth rate depression, increased female liver and kidney
 weights,  and increased testes weight.  These  were all detected with the
 13-week study.   The  hemoglobin level was also depressed  at the high dose
 in  the 2-year study,  but, in  contrast to the  13-week study, results
 occurred  only in the  female rats.  Thus, in  this study there was a good
 prediction of chronic (2-year) effects  from  the 13-week  data,  suggesting
 that  the  13-week study would  have  been  sufficient.
      Lawrence et  al.  (1972) studied  epichlorohydrin in the rat by  intra-
peritoneal injection  for duration's of 30 daily injections (with doses*of
0.00955 and 0.01910 mL/kg) or 12 weeks  of three injections per week
 (with doses of 0.00955,  0.01910, and 0.04774  mL/kg).   In the cumulative
toxicity  test  (30 daily exposures),  weight gain was significantly

-------
                                  137
decreased at the high dose by day 15 and at the low dose by day 20
(Table 4.18).  In the 12-week study, the high dose exhibited a signifi-
cant decrease in weight gain until the last week (Table 4.19).  The
hematology and biochemical parameters showed no significant trends in
most tests.  However, hemoglobin levels did show a significant decrease
at the high dose at the end of the cumulative toxicity test and a dose-
dependent decrease after the 12-week study.  The measurement of organ
weights also showed some changes attributed to toxic effects.  In the
30-day study, the kidney weight decreased significantly at both doses
(Table 4.20).  After 12 weeks of dosing at the high dose, the kidney,
liver, and heart all showed significant weight increases, and the brain
showed a significant weight decrease (Table 4.21).  In comparison of
these two experiments, the total number of injection exposures is
similar (30 vs 36), but the duration of exposure is not  (4 weeks vs
12 weeks).  The results from both studies are quite similar, which
suggests either that the shorter duration adequately predicts the results
of the longer duration or that the number of doses given is  the key
factor in a repeated-dose study.

       Table 4.18.   Cumulative toxicity of epichlorohydrin:  body
             weight gain in grams (mean ± statistical error)
Day
5
10
15
20
25
30
Cottonseed
oil control
28.17 ± 1.96
66.00 ± 2.44
111.25 ± 3.39
145.67 ± 3.75
157.25 ± 4.38
183.33 ± 5.59
Epichlorohydrin
0.00955 mL/kg
36.25 ± 5.73
71.92 ± 6.15
105.25 ± 7.24
123.42 ± 4.83a
145.67 ± 6.44
167.67 ± 7.27
0.01910 mL/kg
26.00 ± 1.54
63.67 ± 2.68
91.33 ± 4.33a
131.58 ± 2.87a
153.92 ± 7.33
158.50 ± 6.82fc
           Significantly different from controls at 99%  level
      (P = 0.01) by Student's t test.
            Significantly different from controls at 95%  level
      (P = 0.05) by Student's t test.
           Source:  Adapted from Lawrence et al., 1972.   Repro-
      duced with permission of the copyright owner.

-------
                                   138
           Table 4.19.  Subacute toxicity of epichlorohydrin:  body
                weight gain  in grams (mean ± statistical error)
Week
1
2
3
4
5
6
7
8
9
10
11
12
Cottonseed
oil control
46.58 ± 2.88
76.50 ± 6.15
105.33 ± 5.86
186.17 ± 5.74
214.25 ± 8.14
254.83 ± 6.52
279.25 ± 7.16
296.17 ± 7.10
333.50 ± 9.72
345.50 ± 13.49
356.67 ± 14.75
358.92 ± 13.27

0.0095 mL/kg
38.58 ± 4.50
94.42 ± 3.05*
119.83 ± 3.72*
193.42 ± 4.74
228.08 ± 6.80
254.33 ± 11.07
267.08 ± 9.11
289.92 ± 5.98
330.75 ± 9.59
359.83 ± 9.64
370.42 ± 10.82
383.42 ± 11.22
Epichlorohydrin
0.0190 mL/kg
41.42 ± 2.40
86.50 ± 5.14
111.83 ± 6.79
175.58 ± 11.02
203.50 ± 10.73
242.75 ± 11.44
261.25 ± 12.27
276.00 ± 11.47
315.67 ± 21.92
334.00 ± 13.81
371.17 ± 12.08
380.92 ± 13.34

0.04774 mL/kg
24.00 ± 3.14a
55.09 ± 10.23
97.00 ± 9.27
128.30 ± 11.54a
172.00 ± 11.37*
195.60 ± 10.29a
218.50 ± 11.14a
246.80 ± 12.16a
266.60 ± 13.19a
293.70 ± 14.19*
286.70 ± 21.91*
328.33 ± 18.19
     aSignificantly different from controls at 99% level  (P = 0.01)  by
Student's t  test.
      Significantly different from controls at 95% level  (P = 0.05)  by
Student's t  test.
     Source:  Adapted from Lawrence et al., 1972.  Reproduced with permission
of the copyright owner.
     In  two  companion studies, Kociba et al.  (1976;  1978)  evaluated the
toxicity potential of 2,3,7,8-tetrachlorodibenzo-p-dioxin  (TCDD)  admin-
istered  in the diet of rats for 13 weeks (1.0 and  0.1  yg/kg)  and  2 years
(0.1 and 0.01 yg/kg).| As shown in Table 4.22 the  results  were quite
similar  for  mortality, body weight gain, alkaline  phosphatase, packed
cell volume,  red blood cell count, hemoglobin concentration,  relative
weights  of the liver and thymus, and histopathologic changes  in the
liver, lung,  thymus, uteri, and lymphoid tissues.   Differences between
the two  studies consisted mainly of the more  transient effects indicated
by the subchronic study, with only a few different effects indicated by
the chronic  study.  Although there were minor differences  in  serum
enzyme levels,  hematology values, and relative  organ weight changes, the
subchronic study did evaluate the major effects found  in the  chronic
study.

-------
                                    139

         Table  4.20.   Cumulative toxicity of epichlorohydrin:
             percent  organ weight  to  body weight of rats
                      (mean ± statistical error)
Organ
Adrenals
Brain
Gonads
Heart
Kidneys
Liver
Lungs
Spleen
Cottonseed
oil control
0.014 ± 0.001
0.475 ± 0.017
0.975 ± 0.037
0.273 ± 0.006
0.654 ± 0.023
3.960 ± 0.140
0.381 ± 0.027
0.236 ± 0.010
Epichlorohydrin
0.00955 mL/kg
0.014 ± 0.001
0.567 ± 0.041
0.995 ± 0.051
0.309 ± 0.022
0.750 ± 0.025a
4.150 ± 0.230
0.409 ± 0.028
0.266 ± 0.017
0.01910 mL/kg
0.013 ± 0.002
0.543 ± 0.033
0.971 ± 0.037
0.378 ± 0.078
0.786 ± 0.035a
3.916 ± 0.056
0.407 ± 0.015
0.253 ± 0.012

    (P = 0.05)  by Student's t  test.

         Source:   Adapted from Lawrence et al., 1972.   Reproduced
    with permission of the copyright owner.
      Table 4.21.  Subacute toxicity of epichlorohydrin:   percent  organ
          weight to body weight of  ratsa  (mean ± statistical error)
Organ
Adrenals
Brain
Gonads
Heart
Kidneys
Liver
Spleen
Cottonseed
oil control
0.023 ± 0.013
0.444 ± 0.018
0.785 ± 0.028
0.291 ± 0.011
0.670 ± 0.034
2.983 ± 0.219
0.174 ± 0.012

0.0095 mL/kg
0.011 ± 0.001
0.426 ± 0.012
0.707 ± 0.034
0.302 ± 0.018
0.636 ± 0.032
3.232 ± 0.157
0.194 ± 0.010
Epichlorohydrin
0.0190 mL/kg
0.010 ± 0.001
0.439 ± 0.029
0.866 ± 0.054
0.306 ± 0.030
0.728 ± 0.049
3.276 ± 0.186
0.208 ± 0.031

0.04774 mL/kg
0.013 ± 0.002
0.324 ± 0.027fc
o
0.402 ± 0.029b
0.917 ± 0.054b
4.070 ± 0.276d
0.225 ± 0.021
a
cent organ weight  to body weight.

      Significantly different from controls at 99%  level  (P = 0.01) by
Student's t test.
     Q
      Gonadal weights were not determined in this group.

      Significantly different from controls at 95%  level  (P = 0.05) by
Student's t test.

      Source:  Adapted  from Lawrence et al., 1972.   Reproduced with permission
of the copyright owner.

-------
                                    140
                  Table 4.22.  Comparison of 13-week and 2-year
                2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) studies
Test parameters with
significant changes
Mortality
Body weight
Food consumption
Packed cell volume
Red blood cell count
Hemoglobin concentration
Reticulocyte count
Thrombocyte count
Total leucocyte count
Total and direct bilirubin
Glutamic-pyruvic transaminase
Alkaline phosphatase
Relative thymus weight
Relative liver weight
Histopathologic changes6 in:
Thymus
Other lymphoid tissue
Uteri
Ovaries
Liver
Lung
Brain
Pituitary
13-week
1.0 ug/kg
*5
\d
\
4-M; tF
4-M; tF
4-M; tF
t
t
tF
t

t
4
t

M, F
M, F
F
F
M, F
M, F


study0
0.1 yg/kg

\
I
4-M
4-M
4-M



tF

tF
4-
t

M, F
M, F


M, F



2-year study
0.1 yg/kg 0.01 yg/kg
F
4- 4-F

4-F
4M
4-




tF
tF
4-F
tF tF

F

F

M, F M, F
M, F F
F
F
     QAdapted from Kociba et al., 1976.
      Adapted from Kociba et al., 1978.
     Q
      F — females only; M — males only.
      4- — decrease in value vs control; t — increase in value vs control.
     o
      Histopathologic changes exclusive of carcinogenic tumors.
     In  another study of TCDD,  Harris et al. (1973) tested the toxic
effects  in female rats from  gastric intubation for  31  consecutive days
with doses of 0.1, 1, and 10 mg/kg.  At the high dose,  significant
weight loss (Table 4.23) occurred by the 7th day, and  15 of 16 rats died
by a mean  of 21.8 days after the start of dosing.   For the intermediate
dose, weight gain was depressed significantly during the first 35 days,
but reversed to yield significant growth increases  during a recovery
period on  days 35 to 63.  Organ weight changes occurred only in the
liver and  the thymus (Table  4.24).  Liver weight increased at all three
dose levels (although statistically erratic) by the 10th day of exposure.
Livers at  the high dose showed  a decrease in weight at the 17th and 24th

-------
                 Table  4.23.  Weight  gain of  female rats  receiving  31  daily  doses  of
                              2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)
 Daily      Initial                                                     Weight  gain
 TCDD       number         Initial  body weight                      (g  ±  standard deviation)
 dose         of         (g ±  standard  deviation)
 (yg/kg)     animals                                    Days  1  to  7       Days  1  to  35a     Days  35  to 63a

   0          16                183.3  ±  6.28            15.8  ±  6.6       65.8  ±  8.0       28.4  ± 9.7
   1          12                185.6  ±  5.04            12.1  ±  6.4       34.8  ±  12.2b      42.0  ± 11.9°
  10          16                184.0  ±  8.00          -21.4  ±  14.9fc
     Values based on 5 and 8 rats  at 0 and  1 yg/kg  respectively; 15 of 16 rats died or were killed
when moribund at mean of 21.8 days.
     bP < 0.01.
     °P < 0.06.
     Source:  Adapted from Harris et  al., 1973.

-------
    Table 4.24.  Liver and thymus weights of female rats receiving daily doses of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)
Number
of
doses
3
6
10
13
17
24
31
Number
of
animals
4
4
4
4
4
4
4
Statistical
*A11
°P<
0 yg/kg
Liver
(g ± SE?)
7.80 ± 0.48
8.41 ± 0.36
9.14 ± 0.21
7.90 ± 0.31
9.26 ± 0.42
9.45 ± 0.31
9.70 ± 0.49
error.
dose response tests for
0.05.

Thymus ,
(mg ± SE)ft



530 ± 48
615 ± 59
525 ± 43
558 ± 87

0.1
Liver
(g ± SE)
8.62 ± 0.41
10.35 ± 0.45C
10.78 ± 0.41C
10.17 ± 0.94°
10.91 ± 0.37C
11.40 ± 1.05
11.78 ± 0.81

Mg/kg

Thymus
(mg ± SE)fc



492 ±
468 ±
412 ±
475 ±




81
22
28°
49

1.0 vg/kg

Liver Thymus
(g ± SE) (mg ± SE)fc
9.32 ± 0.45
9.00 ± 0.14,
11.53 + 0.40
9.76 ± 0.62,
11.33 ± 0.29d
11.39 ± 0.98
12.84 ± 0.84C




390 ± 58
270 ± 11"
372 ± 28^
260 ± 41a


10 yg/kg
Liver
(g ± SE)
9.58
10.84
10.53
9.80
7.52
4.67


± 0.43
± 0.97
± 0.36°
± 1.08
± 1.37,
± 0.21a



Thymus
(mg ± SE)b



142 ± 12°
110 ± 48°
32 ± lQd


thymus significant at P < 0.01.









 P < 0.01.
Source:  Adapted from Harris et al., 1973.

-------
                                  143
days of exposure, caused primarily by reduced body weight.   The thymus
weight was significantly decreased by day 17 at both the high and inter-
mediate doses.  Results of this study indicated that the 30-day duration
would have been sufficient to detect body and organ weight  changes and that
even 24 days would have indicated the majority of effects.   The results of
this 30-day study also correlate relatively well with the results of the
longer 13-week and 2-year TCDD toxicity studies by Kociba et al. (1976;
1978).
     Rosenkrantz et al. (1975) assessed the oral toxicity of A9-tetra-
hydrocannabinol, the major active ingredient in marijuana,  in rats
exposed for 28, 90, and 180 days at concentrations of 2, 10, and 50 mg/kg
per day.   Body growth depression was one of the primary effects, with an
8% to 12% rate decline at 28 and 90 days, increasing to 9%  to 17% by
180 days.  Hematological and biochemical parameters showed  several
trends.  At 28 days, although there were no statistically significant
changes,  there were increases in lymphocyte levels of males, increases
in reticulocyte levels of females, and decreases in polymorphonuclear
cells of males.  At 90 days, red blood cell numbers increased 11%,
hematocrit levels increased 8%, and for females the white blood cell
levels increased 19% to 74% in a dose-dependent manner.  At 180 days,
hematocrit and red blood cells rose 11% and 7%, respectively, in the
females.   Serum glutamic oxaloacetic transaminase (SCOT) and serum
glutamic pyruvic transaminase  (SGPT) levels rose in the males at rates
of 44% and 65%.  The relative adrenal weight increased 30% at 28 days,
and for males the pancreas weight increased 10% and the prostate
weight decreased 30%.  After 90 days of  treatment,.the brain weight  in-
creased 3% to 25%, and the kidney weight increased 7% to 18%.   Adrenal
weight remained elevated while the prostate weight continued to decrease,
but only at 6% to  21%.  At 180 days, more  organs revealed weight  changes
including increases  in the brain  (16%),  lung  (10% to  23%),  kidney (11%
to 20%), heart  (12%),  liver  (10%), and  adrenal glands  (34%  to  56%).
Changes in organ weight  in  the males were  as  follows:   the  thymus decreased
17%, the pancreas  increased  96%,  the prostate increased 13%,  and  the
testes increased 25%.  In the  females  the  weight  of  the uterus decreased
14%.  Although many  of these changes at 180 days  were dose  dependent,

-------
                                   144
 the significance was borderline.   None of the histopathological changes
 was considered significant,  with lesions occurring only incidentally in
 both control and treatment animals.   In this study, the 28-day duration
 revealed the same changes as the longer studies for growth rate depression
 and weight changes in minor organs.   However, the study failed to detect
 weight changes in major organs and in hematology values and suggests
 that a 28-day test does not adequately predict what happens at 90 days.
 By  the 90th day,  toxic effects in these categories were detected.
 Although the 180-day duration revealed more  changes,  the indication  of
 toxicity was sufficient at 90 days to suggest the  potential  danger of
 the compound.
      In a study of the oral  toxicity  of fominoben-HCl,  Kast  et  al.
 (1975&)  dosed rats daily for 1 and 6  months.   Even though  the  6-month
 test is not a subchronic duration, this study is useful for  evaluating
 the effectiveness of short-term tests (less  than 90 days)  to predict
 long-term effects.   In the 1-month study,  dose levels of 250,  500, 1000,
 and 2000 rag/kg were used,  and the 6-month study used levels  of  50, 100,
 and 200 mg/kg.   In the shorter test,  mortality was evident by  the
 second week at the high dose,  with 9  of 30 rats dying.   Body weight  gain
 was depressed from the first week until the  end of the  1-month  test.
 Other effects detected at  the high dose were increases  in  SCOT,  SGPT,
 serum alkaline phosphatase,  serum bilirubin,  and serum  cholesterol;  a
 decrease in total protein;  liver  pathology;  atrophy of  the gastric
 mucosa;  and decreases in thymus and testes weights in male rats.  The
 liver weight increased at  a  dose-dependent rate.   In the 6-month experi-
 ment,  no mortality occurred,  but  body weight  gain  was decreased at the
 high dose.   The biochemical  and hematological parameters were  affected
 less in  the longer study than in  the  1-month study,  with increases in
 SCOT and SGPT activities being the only significant changes.   The
 pathology was  limited to liver damage at the  high  dose,  and  liver weight
 increased at a  dose-dependent  rate.   In comparing  the results of the two
 duration periods,  it  is  apparent  that the  shorter  test  indicated the
major  effects,  particularly  the liver damage,  that were revealed in  the
6-month  study.  Therefore, in  this study,  the shorter test would have
been a sufficient predictor of  long-term effects.

-------
                                   145
     In another study by Kast et al. (1975a) the same experimental
design was used to test the toxicity of fenoterol-HBr.   The doses used
were 15, 150, and 1500 mg/kg (1 month) and 3, 30, and 300 mg/kg (6 months).
In the shorter study, mortality at the high dose was high (64% for
males, 24% for females), although in the chronic study only one substance-
related death occurred.  The body weight gain was significantly decreased
at the high dose in the 1-month test, but showed a significant increase
in the female after 6 months of testing.  The hematological and bio-
chemical tests performed for the 1-month duration revealed, at the high
dose, decreases in platelet count (also at intermediate dose) and blood
glucose levels and increases in blood urea nitrogen, Ca+, SCOT, and SGPT
levels.  In the 6-month study, an alkaline phosphatase increase at the
high dose was the only significant change, although blood glucose levels
showed a slight depression.  Organ weights increased for the salivary
glands (high dose), heart  (high and intermediate doses), and liver
(females at high and intermediate doses).  However, the males showed a
decrease in liver and testes weights at the high dose.  In the 6-month
study, only female heart weight increased at high and intermediate
doses.  In the shorter test, histopathological examination revealed
salivary gland and heart (primarily ischemic lesions) damage at  the high
dose.  Testes were also atrophied in three males at the high dose.  In
the 6-month test, only the heart damage  (again with a predominance of
ischemic lesions) occurred at the high dose.  Comparing the two  experi-
ments, it appears the shorter test  indicated more toxicity than  the
longer evaluation.  This may be caused by different dose magnitudes or
the effects of fenoterol-HBr being  acute in nature, or it may  indicate
an adaptation to the toxic effects  which reduced the final effects  in
the chronic study.
     Kruysse et al.  (1977) studied  the  inhalation  toxicity of  peroxyacetyl
nitrate  (PAN) by exposing  the rat  for 4  and  13 weeks.  The exposure
period was 6 h per day, 5  days  per  week, with dose levels of 0.9, 4.1,
and 11.8 ppm  (4 weeks) and 0.2, 1.0,  and 4.6 ppm (13 weeks).   In a com-
parison of the effects of  the two  durations,  both  produced quite similar
results, especially  at  the high doses.   In the  4-week test,  mortality
was observed at the  high dose,  with 9 of 20 rats dying.   No mortality

-------
                                   146
was  seen  after  the  13-week  exposure.  Growth depression occurred at the
high dose (4  and  13 weeks)  and  irregularly at  the intermediate dose (4
weeks).   This was accompanied in both tests by reduced food consumption.
In the 4-week test  (high  dose)  hemoglobin, hematocrit, and red blood
cell counts were  all  significantly  elevated; in the 13-week test (high
dose) hemoglobin  and  neutrophil levels were increased and lymphocyte
levels decreased.   Relative organ weights were also affected, but the
authors stated  that this  may be due to severe  growth depression, because
absolute  organ  values were  below control values.  After 4 weeks at the
high dose, heart, kidney, and lung weights increased, and the spleen
weight decreased.   After  13 weeks at  the high  dose, testes, thyroid, and
lung weights  all  increased.  Histopathology revealed lung and tracheal
damage after  4  and  13 weeks at  the high and intermediate doses.  The
degree and depth  of damage  increased  with higher doses.  The authors
concluded that  "comparable  PAN  effects" were observed at the high dose
in 4- and 13-week tests.  It appears  that the  4-week test would be
sufficient in this  case.
     Gray et  al.  (1977) studied the toxicity of di-(2-ethylhexyl)phthalate,
a plasticizer,  in the diet  of the rat for 17 weeks at concentrations of
0.2%, 1.0%, and 2.0%.  Toxic effects  were evident early in the study,
particularly  at the high  dose.  Loss  of body fur was noticeable by the
1st  week  at the high  dose and continued until  the 17th week, with the
affected  rats quite emaciated at that time.  Decreases in the body
growth rate (Table  4.25)  appeared by  the 2nd day at the high dose and at
the  intermediate  dose by  day 6  in the males.   This decrease was caused
in part by lowered  food consumption,  which was especially prominent
during the first  48 h.  As  shown in Table 4.26, there were irregular
fluctuations  in the packed  cell volume, hemoglobin, and red blood cell
values throughout the study.  The decrease in  packed cell volume was the
only significant  change occurring at  both 6 and 17 weeks.  As shown in
Table 4.27, changes in relative organ weights  occurred by the 2nd week
of dosing.  Increased weights were noted for the brain (females), liver,
stomach,  small  intestine  (males), caecum (males), thyroid (males), and
adrenals.  Decreases  in weight  were noted for  the heart, spleen, and
testes.   At 6 weeks,  the  only additional changes were an increase in

-------
          Table 4.25.  Mean body weights  and water  intake  of rats  fed diets containing 0% to 2.0%
                           di-(2-ethylhexyl)phthalate  (DEHP) for up  to 17 weeks
Dietary
level
(%)

0.0
0.2
1.0
2.0

0.0
0.2
1.0
2.0
aBody
Body weighta

0° 1 27

96 105 340
98 105 325,
98 100 297*
99 99 187

85 92 214
88 95 216
87 90 210
88 87 131*
in grams at day

55

478
455^

300*

273
277
259
164*

90

569
539
493*
413*

309
308-
2847
191*

120

628
588
546
447*

329
325d
201*
Water intake**

0°

18.3
17.6
18.0
18.4

15.9
17.9
17.4
18.3

1
Male
18.5
19.7
15.1
15.7
Female
15.7
18.9
15.4
14.8
in mL per rat per day at day

27

37.1
37.3
34.3,
24.9

21.5
24.9.
26. 6J
21.1

55

38.0
36.3,
27.9*
30.9

21.9
34.5
24.5
21.3

90

28.5
32.3
29.0,
34.4

18.1
26.5-
25. 97
18.9
Well.!
120 (mL per

26.3
24.7
27.7
26.9

19.4
22.1
22.6
16.7
Mean
>r intake
rat per day)

30.1
30.1
27.8-
25.7^

22.3-
25. 07
24. 37
19.7
weights are the means for 15 animals.
Values for water consumption
the 24-h period preceding the
*First
dp < o
day of treatment.
.01.
day


are the
shown.


means



for three cages






of five



animals



and were measured over









 P < 0.001.

fp < 0.05.
Source:  Adapted from Gray et al., 1977.  Reprinted with permission of the publisher.

-------
                                       148
       Table 4.26.  Mean hematological values2 for rats fed diets containing OX to 2.OX
                   di-(2-ethylhexyl)phthalate (DEHP) for 2, 6, or 17 weeks
Sex and
dietary
level

Male
0
1.0
2.0
Female
0
1.0
2.0

Male
0
1.0
2.0
Female
0
1.0
2.0

Male
0
0.2
1.0
2.0
Female
0
0.2
1.0
2.0
Number
of rats


5
5
5

5
5
5


5
5
5

5
5
5


15
15
15
15

15
15
15
15
Hb*
(g per 100 mL)


14.8
13.0^
14.8

15.1
14.4
14.8


15.1
15.0
14.9

15.8
15.4
14.4


16.0
15.4
14.5*
14. 5*

14.9
14.9
14.4
13.8
PCV0
(X)
Week

45
40
42

43
38
41
Week

48
45
46

49.
44*
431
Week

46
45
43?
43l

45
44
42*
42»
RBCd
(106 per mm3)
2

6.26
5.77^
6.48

6.50
6.24
6.82
6

7.08
6.96
6.86

7.81
7.90
7.49
17

7.57
7.44
6.97
7.60

7.14
7.05
7.26
6.78
Reticse
(X of RBC)


2.0
2.7
1.3

1.6
2.2.
0.8*


1.4
1.6
1.3

1.2
1.0
1.2


0.9
0.6
0.8
0.9

0.9
0.8
1.0
0.8
WB(/
(103 per mm3)


7.2
5.6
5.3

4.9
4.2
6.4


6.2
6.0
5.0

6.9
4.7
4.5


6.4
7.5
6.5
6.5

4.7
4.4
5.4
5.5
      Figures are means for the numbers of rats shown.
      Hb — Hemoglobin.
     CPCV — packed cell volume.
     TIBC — red blood cells.
     a
      Retics — reticulocytes.
     f                                ' '
     •'WBC — white blood cells.
     9P < 0.01.
     hP < 0.05.
     1P < 0.001.
     Source:  Adapted from Gray et al., 1977.  Reprinted with permission of the publisher.
heart weight, an  increase  in male brain weight, an  increase  in female
kidney weight, and  an increase in ovary weight.  However, by the 17th
week, no new organs were affected,  although  there were quantitative
changes.  An examination of the data in this study  reveals that the
majority of  effects were indicated  by the 2-week study, and  certainly by

-------
Table 4.27.  Relative organ weights of rats fed diets containing OX to 2.0% di-(2-ethylhexyl)phthalate (DEHP) for 2, 6, or 17 weeks
Sex and
dietary
level
tt)

Male
0
1.0
2.0
Female
0
1.0
2.0

Male
0
1.0
2.0
Female
0
1.0
2.0

Male
0
0.2
1.0
2.0
Female
0
0.2
1.2
2.0
Number
of rats
examined Brain


5 1.12
5 1.17
5 1.75

5 1.19
5 1.21,
5 2.041


5 0.56
5 0.62
5 1.03

5 0.80
5 0.89
5 1.63


15 0.37
15 0.39
15 0.42^
15 0.50°

15 0.64
15 0.64
15 0.69j
15 1.10
Relative organ weight (g per 100 g of body weight)
Heart Liver Spleen


0.46 3.15,
0.42 6.197,
0.42e 6.47a

0.45 3.61^
0.42* 5.28*
0.40 6.62°


0.35 2.99j
0.40* 4.72^
0.43d 6.76*

0.36 2.88,
0.38 4.40,
0.41* 5.83rf


0.27 2.31,
0.27 2.72,
0.28 3.450
0.30 4.12

0.30 2.25
0 . 30 2 . 60
0.31, 3.46^
0.39d 4.59d
""Relative weights of these organs are
Relative weights of the
°P <
dP<
0.01.
0.001.
female gonads




0.30
0.32
0.24

0.29
0.27
0.24e


0.21
0.19
0.28

0.26
0.24
0.25


0.14
0.15
0.14
0.16

0.16
0.17
0.17
0.19g
expressed
Kidneys


0.95
1.01
1.05

1.07
1.00
1.06


0.78
0.79
0.86

0.80
0.82
0.95"


0.56
0.59
0.64 ',
0.70 '

0.57
0.60,
0.65,
0.71"
in mg per
are expressed in mg




c. . Small
Stomach intestlne
Week

0.70
0.70
l.Ol''

0.69
0.65
0.98 '
Wet-k

0.45
0.47
0.78'

0.58
0.60 ,
0.98
Week

0.30
O.J1
0.33';
0.38

0.39
0.39
0.42^
0.72*
100 g of
per 100


2

3.39
J.76
4.41 '

3.62
3.79
4.39
6

2.42
3.15'^
4. 58*'

2.80
3.16 ,
3.80 '
17

1.45
1.55
1.66',
1 .91 '

2.01
2.09
3'.13'7
body weight.
Cecum


0.54
0.55
0.67 '

0.56
0.57
0.65


0.42
0.47
0.54

0.43
0.51
0.52


0.25
0.24
0.25
0.296

0.29
0.32^
0 . 34 ,
0 . 44"

Adrenals0


20.1
19.9
26.9"

36.0
25.8''
3 J.I


12.3
11.1
20.0''

22.5
23.0
27.7


8.9
9.5
10.1
10.9

17.2
19.0
21.9 '
20.4

Gonads


1.08
0.90
0.59^

57
50
41


0.88 ,
0. JO,
0.44

5r>
61
39''


0.60
0.61 y
0.41 ,
0.23'

30
29
37
25

Pituitary0


4.1
3.9
4.0

5.4
4.H
4.1


2.6
2.4
3.0

4.5
S.4
4.7


1.9
1.9
2.3 .
2 . 8

4.2
4.0
4. ft
4.0

Thyroid0


6.6
8.3
12.0 '

8.2
9. J
10.1


i.ft
4.7
H.I

6.2
7.1
10.1 '


J.8
i.4
5.1
5.1

7.5
6.5
7.8
9.1

g of body weight.












                                                                                                                                             vo
eP <  0.05.
Source:   Adapted from Gray et al.,  1977.   Reprinted with permission of the publisher.

-------
                                   150

 the  6th week.   The  only significant  change  found with 17 weeks of
 testing was  the quantitative degree  of  toxicity in  some organs.

 4.3.4  Conclusions
     The  literature contains many  well-designed studies evaluating the
 predictability of chronic  (lifetime)  effects  from subchronic data.  The
 review papers  by Weil and McCollister (1963)  and McNamara  (1976) presented
 compilations of data demonstrating that the 90-day  or 90-exposure test
 is a significant predictor  of  chronic effects.  Other authors generally
 agreed with  this conclusion, with  many  suggesting a duration of 3 to
 6 months  as  sufficient (Barnes and Denz,  1954; Bein, 1963; Boyd, 1968;
 Davey, 1964; Peck,  1968; World Health Organization  Technical Report,
 1966). In addition,  several primary studies  showed that chronic effects
 are  indicated  at a  statistically significant  level  by tests of 90 days
 (Ambrose  et  al., 1972;  Gaunt et al.,  1965;  Kociba et al.,  1976, 1978;
 Rosenkrantz  et al.,  1975).  Thus,  the evidence in the literature supports
 the  conclusion that a 90-day test  duration  does predict most chronic
 effects.
     There are many comparisons in the  literature concerning test
 durations shorter than 90 days.  Generally, these conclude that the
 shorter tests  do indicate toxic effect  patterns.  However, one must
 consider  that  many  of the primary  studies of  short  duration produced
 effects only at the high dose,  which reduced  the reliability of their
 conclusions.   Papers  by Smith  (1950)  and Weil et al. (1969) concluded
 that short tests (7  to 14 days)  could be sufficient to indicate toxic
 effects and  predict  safe dose  levels.   Two  papers by Kast  et al. (1975a,
 19753) indicated that a 1-month duration would adequately  predict
 6-month effects.  Thirty-day durations  have also been evaluated and
 generally recommended as sufficient  to  indicate toxicity or dose levels
 (Gray  et  al.,  1977; Hall, Austin,  and Fairweather,  1966; Harris et al.,
 1973;  Hodge, Downs, Panner, Smith, Maynard, Clayton, and Rhodes, 1967;
Krussye et al.,  1977; Misu  et  al., 1966).   However, Hayes  (1972) con-
cluded that 30-day durations were  insufficient, but in an  early paper
 (Hayes, 1967&) did agree that  a  90-day  duration was a good indicator of

-------
                                  151
chronic effects.  Thus, data from primary studies  generally support the
use of 30-day duration, although there is little concurrence on what
toxic effects might be missed.   Perhaps in the future,  with more informa-
tion, specific recommendations can be made.   In any case,  the acceptance
of a 90-day test as a significant evaluation of chronic effects can
reduce the cost and time factors.  This could reduce costs from approxi-
mately $255 800 for a complete chronic and subchronic study to $100 500
for a subchronic study design (Gehring, Rowe, and  McCollister, 1973).

4.4  ROUTE OF EXPOSURE

4.4.1  Introduction
     In subchronic test designs, various routes of administering the
chemical to the test animals have been utilized.  The routes most often
employed are oral and inhalation.  Alternate exposure methods such as
percutaneous application are used if advantageous  for a specific chemical.
In 183 subchronic studies taken from the literature, the frequency for
employment of each route is as follows:  oral, 51%; inhalation, 19%;
percutaneous, 2%; intravenous, 8%; intraperitoneal, 8%; subcutaneous,
4%; and others, 8%.
     The choice of which route to use should be based on the expected
route of human exposure (Benitz, 1970; Federal Register, 1978; Food
Safety Council, 1978; Ministry of Health and Welfare Canada, 1975;
National Academy of Sciences, 1975, 1977; Peck, 1974; World Health
Organization, 1978).  In addition to the metabolic reasons for using the
expected human exposure route, it is easier  to extrapolate safe dose
levels from test data to the actual human conditions if the same routes
are used.  However, in some cases it may not be advisable  to  use only
the expected human route.  If there are several potential  routes of
exposure, it may be necessary to  test  each one  to  determine the most
toxic route.  This can be important when the major route  of exposure  is
not the most toxic  (U.S. Environmental Protection  Agency,  1979).   The
effect at the site of exposure  is another important consideration  since
the concentration is controlled  by  the mode  of  exposure (U.S. Environmental

-------
                                   152
Protection Agency, 1979; Weisburger,  1975; World Health Organization,
1978).  Also,  the availability of  testing facilities may determine the
choice of exposure route,  especially  for inhalation studies.  In most
cases, however,  there  is little or no conflict among these variables,
and  the route  chosen is the  expected  route of human exposure.
     The following section will discuss aspects of individual routes
used most frequently in subchronic tests.  An additional section will
briefly consider the primary literature base for comparison of routes
and  the validity of extrapolation  between routes.

4.4.2  Route Discussions
     Oral administration of  chemicals is the principal route used in
most toxicity  experiments.   This predominance results from both practical
considerations,  including  ease of  application and low cost factors,
and  experimental requirements (oral exposure represents the expected
primary route  of human exposure for many substances).  In general, oral
exposures produce quick responses  of  intermediate toxicity.  Hayes
(1967a) rates  oral exposure  as more toxic than dermal applications,
while the World  Health Organization  (1978) rates oral (gavage) as more
toxic than dermal but  less than inhalation exposures.  On a more specific
level, oral  administration of quickly absorbed chemicals produces more
liver toxicity than any other route  (Loomis, 1974; World Health Organiza-
tion, 1978); slowly absorbed chemicals produce more gastrointestinal
damage (Ministry of Health and Welfare Canada, 1975).
     Oral administration can be performed by various techniques, including
mixture in diet  (food  or drinking  water), gavage (gastric intubation),
and  gelatin  capsules or pressed tablets.  The relative frequency of use
for  these techniques in 95 oral application studies surveyed was as
follows:  gavage, 27%; capsule, 11%;  and diet, 60%  (89% in food; 11%  in
water).
     The most  common technique for oral exposure of chemicals is by
mixing the test  substance  into the diet.  One of the advantages of this
method is the  reduced  handling time due to the simplicity of administra-
tion.  This saves valuable personnel  time and disturbs the test animals

-------
                                  153
less (Barnes and Denz, 1954).   Diet mixtures also produce a low,
prolonged exposure with few concentration peaks (Benitz,  1970).   This
exposure pattern is more representative of chronic human  exposure than
other oral routes (Sontag, Page, and Saffiotti, 1976).  However,  one
must be aware of possible shortcomings or hazards involved in the use of
diet mixtures.  These include:  limited duration for stability of the
drug in the diet (Benitz, 1970; World Health Organization, 1978); adjust-
ment of the dose with animal maturation to maintain proper concentration
levels (Barnes and Denz, 1954; National Academy of Sciences, 1977; World
Health Organization, 1978); alteration of nutrient availability or
quality in the diet (Barnes and Denz, 1954; Food Safety Council,  1978);
unpalatability of the diet mixture to test animals affecting growth
rates (Barnes and Denz, 1954); nonhomogeneity of the diet-chemical
mixture (Sontag, Page, and Saffiotti, 1976); and measurement of daily
food consumption to calculate daily exposures  (Barnes and Denz, 1954;
Benitz, 1970; Ministry of Health and Welfare Canada, 1975).  The choice
between diet and gavage or capsule application is often based on such
considerations.  Use of gavage or capsules eliminates problems of
palatability, drug stability, nutrient integrity, and consumption
calculations associated with  diet administration.  However,  gavage  or
capsules tend to produce peaks in substance concentrations,  giving  an
irregular cycle to blood and  tissue levels  (Benitz, 1970).   Also, only
small amounts of a compound can be administered by gavage,  thus  limiting
its use to highly toxic substances  (Food  Safety Council,  1978).   Other
disadvantages include higher  mortality rates and more frequent need to
use a solvent to administer the chemical  (Sontag, Page,  and Saffiotti,
1976).
     Often the best choice depends  on the chemical being tested.  Worden
and Harper  (1963) and Bein (1963)  gave examples  of  chemicals that are
more toxic by feed than by gavage  and vice versa.   If  there is little
difference between the  techniques,  diet  is to  be preferred because of
its simplicity  (World Health  Organization, 1978).   However, either
method is acceptable  for  exposure  in tests involving subchronic  durations
 (Ministry of Health and Welfare Canada,  1975;  National Academy of Sciences,
1977).

-------
                                   154
     Inhalation exposure  is used in studies where the primary human
exposure  is  to be by  inhalation or when information on the specific site
of entry  (lung) damage  is desired  (Roe, 1968).  The total effects of
inhalation can rarely be  predicted  (with a high degree of certainty)
from oral or parenteral studies, which necessitates the use of inhala-
tion exposure (Gage,  1970; World Health Organization, 1978).  However,
unlike  oral  administration, use of  inhalation exposure is neither simple
nor inexpensive.
     For  an  adequate  study of  inhalation toxicity, specially designed
inhalation chambers are needed.  The  various chamber designs and advan-
tages are too numerous  to discuss  in  this document, but Roe (1968), the
National  Academy of Sciences  (1977),  and the World Health Organization
(1978)  provide good reviews of this area.  Additional important considera-
tions are the increased cost that  this route of exposure entails and the
potential bottleneck  represented by the limited number of quality
facilities available  for  testing.
     One  aspect of the  test design that must be considered is the diffi-
culty associated with the determination of dose levels in an inhalation
study.  The  dose is usually measured  by monitoring the chamber concen-
tration (a)  of the toxicant and the time  (£) that the test animal is in
the chamber  (MacFarland,  1968; Roe, 1968).  The dose is expressed as the
product (at)  of these two variables  (Ministry of Health and Welfare
Canada, 1975) and is  used in lieu  of  actual dosage levels (National
Academy of Sciences,  1977).  However, doses given in this manner are
difficult, if not impossible,  to correlate with oral or injection studies
where the dose is expressed as milligrams per kilogram of body weight
(Barnes and  Denz, 1954; Hayes  1967a).  Additional problems associated
with inhalation dosage  are the uncertainties regarding the actual amount
of the  toxicant inhaled by the test animal, the amount of toxicant that
adsorbs onto  the chamber  walls, and possible variations in the flow rate
of the  toxicant into  the  chamber (National Academy of Sciences, 1977;
World Health  Organization, 1978).  The amount of toxicant that enters
the test  animal is influenced  by its  ventilation rate, the particle size
or vapor  pressure of  the  toxicant, and the behavioral traits of the test
species (National Academy of Sciences, 1977; Roe, 1968).

-------
                                  155

     Another factor of importance in the chamber design is the expected
schedule of exposure durations.  If continuous exposures (22 to 24 h/day,
7 days/week) are to be used, then the chamber must provide all food,
water, and other needs of the test animals.   If intermittent exposures
(6 to 8 h/day, 5 days/week) are used, chambers of a simpler design can
be used.  These two duration patterns produce different effects.  The
nonexposure time in an intermittent test design allows the animal to
recover and tends to produce a pulsed chemical concentration pattern
(World Health Organization, 1978).  In contrast, the continuous exposure
results in a steady-state pattern of toxin concentrations.  The choice
of duration schedules should be related to the expected human exposure
pattern.
     The generation and characterization of aerosols is another complexity
involved with inhalation studies.  This is of little concern in vapor or
gas  studies, but in aerosol mist or dust studies the aerosol particles
must be evaluated.  Specialized equipment is required to generate the
aerosol particles, and some control is necessary to ensure uniform
particle size and density, at  least within a size range that is suitable
for  biological action  (Ministry of Health and Welfare Canada,  1975;
National Academy of Sciences,  1977; Roe, 1968).  Particle  size must  also
be monitored  in the chamber itself to ensure that the exposure levels
remain  consistent.  The deposition sites (nose,  trachea, bronchi, or
alveoli) and  the degree of biological action of  the inhaled particles
are  determined by  their solubility in tissue  fluids,  their reactivity
with lung  tissue,  and  their size  (Roe,  1968; World Health Organization,
1978).  Generally  the  larger  particles,  5 to  10 urn in diameter, are
trapped in the upper  respiratory  tract  and nasal cavity (Ministry of
Health  and Welfare Canada, 1975;  World  Health Organization, 1978).
Particles  of  less  than 5  um disperse further into the lower respiratory
tract,  with the depth of  penetration increasing with  decreasing particle
size (World Health Organization,  1978).   It  has been  estimated that
approximately 25%  of  the  inhaled particles  are immediately expelled, 50%
are  trapped in the upper  respiratory tract,  and only 25% reach the
alveoli (Morrow et al.,  1966).

-------
                                  156
     After inhalation exposure, two types of toxic effects can occur.
Systemic effects, not related to the site of entry, are often observed
and in most studies represent the major toxic effects (Barnes and Denz,
1954; World Health Organization, 1978).  These effects are quite similar
to parenteral or intravenous injection effects, at least in qualitative
data (Gage, 1970; World Health Organization, 1978).  In contrast to oral
exposure, the circulatory transport of toxins entering through the lung
does not carry them through the liver first.  Thus, other organs (e.g.,
brain, heart, and endocrine glands) are often sites of damage after
inhalation exposure (World Health Organization, 1978).  Specific site of
entry effects (lung damage) can be studied only with inhalation exposure
(Roe, 1968).  These effects result from both physical impairment of
respiratory function and slow metabolic transformation and transport by
lung epithelial tissue.  Tests to evaluate this specific lung and
tracheal damage are reviewed by Roe (1968) and the World Health Organiza-
tion (1978).  These include respiratory function tests, as well as
pathological damage evaluations.
     A complication in evaluating toxic effects from inhalation is the
clearance mechanism.  The mucociliary system often transports particles
out of the lung.  These are either expelled or swallowed.  If expelled,
the dose levels are not as potent as originally designed.  If swallowed,
oral exposure results, which produces different types of toxic damage
(World Health Organization, 1978).  These problems are most often
associated with the dust or aerosol studies.
     The unique value of inhalation exposure in subchronic test designs
is primarily for the assessment of specific lung damage.  Because of the
high cost, limited facilities, and additional personnel time involved,
inhalation exposure should be limited to such assessments and not used
for general assessment of systematic toxicity.
     Other routes of exposure are infrequently used in subchronic studies,
including percutaneous and injection methods.  Percutaneous or dermal
application is usually performed as a separate study.  When percutaneous
exposure is used, several factors influence the test design and data
interpretation.   These include slower activity and lower exposures than

-------
                                   157

with oral exposure, difficulty of measuring absorbed dose,  necessity of
using a vehicle or solvent in many cases, and ingestion by  animals
during self-cleaning of doses applied to the skin (Benitz,  1970; Loomis,
1974; Ministry of Health and Welfare Canada, 1975; National Academy of
Sciences, 1977).
     Parenteral injections are also used occasionally in subchronic
tests.  These include intradermal, subcutaneous, intramuscular, intra-
venous, intraperitoneal, and intrathecal (Loomis, 1974).  Intravenous
administration produces rapid distribution of toxins, with the degree of
metabolic transformation dependent upon the location of the used blood
vessel in the body.  The other injection methods concentrate doses in
selected areas with slow diffusion to other tissues (Loomis, 1974).  In
any injection method, rotation of injection sites is necessary  (Benitz,
1970).  Although this prevents excessive entry site damage, the use of
different target sites complicates the analysis of effects.  In most
subchronic studies, the use of parenteral administration is not the
preferred route, because it is rarely an expected route of human exposure.

4.4.3  Route Comparisons
     Few papers in the literature discuss in detail the comparative
value of exposure  routes.  The review articles used in  Sect. 4.4.2
contain most of the available information.  Primary studies designed  to
evaluate this area are almost nonexistent.  Even within individual
chemicals, comparisons are difficult.  The  use of different species,
doses, durations,  and parameters of  evaluation  (pathology, hematology,
etc.) all combine  to make route comparisons feasible  only  at a general
level.  A few brief comparisons within chemicals  are  included  as  examples
of the difficulties.  These  are summarized  in Table  4.28.   Following the
chemical comparisons will be a brief discussion of  route-to-route
extrapolation.
     4.4.3.1  Arsenic — Bencko and Symon (1969)  studied tissue levels of
arsenic  in hairless mice  following administration of white arsenic
 (AS203)  in drinking water.   Continuous  exposure to 50 mg or 250 mg of

-------
                                 Table A.28.  Summary of chemical examples tested  for toxiclty by various routes of  administration
Chemical
Arsenic




Beryllium salts

Insoluble salts



Soluble compounds


Beryllium chloride
Beryllium nitrate
Beryllium sulfate
Beryllium oxyfluorlde
Beryllium oxide
Beryllium phosphate
Beryllium




Beryllium sulfate


Beryllium fluoride


Beryllium sulfate


Beryllium oxide


Route
Oral

Inhalation


Oral

Inhalation " ~~



Inhalation


Intraperitoneal
Intraperitoneal
Intraperitoneal
Intraperitoneal
Intraperitoneal
Intraperitoneal
Oral




Inhalation


Inhalation


Inhalation


Inhalation


Species
Mouse

Mouse


Guinea pig

Guinea pig



Guinea pig


Guinea pig
Guinea pig
Guinea pig
Guinea pig
Guinea pig
Guinea pig
Rat




Rat


Rabbit, dog


Dog, rat


Rabbit


Dose
10 to 15 mg/
kg/day
179. A UK As
per m*

0 to 30 mg/
kg/day
188.9 and 233
mg/ra3


27 and A3
mg/m3

0.1 g/day
0.1 g/day
0.1 g/day
0.1 g/day
0.1 g/day
0.1 g/day
6.6 and 66.6
Ug/day



34.25 pg
Be per m3

2.2 mg/m3


A mg/m3


80 mg/m3


Duration
32 days

6 h/day,
5 day /week.
6 weeks
11 to 29 weeks

30 to AO min/
day, 6 days/
week, 1 day to
15 weeks
30 to AO rain/day,
6 days/week,
1 and 10 days
A days
A days
A days
A days
4 months
A months
6, 12, 18, 2A
months



7 h/day, 5
days/week,
72 weeks
6 h/day, 5
days/week,
6 to 23 weeks
6 h/day, 5
days/week,
6 to 23 weeks
6 h/day, 5
days/week,
6 to 23 weeks
Effects noted by author
Transient accumulation in skin
and liver; weight loss
Transient accumulation in skin.
liver, and kidney

No adverse effects; low tissue
levels
No adverse effects; low tissue
levels


Severe symptoms; low tissue
levels

100Z mortality
100Z mortality
100Z mortality
100Z mortality
No mortality
No mortality
Decreased body weight; accumu-
lation in gastrointestinal
tract; skeleton, blood,
liver. No hepatic cell
destruction.
Inflammatory response; lung
tumors

Macrocytic anemia


Macrocytlc anemia


Macrocytic anemia


Reference
Bencko and Symon,
1969
Bencko and Symon,
1970

Hyslop, 19A3

Hyslop, 19A3



Hyslop, 19A3


Hyslop, 19A3
Hyslop, 19A3
Hyslop, 19A3
Hyslop, 19A3
Hyslop, 1943 M
Hyslop, 19A3 Ui
CO
Reeves, 1965




Reeves, Dei ten.
and Vorwald,
1967
Stokinger and
Stroud, 1951

Stokinger and
Stroud, 1951

Stokinger and
Stroud, 1951

Carbon tetrachlorlde      Inhalation
                          Inhalation
                                             Rat
                                             Rat
Not given
10, SO, 100
  ppm/day
3 to 5 min, 3
  times per day
  on alternate
  days In 35 days
3 h/day, 6 to 8
  weeks
                                                                                                  Cirrhosis of liver
Elevated liver triglycerides,
SCOT,0 SGPT* unaffected
Reddy,
  Krishnamurthy,
  and Bhaskar,
  1962
Shimizu, Nagase,
  and Kawal, 1973

-------
                                                                     Table 4.28  (continued)
Chemical























Dimethyl terephthalate




Fenoterol HBr









2-Methyl-A-
chlorophenoxy
Route
Oral
Oral

Oral


Subcutaneous



Subcutaneous

Subcutaneous

Subcutaneous

Subcutaneous

Intraperitoneal




Oral

Inhalation


Oral





Inhalation

Intravenous

Dermal

Species
Rat
Rat

Rat


Rat



Rat

Rat

Rat

Rat

Rat




Rat

Rat


House, rat





Rat

Rabbit

Rabbit

Dose
2 iiL per 100 g
150 to S20 ppm/
day
0.2 mL/kg/day


0.1 mL/100 g



1.3 mL/kg/day

1.3 mL/kg/day

1.3 mL/kg/day

1.3 mL/kg/day

0.06 mL per
100 kg



0.25, 0.5, IX
per day
16.5 and 86.4
mg/m3

15, 150, 1500
mg/kg/day
(rat)
1.5, 15, 150
mg/kg/day
(mouse)
0.01 to 1.0 rag/
kg/day
1.0 and 25 to
50 mg/kg/day
0.5, 1.0, 2.0
g/kg/day
Duration
1 day (acute)
6 weeks

3 days/week,
7 weeks

1 dose per
3 days in
90 days

2 days/week.
12 weeks
2 days/week,
12 weeks
2 days /week,
12 weeks
2 days /week.
12 weeks
I to 10 injec-
tions in
119 days


96 days

4 h/day, 5
days/week, 58
exposures
30 days


30 days


30 days

30 days

3 weeks

Effects noted by author
(Single dose) elevated SGOT,a
SGPT6
Trlglyceride accumulation in
liver
Increased liver fat


Cirrhosis



Cholanglofibrosis

Hepatic vein thrombosis

Neoplasia

Cirrhosis

Retardation of growth; elevated
SGPT; hepatic necrosis (tran-
sient); Inhibition of normal
mineralization of tooth
dentine
Decreased body weight

No adverse effects


Enlarged salivary glands;
enlarged hearts. Mortality:
rats — 642 male, 24X female;
mouse — 3X male and 3X female


No adverse effects; <1X
mortality
Enlarged salivary glands;
enlarged hearts; 6X mortality
Transient growth retardation;
erythema; decreased leuko-
Reference
Shimizu, Nagase,
and Kawai, 1973
Alumot et al. ,
1976
Friedman, Sage,
and Blendermann
1970
Reddy,
Krishnamur thy ,
and Bhaskar,
1962
Reuber and Glover,
1967a
Reuber and Glover,
1967Z>
Reuber and Glover,
1967e
Reuber and Glover,
1968
Hals, Bjorlln, and
Jacobsen, 1973
(-•
Ui
\0
Krasavage, Yanno,
and Terhaar, 1973
Krasavage, Yanno,
and Terhaar, 1973

Kast et al., 1975a





Kast et al., 1975a

Kast et al., 1975a

Verschuuren, Kroes,
and Tonkelaar,
i nt e
acetic acid
(MCPA)
  cytes; hyperplaaia, hyper-
  keratoais, and loss of
  elasticity of skin
100X and 75X mortality in high
  dose groups
                                                                                                                                     1975

-------
                                                                      Table 4.28  (continued)
       Chemical
                             Route
                                                 Species
                                                                   Dose
                                                         Duration
                                        Effects noted by author
                                                                                                                                         Reference
2-Methyl-A-
  chlorophenoxy
  propionic acid
  (MCPP)
                          Oral
Dermal
Oral
                                             Rat
                   Rabbit
                                             Rat
                                                              50, 400, 3200    90 days
                                                                ppm
0.5, 1.0, 2.0    3 weeks
  g/kg per day

50, 400, 3200    90 days
  ppm
Growth retardation
Increased kidney weight
Increased erythrocyte size
Increased hemoglobin content

Transient growth retardation;
  erythema, transient loss of
  skin elasticity
Decreased red blood cell count;
  decreased hemoglobin content;
  increased alkaline phosphatase
  activity, decreased hematocrit
  and leucocyte values (male
  only); Increased kidney weights
  depression of ovary and
  prostate weights
                                                                                                          Verschuuren, Kroes,
                                                                                                            and Tonkelaar,
                                                                                                            1975
Verschuuren, Kroes,
  and Tonkelaar,
  1975
Verschuuren, Kroes,
  and Tonkelaar,
  1975
Nefopam hydrochloride Intraperitoneal

Intramuscular

Oral


Intravenous

Oral


Intramuscular


A9-Tetrahydrocannablnol Oral


Intravenous


Rat

Rat

Rat


Dog

Dog


Dog


Rhesus monkey





2 and 10 mg/
kg per day
1 and 2 mg/
kg per day
20, 40, 80
mg/kg per
day
1 and 5 mg/
kg per day
4, 10, 40
mg/kg
per day
1.5 and 3.0
mg/kg
per day
50, 250, 500
mg/kg
per day
5, 15, 45
mg/kg
per day
5 days/week.
4 weeks
7 days /week,
2 weeks
4 weeks


7 days/week.
4 weeks
7 days/week,
4 weeks

7 days/week,
3 weeks

28 days


28 days


Increased liver weights

No adverse effects

No adverse effects


Slight weight loss

Slight weight loss


No adverse effects


Summarized In Table 4.29





Case, Smith, and
Nelson, 1975
Case, Smith, and
Nelson, 1975
Case, Smith, and
Nelson, 1975

Case, Smith, and
Nelson, 1975
Case, Smith, and
Nelson, 1975

Case, Smith, and
Nelson. 1975

Thompson et al . ,
1974




       SCOT — serum glutamlc oxaloacetic transaminase.
       SGPT — serum glutamlc pyruvic transaminase.

-------
                                   161
arsenic per liter (10 to 15 mg/kg per day)  for 32  days  produced levels
of the chemical in the skin and liver that  peaked  at 16 days,  then
decreased significantly by 32 days.
     In an inhalation experiment (Bencko and Symon,  1970),  hairless mice
were exposed in a dust chamber to As203 dried onto fly  ash  at  a mean
concentration of 179.4 yg arsenic per m3 for 5 days per week,  6 h daily,
for 6 weeks.  Concentrations of arsenic in  the liver, kidney,  and skin
were measured after 1, 2, 4, and 6 weeks of exposure.  Arsenic levels
increased rapidly in the liver and kidney up to 2  weeks, then  decreased
sharply during the third and fourth weeks.   Skin values peaked similarly
as a result of direct exposure in the dust  chamber, but decreased more
gradually.  The authors recognized that effects of inhaled particles
cannot be distinguished from effects of particles  ingested as  a result
of mucociliary clearance or licked from the skin during grooming, and
they concluded that the course of arsenic accumulation is similar fol-
lowing oral administration and inhalation.   The drop in arsenic accumula-
tion in tissues after the initial peak was subsequently shown to be
caused by a stimulation of the excretory mechanism, which increased the
excretory rate of the animals  (Bencko, Dvorak, and Symon, 1973).
     4.4.3.2  Beryllium — In an extensive test for beryllium toxicity
(Hyslop, 1943), guinea pigs ingested up to 40 mg per kg of body weight
per day of various beryllium salts and exhibited no adverse effects
after 11 to 29 weeks of exposure.  The total amounts of beryllium
retained in body tissues were  small in comparison to the quantities
ingested.  Inhalation of insoluble beryllium carbonate (188.9 mg/m3  or
233 mg/m3) 30 to 40 min/day, 6 days/week for 1 day  to  15 weeks,  failed
to produce toxic effects in guinea pigs.  However,  inhalation  of soluble
compounds induced severe symptoms  in a short time:   27 mg/m3 of  beryllium
oxyfluoride and 43 mg/m3 beryllium sulfate  induced  67% mortality after
10 days and 1 day of exposure, respectively.  Distribution of  the
chemical in tissues remained remarkably low following  inhalation of both
soluble and insoluble beryllium  salts.  Intraperitoneal injections of
0.1 g of beryllium chloride, beryllium nitrate, beryllium sulfate, or
beryllium oxyfluoride produced 100% mortality in  guinea pigs  after 4
days; beryllium oxide and  phosphate were not lethal after 4 months.

-------
                                   162
     Other studies of beryllium toxicity are summarized in Table 4.28.
These are of little value for route comparisons because of dose, duration,
and species differences.
     4.4.3.3  Carbon Tetrachloride  (CCl^) — Reddy , Kr ishnamurthy , and
Bhaskar  (1962) exposed albino Wistar rats to carbon tetrachloride
vapors for  3 to 5 h,  three  times per day on alternate days for 35 days.
Inhalation  of the vapors  caused cirrhosis of the liver in 8 of 9 males
and  5 of 15 females  tested.  Eight of  the 15 females developed patchy
fibrosis of the liver.  Inhalation of  10, 50, or 100 ppm CClit per day,
3 h/day  for 6 to 8 weeks  resulted in temporarily elevated triglyceride
levels in the livers of male Sprague-Dawley rats, but no progressive or
cumulative  changes were observed  (Shimuzu, Nagase, and Kawai, 1973).
Transaminase activity was not affected by inhalation exposure, although
a single oral dose of CClt4  produced increased SCOT and SGPT values.
     Elevated levels of liver fat content have been observed in rats
following oral administration of CCl^.  After 6 weeks of feeding
fumigated mash containing a daily dose of 1% of the LD50 (5 g per kg of
body weight) , triglyceride  accumulation in the liver was found to be the
most sensitive sign  of chronic CCl^ poisoning  (Alumot et al. , 1976).
Oral intubation of 0.2 mL CCl^ per kg  of body weight three times a week
for  7 weeks also caused an  increase in the liver fat content of rats
(Friedman,  Sage, and Blendermann, 1970).
     Subcutaneous injection of 0.1 mL  per 100 g of body weight every
3 days for  90 days produced cirrhosis  in 9 of 9 male rats and in 6 of
15 female rats (Reddy, Kr ishnamurthy,  and Bhaskar, 1962).  Five females
developed patchy fibrosis of the liver, and four females did not develop
liver lesions.  The  authors concluded  that the frequency of CCl^-induced
cirrhosis was the same following inhalation (described previously) and
subcutaneous exposure.  In  a series of separate studies in which CClt*
was  administered subcutaneously to Buffalo strain male and female rats,
effects of  the chemical were manifested as a function of age and sex of
the  animals (Reuber  and Glover, 1967a, 19672>, 1967c, 1968).
     Intraperitoneal injection of 0.06 mL of CCli, per 100 kg of body
weight (up  to seven  injections during  119 days) into albino rats induced

-------
                                  163

an inhibition of weight gain, elevated SGPT values,  and hepatic necrosis,
which then regenerated (Hals, Bjorling, and Jacobsen,  1973).   Concom-
itantly, inhibition of normal mineralization of the  dentine layer of
rat incisors was observed.
     These ten studies on carbon tetrachloride, although using the same
species, contain wide variations in dose and duration.  Also, the use of
different age groups in some studies further complicates the comparisons.
Thus, comparisons of routes using these data would be difficult.
     4.4.3.4  Dimethyl Terephthalate (DMT) — DMT, shown previously to be
of a low order of toxicity when administered intragastrically, intraperi-
toneally, and subcutaneously to rats and mice  (Slyusar and Cherkasov,
1964, and Prusakov, 1966, as cited in Krasavage, Yanno, and Terhaar,
1973), was further evaluated in various acute  studies and in subchronic
feeding and inhalation studies by Krasavage, Yanno,  and Terhaar  (1973).
Neither feeding nor inhalation caused mortality.  The toxicological
effect seen in either study was reduced weight gain in high dose animals
of the feeding study.  Although this study was well designed for a
comparison of routes, the lack of toxic effects precludes comparisons.
     4.4.3.5  Fenoterol HBr — Kast et  al.  (1975a) tested the beta-
sympathomimetic,  fenoterol HBr, for toxic properties.   Subacute exposures
 (oral,  intravenous, and inhalation) continued  for 1 month.   Inhalation
produced no symptoms  in rats that could be  related  to fenoterol.  However,
after oral administration enlarged salivary glands  and  enlarged hearts
 (with some myocardial scarring) were  evident in both  rats  and  mice.
Less than 1% of  the rats  in  the inhalation  experiment died  during the
exposure period,  while oral  administration of  high  doses  produced mortality
rates of 64% for male rats,  24% for  female rats,  and  3% for male and
female  mice.  Rabbits receiving the  drug  intravenously developed enlarged
salivary glands  and  enlarged hearts.   Six percent of  the  rabbits died
during  the  third week of  the trial.   In this study, the durations were
all  comparable,  but  the use  of various species and  different doses
 reduced the  reliability of the  comparisons.

-------
                                   164
     4.4.3.6  2-Methyl-4-Chlorophenoxy Acetic Acid  (MCPA) — In a subacute
dermal study, doses of 0, 0.5,  1.0, and 2.0 g MCPA  per kg of body weight
were applied to  the skin of  chinchilla rabbits in an aqueous paste
(Verschuuren, Kroes,  and Tonkelaar, 1975).  Four of four rabbits treated
with 2 g MCPA and  three of four treated with 1 g MCPA died during the
treatment  period,  possibly as  the  result of dysbacteria  (an illness
often seen in rabbits of that  institute).  Growth retardation was observed
in  animals of the  0.5-g/kg group.  However, growth  rates improved during
the recovery period.  Other  major  changes noted and attributed to dermal
application of MCPA were erythema; a  decrease in the number of lympho-
cytes in the 0.5-g/kg group; and hyperplasia, hyperkeratosis, and loss
of  elasticity of the  skin.
     In the subchronic oral  study, male and female  rats received 0, 50,
400, or 3200 ppm MCPA in their food for 90 days (Verschuuren, Kroes, and
Tonkelaar, 1975).  During the  first week's feeding  of 3200 ppm the rats
exhibited  unhealthy fur and  cold extremities.  There was no mortality
attributed to dosage.  Decreased food consumption,  decreased body weight
gain, and  increased kidney weights were characteristic of the 3200-ppm
group.  There was  also an increase in erythrocyte size and a proportional
increase in hemoglobin content in  rats of the high  dose group.
     4.4.3.7  2-Methyl-4-Chlorophenoxy Propionic Acid (MCPP) — In a sub-
acute dermal study, doses of 0,  0.5,  1.0, and 2.0 g MCPP per kg of body
weight were applied to the skin of chinchilla rabbits in an aqueous
paste (Verschuuren, Kroes, and Tonkelaar, 1975).  One rabbit of the
1.0-g/kg group and one rabbit  of the  2.0-g/kg group died.  Growth retarda-
tion was seen in all  groups, but some recovery was  observed during the
2 weeks after termination of treatment.  Erythema was observed and was
dose related.  The skin lost its elasticity, but reverted to normal
condition  during the  recovery  period.  Organ weights of the treated
animals were no  different from those  of the controls.
     In a  subchronic  oral study, male and female rats received 0, 50,
400, or 3200 ppm MCPP in their  food for 90 days (Verschuuren, Kroes, and
Tonkelaar,  1975).  Rats of both  sexes treated with  3200 ppm had unhealthy
fur, decreased hemoglobin content, decreased erythrocyte counts, and

-------
                                  165
increased alkaline phosphatase activity;  hematocrit and leucocyte values
were decreased in males only.  Some of the changes observed also occurred
with 400 ppm.  Increased kidney weights were seen in rats at levels of
3200 and 400 ppm, and depression of ovary and prostate weights was seen
with 3200-ppm feedings.
     These two studies of MCPA and MCPP are typical of many primary-
studies in the literature.  Although two  routes of exposure are used,
they differ in species, duration, and dose.  This makes them of minimal
value for comparisons.
     4.4.3.8  Nefopam Hydrochloride — Nefopam, a compound with nonnarcotic
analgesic activity, was tested for subacute toxic properties by Case,
Smith, and Nelson (1975).  Carworth CFN rats were administered the drug
by three routes:  intraperitoneally, in daily doses of 0, 2, and
10 mg/kg (5 days per week for 4 weeks); intramuscularly, in daily doses
of 0, 1, and 2 mg/kg  (7 days per week for 2 weeks); and orally, in the
diet, in daily doses of 0, 20, 40, and 80 mg/kg for 4 weeks.  Increased
liver weights were observed in female rats receiving 10 mg/kg of the
drug intraperitoneally (approximately 30% of the intravenous LDsg).
No indications of toxicity were seen in any of the other groups receiving
up to 2 mg/kg per day intramuscularly  (approximately 3.5% of the intra-
muscular LDso) or up  to 80 mg/kg per day orally  (approximately  65% of
the oral U^g),
     Mongrel dogs showed only slight weight loss after daily  intravenous
administration of 5 mg/kg  (approximately 25%  of  the  intravenous LDso)
and after daily  oral  administration of 4 and  40  to 80  mg/kg (25% to  50%
of the oral LDsg) of  nefopam.  No toxic  effects  were observed following
daily intramuscular injections of up  to  3  mg/kg  of body weight  (approxi-
mately 10% of the intramuscular LDso)  °f t*ie  drug.
     Despite using three  routes  of  exposure in both the rat and the dog,
the study by Case, Smith,  and Nelson  (1975)  still differs in dose and
duration so  that comparisons would  be difficult.
     4.4.3.9  A9-Tetrahydrocannabinol (A9-THC) — Thompson et al. (1974)
described the toxic effects  of  A^-tetrahydrocannabinol, the active
ingredient in marijuana,  when  adminstered intravenously or orally to

-------
                                    166
primates  for 28 days.  In a preliminary acute study rhesus monkeys
survived  up to 9000 mg/kg orally, while 128 mg/kg intravenously produced
100% mortality (two of two).  The subacute study consisted of male and
female monkeys, either treated with oral doses of 0, 50, 250, or
500 mg/kg per day or injected intravenously with 0, 5, 15, or 45 mg/kg
per day.   During oral administration,  two male monkeys treated  with
500 mg/kg per day and one monkey treated with 50 mg/kg became moribund
and were  sacrificed on days 10, 14,  and 16, respectively.  Deaths in the
intravenous trial caused by acute hemorrhagic pneumonia occurred only in
the 45 mg/kg per day group at days  8 and 19.  Table 4.29 summarizes the
main toxic effects observed in intravenously or orally treated  monkeys.
              Table 4.29.   Main toxic effects produced by oral or
                       intravenous administration of
                         A9-tetrahydrocannabinola
                                       Routes of administration
                                         Oral       Intravenous
                               Organ size
            Thymus                      Atrophic      Atrophic
            Pancreas                    Atrophic
            Testes                      Atrophic
            Adrenal                     Enlarged
            Liver                       Enlarged      Enlarged
            Body weight                 Decreased
                             Clinical effect
Leukocytosis
Myeloid hyperplasia
Anemia
Bromosulfophthalein
retention
Electrolyte balance
Proteinuria
Bradypnea
Anorexia
Ulcerative colitis
Other
Behavioral changes
Development of tolerance
Yes
Yes
No
No

Altered

Yes
Yes
Yes
effects
Yes
Yes
Yes

Yes
Yes


Yes
Yes
Yes


Yes
Yes
                
-------
                                  167
This study has differences in dose levels that  make  the  conclusions  of
the authors questionable.
     The preceding examples are typical of within-chemical comparisons.
Almost every comparison was complicated by test design variables.  Any
conclusions that could be drawn from these would be  tenuous at best.
However, this is unfortunately the current status of information on  this
subject.  A computer and manual search of over  30 000 titles and abstracts
produced no studies specifically designed to compare exposure routes for
subchronic toxicity tests.  Without sufficient  literature data, an
evaluation of the route-to-route extrapolations is limited to a general
discussion.
     The value of extrapolating data from one route to another is limited.
The oral route of exposure has been sufficiently developed to the point
that it is easy to perform, requires a minimum of specialized equipment,
and produces good toxicity data.  Thus, there is little need to extrapo-
late data from other routes to determine oral toxicity for most chemicals.
Injections are rarely primary routes of exposure and are relatively easy
to perform for durations of a subchronic nature.  Extrapolation from
another route is not needed here.
     Dermal and inhalation studies are the principal routes for which
extrapolation might prove useful.  Difficulty in calculating  exact  dos-
ages taken in by test animals applies to both routes.  Also the anatomy
and physiology of the entry sites for these routes differ  between the
test species and man  (especially  for inhalation), thus affecting the
predictive value of data from these two  routes.  For  inhalation, the
additional problem of special equipment  requirements  limits  the number
of  studies that can be  performed.  Extrapolation might be useful in
these  routes, if the  substituted  methods solve these problems. For
example,  an  intravenous injection could  be used to  replace an inhalation
evaluation  (at  least  for systemic toxicity)  thus reducing the problem of
limited  facilities.   Dosages  could also  be more stringently controlled.
Another  alternative route could be the oral route.   This would require
previous  testing  to ascertain if  any significant metabolic transforma-
tions  by the gastrointestinal tract occur for  that chemical.  Of course,
these  extrapolations  would not provide information on toxicity at  the

-------
                                   168
site of entry.  These data would have to be obtained from preliminary
studies (acute or subacute).
     Before any of these route extrapolations can be employed, much lab-
oratory research needs to be performed.  Specific comparisons over a
wide range of chemicals need to be carried out so that the background
data would be available.  A basis for extrapolation should be sought in
pharmacokinetic and metabolism studies that could justify the quantita-
tive and qualitative validity from one route to the other.  This research
would help to define the types of toxicants and the aspects of each
route, for which extrapolation would not be useful.

4.4.4  Conclusions
     In selecting the route of exposure for subchronic toxicity tests,
the route by which human exposure is most probable is the preferred
choice.  The routes used in most subchronic testing are oral and inhala-
tion exposures.  Oral exposure can be by diet mixtures, gavage, or
capsule, all of which are relatively simple and effective.  Inhalation
exposure requires special equipment, which results in higher costs per
test and acts as the limiting factor in determining the number of tests
that can be performed.  Occasional use is made of percutaneous and
intraperitoneal exposures, depending on the chemical, its metabolic
pattern, and the expected human exposures.
     In some cases, extrapolation of data from one route to determine
potential toxicity by another route may be feasible.  Some substitution
for dermal or inhalation studies could theoretically occur by using data
from intravenous or oral studies.  However, the literature is currently
insufficient to evaluate this area.  The lack of data in the literature
stresses the need for research on route-to-route comparisons and
extrapolations.

4.5  PATHOLOGY

4.5.1  Introduction
     Gross and microscopic pathology examinations are currently an
integral part of most toxicity tests.  Particularly for the subchronic

-------
                                    169

evaluations,  extensive organ  and tissue examination is required or

recommended.   The EPA guideline for pesticide evaluations (Federal

Register,  1978) is a good  example of this, recommending for oral  admin-

istration  the microscopic  examination of approximately 30 organs  or

tissues  from all animals at  the control and high doses.  Additionally,

any  lesions on other organs  revealed by gross examination require a

histopathological follow-up.   For intermediate  and low doses,  the tissues

to be  examined depend on the toxic effect, target organ, and lesions

found  in the gross examination or in the high-dose histology procedures.

Table  4.30 gives a typical list of tissues to be examined (Peck,  1968).



                    Table 4.30.  Postmortem studies
                                  Gross
                 External lesions      Oral tissues
                 Tumors                Voluntary muscle
                 Abdominal contents    Bones
                 Pelvic contents       Brain
                 Thoracic contents     Spinal cord
                 Cervical tissues

                              Organ weights
                             Histomorphology

                 Thytnus                Heart
                 Lymph node            Large and small
                 Thyroid                  arteries
                 Parathyroid           Lung
                 Adrenal               Stomach
                 Pancreas              Duodenum
                 Liver                Jej unum
                 Gallbladder           Ileum
                 Kidney                Colon
                 Urinary bladder       Bone  (with marrow)
                 Ovary                Marrow smears
                 Uterus                Brain
                 Testes                Pituitary
                 Prostate              Voluntary muscle
                 Seminal vesicle       Eyes
                 Mammary gland         Other  tissues  as
                                         indicated by
                                         gross  observations
                                         and  by drug  activity

                      Source:  Adapted from Peck,  1968 —  An
                 Appraisal  of Drug safety Evaluation in
                 Animals and the  Extrapolation of  Results to
                 Man.  In:  Importance of Fundamental
                 Principles in Drug Evaluation,  D.  H.  Tedeschi
                 and R.  E.  Tedeschi, eds.  Raven Press, New
                 York.  pp. 450-471.  Reprinted with permis-
                 sion of the publisher.

-------
                                   170
 Besides the general appearance and microscopic  condition of the organs,
 the organ weight change is  also monitored  during  and at the end of the
 exposure period.  An extensive, detailed protocol for  organ examination,
 removal, fixation, and interpretation is given  by Prieur et al. (1973).
 The value of such extensive examinations is  debatable, however, with
 evidence and opinions supporting both the  pro and con  arguments.  The
 purpose of this section of  the literature  review  is to give an overview
 of  opinions concerning the  use of pathology  and the relative predictive
 efficiency for each organ.

 4.5.2  Use  of  Pathology
     In  some earlier short-term tests, microscopic examination was not
 performed because  either the exposure periods were considered too short
 to  affect organ tissues  (Bratton,  1945) or the  ability of the histopatho-
 logic examination  to add quantitative information to the other observa-
 tions was doubted  (Smyth and Carpenter, 1948).  The significance of
 histopathology was considered  to increase somewhat when the results of
 the short test were to be a preliminary step for  a 2-year study (Smyth
 and Carpenter,  1948).  Smith (1950) felt that histopathology could also
 be  useful if a series of compounds were being compared or ranked for
 relative toxicities.  Even  in  these cases, the  tissues to be examined
 were few, generally restricted to  the liver, kidney, heart, and any
 lesion-bearing organs.  This trend toward reduced histopathology in
 earlier  short-term tests was continued by Smyth,  Carpenter, and Weil
 (1951) in their development of the range-finding  test.  As shown in
 Table 4.31, these  researchers  found no material that caused microscopic
 tissue damage  as the first  effect  (at the lowest  dose).  They did find
 that organ weight  could be  a significant early  indicator of toxic
 effects, particularly weight changes  in the liver and kidney.
     Barnes and Denz (1954), in a  general review  of chronic toxicity,
also expressed  reservations about  the value of  pathology, particularly
histopathology.  They identified some of the sources of variation in
pathological results  as:  the  subjective nature of the examination; the
occurrence of natural disease;  and the problems of age changes in the
test species.   They  saw little value in routine sectioning and histo-

-------
                                           Table 4.31.  Range-finding data on subacute oral toxicitya
Least dally dose causing symptom (g/kg)
Material studied
Acroleln<*
Aldol
Allyl alcohol"
l,3-Butanediol«
"Cellosolve" (2-ethoxyethanol)d'e
Dichloral urea
2,4-Dichlorophenoxy ethyl
sulfate, sodium
Diethanolaalne
(2,2'-lmlnodiethanol)e
Dl(2-ethylhexyl) adipate*
Di(2-ethylhexyl)
tetrahydrophthalate8
2-Dimethylaminoethanol
(dloethylethanolamlne)*
2 , 4-Dimethy 1-2-oethylene-
1,2,4-Chlodiazolidine thlone
Dimethyl methylene diphenol-p,p'e
Dimethyl methylene diphenol-p,p',
diglycidyl ether
Ethylene chlorhydrin
(2-chloroethanol)d
Ethylenediamlne"''
2-Ethyl-l , 3-hexanediole
2-Ethylhexanimido (diethyl-
2-ethyl hexanoate)
Glycidyl sorbate dimer
2-Hethyl-2,4-pentanedlol
Monoethanolamlne (2-amlnoethanol)e
Monolsopropanolamine8
Tetrabutyl thiodlsucclnate*
1,2,3, 4-Tet rathla-6 , 9-diazecane-
5,10-dithione
Triethanolamlne6
Maximum
dose
(g/kg)
0.0015
1.58
0.0097
5.60
1.89
5.33

0.66

0.68
4.74

2.67

0.89

0.10
0.52

0.35
0.042
0.31
0.70

1.53
0.27
0.31
2.67
2.22
10.3

0.55
2.61
Minimum
dose
(g/kg)
0.00017
0.026
0.0013
0.32
0.052
0.012

0.011

0.0051
0.16

0.053

0.045

0.0025
0.002

0.014
0.0024
0.036
0.20

0.10
0.012
0.043
0.16
0.14
0.26

0.04
0.0050
Reduced Reduced
growth appetite
0.0015
1.58
0.0097
0.74 0.74
0.18 5.33

0.66

0.68
2.92 2.92

2.67



0.10 0.10




0.31 0.12
0.70


0.27
10.3

0.55
1.27
Altered .
sf S
0.0015

0.0097
0.74 0.74
0.18 2.55

0.20

0.090 0.17
2.92 2.92

0.84

0.89

0.039



0.012
0.31 0.12


1.53
0.012 0.27
0.64 1.28
2.22
1.08 10.3

0.17
0.17 0.73
Death


0.0097
1.89
2.55

0.66

0.17
4.74










0.12



0.27
1.28
10.3


0.73
Maximum
dose Single-dose
having no oral U>so°
effect (g/kg)
(g/kg)
0.00017
0.43
0.0040
5.60J
0.21
0.048

0.047

0.020
0.61

0.19

0.18

0.010
0.52.T
t
0.35'
0.0024
0.036
0.48

0.44
0.0129
0.3V
0.32
0.60
0.26

0.04
0.08
0.046 (0.039-0.056)
2.18 (2.00-2.38)
0.064 (0.056-0.074)
22.8 (21.8-23.9)
3.00 (2.51-3.59)
32 RF

1.41 RF

1.82 (1.66-2.00)
9.11 (7.28-11.4)

114 RF

2.34 (2.26-2.42)

0.50 (0.48-0.52)
4.04 (3.73-4.38)

8.14 (7.25-9.14)
0.089 (0.067-0.117)
1.16 (0.98-1.37)
2.71 (2.52-2.93)

40 RF
5.75 (5.28-6.25)
4.76 (4.27-5.30)
2.74 (2.39-3.15)
4.26 (3.89-4.67)
100 RF

8.53 (6.12-11.9)
9.11 (8.45-9.82)
     "Materials  administered  in  the  food  to  groups  of  ten  rats  for  30  days  on  each  dose  level unless noted otherwise.
     fcThe dose producing microscopic lesions in  liver,  kidney,  spleen  or  testis of  any rat.
     "When LDSO  was derived from an  advanced test,  the  range  of 1.96 times  the standard  deviation is shown in parentheses.  The letters "RF"
refer to data derived  from a  range-finding test.
     ^The dose was administered  in the drinking water.   In  this case appetite  is judged  on the basis of mllllllters of water consumed.
     *Doses administered for  90  days.
     ^The dose shown is the maximum  that was  administered,  and  It caused no symptoms.
     sThe dose shown is the minimum  administered, and it caused some symptoms.
     Source:  Adapted  from Smyth, Carpenter,  and Weil,  1951.  Copyright 1951,  American Medical Association.  Reprinted with permission
cf the publisher.

-------
                                   172
logical examination of 20 major organs and suggested that a thorough
gross examination would suffice.  They also doubted the value of organ
weight changes as indicators of toxicity.  Barnes and Denz cited Cameron
(1952) concerning the increase in organ weights caused by hypertrophy
without true damage to the tissues.  They suggested that organ weights
be used as toxicity indicators only if histology also shows damage.
     Zbinden (1963) also questioned the practice of examining all tissues
histologically.  He felt that to reduce the extensive technical work
histology should be done only on high-dose animals and 25% to 50% of the
control animals.  Further examination would be limited to organs showing
changes.  In addition, Zbinden recommended a  grading system that ranks
histopathological findings,  thus increasing the accuracy for initial or
subtle effects.  The  role of organ weight changes is secondary when
autopsy and histology are done carefully.
      In contrast to these earlier authors, more emphasis is placed on
organ weight changes  than on histopathology as the predominant indicator
of pathological damage by Weil and McCollister  (1963) in their eval-
uation of subchronic  and chronic tests.  As shown in Table 4.32, out of
33 short-term  experiments only twice was histopathology the sole toxic
indicator at the lowest dose.  However, organ weight change was the sole
low-dose toxicity indicator  seven times.  The same trend is shown for
2-year studies,  indicating  that  histopathology  is a  poor short-term
indicator of chronic  toxicity.   Specifically, among  the organs examined
only  the liver, kidney, testes,  and spleen showed weight changes indic-
ating histologic damage.  The weight changes  of the  liver and the kidney
increased their value as toxicity predictors  in the  longer 2-year
studies.  Based on these findings, Weil and McCollister recommended that
for determination of  lowest  dose level of effect, the liver and kidney
should be examined for weight changes and histological damage.  Although
not as useful  as organ weight, histology of the liver and kidney did
increase the predictive efficiency.  These factors,  plus body weight
gain, would serve to  adequately  indicate toxicity.
      In a later paper, Weil  et al.  (1969) again commented on the value
of histopathology.  In comparing 90-day, 1-week, and single exposure
tests, they used primarily the histology and  weight  changes of the

-------
Table 4.32.  Summary of observations of effects detected in short-term and two-year oral studies
                  (At lowest dosage level in which any effect was detected)
Short-term
Criterion
of effect
Mortality
Food intake
Body weight
Organ weights
Liver
Kidney
Heart
Spleen
Testes
Lung
Brain
Thyroid
Stomach
Adrenal
Gross pathology
Micropathology
Liver
Kidney
Heart
Spleen
Testes
Lung
Adrenal
Pancreas
Bone marrow
Voluntary
muscle
Stomach
Intestine
Bladder
Brain
Sciatic nerve
Number of
studies in
which this
criterion
was
followed
33
30
33

27
27
17
17
16
15
3
0
1
0
33

30
30
22
21
21
20
16
16
1

1
3
3
2
3
2
Number of
pertinent
studies0
27
26
27

22
22
13
17
13
12
2
0
0
0
27

24
24
17
16
17
16
12
12
0

1
1
1
2
3
2
Number in
which this
criterion
was
affected
1
2
10

8
7
0
1
0
0
0
0
0
0
0

3
1
0
0
1
0
0
0
0

0
0
0
0
0
0
Number
in which
it was
the sole
effect
0
0
6

4
3
0
0
0
0
0
0
0
0
0

1
0
0
0
1
0
0
0
0

0
0
0
0
0
0
Number of
studies in
which this
criterion
was
followed
33
29
33

30
30
17
16
18
12
4
2
1
3
3

33
33
29
23
33
32
27
23
11

6
15
21
12
11
0
Two-year
Number of
pertinent
studies'2
25
21
25

22
22
13
10
12
10
3
1
0
0
25

25
25
21
16
25
25
20
17
6

3
10
14
9
8
0
Number in
which this
criterion
was
affected
2''
0
15

6
5
0
0
0
0
0
0
0
0
0

9
8
0
1
0
0
0
0
0

0
0
0
0
0
0
Number
in which
it was
the sole
effect
0
0
7

0
3
0
0
0
0
0
0
0
0
0

1
0
0
0
0
0
0
0
0

0
0
0
0
0
0
Number this
criterion
affected at
next higher
dosage level*'
Short-
term
0
3
9

4
2
0
1
1
0
0
0
0
0
0

}
2
0
0
0
0
0
0
0

0
0
0
0
0
0
Two-
year
0
1
2

1
1
0
0
0
0
0
0
0
0
0

3
2
0
0
0
0
0
0
0

0
0
0
0
0
0

-------
                                                      Table 4.32 (continued)
Short-term
Criterion
of effect
Hematology
Blood urea
nitrogen
Clinical urine
analysis
Central nervous
system
Neoplasm
Fertility
Total number
a-lcm-lf Icanf-lv nlf-m
Number of
studies in
which this
criterion
was
followed
19

10

1

1
33
0
of studies
"•oH at* anv t
Number of
pertinent
studiesa

16

7

1

1
27
0
in which this
1n*ana*j*> 1 o-iral
Number in
which this
criterion
was
affected
1

0

0

1
0
0
criterion was

Number
in which
it was
the sole
effect
0

0

0

1
0
0
followed

Number of
studies in N
criterion
was
followed
31

6

7

0
33
5
minus the studies

Two-year
lumber of
ertinent
studies0
24

3

5

0
25
3
in which

Number in
which this
criterion
was
affected
0

0

0

0
0
1
none of the

Number
in which
it was
the sole
effect
0

0

0

0
0
0
criteria were

Number this
criterion
affected at
next higher,
dosage level
Short-
term
0

0

0

0
0
0


Two-
year
0

0

0

0
0
0
•vj
-t-
      Number of times that effects were demonstrated by the criteria when they were not affected at the next lower dosage level.

      Highest level fed was the only one affected.

     Source:  Adapted from Weil and McCollister, 1963.  Reprinted with permission of the Journal of Agricultural and Food Chemistry.
Copyright by the American Chemical Society.

-------
                                   175
kidney and liver as indicators of toxic dosages.   However, for the 90-day
studies, they examined additional organs for toxic changes.  The results
from the additional organs were less sensitive than the other criteria
selected, and they again suggested that histopathology be limited to
certain organs such as the liver, kidney, or spleen.
     Peck (1968), in his discussion of pathology in drug evaluations,
questioned the value of organ weight changes without a histopathological
examination as a follow-up.  He stated that, when organ weight changes
are reflective of toxic effect, histopathology will also show results.
But if histopathology is negative, the organ weight change is most
likely not due to the toxic action of the drug.  Therefore, to be sure
that the effect indicated by organ weight change is a result of the
tested substance, a histopathology examination is necessary.  Peck also
stated that histopathology is useful in determining the significance of
spontaneous species-specific lesions.  If the commonly occurring lesions
for a species are altered in time of appearance,  number, character, or
severity, this could be due to the activity of the drug.  However, as a
caution to the use of histopathology, Peck stressed the difficulty in
interpreting results (particularly for electron microscopy) and in
extrapolating results from the test species to man.
     Benitz (1970), in his review of chronic toxicity, also stressed  the
need for a complete histopathological examination.  He emphasized that
to achieve the maximum results from pathology  techniques,  a thorough
gross necropsy is the critical starting procedure.  If the organs are
excised improperly the extent of  later microscopic examination  can be
irrelevant.  This includes, of course, all  organs, because in his
opinion restricting histology  to  organs with gross lesions significantly
decreases the determination of complete  toxicity.  He also rejects the
procedures of examining only the  liver and  kidney  since morphological
effects or target organs  for new  drugs are  unknown; of  examining  only
the major organs because  this  is  too  imprecise and varies with  the
pathologist's definition  of major;  and of examining organs with gross
lesions and all  organs  from a  few representative test animals as  this
may not yield the optimum amount  of morphological  data.   Benitz does
allow for deviations  from histopathological examination of all  organs if

-------
                                   176
previous studies for that chemical indicate certain toxicity patterns or
if results are needed only for certain organ systems.  He stressed,
however, that all organ systems have demonstrated toxic effects from
chemicals, as is shown in Table 4.33.  These effects can be characterized
as follows:  76.5% were degenerative changes, 14% were inflammatory
lesions, 7% were circulatory disorders, and 1.8% were neoplastic, precan-
cerous lesions.  Benitz generally recommended weighing of the organs as
           Table  4.33.   Incidence (in percent)  of  drug-induced
             pathological changes encountered over a ten-year
                 period in approximately  14  000 animals
System and organ
Cardiovascular system
Respiratory system
Alimentary system
Liver
Gastrointestinal tract
Pancreas
Urinary system
Kidney
Ureter and urinary bladder
Reproductive system
Males
Females
Ductless glands
Thyroid
Adrenal
Pituitary
Islets of Langerhans
Hematopoietic system
Lymphoid and reticuloendothelial
system
Central nervous system
Sensory system
Eye
Ear
Locomotor system
Skin

Incidence
By organ By system


15.8
3.8
1.0
12.0
1.0
4.8
2.9
11.5
4.8
5.3
0.5



1.4
0.5


4.3
5.3
20.6
13.0
7.7
22.1
3.3
7.2
3.3
1.9
10.5
1.0
           Source:  Adapted from Benitz, 1970.  Reprinted with
      permission of the publisher.

-------
                                  177
an indicator of toxicity.  However,  he felt that  use of  relative organ
weights could be misleading, and if  they are used the absolute organ and
body weights should also be given.   This eliminates  any  uncertainty
regarding whether the change is caused by reduced body weight or actual
organ weight change.
     Benitz, in his discussion of the significance of organ weight
changes, deals with the question raised by Peck (1968) concerning how
often organ weight changes correspond with positive  histopathology
findings.  Benitz cited Jackson and Cappiello (1964) and their conclu-
sion that in beagles 80% of the organ weight changes correlates with
histopathological damage.  In assessing the extent of histological
damage, Benitz discussed both the role of electron microscopy and the
quantification of morphological changes.  He felt electron microscopy is
useful  for:  (1) determining the earlier stages of morphological changes,
(2) better interpreting visible lesion damage, and  (3) detecting ultra-
structural changes not visible through normal techniques.  However,
because of the time and  effort involved, the use of electron microscopy
must be adequately justified in each instance.  Quantification  of
morphological changes is of value because results are more precise  and
comparable.  Table 4.34  describes several generally used quantification
methods.  As can be discerned from his  lengthy treatment of histopathology,
Benitz  felt it is highly important to the determination of  toxicity.
Although  some of this may be due to his consideration of chronic as well
as  subchronic tests, the principles discussed apply equally to  both
topics.
     Changes in body weight can  often give misleading results for relative
organ weight change.  This  can  lead  to  errors  in assessing toxic effects.
Stevens (1976, 1977) examined  this  problem and proposed a  simple modifica-
tion of the standard relative weight determination.  By obtaining initial
and terminal body  and organ weights  for untreated test  animals, Stevens
constructed a  simple linear regression of body weight and  organ weight.
This regression  is used to  predict  the expected  mean organ weight for a
test group based on their mean body weight.  Comparisons can be made
between the expected and the  actual relative body weights.  Differences

-------
                                        178
        Table 4.34.  Summary of histological quantification methods and their
                     applications for chemically induced lesions
   Procedure
   Histological substrate
                                                                Comments
Counts
Linear
  measurements
Area measurements
Nuclear volume
Composition of
  organs
Nuclear density
Internal surface
  measurements
Bile plugs, giant cells, mast
cells, calcification sites,
mitoses, etc.

Thinning of femoral epi-
physeal disc; hyperplasia of
zona glomerulosa in adrenal
cortex; reduction in height
of spermatogenic epithelium;
decrease of tubular diameter
of testes

Muscles:  necrosis
Uterus:  atrophy of mucosa
and myometrium
Brain:  hydrocephalus

Hyperplasia of thyroid gland;
dystrophy of adrenal cortex;
atrophy of liver due to
starvation

Hyperplasia of thyroid gland:
epithelium colloid, open
capillaries, etc.
Atrophy of ovaries: ratio of
primary follicles to corpora
lutea
Cirrhosis of liver: density
of reticulum fibers
Composition of submaxillary
glands:  ratio of mucous to
serous terminal portions

Dystrophy in zona fascicu-
lata of adrenal cortex

Alveolar surface in lungs
altered by inflammation,
fibrosis, or emphysema.
Trabecular surface in femoral
epiphysis altered by osteo-
dystrophy
Expressed as average number
per field; total number per
section, etc.

Calibrated micrometer,
results in micrometer units
or microns
Components expressed in
planimeter values obtained
from tracings or projections
Two perpendicular measure-
ments using computerized
formula of rotation ellip-
soid for calculating v3
                                                       Point sampling,
                                                       Components expressed in
                                                       percentage of organ volumes
Line sampling
Results expressed in
mm*/mm3 of organ volume
     ^Por methodology and examples see Hennig, 1959, and Benitz and Dambach, 1964.

     Source:  Adapted from Benitz, 1970.  Reprinted with permission of the
publisher.

-------
                                  179
indicate the actual toxic effect.   By analyzing simulated results from
rat organ weights, Stevens was able to test this modification and found
it to be a significant improvement in relative organ weight data.
     Several recent committee reviews of toxicity tests have discussed
procedures for pathology examinations.   The National Academy of Sciences
review (1977) suggested that as a  minimum all organs of animals at the
high dose and controls should be examined microscopically.   Further
tissue examination at other dose levels should be based on gross obser-
vations and results from the minimal microscopic examinations.   The Food
Safety Council (1978) also recommends this procedure, with additional
emphasis on gross examination.
     The review by the World Health Organization (1978) is more exten-
sive in its discussions.  It generally recommends the procedures endorsed
by the National Academy of Sciences and Food Safety Council, but points
out more hazards in data evaluation.  In interpreting the data, the
pathologist should be aware of the rate of occurrence for spontaneous
lesions and allow for these in his examination.  Also organ weight
changes should be evaluated carefully to determine if they resulted from
the toxin or as a consequence of stress phenomena or metabolic overloading.
Finally, they emphasize that microscopic examinations should be performed
only after all data from the gross examination, organ weight determina-
tions, and biochemical-hematological tests have been properly interpreted.
These data will help direct the microscopic examination and reduce
redundant efforts.
     The U.S. Environmental Protection Agency  (1979), in a support
document for chronic toxicity testing guidelines, follows the recommenda-
tions of the National Cancer Institute (Sontag, Page, and Saffiotti,
1976), which suggested microscopic examination of all test animals  (both
control and treatment groups) in chronic studies.  For subchronic studies,
Sontag, Page, and Saffiotti  (1976) suggested that all animals from  the
control, highest-dose, and next highest-dose levels be subjected to a
histopathologic examination covering 30 to 40  tissues plus  tissues with
gross lesions.  They feel the results obtained with  this added  pathology
are worth the increased time and costs, which  can account for 40% of  the

-------
                                  180
overall study cost (Page, 1977).  This represents the maximum recommended
microscopic examination recorded in the literature.

4.5.3  Basis for a Minimum Pathology Screen
     The value of histopathology in subchronic tests is related to the
sensitivity of each organ to different chemicals.  If certain organs
consistently show toxic effects in subchronic tests, they should be
routinely included in most histopathologic examinations.  These organs
could comprise a minimum, basic pathology screen to which other organs
could be added depending on the suspected toxic effects from the test
chemical.
     To aid in evaluating this concept, a rough tabulation of organ
testing frequency and frequency of positive findings was constructed
from 54 subchronic studies (Table 4.35).  These subchronic studies
tested a wide range of chemicals (Appendix B), including various drugs,
herbicides, pesticides, fungicides, industrial chemicals and solvents,
food additives, resins, and natural substances.  The cumulative data for
each organ are given in this table, combining three species (dog, rat,
and monkey) and three routes (oral, inhalation, and injection).
Appendixes B and C contain specific information on routes of exposure,
test species, chemicals used, and literature references for each organ
in Table 4.35.  The table gives an indication of how often each organ is
examined and how informative it is in subchronic studies.  The "total
studies" column refers to the number of studies in the literature that
were examined for these data.  The "studies using organ" column refers
to the actual number of studies for which that specific organ was examined
for pathologic changes.  The "positive results" column refers to the
number of studies in which pathologic changes were found in that organ.
The "percent used" column refers to the number of studies using that
organ as a toxicity indicator, out of the total number of studies reviewed.
The "percent positive" column refers to the percentage of studies finding
pathological effects in an organ out of the total number of studies
using change in that organ as a criteria of toxicity.  For example, the
brain was evaluated for pathologic changes in 46 of 54 studies for a

-------
                                   181
 Table 4.35.   Comparison of the usefulness  of  Individual organs  to  indicate
         toxicity when pathologically examined in a subchronic study
Organa
Adrenal
Aorta
Bone
Bone marrow
Brain
Esophagus
Eye
Gallbladder
Heart
Kidney
Large intestine
Liver
Lung
Lymph node
Mammary gland
Muscle tissue
Nerve tissue
Ovaries
Pancreas
Pituitary
Prostrate
Salivary gland
Sciatic nerve
Skin
Small intestine
Spinal cord
Spleen
Stomach
Testes
Thymus
Thyroid
Trachea
Urinary bladder
Uterus
Total ,
studies
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
54
Studies
using
organ5
46
17
13
22
46
18
14
9
53
53
38
54
50
31
8
22
15
36
36
26
19
26
4
11
39
16
49
38
40
24
36
18
33
20
Positive
results1^
13
0
1
2
7
0
0
1
8
29
3
28
15
5
0
0
6
6
1
1
3
2
1
1
1
4
12
3
12
6
5
4
0
1
Percent
used6
85
31
24
41
85
33
26
17
98
98
70
100
93
57
15
41
28
67
67
48
35
48
7
20
72
30
91
70
74
44
67
33
61
37
Percent
positive-'
28
0
8
9
15
0
0
11
15
55
8
52
30
16
0
0
40
17
3
4
16
8
25
9
3
25
24
8
30
25
14
22
0
5
      Appendices B and C contain specific information for each organ con-
cerning test species, routes of exposure, chemicals tested, and literature
references.

      Total number of studies reviewed for the inclusion of an organ as an
indicator of pathologic changes.
     Q
      Total number of studies in which an organ was used to assess patho-
logic changes.

     lumber of studies in which pathologic changes were detected in an
organ.
     O
      Frequency that an organ was used as a pathologic indicator
(b/a x 100).
     f
     •* Frequency that positive pathologic changes were found in an organ
(c/b x 100).

-------
                                  182
frequency of use percentage of 85.  However, out of the 46 studies
utilizing the brain in this manner only 7 showed positive pathological
data.  Thus, the frequency of positive results for the brain was 15%.
With the relative  criteria of a 20% use frequency and a 5% positive
result frequency,  a minimum screen would consist of the following
organs:  adrenal,  bone, bone marrow, brain, heart, kidney, large
intestine,  liver,  lung, lymph node, nerve tissue, ovaries or testes,
prostate, salivary gland, skin, spinal cord, spleen, stomach, thymus,
thyroid, trachea,  and uterus.  This minimum screen does include repre-
sentative organs from all of the major systems, except for sensory  (e.g.,
eye) and would  seem to be a logical start for composing a pathology
evaluation.

4.5.4  Conclusions
     In reviewing  the literature on the use of pathology, the wide
variety of  protocols is striking.  In few cases"do the discussions agree
concerning  the  amount and type of pathology necessary.  The opinions
regarding the value of gross examination are perhaps the closest to a
consensus.  Most reviews recommend gross examination of all organs at
all dose levels (Barnes and Denz, 1954; National Academy of Sciences,
1977; Peck, 1968;  World Health Organization, 1978), although the defini-
tion of all organs is often not specified.  This procedure would seem to
be the best option, as time and cost factors are not excessive and the
information gained can be substantial (Food Safety Council, 1978).
     The recommendations for microscopic examination are not as simple
or similar as for  the gross examination.  Initially the importance of
microscopic examination for subchronic tests was rated low, with few
tissues examined (Smyth and Carpenter, 1948; Smyth, Carpenter, and Weil,
1951; Weil and  McCollister, 1963; Weil et al., 1969).  As toxicity tests
became more extensive, the role of microscopic observations became
greater.  Many  reviewers recommend microscopic examination of selected
tissues (25 or  less) at the high- and control-dose levels (Barnes and
Denz, 1954; Zbinden, 1963).  These tissues are usually major organs and
lesion-bearing  organs observed in gross examination.  Other researchers

-------
                                  183
believe that all organs (30 to 40) should be sampled at high and control
doses to adequately determine toxicity (Benitz,  1970;  Food Safety
Council, 1978; National Academy of Sciences, 1977;  World Health Organi-
zation, 1978).  Yet other researchers believe that  this is still insuf-
ficient and recommend examination of all tissues at all levels (Sontag,
Page, and Saffiotti, 1976; U.S. Environmental Protection Agency, 1979).
This last protocol is obviously the most sensitive  in detecting toxicity,
but it is also the most costly.  The data in Table  4.35 and the recom-
mendations of other researchers also indicate that  the examination of
all tissues at two dose levels can be excessive, because some tissues
are poor indicators of toxicity.
     The majority of researchers agree that a highly efficient protocol,
in terms of toxicity detection per time and personnel cost, is the
microscopic examination of specified organs and tissues at the high-
and control-dose levels with added examination of grossly visible lesion-
bearing organs.  The exact number and type of tissues to be examined
would vary with the chemical and  its suspected target organs, but,
at the least, every organ system  should be represented.
     The value of organ weight determinations is another  controversial
aspect of pathology.  Although on occasion changes  in organ weights may
not indicate specific toxicity, since they may reflect problems  such as
functional hypertrophy or metabolic  overloading, they may still  provide
information valuable in a toxicological  assessment.  Furthermore, many
researchers feel that it  can be more signifcant  than histopathology.
Since  the procedures for  determining organ weight  (relative or absolute)
are relatively easy, the  most  efficient  protocol would include them.
Even if considered  insufficient  alone as toxicity  measures, changes  in
organ weight  can indicate the  general trends and suggest specific organs
for microscopic examination.
     Thus, the data presented  in the literature surveyed indicate that
an efficient, yet relatively inexpensive, pathology protocol would
consist of a  gross  examination of all tissues and  organs at all dose
levels, a microscopic  examination of designated tissues at the high and
control levels, organ weights  for major organs, and supplemental

-------
                                  184
microscopic examination of tissues at other dose levels based on the
gross observations and the organ weight data.

4.6  CLINICAL LABORATORY TESTS

4.6.1  Introduction
     Recent developments in automation, methodology refinement, and data
interpretation have greatly increased the predictive role of biochemical
tests on blood, urine, and enzyme systems.  Almost all current testing
schemes include some biochemical tests requiring at least pre- and post-
treatment sample evaluation.  If more thorough toxic effect data are
required, interim sacrifices or periodic sample removals are needed to
provide sufficient data for evaluation.  Despite automation, this increases
time and cost factors in subchronic toxicity tests.  The EPA guidelines
for pesticide evaluations (Federal Register^ 1978) recommend the following
31 biochemical tests:  hematocrit, hemoglobin, erythrocyte count, total
and differential leukocyte counts, platelet count, reticulocyte count,
serum calcium, serum potassium, serum lactate dehydrogenase (LDH),
SGPT, SCOT, glucose, blood urea nitrogen, direct and total bilirubin,
serum alkaline phosphatase, total cholesterol, albumin, globulin, total
protein, serum chlorine, uric acid, blood creatinine, urine specific
gravity, urine pH, urine protein, urine glucose, urine ketones, urine
bilirubin, and urobilinogen.  In addition to these, some toxicologists
also use the following determinations to assess toxic effects:  ornithine
carbamyl transferase, creatinine phosphokinase, lipase, amylase, sodium,
prothrombin time, serum thyroxine, cholinesterase activities, packed
cell volume, mean corpuscle hemoglobin, mean corpuscle volume, glucose-
6-phosphatase, hexobarbital oxidase, methemoglobin, and many others.
All of these tests are used to enhance early detection of toxicity or to
help pinpoint toxic mechanisms.  However, the actual value of this large
number of tests has been discussed infrequently in the literature.  The
purpose of this section is to summarize the available literature on the%
use of hematological, biochemical, enzymatic, and urinalysis tests in
subchronic tests and to present some data on the predictive efficiencies
of these tests.

-------
                                  185
4.6.2  General Clinical Testing
     Weil and McCollister (1963),  in their comparison of  short-  and
long-term feeding studies, evaluated the efficiency of clinical  uri-
nalysis, blood urea nitrogen, and  hematology as  primary indicators of
toxic effects.  As shown in Table  4.32,  of 24 pertinent studies  using
these criteria only one showed any effects.   Furthermore, the effect
noticed was not at the lowest toxic dose, indicating that it contributed
only secondary toxicity information.  They concluded that efficient
detection of toxicity did not need these types of tests for experiments
of a subchronic nature.
     Peck (1968), in reviewing the state of drug safety evaluations,
commented on the value of biochemical, hematologic, and function tests.
He cited the Coggeshall Report of the Committee on Drug Safety (1964),
attributing the origin of biochemical toxicity tests as beginning with
disease evaluation in man.  These tests, originally designed for human
systems, were incorporated in evaluations of animal toxicity effects,
with an assumption that the tests would give comparable results in both
man and test species.  Both the Coggeshall report and Peck questioned
the original validity of  this assumption, especially without verifying
studies.  Peck also stated that the  interpretation of data from these
tests needs improvement and refining.
     The use of tests dealing with  changes in organ  function or enzymatic
activities for drug safety evaluations without  supporting pathological
data is especially questionable.  Here Peck's opinion is essentially the
same as his opinion on using organ  weight changes  without correlating
histopathology.   In both  situations, it  is necessary to  utilize data
from several methods of toxicity  assessment  if  the evaluation is to be
conclusive.  Peck felt  that  experience  has  shown that the tests are
generally valid for man and  the test species, but any toxicity study
should  consider the test  development history in making evaluations or
recommendations.
     The occurrence of abnormal results is  also a problem in the interpre-
tation  of biochemical  or  function tests.  Average values within two
standard deviations and a knowledge of  past aberrant biochemical test

-------
                                   186
values  for  the species under study are essential to a proper evaluation
of data.  This will help eliminate concern over spontaneous abnormal
values.   If these  factors are considered and compensated for, then the
use  of  biochemical, hematologic, and  function tests can be valuable in
determining toxicity.
     Benitz (1970), in a review of chronic toxicity, discussed the value
of clinical chemistry, hematology, and serum drug levels in evaluating
toxicity.   In contrast to some authors, he stated that a large battery
of tests  is not  always better than a  smaller set.  If the small set is
constructed of tests better suited to evaluate the chemical's toxicity,
as indicated in  short-term tests, the results will be more meaningful.
Use  of  interim function and clinical  tests is helpful primarily in
aiding  the  postmortem evaluation, because these tests indicate the sites
of toxic  damages.  Benitz also suggested the use of tests similar to
those used  in humans, with the stipulation that they be equally effective.
This agrees in principle with Peck's  (1968) experiences in the use of
human tests in experimental species.  As an aid in evaluating the test
results,  Benitz  suggested that baseline studies be done prior to testing
rather  than using  "normal" values from the literature or other laboratories.
Another important  role of the pretest baseline examination is to determine
the  health  status  of the test animal, which allows the researcher to
remove  any  potentially questionable subjects before treatment starts.
     In assessing  hematological data, one should be aware of the varia-
bility  between samples taken from different sites in the vascular system
and  of  differences in reliability between methods.  In addition to
numerical data, morphological or qualitative data can aid in assessing
toxicity.   Serum levels of drugs are  most useful for determining toxic
dose level  and for comparison with levels to be used in man.  By assess-
ing  the serum level at which toxic effects occur, comparisons between
human clinical trials and animal toxicity studies are facilitated.  In
addition, Benitz warned against a rigid predetermined schedule of
sampling.  He stated that "these observations should be made when nec-
essary."  "When necessary" is determined during the trial by toxic
effects from the high dose and from pretest short-term studies.  To set

-------
                                  187

up an inflexible schedule of sampling  only invites  unnecessary evaluations
and a waste of time and money.   In conclusion,  Benitz  supported Peck's
(1968) association of pathology evaluations and clinical-hematologic
tests, stating that toxic effects are  not always indicated  only by
morphological damage or only by functional impairment.
     McNamara (1976) briefly discussed hematology and  biochemical tests,
indicating that he supported their inclusion in a test design.  He
listed these tests as minimal requirements:  blood  urea nitrogen; sugar,
glutamic oxalacetic transaminase; alkaline phosphatase; glucose-6-
phosphatase; cholinesterase; lactic acid dehydrogenase; potassium
(albumin, globulin); sodium; calcium;  chloride; carbon dioxide; hema-
tocrit; hemoglobin; total and differential white blood cells; and
erythrocytes.  He also suggested that these tests be performed at expo-
sure days -14, -7, +3, +7, +30, +60, and +90.  This schedule allows for
establishment of pretreatment baseline values and ensures a sufficient
number of determinations to detect most transient changes.
     The National Academy of Sciences  (1977) review on toxicity testing
briefly discussed biochemical and hematological examinations.  These
investigators suggested selected use of clinical tests based  on prior
test data or chemical class.  They agreed with Benitz  (1970)  in recom-
mending that a rigid schedule of sampling  is unnecessary; a  flexible
schedule that can be modified by the health status of  the test  animals
is more practical.  They suggested using  large  animals (nonrodent)  for
most  tests, especially organ function  tests or  special clinical examina-
tions  (e.g., x rays or respiratory rate  determinations).  They did  not
recommend any specific tests,  suggesting that  this be  left  to the
investigators.
      The Food Safety Council  (1978) stated that biochemical and hematolog-
ical  examinations  should be a  part  of  every study, with the degree  of
emphasis dependent on  the  substance.   They also recommended a flexible
sampling schedule,  with  shorter intervals between  samples  when the
toxicity potential is  unknown  or unpredictable. They did  not recommend
any  specific  set of tests  but  suggested a variety  of  tests including
some  biochemical,  hematological, urine,  and fecal  evaluations.  However,

-------
                                   188
in contrast to other reviews, they did not recommend baseline studies
but suggested that historic colony values are acceptable.  They also
suggested that the final protocol be determined by the investigator.
     In addition to these general comments on clinical tests, there are
comments in the literature concerning specific hematology, organ/enzyme
function, and urinalysis evaluations, which are discussed in the following
sections.

4.6.3  Hematology
     Zbinden (1963) discussed in his review of drug toxicity the various
tests used for evaluations of hematological toxicity.  For evaluation of
total hematological effect, he recommended starting with a group of
routine tests (hemoglobin, hematocrit, total red blood cells, total
leucocytes, and differential leucocytes).  As the study progresses,
special tests (reticulocyte counts, Heinz-Ehrlich bodies, methemoglobin,
sulfhemoglobin, thrombocyte count, and sedimentation rate) are added
when indicated.  The tests should be performed at monthly intervals,
evaluating some animals from all dose levels for nonrodents and from the
high-dose level for rodents.
     Saslaw and Carlisle  (1969) reviewed their toxicity data on various
chemicals and drugs in which they utilized nonhuman primates (primarily
rhesus monkeys) and monitored hematology parameters.  They determined
that for hematology effects the monkey was a good animal model for
predicting effects in humans.  Also,  they discussed the value of
hematology tests in determining general toxicity.  In their opinion,
hematology should be included in toxicity tests, as the laboratory work
is not too excessive and  "this effort could possibly be rewarded with
observations that could be meaningful to groups or agencies faced with
the awesome task of predicting or assessing hematoxicity of the increas-
ingly complex preparations intended for eventual use in man."  However,
few definitive studies in support of  this statement are included in the
discussion.  Saslaw and Carlisle also commented on the need for pre-
liminary baseline studies for each specimen to be used in the toxicity
tests.   This would supply significant pretreatment "normal" values for

-------
                                  189
the blood factors,  which would allow easier assessment of changes
occurring during treatment.   The use of  "mean average count" data  is not
as efficient as the individual baseline  data, and  its use should be
discouraged.
     Bushby (1970)  evaluated the role of hematological tests in toxicity
studies.  In subchronic studies, the essential hematological tests
should detect anemia and alterations in  leucocytes and platelets.   If
these changes are observed,  more testing should  be performed to deter-
mine the causes.  Bushby broke these two types of  hematology tests
(detection of effects tests  and site of  action tests) into  the three
groups listed in Table 4.36.
     To detect anemia, Bushby recommended a hemoglobin count,  a red
blood cell count, and a determination of the  packed cell volume.   All
three tests are interrelated, and a change in any  of the three would
indicate a possible anemia.   Determination of the  packed cell  volume
is easier and more reliable  than the red blood  cell count,  so  it  is
preferred.  Additional tests that can pinpoint  the type  of anemia or
site of action include mean  corpuscular hemoglobin concentration, mean
corpuscular hemoglobin, circulating reticulocyte numbers, bilirubin,  and
methemoglobin.  For detecting changes caused by organic disease,  Bushby
recommended use of the sedimentation rate of the red blood cell count,
an easily performed procedure.
     For detection of alterations in the leucocytes and platelets,
Bushby suggested a direct count of  their numbers.  An examination of  a
stained blood film will supply  information on any  gross morphological
changes in platelets and leucocytes.  The type  and number of  leucocytes
present can also be determined with the  film.   This differential
leucocyte count should always be converted to absolute  figures using  the
total leucocyte numbers.  Bushby did not recommend a routine  test for
the determination of hemorrhagic disorders.  If changes are indicated by
clinical evidence or platelet counts, the prothrombin and partial
thromboplastin  times should be  determined.  Bushby concluded  that in
most prolonged  studies a few hematological parameter evaluations  will
detect general  blood damage but, if site of action information is desired,
more extensive  and different tests  are  necessary.

-------
                             190

  Table 4.36.  Hematological studies during toxicity tests


                            Group

Primary tests and estimations

  Hemoglobin estimation
  Packed cell volume
  Total and differential leucocyte count
  Platelet count
  Examination of stained film for polychromasia and abnormal
    leucocytes and platelets
  Sedimentation rate

Secondary tests

  Red cell count
  Calculation of mean corpuscular volume, mean corpuscular
    hemoglobin, and mean corpuscular hemoglobin
    concentration
  Reticulocyte count
  Detection of Heinz bodies
  Examination for methemoglobin
  Hemoglobinemia
  Bilirubinemia
  Bone marrow
  Coagulation time
  Bleeding time
  Clot retraction
  Prothrombin time
  Partial thromboplastin (cephalin) test
  Fragility test

Follow-up investigations

  Schumm's test for methemoglobin
  Coproporphyrins I and III in urine
  Thromboplastin generation test
  Platelet adhesion
  Antiglobulin test for autoantibodies
  Tourniquet or vacuum test
  Osmotic fragility
  FIGLU test
  Folic acid and B12 deficiencies
  Estimation of life span of red cell
  Iron metabolism
    Ferrioxamine test
    Serum iron estimation
    Serum iron-binding capacity
    Siderocytes in bone marrow
  Estimation of haptoglobin
  Blood volume

     Source:  Adapted from Bushby, 1970.  Reprinted with
permission of the publisher.

-------
                                  191
     Arnold et al. (1977) commented on hematological procedures in their
discussion of animal health monitoring in chronic studies.   Generally,
they recommended testing on a preset schedule or in response to changes
in animal health.  They also recommended using,  but did not specify, a
basic test set with incorporation of additional  tests when any of these
conditions are observed:  obvious clinical anemia; preliminary conditions
of anemia (e.g., vaginal bleeding or hematuria); enlargement of the
spleen, liver, or lymph nodes; and signs of physical trauma.
     The monitoring of hematological effects should begin with a gross
observation of the blood sample to detect changes such as altered color
or transparency.  Next, standard erythrocyte and leucocyte evaluations
should be performed.  Arnold et al. stressed the need for interrelated
and overlapping evaluations, especially the correlation of data from
cell counts and blood smear examinations.  Similarly, differential
leucocyte counts must be translated into absolute numbers and given with
the total leucocyte counts.  To screen for hemorrhagic effects, they
recommended the use of prothrombin time (one stage) and partial thrombo-
plastin time  (activated).  If additional information is needed, the
platelets should be counted and assessed for morphological changes, and
bleeding time should be determined.  If these procedures are followed,
hematology can be a valuable tool in determining the type and degree of
toxicity.
     Another review that provides some detailed comments on hematogical
data is a World Health Organization  (1978) report on evaluations of
chemical toxicity.  These investigators recommended sampling at 30-day
intervals with subgroups of rodent and nonrodent  test  species.  They
stated that to adequately assess hematological parameters both quantifi-
cation and morphology of the blood cells are needed.   They  recommended  a
standard protocol that included the  determination of erythrocyte numbers,
reticulocyte numbers, total and differential leucocyte numbers, hema-
tocrit, and hemoglobin.  The erythrocytes must  also be examined for
morphological changes.  For evaluating hemorrhagic effects, a  screen
should consist of platelet counts, clot retraction, prothrombin time
(one stage),  and partial thromboplastin time  (activiated).  A  more

-------
                                  192
detailed evaluation of hemorrhagic effects includes factor assays,
thrombin time, fibrinogin determination, platelet aggregation, pro-
thrombin consumption time, and euglobulin clot lysis time.

4.6.4  Biochemical and Organ Function
     Wr6blewski and LaDue (1955) evaluated SCOT as an index of liver
cell injury.  They primarily examined human patients with varying types
of liver diseases, but also included some rat studies.  Among the aims
of their paper was to discover if the degree of SCOT activity was
related to liver cell destruction; whether SCOT activity in chronic
liver disorders indicated active liver cell destruction; what correla-
tion existed between SCOT activity and various tests of liver dysfunc-
tion; and whether changes in SCOT reflected the presence of liver
cancers.  After evaluating their data, Wrdblewski and LaDue reached the
following conclusions:  a rough relationship exists between the amount
of a toxin in the animal and the degree of elevation of SCOT; the SCOT
activity does not alter at the same rate or in the same direction as
other liver function tests; SCOT appears to be an index of liver cell
injury and not liver cell function changes; and an increase in SCOT
activity indicates the presence of liver metastases.  Based on this
report, SCOT determinations appear to be a useful index of liver damage.
     In addition to discussing hematology tests, Zbinden  (1963) reviewed
the use of clinical chemistry and organ function tests.  The tests he
listed as commonly utilized included SCOT; SGPT; serum lactate dehydro-
genase; blood urea nitrogen; blood glucose; serum cholesterol; alkaline
phosphatase; thymol turbidity; serum bilirubin; bromosulfalein excretion;
nonprotein nitrogen; and phenolsulfonphthalein.  However, he had reserva-
tions about their usefulness, stating, "Despite the widespread use of
these tests, there is still not enough information about their signifi-
cance in toxicology."  Liver function tests are slightly more reliable
than most of the tests and usually give results in the pathological
range that correlate well with the degree of organ damage.  However, not
all degenerative changes that occur in the liver are detected.  Thus,
the significance of these tests should not be overrated either.  Their

-------
                                  193
use should obviously be as a confirming or indicator test.   Zbinden also
stressed the usefulness of drug blood level determinations.   These allow
more reliable extrapolation of "safe" dose levels from test  species to
man.
     Hoe and O'Shea (1965) evaluated the predictive efficiency of bio-
chemistry tests for detection of kidney disease in the dog.   They used
known kidney lesion-producing chemicals to assess the degree of correla-
tion between biochemical tests and histopathology.  The biochemical
tests evaluated included blood urea; blood creatinine; inorganic phosphate;
serum protein; calcium; serum alkaline phosphatase; sodium;  potassium;
chlorine; cholesterol; SGPT; and SCOT.  Each was tested in dogs after
exposure to the chemicals.  The results of the tests for calcium,
chlorine, and serum proteins did not correlate with kidney damage and
were of no predictive value.  The evaluations of alkaline phosphatase,
SCOT, and SGPT were inconclusive regarding kidney damage due to exces-
sive interference resulting from liver damage.  However, the other tests
did correlate with kidney damage.  Blood urea increased in a parallel
fashion with kidney damage.  However, the authors warned that other
researchers (Bloom, 1954) have found that urea values do not always cor-
relate in cases of nephritis.  Creatinine also indicated kidney damage
but not as reliably as did urea changes.  Inorganic phosphate showed a
correlation, as did sodium and potassium.  But Hoe and O'Shea cautioned
that interpretation of these tests is complex, and careful evaluation is
necessary.  In conclusion, the authors suggested  that by combining the
biochemical tests and using urinalysis a forecast of  90% accuracy can
be achieved for kidney damage.  They suggested that general protocols
cannot be developed and that every case or chemical should be assessed
on an individual basis.
     Peck et al. (1967) discussed several aspects of  the interpretation
of serum enzyme tests.  They warn against assuming that a change  in  the
test value automatically indicates a toxic effect.  The change  could be
indicative only of reversible pharmacologic or physiologic actions of
the test chemical.  To properly assess the significance of the  changes,
one should consider the magnitude of the change,  its  incidence  rate  in

-------
                                  194
the test groups, the presence or absence of a dose-response pattern,
the results after continued repeated exposure, and the results of
pharmacology and pathology evaluations.  Another problem to be aware of
is the potential deleterious loss of blood with repeated sampling.  To
prevent this, special studies or additional sample groups should be
used, rather than jeopardizing the main study by oversampling.
     Street (1970) reviewed biochemical tests in toxicology and stated
that their primary functions are as early indicators of toxicity and for
identification of target organs and evaluation of reversibility of
changes.  Most of the techniques employed are modified from human tests
and can be done on an automated basis.  For proper evaluation of the
results, initial values and values from concurrent control groups are
needed for comparison.  The initial values should be taken twice during
the acclimation period.  Normal values published in the literature are
of limited value because of high variability.  Every toxicologist should
establish the "normal" values at his own laboratory using his methods
and species.  These can then be used to aid in evaluation.
     The usefulness and shortcomings of individual tests are discussed,
and Street recommended analysis techniques for each one.  For liver
damage, he suggested the use of SCOT, SGFT, and alkaline phosphatase
evaluations.  Additional tests that can add information on other effects
besides liver damage include bilirubin and protein electrophoresis.
Street also recommended the use of isocitric dehydrogenase in place of
the dye tests, unless the dye tests can be designed to give full elimina-
tion curves.  For the assessment of kidney damage, he recommended the
blood urea test correlated with urinalysis results.  Other tests reviewed
include electrolyte determinations, blood lipids, and cholinesterase.
In conclusion, Street cautioned that biochemical test results usually
suggest the use of additional confirmatory tests and in any case their
findings should be checked against histopathology for correspondence of
effects.
     Cornish (1971) reviewed the literature associated with the use of
serum enzyme changes as indicators of toxic effects.  Cornish did not
limit his discussion to tests that indicate liver and kidney damage, but

-------
                                  195
also reviewed tests that may indicate heart or lung damage.   In general,
he refrained from recommending specific tests for each organ, preferring
to emphasize the advantages and disadvantages found by researchers for
each test.  Several factors of test design are repeatedly stressed in
his discussion.  A principal factor is the need for frequent sampling or
sampling during the periods when the changes in serum enzyme levels
would be most .probable.  If sampling is done at inappropriate times,
many of the changes may be missed, since the toxicant stimulates a
short-term elevation of serum levels.  Despite its often transient
nature, this immediate response can be a sensitive indicator of func-
tional and/or morphological damage.  Although this might require a
predosing determination of enzyme level patterns or a more frequent
sampling schedule after dosing, the enzyme tests require only small
samples of serum and can eliminate the need for the more costly interim
sacrifices of test animals used to determine the onset of organ damage.
Also, the toxicologist should consider other factors, such as stress and
alterations in dietary intake, that can affect the serum enzyme levels
regardless of any organ damage.
     In addition to the usual enzyme tests, Cornish also discussed
the potential value of serum isoenzyme determinations.  Isoenzymes
usually have a distinctive distribution pattern for each organ, and the
pattern found in the blood after a chemical insult can indicate damage
to individual organs.  Because most of the other enzyme tests are less
specific concerning the organ damaged, Cornish feels  that the use of
isoenzyme determinations has a great potential in  toxicology.   In addi-
tion to pinpointing damage to specific organs, isoenzyme patterns can
often be more sensitive indicators of overall tissue  damage  than other
enzyme tests.  However, as with serum enzyme tests, the schedule of
sampling for isoenzymes is important, because the  pattern can change
with time and alter the resulting  diagnosis.
     Grice et al.  (1971) compared  the sensitivity  of  changes in SCOT,
lactate dehydrogenase  (LDH), and LDH isozyme patterns with histopathology
to determine their usefulness in detecting  liver and  kidney  damage.
After exposure of  four toxic chemicals in rats,  the  test results  showed

-------
                                   196
 that morphological damage occurs at lower doses and before any change in
 the serum enzyme levels.  The SCOT test was somewhat more sensitive than
 LDH, but lacked the specificity of the isozyme pattern.  Grice et al.
 concluded that the serum enzyme levels and isozyme patterns could
 contribute  supplemental information, but they are not substitutes for
 pathology evaluations.
     Korsrud et al. (1973) also used histopathology to assess the sensi-
 tivity of changes in several serum enzymes to indicate liver toxicity.
 They used acute doses of three toxic chemicals and monitored the changes
 in isocitrate dehydrogenase (ICD), fructose-1-P aldose (F-l-P ALD),
 sorbitol dehydrogenase  (SDH), glutamic-pyruvic transaminase  (GPT),
 glutamic-oxaloacetic transminase (GOT), malic dehydrogenase  (MDH), LDH,
 glutamic dehydrogenase  (GDH), fructose-l,6-diphosphate aldolase  (F-1,6-
 diP ALD), and ornithine carbamyl transferase  (OCT) in rats.  As shown in
 Table 4.37, histopathology was repeatedly the most sensitive index.  Of
 the enzyme  tests, SDH was the most sensitive.  The next most reliable
 indicators  were ICD, GPT, GOT, and F-l-P ALD.  Korsrud et al. identified
 several factors that could account for the differences in sensitivity,
 such as different intracellular locations of the enzymes in  the liver,
 high control values for LDH, different release rates into the blood for
 each enzyme, different  turnover rates for the enzymes, and the enzyme
 activity in the cell prior to release.  The responses were also
 dependent on the time of sampling.  Korsrud et al. concluded that SDH
 was the best choice for assessing minimal liver damage when  using serum
 enzyme levels.
     Gray et al. (1972), in an evaluation of the toxicity of clindamycin
 in dogs, discussed the value of serum transaminase tests.  In particular,
 they concentrated on the predictive value of changes in SGPT.  They felt
 that SGPT changes were not so much an indication of actual liver damage
 as an indication of liver intolerance.  They based this on the role of
 SGPT in the transport efficiency of the membrane.  They concluded that
no definite morphological effect could be related to SGPT changes;
therefore, SGPT could not be used as a predictor of liver damage.
     Arnold et al. (1977) briefly commented on the usefulness of serum
enzyme determinations.  They cautioned that the removal of blood and the

-------
                                   197
              Table 4.37.   Relative  sensitivity of  liver
                             enzyme  tests
                               Lowest  dose at which significant
                                       change occurred
             Testa                         (mg/kg)
TA^ DMN*2 DBA
ICD
F-l-P ALD
SDH
GPT
GOT
MDH
LDH
GDH
OCT
F-l, 6-diP ALD
Histopathology
25.4
25.4
9.4
25.4
25.4
68.6
68.6
NCe
68.6
NC
3.5
13.7
13.7
5.1
13.7
13.7
37.0
37.0
NC
37.0
NC
1.9
800
800
800
800
800
800
1600
1600
1600
1600
400
               ICD — isocitrate dehydrogenase;  F-l-P ALD —
         fructose-1-P-aldolase; SDH — sorbitol  dehydrogenase;
         GPT — glutamic-pyruvic transaminase; GOT — glutamic-
         oxaloacetic transaminase;  MDH — malic  dehydrogenase;
         LDH — lactic dehydrogenase;  GDH — glutamic dehydro-
         genase;  OCT — ornithine carbamyl transferase;  F-l,
         6-diP ALD — fructose-l,-diphosphate aldolase.
               TA — thioacetamide.
               DMN — dimethylnitrosamine.
               uEA — diethanolamine.
               No change.
              Source:  Adapted from Korsrud et  al., 1973.
trauma associated with it can significantly affect the test animal's
health.  Therefore, serum enzyme tests should be performed only when
necessary.  Also, many of the tests fail to detect minor damage or may
reflect only transient changes.  As a result of these disadvantages,
the use of biochemical tests is limited to a confirmatory role.
     These same disadvantages and cautions are also voiced by the World
Health Organization (1978).  They suggested the use of large test spe-
cies, such as the dog, to reduce sample removal effects.  They recom-
mended a set sampling schedule with serum withdrawal prior to the test,

-------
                                   198
 3 and 10 days after the start  of  dosing,  and  continuing  at  30-day
 intervals.   They concluded that,  in  general,  these  tests cannot  detect
 minor or initial changes in organ function but  can  act as a guide  for
 further assessments.
      Clampitt (1978)  investigated the predictive value of various  enzyme
 tests for liver damage in the  rat.   He compared these results with
 histopathological evaluations  to  determine their effectiveness.  He
 found that  no single test gave an unquestionable indication of liver
 damage.  Many of the conventional liver damage  tests were not the  most
 sensitive indicators of damage.   These include  alkaline  phosphatase,
 alanine transaminase, and aspartate  transaminase.  Clampitt did  recom-
 mend that to assess lipid metabolism changes, plasma cholesterol and
 triglyceride levels should be  monitored.   Also, the time at which  blood
 samples are taken should be carefully monitored if  transient effects  are
 desired.  In conclusion, Clampitt suggested that minimal changes can  be
 detected if the "appropriate plasma  constituents" are measured and used
 with knowledge of the "intracellular location of the diagnostic  enzymes."

 4.6.5  Urinalysis
      Balazs et al. (1963) discussed  the value of urinalysis in assessing
 renal damage.  In general the  authors believed  that although kidney
 function tests are not easily  adapted to routine toxicity studies  using
 the rat, they are potentially  very useful.  Their data  suggested that
 the proximal convoluted tubules were the most vulnerable part of the
 kidney.  Their tests were therefore  designed to evaluate damage  in this
area.   They found that the renal  tests that provide the  most reliable
information were the  Addis count,  urinary glutamic  oxaloacetic trans-
aminase test (UGOT),  and urinary  concentration  test.  Blood urea nitrogen
was erratic and less  reliable  than the UGOT test because of extrarenal
interference.   Also,  the standard concentration tests were  inappropriate,
and the authors suggested using a 6-h determination.  This  allowed minor
effects to  be detected.   Additional  evidence  of renal damage can be
obtained from observation of renal epithelial cells and  casts in the
urine.

-------
                                  199
     Hoe and O'Shea (1965)  evaluated biochemical  analysis  of renal dam-
age in the dog, including the value  of  urinalysis techniques.   They rec-
ommended the use of urine specific gravity,  proteinuria, and the presence
of casts in the urine to assess renal damage.   As with their recommenda-
tions on enzyme tests, Hoe and O'Shea believed a  combination of urinalysis
and function tests was necessary for a  complete assessment of renal
damage.  In this way, 90% of the cases  could be diagnosed  by biochemical
tests alone.
     Urinalysis and its diagnostic value were  discussed by Street (1970).
The use of the simple tablet or dipstick analyses techniques in any
capacity greater than a preliminary  screen is  questioned by Street.  He
feels that the more complex tests add significant information, especially
for renal and hepatic damage.  The recommended tests include observations
on the color and volume, pH (by meter,  not paper), specific gravity,
protein electrophoresis, examination of deposits, and 16-h concentration
tests (in the dog).  Simple screen tests can also be performed to check
for glucose, ketones, free hemoglobin,  bile, and reducing substances.
Unlike Balazs et al.  (1963), Street did not recommend the UGOT and Addis
counts as standard tests.  The specific gravity and concentration tests
are sufficient to detect most renal damage.  Urinary ascorbic acid can
be used to test for liver enlargement, but it requires a. series of
comparative assays.  Overall, Street recommended  the inclusion of urin-
alysis tests for a complete assessment of toxicity.
     Grice  (1972) reviewed some aspects of urinalysis, particularly
tests derived  from human renal assessments.   In  general he  feels  the
tests to be used on small laboratory animals have not reached  a  similar
level of sophistication and are not as valuable  as for humans.   A major
problem, besides the  interpretation of test results, is obtaining a
suitable sample.  Collection is difficult, since  the test animals  "will
not urinate on command" and contamination with feces or other  materials
is hard to prevent.  He also discusses other  techniques to  evaluate
renal damage,  such as renal cell  excretion rates  (useful  for  assessing
the degree of  injury) and palpation of the kidney (often  more  sensitive
than either urinalysis or urinary enzyme tests).  However,  he  concludes
that presently the tests are less sensitive and  less reliable  than
histopathology.

-------
                                    200

      Wright and Plummer (1974)  evaluated the excretion rate of  four
 urinary enzymes (lactate dehydrogenase,  alkaline phosphatase, acid
 phosphatase, and glutamate dehydrogenase)  as indicators of  acute kidney
 damage by administering several known nephrotoxic agents.   The  enzymes
 were selected for their utility as "markers" for renal injury to specific
 regions in the cell,  and the changes were compared to baseline  excretion
 rates.  They found that lactate dehydrogenase and alkaline  phosphatase
 were excreted at significantly  higher rates  after renal insult,  and
 these increases seemed to correlate with renal damage.  The other two
 enzymes were not appreciably affected.   They concluded that excretion
 rates of some enzymes could effectively  detect acute renal  damage.
      Cottrell et al.  (1976) conducted similar studies on the value of
 excretion rates of creatinine,  alkaline  phosphatase, lactic dehydro-
 genase, and leucine aminopeptidase after acute and repeated-dose expo-
 sure to nephrotoxicants.  They  used both histology and histochemistry to
 evaluate the renal damage.  They found that  for acute exposure,  all the
 enzymes but creatinine were affected, tactic dehydrogenase was the most
 sensitive indicator.   However,  the excretion pattern after  repeated
 exposure to the nephrotoxicants did not  continue to indicate the
 increasing renal damage shown by histology and returned to  normal rates
 after three days.  Therefore, they concluded that currently the value of
 urinary excretion rates is confined to acute and very short-term expo-
 sures.  Their utility in subchronic tests would be quite limited.
      Berndt (1976) reviewed the tests necessary for confirmation of the
 kidney as a target organ.  In addition to recommending the  hematology
 tests and blood urea nitrogen and plasma creatinine tests,  Berndt recom-
 mended a urinalysis determination.  Specifically he stressed the use of
 changes in urinary glucose levels as a good  indicator of renal  damage.
 Also,  the excretion of p-aminohipparate  (PAH) and tetraethylammonia
 (TEA)  can be detected in the urine to indicate damage.  These tests are
 particularly good for evaluating the toxic effect on the nephrons.
 Other  recommended tests include the volume and concentration determina-
 tions.   Protein determinations  will indicate the effects on the glomerulus.
 Berndt  concluded that,  in comparison with  urinalysis,  histological and
 anatomical  techniques are the "least useful  criterion for evaluation of
renal damage."

-------
                                  201

     Arnold et al.  (1977)  also discussed the use  of  urinalysis in tox-
icity diagnosis.   They suggested collection of  urine overnight or at
24-h interims for determinations of  volume and  the excretion patterns of
calcium, phosphate, creatinine, chloride,  sodium,  and potassium.   Fresh
samples were needed, however,  for pH,  osmolarity,  and the presence of
malignant cells.   As a routine examination, the authors  suggested the
use of strip tests covering pH, protein, glucose,  ketones, blood, and
bilirubin.  Depending on the results,  more specific  tests or more com-
plex analysis could be incorporated, including  microscopic evaluations
of urine sediments.  These urinalysis  tests will  elucidate renal
problems such as anuria, hematuria,  or hypertrophic  changes.  Thus, the
authors recommended the inclusion of at least preliminary examination of
the urine for all toxicity assessments.
     A recent review of renal function tests evaluated the effectiveness
of changes in glomerular filtration rate (GFR), renal plasma  (RPF) and
red blood flow (RBF), total urinary protein excretion, urinary concentra-
tion, urinary acidification, urinary enzyme excretion rates, urinary
sodium excretion rate, and microscopic examination of sediments as
indicators of nephrotoxic effects (Digzi and Biollaz, 1979).  The
literature they reviewed indicated that for GFR determinations urea and
creatinine excretion rates were poor tests, and clearance rates  for
marker substances were preferred; RBF and  RPF were extremely  difficult
to interpret, especially for rodents; total protein and  urinary  concen-
tration can be useful screens but do not give  site-specific information;
urinary enzyme excretion has been shown useful but because  of a  lack  of
correlation with increases  in  renal damage,  its utility  is  uncertain;
and sodium excretion and urinary sedimentation are useful only as
screening tools.   They  concluded the predictive value for human  toxicity
of these elaborate tests is uncertain  and  their routine  use is not
suggested.  Also,  the difficulty of sample collection and interpretation
of normal variations in values makes the  general  use of  these tests less
practical.  However, these  tests can be used for  more detailed informa-
tion when prior studies indicate renal damage  or  when the structure of
the chemical  to be investigated suggests  possible renal  damage.
     U.S. Environmental Protection  Agency investigators, in a support
document for  chronic  tests  (1979),  stressed the  value of urinalysis.

-------
                                   202

 They recommended a routine screening (semiquantitative) with  a more
 detailed follow-up for positive results.   They  suggested  the  use  of
 urinalysis to detect early renal damage,  as  most  enzyme function  tests
 do not adequately assess this.   Although  the EPA  document evaluated
 chronic test applications, the  principles should  also  apply to sub-
 chronic applications.

 4.6.6  Conclusions
      Biochemical tests have become a large part of  the modern toxicity
 evaluation.   In general, they provide early  indications of the presence
 of toxic effects and suggest specific target organs or contribute con-
 firmatory evidence on the toxic effect.   The wide variety of  tests
 available has presented the toxicologist  with the problem of  test selec-
 tion.  Most of the literature reviewed did not  suggest a  standard list
 of required biochemical tests,  but rather suggested the inclusion of
 hematology,  enzyme, and urinalysis evaluations.  This  is  perhaps  a good
 policy because the effectiveness and applicability  of  the tests change
 with the toxicologist (particularly his experience  with these tests) and
 the chemical to be examined.
      For hematology evaluations  there was  general agreement that  a set
 of  basic hematology tests should provide  information on cellular  damage
 and hemorrhagic effects.   To properly  evaluate  these parameters,  several
 lists of tests  were suggested.   Most schemes used a small routine set of
 tests with more complex evaluations  added as indicated by the results.
 The routine  tests included morphological  and quantitative evaluations of
 erythrocytes and leucocytes plus a temporal  evaluation of hemorrhagic
 effects.   The scheme in Table 4.36 is  representative of this.
      The question of baseline studies  or  normal values for comparison
was  discussed in terms  of hematology tests,  although it also  applies to
enzyme function tests.   Most reviews suggested  that for nonrodent
species,  baseline data  compiled  before  treatment were  more valuable.  A
pretreatment  examination also indicates the  health  status of  the  test
animals,  confirming their suitability  for test  use.  The  use  of normal
values was less  acceptable because of variations between  individual  test
animals and between laboratories.  If normal values are to be used,  they

-------
                                  203

should be developed at the individual laboratory.   The  same applies to
the use of historic colony values for rat  studies.   The use of baseline
or normal values for comparison would be in addition to the use of
concurrent properly designed control groups.
     The sampling schedule was another area of discussion.   Many reviews
recommended sampling at 30-day intervals with initial samples at 1 day
and 2 weeks.  Others stress the value of flexible  schedules adaptable to
changes observed in the animals' health.  The best solution would be an
initial schedule of routine tests performed on a 30-day basis that is
flexible enough to incorporate additional  tests as the  study progresses.
The effect of sampling is greater for rodents than for  nonrodents,
because of size and blood volume considerations.  A recent paper by
Cardy and Warner (1979) discussed the effect of sequential bleeding on
rats with the removal of 1 mL of blood every 2 weeks over a 23-week
testing period.  They found that the hematological values were not
significantly altered but that body weight gain was significantly
reduced on this sampling regime.  Thus, if frequent sequential samples
are taken, consideration must be given to their effect on all parameters.
     Enzyme and organ function tests are usually also performed when
hematology samples are taken with the emphasis on detecting liver and
kidney damage.  The literature recommends  several procedures for asses-
sing these kinds of damage.  Liver tests usually recommended include
SCOT, SGPT, SDH, and alkaline phosphatase, with the emphasis varying
among species and chemicals.  Kidney evaluations were primarily by blood
urea and creatinine tests associated with urinalysis results.  As
indicated by several reviews, these results should be interpreted  care-
fully and only used in connection with histopathological follow-up.
     The value of urinalysis is not quite as accepted as hematology or
enzyme tests.  The National Academy of Sciences (1977) saw  little  value
in routine urinalysis tests.  The use of urinary enzyme excretion
changes is particularly limited  for subchronic  studies.  However,  other
reviews suggested the use of at  least a dipstick check of  the urine as  a
preliminary screen.  Based on evidence  in several  articles,  it  seems
that detailed assessments of kidney damage are  best  evaluated by  using

-------
                                   204
urinalysis tests and a few enzyme determinations.  If used, the urin-
alysis examination should check physical (e.g., volume and specific
gravity), enzymatic (e.g., lactic dehydrogenase), and chemical (e.g.,
glucose and phosphate) parameters.  However, because of problems with
sample collection and interpretation of test results, the routine use of
urinalysis is questionable and should be left to the discretion of the
investigator.
     Thus, the value of hematology, enzyme function, and urinalysis
techniques is apparently sufficient to recommend that they be included
in standard subchronic toxicity protocols.  The extent that each is
utilized should be left up to the investigator.  Perhaps the best
utilization of the biochemical evaluations would be in conjunction with
the terminal pathology examination.  With refinement, the biochemical
tests might suggest target organs and types of effects that could be
checked by pathology.  The effectiveness would thus increase for both
procedures.

-------
                                  205

                       REFERENCES FOR SECTION 4


Abrams, W. B., G. Zbinden, and R. Bagdon.   1965.   Investigative Methods
  in Clinical Toxicology.  J.  New Drugs 5:199-207.

Alumot, E., E. Nachtomi, E. Mandel,  and P-  Holstein.   1976.  Tolerance
  and Acceptable Daily Intake of Chlorinated Fumigants in the Rat Diet.
  Food Cosmet. Toxicol. 14:105-110.

Ambrose, A. M., P. S. Larson,  J. F.  Borzelleca, and G. R. Hennigar, Jr.
  1972.  Toxicologic Studies on 3',4'-Dichloropropionanilide.  Toxicol.
  Appl. Pharmacol. 23:650-659.

Anderson, M. E., R. A. Jones,  R. G.  Mehl,  T. A. Hill, L. Kurlansik, and
  L. J. Jenkins, Jr. 1977.  The Inhalation Toxicity of Sulfolane (Tetra-
  hydrothiophene-1,1,-Dioxide).  Toxicol.   Appl.  Pharmacol.  40:463-472.

Ansbacher, S., W. C. Corwin, and B.G.H. Thomas.  1942.  Toxicity of Mena-
  dione, Menadiol, and Esters.  J. Pharmacol. Exp.  Ther. 75:111-121.

Arnold, D. L., S. M. Charbonneau, Z. Z. Zawidzka, and H. C. Grice.  1977.
  Monitoring Animal Health During Chronic Toxicity Studies.  J. Environ.
  Pathol. Toxicol. 1:227-239.

Atkinson, R. M., J. D. Caisey, J. P. Currie, T. R.  Middleton, D.A.H. Pratt,
  H. M. Sharpe, and E. G. Tomich.  1966.  Subacute Toxicity of Cephalori-
  dine to Various Species.  Toxicol. Appl. Pharmacol. 8:407-428.

Aviado, D. M.  1978.  Overwhelming Similarities and Minimal Differences
  in Toxic Responses of Rats and Dogs  (Part I:  1974-1978  and Part  II:
  1966-1974)  (unpublished).

Balazs, T.  1976.  Assessment of  the Value  of  Systematic Toxicity  Studies
  in Experimental Animals.  In:   Advances in Modern  Toxicology, Vol.  1,
  Part 1:  New Concepts  in Safety Evaluation.  M. A.  Mehlman, R. E.  Shapiro,
  and H.  Blumenthal, eds.  John Wiley  and Sons, New  York,  pp. 141-153.

Balazs, T., A. Hatch, Z.  Zawidzka, and H. C. Grice.   1963.  Renal  Tests
  in Toxicity Studies on Rats.   Toxicol. Appl. Pharmacol.  5:661-674.

Barnes, J. M., and F. A.  Denz.   1954.   Experimental  Methods  Used in Deter-
  mining  Chronic Toxicity.  Pharmcol.  Rev.  6:191-242.

Bein, H.  J.   1963.   Rational  and Irrational Numbers  in  Toxicology.   Proc.
  Eur. Soc.  Study Drug  Toxic.  2:15-26.

Bencko, V., V. Dvorak,  and K.  Symon.   1973.   Organ Retention of Parenter-
  ally Administered  Arsenic  (Labelled  with  7lfAs) in  Mice Preliminarily
  Exposed to  the Element in Drinking Water:  A Study in Arsenic Tolerance.
  J. Hyg.  Epidemiol. Microbiol.  Immunol.  (Prague)  17:165-168.

-------
                                  206

Bencko, V., and K. Symon.  1969.  Dynamics of Arsenic Cumulation in Hair-
  less Mice After Peroral Administration.  J. Hyg. Epidemiol. Microbiol.
  Immunol. (Prague) 13:248-253.

Bencko, V., and K. Symon.  1970.  The Cumulation Dynamics in Some Tissue
  of Hairless Mice Inhaling Arsenic.  Atmos. Environ. 4:157-161.

Benitz, K. F.  1970.  Measurement of Chronic Toxicity.  In:  Methods in
  Toxicology, G. E. Paget, ed.  F. A. Davis Company, Philadelphia.
  pp. 82-131.

Benitz, K. F., and Dambach, G.  1964.  Quantification of Drug-Induced
  Morphologic Changes with Hennig's Point- and Line-Sampling Methods
  Using Zeiss Integrating Eyepieces I and II.  Arzneimittel-Fovsdh.
  14:929-935.

Benitz, K. F., G.K.S. Roberts, and A. Yusa.  1967.  Morphologic Effects of
  Minocycline in Laboratory Animals.  Toxicol. Appl. Pharmacol. 11:150-170.

Berndt, W. 0.  1976.  Renal Function Tests:  What Do They Mean?  A Review
  of Renal Anatomy, Biochemistry, and Physiology.  Environ. Health
  Perspect.  15:55-71.

Bloom, F.  1954.  Pathology of the Dog and Cat.  American Veterinary Pub-
  lications, Inc., Evanston, Illinois.

Boyd, E. M.  1961.  Toxicological Studies.  J. New Drugs 1:104-109.

Boyd, E. M.  1968.  Predictive Drug Toxicity:  Assessment of Drug Safety
  Before Human Use.  Can. Med. Assoc. J. 98:278-293.

Bratton, A. C., Jr.  1945.  A Short-Term Chronic Toxicity Test Employing
  Mice.  J. Pharmacol. Exp. Ther. 85:111-118.

Bushby, S.R.M.  1970.  Haematological Studies During Toxicity Tests.  In:
  Methods in Toxicology, G. E. Paget, ed.  F. A. Davis Company, Phila-
  delphia,  pp. 338-371.

Bushby, S.R.M., P. Lechat, and R. Santarato.  1966.  Haematological Inves-
  tigations in the Toxicity Testing of Drugs.  Results of a Questionnaire
  Circulated to Members  of the European  Society for the Study of Drug
  Toxicity.  Proc. Eur.  Soc. Study Drug  Toxic. 7:208-215.

Cameron, G. R.  1952.  Pathology of the  Cell.  Oliver and Boyd, Ltd.,
  Edinburgh.  120 pp.

Cardy, R. H., and J. W.  Warner.  1979.   Effect of Sequential Bleeding on
  Body Weight Gain in Rats.  Lab. Anim.  Sci. 29(2):179-181.

Case, M. T., J.  K. Smith, and R. A. Nelson.  1975.  Reproductive, Acute
  and Subacute Toxicity  Studies with Nefopam in Laboratory Animals.
  Toxicol. Appl. Pharmacol. 33:46-51.

-------
                                  207

Cavender, F. L.,  W.  H.  Steinhagen, C. E.  Ulrich,  W.  M.  Busey, B. Y.
  Cockrell, J. K. Haseman, M.  D.  Hogan,  and R.  T. Drew.  1977.  Effects
  in Rats and Guinea Pigs of Short-Term Exposures to Sulfuric Acid Mist,
  Ozone, and Their Combination.   J. Toxicol. Environ. Health 3:521-533.

Clampitt, R. B.  1978.   An Investigation into the Value of Some Clinical
  Biochemical Tests in the Detection of Minimal Changes in Liver Morphol-
  ogy and Function in the Rat.  Arch. Toxicol.  Suppl. 1:1-13.

Coggeshall, L. T.  1964.  Report of the Commission on Drug Safety.  Library
  of Congress Catalog Number 64-16404, Washington, D.C.  228 pp.

Cornish, H. H.  1971.  Problems Posed by Observations of Serum Enzyme
  Changes in Toxicology.  Crit.  Rev. Toxicol. 1(1):1-32.

Cottrell, R. C.,  C. E.  Agrelo, S. D. Gangolli, and P. Grasso.  1976.
  Histochemical and Biochemical Studies of Chemically Induced Acute Kidney
  Damage in the Rat.  Food Cosmet. Toxicol. 14:593-598.

Cummings, E. G.,  R. Armstrong, R. L. Farrand, and B. P. McNamara.  1979.
  Physiological and Behavioral Methodology for Evaluating Chemical Effects
  on Unanesthetized Rats.  U.S. Army Armament Research and Development
  Command, Technical Report ARCSL-TR-79015.  33 pp.

Davey, D. G.   1964.  The Study of  the Toxicity of a Potential Drug:
  Basic Principles.  Eur. Soc. Study Drug Toxic. Suppl. 6:1-13.

Digzi, J., and J. Biollaz.  1979.  Renal Function Tests in Experimental
  Toxicity Studies.  Pharmac. Ther.  5:135-145.

Earl, F. L., B. E. Melveger,  J. E. Reinwall, G.  W. Bierbower, and
  J. M. Curtis.  1971.  Diazinon  Toxicity — Comparative Studies in Dogs
  and Miniature Swine.  Toxicol.  Appl. Pharmacol. 18:285-295.

Earl, F. L., A. S. Tegeris, G. E.  Whitmore,  R. Morison, and  0.  G. Fitzhugh.
  1964.  The Use of  Swine in  Drug Toxicity  Studies.  Ann. N.Y.  Acad.  Sci.
  111:671-688.

Fancher, 0. E.   1978.   Species Selection and Animal  Models  for  Toxicologic
  Studies.  Clin. Toxicol.  12(2):239-247.

Federal Register.   1978.  Proposed Guidelines  for Registering Pesticides
  in the U.S.  Hazard Evaluation:   Humans and Domestic  Animals.   U.S.  Envi-
  ronmental Protection Agency [40 CFR Part  163]  43(163):37336-37403.

Feron, V.  J.,  A. Kruysse, H.  P.  Til, and H. R.  Immel.   1978.  Repeated
  Exposure to Acrolein Vapour:   Subacute'Studies in Hamsters, Rats, and
  Rabbits.  Toxicology 9:47-57.

-------
                                   208

Food and Agriculture Organization and World Health Organization Expert
  Committee on Food Additives.  1958.  Procedures for the Testing of
  Intentional Food Additives to Establish Their Safety for Use.  WHO
  Tech. Rep. Ser. No. 144.  19 pp.

Food Safety Council.  1978.  Proposed System for Food Safety Assessment.
  Columbia, Maryland.  136 pp.

Freireich, E. J., E. A. Gehan, D. P. Rail, L. H. Schmidt, and H. E. Skipper.
  1966.  Quantitative Comparison of Toxicity of Anticancer Agents in Mouse,
  Rat, Hamster, Dog, Monkey, and Man.  Cancer Chemother. Rep. 50(4):219-244.

Friedman, L., J. Sage, and E. M. Blendermann.  1970.  Growth and Liver
  Response of Chicks and Rats to Carbon Tetrachloride and Ethanol.  Poult.
  Sci. 49(1):298-309.

Gage, J. C.  1970.  The Subacute Inhalation Toxicity of 109 Industrial
  Chemicals.  Br. J. Ind. Med. 27:1-18.

Gaunt, I. F., G. Feuer, F. A. Fairweather, and D. Gilbert.  1965.   Liver
  Response Tests.  IV.  Application to Short-Term Feeding Studies with
  Butylated Hydroxytoluene (BHT) and Butylated Hydroxyanisole  (BHA).
  Food Cosmet. Toxicol. 3:433-443.

Gehring, P. J., V. K. Rowe, and S. B. McCollister.  1973.  Toxicology:
  Cost/Time.  Food Cosmet. Toxicol. 11:1097-1110.

Gray, J. E., R. N. Weaver, J. A. Bollert, and E. S. Feenstra.  1972.  The
  Oral Toxicity of Clindamycin in Laboratory Animals.  Toxicol. Appl.
  Pharmacol. 21:516-531.

Gray, T.J.B., K. R. Butterworth, I. F. Gaunt, P. Grasso, and S. D.  Gangolli.
  1977.  Short-Term Toxicity Study of Di-(2-Ethylhexyl) Phthalate in Rats.
  Food Cosmet. Toxicol. 15:389-399.

Grice, H. C.  1972.  The Changing Role of Pathology in Modern Safety
  Evaluation.  Crit. Rev. Toxicol. 1(1): 119-152.

Grice, H. C., M. L. Barth, H. H. Cornish, G. V. Foster, and R. H. Gray.
  1971.  Correlation Between Serum Enzymes, Isoenzyme Patterns and  Histo-
  logically Detectable Organ Damage.  Food Cosmet. Toxicol. 9:847-855.

Guarino, A. M.  1979.  Pharmacologic and Toxicologic Studies of Anticancer
  Drugs:  Of Sharks, Mice, and Men (and Dogs and Monkeys).  Methods Cancer
  Res. 17:93-174.

Hagan, E. C., W. H. Hansen, 0. G. Fitzhugh, P. M. Jenner, W. I. Jones, J. M.
  Taylor, E. L. Long, A. A. Nelson, and J. B. Brouwer.  1967.  Food Flavor-
  ings and Compounds of Related Structure.  II.  Subacute and Chronic Tox-
  icity.  Food Cosmet. Toxicol. 5:141-157.

-------
                                  209

Hall, D. E., P. Austin, and F.  A.  Fairweather.   1966.   Acute (Mouse and
  Rat) and Short-Term (Rat) Toxicity Studies on Dibutyl (Diethylene Glycol
  Bisphthalate).  Food Cosmet.  Toxicol.  4:383-388.

Hals, E., G. Bjorlin, and K. Jacobsen.   1973.  Histopathological Changes
  in Rat Incisors in Experimental Carbon Tetrachloride Intoxication.
  Odontol. Revy 24:5-26.

Harris, M. W., J. A. Moore, J.  G.  Vos,  and B. N. Gupta.  1973.  General
  Biological Effects of TCDD in Laboratory Animals.  Environ. Health
  Perspect.  5:101-109.

Hartnagel, R. E., B. M. Phillips, P. J.  Kraus, R. L. Kowalski, and
  E. H. Fonesca.  1975.  A Subchronic Study of the Toxicity of an Orally
  Administered Benzoquinolizinyl Derivative in the Rat and Dog.  Tox-
  icology 4:215-222.

Hayes, W. J., Jr.  1967a.  Toxicity of Pesticides to Man:  Risks from
  Present Levels.  Proc. R. Soc., B. 167:101-127.

Hayes, W. J., Jr.  1967Z?.  The 90-Dose LD50 and a Chronicity Factor as
  Measures' of Toxicity.  Toxicol. Appl. Pharmacol. 11:327-335.

Hayes, W. J., Jr.  1972.  Tests for Detecting and Measuring Long-Term
  Toxicity.  Essays Toxicol. 3:65-77.
                                                1
Hayes, W. J., Jr.  1975.  Toxicology of Pesticides.  Williams  and Wilkins
  Co., Baltimore.  580 pp.

Hebold, G.   1972.  Guidelines for the Testing of Drugs in Various Countries.
  In:  World Congress  of Anatomic and Clinical  Pathology, M.  Nordman,
  G. Menten, and H. Lommel, eds.  Elsevier,  New York.   pp.  145-159.

Banning, A.  1959.  A  Critical Survey of  Volume and Surface Measurements
  in Microscopy.  Zeiss-Werkz.  6:3-12.

Hodge, H. C., A. M. Boyce,  W. B. Deichmanne, and H. F. Kraybill.   1967.
  Toxicology and No-Effect Levels of Aldrin and Dieldrin.   Toxicol. Appl.
  Pharmacol. 10:613-644.

Hodge, H. C., W. L. Downs,  B.  S. Fanner,  D.  W.  Smith,  E. A. Maynard, J. W.
  Clayton,  Jr.,  and R.  C.  Rhodes.   1967.   Oral Toxicity and Metabolism of
  Diuron  (^-(3,4-Dichlorophenyl)^l,^'-dimethylurea) in Rats and Dogs.
  Food  Cosmet.  Toxicol.  5:513-531.

Hoe, C. M.,  and J. D.  O'Shea.   1965.  The Correlation of Biochemistry and
  Histopathology in Kidney Disease  in  the-Dog.   Vet.  Rec.  77(8):210-217.

Homan,  E. R. 1972.   Quantitative  Relationships Between Toxic Doses of
  Antitumor Chemotherapeutic Agents in  Animals and Man.  Cancer Chemother.
  Rep., Part 3.   3(1):13-19.

-------
                                  210

Hyslop, F., E. D. Palmes, W. C. Alford, A. R. Monaco, and L. T. Fairhall.
  1943.  The Toxicology of Beryllium.  National Institutes of Health
  Bull. No. 181.  56 pp.

Jackson, B., and V. P. Cappiello.  1964.  Ranges of Normal Organ Weights
  of Dogs.  Toxicol. Appl. Pharmacol. 6:664-668.

Jones, R. A., J. A. Strickland, and J. Siegel.  1972.  Toxicity of Propy-
  lene Glycol 1,2-Dinitrate in Experimental Animals.  Toxicol. Appl.
  Pharmacol. 22:128-137.

Kast, A., Y. Tsunenari, M. Honma, J. Nishikawa, T. Shibata, and M. Torii.
  1975a.  Acute, Subacute, and Chronic Toxicity Studies of the Beta-
  Sympathomimetic, Fenoterol HBr on Rats, Mice, and Rabbits.  Oyo Yakuri.
  Sendai 10(1):45-71.

Kast, A., Y. Tsunenari, M. Honma, J. Nishikawa, T. Shibata, and M. Torii.
  19752?.  Acute, Subacute, and Chronic Toxicity Studies of an Amino-
  Halogen-Substituted Benzylamine (Fominoben) in Rats and Mice.  Oyo
  Yakuri. Sendai 10(1):31-43.

King, M. E., A. M. Shefner, and R. R. Bates.  1973.  Carcinogenesis Bio-
  assay of Chlorinated Dibenzodioxins and Related Chemicals.  Environ.
  Health Persp. 5:163-170.

Knapp, W. K., Jr., W. M. Busey, and W. Kundzins.  1971.  Subacute Oral
  Toxicity of Monochlorobenzene in Dogs and Rats.  Toxicol. Appl.
  Pharmacol. Abstracts:  Tenth Annual Meeting 19:393.

Kociba, R. J., P. A. Keeler, C. N. Park, and P. J. Gehring.  1976.
  2,3,7,8-Tetrachlorodibenzo-p-Dioxin  (TCDD):  Results of a 13-week Oral
  Toxicity Study in Rats.  Toxicol. Appl. Pharmacol. 35:553-574.

Kociba, R. J., D. G. Keyes, J. E. Beyer, R. M. Carreon, C. E. Wade, D. A.
  Dittenber, R. P. Kalnins, L. E. Frauson, C. N. Park, S. D. Barnard,
  R. A. Hummel, and C. G. Humiston.  1978.  Results of a Two-Year Chronic
  Toxicity and Oncogenicity Study of 2,3,7,8-Tetrachlorodibenzo-p-Dioxin
  in Rats.  Toxicol. Appl. Pharmacol. 46-279-303.

Koeferl, M. T., C. D. Port, P. J. Garvin, and J. L. Dorner.  1976.  Sub-
  acute Toxicity of Cyclohexanone in Rats, Dogs, and Monkeys.  Toxicol.
  Appl. Pharmacol. Abstracts:  Fifteenth Annual Meeting 37(1):115.

Kohn, F. E., J. H. Kay, and J. C. Calandra.  1965.  Subacute Oral Toxicity
  of Zinophos.  Toxicol. Appl. Pharmacol. Abstracts:  Fourth Annual
  Meeting 7:488-489.

Korsrud, G. 0., H. C. Grice, T. Kuiper Goodman, J. E. Knipfel, and J. M.
  Mclaughlin.  1973.  Sensitivity of Several Serum Enzymes for the Detec-
  tion of Thioacetamide-, DimethyInitrosamine-, and Diethanolamine-Induced
  Liver Damage in Rats.  Toxicol. Appl. Pharmacol. 26:299-313.

-------
                                 211

Krasavage, W. J., F. J. Yanno, and C.  J.  Terhaar.   1973.   Dimethyl Tere-
  phthalate (DMT):  Acute Toxicity, Subacute Feeding and  Inhalation Studies
  in Male Rats.  Am. Ind. Hyg. Assoc.  J.  34(1):455-462.

Kruysse, A., V. J. Feron, H. R. Immel, B. J. Spit, and G.  J. Van Esch.
  1977.  Short-Term Inhalation Toxicity Studies with Peroxyacetyl Nitrate
  in Rats.  Toxicology 8:231-249.

Lawrence, W. H., M. Malik, J. E. Turner,  and J.  Autian.   1972.  Toxicity
  Profile of Epichlorohydrin.  J.  Pharm.  Sci. 61(11):1712-1717.

Litchfield, J. T., Jr.  1961.  Forecasting Drug Effects in Man from Studies
  in Laboratory Animals.  J. Am. Med.  Assoc. 177:104-108.

Litterst, C. L., T. E. Gram, E. G. Mimnaugh, P. Leber, D.  Emmerling, and
  R. I. Freudenthal.  1976.  A Comprehensive Study of In Vitro Drug Metab-
  olism in Several Laboratory Species.  Drug Metab. Dispos. 4(3):203-207.

Loomis, T.  1974.  Essentials of Toxicology, 2nd edition.   Lea and Febiger,
  Philadelphia.  223 pp.

Lyon, J. P., L. J. Jenkins, Jr., R. A. Jones, R. A. Coon, and J. Siegel.
  1970.  Repeated and Continuous Exposure of Laboratory Animals  to Acro-
  lein.  Toxicol. Appl. Pharmacol. 17:726-732.

MacFarland, H. N.   1968.  Exposure Chambers — Design  and Operation.   In:
  Proceedings  of  the 7th Annual Technical Meeting of  the American Associa-
  tion of Contamination Control, Chicago, 1968.  pp.  19-25.

McNamara, B. P.   1976.  Concepts in Health  Evaluation of Commercial and
  Industrial Chemicals.  In:  Advances in Modern Toxicology Vol. 1, Part
  1:  New Concepts  in  Safety  Evaluation, M.  A. Mehlman, R.  E.  Shapiro,
  and H. Blumenthal, eds.   John Wiley and Sons, New York.   pp.  61-140.

McNerney, J. M.,  and J. D.  MacEwen.   1965.   Comparative Toxicity Studies
  at Reduced and  Ambient Pressures.   I.  Acute  Response.   Am.  Ind.  Hyg.
  Assoc. J.  26:568-573.

Ministry of  of Health  and  Welfare  Canada.   1975.   The Testing of Chemicals
  for  Carcinogenicity,  Mutagenicity,  and Teratogenicity.   Ottawa.   183 pp.

Misu, Y., T. Segawa,  I.  Kuruma, M. Kojima,  and H.  Takagi.   1966.  Subacute
  Toxicity  of  0,0-Dimethyl-0-(3-Methyl-4-Nitrophenyl)phosphorothioate
   (Sumithion)  in the Rat.   Toxicol. Appl. Pharmacol.  9:17-26.

Morrow, P.  E., H.  C. Hodge, W.  F.  Newman, E. A. Maynard,  H. J. Blanchet,
  Jr., D. W. Fasset,  R.  E.  Birk,  and  S.  Mavrodt.   1966.   Deposition and
  Retention Models for Internal Dosimetry of the  Human Respiratory Tract.
  Health  Phys. 12:173-207.

-------
                                  212

National Academy of Sciences.  1975.  Principles for Evaluating Chemicals
  in the Environment.  National Academy of Sciences, Washington, B.C.
  454 pp.

National Academy of Sciences.  Committee for the Revision of NAS Publ.
  1138.  1977.  Principles and Procedures for Evaluating the Toxicity of
  Household Substances.  National Academy of Sciences, Washington, D.C.
  130 pp.

Newberne, J. W., J. P. Gibson, and P. M. Newberne.  1967.  Variation in
  Toxicologic Response of Species to an Analgesic.  Toxicol. Appl.
  Pharmacol. 10:233-243.

Owens, A. H., Jr.  1962.  Predicting Anticancer Drug Effects in Man from
  Laboratory Animal Studies.  J. Chronic Dis. 15:223-228.

Page, N. P.  1977.  Concepts of a Bioassay Program in Environmental Car-
  cinogenesis.  In:  Advances in Modern Toxicology, Vol. 3:  Environmental
  Cancer, H. F. Kraybill and M. A. Mehlman, eds.  John Wiley and Sons,
  New York.  pp. 87-171.

Page, N. P.  1979.  Review of Current Activities in Test Standards Devel-
  opment.  Presentation at EPA Subchronic Toxicity Workshop.  Denver
  (unpublished).

Peck, H. M.  1968.  An Appraisal of Drug Safety Evaluation in Animals and
  the Extrapolation of Results to Man.  In:  Importance of Fundamental
  Principles in Drug Evaluation, D. H. Tedeschi and R. E. Tedeschi, eds.
  Raven Press, New York.  pp. 450-471.

Peck, H. M.  1974.  Design of Experiments to Detect Carcinogenic Effects
  of Drugs.  In:  Carcinogenesis Testing of Chemicals, L. Goldberg, ed.
  CRC Press, Inc., Cleveland, Ohio.  pp. 1-14.

Peck, H. M., P. A. Mattis, P- F. Stoner, and R. E. Zwickey.  1967.
  Toxicology.  Drug. Inf. Bull. 1967:32-47.

Pinkel, D.  1956.  The Use of Body Surface Area As a Criterion of Drug
  Dosage in Cancer Chemotherapy.  Cancer Res. 18:853-856.

Prieur, D. J., D. M. Young, R. D. Davis, D. A. Cooney, E. R. Roman, R. L.
  Dixon, and A. M. Guarino.  1973.  Procedures for Preclinical Toxicologic
  Evaluation of Cancer Chemotherapeutic Agents:  Protocols of the Labora-
  tory of Toxicology.  Cancer Chemother. Rep., Part 3, 4(1):1-30.

Reddy, D. G., K. R. Krishnamurthy, and G. R. Bhaskar.  1962.  Carbon Tetra-
  chloride Cirrhosis in Rats.  Arch. Pathol. 74:73-80.

Reeves, A. L.  1965.  The Absorption of Beryllium from the Gastrointestinal
  Tract.  Arch. Environ. Health 11:209-214.

-------
                                   213

Reeves, A. L.,  D.  Deitch,  and A.  J. Vorwald.   1967.   Beryllium Carcinogen-
  esis.  I.  Inhalation Exposure  of Rats to Beryllium Sulfate Aerosol.
  Cancer Res.  27 (Part l):439-445.

Reuber, M. D.,  and E.  L.  Glover.   1967a.  Cholangiofibrosis in the Liver
  of Buffalo Strain Rats Injected with  Carbon Tetrachloride.   Br.  J.  Exp.
  Pathol. 48:319-322.

Reuber, M. D.,  and E.  L.  Glover,   1967h.  Thrombosis of Hepatic Veins
  Accompanying Carbon Tetrachloride Induced Cirrhosis.  Arch. Pathol.
  83:267-270.

Reuber, M. D.,  and E.  L.  Glover.   1967c.  Hyperplastic and Early Neopla-
  stic Lesions of the Liver in Buffalo  Strain Rats of Various Ages Given
  Subcutaneous Carbon Tetrachloride. J. Nat. Cancer Inst. 38(1):891-899.

Reuber, M. D.,  and E.  L.  Glover.   1968.  Carbon Tetrachloride Induced
  Cirrhosis.  Effect of Age and Sex. Arch. Pathol.  85:275-279.

Robbins, G., and D. Tettenborn.  1976.   Toxicity of Sisomicin in Animals.
  Infection 4 (Suppl.  4):349-354.

Roe, F.J.C.  1968.  Inhalation Tests.  In:  Modern Trends in Toxicology,
  Vol. I., E.  Boyland and R. Goulding,  eds.  Appleton-Century-Crofts,
  New York.  pp. 39-74.

Rosenkrantz, H., R. A. Sprague, R. W. Fleischman, and M. C. Braude.   1975.
  Oral A9-Tetrahydrocannabinol Toxicity in Rats Treated for Periods up to
  Six Months.   Toxicol. Appl. Pharmacol. 32:399-417.

Rowe, V. K., H. C. Spencer, D. D. McCollister, R. L. Hollingsworth, and
  E. M. Adams.  1952.  Toxicity of Ethylene Dibromide Determined on Exper-
  imental Animals.  Arch. Ind. Hyg. Occup. Med. 6:158-173.

Saslaw, S., and H. N. Carlisle.  1969.   llonhuman Primates  in  Evaluation
  of Hematotoxicity.  Ann. N.Y. Acad.  Sci. 162(1):646-658.

Schein, P.  S., R. D. Davis, S. Carter,  J.  Newman, D.  R. Schein, and  D. P.
  Rail.  1970.  The Evaluation of  Anticancer Drugs  in Dogs and Monkeys
  for the Prediction of Qualitative Toxicities in Man.  Clin.  Pharmacol.
  Therap. 11(1):3-40.

Shimizu, Y., C. Nagase, and K. Kawai.   1973.  Accumulation and Toxicity
  of Carbon Tetrachloride After Repeated  Inhalation in  Rats.   Ind. Health
  11:48-54.

Smith, C. C.  1950.  A Short-Term  Chronic Toxicity  Test.   J.  Pharmcol.
  Exp. Ther.  100:408-420.

Smith, C. C. 1979.  Extrapolation  of Drug Metabolism Data from Animals  to
  Man.   In:  Fundamentals and Principles  of  Clinical Pharmacology, H. P.
  Kuemmerle et  al., eds.  Urben  and Swartzenberg, Baltimore  (in preparation).

-------
                                  214

Smyth, H. F., Jr., and C. P. Carpenter.  1948.  Further Experience with
  the Range Finding Test in the Industrial Toxicology Laboratory.  J.
  Ind. Hyg. Toxicol. 30(1):63-68.

Smyth, H. F., Jr., C. P. Carpenter, and C. S. Weil.  1951.  Range-Finding
  Toxicity Data:  List IV.  Arch. Ind. Hyg. Occup. Med. 4:119-122.

Sontag, J. M., N. P. Page, and U. Saffiotti.  1976.  Guidelines for Car-
  cinogen Bioassay in Small Rodents.  U.S. Department of Health, Education,
  and Welfare Publ. No.  (NIH) 76-801.  65 pp.

Stevens, M. T.  1976.  The Value of Relative Organ Weights.  Toxicology
  5(3):311-318.

Stevens, M. T.  1977.  An Alternative Method for the Evaluation of Organ
  Weight Experiments.  Toxicology 7:275-281.

Stokinger, H. E., G. F.  Sprague, R. H. Hall, N. J. Ashenburg, J. K. Scott,
  and L. T. Steadman.  1950.  Acute Inhalation Toxicity of Beryllium.
  Arch. Ind. Hyg. Occup. Med. 1(4):379-397.

Stokinger, H. E., and C. A. Stroud.  1951.  Anemia in Acute Experimental
  Beryllium Poisoning.   J. Lab. Clin. Med. 38:173-182.

Street, A. E.   1970.  Biochemical Tests in Toxicology.  In:  Methods in
  Toxicology, G. E. Paget, ed.  F. A. Davis Company, Philadelphia.
  pp. 313-337.

Thompson, G. R., R. W. Fleischman, H. Rosenkrantz, and M. C. Braude.
  1974.  Oral and Intravenous Toxicity of A9-Tetrahydrocannabinol in
  Rhesus Monkeys.  Toxicol. Appl. Pharmacol. 27:648-665.

U.S. Environmental Protection Agency.  1979.  Support Document Test Data
  Development Standards:   Chronic Health Effects.  Toxic  Substances Con-
  trol Act, Section 4, EPA 560/11-79-001, Washington, D.C.  154 pp.

Verschuuren, H.  G., R. Kroes, and E.M.D. Tonkelaar.  1973.  Toxicity
  Studies on Tetrasul II.  Short-Term Comparative  Studies in 6 Animal
  Species.  Toxicology 1:103-112.

Verschuuren, H.  G., R. Kroes, and E.M.D. Tonkelaar.  1975.  Short-Term
  Oral and Dermal Toxicity of MCPA and MCPP.  Toxicology  3(3):349-359.

Villeneuve, D.  C., and W.  H. Newsome.  1975.  Toxicity and Tissue Levels
  in the Rat and Guinea  Pig Following Acute Hexachlorobenzene Administra-
  tion.  Bull.  Environ.  Contain. Toxicol. 14(3):297-300.

Vogin, E. E., S. Carson, A. Palanker, and G. E. Cannon.   1970.  Toxico-
  logic, Reproductive, and Teratogenic Studies with a Tetracycline Phos-
  phate Complex — Sulfamethizole Formulation.  Toxicol. Appl. Pharmacol.
  16:453-458.

-------
                                  215

Weil, C. S., and D. D. McCollister.   1963.   Relationship Between Short-
  and Long-Term Feeding Studies in Designing an Effective Toxicity Test.
  J. Agric. Food Chem. 11(1):486-491.

Weil, C. S., M. D. Woodside, J. R. Bernard, and C. P. Carpenter.  1969.
  Relationship Between Single-Peroral, One-Week, and Ninety-Day Rat
  Feeding Studies.  Toxicol. Appl. Pharmacol. 14:426-431.

Weisburger, J. H.  1975.  Chemical Carcinogenesis.  In:  Toxicology:  The
  Basic Science of Poisons, L. Casarett and J. Doull, eds.  McMillan,
  New York.  pp. 333-378.

Wiselogle, F. Y., ed.  1946.  A Survey of Antimalarial Drugs:  1941-1945.
  J. W. Edwards, Ann Arbor, Michigan.  1863 pp.

Worden, A. N., and K. H. Harper.  1963.  Oral Toxicity As Influenced by
  Method of Administration.  Proc. Eur. Soc. Study Drug Toxic. 2:15-26.

World Health Organization.  1966.  Principles for Pre-Clinical Testing
  of Drug Safety.  WHO Tech. Rep. Ser. 341, Geneva,  pp. 3-12.

World Health Organization.  1978.  Principles and Methods for Evaluating
  the Toxicity of Chemicals:  Part I.  Environmental Health Criteria 6.
  Geneva.  273 pp.

Worth, H. M., D. B. Meyers, W. R. Gibson, and G. C. Todd.  1970.  Acute
  and Subacute Toxicity of A204.  Antimicrob. Agents Chemother.  357-360.

Wright, P. J., and D. T. Plummer.  1974.  The Use of Urinary Enzyme
  Measurements to Detect Renal Damage  Caused by Nephrotoxic Compounds.
  Biochem. Pharmacol. 23:65-73.

Wroblewski, F., and J.  S. LaDue.  1955.  Serum Glutamic Oxalacetic  Trans-
  aminase Activity as an Index of Liver Cell  Injury:   A Preliminary Report.
  Ann.  Intern. Med. 43:345-360.

Yeary,  R. A.,  C. A. Brahm,  and D. L. Miller.   1965.  Acute  and  Subacute
  Toxicity of  an Adduct  of  Hydralazine and a 3-Ketoalkylthiazide.   Toxicol.
  Appl. Pharmacol. 7:598-605.

Zbinden, G.   1963.  Experimental and Clinical Aspects  of Drug Toxicity.
  Adv.  Pharmcol.  2:1-112.

-------
Appendix A

-------
        SUBCHRONIC TOXICITY TESTS AS DOSE-LEVEL-SETTING STUDIES
                FOR CHRONIC CARCINOGENICITY EVALUATIONS
                              David Prejean
INTRODUCTION

     A subchronic toxicity test  can be  defined  as  an  intermediate-length,
repeated-dose study designed to  indicate the  potential  long-term toxic
effects of a chemical.   If the study provides a sufficient  basis for the
estimation of safe exposure conditions, it may  function as  an "end
result" study; however, if the data obtained  are insufficient to esti-
mate safety and a longer chronic study  is required for  evaluation, the
subchronic toxicity test becomes a "dose-level—setting" study — the
topic of this discussion.
     Subchronic toxicity tests as dose-level-setting  studies have a
variety of applications, particularly as prechronic studies in terato-
genicity, reproduction, longevity, or carcinogenesis  evaluations; however,
it is in the area of carcinogenicity that they have most recently been
applied.  Therefore, the discussion that follows centers on the use of
subchronic toxicity tests to establish a maximum tolerated dose for
each sex and species used in a chronic carcinogenicity study.

TEST MATERIAL

     It is essential that the chemical being assayed be characterized as
to purity and pertinent physical properties  such  as  stability,  solubility,
and vapor pressure.  To ensure consistency,  it  is preferable, when pos-
sible, to use the same manufacturer's  lot  number  for both  the subchronic
and chronic  studies.   In  addition,  it  is  absolutely  necessary to deter-
mine the accuracy and  consistency with which the  chemicals/vehicles
are prepared (Page, 1977).
     Although not all  of  the  above  requirements have been  specified in
performing the subchronic dose-level-setting studies in the past (National
Institutes of Health,  1978),  current specifications  for conducting
carcinogenesis bioassay  studies,  as set forth  by  both  EPA  (Federal
                                   219

-------
                                  220
Register, 1978) and the National Cancer Institute (Sontag, Page, and
Saffiotti, 1976), do require or recommend each of the above.

TEST ANIMALS

Species and Strain
     Since the subchronic test is a dose-level-setting study, the spe-
cies, strains, and sexes selected for use should be the same as will be
used in the long-term study that follows.  For chronic toxicity tests,
two species have usually been recommended — preferably a rodent and a
nonrodent (Page, 1977), with the rat and dog historically the most popu-
lar.  Carcinogenicity studies, however, present a unique problem.
Whereas toxicity studies, even long-term ones, require relatively small
numbers of animals, carcinogenicity studies require a relatively large
number for statistical purposes.  Thus, because of the cost and time
factors, small laboratory rodents, specifically the rat and mouse, have
emerged as the most expedient test species.
     As with the selection of the test animals for toxicity studies, the
designation of a particular species to be used in a carcinogenesis study
is only the beginning.  Other questions, such as the use of inbred or
random bred strains and the use of germ-free, specific pathogen-free,
or conventional animals, must be answered.  Although a large number of
different rat and mouse strains have been utilized in the past  (Boyland
and Sydnor, 1962; Dunning, Curtis, and Madsen, 1947; National Institutes
of Health, 1977; Pietra, Rappaport, and Shubik, 1961; Shimkin et al.,
1962), the Fischer-344 rat and B6C3F1 hybrid mouse have become  the
standard for the National Cancer Institute Carcinogenesis Bioassay
Program  (National Institutes of Health, 1977; National Institutes of
Health, 1978; Sontag, Page, and Saffiotti, 1976).  Thus, although the
controversy over the use of inbred or outbred strains continues, for the
vast majority of long-term carcinogenicity studies performed between
1972 and 1979, the inbred and hybrid strains have been used.
     The final consideration in the selection of a test animal  has been
the health status.  For the most part, germ-free animals have rarely
been used in chronic carcinogenesis testing because of the extensive

-------
                                  221
labor requirements.   Gnotobiotic,  specific  pathogen-free,  and "extremely
clean" conventional animals have been used  most  frequently in long-
term carcinogenesis tests and thus have also  been used most widely in
subchronic tests designed to establish chronic dose levels (National
Institutes of Health, 1978; Sontag, Page, and Saffiotti,  1976).
     One final point should be made before  leaving the area of species
and strain.  With rare exceptions, the majority  of long-term  carcinogen-
esis bioassay studies have been performed utilizing cost-effective
systems rather than pursuing the question of  carcinogenicity  from the
standpoint of the response of the test system to the chemical in question
and the similarity to known responses in man. Future investigations
should attempt to tailor the subchronic study to delineate one or more
of a series of species and/or strains which would provide the most
suitable test system.

Number
     The number of animals required for a subchronic dose-level—setting
study has varied considerably in the past.   At one time the required
number of animals per dose level was five animals per species, and sex
was not taken into consideration  (Prejean,  1979).  Understandably, the
resulting dose levels selected for chronic studies were, at best,
guesses.  In some cases the levels selected proved correct but, more
often than not, incorrect dosages were selected, producing invalid
chronic data.
     The current trend in subchronic studies has been to use  a minimum
of ten animals of each sex and each species  in  the long-term  study
(Sontag, Page, and Saffiotti, 1976).  This number has proved  sufficient
as long as the parameters  for evaluation were limited to mortality,  body
weight gain, clinical signs, postmortem observation, and pathologic
diagnosis; however,  the recent expansion of  these parameters  to  include
hematology, clinical chemistry, and pharmacokinetic  studies will,  of
necessity, produce an increase in the number of animals required for the
subchronic study.  There are indications that the  role of  the subchronic
study in the carcinogenesis bioassay program is likely to  require more

-------
                                  222
animals, probably a minimum of 20 animals per sex per species (National
Cancer Institute, 1979).

DOSES AND DOSE LEVELS

     The doses selected for use in the subchronic study are usually
based on data obtained from other shorter-term studies (Sontag, Page,
and Saffiotti, 1976) but may, in some instances, be based on literature
information.  In selecting doses to be used in the subchronic study, it
is important to remember that the ultimate objective of the study is to
establish the maximum tolerated dose(s) for use in the chronic study.
Thus, the more data available from the subchronic study, the more
accurately the chronic dose levels may be set.  For this reason, it is
extremely important to select as the highest dose a level that will
produce an easily discernible, reproducible toxic effect and a series of
lower doses that provide a complete spectra of dose responses (Weisburger
and Weisburger, 1967) from toxic to "nontoxic."  In this way, not only
can the dose levels for the chronic study be selected with more accuracy,
but representative toxic lesions and symptoms may be observed that could
be of value later in the chronic study.
     Subchronic dose-level-setting studies should include, as a minimum,
a range of five equally spaced dose levels (Sontag, Page, and Saffiotti,
1976).  The intervals used most frequently have been half-doses (100,
50, 25, etc.)  (Sontag, Page, and Saffiotti, 1976) or half-log10 doses
(100, 31.6, 10, 3.16, etc.).  Thus, assuming five dose levels, the
lowest level tested would be approximately 6/100 of the top dose if half-
doses are used or 1/100 of the top dose if half-log}Q doses are used;
either of these selections provides a reasonable span of doses from
which to gather toxicity data.

ROUTE
                                                                      t
     Administration of the chemical in the subchronic study should
closely simulate the route by which most human exposure occurs and
should be identical to that which will be used for the chronic study.

-------
                                  223
With the exception of certain drugs,  most chemicals  enter the body by
one of three routes — orally, by inhalation,  or by absorption through
the skin (Page, 1977).  These then are the most common routes used to
test chemicals for carcinogenicity.   When exposure occurs by more than
one route of administration, as in the case of pesticides, the route of
exposure chosen is usually based on pharmacokinetic  data, which could
necessitate performing several subchronic studies before selecting the
route and dose levels for the chronic study.
     In general, chemical vapors and particulate materials such as smoke
and asbestos are administered by inhalation,  whereas food additives and
pesticides are examples of chemicals that would be administered orally
either by way of gastric intubation, the drinking water, or the feed.
Cutaneous application, though not so common as oral administration, is
the preferred route for administering cosmetics and certain drugs.

DURATION

     Although the literature abounds with various specifications  regard-
ing the appropriate length for an end result subchronic  study  (Peck,
1968; Smith, 1950; Weil and McCollister, 1963),  the majority of  the  evi-
dence supports the conclusion that a 90-day test will predict  most
chronic effects exclusive of carcinogenicity and  teratogenicity  (McNamara,
1976).  Since the function  of a dose-level-setting  subchronic  test  is to
establish appropriate dose  levels for a  chronic study  through  the iden-
tification of subchronic toxic effects and their extrapolation to chronic
toxic effects, a 90-day test should be just  as appropriate for a dose-
level-setting subchronic study as it  is  for  an end  result study.  Of
course, if prior information suggests  that the chemical or its metabolites
may be rapidly or  slowly eliminated  from the test species, then the
duration may be reduced or  extended  as necessary.  This has, in fact,
proven to be the case.  In  the  Guidelines for* Carcinogen Bioaseay in
Small Rodents  the National  Cancer Institute  recommends that in a subchronic
study "The test agent should be administered to the animals for 90 days"
 (Sontag, Page, and  Saffiotti,  1976).   Thus,  as with the other parameters

-------
                                   224
already discussed, the duration of end result and dose-level-setting
subchronic studies is compatible.

DATA COLLECTION

     Of all the aspects of a subchronic test, regardless of whether it
is an end result study or a dose-level-setting study, the most important
is data collection.  Without the accumulation of adequate and accurate
observations during the test period and pathological examinations, the
establishment of reliable chronic dose levels and/or the estimation of
the long-term toxic effects is very difficult.  What then are the impor-
tant data collection points for a subchronic dose-level-setting study?

Clinical Observations, Body Weights, and Food and Water Consumption
     During the course of a subchronic study much of the data must be
collected on a daily or, at most, a weekly basis.  Clinical observations
as to the general health of each animal should be recorded twice daily,
7 days a week by a qualified technician (National Academy of Sciences,
1975).  These observations should include an assessment of animal activ-
ity, posture, body temperature (extremes only), appearance of eyes and
hair or coat, excreta and bedding, and respiratory function.
     In addition to the above observations, each animal should be weighed
weekly (Sontag, Page, and Saffiotti, 1976).  Except in unusual instances,
palpation for subcutaneous or intraperitoneal masses would not be required.
     Food and water consumption should be measured at least weekly and if
possible daily for water consumption.  These data are especially useful
for those studies where the test chemical is administered by dosed-feed
or dosed-water.  Not only are the data necessary for dose calculations,
they also provide an excellent indication of the effect of the chemical
on the palatability of the food and water and indicate the general
health status of the test animals.

-------
                                  225
Pathology
     The gross and microscopic examination of test animal tissues and
organs is an extremely important aspect of data collection on a subchronic
dose-level-setting study.  Complete  necropsies should be performed on
all animals (Sontag, Page, and Saffiotti,  1976).  These necropsies
should be performed under the supervision of a qualified pathologist,
preferably board-certified.  As a minimum, samples of the approximately
30 organs or tissues listed in the Guidelines for Carcinogen Bioaseay in
Small Rodents should be preserved (Sontag, Page, and Saffiotti, 1976).
In addition to a careful examination of all internal organs and tissues
prior to their removal from the animal, a gross necropsy should include
an external examination of all body surfaces and orifices.
     A brief review of the literature indicates that there is general
agreement concerning the gross observations recommended above  (Peck,
1968; World Health Organization, 1978).  The recommendations from the
microscopic examination vary widely, however, ranging from none  (Barnes
and Denz, 1954) to tissues from all control animals, the highest dose-
level animals without mortality, and the next higher dose-level  group
(Sontag, Page, and Saffiotti, 1976).  In practice, results of  subchronic
toxicity studies performed with token microscopic examinations compared
to those with extensive  examinations indicate that the  selections of
dose levels for chronic  studies were more  accurate when a reasonable
amount of histopathology was available  (National  Institutes  of Health,
1977; National Institutes  of Health, 1978; Prejean,  1979).

Other Data
     Recently, consideration has been  given to the addition of biochemical
tests to subchronic dose-level-setting studies.  In general, it is felt
that these  tests  could be extremely valuable in evaluating specific
toxic effects of  selected chemicals; however,  for the most part, the
literature  does not  specify a  standard set of tests but recommends the
inclusion of hematology, clinical chemistry, and pharmacokinetic and
behavioral  studies  for each chemical.

-------
                                  226
CONCLUSIONS

     In reviewing the general parameters for a subchronic toxicity test
designed to establish dose levels for a chronic carcinogenicity study,
the degree of similarity between this type of study and one designed as
an end result study is remarkable.  Both types require characterization
of the test chemical, an adequate number of test dose levels (usually
five), the selection of route based on exposure data in man, exposure
over a 90-day period, and the collection of pertinent data such as
clinical and gross observations and histopathologic evaluations.  Although
the use of at least two species is recommended for both types of studies,
the end result test has usually relied on a rodent and a nonrodent,
whereas the dose-level-setting test has used two species of rodents
because of the economic considerations of the long-term study that will
follow.  The only other major area of disagreement has been in the use
of biochemical tests.  End result studies have relied on these tests for
several years, but it has been only recently that dose-level-setting
studies have also begun to take advantage of the data available in
selected hematologic or clinical chemistry tests.  Thus, unless the
current trend changes, it appears that, except for the species difference,
which will probably always remain, end result and dose-level-setting sub-
chronic toxicity tests will be virtually identical in the next few
years.

-------
                                  227

                       REFERENCES FOR APPENDIX A


Barnes, J. M.,  and F. A.  Denz.   1954.   Experimental  Methods  Used in
  Determining Chronic Toxicity.   Pharmcol.  Rev. 6:191-242.

Boyland, E., and K. Sydnor.   1962.  The Induction of Mammary Cancer in
  Rats. Br. J.  Cancer 16:731-739.

Dunning, W., M. Curtis, and M.  Madsen.  1947.   Induction of  Neoplasms in
  Five Strains of Rats with Acetylaminofluorene.  Cancer Res. 7:134-140.

Federal Register.  1978.   Nonclinical Laboratory Studies: Good Labora-
  tory Practice Regulations. 43Fr(247):59986-60025.

McNamara, B.  1976.  Concepts in Health Evaluation of Commercial and
  Industrial Chemicals.  In:  Advances in Modern Toxicology, Vol. 1,
  Part 1:  New Concepts in Safety Evaluation,  M. A.  Mehlman, R. E.
  Shapiro, and H. Blumenthal, eds.  John Wiley and Sons, New York.
  pp. 61-140.

National Academy of Sciences.  1975.  Acute and Subchronic Toxicity.
  In:  Principles for Evaluating Chemicals in the Environment.  National
  Academy of Sciences, Washington, D.C.  pp. 103-104, 327-330.

National Cancer Institute.  1979.  Carcinogenicity Studies in Rodents.
  National Institutes of Health.  RFP No. N01 CP 95619-62.  Bethesda,
  Maryland.

National Institutes of Health.   1977.  Bioassay of Tolbutamide  for Pos-
  sible Carcinogenicity.  National Cancer Institute Carcinogenesis
  Technical Report Series No. 47.  Department  of Health, Education,  and
  Welfare Publication No. (NIH)  77-831.  Bethesda, Maryland.   83 pp.

National Institutes of Health.   1978.  Bioassay of  4,4'-Thiodianiline
  for Possible Carcinogenicity.   National Cancer  Institute  Carcinogenesis
  Technical Report Series No. 47.  Department  of  Health, Education,  and
  Welfare Publication No. (NIH)  78-847.  Bethesda,  Maryland.   106 pp.

Page, N.   1977.  Chronic Toxicity and Carcinogenicity  Guidelines.  J.
  Environ. Pathol. Toxicol.  1:161-182.

Peck, H.   1968.  An Appraisal of Drug Safety  Evaluation in  Animals and
  the Extrapolation  of Results  to Man.   In:   Importance of  Fundamental
  Principles in Drug Evaluation, D.  H.  Tedeschi and R.  E. Tedeschi, eds.
  Raven Press, New York.  pp. 450-471.

Pietra, G., H. Rappaport, and P. Shubik.   1961.  The Effects of Carcino-
  genic Chemicals  in Newborn Mice.   Cancer 14:308-317.

Prejean,  J.  1979.   Unpublished data.  Southern Research Institute.
  Birmingham,  Alabama.

-------
                                   228

Shimkin, M., S. Smith, P. Shimkin, and H. Andervont.  1962.  J. Natl.
  Cancer Inst. 28:1219-

Smith, C.  1950.  A Short-Tern Chronic Toxicity Test.  J. Pharmcol. Exp.
  Ther. 100:408-420.

Sontag, J., M. Page, and U. Saffiotti.  1976.  Guidelines for Carcinogen
  Bioassay in Small Rodents.  Department of Health, Education, and Welfare
  Publication No. (NTH) 76-801.  National Cancer Institute, Bethesda,
  Maryland.  65 pp.

Weil, C., and D. McCollister.  1963.  Relationship Between Short- and
  Long-Tena Feeding Studies in Designing an Effective Toxicity Test.  J.
  Agric. Food Chem. 11(1):486-491.

Weisburger, J., and E. Weisburger.  1967.  Tests for Chemical Carcinogens.
  In:  Methods in Cancer Research, Vol. I, H. Busch, ed. Academic Press,
  New York.  pp. 307-398.

World Health Organization.  1978.  Acute, Subacute, and Chronic Toxicity
  Tests.  In:  Principles and Methods for Evaluating the Toxicity of
  Chemicals.  Geneva,  pp. 95-115.

-------
Appendix B

-------
                      Appendix B.  Chemicals surveyed for Table 4.35
                 Namea
       Primary
                                                    use
                                                      ,b
                                                                              Reference
Carbon tetrachloride
3',4'-Dichloropropion anilide
Tetrahydrothiophene-1,1-dioxide;
  Sulfolane
Calcium carbimide

1,2,4-Trichlorobenzene
Acrolein; aerylaldehyde, 2-propenal

Ethylenebisisothiocyanate sulfide
Triphenyl tin hydroxide; fentin
  hydroxide
Isooctyl isodecyl nylonate
Ponceau 4R; trisodium salt of l-(4-
  sulpho-1-naphthylazo)-2-naphthol-
  6,8-disulphonic acid
Orange G; disodium salt of 1-phenylazo-
  2-naphthol-6,8-disulphonic acid
Hexachlorobenzene; HCB
Clindamycin hydrochloride; Cleocin
Trlethyl phosphate

Ponceau MX; disodium salt of l-(2,4-
  xylylazo)-2-naphthol-3,6-disulphonic
  acid
Dibutyl(diethylene glycol bisphtha-
  late); DDGB
2,3,7,8-Tetrachlorodibenzo-p-dioxin;
  TCDD

Quanethidine
l-Methyl-3-keto-4-phenylquinuclidinium
  bromide; MA 540
TR2379; trans isomer of N-(l,3,4,6,7-
  hexahydro-llbH-benzo[a]quinolizin-
  2-y1)propionanilide hydrochloride
ff-Methyl-ff-(1-naphthyl)fluoroaceta-
  mlde; Nissol; MNFA
2-Methyl-4-chlorophenoxy acetic acid;
  MCPA

2,5,4*-Trichlorobiphenyl

Fenterol-HBr; 1-(3,5-dihydroxy-phenyl)-
  2-{[l-(4-hydroxybenzyl)-ethyl]-amino}
  ethanol hydrobromide
Industrial chemical
Herbicide
Solvent

Antialcoholic drug

Solvent
Cigarette smoke
  component
Fungicide
Insect reproduction
  inhibitor
Plasticizer
Food color
Food color
Fungicide and indus-
  trial by-product
Antibiotic drug
Whipping agent

Food color

Plasticizer

Impurity in pesticides

Antihypertensive drug
Antihypertensive drug

Antihypertensive drug

Pesticide

Herbicide
Transformer dielectric
   fluid
Beta-sympathomimetic
   drug
Adams et al., 1952
Ambrose et al., 1972
Anderson et al., 1977

Benitz, Kramer, and Dambach,
  1965
Coate et al., 1977
Feron et al., 1978;
  Lyon et al., 1970
Freudenthal et al., 1977
Gaines and Kimbrough, 1968

Gaunt et al., 1969
Gaunt et al., 1967
Gaunt et al., 1971

Gralla et al., 1977;
  latropoulos et al., 1978;
  latropoulos et al., 1976;
  Kimbrough and Linder, 1974;
  Kuiper-Goodman et al.,
  1977
Gray et al., 1972
Gumbman, Gagne, and Williams,
  1968
Hall, Lee, and Fairweather,
  1966
Hall, Austin, and
  Fairweather,  1966
Harris  et  al.,  1973;  Kociba
  et al.,  1976;  Zinkl et  al.,
  1973
Hartnagel  et  al.,  1976
Hartnagel  et  al.,  1976

Hartnagel  et  al.,  1975
 Hashimoto et al., 1968

 Hattula et al., 1977;
   Verschuuren, Kroes, and
   Den Tonkelaar, 1975
 latropoulos et al., 1977

 Kast et al., 1975a
                                             231

-------
                                             232

                                     Appendix B (continued)
                 Namea
                                                  Primary
                                   Reference
Fominoben-HCl; 3'-chloro-2'-[N-methyl-
  [(morpholino-carbonyl)methyl]-amino-
  methyl]benzanilide-hydrochlorlde
Mycotoxin produced by Aepergillus
  fianigatua 121

Peroxyacetyl nitrate; PAN
Epichlorohydrin; l-chloro-2,3-
  epoxypropane
Ferric dimethyl dithiocarbamate; Ferbam


Tetramethylthiuram disulfide; Thiram


AHR-2438B; sodium salt of a. lignosul-
  phonate

Barthrin; 6-chloropiperonyl ester of
  chrysanthemumic acid

Dimethrin; 2,4-dimethylbenzyl ester of
  chrysanthemumic acid
Hexachlorophene; 2,2'-methylenebis
  (3,A,6-trichlorophenol)

Pyridoxine hydrochloride
A9-Tetrahydrocannabinol; A9-THC


2-Methyl-4-chlorophenoxy propionlc
  acid; MCPP
Antitussive drug


Animal feed impurity


Photochemical product
  in smog

Epoxy resin

Fungicide


Fungicide


Animal feed impurity

Insecticide

Insecticide

Antibacterial and anti-
  fungicidal agent

Antischizophrenic drug

Natural ingredient in
  marijuana plant

Herbicide
Kast et al., 1975Z>


Khor et al., 1976


Kruysse et al., 1977

Lawrence et al., 1972

Lee, Russell, and Minor,
  1978

Lee, Russell, and Minor,
  1978

Luscombe and Nicholls, 1973

Masri et al., 1964

Masri et al., 1964


Nakaue, Dost, and Buhler,
  1973

Phillips et al., 1978

Thompson et al., 1974

Verschuuren, Kroes, and
  Den Tonkelaar, 1975
      rirst name listed is the one used in Appendix C.

      Primary use as listed by the reference source.

-------
                                  233

                       REFERENCES FOR APPENDIX B
Adams, E. M., H. C. Spencer, V. K. Rowe, D. D. McCollister, and
  D. D. Irish.  1952.  Vapor Toxicity of Carbon Tetrachloride Deter-
  mined by Experiments on Laboratory Animals.  Ind. Hyg. Occup. Med.
  6:50-66.

Ambrose, A. M., P. S. Larson, J. F. Borzelleca, and R. G. Hennigar, Jr.
  1972.  Toxicologic Studies on 3',4'-Dichloropropionanilide.  Toxicol.
  Appl. Pharmacol. 23:650-659.

Anderson, M. E., R. A. Jones, R. G. Mehl, T. A. Hill, L. Kurlansik,
  and L. J. Jenkins, Jr.  1977.  The Inhalation Toxicity of Sulfolane
  (Tetrahydrothiophene-l,l-Dioxide).  Toxicol. Appl. Pharmacol.
  40:463-472.

Benitz, K. F., A. W. Kramer, Jr., and G. Dambach.  1965.  Comparative
  Studies on  the Morphologic Effects of Calcium Carbimide, Propylthiouracil,
  and Disulfiram in Male Rats.  Toxicol. Appl. Pharmacol. 7:128-162.

Coate, W. B., W. H. Schoenfisch, T. R. Lewis, and W. M. Busey.  1977.
  Chronic Inhalation Exposure of Rats, Rabbits, and Monkeys to 1,2,4-
  Trichlorobenzene.  Arch. Environ. Health 32:249-255.

Cornish, H. H., and R. Hartung.  1969.  The Subacute Toxicity of
  1,1-Dimethylhydrazine.  Toxicol. Appl. Pharmacol. 15:62-68.

Feron, V. J., A. Kruysse, H. P. Til, and H. R. Immel.   1978.  Repeated
  Exposure to Acrolein Vapor:   Subacute Studies in Hamsters, Rats,  and
  Rabbits.  Toxicology 9:47-57.

Freudenthal,  R. I., G. A. Kerchner, R. L. Persing, I.  Baumel,  and
  R. L. Baron.  1977.  Subacute Toxicity of Ethylenebisisothiocyanate
  Sulfide in  the Laboratory Rat.  J. Toxicol. Environ.  Health
  2:1067-1078.

Gaines, T. B.,  and R. D. Kimbrough.  1968.  Toxicity  of Fentin Hydroxide
  to Rats.  Toxicol. Appl. Pharmacol. 12:397-403.

Gaunt, I. F., J. Colley, P. Grasso, M. Creasey,  and  S.  D.  Gangolli.  1969.
  Acute  (Rat  and Mouse) and Short-Term  (Rat)  Toxicity Studies  on
  Isooctyl Isodecyl Nylonate.   Food Cosmet.  Toxicol.  7:115-124.

Gaunt, I. F., M. Farmer, P. Grasso, and  S.  D.  Gangolli.  1967.   Acute
  (Mouse and  Rat)  and Short-Term  (Rat) Toxicity  Studies on Ponceau 4R.
  Food Cosmet.  Toxicol. 5:187-194.

Gaunt, I. F., M. Wright, P. Grasso, and  S.  D.  Gangolli.  1971.  Short-
  Term Toxicity of Orange G in Rats.  Food Cosmet.  Toxicol. 9:329-342.

-------
                                   234

Gralla, E. J., R. W. Fleischman, Y. K.  Luthra, M. Hagopian, J. R. Baker,
  H. Esber, and W. Marcus.  1977.  Toxic Effects of Hexachlorobenzene
  After Daily Administration to Beagle Dogs for One Year.  Toxicol. Appl.
  Pharmacol. 40:227-239.

Gray, J. E., R. N. Weaver, J. A. Bollert, and E. S. Feenstra.  1972.
  The Oral Toxicity of Clindamycin in Laboratory Animals.  Toxicol. Appl.
  Pharmacol. 21:516-531.

Gumbmann, M. R., W. E. Gagne, and S. N. Williams.  1968.  Short-Term
  Toxicity Studies of Rats Fed Triethyl Phosphate in the Diet.  Toxicol.
  Appl. Pharmacol. 12:360-371.

Hall, D. E., F. S. Lee, and F. A. Fairweather.  1966.  Acute  (Mouse and
  Rat) and Short-Term (Rat) Toxicity Studies on Ponceau MX.  Food Cosmet.
  Toxicol. 4:375-382.

Hall, D. E., P. Austin, and F. A. Fairweather.  1966.  Acute  (Mouse and
  Rat) and Short-Term (Rat) Toxicity Studies on Dibutyl  (Diethylene
  Glycol Bisphthalate).  Food Cosmet. Toxicol. 4:383-388.

Harris, M. W., J. A. Moore, J. G. Vos, and B. N. Gupta.  1973.  General
  Biological Effects of TCDD in Laboratory Animals.  Environ. Health
  Perspect. 5:101-109.

Hartnagel, R. E., B. M. Phillips, E. H. Fonseca, and R. L. Kowalski.
  1976.  The Acute and Target Organ Toxicity of l-Methyl-3-Keto-4-
  Phenylquinuclidinium Bromide  (MA 540) and Guanethidine in the Rat
  and Dog.  Arzneim.-Forsch. 26:1671-1672.

Hartnagel, R. E., B. M. Phillips, P. J. Kraus, R. L. Kowalski, and
  E. H. Fonseca.  1975.  A Subchronic Study of the Toxicity of an
  Orally Administered Benzoquinolizinyl Derivative in the Rat and
  Dog.  Toxicology 4:215-222.

Hashimoto, Y., T. Makita, H. Miyata, T. Noguchi, and G.  Ohta.  1968.
  Acute and Subchronic Toxicity of a New Fluorine Pesticide.  #-Methyl-
  tf-(l-Naphthyl)fluoroacetamide.  Toxicol. Appl. Pharmacol. 12:536-547.

Hattula, M. L., H. Elo, H. Reunanen, A. U. Arstila, and  T. E. Sorvari.
  1977.  Acute and Subchronic Toxicity of 2-Methyl-4-Chlorophenoxyacetic
  Acid  (MCPA) in Male Rat.  I.  Light Microscopy and Tissue Concentra-
  tions of MCPA.  Bull. Environ. Contain. Toxicol. 18:152-158.

Hunter, C. G., and D. E. Stevenson.  1967.  Acute and Subacute Oral
  Toxicity of Alromine RU 100 in Rats.  Food Cosmet. Toxicol. 5:491-496.

latropoulos, M. J., J. Bailey, H. P. Adams, F. Coulston, and  W. Hobson.
  1978.  Response of Nursing Infant Rhesus to Clophen A-30 or Hexachloro-
  benzene Given to Their Lactating Mothers.  Environ. Res. 16:38-47.

-------
                                  235

latropoulos, M. J., G. R. Felt, H. P. Adams, F.  Korte,  and F. Coulston.
  1977.  Chronic Toxicity of 2,5,4'-Trichlorobiphenyl in Young Rhesus
  Monkeys.  II.  Histopathology.  Toxicol. Appl. Pharmacol. 41:629-638.

latropoulos, M. J., W. Hobson, V. Knauf, and H.  P. Adams.  1976.
  Morphological Effects of Hexachlorobenzine Toxicity in Female
  Rhesus Monkeys.  Toxicol. Appl. Pharmacol. 37:433-444.

Kast, A., Y. Tsunenari, M. Honma, J. Nishikawa,  T. Shibata, and M. Torii.
  1975a.  Acute, Subacute, and Chronic Toxicity Studies of the Beta-
  Sympathomimetic, Fenterol-HBr on Rats, Mice, and Rabbits.  Oyo Yakuri.
  Sendai 10(1):45-71.

Kast, A., Y. Tsunenari, M. Honma, J. Nishikawa,  T. Shibata, and M. Torii.
  19752?.  Acute, Subacute, and Chronic Toxicity Studies of an Amino-
  Halogen-Substituted Benzylamine (Fominoben) in Rats and Mice.  Oyo
  Yakuri. Sendai 10(1):31-43.

Khor, 6. L., J. C. Alexander, J. H.  Lumsden, and G. J.  Losos.  1976.
  Safety Evaluation of AspergilZus fumigatus Grown on Cassava for Use
  as an Animal Feed.  Can. J. Comp.  Med. 41:428-434.

Kimbrough, R. D., and R. E. Linder.   1974.  The Toxicity of Technical
  Hexachlorobenzene in the Sherman Strain Rat.  A Preliminary Study.
  Res. Commun. Chem. Pathol. Pharmacol. 8(4):653-664.

Kociba, R. J., P. A. Keeler, C. N. Park, and P. J. Gehrin.  1976.
  2,3,7,8-Tetrachlorodibenzo-p-Dioxin  (TCDD):  Results of  a 13-Week
  Oral Toxicity Study in Rats.  Toxicol. Appl. Pharmacol.  35:553-574.

Kruysse, A., V. J. Feron, H. R. Immel, B. J. Spit, and G.  J. Van  Esch.
  1977.  Short-Term Inhalation Toxicity Studies with Peroxyacetyl
  Nitrate in Rats.  Toxicology 8:231-249.

Kuiper-Goodman, T., D. L. Grant, C. A. Moodie, G. 0. Korsrud,  and
  I. C. Munro.  1977.  Subacute Toxicity of  Hexachlorobenzene  in  the
  Rat.  Toxicol. Appl. Pharmacol. 40:529-549.

Lawrence, W. H., M. Malik, J. E. Turner, and J. Autian.   1972.  Toxicity
  Profile of Epichlorohydrin.  J. Pharmacol. Sci.  61(11):1712-1717.

Lee, C-C., J. Q.  Russell, and J. L. Minor.   1978.   Oral Toxicity  of
  Ferric Dimethyl Dithiocarbamate (Ferbam)  and  Tetramethylthiuram
  Bisulfide  (Thiram)  in Rodents.  J. Toxicol. Environ.  Health  4:93-106.

Luscombe, D. K.,  and  P. J. Nicholls.   1973.  Acute and Subacute Oral
  Toxicity of AHR-2438B,  a Purified  Lignosulphonate, in Rats.   Food
  Cosmet. Toxicol. 11:229-237.

Lyon, J. P., L. J. Jenkins,  Jr.,  R.  A.  Jones, R.  A.  Coon, and  J.  Siegel.
  1970.  Repeated and Continuous  Exposure of Laboratory Animals to
  Acrolein.  Toxicol.  Appl.  Pharmacol.  17:726-732.

-------
                                   236

Masri, M. S., A. P. Hendrickson, A. J. Cox, Jr., and F. DeEds.  1964.
  Subacute Toxicity of Two Chrysanthemumic Acid Esters:  Barthrin and
  Dimethrin.  Toxicol. Appl. Pharmacol. 6:716-725.

Nakaue, H. S., F. N. Dost, and D. R. Buhler.  1973.  Studies on the
  Toxicity of Hexachlorophene in the Rat.  Toxicol. Appl. Pharmacol.
  24:239-249.

Phillips, W.E.J., J.H.L. Mills, S. M. Charbonneau, L. Tryphonas,
  6. V. Hatina, Z. Zawidzka, F. R. Bryce, and I. C. Munro.  1978.
  Subacute Toxicity of Pyridoxine Hydrochloride in the Beagle Dog.
  Toxicol. Appl. Pharmacol. 44:323-333.

Thompson, 6. R., R. W. Fleischmann, H. Rosenkrantz, and M. C. Braude.
  1974.  Oral and Intravenous Toxicity of A9-Tetrahydrocannabinol in
  Rhesus Monkeys.  Toxicol. Appl. Pharmacol. 27:648-665.

Verschuuren, H. G., R. Kroes, and E. M. Den Tonkelaar.  1975.  Short-
  Term Oral and Dermal Toxicity of MCPA and MCPP.  Toxicology 3:349-359.

Zinkl, J. G., J. G. Vos, J. A. Moore, and B. N. Gupta.  1973.  Hema-
  tologic and Clinical Chemistry Effects of 2,3,7,8-Tetrachlorodibenzo-
  p-dioxin in Laboratory Animals.  Environ. Health Perspect. 5:111-118.

-------
Appendix C

-------
Appendix C.  Species,  route  of  exposure, chemical, and reference data for Table 4.35
Chemical

Carbon tetrachloride
3, ' ,4'-Dichloropropionanilide
Calcium carbimide
Acrolein
Ethylenebislsothiocyanate sulfide
Triphenyl tin hydroxide
Ponceau 4R
Isooctyl isodecyl nylonate
Orange G
Hexachlorobenzene

Clindamycin hydrochloride

Triethyl phosphate
2,3,7, 8-Tetrachlorodibenzo-p-dloxin
TR2379

Quanethidine
l-Methyl-3-keto-4-phenylquinuclldinium
bromide
ff-Methyl-ff- (1-naphthyl) f luoracetamide
2-Methyl-4-chlorophenoxy acetic acid
2-Methyl-4-chlorophenoxy propionic acid
Fenterol-HBr
Fominoben-HCl
Aspergillus fumlgatus
Peroxyacetyl nitrate
Bpichlorohydrin
Ferric dimethyl dithiocarbamate
Tetramethylthiuram disulf ide
Ponceau MX
DibutyKdiethylene glycol bisphthalate)
AHR-2438B
Acrolein
Barthrin
Dimethrin
Pyridoxine hydrochloride
A9-Tetrahydrocannabinol
Species
Adrenal
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Rat
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Dog
Dog
Rat
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Rat
Rat
Dog
Monkey
Monkey
Route Result

Inhalation
Oral
Oral +
Inhalation +
Oral
Oral
Oral
Oral +
Oral +
Oral
Oral
Oral
Oral +
Oral +
Oral
Oral
Oral +
Oral
Oral
Oral +
Oral +
Oral
Oral
Oral
Oral
Oral
Oral +
Oral
Oral
Oral +
Oral
Oral
Inhalation -
Injection
Oral
Oral
Oral
Oral
Oral
Inhalation
Oral
Oral
Oral
Injection
Oral +
Reference

1
2
4
6
7
8
10
9
11
12
22
24
28
31
13
13
14
17
29
19
19
18
18
18
18
20
21
40
40
25
26
27
30
32
33
33
15
16
34
35
36
36
38
39
39
                                        239

-------
      240




Appendix C (continued)
Chemical

Tetrahydrothiophene-l,l-dioxide


Ac role in
Ethylenebisisothiocyanate sulfide
leooctyl isodecyl nylonate
Ponceau 4R
Clindamycin hydrochloride

TR2379

Fominoben-HCl
Fenterol-HBr
2,3, 7 ,8-Tetrachlorodibenzo-p-dioxin
Peroxyacetyl nitrate
A 9-Tet rahydrocannab ino 1


Calcium carbimide
Ethylenebisisothiocyanate sulfide
TR2379

Quanethidine

l-Methyl-3-keto-A-phenylquinuclidinium
bromide
Hexachlorobenzene

2,5,4' -Trichlorobiphenyl
Ferric dimethyl dithlocarbamate
Tetramethylthiuram disulfide

Carbon tetrachloride
3' ,4'-Dichloropropionanilide
Calcium carbimide
1 , 2 ,4-Trichlorobenzene

Hexachlorobenzene
Clindamycin hydrochloride

TR2379

ff-Methyl-ff- (1-naphthyl) f luoracetamide
Fenterol-HBr
Fominoben-HCl
Asperglllus fumigatus
Species
Aorta
Rat
Dog
Monkey
Rat
Rat
Rat
Rat
Dog
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Monkey
Monkey
Bone
Rat
Rat
Dog
Rat
Rat
Dog
Rat
Dog
Monkey
Monkey
Monkey
Rat
Rat
Bone Marrow
Rat
Rat
Rat
Rat
Monkey
Dog
Dog
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Route Result

Inhalation
Inhalation
Inhalation
Inhalation
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Inhalation
Injection
Oral

Oral +
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral

Inhalation
Oral
Oral +
Inhalation
Inhalation
Oral
Oral
Oral
Oral
Oral
Oral
Oral i
Oral
Oral
Reference

3
3
3
6
7
9
10
13
13
19
19
26
25
29
30
39
39

4
7
19
19
18
18
18
18
24
22
23
33
33

1
2
4
5
5
12
13
13
19
19
20
25
26
27

-------
      241



Appendix C (continued)
Chemical
Ferric dimethyl dithiocarbamate
Tetramethylthiuram disulfide
Ponceau MX
Dibutyl(diethylene glycol biaphthalate)
Pyridoxine hydrochloride
A9-Tetrahydrocannabinol
2,5,4'-Trichlorobiphenyl

Carbon tetrachloride
3' ,4'-Dichloropropionanilide
Tetrahydrothiophene-1 , 1-dloxide
Calcium carbimide
1 , 2 ,4-Trichlorobenzene
Acrolein

Ethylenebisisothlocyanate sulfide
Triphenyl tin hydroxide
Ponceau 4R
Isooctyl iaodecyl nylonate
Orange G
Hexachlorobenzene

Clindamycin hydrochloride
Triethyl phosphate
TR2379
tf-Methyl-tf- (1-naphthyl) f luoracetamide
2-Methyl-A-chlorophenoxy acetic acid
2-Methyl-A-chlorophenoxy propionic acid
Fenterol-HBr
Fominoben-HCl
Aapergillus fumigatus
2,3,7, 8-Tetrachlorodibenzo-p-dioxin
Peroxyacetyl nitrate
Epichlorohydrln
Ferric dimethyl dithiocarbamate
Tetramethylthiuram disulfide
Species
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Monkey
Brain
Rat
Rat
Monkey
Dog
Rat
Rat
Rat
Monkey
Rat
Dog
Monkey
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Rat
Rat
Rat
Dog
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Route Result
Oral
Oral
Oral
Oral
Oral
Oral +
Injection
Oral

Inhalation
Oral
Inhalation
Inhalation
Inhalation
Oral
Inhalation
Inhalation
Inhalation
Inhalation -
Inhalation
Oral
Oral
Oral
Oral
Oral
Oral
Oral +
Oral +
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral +
Oral
Oral
Oral
Oral
Oral
Oral +
Inhalation
Injection
Oral
Oral
Reference
33
33
15
16
38
39
39
23

1
2
3
3
3
4
5
5
6
35
35
7
8
9
10
11
12
22
24
28
31
13
13
14
19
19
20
21
40
40
25
26
27
29
30
32
33
33

-------
       242




Appendix C (continued)
Chemical
Ponceau MX
Dibutyl(diethylene glycol bisphthalate)
AHR-2438B
Hexach lorophene
Pyridoxine hydrochloride
A9-Tetrahydrocannabinol
2,5,4' -Tr ichlorobiphenyl

Tetrahydrothiophene-1 , 1-dioxide
Ethylenebisisothiocyanate sulfide
Clindamycin hydrochloride
TR2379
Hexachlorobenzene
2,5,4' -Trlchlorobiphenyl
Fenterol-HBr
Fominob en-HC 1
2,3,7, 8-Tetrachlorodibenzo-p-dioxin
AHR-2438B
Pyridoxine hydrochloride
A9-Tetrahydrocannabinol
1 , 2 ,4-Trichlorobenzene
Ethylenebisisothiocyanate sulfide
Hexachlorobenzene
2,5,4' -Trichlorophenyl
2,3,7, 8-Tetrachlorodibenzo-p-dioxin
A 9-Tetrahydrocannabinol
Tetrahydrothiophene-1 , 1-dioxide
Hexachlorobenzene
Clindamycin hydrochloride
Triethyl phosphate
A9-Tetrahydracannabinol
Species
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Monkey
Esophagus
Monkey
Dog
Rat
Rat
Dog
Rat
Rat
Dog
Monkey
Monkey
Monkey
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Rat
Monkey
Rat
Dog
Monkey
Monkey
Monkey
Rat
Monkey
Monkey
Gallbladder
Monkey
Dog
Rat
Dog
Dog
Rat
flat
Monkey
Monkey
Route Result
Oral
Oral
Oral
Oral +
Oral +
Oral
Injection
Oral +

Inhalation
Inhalation
Inhalation
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Inj ection
Inhalation
Inhalation
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Inj ection
Inhalation
Inhalation
Inhalation
Oral
Oral +
Oral
Oral
Oral
Injection
Reference
15
16
34
37
38
39
39
23

3
3
3
7
13
13
19
19
22
24
23
25
26
29
34
38
39
39
5
5
7
12
22
24
23
29
39
39
3
3
3
12
13
13
14
39
39

-------
       243
Appendix C (continued)
Chemical

Carbon tetrachloride
3 ' , A ' -Dichloropropionanilide
Tetrahydrothiophene-1 ,1-dioxide


Calcium carbimide
1,2, 4-Tr ichlorobenzene

Acrolein



Ethylenebisisothiocyanate sulfide
Triphenyl tin hydroxide
Ponceau 4R
Isooctyl isodecyl nitrate
Orange G
Hexachlorobenzene




Clindamycin hydrochloride

Triethyl phosphate
2,3,7, 8-Tetrachlorodibenzo-p-dioxin

TR2379

Quanethidine

l-Methyl-3-keto-A-phenylquinuclidinium
bromide
ff-Methyl-ff- (1-naphthyl) f luoracetamide
2-Methyl-4-chlorophenoxy acetic acid

2-Methyl-A-chlorophenoxy propionic acid
Fenterol-HBr
Fomlnoben-HCl
Aspergillus fumigatus
Peroxyacetyl nitrate
Epichlorohydrin
Ferric dimethyl dithiocarbamate
Tetramethylthluram disulfide
Ponceau MX
Dibutyl(diethylene glycol blsphthalate)
AHR-2438B
Barthrin
Species
Heart
Rat
Rat
Rat
Dog
Monkey
Rat
Monkey
Rat
Rat
Rat
Dog
Monkey
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Rat
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Dog
Dog
Rat
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Route Result

Inhalation
Oral +
Inhalation
Inhalation
Inhalation +
Oral
Inhalation
Inhalation
Inhalation +
Inhalation
Inhalation -
Inhalation -
Oral
Oral
Oral
Oral
Oral +
Oral
Oral
Oral
Oral +
Oral
Oral
Oral
Oral
Oral
Oral
Oral +
Oral +
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral +
Oral
Oral
Inhalation
Inj ection
Oral
Oral
Oral
Oral
Oral
Oral
Reference

1
2
3
3
3
4
5
5
6
35
35
35
7
8
10
9
11
12
22
24
28
31
13
13
14
17
29
19
19
18
18
18
18
20
21
40
40
25
26
27
30
32
33
34
15
16
34
36

-------
     244



Appendix C (continued)
Chemical
Dimethrin
Pyridoxine hydrochloride
A9-Tetrahydrocannabinol

2,5,4' -Trichlorobiphenyl

Carbon tetrachloride
3 ' ,4 ' -Dichloropropionanilide
Tetrahydrothiophene-1 , 1-dioxide


Calcium carbimide
1,2, 4-Tr ichlorobenzene

Acrolein



Ethylenebisisothiocyanate sulfide
Triphenyl tin hydroxide
Ponceau 4R
Isooctyl isodecyl nylonate
Orange G
Hexach lorob enz ene




Clindamycin hydrochloride

Trlethyl phosphate
2,3,7, 8-Tetrachlorodlbenzo-p-dioxin

TR2379

Quanethidine

l-Methyl-3-keto-4-phenylquinuclidinium
bromide
ff-Methyl-ff- (1-naphthyl) f luoractemide
2-Methyl-4-chlorophenoxy acetic acid

2-Methyl-4-chlorophenoxy propionic acid
Fenterol-HBr
Fominoben-HCl
Aspergillus fumlgatus
Peroxyacetyl nitrate
Epichlorohydrin
Ferric dimethyl dithiocarbamate
Species
Rat
Dog
Monkey
Monkey
Monkey
Kidney
Rat
Rat
Rat
Monkey
Dog
Rat
Rat
Monkey
Rat
Rat
Monkey
Dog
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Rat
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Dog
Dog
Rat
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Route Result
Oral
Oral
Oral
InJ ection
Oral

Inhalation +
Oral +
Inhalation
Inhalation
Inhalation
Oral
Inhalation +
Inhalation
Inhalation +
Inhalation +
Inhalation +
Inhalation +
Oral +
Oral +
Oral
Oral +
Oral
Oral +
Oral +
Oral +
Oral +
Oral +
Oral +
Oral
Oral
Oral
Oral
Oral +
Oral +
Oral
Oral
Oral
Oral
Oral +
Oral +
Oral +
Oral +
Oral
Oral
Oral +
Inhalation
Injection +
Oral
Reference
36
38
39
39
23

1
2
3
3
3
4
5
5
6
35
35
35
7
8
10
9
11
12
22
24
28
31
13
13
14
17
29
19
19
18
18
18
18
20
21
40
40
25
26
27
30
32
33

-------
     245



Appendix C (continued)
Chemical
Tetramethylthiuram disulfide
Ponceau MX
Dibutyl(diethylene glycol bisphthalate)
AHR-2438B
Barthrin
Dimethrln
Pyridoxine hydrochloride
A9-Tetrahydrocannabinol

2,5 ,4 '-Trichlorobiphenyl
Large
Carbon tetrachloride
3' ,4'-Dichloroproplonanilide
Tetrahydrothiophene-1 , 1 -dioxide


Acrolein
Ethylenebislsothiocyanate sulfide
Isooctyl isodecyl nylonate
Orange G
Hexachlo rob enz ene


Clindamycin hydrochloride

Triethyl phosphate
TR2379

Quanethidine

l-Methyl-3-keto-4-phenylquinuclidinium
bromide
AT-Methyl-ff- (1-naphthyl) f luoracetamide
Fenterol-HBr
Fominoben-HCl
Aspergillus fumigatus
2,3,7, 8-Tetrachlorodibenzo-p-dioxin
Peroxyacetyl nitrate
Ferric dimethyl dithiocarbamate
Tetramethylthiuram disulfide
AHR-2438B
Barthrin
Dimethrin
Pyridoxine hydrochloride
A9-Tetrahydrocannabinol
Species
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Monkey
Intestine
Rat
Rat
Rat
Dog
Monkey
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Dog
Rat
Rat
Rat
Dog
Dog
Rat
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Route Result
Oral
Oral +
Oral
Oral +
Oral +
Oral +
Oral
Oral
Injection
Oral +

Inhalation
Oral
Inhalation
Inhalation
Inhalation
Inhalation
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral +
Oral
Oral +
Oral
Oral
Injection
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Injection +
Reference
33
15
16
34
36
36
38
39
39
23

1
2
3
3
3
6
7
9
11
12
22
24
13
13
14
19
19
18
18
18
18
20
25
26
27
29
30
33
33
34
36
36
38
39
39

-------
    246
Appendix C (continued)
Chemical
2-Methyl-4-chlorophenoxy acetic acid
2-Methyl-4-chlorophenoxy propionic acid
2,5,4' -Trichlorobipheny 1

Carbon tetrachlorlde
3 ' , 4 ' -Dichloropropionanllide
Tetrahydrothiophene-1 , 1-dioxlde


Calcium carbimlde
1 , 2 ,4-Tr ichlorobenzene

Acrolein



Ethylenebislsothiocyanate sulfide
Triphenyl tin hydroxide
Isooctyl isodecyl nylonate
Ponceau 4R
Orange G
Hexachlorobenzene




Clindamycin hydrochloride

Triethyl phosphate
2,3,7, 8-Tetrachlorodibenzo-p-dioxin

TR2379

Quanethidine

l-Methyl-3-keto-4-phenylquinuclidinlum
bromide
ff-Methyl-ff- (1-naphthyl) fluoracetamide
2-Methyl-4-chlorophenoxy acetic acid

2-Methyl-4-chlorophenoxy proplonic acid
Fenterol-HBr
Fomlnoben-HCl
Asperglllus fumigatus
Peroxyacetyl nitrate
Epichlorohydrin
Ferric dimethyl dithiocarbamate
Tetramethylthiuram dlsulfide
Species
Rat
Rat
Monkey
Liver
Rat
Rat
Rat
Dog
Monkey
Rat
Rat
Monkey
Rat
Rat
Dog
Monkey
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Rat
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Dog
Rat
Dog
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Route Result
Oral
Oral
Oral

Inhalation +
Oral +
Inhalation
Inhalation
Inhalation +
Oral +
Inhalation +
Inhalation
Inhalation
Inhalation +
Inhalation +
Inhalation +
Oral
Oral
Oral +
Oral
Oral +
Oral
Oral
Oral +
Oral +
Oral +
Oral
Oral
Oral +
Oral +
Oral +
Oral +
Oral +
Oral
Oral
Oral
Oral
Oral
Oral +
Oral
Oral
Oral +
Oral +
Oral
Inhalation
Injection
Oral
Oral
Reference
40
40
23

1
2
3
3
3
4
5
5
6
35
35
35
7
8
9
10
11
12
22
24
28
31
13
13
14
17
29
19
19
18
18
18
18
20
21
40
40
25
26
27
30
32
33
33

-------
     247



Appendix C (continued)
Chemical
Ponceau MX
Dibutyl(dlethylene glycol bisphthalate)
AHR-2A38B
Barthrin
Dimethrin
Hexachlorophene
Pyrldoxine hydrochloride
A 9 -Tet rahydr o c annab inol
2,5,A'-Trichlorobiphenyl

Carbon tetrachlorlde
3' ,A'-Dichloropropionanilide
Tetrahydrothiophene-1 , 1-dioxide

Calcium carbimlde
1,2, A-Tr ichlorobenzene
Acrolein


Ethylenebisisothiocyanate sulfide
Triphenyl tin hydroxide
Isooctyl isodecyl nylonate
Orange G
Hexachl orob enz ene

Clindamycin hydrochloride
Triethyl phosphate
2,3,7, 8-Tet rachlorodibenzo-p-dioxin
TR2379
Quanethidine
l-Methyl-3-keto-A-phenylquinuclldinium
bromide
ff-Methyl-ff- (1-naphthyl) f luoracetamide
2-Methyl-A-chlorophenoxy acetic acid
2-Methyl-A-chlorophenoxy propionic acid
Fenterol-HBr
Fominoben-HCl
Species
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Monkey
Lung
Rat
Rat
Dog
Rat
Monkey
Rat
Rat
Monkey
Rat
Rat
Dog
Monkey
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Rat
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Dog
Rat
Dog
Rat
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Route Result
Oral +
Oral
Oral +
Oral +
Oral +
Oral
Oral
Oral +
Injection +
Oral +

Inhalation
Oral
Inhalation +
Inhalation +
Inhalation +
Oral
Inhalation
Inhalation -
Inhalation +
Inhalation +
Inhalation +
Inhalation +
Oral +
Oral +
Oral
Oral
Oral
Oral
Oral +
Oral +
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Reference
15
16
3A
36
36
37
38
39
39
23

1
2
3
3
3
A
5
5
6
35
35
36
7
8
9
10
12
2A
22
28
31
13
13
1A
17
29
19
19
18
18
18
18
20
21
AO
AO
25
26

-------
     248
Appendix C (continued)
Chemical
Aspergillus fumigatus
Peroxyacetyl nitrate
Ep ichlorohydr in
Ferric dimethyl dithiocarbamate
Tetramethylthiuram disulfide
AHR-2438B
Barthrin
Dimethrin
Pyridoxine hydrochloride
A9-Tetrahydrocannabinol
2,5,4'-Trichlorobiphenyl
Species
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Monkey
Route Result
Oral
Inhalation +
Injection +
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Injection +
Oral +
Reference
27
30
32
33
33
34
36
36
38
39
39
23
Lymph Node
Carbon tetrachloride
Tetrahydrothiophene-1 , 1 -dioxide
Calcium carbimide
Acrolein
Ethylenebisisothiocyanate sulfide
leooctyl isodecyl nylonate
Orange G
Hexachlorobenzene
Clindamycin hydrochloride
Triethyl phosphate
TR2379
Fenterol-HBr
Fominoben-HCl
Aspergillus fumigatus
2,3, 7 ,8-Tetrachlorodibenzo-p-dioxin
Peroxyacetyl nitrate
Ferric dimethyl dithiocarbamate
Tetramethylthiuram disulfide
AHR-2438B
Pyridoxine hydrochloride
A9-Tetrahydrocannablnol
2-Methyl-4-chlorophenoxy acetic acid
2-Methyl-4-chlorophenoxy propionic acid
2,5,4' -Tr ichlorobiphenyl
Rat
Rat
Dog
Monkey
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Dog
Rat
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Rat
Rat
Monkey
Inhalation
Inhalation
Inhalation
Inhalation
Oral +
Inhalation
Oral
Oral
Oral
Oral +
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral +
Inhalation
Oral +
Oral
Oral +
Oral
Oral
Injection
Oral
Oral
Oral
1
3
3
3
4
6
7
9
11
12
22
24
13
13
14
19
19
25
26
27
29
30
33
33
34
38
39
39 «
40
40
23

-------
    249



Appendix C (continued)
Chemical
Species
Route Result
Reference
Mammary Gland
Ethylenebisisothiocyanate sulfide
Orange G
Hexachlorobenzene
2,5,4' -Trichlorobiphenyl
A 9-Tet rahy drocannabinol
Rat
Rat
Dog
Monkey
Monkey
Monkey
Monkey
Monkey
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Injection
7
11
12
22
24
23
39
39
Muscle Tissue
Carbon tetrachloride
3 ' ,4 '-Dichloropropionanilide
Acrolein
Triphenyl tin hydroxide
Orange G
Clindamycin hydrochloride
TR2379
2-Methyl-4-chlorophenoxy acetic acid
2-Methyl-4-chlorophenoxy propionic acid
Fenterol-HBr
Fomlnoben-HCl
Aspergillus fumlgatus
2,3,7, 8-Tet rachlorodibenzo-p-d ioxin
Peroxyacetyl nitrate
Ferric dimethyl dithiocarbamate
Tetramethylthiuram disulfide
Pyridoxine hydrochloride
A9-Tetrahydrocannabinol
Nerve
Carbon tetrachloride
Quanethidine
l-Methyl-3-keto-4-phenylquinuclidinium
bromide
Hexachlorobenzene
Aspergillus fumlgatus
Pyridoxine hydrochloride
A9-Tetrahydrocannabinol
2-Methyl-4-chlorophenoxy acetic acid
2-Methyl-4-chlorophenoxy propionic acid
2,5,4' -Trichlorobiphenyl
Rat
Rat
Rat
Rat
Rat
Dog
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Tissue
Rat
Dog
Rat
Dog
Rat
Monkey
Monkey
Rat
Rat
Dog
Monkey
Monkey
Rat
Rat
Monkey
Inhalation
Oral
Inhalation
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Inhalation
Oral
Oral
Oral
Oral
Injection
Inhalation
Oral +
Oral +
Oral
Oral
Oral +
Oral +
Oral
Oral
Oral +
Oral
Injection
Oral
Oral
Oral +
1
2
6
8
11
13
13
19
19
21
40
40
25
26
27
29
30
33
33
38
39
39
1
18
18
18
18
22
24
28
27
38
39
39
40
40
23

-------
     250



Appendix C (continued)
Chemical

3' ,4'-Dichloropropionanilide
Calcium carbimide
Acrolein
Ethylenebisisothiocyanate sulfide
Triphenyl tin hydroxide
Ponceau MX
Isooctyl isodecyl nylonate
Orange G
Hexachlorobenzene



Clindamycin hydrochloride

Triethyl phosphate
TR2379

JV-Methyl-ff- (1-naphthyl) f luoracetamide
Fenterol-HBr
Fominoben-HCl
2,3,7, 8-Tetrachlorodibenzo-p-dioxin
Epichlorohydrin
Ferric dimethyl dithiocarbamate
Tetramethylthiuram disulfide
Ponceau MX
Dibutyl (diethylene glycol bisphthalate)
AHR-2438B
Barthrin
Dimethrln
Hexach lorophene
Pyridoxine hydrochloride
A9-Tetrahydrocannabinol

2-Methyl-4-chlorophenoxy acetic acid
2-Methyl-4-chlorophenoxy propionic acid
2,5,4' -Trichlorobiphenyl

Carbon tetrachloride
3* ,4'-Dichloropropionanilide
Tetrahydrothiophene-1 , 1-dioxide


Calcium carbimide
Acrolein
Ethvlenebisisothlocyanate sulfide
Species
Ovaries
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Rat
Dog
Rat
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Rat
Rat
Monkey
Pancreas
Rat
Rat
Rat
Dog
Monkey
Rat
Rat
Rat
Route Result

Oral
Oral +
Inhalation
Oral
Oral
Oral
Oral +
Oral +
Oral
Oral +
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral +
Injection
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Inj ectlon
Oral
Oral +
Oral

Inhalation
Oral
Inhalation
Inhalation
Inhalation
Oral
Inhalation
Oral
Reference

2
4
6
7
8
10
9
11
12
24
22
31
13
13
14
19
19
20
25
26
29
32
33
33
16
17
34
36
36
37
38
39
39
40
40
23

1
2
3
3
3
4
6
7

-------
     251



Appendix C (continued)
Chemical
Isooctyl isodecyl nylonate
Orange G
Hexachlorobenzene
Clindamycin hydrochloride
Triethyl phosphate
TR2379
tf-Methyl-ff- (1-naphthyl) f luoracetamide
2-Methyl-4-chlorophenoxy acetic acid
2-Methyl-4-chlorophenoxy propionic acid
Peroxyacetyl nitrate
Ferric dimethyl dithiocarbamate
Tetramethylthiuram disulfide
AHR-2438B
Barthrin
Dimethrin
A9-Tetrahydrocannabinol
2,5,4' -Tr ichlorobiphenyl

Carbon tetrachloride
Calcium carbimide
Ethylenebisisothiocyanate sulfide
Isooctyl Isodecyl nylonate
Orange G
Hexachlorobenzene
2,5,4' -Trichlorobiphenyl
Clindamycin hydrochloride
Triethyl phosphate
TR2379
Fenterol-HBr
Fominoben-HCl
Aspergillus fumigatus
2,3,7, 8-Tetrach lorodibenzo-p-dioxin
Ferric dimethyl dithiocarbamate
Tetramethylthiuram disulfide
AHR-2438B
Pyrldoxine hydrochloride
Species
Rat
Rat
Dog
Monkey
Monkey
Rat
Rat
Dog
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Monkey
Monkey
Monkey
Pituitary
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Monkey
Dog
Rat
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Route Result
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Inhalation
Oral
Oral
Oral
Oral
Oral
Oral +
Injection
Oral

Oral
Oral +
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Reference
9
11
12
22
24
31
13
13
14
19
19
20
21
40
40
30
33
33
34
36
36
39
39
23

1
4
7
9
11
12
22
24
23
13
13
14
19
19
25
26
27
29
33
33
34
38

-------
     252
Appendix C (continued)
Chemical
A9-Tetrahydrocannabinol

2-Methyl-4-chlorophenoxy acetic acid
2-Methyl-A-chlorophenoxy propionlc acid

Ethylenebislsothiocyanate sulfide
Hexachlorobenzene
Clindamycin hydrochloride

TR2379

2-Methyl-A-chlorophenoxy acetic acid

2-Methyl-A-chlorophenoxy propionic acid
Fenterol-HBr
Fominob en-HC 1
Aspergillus fumigatus
2,3,7, 8-Tet rachlorodibenzo-p-dioxin
Peroxyacetyl nitrate
Ferric dimethyl dithiocarbamate
Tetramethylthiuram disulflde
A^-Tetrahydrocannabinol

2,5,A'-Trichlorobiphenyl
Species
Monkey
Monkey
Rat
Rat
Prostate
Rat
Rat
Rat
Dog
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Monkey
Monkey
Monkey
Route Result
Oral
Injection
Oral
Oral

Oral
Oral
Oral
Oral ff
Oral
Oral
Oral
Oral
Oral +
Oral +
Oral +
Oral
Oral
Inhalation
Oral
Oral
Oral
Injection
Oral
Reference
39
39
40
AO

7
12
13
13
19
19
21
AO
40
25
26
27
29
30
33
33
39
39
23
Salivary Gland
Calcium carbimide
Acrolein
Ethylenebisisothiocyanate sulfide
Triphenyl tin hydroxide
Isooctyl iaodecyl nylonate
Orange G
Hexachlorobenzene


Clindamycin hydrochloride

TR2379

Fenterol-HBr
Fominoben-HCl
Aspergillus fumigatus
2,3,7, 8-Tetrachlorodibenzo-p-dioxin
Peroxyacetyl nitrate
Ferric dimethyl dithiocarbamate
Tetramethylthiuram disulfide
AHR-2A38B
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Rat
Dog
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Oral
Inhalation
Oral
Oral
Oral
Oral
Oral
Oral ,-
Oral *« •
Oral +
Oral
Oral
Oral
Oral +
Oral
Oral
Oral
Inhalation
Oral
Oral
Oral
A
6
7
8
9
11
12
22
2A
13
13
19
19
25
26
27
29
30
33
33
34

-------
      253



Appendix C  (continued)
Chemical
A9-Tetrahydrocannabinol

2,5,4' -Tr ichlorobiphenyl
2-Methyl-4-chlorophenoxy acetic acid
2-Methyl-4-chlorophenoxy propionic acid
Species
Monkey
Monkey
Monkey
Rat
Rat
Route Result
Oral
Injection
Oral
Oral
Oral
Reference
39
39
23
40
40
Sciatic Nerve
Ethylenebisisothiocyanate sulfide
Hexachlorobenzene
2,3,7, 8-Tet rachlorodibenzo-p-dioxin
Pyridoxlne hydrochloride

3* ,4'-Dichloropropionanilide
1,2, 4-Trichlorobenzene

Acrolein
Hexachlorobenzene
Aspergillus fumigatus
2,3, 7 ,8-Tetrachlorodibenzo-p-dioxin
Peroxyacetyl nitrate
AHR-2438B
A9-Tetrahydrocannabinol

Rat
Dog
Rat
Dog
Skin
Rat
Rat
Monkey
Rat
Dog
Rat
Rat
Rat
Rat
Monkey
Monkey
Oral
Oral
Oral
Oral +

Oral
Inhalation
Inhalation
Inhalation
Oral
Oral
Oral
Inhalation
Oral +
Oral
Injection
7
12
29
38

2
5
5
6
12
27
29
30
34
39
39
Small Intestine
Carbon tetrachloride
3' ,4'-Dlchloropropionanilide
Tetrahydrothiophene-1 , 1-dloxlde


Acrolein
Ethylenebisisothiocyanate sulfide
Isooctyl isodecyl nylonate
Orange G
Trlethyl phosphate
Hexachlorobenzene


Clindamycin hydrochloride

TR2379

Quanethldine

l-Methyl-3-keto-4-phenylquinuclidinium
bromide
ff-Methyl-ff- (1-naphthyl) f luoracetamide
2-Methyl-4-chlorophenoxy acetic acid

Rat
Rat
Dog
Rat
Monkey
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Rat
Dog
Rat
Dog
Dog
Rat
Dog
Rat
Rat
Rat
Rat
Inhalation
Oral
Inhalation
Inhalation
Inhalation
Inhalation
Oral
Oral
Oral +
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
1
2
3
3
3
6
7
9
11
14
12
22
24
13
13
19
19
18
18
18
18
20
21
40

-------
      254




Appendix C  (continued)
Chemical
2-Methyl-4-chlorophenoxy propionic acid
Fenterol-HBr
Fominoben-HCl
Aapergillus fumigatus
2,3,7, 8-Tetr achlorodibenzo-p-d ioxin
Peroxyacetyl nitrate
Ferric dimethyl dithiocar hamate
Tetramethylthiuram disulfide
AHR-2438B
Barthrin
Dimethrin
Pyridoxine hydrochloride
A9-Tetrahydrocannabinol
2,5,4' -Tr ichlorobiphenyl

1 , 2 ,4-Trichlorobenzene
Ethylenebisisothiocyanate sulfide
Isooctyl isodecyl nylonate
Hexachlorobenzene
Fenterol-HBr
Fominob en-HC 1
Aspergillus fumigatus
2,3,7, 8-Tetrachlorodibenzo-p-dioxin
Acrolein
Pyridoxine hydrochloride
2,5,4' -Trichlorobiphenyl

Carbon tetrachlorlde
3 ' , 4 ' -Dichloropropionanilide
Tetrahydrothiophene-1 , 1-dioxide
Calcium carbimide
1,2, 4-Tr ichlorobenzene
Acrolein
Ethylenebisisothiocyanate sulfide
Trinhenvl tin hydroxide
Species
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Monkey
Spinal Cord
Rat
Monkey
Rat
Rat
Dog
Monkey
Monkey
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Dog
Monkey
Spleen
Rat
Rat
Rat
Dog
Monkey
Rat
Rat
Monkey
Rat
Rat
Dog
Monkey
Rat
Rat
Route Result
Oral
Oral
Oral
Oral
Oral
Inhalation
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Injection
Oral

Inhalation
Inhalation
Oral
Oral
Oral
Oral +
Oral +
Oral
Oral
Oral
Oral
Oral
Inhalation
Inhalation
Oral +
Oral +

Inhalation +
Oral +
Inhalation
Inhalation
Inhalation
Oral +
Inhalation
Inhalation
Inhalation
Inhalation
Inhalation
Inhalation
Oral
Oral +
Reference
40
25
26
27
29
30
33
33
34
36
36
38
39
39
23

5
5
7
9
12
22
24
31
25
26
27
29
35
35
38
23

1
2
3
3
3
4
5
5
6
35
35
35
7
8

-------
     255



Appendix C (continued)
Chemical
Isooctyl isodecyl nylonate
Ponceau 4R
Orange G
Hexachlorobenzene

Clindamycin hydrochloride
Triethyl phosphate
2,3,7, 8-Tetrachlorodibenzo-p-dioxin
TR2379
tf-Methyl-ff- (1-naphthyl) f luoracetamide
2-Methyl-4-chlorophenoxy acetic acid
2-Methyl-4-chlorophenoxy propionic acid
Fenterol-HBr
Fominoben-HCl
Aspergillus fumigatus
Peroxyacetyl nitrate
Epichlorohydrin
Ferric dimethyl dithiocarbamate
Tetramethylthiuram disulfide
Ponceau MX
Dibutyl(dlethylenc glycol bisphthalate)
AHR-2438B
Barthrln
Dimethrin
Hexachlorophene
Pyridoxine hydrochloride
A9-Tetrahydrocannabinol

Carbon tetrachloride
3' ,4'-Dichloropropionanilide
Tetrahydrothiophene-1 , 1-dioxide
Acrolein
Ethylenebisisothiocyanate sulfide
Isooctyl isodecyl nylonate
Orange 6
Hexachlorob enz ene
Species
Rat
Rat
Rat
Dog
Monkey
Monkey
Rat
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Stomach
Rat
Rat
Rat
Dog
Monkey
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Route Result
Oral
Oral
Oral +
Oral
Oral
Oral
Oral +
Oral +
Oral
Oral
Oral
Oral
Oral +
Oral
Oral
Oral
Oral +
Oral
Oral
Oral
Oral
Oral
Inhalation
Injection
Oral +
Oral
Oral +
Oral
Oral +
Oral
Oral
Oral
Oral
Oral
Inj ection

Inhalation -
Oral
Inhalation
Inhalation
Inhalation
Inhalation
Oral
Oral
Oral
Oral +
Oral
Oral
Reference
9
10
11
12
22
24
28
31
13
13
14
17
29
19
19
20
21
40
40
25
26
27
30
32
33
33
15
16
34
36
36
37
38
39
39

1
2
3
3
3
6
7
9
11
12
22
24

-------
      256




Appendix C  (continued)
Chemical
Clindamycin hydrochloride

Triethyl phosphate
TR2379

Quanethidine

l-Methyl-3-keto-4-phenylquinuclidinium
bromide
#-Methyl-ff-(l-naphthyl)f luoracetamide
Fominoben-HCl
Fenterol-HBr
Aspergillus fumigatus
2,3,7, 8-Tetrachlorodibenzo-p-dioxin
Peroxyacetyl nitrate
Ferric dimethyl dithiocarbamate
Tetramethylthiuram disulfide
AHR-2438B
Barthrin
Dimethrin
Pyridoxine hydrochloride
A9-Tetrahydrocannabinol

2-Methyl-4-chlorophenoxy acetic acid
2-Methyl-A-chlorophenoxy propionic acid
2,5,4' -Tr ichlorob ipheny 1

Carbon tetrachloride
3' ,4 '-Dichloropropionanilide
Calcium carbimide
Acrolein
Ethylenebislsothiocyanate sulfide
Triphenyl tin hydroxide
Isooctyl isodecyl nylonate
Ponceau 4R
Orange G
Hexachlorobenzene


Clindamycin hydrochloride

Triethyl phosphate
2,3,7, 8-Tetrachlorodibenzo-p-dioxin

TR2379

Species
Dog
Rat
Rat
Rat
Dog
Rat
Dog
Rat
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Rat
Rat
Monkey
Testes
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Rat
Dog
Route Result
Oral +
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral +
Oral
Oral
Oral
Inhalation
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Injection
Oral
Oral
Oral

Inhalation +
Oral +
Oral +
Inhalation
Oral
Oral +
Oral
Oral
Oral
Oral +
Oral
Oral
Oral
Oral
Oral
Oral
Oral +
Oral
Oral
Reference
13
13
14
19
19
18
18
18
18
20
26
25
27
29
30
33
33
34
36
36
38
39
39
40
40
23

1
2
4
6
7
8
9
10
11
12
28
31
13
13
14
17
29
19
19

-------
        257
Appendix C (continued)
Chemical
ff-Methyl-ff- (1-naphthyl) f luoracetamlde
2-Methyl-4-chlorophenoxy acetic acid

2-Methyl-4-chlorophenoxy propionic acid
Fenterol-HBr
Fominoben-HCl
Aspergillus fumigatus
Peroxyacetyl nitrate
Epichlorohydrin
Ferric dimethyl dithiocarbamate
Tetramethylthiuram disulfide
Ponceau MX
DibutyKdiethylene glycol bisphthalate)
AHR-2438B
Barthrin
Dimethrin
Hexachlorophene
Fyridoxine hydrochloride
A 9-Tetrahydrocannab inol

2,5,4' -Trichlorobipheny 1

Calcium carbimide
Acrolein
Ethylenebisisothiocyanate sulfide
Isooctyl isodecyl nylonate
Orange G
2,3,7, 8-Tetrachlorodibenzo-p-d ioxin

Quanethidlne

l-Methyl-3-keto-4-phenylquinuclidinium
bromide
AMtethyl-JV-(l-naphthyl)fluoracetamide
Hexachlorobenzene
Fenterol-HBr
Fominoben-HCl
Aspergillus fumigatus
Peroxyacetyl nitrate
TR2379
A9-Tetrahydrocannabinol

2-Methyl-4-chlorophenoxy acetic acid
2-Methvl-4-chlorophenoxy propionic acid
Species
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Monkey
Thymus
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Rat
Dog
Rat
Rat
Monkey
Rat
Rat
Rat
Rat
Rat
Monkey
Monkey
Rat
Rat
Route Result
Oral +
Oral +
Oral
Oral
Oral +
Oral +
Oral
Inhalation +
Injection
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Inj ection
Oral +
Oral

Oral
Inhalation
Oral
Oral
Oral
Oral +
Oral +
Oral
Oral
Oral
Oral
Oral
Oral +
Oral
Oral +
Oral
Inhalation
Oral
Oral +
Injection +
Oral
Oral
Reference
20
21
40
40
25
26
27
30
32
33
33
15
16
34
36
36
37
38
39
39
23

4
6
7
9
11
17
29
18
18
18
18
20
24
25
26
27
30
34
39
39
40
40

-------
     258



Appendix C (continued)
Chemical

3 , ' 4 ' -Dichloropropionanilide
Tetrahydrothiophene-1 ,1-dioxide


Calcium carbimide
Acrolein

Ethylenebisisothiocyanate sulfide
Isooctyl isodecyl nylonate
Orange G
Hexachlorobenzene



Clindamycin hydrochloride

Triethyl phosphate
TR2379

2-Methyl-4-chlorophenoxy acetic acid

2-Methyl-4-chlorophenoxy propionic acid
Fenterol-HBr
Fominoben-HCl
Aspergillus fumigatus
2,3,7, 8-Tetrachlorodibenzo-p-dioxin
Peroxyacetyl nitrate
Ferric dimethyl dlthiocarbamate
Tetramethylthiuram disulfide
AHR-2438B
Barthrln
Dimethrin
Pyridoxine hydrochloride
A9-Tetrahydrocannabinol

2,5,4' -Trichlorobiphenyl

Tetrahydrothiophene-1 ,1-dioxide


Acrolein


Ethylenebisisothiocyanate sulfide
Orange G
Hexachlo rob enz ene

Species
Thyroid
Rat
Rat
Dog
Monkey
Rat
Rat
Dog
Rat
Rat
Rat
Dog
Monkey
Monkey
Rat
Rat
Dog
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Monkey
Trachea
Rat
Dog
Monkey
Rat
Dog
Monkey
Rat
Rat
Monkey
Monkey
Route Result

Oral
Inhalation
Inhalation
Inhalation
Oral +
Inhalation
Inhalation
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral +
Oral +
Oral
Oral
Oral
Oral
Oral +
Oral
Oral
Inhalation +
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Injection
Oral

Inhalation
Inhalation
Inhalation
Inhalation +
Inhalation +
Inhalation +
Oral +
Oral
Oral
Oral
Reference

2
3
3
3
4
6
35
7
9
11
12
22
24
31
13
13
14
19
19
21
40
40
25
26
27
29
30
33
33
34
36
36
38
39
39
23

3
3
3
6
35
35
7
11
22
24

-------
     259
Appendix C (continued)
Chemical
2,5,4' -Trichlorobiphenyl
Fenterol-HBr
Fominoben-HCl
Aspergillus fumigatus
2,3,7, 8-Tetrachlorodibenzo-p-dioxin
Peroxyacetyl nitrate
A^-Tetrahydrocannabinol
Carbon tetrachloride
3' ,4'-Dichloropropionanilide
Tetrahydrothiophene-1 , 1-dioxide
Acrolein
Ethylenebisisothiocyanate sulfide
Isooctyl isodecyl nylonate
Orange G
Hexachlorobenzene
Clindamycin hydrochloride
Triethyl phosphate
TR2379
Fenterol-HBr
Fominoben-HCl
Aspergillus fumigatus
2,3,7, 8-Tetrachlorodibenzo-p-dioxin
Peroxyacetyl nitrate
Ferric dimethyl dithiocarbamate
Tetramethylthiuram disulfide
AHR-2438B
Barthrin
Dimethrin
Pyrldoxine hydrochloride
A9-Tetrahydrocannabinol
2,5,4' -Trichlorobiphenyl
Species
Monkey
Rat
Rat
Rat
Rat
Rat
Monkey
Monkey
Urinary Bladder
Rat
Rat
Rat
Dog
Monkey
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Dog
Rat
Rat
Rat
Dog
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Monkey
2-Methyl-4-chlorophenoxy acetic acid Rat
2-Methyl-4-chlorophenoxy propionic
acid Rat
Route Result
Oral
Oral
Oral
Oral
Oral
Inhalation
Injection
Oral
Inhalation
Oral
Inhalation
Inhalation
Inhalation
Inhalation
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Inhalation
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Injection
Oral
Oral
Oral
Reference
23
25
26
27
29
30
39
39
1
2
3
3
3
6
7
9
11
12
22
24
13
13
14
19
19
25
26
27
29
30
33
33
34
36
36
38
39
39
23
40
40

-------
     260



Appendix C (continued)
Chemical

Acrolein
Ethylenebisisothiocyanate sulfide
Isooctyl isodecyl nylonate
Orange G
Hexachlorobenzene
2,5,4' -Trichlorobiphenyl
Clindamycin hydrochloride
TR2379
2,3,7, 8-Tetrachlorodibenzo-p-dioxin
Ferric dimethyl dithiocarbamate
Tetramethylthiuram disulfide
Pyridoxine hydrochloride
A9-Tetrahydrocannabinol
2-Methyl-A-chlorophenoxy acetic acid
2-Methyl-4-chlorophenoxy propionic acid
Species
Uterus
Rat
Rat
Rat
Rat
Dog
Monkey
Monkey
Monkey
Rat
Dog
Rat
Dog
Rat
Rat
Rat
Dog
Monkey
Monkey
Rat
Rat
Route Result

Inhalation
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral
Oral +
Oral
Oral
Oral
Oral
Injection
Oral
Oral
Reference

6
7
9
11
12
22
24
23
13
13
19
19
29
33
33
38
39
39
40
40

-------
                                  261

                       REFERENCES  FOR APPENDIX  C
 1.   Adams,  E.  M.,  H.  C.  Spencer,  V.  K.  Rowe,  D. D. McCollister,  and
     D.  D.  Irish.   1952.   Vapor Toxicity of  Carbon Tetrachloride  Deter-
     mined  by Experiments on Laboratory  Animals.  Ind. Hyg.  Occup. Med.
     6:50-66.

 2.   Ambrose, A. M.,  P.  S. Larson, J.  F.  Borzelleca,  and  R.  G.  Hennigar,  Jr.
     1972.   Toxicologic  Studies on 3',4'-Dichloropropionanilide.  Toxicol.
     Appl.  Pharmacol.  23:650-659.

 3.   Anderson,  M.  E.,  R.  A.  Jones, R.  G.  Mehl, T. A.  Hill, L. Kurlansik,
     and L.  J.  Jenkins,  Jr.   1977.  The  Inhalation Toxicity  of  Sulfolane
     (Tetrahydrothiophene-l,l-Dioxide).   Toxicol. Appl. Pharmacol.
     40:463-472.

 4.   Benitz, K. F., A. W. Kramer,  Jr., and G.  Dambach.  1965.   Comparative
     Studies on the Morphologic Effects  of Calcium Carbimide, Propylthio-
     uracil, and Disulfiram in Male Rats. Toxicol. Appl. Pharmacol.
     7:128-162.

 5.   Coate,  W.  B.,  W.  H.  Schoenfisch, T.  R.  Lewis, and W. M. Busey.   1977.
     Chronic, Inhalation Exposure of Rats, Rabbits, and Monkeys to
     1,2,4-Trichlorobenzene.  Arch. Environ. Health  23:249-255.

 6.   Feron, V.  J., A.  Kruysse, H.  P. Til, and H.  R.  Immel.  1978.  Repeated
     Exposure to Acrolein Vapour:   Subacute  Studies  in Hamsters,  Rats,
     and Rabbits.   Toxicology 9:47-57.

 7.   Freudenthal,  R. I., G. A. Kerchner, R.  L. Persing,  I. Baumel, and
     R.  L.  Baron.   1977.  Subacute Toxicity  of Ethylenebisisothiocyanate
     Sulfide in the Laboratory Rat.  J.  Toxicol.  Environ. Health
     2:1067-1078.

 8.   Gaines, T. B., and R. D. Kimbrough.  1968.  Toxicity of Fentin Hydroxide
     to Rats.  Toxicol.  Appl. Pharmacol. 12:397-403.

 9.   Gaunt, I. F., J.  Colley, P. Grasso, M.  Creasey,  and S.  D.  Gangolli.
     1969.   Acute  (Rat and Mouse) and Short-Term (Rat) Toxicity Studies
     on Isooctyl Isodecyl Nylonate.  Food Cosmet.  Toxicol. 7:115-124.

10.   Gaunt, I. F., M.  Farmer, P. Grasso, and S. D.  Gangolli.  1967.  Acute
     (Mouse and Rat) and Short-Term (Rat) Toxicity Studies on Ponceau 4R.
     Food Cosmet.  Toxicol. 5:187-194.

11.   Gaunt, I. F., M. Wright, P. Grasso, and S. D.  Gangolli.  1971.   Short-
     Term Toxicity of drange G in Rats.  Food Cosmet. Toxicol. 9:329-342.

-------
                                   262

12.  Gralla, E. J., R. W. Fleischman, Y. K. Luthra, M. Hagopian,
     J. R. Baker, H. Esber, and W. Marcus.  1977.  Toxic Effects of
     Hexachlorobenzene After Daily Administration to Beagle Dogs for One
     Year.  Toxicol. Appl. Pharmacol. 40:227-239.

13.  Gray, J. E., R. N. Weaver, J. A. Bollert, and E. S. Feenstra.
     1972.  The Oral Toxicity of Clindamycin in Laboratory Animals.
     Toxicol. Appl. Pharmacol. 21:516-531.

14.  Gumbmann, M. R., W. E. Gagne, and S. N. Williams.  1968.  Short-Term
     Toxicity Studies of Rats Fed Triethyl Phosphate in the Diet.
     Toxicol. Appl. Pharmacol. 12:360-371.

15.  Hall, D. E., F. S. Lee, and F. A. Fairweather.  1966.  Acute  (Mouse
     and Rat) and Short-Term (Rat) Toxicity Studies on Ponceau MX.  Food
     Cosmet. Toxicol. 4:375-382.

16.  Hall, D. E., P. Austin, and F. A. Fairweather.  1966.  Acute  (Mouse
     and Rat) and Short-Term (Rat) Toxicity Studies on Dibutyl  (Diethylene
     Glycol Bisphthalate).  Food Cosmet. Toxicol. 4:383-388.

17.  Harris, M. W., J. A. Moore, J. G. Vos, and B. N. Gupta.  1973.
     General Biological Effects of TCDD in Laboratory Animals.  Environ.
     Health Perspect. 5:101-109.

18.  Hartnagel, R. E., B. M. Phillips, E. H. Fonseca, and R. L. Kowalski.
     1976.  The Acute and Target Organ Toxicity of l-Methyl-3-Keto-4-
     Phenylquinuclidinium Bromide  (MA 540) and Guanethidine in  the Rat
     and Dog.  Arzneim.-Forsch. 26:1671-1672.

19.  Hartnagel, R. E., B. M. Phillips, P. J. Kraus, R. L. Kowalski, and
     E. H. Fonseca.   1975.  A  Subchronic  Study of the Toxicity  of  an
     Orally Administered Benzoquinolizinyl Derivative in the Rat and
     Dog.  Toxicology. 4:215-222.

20.  Hashimoto, Y., T. Makita, H. Miyata, T. Noguchi, and G. Ohta.  1968.
     Acute and  Subchronic Toxicity of a New Fluorine Pesticide, #-Methyl-
     tf-(l-Naphtyl)fluoracetamide.  Toxicol. Appl. Pharmacol. 12:536-547.

21.  Hattula, M. L.,  H. Elo, H. Reunanen, A. U. Arstila, and T. E. Sorvari.
     1977.  Acute  and Subchronic Toxicity of 2-Methyl-4-Chlorophenoxy
     Acetic Acid  (MCPA) in Male Rat.  I.  Light Microscopy and  Tissue
     Concentrations of MCPA.   Bull. Environ. Contain. Toxicol. 18:152-158.

22.  latropoulos, M. J., J. Bailey, H. P. Adams, F. Coulston, and
     W. Hobson.  1978.  Response of Nursing Infant Rhesus to Clophen
     A-30 or Hexachlorobenzene Given  to Their Lactating Mothers.   Environ.
     Res. 16:38-47.

23.  latropoulos, M. J., G. R. Felt,  H. P. Adams, F. Korte, and F. Coulston.
     1977.  Chronic Toxicity of 2,5,4'-Trichlorobiphenyl in Young  Rhesus
     Monkeys.  II.  Histopathology.   Toxicol. Appl. Pharmacol.  41:629-638.

-------
                                  263

24.  latropoulos, M. J., W. Hobson,  V.  Knauf,  and H.  P.  Adams.   1976.
     Morphological Effects of Hexachlorobenzene Toxicity in Female Rhesus
     Monkeys.  Toxicol. Appl. Pharmacol. 37:433-444.

25.  Kast, A., Y. Tsunerari, M. Honma,  J. Nishikawa,  T.  Shibata, and
     M. Torii.  1975a.  Acute, Subacute, and Chronic  Toxicity Studies of
     the Beta-Sympathomimetic, Fenterol-HBr on Rats,  Mice, and Rabbits.
     Oyo Yakuri. Sendai 10(1):45-71.

26.  Kast, A., Y. Tsunenari, M. Honma,  J. Nishikawa,  T.  Shibata, and
     M. Torii.   1975&.  Acute, Subacute, and Chronic Toxicity Studies
     of an Amino-Halogen-Substituted Benzylamine (Fominoben) in Rats and
     Mice.  Oyo Yakuri. Sendai 10(1):31-43.

27.  Khor, G. L., J. C. Alexander, J. H. Lumsden, and G. J. Losos.
     1976.  Safety Evaluation of Aspergillus fwrrlgatua Grown on Cassava
     for Use as an Animal Feed.  Can. J. Comp. Med. 41:428-434.

28.  Kimbrough, R. D., and R. E. Linder.  1974.  The Toxicity of Tech-
     nical Hexachlorobenzene in the Sherman Strain Rat.   A Preliminary
     Study.  Res. Commun. Chem. Pathol. Pharmacol. 8(4):653-664.

29.  Kociba, R. J., P. A. Keeler, C. N. Park, and P.  J.  Gehring.  1976.
     2,3,7,8-Tetrachlorodibenzo-p-dioxin  (TCDD):  Results of a 13-Week
     Oral Toxicity Study in Rats.  Toxicol. Appl. Pharmacol. 35:553-574.

30.  Kruysse, A., V.  J. Feron, H. R. Immel, B.  J. Spit, and G. J. Van  Esch.
     1977.  Short-Term Inhalation Toxicity Studies with Peroxyacetyl
     Nitrate in  Rats.  Toxicology 8:231-249.

31.  Kuiper-Goodman,  T., D. L. Grant,  C.  A. Moodie, G.  0. Korsrud,  and
     I. C. Munro.   1977.   Subacute Toxicity of  Hexachlorobenzene  in the
     Rat.  Toxicol. Appl.  Pharmacol. 40:529-549.

32.  Lawrence, W. H.,  M. Malik, J. E.  Turner, and J. Autian.   1972.
     Toxicity Profile of Epichlorohydrin.  J. Pharmacol.  Sci.  61(11):
     1712-1717.

33.  Lee, C-C.,  J.  Q.  Russell, and J.  L.  Minor.   1978.  Oral  Toxicity of
     Ferric Dimethyl  Dithiocarbamate (Ferbam) and Tetramethylthiuram
     Disulfide  (Thiram) in Rodents.  J.  Toxicol.  Environ.  Health  4:93-106.

34.  Luscombe, D. K.,  and  P.  J. Nicholls.   1973.  Acute and Subacute Oral
     Toxicity of AHR-2438B,  a Purified Lignosulphonate, in Rats.   Food
     Cosmet. Toxicol.  11:229-237.

35.  Lyon, J. P., L.  J. Jenkins,  Jr.,  R.  A.  Jones, R. A.  Coon, and J. Siegel.
     1970.   Repeated  and Continuous  Exposure of Laboratory Animals to
     Acrolein.   Toxicol. Appl.  Pharmacol. 17:726-732.

-------
                                  264

36.  Masri, M. S., A. P. Hendrickson, A. J. Cox, Jr., and F. DeEds.  1964.
     Subacute Toxicity of Two Chrysanthemumic Acid Esters:  Barthrin and
     Dimethrin.  Toxicol. Appl. Pharmacol. 6:716-725.

37.  Nakaue, H. S., F. N. Dost, and D. R. Buhler.  1973.  Studies on the
     Toxicity of Hexachlorophene in the Rat.  Toxicol. Appl. Pharmacol.
     24:239-249.

38.  Phillips, W.E.J., J.H.L. Mills, S. M. Charbonneau, L. Tryphonas,
     G. V. Hatina, Z. Zawidzka, F. R. Bryce, and I. C. Munro.  1978.
     Subacute Toxicity of Pyridoxine Hydrochloride in the Beagle Dog.
     Toxicol. Appl. Pharmacol. 44:323-333.

39.  Thompson, G. R., R. W. Fleischmann, H. Rosenkrantz, and M. C. Braude.
     1974.  Oral and Intravenous Toxicity of A9-Tetrahydrocannabinol in
     Rhesus Monkeys.  Toxicol. Appl. Pharmacol. 27:648-665.

40.  Verschuuren, H. G., R. Kroes, and E. M. Den Tonkelaar.  1975.  Short-
     Term Oral and Dermal Toxicity of MCPA and MCPP.  Toxicology
     3:349-359.

-------
            5.  CHRONIC TOXICITY AND CARCINOGEN1CITY TESTING

5.1  INTRODUCTION

     Tests designed to evaluate chronic toxicity and those designed to
evaluate carcinogenic effects have many similar considerations.   Both
are subject to various design factors, including species,  strain, sex,
age, and number of the test and control animals; duration  of test;  route
of exposures; dosage levels; and use of data evaluations.   Both studies
are costly and involve extensive manpower and time requirements.   However,
the two tests are also different in many ways.   There is little uniformity
in the literature regarding the purposes, methods, and nomenclature of
tests for chronic toxicity.  Some investigators seek only  the demonstration
of a "safe" or "harmless" dose, while other workers attempt to observe
the nature and severity of toxic effects as well as to determine a maximum
"no-observed-adverse-effect" dose level (Friedman, 1973).   In contrast,
for carcinogenicity tests, tumor1 formation and increased  tumor incidence
are the major end points.  Tumor incidence, tumor latency, and in some
cases tumor multiplicity are the parameters used to determine sensitivity
of the test animals to the carcinogenic challenge.  Short-term in vivo
tests can also be useful, and several types are included as examples of
intermediates between the lifetime studies and the various short-term in
vitro tests.  Therefore, in the following subsections, chronic toxicity
and carcinogenicity tests and the factors that affect them will be dis-
cussed together when possible, pointing out both the similarities and
dissimilarities.  There will also be discussions of topics that  pertain
only to each individual test.  Additionally, some topics usually considered
part of chronic toxicity testing, such as reproductive and behavioral
effects, neurotoxicity, and teratogenicity, are discussed elsewhere  in
this report or later publications and are not included in this section.
         this report, the general term "tumor" will apply  to  either
benign or malignant neoplasms.
                                   265

-------
                                   266
5.2  TEST ANIMALS

     Various aspects of the selection of species and strain of experimental
animal will be discussed in the context of long-term toxicity tests, which
include assays for chronic toxicity and carcinogenicity.  Species- and
strain-specific responses and spontaneous tumor incidence in test animals
will be examined through a review of  the literature.

5.2.1  Species
     Because the usual purpose of chronic toxicity and carcinogenicity
testing is to predict adverse effects of chemicals in man, test animals
should be chosen that closely resemble man with respect to absorption,
distribution, metabolism, excretion,  and target site effect of the toxic
substance (Weil, 1972).  Unless information  to the contrary is available
from previous metabolic, pharmacodynamic, or subchronic studies, the most
sensitive species and strain should be selected for chronic toxicity and
carcinogenicity testing (National Academy of Sciences, 1977).
     No test animal has been found to be an  ideal surrogate for man
under all test conditions (Krasovskii, 1976; Rail, 1969; Shubik, 1972);
furthermore, great variability of response exists among different species
(Hodge et al., 1967).  Nevertheless,  a variety of species can respond to
individual toxicants in a manner useful for  chronic testing.  Thus, dogs,
cats, and nonhuman primates, but not mice or rats, have been found suitable
for testing methylmercury compounds (National Academy of Sciences, 1977),
and the rhesus monkey is more similar to humans in the dermal absorption
of certain compounds such as benzoic  acid, hydrocortisone, and testosterone
than is the pig, rat, or rabbit (Fancher, 1978).  The chicken more closely
resembles man than other species tested for  response to demyelinating effects
of triorthocresyl phosphate and other organophosphorus compounds (Barnes
and Denz, 1954).  In a similar vein, monkeys are recommended for inhalation
experiments because of anatomical similarities to man, cats for compounds
likely to produce methemoglobinemia, pigtail monkeys for methanol toxicity,
and dogs or rats for cholinesterase inhibition studies (Fancher, 1978).

-------
                                   267
     5.2.1.1  Chronic Toxicity Testing - Despite the need for manlike
metabolic responses in test animals, in the ultimate analysis practical
considerations drastically limit the choice of animals for chronic
testing.  Because the test period must encompass a major portion of the
life span of the test species, short-lived mammalian species usually
receive first consideration.  Precedent, convenience, and economic
considerations dictate the use of rats or mice in the majority of all
chronic toxicity tests (Stevenson, 1979).  For example, in more than
100 computer-selected publications on noncarcinogenic chronic toxicity
examined during the preparation of this chapter (listed in the Bibliog-
raphy) , the frequency of occurrence of test animals was:  rat, 54%; dog,
24%; monkey, 9%; mouse, 6%; rabbit, 4%; guinea pig, 3%; and gerbil, 1%.
Only a few of these studies exceeded 2 years in duration.  On the basis
of animals exposed to the test material over a major portion of their
life spans, the above data on animal frequency show a strong bias with
respect to short-lived rodents: rat, 88%; mouse, 8%; dog, 2%; and gerbil,
2%.
     The dominating effect of practical considerations in the choice of
test animals for chronic toxicity testing is further reflected in animals
used in 134 long-term studies published in Toxicology and Applied Pharma-
cology between 1959 and 1966  (Benitz, 1970):  rat, 43.3%; dog, 38.1%;
monkey, 6.7%; mouse, 3.7%; rabbit, 3.0%; chicken and guinea pig, 1.5%;
and gerbil, 0.7%.  A survey of all studies published in  the same journal
during 1975 revealed a continuing preference for use of  short-lived
rodents:  rat, 43.7%; mouse,  15.9%; rabbit, 7.6%;  dog,  7.5%;  primate,
7.3%; guinea pig, 3.7%; human, 3.4%; fish, 3.0%; cat,  2.2%; hamster,
1.1%; chicken, 0.9%; swine, 0.7%; duck, 0.6%; quail, 0.6%;  cow,  0.4%;
sheep, 0.4%; frog, 0.2%; goat, 0.2%; sea lion,  0.2%; and snail,  0.2%
(Fancher, 1978).
     When relatively long-lived nonrodent  animals  are  the test  species
of choice and it is not feasible  to expose the  animals to the test
material over a major  fraction of  their life  span, some authorities
recommend conducting kinetic  studies to determine  when steady-state
tissue concentrations  of  the  test  chemical and  its metabolites have been
achieved.  Treatment of the animal for a substantial period after attaining

-------
                                   268
 steady-state  kinetics would  then  partially  substitute  for a  lifetime
 study  and  provide  added  assurance of  the validity of experimental  conclu-
 sions  (National Academy  of Sciences,  1977;  World Health Organization,
 1978).
     When  a rodent is chosen as the principal  test  animal in a  chronic
 toxicity test, most authorities recommend including a  second species
 that is not a rodent in  order to  reveal a broader range of toxic effects
 (Barnes and Denz,  1954;  Zbinden,  1973).  Usually, the  dog is chosen
 (Federal Register,  1978; Goldenthal,  1968;  Page, 1977Z?), but other
 species may be selected  if their  metabolic  processes are thought to
 resemble those of  man.
     5.2.1.2  Carcinogenicity Testing — Rodents, mainly the  rat, mouse,
 and Syrian hamster, are  generally selected  for large-scale screening of
 suspected  carcinogens (Ministry of Health and  Welfare  Canada, 1975;
 National Academy of Sciences, 1975; Page, 19772?).   Utilization  of  these
 species is based neither on  the established similarities to  man nor on
 biochemical,  physiological,  or anatomical characteristics but on the
 ability to test large numbers of  compounds  in  these animals  in  a rela-
 tively short  time.  Although the  susceptibility of  simians and  the dog
 to chemical carcinogenesis has been established with several groups of
 chemicals, the long latency  period in these species discourages their
 use in carcinogenicity screening  (Ministry  of  Health and Welfare Canada,
 1975;  National Academy of Sciences, 1975; U.S. Food and Drug Administration,
 1971).  Dogs  and monkeys may be useful for  extrapolation purposes, but
 only in highly select situations  (Page, 1977&).
     The selection of the most appropriate  animal system is  an  important
 factor in  tests for carcinogenicity,  and species differences in response
 to various chemical carcinogens must  be a major consideration in that
 selection.  Responses of various  mammalian  species  to  chemical  carcinogens
within selected chemical classes  are  reviewed  in the following  subsections.
     5.2.1.2.1  Polynuclear  aromatic  carcinogens —  Tumor induction with
polynuclear aromatic compounds has been demonstrated in a variety  of
species using different  routes of administration.   The following section
includes comparisons of  species'  responses  to  a few of these compounds.
The tumorigenic response of  animals to a particular carcinogen  can vary

-------
                                   269
according to route of administration of the test material, and this is
demonstrated in Sect. 5.3; thus, valid comparisons can be made between
species only when the same route of exposure is used.   Therefore, the
studies are grouped according to mode, of administration of the carcinogen.
     Tumors have been induced in many species by subcutaneous injection
of polycyclic aromatic carcinogens or by subcutaneous implantation of
special discs which have been impregnated with the carcinogens.  For
instance, when injected subcutaneously into newborn mice, 7,12-dimethyl-
benz(a)anthracene has been found to be a potent leukemogen (Pietra,
Rappaport, and Shubik, 1961).  Of 27 Swiss albino mice injected with 30
to 40 yg dimethylbenzanthracene in 1% gelatin, 29.6% developed malignant
lymphoma by 12 to 27 weeks after injection, and 85.4% developed pulmonary
tumors.  However, when newborn Lewis rats were injected subcutaneously
with 10 to 1000 yg of dimethylbenzanthracene, the only tumors related to
the treatment were sarcomas induced at the site of injection (Toth and
Shubik, 1963).  The number and latent periods of the tumors were dose
related.  Thus, it seems that newborn Swiss mice respond systemically to
subcutaneously administered dimethylbenzanthracene, while newborn Lewis
rats respond locally.
     Pott, Brockhaus, and Huth (1973) found the subcutaneous tissue of
the NMRI mouse to be more sensitive than that of the Wistar rat to
benzo(a)pyrene.  Female NMRI mice developed malignant sarcomas at the
site of injection of benzo(a)pyrene in tricaprylin (Pott, Tomingas, and
Misfeld, 1977).  Single doses of 3.3 to 270 yg of the carcinogen were
administered.  A linear dose relationship was observed in the range of
3.3 to 90 yg for an induction time of 20 to 41 weeks.  Survival time was
21 to 84 weeks, and the tumor incidence was 8% to 80%.
     When sensitive strains of Syrian hamsters were injected subcutane-
ously with 500 yg of benzo(a)pyrene in tricaprylin, sarcomas were  palpa-
ble at 20 weeks (Homburger et al., 1972).  In the experiments  of Pott  et
al. and Homburger et al. the average induction time for  the appearance
of subcutaneous tumors was the same for mice and hamsters.  However,  the
dose required to produce tumors in hamsters  in 20 weeks  was  two  times
the dose required to produce tumors in mice  in the same  time.   If  one
assumes that the dose response in hamsters is linear,  the mice would be

-------
                                   270
 more sensitive.   In the same hamster study,  500 yg of 3-methylcholanthrene
 induced sarcomas at the injection site  in 17 weeks;  in an earlier
 experiment (Homburger and Hsuch,  1970),  sarcomas appeared in hamsters
 only 3 weeks after subcutaneous injection of 7,12-dimethylbenzanthracene
 in tricaprylin.
      Hartley guinea pigs, on the  other  hand, exhibited relatively pro-
 longed induction times of 7 to 17 months after methylcholanthrene was
 injected into the abdominal wall  (Dale  et al., 1973).  One or two doses
 of 4 mg of the carcinogen in sesame  oil  produced sarcomas in 30% of the
 animals surviving for 6 months.  The dose of methylcholanthrene admin-
 istered was 8 to 16 times higher  than the dose that,  in the study of
 Homburger et al.  (1972), induced  subcutaneous tumors  in hamsters in 17
 weeks.  Thus, the guinea pig seems relatively resistant to subcutaneous
 tumor induction with 3-methylcholanthrene.
      A few years earlier O'Gara and  Kelly (1965) attempted to produce
 tumors in rhesus and cynomalgus monkeys  using conditions known to produce
 malignant tumors in rodents.  Dibenzo(a,i)pyrene and  3-methylcholanthrene
 injected subcutaneously or intradermally induced local keratoacanthomas,
 papillomas, and giant cell granulomas but no sarcomas or carcinomas.
      The squirrel monkey (Saimiri soireus)  is the second most widely
 used subhuman primate in biomedical  research,  and by  1969 no reports had
 been made of tumor induction in this animal.  Steinmuller, Dillingham,
 and Prehn (1969)  attempted to induce tumors  with subcutaneously implanted
 discs of paraffin impregnated with 5% 3-methylcholanthrene (one disc
 per 25 g of body weight).  No tumors or neoplastic changes were found
 4  years later in any of eight monkeys tested.   In addition, autochthonous
 skin fragments implanted subcutaneously with 3-methylcholanthrene crystals
 failed to give rise to tumors in  4 years in  the three monkeys treated.
 No  signs of toxicity were seen in monolayers of monkey kidney epithelial
 cells  grown in vitro with 5% 3-methylcholanthrene discs during 3 weeks
 of  exposure,  although Alfred et al.  (1964),  using identical techniques,
had  produced  marked toxicity in mouse cells.
     The most frequent explanation for  the differences in tumor response
between  rodents and primates is based on the assumption of a direct
relationship  between the induction time  of tumors and the life span of

-------
                                   271
the animal.  However, experiments of Sugiura, Smith, and Sunderland
(1956) do not support this but tend to emphasize differences In sensi-
tivity among the species to a particular carcinogen.  Thus, skin painting
with MH101, a high boiling catalytically cracked oil, in repeated doses
produced papillomas in six rhesus monkeys and, later, malignant change
in three of them.  This was the first demonstration of tumor induction
in Macaaus rhesus.  In the same experiment, mice and rabbits developed
papillomas and cancers, but rats and guinea pigs did not.  Table 5.1
demonstrates the lack of a relationship between life span and tumor
induction in various species (e.g., papilloma induction required 1/25 of
the life span of the mouse, 1/18 of the life span of the monkey, but
only 1/70 of the life span of the rabbit).  Steinmuller, Dillingham, and
Prehn (1969) concluded from the experiment of Sugiura, Smith, and
Sunderland (1956) and from their own data that the species-specific
response to carcinogens was more evident than a relationship between
induction time and life span.  They also suggested that primates are
more resistant than rodents to the polycyclic hydrocarbons.
     In contrast to squirrel monkeys and rhesus monkeys, primates lower
in the phylogenetic scale seem to be responsive to the carcinogenic
effects of polycyclic hydrocarbons (Adamson, Cooper, and O'Gara, 1970)
with the difference in response occurring approximately at the evolu-
tionary level of the marmoset.  Levy (1963) injected a marmoset subcu-
taneously with 2 mg of 3-methylcholarithrene, and a fibrosarcoma appeared
in 10 months; Noyes  (1969) induced a fibrosarcoma and a rhabdomyosarcoma
in 16 months after injection of benzo(a)pyrene and dimethylbenzanthracene,
respectively, into opposite flanks of a cottontop marmoset.  Also, Noyes
(1968) observed  fibrosarcomas in three tree shrews  (Tup-ia glis) 6 months
after injecting  benzo(a)pyrene.  In a similar study, Adamson, Cooper,
and O'Gara (1970) injected six tree shrews with single  subcutaneous
doses of 10 mg methylcholanthrene dissolved  in 1 mL  olive  oil.  At  the
same time, 12 galagos were injected intradermally or subcutaneously with
3 to 10 mg benzo(a)pyrene.  The three tree shrews that  survived developed
tumors at  the injection site, the first appearing 14 months  after  injec-
tion.  Of  the surviving 12 galagos, one developed a tumor  at the site  of
injection  in 26  months.  The fibrosarcomatous nature of the  tumors

-------
             Table 5.1.   Some factors in the experiments with MH 101 upon a variety of species
Species

Mouse
Rat

Guinea pig
Rabbit
Monkey
Time to first
appearance
(days)

Papilloma
20 (l/25)fc
0

0
26 (1/70)
322 (1/18)


Approximate
Dose weight of
(g) animal
tn\
Dose per kg
(g)
Area
treated*2
(cm2)
Dose per cm2
(g)
Approximate
life span
(years)
Cancer V6'
70
0

0
411
1373
(1/10)



(1/4)
(1/4)
0.015
0.1

0.1
0.5
0.2-1.0
25
200

400
3000
7000
0.6
0.5

0.25
0.17
0.03-0.14
1
4

4
16
4-35
0.
0.

0.
0.
0.
015
025

025
031
05-0.03
2
3

6
5
16

NJ
•vl
Ni


 Refers to a single painted site.
 Numbers in parenthesis represent portion of life span required for tumor induction.
Source:  Adapted from Suguira, Smith, and Sunderland, 1956.  Reprinted with permission of the publisher.

-------
                                   273
induced in these species [and the failure to induce sarcomas in rhesus
and cynamolgus monkeys (O'Gara and Kelly, 1965) and in squirrel monkeys
(Steinmuller, Dillingham, and Prehn, 1969)] led Adamson et al. to suggest
that prosimians, which include tree shrews, lemurs, lorises, and tarsiers,
may resemble rodents more than the higher primates in their reactions to
polycyclic hydrocarbon carcinogens.
     Although many skin carcinogenesis experiments have been conducted
using the two-stage, initiation-promotion technique, promotion studies
are omitted from this section to simplify discussion.  Skin tumor induc-
tion times are compared for mice, rats, and hamsters in the following
examples.
     Weekly cutaneous applications of 0.1% (or 0.025 mg per dose) 9,10-
dimethyl-l,2-benzanthracene to the skin of noninbred mice induced papil-
lomas in 5 to 6 weeks and carcinomas in 20 to 40 weeks (Neiman, 1968).
The induction times for both papillomas and carcinomas were reduced con-
siderably by presensitization with nontumorigenic doses of the carcin-
ogen.  In a later study with C57 B1/6J mice, Wislocki et al.  (1977)
noted the appearance of squamous cell carcinomas after 30 weeks of
biweekly applications (15 skin paintings) of benzo(a)pyrene.
     Similar experiments have been conducted in rats and hamsters.  Mul-
tiple weekly doses of 1% dimethylbenzanthracene applied to the skin of
hamsters gave rise to papillomas and squamous cell carcinomas.  The
average latent period was 16 weeks (Delia Porta et al., 1956).  Doses of
0.5% of the same carcinogen applied to the skin of Fischer rats produced
squamous cell carcinomas after 6 months of skin painting (Zackheim,
1964).  Table 5.2 summarizes the results of the three experiments with
dimethylbenzanthracene.  The differences in dose administered in the
three studies increase the difficulty of comparing responses  of the
animals.  Nonetheless, the limited data indicate that mice are more
sensitive than hamsters and rats to skin carcinogenesis with  9,10-
dimethyl-l,2-benzanthracene, supporting the view that mouse skin generally
exhibits greater sensitivity than rat skin to carcinogenesis  with poly-
cyclic aromatic hydrocarbons (Weisburger, 1976).
     Tumors of the respiratory tract have been  induced by polycyclic
aromatic hydrocarbons in the various species by special techniques,

-------
                                   274
            Table  5.2.   Induction  of  skin  carcinomas  in mice,
             hamsters,  and  rats with  weekly applications of
                    9,10-dimethyl-l,2-benzanthracene
        Species
Dose
Induction time
 Number of
applications
Inbred mice
Syrian
hamsters
Fischer rats
0.1% (0.25 mg)
1%
0.5%
5
4
6
to 10
months
months
, average
months , average
20
16
24
to 40


 which include intratracheal injection.   In the first report of tumor
 induction by the intratracheal route of administration,  weekly instil-
 lations  of 50 yg dimethylbenzanthracene in 1% gelatin for up to 45 weeks
 produced tracheobronchial  carcinomas in hamsters (Delia  Porta, Kolb, and
 Shubik,  1958).   In  a recent study Kektar et al.  (1978) instilled benzo-
 (a)pyrene,  weekly,  in Syrian golden hamsters at  doses of 0.1,  0.33, or
 1.0 mg (in bovine serum albumin).   Respiratory tract neoplasms were
 found in 19% to  22% of the males  and 20% to 40%  of the females treated.
 Survival times of 10 to 40 weeks  were dose related,  but  tumor  incidence
 was not.
      Although hamsters have been  the animal of choice for intratracheal
 studies  in  the past,  pneumonia-free specific pathogen-free (SPF) rats
 are now  used frequently because of their low incidence of infectious
 diseases (Nettesheim and Griesemer,  1978).   Davis  et al.  (1975)  induced
 squamous carcinomata in the lungs  of Wistar rats with intratracheal
 injections  of benzo(a)pyrene.   Using infusine as the vehicle,  0.5-, 1.0-,
 and 2.0-mg  doses  were administered once every 2  weeks.   Twenty-four of
 48 rats  developed squamous neoplasms of the lung.   Unlike the  results
with  the hamster  system (Ketkar et al.,  1978),  the tumor incidence in
 rats was dose related.   Tumors  have  also been induced by intratracheal
 instillation of carcinogens in  mice  (Nettesheim  and  Hammons, 1971), in
prosimian galagos (Crocker et al.,  1970),  and in rabbits (Hirao  et al.,
1968).

-------
                                   275
     Another mode of administration of chemical carcinogens is by direct
injection into the salivary gland.   Glucksman and Cherry (1971) injected
0.1 mL of a 2% suspension of 9,10-dimethyl-l,2-benzanthracene in acetone
into the salivary glands of male and female Lister rats (18 males and 10
females) and observed carcinomas and/or sarcomas of the glands in 16 of
18 males and 10 out of 10 females.   Carcinomas peaked at 100 days, and
sarcomas appeared as late as 770 days.
     Wigley, Amos, and Brookes (1978) induced tumors in the salivary
glands of C57BL male mice by direct injection of 0.1, 0.5,  or 2.0 mg
benzo(a)pyrene.  Forty percent of the animals injected with 0.1 mg, 90%
injected with 0.5 mg, and 56% injected with 2 mg developed fibrosarcomas,
carcinomas, and/or rhabdomyosarcomas.  (Two lymphosarcomas developed at
sites other  than  the  injection site.)  The  first  two  tumors appeared ap-
proximately  110 days  after  injection of benzo(a)pyrene.  The  responses
of mice  and  rats  to polynuclear aromatic carcinogens  injected locally
into the salivary glands  seem to be  similar.
     A broad spectrum of  tumor types can be elicited  in laboratory
animals  with polycyclic aromatic hydrocarbons administered systemically.
For example,  mice have been shown  to develop gastric  carcinomas,  pulmonary
adenomas,  and/or  leukemias  and lymphomas following  oral administration
of benzo(a)pyrene; the organ specificity of the  response is clearly
related  to strain (Rigdon and Neal 1966; Rigdon  et  al., 1969).   To be
specific,  of 81 male  Wistar rats given daily intragastric  instillations
of 5 mg  3-methylcholanthrene, 28%  developed 26 malignant neoplasms (10
mammary  carcinomas, 4 sebaceous gland carcinomas,  7 cutaneous carcinomas,
and 5 leukemias).  Of 122 female rats, 115  (98%)  developed 117 malignant
neoplasms  (106 mammary carcinomas, 10 leukemias,  1  sebaceous  gland
carcinoma,  and no skin carcinomas)  (Gruenstein,  Meranze, and  Shimkin,
1966).
     Huggins, Grand,  and  Brillantes  (1961)  observed preferential mammary
cancers  in 100% of the 50-day-old  Sprague-Dawley female rats  28 to 59
and 34  to  70 days after  feeding  them sesame oil  solutions  of  20 and 10
mg of 7,12-dimethyl-benzanthracene,  respectively.  Only ten  animals per
group were used.   Control groups were not  included in the  experiment,
but the investigators reported  that  only  two  mammary cancers had been
observed in 20 000 females  less  than 8 months  old in their colony.

-------
                                    276
 Thus, female rats seem to be highly susceptible to mammary carcinogenesis
 following oral administration of polycyclic aromatics.
      The carcinogenic response of hamsters to 3-methylcholanthrene was
 tested by Homburger et al. (1972), who administered, by gavage, 5 mg of
 the chemical three times per week for 17 weeks and observed carcinomas
 in males of the most susceptible strain tested 16 to 31 weeks later.
 Tumors were found in the forestomach (20%), stomach (40%), small intestine
 (45%), and large intestine (65%).  Eighty-seven percent of the female
 hamsters of that strain developed mammary tumors of various histological
 types.
      5.2.1.2.2  Aromatic amines — Bladder cancer in humans has been
 reported in all countries with established chemical industries.  A
 correlation between aromatic amine exposure and human bladder cancer was
 first reported by Rehn in 1895.  Tumor induction in animals by aromatic
 amines and subsequent- epidemiological studies have confirmed the corre-
 lation reported by Rehn.  Experimental details for the studies discussed
 in this section are shown in Table 5.3.
      Early attempts to produce bladder cancer in various species were
 unsuccessful, but in 1937 Heuper and Wolfe reported bladder tumors in
 dogs following oral and subcutaneous administration of 70 g of 2-naph-
 thylamine.  The tumors were first diagnosed by use of cystoscopy 21
 months after the start of treatment.
      Nelson and Woodard (1953) demonstrated the susceptibility of dogs
 to o-aminoazotoluene (o-AAT).   Administered orally for 30 to 62 months,
 0-AAT produced urinary bladder tumors in two of four animals studied.
 The other two dogs developed liver and gallbladder tumors.
      Another aromatic amine, 4-aminodiphenyl, was used by Walpole,
 Williams,  and Roberts (1954) to produce bladder cancer in the dog.
 Cumulative doses of 30 to 33 g, orally administered, induced tumors in
 the two  dogs tested within 2% years.  Thirty control beagles developed
 no  bladder tumors during 3 to 9 years of observation.
     Walpole's  results were confirmed in a 3-year study by Deichmannvet
 al.  (1965),  who were able to induce bladder tumors in 100% of six dogs
with smaller cumulative doses  (5.4 to 7.3 g per dog) of 4-aminodiphenyl.

-------
                                   Table 5.3.  Tumor Induction in various species with aromatic amines
Chemical/species
2-Naphthylamlne
Dog

Syrian hamster

Mouse
IF mouse


CBA mouse


CBA mouse

IF mouse

Albino rat


Rabbit

2-Acetvlamino-
fluorene
Dog

Mouse


Guinea pig


Dose

70 g

l.OZ

1-2 mg per pellet
200 mg/kg body weight
in arachis oil
twice weekly
120 mg/kg body weight
in arachis oil
twice weekly
160 mg/kg body weight
once weekly
Saturated solution
in benzene
310 mg/kg body weight
per week

200 mg
twice per week


4-12 mg/kg body weight
per day
0.05Z


0.045Z or 0.032Z

1.5 and 1.6 mg per
Duration of
exposure
(weeks)



>36

22-39
30-72


30-89


40-89

40-99

20->90


140-272



272-364

56


112-128

9-22
Route of
exposure

Oral;
subcutaneous
Oral, diet

Bladder pellet
Oral, gavage


Oral , gavage


Oral, gavage

Skin

Oral, diet
(reduced
protein)
Oral, spoon



Oral, diet

Oral, diet


Oral, diet

Intraperitoneal
Tumor incidence

Bladder

50Z Bladder
carcinomas
0/8
10/25 Liver
cholangiomas

13/23 Hepatomas;
1 cholangloma

11/26 Hepatomas

0/25

3/15 Bladder
papillomas

1/6 Bladder
papilloma


4/4 Bladder,
liver
14/49 Bladder
carcinoma or
papilloma
0/16

0/26
Tumor
latency Reference
(weeks)

84 Heuper and Wolf,
1937
40 Saffiotti, 1966

Bonser et al. , 1952
50-72 Bonser et al., 1952


50-89 Bonser et al., 1952


60-89 Bonser et al., 1952

Bonser et al. , 1952

60-90 Bonser et al. , 1952


247 Bonser et al., 1952



272-364 Morris and Eyes tone,
1953
48 Miller, Miller, and
Enomota, 1964

Miller, Miller, and
Enomota, 1964
Miller, Miller, and
Monkey
  100 g body weight
  2-3 times per week
Dosage increased
  with time from
  10-200 rag/kg
  body weight per
  day, S days per
  week
                                              52-182
Oral, diet
                                                                                   0/16
  Enomota, 1964


Dyer, Kelly, and
  O'Cara, 1966
                                                                                                                                                     ro

-------
                                                           Table 5.3.   (continued)
Chemical/species
2-Acetylamino-
fluorene
Hamster





o-Aminoazo-
toluene
Hamster


Rat
C3H mouse
Scraln C mouse
Strain A mouse
FI hybrids of
above
Dog



4-Aminodlphenyl
Dog





Duration of
Dose exposure
(weeks)


0.03X 30


1.5 mg per 100 g body 24-32
weight per day
3 times per week


0.1X 49


1.4-3.4 g total dose 21-49
10 mg once per 40
4 weeks in glycerol



20 rag/kg per day 8

5 mg/kg per day 128-248


Varied 140
5-20 mg/kg body weight
6 days per week
(Total - 2.9-3.3 g/kg)
1.0 mg/kg body weight 156
5 times per week
Route of Tumor incidence
exposure


Oral, diet 3/18 Liver
bile duct
carcinomas
Intraperitoneal 1/18 Small intestine
adenocarcinoma



Oral, diet 20/25 Bladder
19/24 Liver cell
3/15 Mammary
Oral 7/8 Hepatomas
Subcutaneous Varied with strain
Hepatic
Pulmonary
Hemangio-
endotheliomas
Oral 0/5 (all died
with liver damage)
Oral 2/5 Bladder
carcinomas

Oral, capsule 2/2 Bladder



Oral, capsule 6/6 Bladder
(4 malignant,
Tumor
latency Reference
(weeks)


52 Miller, Miller, and
Enomota, 1964

64 Miller, Miller, and
Enomota, 1964



45 Tomatis, Delia Porta,
and Shublk, 1961

21 Yoshida, 1932
Andervont, Crady, and
Edwards, 1942



Nelson and Woodard,
1953
Walpole, Williams,
and Roberts, 1954

Walpole, Williams,
and Roberts, 1954


Deichmann et al., 196:

                                                                                                                                                     to
                                                                                                                                                     «^l
                                                                                                                                                     00
Rat
3.6-5.8 g/kg body
  weight total
                                              36-53
                                                                Subcutaneous
17/23 (Variety
  of sites.  No
  bladder tumor.)
Walpole, Williams,
  and Roberts, 1952

-------
                                   279
Similar doses of 2-naphthylamine failed to produce neoplasms in six
dogs, thus establishing 4-aminodiphenyl as the more potent carcinogen in
this study.
     Morris and Eyestone (1953) induced liver and bladder tumors in each
of four dogs fed a total dose of 90 to 198 g of 2-acetylaminofluorene,
administered orally for 68 to 91 months.  Experiments in which bladder
cancer has been induced in dogs by other aromatic amines are listed in
Table 5.4.
     A distinct difference exists among the various species in their
responsiveness to carcinogenesis by aromatic amines.  The following
demonstrate some of those differences:   (1) 2-naphthylamine produced
bladder cancer in hamsters (Saffiotti et al., 1966) but not in rats,
mice, and rabbits (Bonser et al., 1952); (2)  o-ATT produced bladder
cancer in hamsters (Tomatis, Delia Porta,  and Shubik, 1961), but not in
rats (Yoshida, 1932) or mice (Andervont, Grady, and Edwards, 1942;
Andervont, 1950); (3) 4-aminodiphenyl failed to induce bladder tumors in
rats (Walpole, Williams, and Roberts, 1952);  and (4) 2-fluorenylacetamide
produced bladder tumors in mice but not in hamsters, guinea pigs (Miller,
Miller, and Enomota, 1964), or monkeys (Dyer, Kelly, and O'Gara, 1966).
     Because of the relative resistance encountered in other species, it
is felt that the dog should be used for testing carcinogenicity of
substances related to the aromatic amines (U.S. Food and Drug Adminis-
tration, 1971).  However, the hamster can develop aromatic amine-induced
bladder tumors having short latent periods and may be a likely substitute
(Gak, Graillot, and Truhaut, 1976).
     5.2.1.2.3  ff-Nitroso compounds — About 100 #-nitroso compounds have
been shown to be carcinogenic in experimental animals.  The carcinogenic
properties of some of these compounds are compared in different species.
     Diethylnitrosamine and dimethylnitrosamine — In 1967 Schmahl and
Osswald reviewed the effects of diethylnitrosamine in 11 different
animal species, paying particular attention to the organotropic activity
of the carcinogen.  The data of Schmahl and Osswald  (1967) and those of
other investigators are summarized in Table 5.5, which includes the
total dose (mg per kg of body weight) required to produce cancer in  the
various species.  Tumors were induced in all species and in almost 100%

-------
             Table 5.4.  Compounds, not previously listed, which have produced bladder cancer  in  the dog
       Compound
   Individual doses
Total dose per animal
that produced tumors
         (g)
 Incidence of tumors
  Latent period
Benzidine
4-Dimethylamino-
  azobenzene
4-Nitrobiphenyl
2-Naphthylamine plus
  4-nitrobiphenyl

2-Naphthylamine plus
  4-nitrobiphenyl  plus
  4-aminobiphenyl
200 mg per dog per day
  (in capsules), 6 days/
  week for 15 months,
  followed by 300 mg per
  dog, 6 days/week for
  45 months; then treat-
  ment discontinued

20 mg/kg per dog per
  daya (dry or in corn
  oil in capsules)
0.3 g per dog three
  times a week (in
  capsules)

0.1 g of each compound
  per dog, three times
  a week (in capsules)
0.1 g of each compound
  per dog, three times
  a week (in capsules)
  325
1 of 6 mongrels
  (1 male, 5 females)
7.5 years
  98 to 129
  Total:  17 to 20
  Total:  22 to 25
2 (1 male and 1
  female) of 10
  mongrels

3 of 4 female
  mongrels


5 of 5 female
  beagles


3 of 3 female
  beagles
3 years, 2 months
  to 4 years


2 years, 1 month
  to 2 years, 9
  months

2 years, 5 months
  to 2 years, 7
  months

2 years, 1 month
  to 2 years, 4
  months
     ait  is not  clear whether  a  certain dosage  per  day  was  given  five  or  seven  times  per  week.
     Source:  Adapted from Deichmann  and Radomskl,  1963.  Data  collected  from several sources.
                                                                                                                        oo
                                                                                                                        o

-------
     Table 5.5.  The carcinogenic action  of diethylnitrosamine  in different animal  species
Species
Mouse
Rat
Hamster
Guinea pig
Rabbit
Dog
Pig
Monkey
Grass
parakeet
Brachydanio
rerio
Trout
Route of
administration
Oral
Oral
Oral
Oral
Oral
Oral and
subcutaneous
Oral
Oral
Intramuscular
Oral

Daily dose
(mg/kg)
3
3
40 (weekly)
3
3.4
3
4.4
2 to 50
70 (weekly)
10 to 100 ppm

Total dose (D50)
(mg/kg)
871 ± 124
700 ± 53
640
1200 ± 100
2500
560
1400
1400 to 25 700
2800 ± 400
10 to 13 weeks

Type of tumor
Hemangioendotheliomas
the liver
Hepatocarcinomas
Hepatocarcinomas
Hepatocarcinomas
Hepatocarcinomas
Leimyosarcoma of the

of




liver
Reticulosarcomas of the
liver
Hepatocarcinomas
Hepatocarcinomas
Hepatocarcinomas and
cholangiomas
Hepatocarcinomas




     Source:  Adapted from Schmahl and Osswald, 1967.  Reprinted with permission  of  the
publisher.
                                                                                                     00

-------
                                   282
 of  the animals  tested.   Rabbits,  dogs,  and  pigs  treated with  approxi-
 mately 3  mg/kg  diethylnitrosamine developed cirrhosis  and  liver  cancer,
 while  mice,  rats,  and guinea pigs developed only liver cancer.
     Although Schmahl and  Osswald were  unable  to produce hepatomas  in
 monkeys,  they referred to  Kelly et al.  (1966), who  had done so in 6 of
 15  monkeys  (cynamolgus,  rhesus, and capuchin) with  oral doses of diethyl-
 nitrosamine  administered for more than  1  year.   Experimental  details of
 this and  the following studies with diethylnitrosamine and dimethylnitro-
 samine are listed  in  Table 5.6.
     Two  cercopithecus monkeys  given 1.6  and 2.2 g  of  diethylnitrosamine
 intraperitoneally  also developed  hepatocarcinomas,  with  induction  times
 of  25  and 27 months (Kelly et al., 1966).
     The  monkeys in the preceding experiments  did not  develop tumors in
 organs other than  the liver. A strain  of inbred guinea  pigs, however,
 developed tumors in both the lung and the liver  when diethylnitrosamine
 was administered in their  drinking water  (Argus  and Hoch-Ligeti, 1963).
 In  these  animals the first liver  carcinoma and the  first  lung papilloma
 were diagnosed  at  16 weeks and  45 weeks,  respectively.  In a  later  study
 with outbred Hartley guinea pigs, diethylnitrosamine in  the water  induced
 only hepatomas, with a mean induction time of  15 months  (Dale et al.,
 1973).
     Rats,  on the  other hand, developed tumors in a variety of organs
 following oral  administration of  diethylnitrosamine (Lijinsky and Taylor,
 1978). Diethylnitrosamine was  administered in drinking  water to groups
 of  6 to 15  Sprague-Dawley  rats  for up to  29 weeks.   The  animals  survived
 for 33 weeks, and  100% of  them  developed  tumors  of  the liver  (carcinomas
 and sarcomas),  nasal  turbinates,  and/or esophagus.   In a study by Reuber
 (1976), even more  extensive lesions were  observed in Buffalo  strain rats
 of  various ages fed diethylnitrosamine  in their  diet for 26 weeks.
 Malignant tumors appeared  in the  esophagus, liver (carcinomas and  sar-
 comas), and  prostate  gland; the type and  distribution  of  tumors  depended
 on  the sex and  age of the  animals.  The carcinogen  was incorporated into
 the diet  in  the amount of  0.0114%, and  the first tumors were  seen  24 to
 36 weeks  after  the start of the experiment.
     Similar  to rats,  BALB/c mice developed tumors  of  the  liver  and
esophagus as  a  result  of ingesting diethylnitrosamine  in drinking water

-------
                  Table  5.6.   Tumor  induction  in  various  species with  diethylnitrosamine  and dimethyInitrosamine
Chemical /species
Diethylnitrosamine
Monkey
Monkey
Inbred guinea
Pig
Outbred Hartley
guinea pig
Sprague-Dawley
Dose

2-5 mg/kg body
weight/day
Total: 6-24 g
20-40 rag per 2 weeks
Total: 1.6-2 g
1.1-4.2 rag/day
Total: 200-300 mg
About 3 mg/day
3 days/week
Total: 203 mg
Duration
of
exposure
(weeks)

108
108
16 to 40
Up to 68
29
Route of
exposure

Oral
Intraperitoneal
Oral
Oral
Oral
Tumor incidence

6/18 hepatomas
2/2 hepatomas
14/15 hepatomas
7/15 lung tumors
2/15 bronchial
papillomas
35/35 hepatomas
10/12 liver tumors
Tumor
latency Reference
(weeks)

92 Kelly et al. , 1966
104 Kelly et al., 1966
16 to 46 Argus and Hoch-Ligeti
1963
60 Dale et al., 1973
About 20 Lijinsky and Taylor,
  rat
Buffalo strain
  rat (12 weeks
  old)


Mice BALB/c
0.0114X in diet       26
Total:  515 mg/kg     41
  body weight
                     Total:  1010 mg/kg    41
                       body weight
                                                       Oral
Oral
                                  Oral
2/12 nasal
  turbinate tumors
6/12 esophageal
  papilloma
1/12 esophageal
  carcinoma

1/7 prostate tumor
12/26 esophageal
  carcinoma
Unclear liver

4/15 liver
  heraangiosarcomas
6/15 esophageal
  tumors
3/15 stomach tumors
0/15 lung tumors
14/60 liver
  hemangiosarcomas
35/60 stomach tumors
2/60 lung tumors
                                                                                                              1978
                                         24 to 36    Reuber, 1976
Unclear     Clapp, Tyndall, and
              Otten, 1971
                                         Unclear     Clapp, Tyndall, and
                                                       Otten, 1971
                                                                                                                                     ro
                                                                                                                                     oo

-------
                                                        Table 5.6 (continued)
 Chemical/species
       Dose
Duration
   of
exposure
 (weeks)
   Route of
   exposure
  Tumor incidence
 Tumor
latency
(weeks)
Reference
  Mice, Swiss
DimethyInitrosamine
  Mice, BALB/c



  Mice, Swiss
  Chinese hamster
6 mg/kg body
  weight/week
10
Intraperitoneal
Total:  300 mg/kg
  body weight

6 mg/kg body
  weight/week
Up to 41    Oral
10
Intraperitoneal
0.89-3.54 mg/kg
  body weight/week
Total:  31-105 mg
            Subcutaneous
30/38 lung tumors     66
4/38 vascular
  tumors
3/38 forestomach
  papillomas
0/38 kidney tumors
10/38 lymphomata
1/38 mammary
  adenocarc inoma
3/15 hemangio-        Unclear
  sarcoma
7/15 lung tumors

37/39 lung tumors     59
12/39 vascular
  tumors
0/39 forestomach
  papilloma
4/39 kidney tumors
4/39 lymphomata
1/39 mammary
  adenocarcinoma
100/108 liver         26
  tumors
1/108 lung tumors
1/108 nasal tumor
           Cardesa et al.,  1974
           Clapp,  Tyndall,  and
             Otten,  1971


           Cardesa et al.,  1974
                                                                                        N>
                                                                                        oo
                                                     Reznik,  1975

-------
                                    285
(Clapp, Tyndall, and Otten, 1971).   Tumors were also found in the lung
and stomach.  Dimethylnitrosamine,  however, produced tumors only in the
lung and liver.  Differences in susceptibility of the lung to diethyl-
nitrosamine (3% tumor incidence) and dimethylnitrosamine (47% tumor
incidence) indicate that different  enzyme systems may be activated for
metabolism of each of the chemicals and/or that the system for metabo-
lizing diethylnitrosamine is not present in sufficient quantity in the
lung of the BALB/c mouse.  Swiss mice, a strain having a 26% spontaneous
lung tumor incidence, also developed more lung tumors after intraperito-
neal injection of dimethylnitrosamine than after diethylnitrosamine, but
the difference in tumor incidence was less pronounced (Cardesa et al.,
1974). Liver tumors in both strains of mice were of vascular rather than
of hepatocellular origin.  A similar response was reported in Chinese
hamsters injected subcutaneously with three doses of dimethylnitrosamine
(Reznik, 1975).  Tumors were mainly in the liver and were all of vascular
origin (hemangioendotheliomas).  Tumor incidence was nearly 100% and was
not dose dependent; the first liver tumor occurred in the 26th exper-
imental week.
     Thus, in  the above examples the most obvious species-related differ-
ence in the response of animals to diethylnitrosamine and dimethylnitro-
samine was in  the type of liver tumors induced.  Those in monkeys, rats,
guinea pigs, and hamsters were of vascular and/or liver cell origin,
while those in mice were of vascular origin only.
     Other jy-nitroso compounds — In separate studies, species differences
were observed between rats and hamsters treated with diisopropanol-
nitrosamine (Mdhr, Reznik, and Pour, 1977; Pour, Kruger, and Althoff,
1974, 1975).   Sprague-Dawley rats were given subcutaneous injections  of
178 to 1425 mg per kg of body weight once weekly for 20 weeks, and
hamsters received 125 to 500 mg per kg of body weight per week for life.
The chief differences noted were:  (1) pancreatic tumors were diagnosed
in all hamsters, but only one such tumor was observed among 150  rats;
(2) cholangiomas or cholangiocarcinomas were, found in the hamster but
not in the rat; and (3) adenomas and adenocarcinomas were found  in the
thyroid glands of rats but not in hamsters.

-------
                                    286
     Lijinsky et al. (1967, 1970) tested the carcinogenicity of a cyclic
nitrosamine, nitroazetidine, in rats, mice, and hamsters.  Five milli-
grams of the compound per day administered in drinking water for 46 days
(total dose 230 mg) induced tumors in 15 out of 20 Wistar rats, and 0.5
mg per day (total dose 23 mg) induced tumors in 15 out of 40 mice.
Hamsters were resistant to total doses of up to 500 mg.  However, both
rats and hamsters were responsive to tumor induction by another cyclic
nitrosamine, nitrosoheptamethyleneimine (Lijinsky et al., 1967, 1970).
     It may be concluded from the above examples of #-nitroso compound
carcinogenesis that the organ specificity of these chemicals is approxi-
mately the same in all species that respond to them, regardless of
route.  However, differences do exist  in general susceptibility of the
various species to specific compounds  of this class of chemicals.
     5.2.1.2.4  Miscellaneous compounds — Brief mention is made below of
species differences encountered in carcinogenesis studies with miscel-
laneous compounds of chemical classes  other than those previously men-
tioned.
     Minerals and Metals — Exposure in inhalation chambers to amosite,
crocidolite, and chrysotile was the basis for a comparison of the various
responses of rats  (Charles River), guinea pigs  (Camm-Hartley), rabbits
(Shankin Farms Dutch), mice  (Swiss), and gerbils (Mongolian) of both
sexes  (Reeves, Puro, and Smith, 1974).  The animals were exposed 4 h per
day, 4 days per week for 2 years  (1480 h cumulative total exposure).
Some animals were sacrificed after 3 to 6 months, but most were allowed
to survive until the exposure period ended.  Table 5.7 contains results
of the study.
     Reeves and associates were the first to obtain lung carcinoma and
pleural mesothelioma following exposure by inhalation to amosite.  Rats
seemed to be the only species that displayed an increased incidence of
neoplasia of the respiratory tract  (mice were excluded because of their
tendency to develop spontaneous bronchial cancers).  It was  also noted
that after equivalent asbestos exposure, pulmonary ferruginous bodies
were abundant in the guinea pig and rare in the rat.  The authors sug-
gested the frequency of ferruginous bodies is inversely  related to
carcinogenic effects attributable to asbestos inhalation.

-------
                  Table 5.7.  Carcinogenicity of different forms of asbestos in various species
Species
Mouse*2

Gerbil
Rat
Rabbit
Guinea pig

Number
surviving
19

50
43
11
14
Chrysotile
Effect
None

None
One papillary carcinoma
of lung
One squamous carcinoma
of lung
One mesothelial fibro-
sarcoma of mediastinum
None
None
Crocidolite
Number
surviving
18

49
46
9
14
Effect
Two papillary
carcinomas of
bronchus
None
One adenocarci-
noma of lung
Three squamous
carcinomas of
lung
One papillary
adenocarc inoma
of lung
None
None
Number
surviving
17

51
46
9
13
Amosite
Effect
None

None
One metastatic osteo-
sarcoma of lung
One mesothelial fibro-
sarcoma of lung and
pleura
One fibrous mesothelial
of pleura
None
None
                                                                                                               00
                                                                                                               ^1
aControl mouse had one papillary carcinoma of the bronchus.  All other controls were free of neoplasma.

Source:  Adapted from Reeves, Puro, and Smith, 1974.  Reprinted with permission of the publisher.

-------
                                    288
      Sunderman (1971) reviewed the carcinogenic effects of beryllium,
 cadmium,  chromium, cobalt, iron, lead, nickel, selenium, zinc, and
 titanium in various species by different routes of exposure.
      jL>l-Trichloro-2,2-bis(p-chlgrophenyl) ethane (DDT) — DDT has been
 shown to be primarily a hepatocarcinogen in mice, although tumors have
 developed in other organs of more susceptible mouse strains (Innes et
 al.,  1969;  Terracini et al., 1973).   Tomatis and Turusov (1975) demon-
 strated that the tumorigenicity of DDT in mice is dose related.  In rats,
 however,  DDT has been noncarcinogenic (Cameron and Cheng,  1951) or mar-
 ginally carcinogenic (Deichmann et al.,  1967) and has had no effect on
 hamsters and monkeys (Agthe et al.,  1970; Durham, Ortega,  and Hays,
 1963).
      Aflatoxin Bj — The hepatocarcinogenicity of aflatoxin Bj had been
 demonstrated in rats (Newberne and Rogers, 1972; Wogan and Shank, 1971)
 but not in mice until Vessilnovitch et al. (1972) induced heptatomas in
 newborn mice with intraperitoneal injection of a total of 1.5 to 6 yg
 per g of body weight (in one to five doses).  Tumors were observed 52
 weeks after exposure.
      Adamson, Correa, and Dalgard (1973) reported the induction of liver
 tumors in a rhesus monkey with aflatoxin BI} and carcinogenicity of
 aflatoxin in the hamster has been described by Herrold (1969).  Table
 5.8 lists other species and target organs in which the tumorigenic
 activity of aflatoxin has been demonstrated.
      [Although this report is primarily concerned with mammalian tests
 for carcinogenicity,  Table 5.8 has been included because it concisely
 relates the association between exposure to a chemical or a chemical
 process and the occurrence of cancer in humans as reported by the Inter-
 national  Agency for Research on Cancer (Tomatis et al., 1978).  These
 data  are  correlated with the results of  animal tests.]

 5.2.2   Strain
     The  use  of  random-bred,  rather  than highly inbred, strains in
chronic toxicity  and  carcinogenicity  testing remains controversial.   In
broad-scale studies where  little prior information exists,  some workers

-------
Table 5.8.  Chemicals or industrial processes associated with cancer induction in humans:
      comparison of target organs and main routes of exposure in animals and humans
Chemical or
industrial process
Aflatoxins








4-Aminobiphenyl



Arsenic Compounds


Asbestos




Auramine
(manufacture of)


Benzene
Benzidine





Bis(chloromethyl)
ether


Humans
of^xposu're*
Environmental, Liver
occupational







Occupational Bladder



Occupational, Skin, lung, liver
medicinal, and
environmental
Occupational Lung, pleural cavity,
gastrointestinal
tract


Occupational Bladder



Occupational Hemopoietic system
Occupational Bladder





Occupational Lung




Main route. A_<_«I
of exposure**0 Animal
P.O., i.h. Rat

Fish, duck, mar-
moset, tree
shrew, monkey
Rat

Mouse, rat
Mouse
i.h., s. , Mouse, rabbit, dog
p.o. Newborn mouse
Rat

i.h., P.O., Mouse, rat, dog
s . Mouse

i.h., p.o. Mouse, rat, hamster,
rabbit
Rat, hamster
Rat

i.h., s., p.o. Mouse, rat
Rabbit, dog
Rat

i.h., s. Mouse
i.h., s., p.o. Mouse
Rat


Hamster
Dog
i.h. Mouse, rat
Mouse

Rat
Animals
Target organ
Liver, stomach,
colon, kidney
Liver


Liver, trachea
Liver
Local
Lung
Bladder
Liver
Mammary gland,
intestine
Inadequate, negative
Inadequate, negative

Lung, pleura

Local
Local ,
Various sites
Liver
Negative
Local, liver,
intestine
Inadequate
Liver
Liver
Zymbal gland, liver,
colon
Liver
Bladder
Lung, nasal cavity
Skin
Local, lung
Local

Route of
exposure"
p.o.

p.o.


i.t.
i.p.
s.c.
i.p.
p.o.
s.c.
s.c.

p.O. N3
t i v 00
t., l.V. ^

i.h. or i.t.

ipl.
i.p., s.c.
p.o.
p.o.
p.o.
s.c.

t., s.c.
s.c.
p.o.
s.c.

p.o.
p.o.
i.h.
t.
s.c.
s.c.

-------
Table 5.8 (continued)
Chemical or
industrial process
Cadmium-using indus-
tries (possibly
cadmium oxide)
Chloramphenicol
Chloromethyl methyl
ether (possibly
associated with
bis (chloromethyl) -
ether
Chromium (chromate-
producing
industries)
Cyclophosphamide





Die thy Is t ilbes t rol








Hematite mining
(? radon)

Isopropyl oils
Melphalan


Main type
of exposure*2
Occupational


Medicinal
Occupational




Occupational


Medicinal





Medicinal








Occupational


Occupational
Medicinal

Humans
^SS"
Prostate, lung** i.h., p.o. Rat


Hemopoietic system P.O., i.j.
Lung i.h. Mouse


Rat

Lung, nasal cavities^ i.h. Mouse, rat
Rat

Bladder p-c... i.j. Mouse


Rat


Uterus, vagina p.o. Mouse
Mouse


Rat

Hamster

Squirrel monkey
Lung i.h. Mouse, hamster,
guinea pig
Rat
Nasal cavity, larynx i.h.
Hemopoietic system P.O., i.j. Mouse
Rat
Animals
Target organ
Local, testis


No adequate tests
Initiator
Lungd
Local,, lung"
Local"

Local
Lung

Hemopoietic system,
lung
Various sites
Bladder^
Mammary gland
Various sites
Mammary
Mammary, lymph-
oreticular, testis
Vaeina
•«*o ,
Mammary, hypophysis"
bladder
Kidney

Uterine serosa
Negative

Negative
No adequate tests
Initiator
Lung, lymphosarcomas
Local

Route of
exposure0
s.c. or i.m.



s.
i.h.
s.c.
s.c.

s.c. , i.m.
i.b. impl.

i.p., s.c.
N)
vO
p.O. o
i.p.
i.p.
i.v.
p.o.
s.c. ,
s.c. impl .
Local
s.c. impl.

B.C.;
s.c. impl .
s.c. impl .
i.h., i.t.

s.c.

s.
i.p.
i.p.

-------
                                                              Table 5.8 (continued)
Chemical or
industrial process
Mustard gas

2-Naphthylamine



Nickel (nickel
refining)

/V,#-Bis(2-chlpro-
ethyl) 2-naphthy 1-
lamine
Oxymetholone
Phenacetin
Phenytoin

Soot, tars, and oils

t
Vinyl chloride




Main type
of exposure"
Occupational

Occupational



Occupational


Medicinal


Medicinal
Medicinal
Medicinal

Occupational,
environmental
Occupational



Humans
Target organ
Lung, larynx

Bladder



Nasal cavity, lung


Bladder


Liver
Kidney
Lymphoreticular
tissues
Lung, skin (scrotum)

Liver, braind, lungd




Main route A-.IIMI
of exposure* ^iaai
i.h. Mouse

i.h., s. , p.o. Hamster, dog,
monkey
Mouse
Rat, rabbit
I.h. Rat
Mouse, rat, hamster
Mouse, rat
p.o. Mouse
Rat

p.o.
p.o.
P.O., i.j. Mouse

i.h., s. Mouse, rabbit

i.h., s. Mouse, rat



Animals
Target organ
Lung
Local , mammary
Bladder

Liver, lung
Inadequate
Lung
Local
Local
Lung
Local

No adequate tests
No adequate tests6
Lymphoreticular
tissues
Skin

Lung, liver, blood
vessels, mammary,
Zymbal gland,
kidney

Route of
exposure0
i.h., i.v.
s.c.
p.o.

s.c.
p.o.
i.h.
s.c., i.m.
i.m. impl.
i.p.
s.c.


NS
\£>
P.O., i.p.

t.

i.h.



     aThe main types of exposures mentioned are  those by which  the association has been demonstrated; exposures other than those mentioned
may also occur.
      The main routes of exposure given may not  be  the only ones by which such effects could occur.
     °p.o. - peroral; i.t. - intratracheal; i.p. -  intraperitoneal; s.c. — subcutaneous; t. - topical; i.v. — intravenous; ipl. - intra-
pleural; i.h. - inhalation; i.m. - intramuscular; s. - skin; s.c. impl. - subcutaneous implantation; i.b. - intrabronchial implantation;
i.J. — injection.
     d.
1977).
      Indicative evidence.
      The induction of tumors of the nasal cavities in rats given phenacetln has been reported recently  (S. Odashima, personal communication,

-------
                                   292
 recommend using random-bred strains  to ensure observing a species-
 characteristic response (Benitz,  1970).   Other investigators urge the
 use of inbred animals whose genetic  stability tends to reduce variations
 in the incidence of spontaneous disease and contributes to greater
 experimental precision (Food Safety  Council, 1978;  Womack, 1979).
 However,  Greenman,  Delongchamp, and  Highman (1979)  warn that easy general-
 ization cannot be made about the variability of inbred and hybrid mice.
 Whether or not inbred or random-bred strains are used, it is important
 that background clinical and pathological information be available for
 test animals so that results in both control and treated animals can be
 compared with known literature values (Fancher, 1978; World Health
 Organization, 1978).  Germ-free animals are rarely  used in chronic
 toxicity and carcinogenicity testing because maintenance is too laborious
 and costly for long-term studies, but specific pathogen-free and gnoto-
 biotic animals have been found useful by some investigators (Benitz,
 1970).
      Selection of the appropriate animal strain is  an important part of
 experimental design in bioassays for carcinogenicity.  In terms of
 numbers of strains  available, selection is not easy.   For example, there
 are over 250 mouse  strains, 100 rat  strains, and 30 hamster strains
 (Butler,  1979).   Weisburger (1976) suggested several factors that deserve
 consideration in choosing a strain.   These include  maintenance require-
 ments, sensitivity  to test chemicals, and occurrence of diseases, both
 nonneoplastic and neoplastic.  In addition, he reviewed information
 about some of the strains of rats, mice, and hamsters used in bioassays
 of  carcinogenic  agents.   Because of  variation in response to such agents
 by  animals of different strains,  it  is generally advisable to test the
 agents in several strains of a species (Butler, 1979).  Several studies
 will  expand upon some of these considerations.
      5.2.2.1  Hamsters and Guinea Pigs — Inbred strains of Syrian golden
 hamsters  respond differently to subcutaneously injected methylcholanthrene
 and benzopyrene  (Homburger et al., 1972).   The less sensitive strains
 (BIO 82.73,  BIO  1.5)  have  longer  induction times than does the more sen-
sitive 87.20 line.   This  fact is  important for determining the duration
of an experiment and,  if not  properly understood, could lead to misinter-
pretation of  data.  Another  consideration  in selection of an appropriate

-------
                                   293
 strain relates to tumor type.  For example, some strains of hamsters
 (BIO 54.7, BIO 82.73, BIO 86.93) are not susceptible to induction of
 intestinal neoplasms and thus should not be used for bioassays concerned
 with such tumors (Homburger et al., 1972).
     A strain may be sensitive to one chemical and not another, as seen
 in the Hartley and inbred ICRF guinea pigs.  Diethylnitrosamine is an
effective carcinogen in both strains;  methylcholanthrene is inconsistent
in the Hartley strain,  but less so in the ICRF animals (Dale et al.,
1973).
     5.2.2.2  Mice — The influence of genetic factors has been clearly
demonstrated in two strains of mice,  BALB/c and C57BL/6, which after
treatment with diethylstilbestrol developed testicular teratomas and
pituitary neoplasms,  respectively.   The F^  hybrids of these strains
developed both tumors in response to the carcinogen (Greenman and
Delongchamp, 1979).   In addition to variation in tumor types among
different strains,  incidence, induction time, and susceptibility to an
agent can vary.   In an extensive study of transplacentally injected 1-
ethyl-1-nitrosurea,  Diwan and Meier (1974)  demonstrated clear differences
in all these parameters for five mouse strains (AKR/J, SWR/J, DBA/2J,
C57L/6J,  and C57L/J).  They concluded that responses to the carcinogen
were dependent on both the strain involved and the fetal age at the time
of injection.  The latent period for malignant lymphoma in "101" mice
treated with dimethylbenzanthracene (DMBA)  was shorter than that of CBA
mice (Roe, Rowsen,  and Salaman, 1961).  Thiery and van Gijsegem (1965)
studied the induction of squamous cell carcinoma of the cervico-vaginal
epithelium by 3,4-benzo(a)pyrene in 12 mouse strains.  On the basis of
tumor yield and induction time, strains C3H, A, 0.20, N, R, K, Q, and Mo
were highly sensitive,  S and AKR were moderately sensitive, and H and W
were relatively insensitive to the carcinogen.  Holland, Gosslee, and
Williams (1979)  demonstrated that C57BL/6 mice were 2.4 times more
sensitive to the epidermal carcinogens bis(2,3-epoxycyclopentyl)ether,
2,2-bis(p-glycidyloxphenyl) propane, and m-phenylenediamine.  Holland et
al. also noted that the sensitivity of an assay could be affected by the
choice of strain in the case of very weak carcinogens.

-------
                                   294

      5.2.2.3  Rats — Within a few weeks after one dose of dimetbylbenz-
 anthracene, Sprague-Dawley rats developed high incidence of multiple
 mammary gland tumors.  In contrast,  Long-Evans rats receiving the same
 treatment had low incidence, few tumors, extended induction time, and,
 in some cases, tumor regression.  The sensitivity of the mammary glands
 to the hydrocarbon can be correlated with inherited variation in pituitary
 function.   Fibroadenoma incidences after DMBA in Fj hybrids from recip-
 rocal crosses of the two strains were nearly equal; hybrids from back-'
 crosses in which the strain of origin was SD had high tumor incidence as
 opposed to low ones from the LE strain.  Foster nursing had no effect on
 tumor incidence (Sydnor et al., 1962).  Sydnor (1973) also tested nine
 inbred strains of rats for susceptibility to benzo(a)pyrene.  Sarcoma
 incidence ranged from 38% in Long-Evans rats to 97% in Fischer rats.  The
 most  sensitive strains in terms of incidence and induction time were
 Sprague-Dawley, Fischer, and Werck-Stewart,  and the least sensitive were
 the Long-Evans rats.  Sydnor concluded that  susceptibility is genetically
 determined by carcinogen dose.
      In conclusion, careful selection of appropriate strains for experi-
 mentation and substance evaluations  is a prerequisite for meaningful
 interpretation of data.  Strain-related parameters that relate to carci-
 nogenesis include spontaneous tumor  incidence (discussed elsewhere),
 age,  induction time, and susceptibility.  Superimposed on the genetic
 framework are the many environmental factors that can modify the response
 of a  strain to a particular stimulus.  These factors must be controlled
 as extensively as possible.  The great value of strain differences lies
 in the potential for eventually determining  the molecular mechanisms
 that  vary between strains.  Ultimately, such knowledge may help to
 determine  those mechanisms that cause variation among individuals.

 5.2.3  Spontaneous Tumors
      Spontaneous tumors are relatively common among experimental animals,
both  those bred for laboratory studies as well as wild animals maintained
 in  the  laboratory (Andervont and Dunn, 1962).  Such lesions occur in a
variety of organs and at different ages.  Snell (1965) defined spontaneous
lesions as those for which a cause cannot be ascertained.  Such neoplastic

-------
                                  295

lesions appear in animals that have not been experimentally exposed  to  a
carcinogenic agent,  Hoag (1963) pointed out that spontaneous tumors are
generally considered "naturally occurring"; however, when the etiology
of the particular tumor is understood, it can be induced experimentally.
Spontaneous tumor incidences in the various organs of animals used in
the national Cancer Institute Bioassay Program have been summarized  by
Page (1977a) and are listed in Table 5.9.  The B6C3Fj mouse, an PI
hybrid cross between the C57BL/6 female and the C3H male, has a rela-
tively high incidence of spontaneous tumors of the lung and liver.   The
F/344 rat has a high incidence of testicular tumors in the male and  of
pituitary tumors in the female.  These strains have been used extensively.
They have the advantages of good survival, disease resistance, and a
relatively low spontaneous tumor incidence in organs other than those
mentioned.
     In order to make valid interpretations and comparisons of results
in a variety of experimental situations — whether they are designed  to
test carcinogenicity of specific agents, to screen substances for anti-
carcinogenic activity, or to look at mechanisms of neoplastic growth —
the investigators must be cognizant of the incidence of spontaneous
tumors in the animal tested.  For example, the tumor response (i.e., the
type, frequency, latency, etc.) to carcinogenic agents may vary among
animals with different spontaneous tumor rates.  For more information
relating to this point, see the discussion of strain differences.
Various factors can influence frequency, distribution, latency, and
morphology of spontaneously occurring tumors.  Some variables known  to
have effects are:  breeding methods, genetics, age, diet, maintenance,
histologic screening, and statistical evaluation (Pollard and Kajima,
1970; Pour et al., 1976; Sass et al., 1975), as well as geophysical
parameters such as geography and climate (Gilbert et al., 1958).  Sex
and age incidence of characteristic tumors for certain inbred strains of
mice have been summarized by Weisburger and Weisburger in Table 5,10.
Spontaneous tumor incidence can generally be viewed as the  result of
both genetic and environmental influences whose relative contributions
must be defined for a particular animal strain.  Genetic or strain
differences in spontaneous tumor frequency can be best evaluated  when
environmental factors have been characterized  (Gilbert et al.,  1958).

-------
Table 5.9.  Spontaneous tumor incidence  in  animals  used  in  Che  National  Cancer Institute Bioassay Program
B6C3F1 Fischer
mouse0 344 rat*1
fl v rr in
Males
Brain <1
Skin/ subcutaneous 1.0
Mammary gland
Spleen <1
Lung/ trachea 9.2
Heart
Liver 15.7
Pancreas <1
Stomach/ intestines 1.3
Kidney <1
Urinary bladder
Testis <1
Ovary NA
Uterus NA
Pituitary <1
Adrenal <1
Thyroid 1.1
Parathyroid
Pancreatic islets <1
Thymus <1
Body cavities <1
Leukemia/lymphoma 1.6
Females Males
1176 846
1.3
<1 5.7
<1 1.0
<1 <1
3.5 2.4
1
2.5 1.2
<1 <1
<1 <1
<1 <1
<1 j <1
NAfl 76.2
<1 NA
1.9 NA
3.5 10.2
<1 8.7
<1 5.1

<1 3.2

<1 <1
6.8 6.5
Females
840

-------
                                  297
       Table 5.10.
Sex and age incidence of characteristic tumors
        of inbred strains
Tumor type
and strain
Mammary gland tumor
C3H

DBA/ 2

A

DD

Lymphocytic leukemia
AKR
C58
Primary lung tumor
A
SWR
Hepatoma
C3HeB


Reticulum cell
neoplasm, type B SJL

Incidence
(%)

99
100
77
Lower
84
5
84
75

92
90

90
80

91
58
30

91
High
Type mouse

Breeding 99
Virgin 99
Breeding 99
Virgin 99
Breeding 99
Virgin 99
Breeding 99
Virgin 99







Breeding 
-------
                                   298
                           Table  5.10  (continued)
        Tumor type
        and strain
Incidence
                 Type mouse
                          Age
                       (months)
  Testicular teratoma
    129
 Myoepithelioma
     1
    82
           66
Male gonadal
  ridges trans-
  planted to adult
  testes
Congenital
BALB/c
A
Skin papilloma
HR/De
Skin carcinoma
HR/De
Subcutaneous sarcoma
C3H/J
Harderian gland tumor
C3H
4
Similar

9 Hairless

3 Hairless

3 99

1



Av.

Av.

Av.

Av.



22

18

25

20.7
      Source:  Weisburger and Weisburger,  1967,  as adapted  from Murphy,
 1966.  Reprinted with  permission of  the publisher.
     Genetic  control of  susceptibility to  ileocecal  immunocytomas in
certain  strains  of  rats  appears to  reflect one  or more  dominant  genes
for susceptibility  in the LOD/c strain and one  or more  loci of resistance
in the OKA  strain.   Fj progeny from crosses of  these strains have null
tumor incidence  (Beckers and Bazin,  1978).   High incidence  of spontaneous
congenital  testicular teratomas in  an inbred subline of mice is  probably
due to a single  gene mutation (Stevens,  1973).
     Evidence of genetic involvement at the molecular level is evident
in the inhibition of mammary tumors in mice by  treatment with a  bacterial
cell wall skeleton  preparation which reduces the level  of DNA synthesis
in normal mammary glands and lowers circulating prolactin levels, both
major factors in tumorigenesis (Nagasawa,  Yanai,  and Azuma,  1978).
     Genetic  influence on susceptibility has been demonstrated in certain
                                                                  vy
strains  of C3H mice prone to hepatomas and mammary tumors.   The  A   gene

-------
                                  299
increases the Incidence of both tumor types, perhaps by enhancing the
virulence or transmission of the mammary tumor virus by either parent or
by increasing the responsiveness of tissues.  The gene's effects are
also correlated with increased body weight and are enhanced by hormonal
factors (Heston and Vlahakis, 1968; Vlahakis, Beston, and Smith, 1970).
     Interesting results with this same CSH-A^ system have been reported
by Sabine, Horton, and Wicks (1973).  Mice bred in Australia had 0%
incidence of mammary tumors compared with the incidence of nearly 100%
found in the united States.  By using feed and bedding from the United
States, the high tumor incidence can be restored.  The interactions of
genetic and environmental factors are clearly complex, and both deserve
careful attention.
     Examples of the modification of spontaneous tumor incidence by
environmental factors have been reported in the C3H mouse hepatic tumor
system, which can be influenced by sex, population density, and diet
(level of protein and caloric intake) (Grahn and Hamilton, 1964; Heston,
Vlahakis, and Deringer, 1960; Peraino, Fry, and Staffeldt, 1973; Silverstone
and Tannenbaum, 1951; Tannenbaum and Silverstone, 1949).  Heston (1958),
in studying four substrains of C3H mice, found that mammary tumor inci-
dence was influenced not only by genetic susceptibility, but also by
hormonal stimulation during breeding and by age.  Riley  (1975) described
a shortening of latent period.for mammary tumors in C3H/He mice carrying
the Bittner oncogenic virus and exposed to environmental stress factors.
     Age of the animals and latency periods of the tumors can present
problems in design and interpretation of experiments.  Tumor incidence
data from rats at 2 to 2.5 years of age could give a different picture
from that which might be seen if the animals lived to old age.  If the
animals are more susceptible to tumors late in life, then testing only
in young animals could prevent detection of neoplasms.  Studies over the
life span of the animal are necessary for the evaluation of the biologic
behavior of spontaneous tumors and, possibly, of some carcinogen-induced
tumors (Burek and Hollander, 1977).  In addition to the value of life
span data, one must consider the latency period before the effect of a
carcinogen is detectable as well as the dose (which in some cases deter-
mines latency).  Laboratory animals tend to be more susceptible to

-------
                                   300
 tumors as they age, but, as discussed earlier, additional factors are
 involved (Gilbert et al., 1958).  Table 5.11 summarizes some pertinent
 literature on this subject.

 5.2.4  Number
      The number of animals used in chronic toxicity and carcinogenicity
 tests represents a compromise between the need for a sufficiently large
 number of animals to allow adequate statistical precision of the conclu-
 sions and the need to place a reasonable limit on costs and the experi-
 mental work load.  For example, if the true frequency of a toxic effect
 in test animals is 5%, a test group numbering at least 58 must be used
 to discover the effect with a probability of P = 0.05, but 90 animals
 are needed at the P = 0.01 level of significance, and 134 animals must
 be used to achieve the P = 0.001 level of confidence (Barnes and Denz,
 1954).   Although high levels of statistical probability are obviously
 desirable, large increases in group size may diminish the thoroughness
 and care that are necessary for a successful completion of the study and
 may be counterproductive.  In general, more useful information can be
 obtained by conducting thorough studies with a relatively small number
 of animals than by performing incomplete experiments using an excessive
 number of animals (Benitz, 1970).
      Early investigators of chronic noncarcinogenic toxicity tended to
 use 10 or fewer test animals (rodents) per group (Barnes and Denz,
 1954);  later, groups of 20 to 30 rodents per sex and dose level were
 considered  sufficient  for practical  purposes  (Benitz,  1970).  More
 recently, a minimum of 50 rodents of each species and  sex has been
 considered  necessary,  especially in  work subject  to federal  agency
 regulation  (Federal Register,  1978,  1979;  Food Safety  Council,  1978;
National Academy  of Sciences,  1977).   Fewer rodents per  group may be
used if the number  of  dose levels is increased; however,  little current
literature  support  exists  for  groups smaller  than 20 animals.
     Test groups  are generally smaller than indicated  in the previous
paragraph when the  test  species are  nonrodents.   If dogs,  cats,  or non-
human primates are  used  in chronic toxicity studies, a minimum  of at
least four animals  per dose and sex  is usually considered necessary

-------
                                             Table 5.11.  Spontaneous  tumors in laboratory animals
          Animal
         Tumor  and  organ
                                                                     Incidence and age
                                                nts
                                                                    Reference
Monkey
  Maaaca phillipieneie and
  Macaaa mulatto
Lymphosarcoma — kidney
Hemangloendothelloma — vertebra
Meningiomatosls — lumbar  cord
Lipoma — chorold  plexus
Endotheliomatosls — lumbar  cord
Overall - 0.08*
prepubertal
12 000 monkeys (1800 of
them cynomolgus) observed
over 3.5 years.  Also looked
at granulomatous processes.
Jungherr, 1963
  Nude mutant, most
  with BALB/c, some
  with C3H or C57/BL/6
  backgrounds

  Nude mutant from
  BALB/c backgrounds
  CD8F,
  (BALB/CXDBA/8F1)'

  SHN


  129/terS



  Strain A
  CD^-lHaM/ICR
Mammary  tumors


None




Lymphoreticular neoplasms
Adenocarclnoma mammary
(also metastasis to lung)

Mammary tumors
Teratomas, testicular
Lung tumors
Lymphoreticular mammary,
pulmonary, osteogenlc sarcoma,
hemanglosarcoma, renal and
hepatic tumors

Liver, mammary tumors
 CSH-AfB
                                                                 Reaches 87%
OX, generally die at
2 to 3 months of age
Estimated from graphic
material, tumor Incidence
for entire group was about
50X at 57 weeks (2Z at 35
weeks)

802, 10 months
41.32 at 7 months, 87.52
at 12 months

302, earliest seen were in
6-day fetuses
02 at 2 months, 402 at 12
months, 77.1Z at 18 months
Total incidence of tumors —
range of 0.77-50Z for age
groups ranging from 0+ to 20+
months

90-1002 or 02, ages from
7 to 22 months
Studied tumor regression
after 5-methyl cytidine.

11 000 mice studied from
birth to 3 months.
Cermfree conditions — can
maintain for over 20 months.
In germfree nu/+ females, no
mammary tumors seen.
Studied chemotherapeutic
activity.

Studied tumor suppression.
High incidence probably
due to a single gene
mutation.

Frequency of lung tumors has
been reproducible for more
than 30 years.

Percentages of a particular
tumor type varied with age.
                                                                                                  Tumor Incidence apparently
                                                                                                  related to type bedding used.
Strong and
Matsunaga, 1975

Rygaard and
Polvsen, 1974
Outzen et al.,
1975
Fugmann et al. ,
1977

Nagasawa, Yanai,
and Azuma, 1978

Stevens, 1973
Shimkin and
Stoner, 1975
Percy and
Jonas, 1971
                                Sabine, Horton,
                                and Wicks, 1973
                                                                                                                                                       U>
                                                                                                                                                       O

-------

Animal
C3Hf

C3Hff

C3Hf/2
C3Hf/An
C3H£B X YBR
C3H£B X YBR
C3Hfb X YBR
YBR X C3UfB
YBR X C3HfB
YBR X C3HfB
YBR X C3HfB


C3H/St

C3HX101
hybrids
A
GR
020
CBA
C3H
DBA
C57BL
Table 5.11
Tumor and organ
Mammary tumors: adeno- 22%,
carcinoma,
Adenoacanthoma, care in o- 29Z,
sarcoma
51Z,
27Z,
Hepatoma 100Z
94Z
63Z
52
95*
43*
41%
*+•*•:-,. QZ

Adenocarcinoma, mammary 41Z,

Os ceosar comas 20X

Lung tumors 71Z,
42Z,
58Z,
18%,
14Z,
9%,
7%,
(continued)
Incidence and age
20 months

19 months

20 months
20 months









4. 5 months



23 months
14 months
21 months
27 months
21 months
21 months
22 months

Comments
Deprived of mammary tumor
agent by foster nursing.




Genotypes: AYA males
Aa males
A^A females
AYA males
Aa males
AYA females
Aa females
Weight and growth factors
considered Important.
Study of effects of As and
Se on tumor incidence.


Authors cited data of
Bentvelzen and Szalay,
1966.





Reference
Heston, 1958





Heston and
Vlahakis, 1966







Schrauzer
et al. , 1978


Shlmkin and
Stoner, 1975





NMRl-Neuherberg
Wild house mice
(Hue mueculue)
Lymphoma
Lung tumors
Glomerulosclerosis
Ovarian neoplasms
Adenomatous hyperplasia of
the glandular stomach

Pulmonary, reticulem-cell
(type B), granuloaa-cell of the
ovary, hepatoma, hemangioendo-
thelioma, mammary, lymphocytic
neoplasm
41Z
16Z
13Z (50Z survival time
12Z  is 600-700 days)
 7Z
36Z, 2-24 months
64X, 25-33 months
                                                                                                                                                   U>
                                                                                                                                                   O
                                                                                                                                                   NJ
                                                                                                                                Luz, 1977
Tumors in mice received
in 1951 and their descendants
until 1961.
Andervont and
Dunn, 1962

-------
                                                          Table 5.11 (continued)
         Animal
          Tumor and organ
      Incidence and  age
                                Comments
                                   Reference
 C3Hf/Anl(Anl 70)
 C3H/HeIH
 GR/N
 BALB/cfC3H
 C57BL/6XC3HF.
 (called     i
 B6C3F.)
 AKR
 BALB/c
 C57BL/6_
 C3H/HeN

 C3H/HeJ
 C3H/He
C3H/He
C3H/A/BOM
C3HeB/Fej
C3H
C3Hf
C3He
 Liver tumors
 Mammary tumors
C3H-A
     vy
C3H-AvyfB
Mammary  tumors
Mammary  tumors
Hepatoma
Hepatoma
Hepatoma
Mammary tumors
Mammary tumors
Females 6-13Z, males 41-
682, before  12 months

99*, 7.2 months
70%, 10 months

<10Z, 8 months
OX, 8 months
32, 15 months
1Z, 15 months
5Z, 8 months

27%, duration of experiment,
2 years.   (In another table,
incidence  appears to be 100Z.)
80-100% within 8 to 18
months
66%, 22-25
  received
  starting
50%, 18-20
  received
  starting
25Z, 12-16
  received
months, controls
rat albumin
at 9 months.
months, controls
adjuvant in PBS
at 2 months.
months, controls
adjuvant in PBS
85%
72%
78Z
                                        All  killed  at  14 months
                                                                100Z,  12 months
100Z, 6-7 months
90Z, 15 months

20Z, 15 months
                      Studied enhancement of tumor
                      incidence.

                      Demonstration of mammary
                      tumor virus antibodies in
                      different strains.  Highest
                      titers in tumor bearing
                      animals.
                                Peraino, Fry, and
                                Staffeldt, 1973

                                Ihle, Authur, and
                                Fine, 1976
Reduction of tumor incidence
by manipulation of estrogen
and prolactin interactions
with administration of 2-
bromo-d-ergocryptine, 17 B-
estradiol, or Enovid.

Strain carrying the Bittner
oncogenic virus incidence
greater under chronic stress.

Study of increased levels of
serum alpha-fetoprotein in
mice with spontaneous liver-
cell cancer.
                      In males.
                      C3Hf male fed Purina
                      chow instead of the NCI
                      pellets had a 57Z incidence.

                      Males (also nearly all
                      females at 16 months).

                      Females with MTV.
                      Female breeders free of MTV
                      but having NIV.
                      Female virgins free of MTV
                      but having NIV.
                                                      Helsch, 1976
                                                      Riley, 1975
Jalanko et al.,
1978
                                                                                                                                                      OJ
                                                                                                                                                      O
                                                                                                                                                      UJ
                                Heston, Vlahakis,
                                and Deringer, 1960
                                                                 Heston and
                                                                 Vlahakis, 1968

-------
                                                           Table 5.11  (continued)
          Animal
                                       Tumor and organ
                                        Incidence and age
                                                                                                             Comments
                                                                                                                                      Reference
Hamster

  Syrian, Eppley colony,
  Hannover colony
  BIO 4.24

  BIO 45.5
Overall tumor Incidence
Primary tumors in respiratory,
digestive, urogenital, endocrine,
vascular, lymphatic systems

Adrenal tumors
32Z, average
41Z survival time for animals
with multiple tumors was 97
and 85 weeks, respectively

52Z, average age at autopsy,
80.3 weeks
17Z
Study of spontaneous tumors     Pour et al.,
and diseases In Syrian hamster  1976
colonies.
                                Hotnburger and
                                Russfield, 1970
Rat
  CD
  CD-I
  BN/BiRij
  LOU/C
  LOU/M
  AXC
  OKA
   (LOU/CXAXC)  F.
   (LOU/CXAUG)F.
   (LOU/CXOKA)F7
   GG
Sarcoma, fibroma, adeno-
carcinoma, adenoma, hemangioma,
fibroadenoma, adenoma, leiomyoma,
neck, Jaw, flank, shoulder,
salivary gland, pituitary gland,
thyroid gland, spleen, mammary
gland, uterus

Papillary  tumors - urinary
bladder
Squamous cell carcinomas -
ureter
 Ileocecal  immunocytomas
 Ileocecal  lymph node  for most
   NB
 Phaeochromocytoma  (adrenal
 gland)  the  major tumor.
 Numerous  others also occurred
 in breast,  pituitary gland,
 thyroid gland,  testis,  pancreas,
 liver,  kidney,  ovary,  salivary
 glands, uterus, and meninges

 Adrenal carcinoma
252, weeks of observation of
tumors ranged from 23 to 80
weeks
Males
28% (bladder), 6Z  (ureter),
ages 7-48 months
Females
2% (bladder), 54*  (ureter),
ages 7-54 months
                                           8-12 months
242
1.7Z
2.6Z
OZ
16Z
122
OZ
 74Z of all males,
 50Z of all females
Controls in study of oral       Lee, Russell, and
toxiclty of organic fungicides. Minor, 1978
 Females  about  52  at  12+
 months
First report of high incidence
of such tumors in rats.  These
spontaneous tumors are rare.
Tumor incidence not homo-
geneous - could get wide
variation among litters of
the same strain.
Tumor incidence influenced
by diet through effects on
endocrine glands, used other
strains also.
Spontaneous  tumors seldom
found  in males.
Boorman and
Hollander, 1974
Beckers and
Bazin, 1978
Gilbert et al.,
1958
Noble, Hochachka,
and King, 1975
                               Breast fibroadenoma
                                                                  About 20Z at 12+ months

-------
                                                          Table 5.11 (continued)
        Animal
         Tumor and organ
     Incidence and age
                                                                                                           Comments
                                   Reference
BDX
Wistar - Af/Han-EMD
BN/Bi
AC1/N
Sarcomas - connective  tissue,
bone, neural system, skin
carcinomas, tumors of  lung,
gastrointestinal tract, genito-
urinary tract, mammary glands,
testis, adrenal glands;
Malignancies of lymphoreticular
system

Tumors of the central  nervous
system:  oligodendroglioma,
estrocytoma, mixed glioma,
pleomorphic glioma, meningioma

Mammary gland — fibroadenoma
Pituitary tumors

Lynphoreticular sarcomas
Adrenal gland, cortical adenoma

Pancreas - islet adenoma

Cervix and vagina, sarcoma

Testes-lnterstitial cell tumors,
adrenal gland, pituitary gland,
urinary bladder, mammary gland,
thymus, lymph nodes, subcutaneous
tissue, heart, vagina, salivary
gland, others
66%, 13-30 months
                                                                                                 Tumors were macroscopic,
                                                                                                 malignant as proven by
                                                                                                 histology and transplantation.
                                Zoller, Matzku,
                                and Goerttler,
                                1978
5.82, mean ages ranged from
796 to 963 days
11Z, 30 months (23-40)
Males, 14*, 28 months (15-42)
Females, 26Z, 31 months (17-39)
Males, 14X, 24 months (19-36)
Males, 12Z, 33 months (27-43)
Females, 19Z, 33 months (23-54)
Males, 152, 33 months (20-42)
Females, 11*, 33 months (23-40)
15Z, 27 months (18-38)

56Z in males
52Z in females
Developed in 169 weeks
Data for other tumor types
Included.  Study of Inci-
dence of benign and malignant
neoplasms at different ages.
                                Sum! et al.,
                                1976
Burek and
Hollander, 1977
                                                                                                                                                     O
                                                                                                                                                     Ui
                                Maekawa and
                                Odashlma, 1975

-------
                                   306

(National Academy of Sciences, 1977); however, currently proposed U.S.
Environmental Protection Agency regulations require six nonrodent animals
per exposure group (Federal Register, 1979).  This number does not
include allowances for interim sacrifices and must be increased if that
practice is followed.  Needless to say, toxic reactions with low incidence
rates are unlikely to be detected with such small groups (Barnes and
Denz, 1954).  Consequently, nonrodent test animals are usually used only
as "second" species in conjunction with larger numbers of rodent test
animals.
     For carcinogenicity testing, the number of animals (rodents) to be
used is influenced by two additional factors not pertinent to chronic
toxicity evaluations (Magee, 1970; Page, 1977b).  First, a significant
number of animals must survive to tumor-bearing age.  Second, the inci-
dence of spontaneous tumors in the control groups must be compensated
for in order to avoid jeopardizing the sensitivity of the test.  Table
5.12 shows, in relation to animal numbers, how such an increase in
spontaneous tumors in the controls will affect the incidence of observed
tumors required for significance.  Experience has shown that in most
general carcinogenicity tests, 50 animals per sex per dose level will be
sufficient to meet these two requirements (National Academy of Sciences,
1977; Page, 1977&; Sontag, Page, and Saffiotti, 1976).  However, in any
case, the number surviving to tumor-bearing age should be at least 25
per sex per level (World Health Organization, 1978).
5.2.5  Controls
     Each  chronic toxicity or carcinogenicity  test  should be evaluated
with reference to a  concurrent control  group composed of animals of  the
same species, age, sex, and weight as the treated groups (Fedeval Register,
1979; U.S. Food and Drug Administration, 1971; Weil, 1962; Weil and
Carpenter, 1969).  These animals should be handled  identically to the
test species except  for treatment with  the test material (World Health
Organization, 1978).  Most investigators require that this negative
control group contain at least as many animals of each sex as  the test
groups; other workers recommend that  it contain twice as many  (National
Academy of Sciences, 1977).  In lieu of a negative  control group, a

-------
                                   307
              Table 5.12.  Incidence of  tumors  In  treated
              groups  required  for  significance  (P  =  0.05)
               depending on experimental group  size  and
                   spontaneous  tumors in controls*2
Incidence of
tumors in controls
(%)
0
10
20
30
40
Number
10
50
70
80
90
100
of
25
20
40
52
64
72
animals
50
12
28
40
52
62
per group^
75 100
8 6
24 21
36 34
47 45
58 55
             Calculations based on tabulations of Mainland  and
       Murray, 1952, as cited in Page, 19772).
             Controls and treated groups of same size.
            Source:  Adapted from Page, 1977Z?.  Reprinted with
       permission of the publisher.

vehicle control group is often required when the test material is admin-
istered by gavage (World Health Organization, 1978).  If the toxic
properties of the vehicle are not well documented, both negative and
vehicle control groups may be recommended (Federal Register, 1979; Page,
19772?).
     The use of "positive controls," animals treated with a chemical of
known toxic or carcinogenic potential utilizing the same or a very
similar test design, is occasionally recommended for chronic toxicity  or
carcinogenicity tests (Page, 1977Z?).  In chronic tests, its usefulness
is not well established (Benitz, 1970), and it is only infrequently
suggested in order to detect borderline effects and minimal enhancement
of normal pathological or age-related conditions (Federal Register,
1979; National Academy of Sciences, 1977).
     For carcinogenicity tests, positive controls can provide more
information, including:  (1) establishing the test animals' ability to
respond to a carcinogenic insult; (2) helping detect genetic drift; (3)
providing baseline information on new test species or strains;  (4)
revealing any accidental inclusion of extraneous factors that could

-------
                                   308

 affect the test; (5) providing an indirect evaluation of the laboratory
 and its general bioassay program; and (6) assessing the relative carcino-
 genic potential of the test chemical (Food Safety Council, 1978; Page,
 1977Z?; World Health Organization, 1978).   Despite all this potential
 information, the applicability of positive controls is limited.   Most
 discussions  suggest that it be applied  only to series of analogous
 chemicals  and need not be done with  every study (Food Safety Council,
 1978;  Page,  1977fc;  Peck, 1974; U.S.  Food  and Drug Administration, 1971).

 5.2.6   Age
     The age at which animals are started on test is an important and
 sometimes  overlooked consideration in testing for chronic toxicity  and
 carcinogenicity.   Because it is a basic principle of chronic toxicity
 and carcinogenicity testing that test animals should be exposed  to  the
 test material for a major portion of their life span, it is  generally
 agreed that  animals should be put on test at the earliest practical age
 (National  Academy of Sciences, 1977).   In most published screening
 studies for  carcinogenesis, weanling or immediately postweanling rats
 and mice have been used (Loomis,  1974;  Magee, 1970; Ministry of  Health
 and Welfare  Canada,  1975;  National Academy of Sciences, 1977; World
 Health Organization, 1978).  Effects of the age of the test  animal  on
 its response to carcinogenic challenge  are well documented,  and  the
 remainder  of this section will be devoted to a discussion of these
 effects.
     The use of weanlings or immediately  postweanlings permits testing
 under  a variety of conditions not present in mature animals  (e.g.,
 during periods of active protein synthesis,  cellular proliferation,  and
 sexual maturation).   In all of these physiological stages the weanling's
 response to  carcinogens is expected  to  be more acute than that of adults
 (Weisburger  and Weisburger, 1967).
     Numerous  studies have demonstrated that infant,  newborn, and even
 in utero rodents  may have  greatly increased  susceptibilities to  certain
carcinogens  compared with  weanlings.  For example,  Pietra, Spencer,  and
Shubik  (1959)  observed a 32% incidence  of malignant lymphomas in 11- to
24-week-old  Swiss mice following  a single 30-iig subcutaneous dose of

-------
                                   309
9,10-dimethyl-l,2-benzanthracene administered at age 12 h or less.  Sim-
ilarly, Kelly and O'Gara (1961) induced pulmonary tumors in essentially
all 18-h-old albino mice treated subcutaneously with 0.02 mg dibenz(o,7z)-
anthracene or 0.011 mg 3-methylcholanthrene.
     Many other workers have also demonstrated the extraordinary sensi-
tivity of newborn mice to certain carcinogens (Axelrad and van der Gaag,
1962; Berenblum, Boiato,  and Trainin, 1966; Chieco-Bianchi et al., 1963;
De Benedictis et al.,  1962; Doell and Games, 1962;  Flaks,  1965, 1966;
Gargus, Paynter, and Reese, 1969; Kaye and Trainin,  1966;  Klein, 1963;
Liebelt, Liebelt, and Lane, 1964; Liebelt, Yoshida,  and Gray, 1961;
Nishizuka, Nakakuki, and Sakakura,  1964;  Roe, Mitchley, and Walters,
1963; Toth, Magee, and Shubik, 1964;  Toth, Rappaport, and Shubik,  1963;
Vesselinovitch and Mihailovich, 1966; Vesselinovitch, Milhailovitch, and
Itze, 1970; Vesselinovitch et al.,  1972,  1975; Vesselinovitch, Rao, and
Milhailovitch, 1975).   Similar and related studies have been performed
with hamsters (Lee, Toth, and Shubik, 1963) and rats (Baba and Takayama
1961; Howell, 1963; Toth and Shubik,  1963).  Many of these studies were
critically reviewed by Toth (1968)  or Delia Porta (1968).
     In view of the apparently accentuated sensitivity of newborn animals
to selected carcinogens,  some early workers believed that a relatively
quick and sensitive screening test for carcinogens might be achieved by
exposing newborn animals to single doses of the test material (Delia
Porta and Terracini, 1969;  Epstein, Andrea, and Jaffe, 1967; Gorrod,
Carter, and Roe, 1968; Roe, Carter, and Adamthwaite, 1969; Roe, Rowsen,
and Salaman, 1961; Toth,  1968).  However, subsequent research indicated
such exposures could be more complex than they first appeared.  For
example, treatment with #-hydroxy-#-2-fluorenylacetamide produced no
detectable tumors in infant rats but did induce tumors in weanlings
under similar conditions (Weisburger et al., 1970).  Also, dimethylnitro-
samine induced renal adenomas in mice exposed as adults, but not in
those exposed as newborns (Terracini et al., 1966).  In retrospect, it
became apparent that since newborns differ from weanlings or adult
animals in metabolic capability, viral susceptibility, and hormonal
status, as well as other anatomical and physiological  characteristics,
it is illogical to expect identical responses from each of these age

-------
                                   310
groups  (Ministry of Health and Welfare Canada, 1975; Rice, 1976).  It is
reasonable  to expect newborns to  respond more sensitively than adults to
ultimate carcinogens because of reduced immunological and hormonal
competence.  But because of the lack  of well-developed metabolic capabil-
ities,  responses of such animals  to proximate carcinogens that require
metabolic activation will obviously be deficient or absent compared with
more mature animals (Page, 19772?).  It is thus clear that negative
results from tests involving newborn  animals cannot be the sole basis of
evaluating  the carcinogenicity of a test material.  Indeed, Roe (1975)
even questioned the wisdom of accepting positive results from such
experiments.
     In principle, test sensitivity and age-related effects should be
maximized if animals are exposed  to the test material during all prenatal
and postnatal phases of their lives (Munro, 1977; Page, 1977fc; U.S. Food
and Drug Administration, 1971).   Such a procedure requires treatment of
the pregnant mother and continuing postnatal exposure of the offspring.
Such a  bigenerational exposure was strongly recommended as a routine
approach to carcinogenesis testing by several national and international
authorities following the discovery of vaginal adenocarcinomas in daugh-
ters of women who had received diethylstilbestrol during pregnancy
(Greenwald et al., 1971; Herbst,  Ulfelder, and Poscanzer, 1971; Munro,
1977).  Furthermore, several subsequent studies showed that prenatal
exposures to certain known carcinogens in fact result in high incidences
of tumors in offspring (Andrianova, 1971; Goerttler and Lohrke, 1977;
Mohr, 1973; Spatz and Laqueur, 1967;  Svenberg et al., 1972; Tomatis et
al., 1971; Turusov et al., 1973).  Ivankovic (1973) reported transpla-
cental  induction of tumors in rats with ethylnitrosourea.  Five to
eighty mg per kg of body weight of the compound given as a single dose,
intravenously or orally, on day 13 to 23 of pregnancy produced a signif-
icant yield of tumors of the nervous  system in the offspring.  However
the 5-mg/kg dose was ineffective  in adult rats observed throughout their
entire life span.  In fact, a notable yield of tumors in the nervous
system of adults was obtained only when 140 to 200 mg/kg were given,
thus demonstrating increased sensitivity of the fetus to carcinogenesis.

-------
                                   311
However, routine testing for carcinogenesis through prenatal and bigen-
erational exposures is complicated by several factors that have not yet
been adequately resolved.  For example, dose determination and regulation
for animals in utero is uncertain, and tolerances are variable and age
dependent (Vesselinovitch,  1973).  Also,  many substances likely to be
tested as carcinogens are teratogenic and may jeopardize the outcome of
the cancer bioassay through reproductive deficiencies unless doses are
carefully selected and timely administered.   Furthermore, certain classes
of compounds likely to be tested for carcinogenicity, such as chlorinated
aliphatic and aromatic hydrocarbons, tend to accumulate in body fat and
liver and may interfere with normal reproductive processes even though
they are not strongly teratogenic.  In an effort to minimize such repro-
ductive interferences, some authorities recommend limiting in utero
exposures to the second half of pregnancy when organogenesis is complete
(Golberg, 1974); otherwise, teratogenicity studies would have to be done
and nonteratogenic doses would have to be used for in utero studies.
However, no generally accepted guidelines exist with respect to in utero
and bigenerational exposures for carcinogenesis testing (Munro, 1977).
Except for special studies, such as effects of certain food additives,
pesticides, and drugs on pregnant women (U.S. Food and Drug Administra-
tion, 1971), bigenerational and in utero exposures are not generally
made a part of routine carcinogenesis testing today.

5.2.7  Sex
     Current practice calls for the use of equal numbers of male and
female test animals in assays for chronic toxicity  (Federal Register,
1978; Food Safety Council,  1978; National Academy of Sciences, 1977;
World Health Organization,  1978), although early workers questioned the
need for equity (Barnes and Denz, 1954).  Essentially all test guide-
lines require that both sexes of the test species be used in carcino-
genicity assays (Page, 1977i>).  However, female mice are preferred by
some researchers because they are less aggressive and survive better
than males; in some instances, male animals may be  preferred when  females
are needed for breeding.  In the following section, data will be presented
that illustrate sex-related differences in the response  of  animals  to

-------
                                   312
 chemical carcinogens and the effects certain intrinsic and extrinsic
 factors of the animal test system have on these differences.   Comparisons
 will be made of the responses of male and female animals to chemicals
 within  selected chemical classes:  polycyclic  hydrocarbons, nitroso
 compounds  and  amines,  and halogenated hydrocarbons.
      Akamatsu  and  Barton (1974)  compared the neoplastic  response of both
 sexes of  five  strains  of mice that had received 1  mg of  3-methylcho-
 lanthrene  intragastrically.   Tumor incidence varied  with strain and,  in
 some cases,  sex.   Sixty-seven percent of the males of the BTO strain
 developed  gastric  tumors while only  30% of the females did, and more
 males (25%)  of the C3H/HeOs  strain had gastric tumors than did the
 females (12%).   The C57BL/60s females had an increased incidence of
 lymphomas,  25%,  while  only 11% of  the treated  males  responded.  More
 females of  the BALB/cOs  strain developed pulmonary tumors (62%) than did
 the  males  (50%); methylcholanthrene-treated males  in all five strains
 developed  amyloidosis  more often than did the  females.   Females of two
 strains of  mice (BTOs  and C3H/HeOs)  had a high incidence of spontaneous
 mammary tumors,  which  was not increased by methylcholanthrene; however,
 strains with low mammary tumor incidence (BALB/cOs and C3H/HeOs),  treated
 with methylcholanthrene,  developed statistically significantly more
 mammary tumors than did  their untreated controls.  The C57BL/60s females,
 and  males  of all strains,  did not  develop any  mammary tumors.   The data
 (Table  5.13) in this experiment  suggest that differences in tumor  response
 of mice to  systemic administration of methylcholanthrene are  sex related
 and  that sex-related differences are strain dependent.
      Wistar  rats given daily  intragastric instillations  of 3-methylcho-
 lanthrene  for  9 weeks  (for a  total of 270 mg)  also displayed  distinct,
 sex-related  differences  in neoplastic responses (Gruenstein,  Meranze,
 and  Shimkin, 1966).  Results  of  the  study are  summarized in Table  5.14.
Of 122  treated  females,  115  (94%)  developed 120 total neoplasms.  Of
 these,  117 were  malignant.  Of 81  treated males, 23  (28%)  developed 26
malignant neoplasms.   The  difference in the tumor  incidences  of male and
female  rats can  be  accounted  for mainly by the difference  in  the numbers
of mammary carcinomas  (87% of  the  females,  12.4% of  the  males).  However,
16% of  the males developed benign  and malignant cutaneous  tumors,  while

-------
                Table 5.13.   Systemic tumor induction in mice with 3-methylcholanthrene by the intragastric route
Strain
BTO
Treated0 male
Untreated*5 male
Treated female
Untreated female
C57B1/60
Treated male
Untreated male
Treated female
Untreated female
BALB/cO
Treated male
Untreated male
Treated female
Untreated female
C3HeB/0
Treated male
Untreated male
Treated female
Untreated female
C3H/HeO
Treated male
Untreated male
Treated female
Untreated female
Number
of
animals

51
93
20
174

85
90
20
40

36
18
42
104

70
142
32
193

24
253
43
166
Tumor incidence (X)
Gastric

67
0
30
0

15
0
20
0

11
6
14
1

34
0
38
0

25
0
12
0
Skin

4
2
5
1

3
0
5
0

3
0
3
4

0
0
6
0

9
0
0
0
Lymphomas

0
4
5
2

11
6
25
3

20
6
31
13

5
5
3
6

4
0
9
1
Hepatomas

4
11
0
1

1
1
0
0

3
0
3
2

3
18
0
0

4
34
0
0
Pulmonary
tumor

4
2
0
1

1
0
5
0

50
0
62
1

3
0
0
3

4
0
0
0
Bladder
tumor

4
0
0
0

0
0
0
0

0
0
0
0

0
0
0
0

0
0
0
0
Mammary
tumor



85
70



0
0



24
12



6
2



93
96
Ovarian
tumor



0
2



0
0



5
4



11
O



0
0
Amyloidosis

28
19
25
2

13
2
10
3

6
0
0
0

11
0
0
0

4
0
0
0
Mice
with
tumors

67
19
95
76

35
7
45
3

50
11
81
36

29
22
56
13

46
34
96
96
aAll treated animals  received  1  mg  3-methylcholanthrene in olive oil.


 Controls received olive oil alone.

Source:  Adapted from Akamatsu and  Barton,  1974.
                                                                                                                                      co
                                                                                                                                      !-•
                                                                                                                                      co

-------
Table 5.14.  Influence of sex on response  to  carcinogens
Species
Wistar rat


Fischer rat

Lister rat





B6C3F} mouse





C3AF} mouse





Buffalo rat



C3H/HeN
mouse

C57BL/6JN
mouse
Albino mouse
Age at start
of experiment
6 weeks


3-6 months

2-3 months





1 day
15 days
42 days
1 day
15 days
42 days
1 day
15 days
42 days
1 day
15 days
42 days
4 weeks
12 weeks
24 weeks
52 weeks
8 weeks


8 weeks

8 weeks
Chemical
3-Methylcholan-
threne

3-Methylcholan-
threne
Dimethylbenz-
anthracene




Benzo(a)pyrene


Benzo(a)pyrene


Benzo(a)pyrene


Benzo (a) py rene


Diethylnitros-
amine


4-Hydroxyamino-
quinoline 1-
oxide (4HAQO)
4-HAQO

4HAQO
Dose
270 mg
(total)

1.6 mg/week

o os)0'5*
°-°T5Ji.o%
^2.0%
0 OS)0*5*
V • U Jl « ABl
. >1.0%
mL)2.0%
75 ug/g body
weight

150 yg/g body
weight

75 pg/g body
weight

150 ug/g body
weight

0.0114%



1 mg


1 mg

1 mg
Route
Intragastric
instillation

Skin painting

Salivary gland


Salivary gland


Intraperitoneal


Intraperitoneal


Intraperitoneal


Intraperitoneal


Diet for 26
weeks


Subcutaneous


Subcutaneous

Subcutaneous
Tumor
type
Sebaceous
gland
Cutaneous
Skin

Carcinoma


Sarcoma


Liver


Liver


Liver


Liver


Total



Sarcoma


Sarcoma

Sarcoma
Tumor
incidence „ ,
Reference
Male
18%

16%
Female
0.8% Gruenstein, Meranze,
and Shimkin, 1966
0.8%
No difference Zacheim, 1964

18%
75%
83%
91%
75%
73%
55%
60%
13%
81%
58%
9%
34%
27%
0%
46%
23%
3%
60%
54%
33%
0%
40%


10%

11%

0% Glucksmann and
33% Cherry, 1971
71%
49%
70%
100%
7% Vesselinovitch et
7% al., 1975
0%
18%
7%
0%
2%
2%
0%
2%
2%
0%
21% Reuber, 1976
38%
23%
0%
20% Shirasu, 1965


10%

9%

-------
Table 5.14  (continued)
Species Afe at s*a" Chemical
K of experiment
Holtzman

Fischer rat

Wistar rat

B6C3Fi mouse 6 weeks


Osborne- 3-4 months
Mendel rat

Osborne- 3-4 months
Mendel rat
Osborne- 3-4 months
Mendel rat

Wistar rat , 12 hours

ACX rat
Intact 2-4 months
Castrate
Castrate-
hormone
Ethionine

Ethionine

Ethionine

Benzidene
hydrochloride

p-Dimethylamino-
benzene-1-azo-
1-naphthalene
p-Dimethylamino-
azobenzene
p-Diraethylamino-
benzene-1-azo-
2-naphthalene
p-Dimethylamino-
azobenzene

2-Diacetylamino-
fluorene


Dose Route
Diet for 7.5
months
Diet for 7.5
months
Diet for 7.5
months
50 ppm Diet
150 ppm Diet

0.075% Diet


0.060% Diet

0.075% Diet


1.2 mg Subcutaneous


166-217 mg/kg Diet
body weight


Tumor
type
Liver

Liver

Liver

Liver
Liver

Liver


Liver

Liver


Liver


Liver



Tumor
incidence
Male
60%

100%

100%

6%
44%

75%


85%

98%


Female
25%

90%

100%

26%
94%

17%


19%

100%


No difference


60%
33%
29%



12.5%
0%
86%

Reference
Farber, 1963





Vesselinovitch, Rao,
and Mihailovicch,
1975
Mulay and O'Gara,
1959






Baba and Takayama,
1961

Morris and
Ferminger, 1956


                                                                                  
-------
                                   316
 the incidence of those tumors in females was less than 1%,  and male rats
 developed more sebaceous gland tumors (18% incidence) than  females
 (<0.8%).   Other types of tumors were induced in this study, but no other
 sex differences were  obvious  in the  responses.   The cutaneous  tumor data
 were at variance with the results of an earlier experiment  in  which
 Zackheim  (1964)  studied the cutaneous response  of several strains  of
 rats to skin  painting with methylcholanthrene,  anthramine,  and 7,12-
 dimethyl(a)benzanthracene.  Although he reported differences in types  of
 skin tumor that were  related  to the  chemicals administered, no significant
 strain or sex-related differences were observed.   Gruenstein et al.
 suggested that the sex differences seen in their experiment were influenced
 by  route  of administration.   In Zackheim's experiment the route was
 external  (skin painting),  whereas the route used in the Gruenstein et  al.
 study was internal (intragastric).
      Glucksmann and Cherry (1971) reported that,  when low concentrations
 of  0.5% or 1.0% 9,10-dimethyl-l,2-benzanthracene were injected into rats
 locally in the salivary gland,  about twice as many sarcomas and carcinomas
 were induced  in males as in females  (Table 5.14).   These effects were
 confirmed when the administration of female hormones, estrogens, to
 males reduced the number of dimethylbenzanthracene-induced  tumors  by one-
 half and  the  administration of a male hormone,  testosterone, doubled the
 number of tumors in carcinogen-exposed females.   However, when 2%  dimethyl-
 benzanthracene was injected,  the sex difference disappeared, suggesting
 that the  sex  differences observed were related  to carcinogen dose  and
 were therefore quantitative rather than qualitative.
      In an experiment designed to test the effects of age,  sex, and
 strain on tumor  induction  with benzo(a)pyrene,  two strains  of  mice of
 various ages  were injected intraperitoneally, with one dose of the carcin-
 ogen  per  mouse (75 or 150  yg  per g of body weight) (Vesselinovitch et
 al.,  1975).   The life span study revealed  specific effects  associated
with  each of  the modifiers (Table 5.14).   Sex-influenced differences
were most  prominent in induction of  liver  and lymphoreticular  tumors^.
Male  rats  of  both strains  developed  significantly more liver tumors than
 females,  whereas  females of both strains developed more lymphoreticular
 tumors than the males.   As  the  animals grew older, the response of the

-------
                                   317
liver to the carcinogen decreased, the differential between the two
sexes decreased, and the sex response therefore appeared to be related
to age.
     Reuber (1976) examined the effects of age and sex on the induction
of preneoplastic and neoplastic lesions in the esophagus of Buffalo
strain rats with diethylnitrosamine.   Starting at 4, 12, 24, and 52
weeks of age, the animals were fed a diet containing 0.0114% diethyl-
nitrosamine for 26 weeks.  At 26 weeks, male rats that had been 4 weeks
old when the treatment was started had a higher incidence of carcinomas
of the esophagus (60%) than females of the same age group (21%).   Among
animals that began receiving treatment at 12 weeks of age, the differ-
ence between the sexes was slight by 28 weeks; in those that received
the first treatment at 24 weeks, tumor incidence between the two sexes
was practically the same by 36 weeks.  The differences in the responses
of the males and females diminished with increasing age.  This finding
supports the previously cited data of Vesselinovitch et al. (1975).  See
Table 5.14.
     As in the experiments of Akamatsu and Barton (1974) with methylcho-
lanthrene, the strain of test species was shown by Shirasu (1965) to
influence the difference in sex response of mice to 4-hydroxyaminoquin-
oline-1-oxide.  Results of the experiment are summarized in Table 5.14.
Nine months after starting subcutaneous injection of 1 mg of the carcin-
ogen (divided into 10 weekly injections), 40% of the male mice of the
C3H/HeN strain had developed sarcomas at the injection site, while only
20% of the females developed the same type of tumor.  Other strains
tested developed sarcomas with incidences of 9% to 40%, but no sex-
related differences were seen.  Papilloma development appeared to be sex
related in the C3H/HeN strain (papillomas in 40% of the females and 0%
of the males), in the C57BL/6JN strain  (papillomas  in 0% of the females
and 20% of the males), and in a noninbred albino strain (papillomas in
9.1% of the females and 0% of the males).  None of  the  females and only
10% of the males of the C57BL/6JN strain had leukemia.  Subcutaneous
injection of 4-nitroquinoline-l-oxide produced sarcomas in  10% of  the
C3H/HeN males but none in the females.
     One other experiment that showed sex-related differences  in response
to amino compounds was that of Farber  (1963), who observed  the response

-------
                                   318

 of Holtzman rats to hepatocarcinogenesis with ethionine.   Feeding the
 chemical in the diet for 7.5 months produced liver cancer in 60% of the
 males and in 25% of the females.   Almost 100% of both sexes of two other
 more  sensitive strains (Fischer and Wistar)  tested in the same experiment
 developed liver cancers (Table 5.14).
      Benzidine dihydrochloride, administered to mice continuously in the
 diet  at  doses of 50,  100,  and 150 ppm from the 6th to the 90th week of
 age,  induced liver tumors  at dose-related incidences of 6% to 44% in
 males and 26% to 94% in females (Vesselinovitch, Rao, and Milhailovich,
 1975).   When the carcinogen was administered intermittently instead of
 continuously, the sex difference was abolished (Table 5.15).   Regardless
 of the schedule of administration, males developed more tumors of the
 Harderian gland than females.  Lung adenomas were found in more males
 (23%) than females (7%), but only when feeding was intermittent.   When
 the animals were fed the carcinogen twice (at 7 and then  at 27 days of
 age), 66% of the males and none of the females developed  liver tumors,
 whereas  feeding at 42 and  62 days produced liver tumors in only 4% of
 the males and, again, none in the females.  These data demonstrate the
 influence of age on tumor  induction in males in particular.
      Mulay and O'Gara (1959) demonstrated that Osborne-Mendel male rats
 fed p-dimethylamino-benzene-1-azo-l-naphthalene (DAN) and p-dimethyl-
 aminoazobenzene (DAB) were more sensitive to liver tumor  induction than
 females.   (See Table 5.14.)   Daily administration of DAN  induced liver
 tumors in 75% of the males and in 17% of the females, whereas DAB induced
 liver tumors in 85% of the males and 19% of  the females.   Male and
 female mice fed p-dimethylaminoazobenzene-l-azo-2-naphthalene (DA-2-N)
 responded with the same incidence of liver tumors (98% and 100%), but
 the tumors appeared more slowly in females,  suggesting that they were
more  resistant to DA-2-N than were males.   Baba and Takayama (1961), on
 the other hand,  were unable to demonstrate sex differences in Wistar
rats  injected subcutaneously with DAB.   The  investigators suggested that
if  the experiment had been stopped on the 200th day, instead of on day
380,  hepatoma incidence would have differed  greatly between the two
sexes, as  induction time was longer for females.  They also speculated
that  if  the doses of  carcinogen had been larger the difference in sex
might have  been  obscured.   The authors  concluded that in  DAB carcino-
genesis  sex of the animal  does not affect the transformation of normal

-------
               Table 5.15.  Comparison of  continuous versus  intermittent administration of BZ-2HC1 on development of
                                                  liver, Harderian gland, and  lung
Dose
(ppm)
50
100
Sex
M
F
M
F

Liver
Continuous
Ratio0
3/50
13/50
11/50
32/50
%
6
26
22
64
tumors
Harderian gland tumors
b
Intermittent
Ratio
3/75
4/75
12/75
17/75
%
4
5
16
23
Continuous
Ratio
9/50
3/50
18/50
3/50
%
18
6
36
6
Intermittent
Ratio
8/75
2/75
11/75
5/75
%
11
3
15
7
Lung adenomas
Continuous
Ratio
2/50
2/50
3/50
2/50
%
4
4
6
4
Intermittent
Ratio
17/75
5/75
19/75
4/75
%
23
7
25
5
      Animals were given food containing specified amounts of BZ«2HC1.

      Mice received BZ-2HC1 stomach intubation in amounts equivalent to specified dosages; mice were intubated twice weekly.  The
amounts at each treatment were 0.5 mg (corresponding to 50 ppm series) or 1.0 mg (corresponding to 100 ppm series) per animal at

treatment.

     ^Number of mice bearing specified tumors per number of BZ*2HCl-treated animals.

     Source:  Adapted from Vesselinovitch, Rao, and Mihailovitch, 1975.  Reprinted with permission of the publisher.
                                                                                                                                      CO
                                                                                                                                      M
                                                                                                                                      vo

-------
                                   320
 cells  to  malignant  ones,  but that  it  does affect proliferation of the
 cells  and thus  appearance of the cancer.   This supports the opinion of
 Glucksmann and  Cherry (1971) that  sex-related differences in tumor
 induction are quantitative rather  than qualitative.
     Morris and Ferminger (1956),  using 2-acetylaminofluorene, induced
 tumors in male  and  female rats (castrated and normal)  that, in some
 cases, were treated with  sex hormones.   Most hepatomas were confined to
 the  livers of intact males and of  castrated females  treated with testos-
 terone propionate.   Intact females,  castrated males,  castrated females,
 and  castrated males treated with diethylstilbesterol  were less suscept-
 ible (Table 5.14).   Therefore, it  appeared that under those experimental
 conditions the  presence of androgen was important for the development of
 hepatomas in the rat.   Estrogen did not seem to have  a protective effect
 against tumor development.
     Weisburger (1977)  tested several halogen compounds for carcinoge-
 nicity in B6C3Fj mice and in Osborne-Mendel rats.  Some of the sex differ-
 ences  seen after 78 weeks of dosing by gavage were:
  1.  Chloroform-treated male rats  had an increased incidence of kidney
     tumors,  and females  developed more thyroid tumors.
  2.  l,2-Dibromo-3-chloropropane caused many mammary  carcinomas in
     female rats.
  3.  1,2-Dichloroethane caused some increase in hepatocellular carcinomas
     and  stomach tumors in male mice  and induced increased numbers of
     mammary  tumors in  female rats.
  4.  lodoform increased thyroid tumors  in male rats and hepatocellular
     carcinomas in  male mice.
     Both sexes of  mice have been  found to be equally  susceptible to the
hepatocellular  effects  of carbon tetrachloride (Andervont,  1958;  Weisburger,
1977),   whereas  female Buffalo strain  rats have appeared to be more
sensitive  than  males  (Reuber and Glover,  1967).

5.2.8   Conclusions
     The need for lifetime  exposures  in chronic  toxicity and carcino-
genicity  tests  limits the choice of primary test animals to relatively

-------
                                  321
short-lived rodents in all but a few exceptional cases.  Rats are the
overwhelming choice of most investigators for chronic testing, with mice
and dogs distant second and third choices,  respectively.
     In addition to the primary test animal, most authorities recommend
the use of a second species in chronic toxicity and carcinogenicity
tests to reveal a broader range of toxic effects.  Most,  but not all,
authorities recommend a nonrodent species for chronic tests.  Among
nonrodents, the dog is most frequently used, but other species may be
selected if their metabolic processes are thought to resemble those of
man more closely.
     Rats, mice, and hamsters are generally selected for carcinogenicity
assays.  No test animal has been found to be an ideal surrogate for man
in testing for long-term toxicity; however many species do respond to a
variety of toxic substances and are reliable subjects for testing.
Selection of the most appropriate species or strain for testing is
highly complex and can depend on sensitivity desired by the experimenter,
physiology, anatomy, and life span of the animal.  Ideally, the test
animal that most closely resembles man would be chosen, but cost and
time requirements do not always allow this.  The choice may be simplified
through such techniques as those involving the use of structure-activity
relationships, metabolism studies, and in vitro screening.  These tech-
niques, in conjunction with standardized animal studies, could aid in
the efficient selection of the appropriate animal species.  However,
literature searches revealed a distinct lack of experiments expressly
designed to demonstrate species differences in response to  chemical
carcinogens.  Thus, it was necessary in this document  to make many
comparisons of nonstandardized data between experiments conducted in
different laboratories.  There is a definite need for  basic,  standard-
ized animal experiments that allow for the comparison  of the  responses
of various species to chemical carcinogens or classes  of carcinogens.
     The number of animals used in chronic toxicity and carcinogenicity
tests represents a compromise between requirements for good statistical
precision and reasonable costs and work loads.   Early  investigators  used
10 or fewer test animals per dose group, but most authorities now recommend

-------
                                   322
 using  50  rodents  or  4  to  8 nonrodents  per  group.   Equal  numbers  of male
 and  female  animals are normally  used.
     Each long-term  toxicity test  should be  evaluated  with  reference  to
 a  concurrent  control group composed  of animals  of  the  same  species, age,
 sex, and  weight as the treated groups.  These animals  should  be  handled
 identically to the test species  except for treatment with the test mate-
 rial.   Each control  group should contain at  least  as many animals as  the
 corresponding test group.  Negative  and vehicle control  groups are com-
 monly  used  in all long-term  tests, but positive controls are  more fre-
 quently used  in carcinogenicity  testing.
     General  agreement exists among  authorities on the need to start
 animals on  test at an  early  age  to provide maximum exposure during
 periods of  active protein synthesis, cellular proliferation,  and sexual
 maturation, as well  as adulthood.  In  most routine screening  tests for
 carcinogenesis, weanlings or immediately postweanling  animals are used.
 There  is  also general  agreement  that newborn animals are sometimes even
 more sensitive to carcinogens than weanlings and adults; however, the
 metabolic,  hormonal, and  immunological incompetence of prenatal  animals
 may  induce  responses uncharacteristic  of more mature animals.  Hence,
 some responses from  newborn  animals, taken alone,  may  not be  reliable
 indicators  of the carcinogenicity of the test material.
     In principle, test sensitivity  and age-related effects should be
 maximized if  animals are  exposed to  the test material  during  all prenatal
 and postnatal phases of their lives.   In practice, difficulties  with
 dose estimation or regulation and  teratogenic or reproductive interfer-
 ences  of  the  test material so limit  the effectiveness  of this approach
 that tests  involving in utero and bigenerational exposures  are performed
 only infrequently.   As a  consequence,  much more experience  with  these
methods will  be required  before  their  general application to  carcino-
 genesis testing can  be considered.  However, use of such tests may be
 indicated whenever the chemistry or biology  of  the test  compound, or  its
pattern of use, suggests  a high  level  of interaction with in  utero,
infant, or preadolescent  children.
     The sex  of the animal can influence the incidence,  site,  and latency
of tumors.  The host environment, which is determined  by factors such as

-------
                                  323
sex, appears to regulate the growth of the induced neoplasm.  The mecha-
nism of sex-influenced effects is not clear, and sex-related differences
cannot be explained on the basis of hormone effects alone or on the
basis of cell susceptibility.  Homburger and Tregier (1960) described
studies in rodents in which tumors induced in males grew larger when
transplanted to males than when transplanted to females.  These and
results of previous tests indicated to them that cells of the male and
female are equally susceptible to carcinogens but that the malignant
cells encounter a less favorable environment in the female than in the
male and that the tumors progress at a slower rate.  Sex-related differ-
ences in the tumor response to chemical carcinogens can be altered by
factors such as strain, species, and age of the animal; route and schedule
of administration of the chemical; dose of the chemical; and duration of
the experiment.  "Therefore, reliable testing of chemicals with unknown
characteristics would be virtually impossible using only one sex of
experimental animal.

5.3  ROUTES OF ADMINISTRATION

     The mode of exposure of experimental animals to chemicals may
influence the results of chronic and carcinogenicity studies and should
closely simulate conditions under which human exposure would most likely
occur (Ministry of Health and Welfare Canada, 1975).  Thus, the oral
route is preferred for testing compounds to which humans may be exposed
through ingestion (Ministry of Health and Welfare Canada,  1975; World
Health Organization, 1978).  For gases, volatile industrial solvents,
and other air contaminants, inhalation studies are recommmended (Clark,
1977; Nettesheim and Griesemer, 1978).  Section 5.3.1 provides an evalu-
ation of advantages and disadvantages of these exposure routes in tests
for chronic toxicity and carcinogenicity and includes a description of
other less common routes which have specific application  in tests for
carcinogenicity.  In Sect. 5.3.2 examples are presented to illustrate
differences or similarities observed in a given species following carcino-
gen exposure by different routes.

-------
                                   324
 5.3.1   Exposure  Routes
     5.3.1.1   Oral  Route — Suitable  for all  species,  the oral  route  of
 exposure  is used for testing compounds to  which humans  may  be  exposed
 through ingestion.   Oral administration may  be effected by  gavage, by
 feeding in dietary  mixtures,  or by administration in  drinking  water.
 All  three methods are useful for long-term administration and  permit
 reliable  quantitation if intake and  body weight are measured routinely
 (Ministry of  Health and Welfare Canada,  1975).   Sontag, Page,  and
 Saffiotti (1976) discussed the advantages  and  disadvantages of the three
 oral routes.
     The  gavage  method  has the advantages  of more effective hazard
 control and better  quantitation.   The test agent can  be freshly prepared
 and, because  less test  agent is needed,  less storage  space  is  required.
 Animal  handling  is  increased,  allowing frequent observation for clinical
 symptoms.  Gavage has certain disadvantages:  (1) intake of the test
 agent may be  less than  maximum,  (2)  a solvent  is often  required,  (3)
 mortality may be increased because of trauma,  and (4) animals  must be
 closely matched  by  weight.
     Feeding  through diet  mix and drinking water has  other  advantages
 and  disadvantages.   Although these methods most closely simulate the
 mode of human exposure  and allow greater total intake,  the  quantity
 ingested  may  vary,  homogenicity and  stability  of the  mix may be incon-
 sistent,  and  palatability  may be altered.  Furthermore, the concentra-
 tion of the test material  in the diet must be  altered weekly or biweekly
 during  the early part of the experiment  to maintain a constant dose
 level (in milligrams per kilogram of body  weight),  because  toxicity  is
 related to dosage per unit  of  body weight  and  food consumption per unit
 of body weight decreases with  increasing animal age (Weil,  1973).  Also,
 potential exposure  of workers  who mix and  dispense feed and water con-
 taining toxic  chemicals must not  be  overlooked.
     Dietary  feeding is generally recommended  for adult animals (Weisburger
and Weisburger,  1967),  and  intubation techniques are  reserved  for new-
born and infant  animals and for  chemically unstable compounds  (Weisburger
and Weisburger,  1967).   Pills  and capsules may be suitable  for oral

-------
                                  325
administration to large species.  The Ministry of Health and Welfare
Canada (1975) suggested experimental use of the oral route of exposure,
in certain instances, even when it is not the primary mode of human
exposure.  However, substitution of the oral route would be feasible
only if levels of the compound in the tissue are higher than those
produced following administration by the normal route for humans.  In
any case, extrapolation from one route of exposure to another must be
approached with extreme caution.
     5.3.1.2  Repiratory Routes — Experimental exposure of the respiratory
tract to chemical carcinogens is usually accomplished through inhalation
or by intratracheal instillation.  These and two other specialized
techniques which are occasionally used for carcinogenicity studies
(pellet implantation and tracheal washing) will be discussed.  The most
significant advantage of inhalation exposure is that it duplicates
conditions under which humans are exposed to air contaminants and it is
the most probable route of industrial exposure to toxic chemicals
(Nettesheim and Griesemer, 1978; Ministry of Health and Welfare Canada,
1975).  The disadvantages of inhalation techniques, some of which add
difficulty to interpretation of results, were listed by Ministry of
Health and Welfare Canada (1975):  (1) methods and equipment are complex
and expensive, (2) particle sizes must be exact, not to exceed 5 y,  (3)
the respiratory anatomy of man and animals differs in several respects,
and (4) it is difficult to determine the actual dose inhaled.
     Of the various species of experimental animals, the rat seems to  be
most suited to inhalation studies.  Size of the animal, economic consider-
ations, and similarities to man (demonstrated by the induction of squamous
cell carcinomas of bronchogenic origin) are desirable characteristics  of
the rat model (Page, 19772?).
     To circumvent the complexity of inhalation procedures,  other useful
methods for direct exposure of the respiratory tract have been developed.
Intratracheal instillation of test material permits administration of  a
controlled dose and can be used in cocarcinogenesis studies  concomitantly
with inhalation exposure (National Academy of Sciences, 1975).   In
addition, large particles and large doses can be administered, the nasal

-------
                                   326
filter in rodents is bypassed, skin contamination is avoided, and there
is less risk to personnel from carcinogen exposure.  Intratracheal
instillation is currently the method  of choice for  testing suspected
respiratory carcinogens  (Nettesheim and Griesemer,  1978).  Disadvantages
of the technique are site-to-site variability in deposition of the test
material and the formation of foci of chronic inflammation in the respi-
ratory tract.  Hamsters  appear to be  the animal most used in experiments
requiring intratracheal  injections.   Size of the animals, their low
incidence of spontaneous respiratory  infections, their low incidence of
spontaneous tumors, and  their susceptibility to various carcinogens make
hamsters suitable for intratracheal instillation.
     Implantation of carcinogen-containing pellets  into respiratory
tissues of the rat produced  squamous  cell carcinomas in the bronchial
epithelium (Kuschner et  al., 1957) and in the epithelium of heterotropic
tracheal transplants (Griesemer et al., 1977).  In  the pellet implant
system the carcinogen acts on a specific limited area determined by the
investigator, and dose-response relationships have  been established.
     The respective carcinogenicities of two occupational pollutants,
chromium compounds and polyurethane,  have been detected using the bron-
chial implant.  However, the acute and chronic traumas afforded by this
method are unavoidable and can be troublesome.
     Schreiber, Schreiber, and Martin (1975) described the induction of
squamous cell carcinomas in  the hamster using repeated localized tracheal
washings with a carcinogen.  Tumors produced by this method are easily
detected (the animal shows respiratory distress as  the tumor becomes
large) and may usually be diagnosed simply by dissecting the trachea
under the dissecting microscope (Nettesheim and Griesemer, 1978).  This
relatively new exposure  technique is  complex and traumatic to the animal,
and little is known to date  of its real potential.
     5.3.1.3  Dermal Route — Of particular interest in carcinogenicity
studies, this route exposes  the skin  directly and may result in exposure
of the whole body following  percutaneous absorption (Ministry of Health
and Welfare Canada, 1975).

-------
                                   327
     An obvious advantage of dermal application is the ease with which
the latency for cancerous or precancerous lesions can be established
(Page, 19772?).   Also, the effective cutaneous dose can be smaller than
the effective oral dose, even when absorbed (however, the maximum dose
that can be given dermally is usually less than the maximum dose that
can be administered orally); tumors arise rapidly, and they are usually
multiple; the statistical analysis in skin carcinogenesis studies is
advanced; the method is well suited to the two-stage carcinogenesis
procedure; and the technique is useful for testing carcinogens that are
destroyed in the gut.  As for disadvantages, the procedure is arduous
and expensive, the absorbed dose is not accurately known (whole body
exposure may occur when the test agent is licked off the skin), lesions
and infections caused by the vehicle may shorten the life span of the
animal, and it is unsuitable for chemicals requiring activation in the
gut (Ministry of Health and Welfare Canada, 1975).
     Weisburger (1976) suggested that mice and rabbits are generally
more responsive to dermal tumorigenesis than are other species.  Magee
(1970), on the other hand, concluded that there seemed to be no inherent
difference in the capacity of mouse and rat skin to undergo malignant
change.  This conclusion was based partly on the results of Graffi,
Hoffman, and Schutt  (1967), who induced skin tumors in mice, rats, and
hamsters by dermal application of tf-nitrosomethylurea.  However, Fare
(1966) demonstrated  that complete reliance on results of mouse skin
carcinogenesis studies could be misleading.  An azo dye, 3-methoxy-4-
dimethylaminoazobenzene, when painted on the skin of ten male  rats for
62 weeks, was a potent carcinogen, producing tumors  in 100% of the
animals.  But the carcinogen was found to be totally ineffectual when
painted on the skin  of male and female mice for averages of 62 and 30
weeks, respectively.
     5.3.1.4  Subcutaneous Injection — Subcutaneous  injection  is  a
sensitive and frequently used mode of administration of  chemical  carcino-
gens  (Page, 19772>).  Doses can be  accurately delivered,  absorption  is
slow, high concentrations  in the blood are  reached  rapidly,  and  the
compound circulates  without those  changes  that might occur in the gut

-------
                                   328
 following  oral  administration.   Repeated injections  can  be  time-consuming
 and  local  reactions  are  likely  to occur (Ministry  of Health and Welfare
 Canada,  1975),  but the appearance of  subcutaneous  sarcomas  at  the  site
 of injection  constitutes the most controversial characteristic of  the
 technique.  The production of subcutaneous sarcomas  has  been an end
 point  in the  testing of  many compounds for carcinogenic  potential.   In
 1941 Turner induced  sarcomas in rats  at the site of  implantation of
 bakelite.  Oppenheimer,  Oppenheimer,  and Stout  (1948) obtained similar
 results  using cellophane.
     Grasso (1976),  following extensive experiments  performed  in an
 attempt  to elucidate the mechanism of induction of subcutaneous tumors
 at the site of  injection or implantation,  concluded  that "a progressive
 type of  connective tissue  reaction, which has been carefully characterized
 histologically,  is responsible  for the malignant tumors  that are produced
 in the subcutaneous  tissue of rats and mice by  the following:   (a)
 solid  implants,  (2?)  injection of hypertonic, acidic,  or  surface active
 solutions, (0)  injection of macromolecular substances or substances  that
 form local deposits."  Thus,  although the subcutaneous techniques  are
 still  quite useful,  caution is  required in the  interpretation  of the
 results  of assays in which they are used.
     Homburger  and Tregier (1960)  observed species differences in  suscep-
 tibility of hamsters, mice,  rats,  and monkeys to subcutaneously administered
 3,4,9,10-dibenzopyrene.  One monkey appeared to be resistant,  but  sarcomas
were induced  in  hamsters,  mice,  and rats.   Also, strain  differences  were
 observed among  the mice  tested.
     5.3.1.5  Other  Exposure Techniques — Intraperitoneal and  intravenous
 injections and bladder implantation of wax pellets are less frequently
used techniques.
     The intraperitoneal route  of  administration is  convenient for
single-dose exposure to  a  carcinogen,  the liver being the first organ
encountered.  With procarcinogens  the tumor spectrum induced is the  same
as by  oral administration  (Weisburger,  1976), and  injection may be done
repeatedly if the carcinogen  is  soluble and if  there is  no  vehicle
accumulation.   Intraperitoneal  injection does not  resemble  the usual
routes of human exposure.

-------
                                   329
     Intravenous injection is a rapid and efficient means of delivery of
test materials to various organs.  However, it is an unlikely route for
human exposure and is not well suited for repeated injections (Ministry
of Health and Welfare Canada, 1975).
     Jull (1951) introduced the technique of surgical implantation of
wax pellets containing carcinogens into the bladders of mice.  Since
then other investigators, including Bonser et al. (1952) and Allen et
al. (1957), have used the technique successfully for testing aromatic
amines, polycyclic hydrocarbons, saccharin, and other potential carcino-
gens.  However, the U.S. Food and Drug Administration (1971) felt that
the number of chemicals tested comparatively by both bladder implantation
and more conventional methods was insufficient to provide sound inter-
pretation of results obtained by the technique.  Therefore, any chemical
clearly carcinogenic by bladder implantation would be considered suspi-
cious but should then be tested by more conventional assays.

5.3.2  Carcinogen Exposure by Different Routes
     5.3.2.1  Diethylnitrosamine in the Hamster ~ The effects of route
of administration on the carcinogenic action of diethylnitrosamine have
been tested in the Syrian hamster (Herrold and Dunham, 1963; Herrold,
1964a, 1964&).  The routes tested were intragastric, intratracheal,
subcutaneous, intradermal, topical, and intraperitoneal.  Results of  the
experiment are summarized in Table 5.16.  Irrespective of the route of
administration of diethylnitrosamine, tumors were induced in the trachea,
bronchi, nasal cavity, and liver of the hamsters.  The frequency of
tumors in the nasal cavity was considerably higher  after subcutaneous
administration of diethylnitrosamine than after  administration by intra-
gastric or intratracheal routes  (Herrold, 1964a).   Although liver tumors
did not develop in animals injected intratradically, atypism of  the
hepatic cells was observed (Herrold and Dunham,  1963).  There was no
local effect on the skin or in subcutaneous  tissue  (Herrold, 19642?).
Transplacental transfer of the carcinogen was  not demonstrated by Herrold,
but Mohr (1973) described extensive experiments  in  which he and  his
associates induced tumors transplacentally with  diethylnitrosamine.

-------
                                     Table 5.16.  Induction of tumors in the Syrian hamster with diethylnitrosamine

Route Number of
animals

Buccal pouch 10

Intradermal 19

Intraperitoneal 18

Topical 8




Dose

0.1 mg in saline
(total - 7.5 mg)
3.5 mg in water
(total - 70-84 mg)
2 mg in saline
(total - 32-56 mg)
Undiluted/' then 1:1
in water



Exposure
schedule

Twice a. week for
about 9 months
Once a week for
5-6 months
Once .1 week for
4-7 months
Once a week for
1 month then
twice a month
for 3 months

Trachea

0/10

19/19

17/18

6/8



Tumor incidence
Nasal

Anterior
0/10 0/10 0/10

10/19 3/19 10/19

4/18 4/18 5/15

2/8 4/8 6/8




cavity

Posterior
0/10

13/19

11/15

4/8




Reterencu
Kidney0

Milievskaja and
Klseleva, 1976
Herrold, 1964i

Herrold, 1964&

Herrold, 1964;


CO
LO
Pregnant female   3
  subcutaneous
5, 8, or 10 mg in
  water
1-2 days preced-
  ing delivery
  (single dose)
3/3
1/3
0/3
1/3
2/3
Herrold, 19642)
Subcutaneous

Intratracheal


Intragastric


15

11 female

14 male
15 female

13 male
2 mg in water
(total - 32-48 mg)
0.05 raL of a 1:14 ,
solution in water

0.4 mL of a 1:250
solution in water

Twice 6 week for
4-6 months
Once a week for
6 months

Twice a week for
7 months

NSU

11/11

14/14
15/15

13/13
NS

4/11

10/14
6/15

4/13
NS

0/11

0/14
10/15

12/13
14/15

3/9

1/13
6/11

4/9


0/11

0/14
7/15

5/13
Herrold, 1964j

Herrold and
Dunham, 1963

Herrold and
Dunham, 1963

     Microscopic lesions that may represent an early stage in tumorigenesis.

      Dose and schedule were changed after animals died during the first month of treatment.

     °NS — tumors observed but incidence not specified.
     Htoses administered intratracheally and intragastrically were approximately the same.

-------
                                  331
     Milievskaja and Kiseleva (1976) attempted to induce tumors in the
buccal pouch of the Syrian hamster with injections of diethylnitrosamine.
Doses of 0.1 mg per animal twice a week for about 9 months failed to
produce local or remote tumors.   The small number of animals (ten) or
the route of administration constitutes possible reasons for the lack of
carcinogenicity of diethylnitrosamine in the experiment.  The total dose
administered to the buccal pouch, however, was only one-tenth of the
intradermal dose required to produce tumors in hamsters (Herrold, 1964&);
therefore, the lack of effect could be attributed to insufficient dose.
     5.3.2.2  Methylcholanthrene in the Mouse — Skin carcinomas have
been induced in mice painted with single large doses (0.1 mL of 0.6%
solution four times in an hour)  or with repeated smaller doses (0.15%
two times per week for 30 weeks) of 3-methylcholanthrene (Bielschowsky
and Bullough, 1949; Shubik, 1950).  Table 5.17 contains experimental
details for local tumor induction with methylchloanthrene.
     When methylcholanthrene is painted on the skin of mice in doses too
low to produce tumors, croton oil, applied repeatedly, enhances the
appearance of tumors (Klein, 1952).  The promoting action of croton oil
in skin carcinogenesis appeared to be a factor in the induction of
sarcomas in albino mice following intramuscular injection of methylcho-
lanthrene (Klein, 1951).  Single injections of 0.024 or 0.004 mg of
methylcholanthrene in croton oil produced tumors in a shorter time and
with a higher incidence than those produced by the same doses of carcino-
gen injected alone.  However, croton oil did not influence  tumor number
or latency when injected intramuscularly with 0.002, 0.0008, or 0.0004
mg methylcholanthrene doses, which produced few or no sarcomas at  the
site of injection (Klein, 1953).
     Subcutaneous administration of polycyclic hydrocarbons has been
used primarily for the induction of connective tissue  tumors.   Epidermal
changes that occur with this route of exposure are not  often described.
Bhisey and Sirsat (1975) monitored histological changes in  the  skin  of
Swiss albino mice at various times after  subcutaneous  injection  of
0.5 mg of 20-methylcholanthrene  in  thiophene-free benzene.   A mild focal
epidermal hyperplasia developed, which  led  to development of squamous

-------
Table 5.17.  Local tumor induction in mice with 3-methylcholanthrene by various routes of administration
Number
Route Strain of
animals
Skin painting Kreyberg 30

CF-1 28


DBA 34






35


Intramuscular Albino A 40

40


32

32


35

36


36

35


33

37


Methylcholanthrene
dose
0.1 mL of 0.6Z in
acetone

0.15% in acetone


0.5* in olive~oil






0.5% in olive oil


0.024 mg in olive
oil
0.024 mg in olive
oil and croton
oil
0.004 mg in olive
oil
0.004 mg in olive
oil and croton
oil
0.002 mg in olive
oil
0.002 mg in olive
oil and croton
oil
0.0008 mg in olive
oil
0.0008 mg in olive
oil and croton
oil
0.0004 mg in olive
oil
0.0004 mg in olive
oil and croton
oil
Exposure schedule
4 applications at
15-min intervals

Twice & week for
30 weeks

Once, followed by
application of
5* croton oil in
olive oil 1-3
times per week
for 53 applica-
tions
Once, with no
additional
treatment
Single injection

Single injection


Single injection

Single injection


Single injection

Single injection


Single injection

Single injection


Single injection

Single injection


Average
induction
time
10.5 weeks

Not clear


59 days









160 days

95 days


168 days

123 days


183 days

185 days


254 days

272 days







Tumor
incidence
7/30 (skin)

23/28 (skin)


27/34 (skin)






0/35 (skin)


20/40 (muscle)

40/40 (muscle)


9/32 (muscle)

22/32 (muscle)


5/35 (muscle)

3/36 (muscle)


1/36 (muscle)

1/35 (muscle)


0/33 (muscle)

0/37 (muscle)


Reference
Comments
Bielschowsky 3/7 carcinomas (par-
and Bullough, tial autopsy)
1949
Shubik,


Klein,






Klein,


Klein,

Klein,


Klein,

Klein,


Klein,

Klein,


Klein,

Klein,


Klein,

Klein,



1950


1952






1952


1951

1951


1951

1951


1953

1953


1953

1953


1953

1953



2 malignancies in
69 total tumors
(partial autopsy)










Fibrosarcomas (par-
tial autopsy)
Fibrosarcomas (par-
tial autopsy)

Fibrosarcomas (par-
tial autopsy)
Fibrosarcomas (par-
tial autopsy)

Fibrosarcomas (par-
tial autopsy)
Fibrosarcomas (par-
tial autopsy)

Fibrosarcomas (par-
tial autopsy)
Fibrosarcomas (par-
tial autopsy)






                                                                                                                              w

-------
                                                           Table 5.17 (continued)
Number „«.!._, u
Route Strain of Methylcholanthrene schedule
animals dose
Subcutaneous Swiss 24 0.5 tag in thio- Single injection
phene-free
benzene
Average T,,n,«r
induction . T^or a
. . incidence
time
13/24 (skin)
Sirsat, 1975
2/24 (muscle)
19/24 (subcuta-
neous connec-
tive tissue)
3/24 (skin)
Reference
Bhisey and
Sirsat,
1975
Comments
Squamous cell carci'
noma (partial
autopsy)
Rhabdomyosarcomas
(partial autopsy)
Fibrosarcomas (par-
tial autopsy)
Papillomas (partial
                                                                                        6/24 (skin)
  autopsy)
Sebaceous cysts (par-
  tial autopsy)
Intratracheal BC3Fj




DBA/2



Bladder Albino
implant of '
pellets





36 0.5 mg in 0.2X
gelatin



50 0.5 mg in 0.2%
gelatin


37 12.5% suspension



38 20% suspension



Once a week for
6 weeks



Once a week for 4
weeks


Continuous up to
4 weeks


Continuous up to
4 weeks


First mouse 31/36 (lung)
died in
4 weeks,
last in
24 weeks
3/18 (lung)



18/37 (urinary
bladder)


22/38 (urinary
bladder)


Nettesheim and
Haramons, 1971



Nettesheim and
Hammons, 1971


Bonser et al. ,
1963


Bonser et al. ,
1963


Squamous cell
carcinomas



Squamous cell car-
cinomas (22
animals were still
alive)
Carcinomas (partial
autopsy) , 12% of
controls developed
tumors
Carcinomas (partial
autopsy) , 4.8% of
controls developed
tumors
                                                                                                                                                  CO
                                                                                                                                                  co
                                                                                                                                                  CO
aNumber of tumor-bearing mice  per  total number of mice at risk.

-------
                                   334

cell carcinomas.  Papillomas, sebaceous  cysts,  rhabdomyosarcomas, and
fibrosarcomas also developed.  Bhisey compared  the overall  lack of
epidermal hyperplasia  seen  in these  experiments to the massive epidermal
hyperplasia  thought to be a required step during neoplastic transformation
of mouse skin topically painted with methylcholanthrene.  He concluded
that "when methylcholanthrene is administered subcutaneously to Swiss
mice, epidermal hyperplasia is not a prerequisite for epidermal tumori-
genesis."
     Examples of  specialized techniques  for  local tumor induction in the
respiratory  tract and  urinary bladder include intratracheal injection
and pellet implantation, respectively.   Repeated intratracheal injections
of 3-methylcholanthrene have induced squamous cell carcinomas in the
respiratory  tracts of  mice  (Nettesheim and Hammons, 1971).   When DBA/2
and BC3Fj mice were injected weekly  with 0.5 mg of 3-methylcholanthrene
for 4 and 6  weeks, respectively, 31  of the 36 BC3Fi mice developed
squamous cell carcinomas, the first  appearing 4 weeks after exposure.
Tumor incidence was much lower in the DBA/2  strain.  Methylcholanthrene-
impregnated  (12%  or 20% methylcholanthrene)  wax pellets implanted in the
urinary bladder of albino mice induced local squamous cell  carcinomas in
approximately 55% of the animals during  25 weeks of observation (partial
autopsy) (Bonser  et al., 1963).
     Systemic effects  of methylcholanthrene  in  mice have been reported
following oral, intravenous, subcutaneous, and  vaginal exposure.  Akamatsu
and Barton (1974) fed, by gavage, 1  mg of 3-methylcholanthrene in olive
oil to five  inbred strains  of mice.   Tumors  were induced at various
sites, including  skin, forestomach,  liver, lung, and lymphatics (Table
5.13).  Treated animals also developed amyloidosis, which was signifi-
cantly correlated with gastric neoplasms.
     Strain  A mice, highly  susceptible to chemical induction of pulmonary
adenomas, developed an increased number  (as  compared with controls) of
lung tumors  4 weeks after intravenous injection of 0.05 to  1.5 mg methyl-
cholanthrene in horse  serum and cholesterol  (Table 5.18)  (Shimkin,
1940).  The  incidence  of tumors was  100% in  3 months.  Subcutaneous
injection of 0.25, 0.5, and 1.0 mg of methylcholanthrene in lard induced
lung tumors  in strain A mice, with 70%,  80%, and 91% incidence developing

-------
           Table  5.18.
Incidence of pulmonary tumors in strain A mice after intravenous
           injection of methylcholanthrene
Dosage
1.5 mg


Time
(weeks)

3
4
5
6
13
20

Number

of
mice

6
7
10
4
5

Number
with

tumors
of the
lungs
1
7
10
4
5

Average
number of

tumors
of the
lungs
1
12
22
55
74


Number

of
mice

9
10
10
10
18
6
0.5 mg
Number
with

tumors
of the
lungs
1
8
10
10
18
6
Average
number of

tumors
of the
lungs
1
5
14
25
30
47

Number

of
mice




7
10
15
0.1 mg
Number
with

tumors
of the
lungs



3
8
15
Average
number of

tumors
of the
lungs



2
4
11
                                                                                                        Ul
     Source:  Adapted from Shimkin, 1940.  Copyright  1940, American Medical Association.
Reprinted with permission of the publisher.

-------
                                   336
by 3 months.  Differences  seen in carcinogenic  potency of  20-methylcho-
lanthrene  (in horse  serum)  and 1,2,5,6-dibenzanthracene (in cholesterol),
when 0.5 mg of  each  compound  was  injected intravenously, disappeared
when 0.5 mg of  each  was  injected  subcutaneously (Tables 5.19 and  5.20).
These  results suggest  that  the subcutaneous  route  is  less  sensitive than
the intravenous route  for  lung tumor induction  in  strain A mice.
   Table 5.19.   Incidence of pulmonary  tumors  in  strain A mice after
    intravenous  injection of 0.5 mg of  methylcholanthrene or 0.5 mg
                          of dibenzanthracene
Methylcholanthrene
Time
(weeks)
3
4
5
6
Number
_ c
or
mice
9
10
10
10
Number
with
tumors of
the lungs
1
8
10
10
Average
number of
tumors of
the lungs
1
5
14
25
Dibenzanthracene
Number
of
mice
10
10
11
10
Number
with
tumors of
the lungs
0
3
10
10
Average
number of
tumors of
the lungs
0
1
3
8
     Source:  Adapted from Shimkin, 1940.  Copyright 1940, American
Medical Association.  Reprinted with permission of the publisher.
    Table 5.20.   Incidence of pulmonary tumors in strain A mice given
         subcutaneously 0.5 mg of methylcholanthrene or 0.5 mg of
  dibenzanthracene dispersed in 0.5 cc of horse serum and cholesterol
Methylcholanthrene
Time

(weeks)
3
4
5
6
Number

mice
10
10
10
10
Number
with

tumors of
the lungs
0
2
5
7
Average
number of

pulmonary
tumors
0
1.0
1.6
2.2
Dibenzanthracene
Number

mice
10
7
6
7
Number
with

tumors of
the lungs
0
0
2
5
Average
number of

pulmonary
tumors
0
0
1.0
2.0
      Source:   Adapted from Shimkin,  1940.   Copyright 1940, American
Medical Association.   Reprinted with permission of the publisher.

-------
                                   337
     Intravaginal exposure of ICR Swiss mice to 20-methylcholanthrene
gave rise to tumors remote from the site of application (Campbell, Yang,
and Bolton, 1965).  During a 6-week period, doses totaling 1.5 to 5.5 mg
of 20-methylcholanthrene were administered, and the mice were observed
for life.  Of 106 female mice, 23% developed lung adenomas (two mice had
adenocarcinomas), 12% developed genital tract cancer,  and genital tract
dysplasia occurred in 27%.  Six of the 24 mice with lung adenomas also
had cancer and/or dysplasia of the vagina and/or cervix uteri.
     5.3.2.3  Aromatic Amines in Mice, Rats, and Rabbits — Bonser et al.
(1952) compared carcinogenesis of 2-naphthylamine and 2-amino-l-naphthol
hydrochloride in several species by several routes of administration.
Implantation of 2-amino-l-naphthol pellets (10% to 15% carcinogen)
induced metaplasia, papilloma, and carcinoma in the bladders of mice,
and subcutaneous injection (5 mg per 100 g of body weight) induced
sarcomas in both mice and rats.  Implantation of 2-naphthylamine pellets
in the bladder and skin painting with the carcinogen failed to produce
bladder tumors in mice, but oral administration (100 to 300 mg/kg weekly)
induced hepatomas in mice and benign bladder tumors in rats and rabbits
(Table 5.21).
     Inhalation of benzidine causes bladder cancer in man (Goldwater,
Rosso, and Kleinfeld, 1965) and has been shown to be carcinogenic in
certain animal species.  Rats, after inhaling benzidine for 13 months,
developed leukemia, fibroadenomas, carcinoma of the mammary glands,
carcinoma of the male mammary gland, and hepatoma, but no bladder tumors
(Zabezhinskii, 1970).  Oral administration of benzidine to rats induced
hepatomas and rectal and acoustic duct carcinomas  (Spitz, Maguigan,  and
Dobriner, 1950), male mammary gland carcinomas, and leukemia  (see
National Institutes of Health, 1968-1973).  The absence of tumors of the
urinary bladder of rats receiving benzidine by inhalation and by subcu-
taneous and oral administration led Zabezhinskii to conclude  that the
character of action of aromatic amines is dependent mainly on the species
of the experimental animal and not the mode of administration of  the
substance.

-------
Table 5.21.  Tumor Induction with aromatic amines by various routes of exposure
Chemical/species
2-Amino-l-naphthol
hydrochlorlde
Mouse


Mouse, "stock"
Mouse, R III

Rat, albino
2-Naphthylamine
Mouse
Mouse, IF
Mouse, CBA
Rat, albino
Mouse, IF
Rabbit
Route of
exposure

Bladder
pellet


Subcutaneous
Subcutaneous

Subcutaneous

Bladder
pellets
Gavage
Gavage
Diet
Diet (low
protein)
Skin painting
Spoon feeding
Dose

1-2 mg per pellet


5 mg per 100 g
body weight
every 2 weeks
5 mg per 100 g
body weight
every 2 weeks

5 mg per 100 g
body weight
every 2 weeks

1-2 mg per pellet
5 mg every 2 weeks
in arachis oil
120 mg/kg body
weight twice
per week
160 mg/kg body
weight per week
310 mg/kg body
weight per week
Saturated solution
100 mg/kg body
weight per week
Duration of
exposure
(weeks)

22 to 28
30 to 39


Up to 80
Up to 80

Up to 80

39
Up to 72
Up to 89
Continuous
>90
Up to 99
Up to about
275
Tumor incidence

0/6 bladder tumor
1/8 bladder
adenoma
5/8 bladder
carcinoma
2/15 subcutaneous
tumors
3/15 leukemias
A/15 hepatomas
1/8 subcutaneous
tumors
1/8 leukemia
(hepatoma and
leukemia in
controls)
5/14 subcutaneous
tumors

0/8 bladder tumor
Cholangioma of
bile duct
13/23 hepatomas
11/26 hepatomas
3/15 bladder
papillomas
0/25 skin tumor
0/25 hepatoma
1/6 bladder
papilloma
Tumor
latency Reference
(weeks)

Bonser et al.,
About 30 1952


Bonser et al. ,
1952
Bonser et al. ,
1952
OJ
00
Bonser et al. ,
1952

Bonser et al. ,
1952
Bonser et al. ,
1952
Bonser et al . ,
1952
About 60 Bonser et al. ,
1952
About 60 Bonser et al.,
1952
Bonser et al . ,
1952
Bonser et al. ,
1952

-------
                                                   Table  5.21  (continued)
Chemical/species
Route of
exposure
Duration of
Dose exposure Tumor incidence
(weeks)
Tumor
latency
(weeks)
Reference
Benzidine

  Rat, noninbred
Benzidine,
  technical
  Rat
Inhalation      4 hours/day, 5         80
                  days/week
Subcutaneous    15 mg/week
Life
Benzidine, pure

  Rat
Benzidine sulfate

  Rat
Subcutaneous    15 mg/week
Life
Subcutaneous    15 mg/week
                       Oral
                15 mg/week
Life
                                                              Life
               5/28 leukemias
               1/28 hepatoma
               2/28 mammary
                 tumors
8/233 hepatomas
0/233 bladder
  tumors
7/385 gastric
  tumors
54/233 auditory
  canal tumors
6/152 hepatomas
0/152 bladder
  tumors
32/152 auditory
  canal tumors
1/153 hepatomas
0/153 bladder
  tumors
16/153 auditory
  canal tumors
1/37 hepatomas
0/37 bladder
  tumors
2/37 auditory
  canal tumors
                     52
Zabizhinakii,
  1970
                                                Spitz, Maguigan,
                                                  and Dobriner,
                                                  1950
                                                Spitz, Maguigan,
                                                  and Dobriner,
                                                  1950
                                                Spitz, Maguigan,
                                                  and Dobriner,
                                                  1950
                                                Spitz, Maguigan,
                                                  and Dobriner,
                                                  1950
                                                                                                                  so

-------
                                   340
 5.3.3  Conclusions
      It is recommended that the route of exposure used in animal studies
 closely resemble the means by which man would encounter the chemical in
 the environment.  Nevertheless, it is sometimes necessary to substitute
 other routes.   Because of the variables inherent in any animal test
 system — metabolic, biochemical, species, and strain differences, for
 example — route-to-route extrapolations should be made with extreme
 caution.  Experiments designed specifically to define the role of route
 of administration of chemicals to animals in carcinogenesis studies are
 not easily found in the literature.   However, it is obvious that route
 of administration is a major modifying factor that can determine site,
 histological type, and incidence of tumors.

 5.4  DOSE AND DURATION

      Dose selection is a critical aspect of chronic toxicity and carcino-
 genicity testing.  If the dose level is set too low, no effects will be
 observed; if it is set too high, excess mortality rates will reduce the
 test population too low for adequate statistical analysis.  Consequently,
 dose rates for long-term testing should be set only after data from
 acute and subchronic tests have been analyzed.  When possible, pharmaco-
 kinetic data should also be considered (Golberg, 1975; Munro,  1977).
 Massive doses  that result in nonlinear pharmacokinetics should be avoided
 unless such data reflect actual conditions of human exposure (Watanabe,
 Young, and Gehring, 1977).
      In early  chronic toxicity testing, the primary purpose was the
 demonstration  of no-observed-effect  level; more recently, data needed
 for dose-response analysis have also been sought (Barnes and Denz, 1954;
 Federal Register, 1979;  National Academy of Sciences, 1977; Winstead,
 1978).   For  the  establishment  of a no-observed-effect level, at  least
 three  dose rates  are  required.   The  highest  level  should  produce some
 signs  of  toxicity without seriously  altering normal  physiological func-
 tion or causing excessive lethality  during the course of  the experiment.
The lowest level  should be a fraction of  the high  dose that is not
expected  to produce evidence of  toxicity.  The remaining  dose  should be

-------
                                  341
set at a level intermediate between the high and low dose  (World Health
Organization, 1978).  Dose levels should also be influenced by the
anticipated level of human exposure as well as the margin  of safety that
may be desired in setting subsequent exposure standards  (National Academy
of Sciences, 1977).  If the determination of a dose-response curve is an
objective of the chronic toxicity test, more than three dose levels may
be required (Federal Register, 1979; Food Safety Council,  1978).
     Although a test for carcinogenic potential might occasionally
entail only a single well-selected dose, tests for risk estimation or
determination of a no-effect level usually require several dose levels
(Page, 1977a).  If more than one dose is used, the highest level should
be within the toxic range but should be consistent with prolonged survival
of the majority of the animals.  The lowest dose selected  ideally should
produce no real increase in tumor incidence over controls  (Ministry of
Health and Welfare Canada, 1975).  The recommendations of  several agencies
regarding the controversial subject of dose selection in carcinogenicity
studies are presented in Table 5.22.
     Weisburger (1976) emphasized the selection of the maximum tolerated
dose (dose at which mortality is low in an 8-week test and which may
depress weight gain 5% to 20%, with an optimal depression  of approxi-
mately 10%).  The rationale given by Munro (1977) for use  of maximum
tolerated dose (MTD) includes:   (1)  when weak carcinogens are tested,
high doses for long periods of time increase chances of  seeing statisti-
cally increased numbers of tumors within the life span of  the animals
(since cancer induction in animals is both time- and dose-related); and
(2) increased confidence can be  placed in negative data  because  the
chemical has been tested at the  maximum dose level compatible with
normal or near normal survival rates.  Munro also points out several
problems associated with the use of MTD:  (1) the fact that MTD may
result in a 10% weight loss is considered to be inappropriate because it
may affect biochemical or physiological processes that make the animal
more or less susceptible to carcinogenic challenge; (2) metabolic over-
loading may result in changes in normal handling of the  test compound;
(3) simultaneously induced toxicity due to the test compound may enhance
or diminish its carcinogenic activity, and further use of MTD favors

-------
                                    342
        Table 5.22.   Summary of recommendations  on dose selection
            Agency
            Recommendation
Health Protection Branch,
  Department of National
  Health and Welfare
  Canada, 1973
International Union Against
  Cancer, 1969
National Cancer Institute,
  1976
U.S. Food and Drug   }
  Administration, 1971
"The highest dose level within  the toxic
  range but consistent with prolonged
  survival.  The lower levels should
  permit .  . .  good health .  .  . until
  tumors develop."

"All substances submitted for carcino-
  genicity testing should be examined
  at 3 or more dose levels."

"The MTDa is defined as the highest dose
  of the test agent given during the
  chronic study that can be predicted
  not to alter the animals' normal lon-
  gevity from effects other than car-
  cinogenicity.  The MTD should be the
  highest dose that causes no more than
  10% weight decrement, as compared to
  the appropriate control groups and
  does not produce mortality,  clinical
  signs of toxicity, or pathologic le-
  sions (other than those that  may be
  related to a neoplastic response) that
  would be predicted to shorten the ani-
  mal's natural life span.  Although de-
  pressed weight gain may be a  clinical
  sign of toxicity, it is acceptable
  when estimating the MTD.  Since  the
  data may not always be easily inter-
  pretable, a degree of judgment is
  often necessary in estimating the
  MTD."

"At several dose levels — one likely to
  yield maximum tumor incidence."
      nfD — Maximum tolerated dose.

     Source:  Adapted from Munro, 1977.  Reprinted with permission of the
publisher.
very  toxic compounds that can be  used only at lower dose levels,  thus

reducing  the chance of tumor formation;  (4) there is difficulty  in

handling  data when the criteria for MTD  are not met; and (5) MTD doses

usually far exceed human exposure levels.

      In tests for carcinogenicity a dose-response relationship should be

established if at all possible, but,  because of the number of animals

-------
                                  343
required, this is highly impractical with doses that produce lower tumor
incidences (Terracini, Magee, and Barnes, 1967).  Also, the limited life
span of the animals may frustrate attempts to establish a low dose
response.  The simple concept of additive effects of repeated doses is
not applicable to every situation.  Page (19772?) suggested that the
effectiveness of increasing exposure might decrease on a per dose basis
for some chemicals especially if the body's ability to retain the chemical
becomes saturated.  This is illustrated by the study of Schepers (1971),
who compared the effects of beryllium oxide in rats after:  (1) continuous
inhalation exposure up to 12 months, (2) continuous exposure for 6
months and (3) brief exposure for 1 month to high concentrations of the
chemical.  The rate of tumor formation was enhanced in animals of the
interrupted (6-months) exposure group, whereas those exposed continuously
for a long period and those exposed continuously for a shorter period
(but to a higher concentration) developed tumors at about the same slow
rate.  Schepers postulated that the effect might be attributed to saturation
of the lung with beryllium oxide.
     Carcinogens are usually administered every day (to simulate human
exposure) over the life span of the animal; some experiments may be
terminated when mice are 18 months old and rats are 24 months old (Ministry
of Health and Welfare Canada, 1975).  Termination of studies must be
carried out with caution since some tumors tend to develop late in the
life of the animal.  For example, Nelson, Fitzhugh, and Calvery (1943)
reported that rats did not begin to develop liver tumors until 18 months
after they had been placed on a diet containing selenium.  The tumors in
11 of 53 animals arose in cirrhotic livers, while control animals had a
spontaneous hepatic tumor incidence of less than 1%.
     In carcinogenicity testing the route of administration often deter-
mines the frequency of exposure necessary for tumor development.  Weisburger
and Weisburger (1967) pointed out that for cutaneous exposure  to strong
carcinogens a weekly or even a single treatment is satisfactory.  More
frequent administration of weak carcinogens is  recommended.   Intraperi-
toneal injections require a repeated  schedule because  of  the  higher
absorption rate of the internal organs, while intravenous administration
allows no more than approximately eight  injections.

-------
                                    344
      In chronic toxicity evaluations the test compound should also be
 administered 7 days per week (Benitz, 1970).   Treatment 5 days a week
 results in a 28.6% decrease of exposure and a 48-h weekly recovery
 period.  The administration of the equivalent of seven doses during 5
 days as recommended by some workers (Boyd, 1968) does not constitute a
 treatment equivalent to daily dosing and may not produce identical
 results (Baker and Alcock, 1965,  as cited in Benitz,  1970; Hayes, 1967).
      Apart from dose rate, length of exposure is the  principal character-
 istic that distinguishes chronic  tests  from other forms of toxicity
 testing.   Most investigators agree that chronic testing should extend
 over a large portion of the test  animal's life span (Boyd, 1968; Food
 Safety Council, 1978; National Academy  of Sciences, 1975, 1977; World
 Health Organization, 1978); however, there is no consensus concerning
 the length of this period.  Conceptually, the duration of a chronic
 toxicity test should reflect the  time for the test material, or its
 active metabolites, to accumulate to toxic levels in  the organism, plus
 the time required for the target  organ  or system to respond once the
 toxic concentration is achieved (Lawrence, 1976).  Generally speaking,
 most effects of chronic testing become  apparent within 3 or 4 months.
 For example, in one study of the  chronic effects of 11 drugs on rats,
 dogs,  and monkeys, all compounds  produced an observable effect within
 the first 2  weeks, and only one compound caused an additional effect
 after  the first 3 months (Peck, 1968).   Similarly,  in a chronic study of
 46  compounds,  the Ciba Drug Company noted all indications of toxicity
 within the first  8 weeks of the test (Bein,  1963, as  cited in McNamara,
 1976).   In this connection, McNamara (1976)  stated, "If no effect occurs
 in  3 months,  there is a low likelihood  that  any effect will occur on
 continued dosing  for 1 year."   Barnes and Denz (1954)  asserted that
 little  useful  information can  be  extracted from chronic toxicity tests
 after  the first 3 to 6 months.  Because of this characteristically early
 detection of most  chronic effects,  many investigators  feel that the
 duration  of  chronic  tests need  not  exceed 6 months  (Barnes and Denz,
 1954; McCollister,  1974,  as cited  in McNamara,  1976;  Paget,  1963,  as
 cited in  McNaraara,  1976;  Weil and McCollister,  1963;  Zbinden,  1973).
However,  this  conclusion  appears  to have gained wider  acceptance among
investigators  dealing with  drugs, to which humans may  be exposed for

-------
                                   345
relatively brief intervals, than with investigators dealing with occupa-
tional and environmental pollutants, to which the population may be
exposed for intervals approximating a lifetime.  For assessing the
effects to humans of lifetime exposures of toxic compounds, some scien-
tists insist that lifetime or near-lifetime exposures of laboratory test
animals are required (Food Safety Council, 1978; Loomis, 1974; National
Academy of Sciences, 1975; World Health Organization, 1978).  Unlike the
shorter acute or subchronic tests, lifetime exposures are said to reveal
toxic effects associated with (1) repetitive exposures to a test material
and  (2) changes occurring during the aging process, such as altered
tissue sensitivity, changing metabolic and physiological capability, and
spontaneous disease (World Health Organization, 1978).  The logic of the
latter argument appears unassailable; however, examples illustrating its
validity do not abound in the literature.
     Only occasionally are chronic effects reported that were not observed
during the first 90 days of treatment (Page, 1977Z?) .  Among more than
100  computer-selected publications on chronic toxicity examined during
the  preparation of this chapter (see Bibliography), only two such papers
were noted:  (1) Rosenkrantz et al. (1975) reported observing hyperglycemia
in rats only after 180 days of treatment with A9-tetrahydrocannabinol,
and (2)  Verschuuren,  Kroes,  and Van Esch (1973)  detected stridor in rats
fed tetrasul only after 4 months of treatment.   In a similar examination
of the literature,  McNamara (1976)  considered 82 long-term toxicity
studies involving 122 test materials and 566 dose levels.   Only three
                f
compounds and 15 dose levels produced toxic signs after, but not before,
3 months exposure.   It thus appears that in most instances all significant
toxic effects are indeed observed within 3 to 4 months, but that excep-
tions occasionally occur.  Because it is not possible to determine a
priori the relative significance of the occasional chronic effect missed
in short-term tests,  confidence in the validity of toxicity test results
can be maximized only by performing life span exposures on test animals,
even though longer tests are much more expensive and will provide no
additional useful information most of the time.  Justification of the
test duration thus becomes a discretionary decision that must be resolved
on the basis of risk-benefit analysis.

-------
                                   346
      When lifetime or near-lifetime exposures of test animals are required
 in chronic toxicity tests, the actual duration of exposure will naturally
 vary with test animal.  Many strains of mice or rats will survive 30
 months or more with good care; consequently, these animals are usually
 exposed for 24 to 27 months, beginning at weaning (Food Safety Council,
 1978; National Academy of Sciences, 1977).  The normal life spans of
 other common test animals are considerably greater than that indicated
 for mice and rats.  Table 5.23 shows that chronic tests of even 2 years
 duration represent only a small fraction of the life span of most nonrodent
 laboratory test animals.  Needless to say, these animals are only rarely
 chosen for life span exposures.  Among the publications examined during
 the preparation of this chapter, no life span exposures of nonrodents
 were found, but one 7-year dog study and one 43-month monkey study were
 noted.
      When a long-lived animal is chosen as a second species to mice or
 rats in chronic toxicity tests, exposures amounting to at least 10% of
 the projected life span of the animal are generally recommended (National
 Academy of Sciences, 1977; World Health Organization, 1978).  On this
 basis, dogs used as a second species in chronic toxicity tests should
 usually be exposed for at least 1 year.  However, several investigators
 have  reported previously unobserved toxic effects in dogs after periods
 of  exposure greater than 1 year (Braun et al.,  1977;  Kaplan and Sherman,
 1977;  Weil et al.,  1971).   Consequently,  some  authorities recommend
 exposing  dogs longer,  for periods  of 2 to 7  years (Goldenthal,  1968;
 Loomis, 1974;  National Academy of  Sciences,  1977).   In the chronic
 toxicity  papers  analyzed in this study the typical duration of  exposure
 of  dogs used as  a  second species was 2 years.
      In conclusion,  dose selection is a critical aspect of long-term
 testing.   Dose rates should be set only after  acute and subchronic test
 data have  been analyzed and pharmacokinetic  effects have been considered.
At  least  three dose rates are  required to establish a no-observed-effect
level.  The  highest  level should produce  some  signs of toxicity without
seriously  altering normal physiological function or causing excessive
lethality  during the course of the experiment.   The lowest level should
be a fraction  of the high dose that  is not expected to produce  evidence
of toxicity.   The remaining dose should be set  at an  intermediate level

-------
          Table 5.23.  Time relationships  among  drug  exposure,  life  span,  and  time  equivalents  in man

of study
(months)

1
2
3
6 •?.•
12 -
24

Life
span
(%)
4.1
8.2
12
25
49
99
Rat
Human
equivalent
(months)
34
67
101
202
404
808

Life
span
(%)
1.5
3.0
4.5
9.0
18
36
Rabbit
Human
equivalent
(months)
12
24
36
72
145
289

Life
span
(%)
0.82
1.6
2.5
4.9
9.8
20
Dog
Human
equivalent
(months)
6.5
14
20
40
81
162

Life
span
(%)
0.82
1.6
2.5
4.9
9.8
20
Pig
Human
equivalent
(months)
6.5
14
20
40
81
162
Monkey
Life
span
(%)
0.55
1.1
1.6
3.3
6.6
13
Human
equivalent
(months)
4.5
9
13
27
53
107
Source:  Adapted from Benitz, 1970.
                                                                                                                       to

-------
                                    348
 between the high and low doses.  Dose levels should also reflect antici-
 pated human exposures as well as the margin of safety that may be desired
 in setting subsequent exposure standards.  If a dose-response relation-
 ship is a goal of the test, then more than three dose levels may be
 required.
      The test compound should be administered 7 days per week; treatment
 5 days a week or administration of the equivalent of seven doses during
 5 days is not equivalent to daily dosing and may not produce identical
 results.  In carcinogenicity studies, the exposure pattern is often
 modified by the route of exposure.
      Most authorities agree that chronic toxicity and carcinogenicity
 studies should extend over a large portion of the test animal's life
 span.  However, there is no consensus concerning the length of this
 period.  Conceptually, the duration of a chronic toxicity or carcino-
 genicity test should reflect the time for the test material, or its
 metabolites, to reach toxic concentrations in the test animal, plus the
 time required for the target organ or system to respond once the toxic
 concentration is achieved.  Generally speaking, most effects of non-
 carcinogenic chronic testing become apparent within 3 or 4 months, and
 many  authorities  feel  there is  only a  low likelihood  that  additional
 effects will be observed  after  6 months  of  treatment.  Nevertheless,  a
 few new effects are  occasionally seen  during  longer periods  of  treatment,
 and in  carcinogenicity studies  the  chance for late development  of  long-
 term  effects is even greater.   Because it is  not  possible  to  determine a
 priori  the  relative  significance of the  occasional chronic effect  missed
 in short-term  tests, confidence in  the validity of toxicity  test results
 can be maximized  only  by  performing life span exposures  on test animals,
 even  though the longer tests are much  more  expensive  and will provide no
 additional useful  information most  of  the time.
     When lifetime or  near-lifetime exposures of  test animals are  required
 in chronic toxic and carcinogenicity tests, the actual duration of
 exposure naturally varies according to species.   The  life  spans of mice
 and rats are about 30  months, and they are  usually exposed for  24  to  27
months, beginning at weaning.   The  nominal  life spans of nonrodents are
much longer — for example,  rabbit,  66 months;  dog or  pig,  120 months;
and monkey, 184 months.  Consequently, these  animals  are rarely subjected

-------
                                  349
to life span exposures.  However, a few literature examples have been
noted of 7-year exposures for dogs.
     When long-lived animals are chosen as second species to mice or
rats in noncarcinogenic chronic toxicity studies, exposures of at least
10% of the projected life span of the animal are usually recommended.
On this basis, dogs used as a second species in chronic toxicity tests
should be exposed for at least a year.   In the noncarcinogenic chronic
toxicity publications analyzed in this study the typical exposure of
dogs used as a second species was 2 years.

5.5  INTERIM SACRIFICE

     Interim sacrifices are generally considered useful in the study of
long-term toxic effects, pathogenesis, and reversible changes.  Timely
sampling may also provide valuable insights into specific clinical chem-
istry or organ function tests that may be required (Food Safety Council,
1978; National Academy of Sciences, 1977).  For these reasons, some
authorities recommend the inclusion of serial sacrifices in chronic
toxicity tests (Fitzhugh, 1955, as cited in National Academy of Sciences,
1975; National Academy of Sciences, 1975; World Health Organization,
1978).  However, other workers consider interim sacrifices a needless
reduction in an otherwise limited number of test and control animals
and recommend that interim sacrifices be avoided in chronic toxicity
tests (Benitz, 1970).  Obviously, extra animals could be taken initially
to compensate for losses in size of the test and control groups through
interim sacrifices.  Frequently, however, similar information is available
from previously performed interim sacrifices during subchronic tests or
from earlier pharmacodynamic studies.  The need  for interim sacrifices
in long-term studies thus varies from study to study and should be
reassessed with each new study.
     In actual practice, few investigators appear to make  extensive  use
of interim sacrifices during chronic toxicity studies.  Among more  than
100 chronic toxicity publications examined during the  preparation of
this chapter, only 12 reported interim sacrifices, and only eight studies
involved the examination of several animals during several intervals of
the test period.  Significant observations derived specifically from
interim sacrifices were cited in only four of  these  reports.

-------
                                   350
     In conclusion, the use of interim sacrifices in chronic toxicity
testing is controversial.  Some authorities recommend the practice as a
valuable aid in the study of toxic effects, clinical chemistry, organ
function tests, and pathogenesis.  Other experts acknowledge the value
of interim sacrifices for the purposes stated, but consider the practice
a needless waste of the relatively limited number of test and control
animals typically available in chronic toxicity tests.  Much of the
information provided by interim sacrifices in chronic toxicity tests can
be provided by previously performed subchronic toxicity and pharma-
codynamic studies.  In actual practice, few workers appear to make
extensive use of interim sacrifices during chronic toxicity studies.  In
the literature sample represented by  the bibliography for this section
interim sacrifices were reported in only 12 papers.  Of these, only eight
studies involved the examination of several animals during several
intervals of the test period, and significant observations based on
interim sacrifices were reported in only four papers.  It thus appears
that interim sacrifices are not a generally used, or needed, feature of
chronic toxicity tests.

5.6  DATA COLLECTION AND EVALUATION

     Long-term tests are unique research experiments that are only
rarely amenable to standardized protocols  (Food Safety Council, 1978).
Nevertheless, the success of a well-planned chronic toxicity or carcino-
genicity study strongly depends on adequate observations of the animal
during the test period and on the efficacy of the ensuing pathological
examinations.  The following subsections address important aspects of
clinical and pathological procedures  essential to the successful execution
of long-term tests.

5.6.1  Food Consumption and Body Weight
     Growth and body weights are important indicators of adverse effects
of the test material, but these data  are not uniformly collected by some
investigators.  All animals should be weighed weekly during periods of
rapid growth and at least monthly thereafter (Benitz, 1970; Food Safety
Council 1978; Loomis, 1974; Magee, 1970; National Academy of Sciences,

-------
                                  351
1977; Sontag, Page, and Saffiotti,  1976;  World Health Organization,
1978).  Food consumption should also be monitored weekly to observe the
effect of the test material on food intake and to provide an accurate
determination of dose when the test material is administered in the
diet.  When the test compound is administered in drinking water,  the
weekly consumption rate of this material is also required for dose
determinations.

5.6.2  Clinical and Laboratory Examinations
     Careful clinical observation of test and control animals appears to
be the most neglected area in experimental toxicology (Food Safety
Council, 1978; World Health Organization, 1978).  To remedy this situation
and minimize the effects of extraneous factors on the course of chronic
toxicity and carcinogenicity experiments, several authorities recommend
that qualified employees observe the general physical condition,  viability,
and adverse behavioral changes of all test and control animals at least
twice daily, 7 days a week (Arnold et al., 1977; Food Safety Council,
1978; Fox, 1977; National Academy of Sciences, 1977; World Health Organi-
zation, 1978).  Animals with obvious life-threatening conditions should
be isolated; those unlikely to survive an additional day should be
sacrificed and necropsied to preserve tissues for histological examination.
Each animal should be completely examined at least once a week by quali-
fied personnel for unusual behavioral patterns, respiratory signs,
bleeding, and palpable masses, as well as for abnormalities of the coat,
eyes, mouth, teeth, nose, and ears (Arnold et al., 1977; National
Academy of Sciences, 1977).  In chronic toxicity  studies, appropriate
biochemical tests are also essential for detecting and assessing  toxic
effects at the clinical level, but no consensus exists regarding  the
extent or frequency of their use.  Loomis  (1974)  recommended  clinical
blood chemistry tests, urinalysis, and blood  cell counts  for  all  animals
at 6- to 12-week intervals, but stated "Routine special  types of  bio-
chemical analyses of sample material, such  as blood  or urine,  from
apparently healthy animals probably are  indicated only when there is
reason to suspect that the chemical under  investigation  is capable of
producing specific toxic effects for which biochemical  methods are
clinically of  diagnostic value."   A  report of the National Academy of

-------
                                   352
 Sciences (1977)  suggests clinical chemistry tests "should be judiciously
 selected,"  based on preliminary or short-term toxicity tests,  or on  the
 known chemical characteristics of the test compound.   The Academy report
 further states that a rigid sampling schedule is impractical and excess-
 ively costly.   Other workers recommend "periodic clinical examinations"
 to  identify and monitor the progression of specific abnormal findings
 (Food Safety Council, 1978).  There appears to be general agreement,
 however, that clinical blood chemistry tests, blood cell counts, and
 urinalysis  tests should be performed on all animals that become ill  or
 show effects from exposure to the test material.   Arnold et  al.  (1977),
 Fox (1977), and Street (1970) provide lengthy discussions of these
 tests,  as well as extensive bibliographies of additional papers.
      Less unanimity exists among investigators of chronic toxicity rela-
 tive to prescheduled organ function  tests.   The minimum recommendation
 of  the  Food Safety Council (1978)  includes tests for  liver,  kidney,  and
 bone marrow function.   The World Health Organization  (1978)  also recom-
 mends the use  of organ function tests but  concedes that "In  general,
 these methods  are not sufficiently standardized or reproducible  to
 detect  minor alterations in organ function."  Benitz  (1970)  and  the
 National Academy of Sciences (1977)  questioned the benefits  of presched-
 uled organ  function tests because of their low sensitivity to organ
 damage.   Instead of organ function tests,  the latter  workers recommended
 exposing extra animals that can be later sacrificed for complete gross
 and  histopathological examinations.   Other workers have also expressed
 reservations about the use of organ  function tests (Peck,  1968).   When
 organ function tests are included in chronic toxicity experiments, they
 are  most  often limited to liver,  kidney, and thyroid  studies in  larger
 animals  (National Academy of Sciences,  1977).

 5.6.3   Pathological Examinations
      Until  recently,  generally  accepted procedures in long-term  studies
 required  the necropsy  of  all nonrodent  test  animals,  but  only a  fraction
 of the  total number  of rodents  (Benitz,  1970;  Zbinden,  1973).  Now, how-
ever, there appears  to be general  agreement  among investigators  that all
animals that die during  the  study, or are  subsequently  killed, should  be
carefully necropsied  (Arnold et  al., 1977; Food  Safety  Council,  1978;

-------
                                  353

National Academy of Sciences, 1977;  Sontag,  Page,  and Saffiotti,  1976;
World Health Organization, 1978).  Furthermore,  current thinking  stresses
the importance of adequate gross examination of  tissues prior to  histo-
logic sampling.  In many instances,  pertinent information missed  during
the macroscopic examinations of the test animal  cannot be recovered in
subsequent microscopic examinations of prepared  tissues,  even if  the
utmost care is used (Benitz, 1970).   Consequently, the necropsy procedure
should be considered one of the most important steps in the experimental
protocol (Food Safety Council, 1978).
     There is general agreement in the current literature that samples
of all major organs and tissues should be taken  from all  test and control
animals for macroscopic examination and fixation (Benitz, 1970; Food
Safety Council, 1978;  Loomis, 1974;  National Academy of Sciences, 1977).
No consensus exists, however, concerning which tissues should be  examined
microscopically.  Early workers recommended  microscopic examination of
all obviously altered tissues plus an additional quantity that generally
did not exceed 20 (Abrams, Zbinden,  and Bagdon,  1965; Barnes and  Denz,
1954).  Other authorities left the microscopic observation of the sup-
plementary tissues to the discretion of the  pathologist in charge (Magee,
1970) or specified "complete and accurate" pathological examination
without indicating more specific requirements (U.S. Food and Drug Adminis-
tration, 1971).  More recent guidelines require microscopic examination
of all major tissues and gross lesions in all high dose and control
animals, as well as grossly altered tissues  in all other dose groups
(Food Safety Council, 1978; National Academy of Sciences, 1977; Peck,
1974; World Health Organization, 1978).  The National Cancer Institute
guidelines for chronic toxicity tests are even more stringent:  they
require microscopic examination of approximately 40 tissues from all
test and control animals, except positive controls which may be exempted
from this requirement  (Sontag, Page, and Saffiotti, 1976).  Obviously,
careful microscopic examination of all tissues is highly desirable  from
the experimental point of view; however, the Incremental increase in
information gained through such a practice must be balanced against the
added economic, temporal, and labor costs incurred by  such a  requirement.
According to recent estimates, the number of trained pathologists avail-
able to perform histologic examinations  of  tissues  from  tests required

-------
                                   354

or expected under the Toxic Substances Control Act and other expanded
government programs is insufficient if microscopic examinations of 40 or
more tissues from all test and control animals are required (Page,
19772?).
     In carcinogenicity testing, special emphasis is placed on the
criteria used to classify the lesions found in microscopic examination
(World Health Organization, 1978).  The investigator should specify how
the classification was done and what is implied by the terms benign,
malignant, neoplastio, preneoplastic, and hyperneoplastio.  Also, as
discussed before, the pathologist  should be aware of the type and location
of the spontaneous  tumors associated with the animal species/strain used
(Magee, 1970).  The value of proper pathology is especially great in
carcinogenicity testing, since other evaluations are less frequently
incorporated  in the test design  (Page, 1977
-------
                                  355
Each animal should be examined at least weekly for unusual behavioral
patterns, respiratory signs, excretory products, and palpable masses, as
well as other abnormalities.  In noncarcinogenic chronic tests there is
general agreement that animals showing obvious signs of illness should
receive blood chemistry and urinalysis tests, but no consensus exists
concerning routine tests for healthy animals.  Even less unanimity
exists relative to prescheduled organ function tests, which many authori-
ties regard as insufficiently standardized and reproducible to detect
minor alterations in organ function.   When performed, these tests are
generally limited to the kidney and liver function of larger,  nonrodent
test animals.
     There is general agreement among experts that samples of all major
organs and tissues should be taken from all test and control animals for
macroscopic examination and fixation, but no consensus exists concerning
which tissues should be examined microscopically.  Obviously, careful
microscopic examination of all sample tissues is highly desirable from
the experimental point of view, but,  according to recent estimates,
there are too few trained pathologists to perform histologic examinations
of tissues from tests required or expected under the Toxic Substances
Control Act and other expanded government programs if microscopic exami-
nations of 40 or more tissues from all test and control animals are
required.  It thus appears that, at least for the next few years, limita-
tions must be placed on (1) the number of long-term tests performed, if
total histologic tissue analysis of all animal samples is required,  (2)
the number of animals to be examined histologically in each long-term
test, or (3) the number of tissues per animal to be examined histologically.
In the literature analyzed in this study, most workers performed histo-
logical examinations of "all major organs and tissues" of high dose  and
control animals, plus any other tissues exhibiting gross lesions.  In
most thorough studies, "all major organs and tissues" correspond to
approximately 30 histological samples.  The analysis of this number  of
samples from the high dose and control groups is  recommended as  the
minimum acceptable level for histological sampling at the  present  time.

-------
                                   356
 5.7   SHORT-TERM TESTS FOR CARCINOGENICITY

      Because of time and cost  requirements,  application of  the  lifetime
 rodent  bioassay to the screening of  all potential  carcinogens is  practi-
 cally impossible.   Short-term  in vitro screening procedures have  been
 developed that provide limited information concerning the potential  car-
 cinogenicity of chemicals,  under reasonable  time and cost constraints.
 Such tests,  designed to detect genotoxic activity  or morphological
 transformation, include the following:   the  Ames Salmonella/microsome
 assay;  the polk+lpolk.- assay (differential toxicity in E. ooli);  "Rec"
 differential toxicity in Bacillus subtilis;  mouse  lymphoma  assay  (mutation
 in cultured  mouse  cells);  and  assays for unscheduled DMA synthesis,  in
 vitro cell transformation,  sex-linked recessive lethal mutation in
 Drosophila,  sister chromatid exchange,  and in vitro chromosome  aberra-
  *-.
 tions.   These assays are being examined closely in the GENE-TOX workshop
 series  (sponsored  by the U.S.  Environmental  Protection Agency)  and
 therefore will not be included in this  discussion  of short-term tests
 for  carcinogenicity.   For evaluation of advantages and limitations of
 the  assays mentioned above,  see Brusick (1978),  Bridges (1976), and
 Stoltz  et al.  (1974).
      The discussion of short-term tests will be limited to  brief  descrip-
 tions of in  vivo (or in vivo-in vitro)  protocols which allow for  meta-
 bolic activation of chemical carcinogens in  the host animal.  These
 tests are designed to shorten  tumor  induction time or to detect cellular
 alterations  or markers that  appear shortly after exposure to carcinogens.

 5.7.1  Embryo  Homograft
      A  promising technique  for producing tumors with chemicals  in a
 relatively short time was  introduced by Peacock in 1962 and was discussed
 in more detail by  Peacock and  Dick (1963).   Using  BALB/c mice,  tissues
 from  various organs of embryos were  implanted surgically into the thigh
muscle  of  adults of the same strain.  Skin,  lung,  stomach,  and  urinary
bladder  yielded 100%  growing implants.   Kidney,  adrenal,  thymus,  and
spleen  implants were  sometimes successful, but  liver and brain  tissues
failed  to  grow.  When  small  quantities  of  each  of  13 solid  polycyclic
hydrocarbons were  introduced along with the  tissue,  malignant tumors

-------
                                   357

formed in the Implants.  The quantity of carcinogen required to induce
tumors was estimated to be no more than 150 pg, and the tumors induced
were mainly squamous cell carcinomas  (with some sarcomas and lung ade-
nomas) ; however, the incidence and types of embryo tissue affected
varied with each hydrocarbon.  Only those hydrocarbons previously reported
to be carcinogens (by any method) produced tumors during the time of
observation.  The results were obtained in 16 weeks.  The surgical
technique used for implantation introduced the problem of possible
effect of the carcinogen on the healing process, but Davies, Major, and
Aberdeen (1971), using a nonsurgical implantation technique, were able
to induce adenomas in embryonic lung tissues exposed to 26 yg of 3,4-
benzo(a)pyrene (BAP) or 1,2,5,6-dibenzanthracene.  Twenty percent of the
implants exposed to benzo(a)pyrene and 62% of the implants exposed to
dimethylbenzanthracene developed adenomas in 16 weeks.  The technique
lacks quantitation, but that disadvantage can perhaps be eliminated by
in vitro exposure of organ cultures to accurately measured amounts of
carcinogen.  For example, lung explants exposed to 20-methylcholanthrene,
3,4-benzo(a)pyrene, or 1,2,5,6-dibenzanthracene and implanted subcutane-
ously into a host animal developed adenomas (some as early as 3 months
later), as demonstrated by Laws and Flaks (1966) and Davies, Major, and
Aberdeen (1970).

5.7.2  jSite Transfer
     Homburger and Baker (1967) described studies in which mice and
hamsters received subcutaneous injections of 25 y an^ 500 y> respectively,
of 3,4,9,10-dibenzopyrene.  At various times after injection, the  injec-
tion sites were removed.  Four sites were pooled, minced, and injected
subcutaneously into recipients of the same age, sex, and strain.   The
test animals were checked weekly for  tumor formation.  Control mice were
injected with a carcinogen, injection sites being left intact, and were
also checked for tumor development.   One group  of hamsters was also in-
jected with carcinogen and the injection sites  were removed  at various
times for histological study.
     Homburger and Baker found that 10% of the  secondary host mice
developed palpable tumors by 10 weeks, whereas  14 to  15 weeks were

-------
                                   358
 required for 10% of the primary hosts to develop palpable tumors.   In
 the hamster study,  tumors palpable 4 weeks after transfer of the injec-
 tion sites later turned out to be malignant fibrosarcomas.   These partic-
 ular injection sites had been transferred 2 days after carcinogen injec-
 tion.
     RNA-loaded  atypical fibroblasts appeared at the  site of  administra-
 tion soon  after  injection and increased  in number until a fibrosarcoma
 was  formed.  Homburger  suggested that the fibroblasts  represent  premalig-
 nant cells which, if left in the original host,  slowly develop into a
 fibrosarcoma but which,  when transferred to a secondary host, express
 their malignant  behavior more readily.   Homburger and  Baker  proposed
 that (by using susceptible inbred strains of hamsters, by developing
 harvesting methods  for  selective collection of  fibroblasts,  and  by
 transfer of such cells  into fresh hamsters)  a test  is  theoretically
 possible requiring  less than 1 month for the demonstration of carcino-
 genic potency  in vivo in adult mammalian cells  that yield tumors as end
 points.

 5.7.3  Partial Hepatectomy
     Using the 1938 study of Alexander Haddow as a  starting  point,  Solt,
 Medline, and Farber (1977) developed a new model for  the  sequential
 analysis of liver carcinogenesis.  In their description of this  model
 Solt et  al.  stated  that cancer cells will emerge under conditions that
 would inhibit  or impair the growth of normal cells.   Hyperplastic liver
 lesions  induced  by  the  administration of hepatocarcinogens are resistant
 to other hepatocarcinogens and hepatotoxins that impair the  growth of
 surrounding normal  liver cells.
     Single doses of diethylnitrosamine  eventually  produce liver cancer,
 and  during the first week after exposure diethylnitrosamine-induced
 nodules  arise  on the surface of the  liver.   The model proposed by Solt,
Medline, and Farber (1977) contains  the  following features:   initiation
by diethylnitrosamine,  selective growth  inhibition  of normal cells by 2-
acetylaminofluorene and  growth stimulation of carcinogen-altered hepato-
cytes by partial hepatectomy.   The test  can be  completed  in  4 weeks.
The procedure  in male Fischer 344 rats was as follows:  (1)  the  rats
were injected  intraperitoneally with 200 mg/kg  diethylnitrosamine,  (2)

-------
                                   359
after a 2-week recovery period the animals were  placed  on  a basal  diet
containing 0.02% acetylaminofluorene,  and (3)  after  1 week on the  2-
acetylaminofluorene diet the rats were  subjected to  two-thirds partial
hepatectomy.  The animals were maintained on the carcinogen diet after
surgery until 24 h before sacrifice or for one more  week.   The liver
nodules continued to proliferate vigorously under these experimental
conditions, were characterized histologically by the investigators,  and
were identified as precursor lesions for some hepatocellular carcinomas.
     In experiments designed to study the cellular components involved
in hepatocarcinogenesis and the progression of such  cells  to malignancy,
Laishes and Farber (1978) isolated the carcinogen-altered  hepatocyte
populations from the livers of treated rats.   The cells,  some bearing a
y-glutamyl transpeptidase (y-GT) marker, were transferred  to the livers
of syngeneic host rats.  Selective proliferation of  the y-GT-positive
hepatocytes was then stimulated in the host rat  liver by  partial hepa-
tectomy, while proliferation of y-GT-negative host hepatocytes was
inhibited by dietary administration of 2-acetylaminofluorene.  The
system is quantitative and rapid — macroscopic foci  were  observed  within
10 days of cell transfer.  In a recent report Laishes,  Fink, and Carr
(1980) described in vitro purification of the carcinogen-altered cell
populations isolated from rats treated with the  diethylnitrosamine/2-
acetylaminofluorene/partial hepatectomy regime.   Primary  hepatocyte
monolayer cultures from carcinogen-treated and untreated  rats were
exposed for 24 h to acetylaminofluorene, to its  derivatives, or to
certain chemicals known to be cytocidal but nonhepatocarcinogenic.
Acetylaminofluorene (AAF), its derivatives N-OH-AAF and N acetoxy-AAF,
and three chemotherapeutic drugs — methotrexate, cyclohexamide, and
adriamycin — were found to be selectively cytocidal to normal rat
hepatocytes.  However, the hepatocyte phenotypes which developed  during
hepato-carcinogenesis were highly resistant to  the effects  of  the chemi-
cals.  Studies are under way in which cells selected in vitro will  be
transferred to syngeneic hosts for determination of colony-forming
ability.
     Tatematsu et al.  (1977) adapted the  system of  Solt,  Medline, and
Farber (1977) as an in vivo screening test for  hepatocarcinogens.  Their
procedure was as follows:   (1) rats were  injected intraperitoneally with

-------
                                   360
 200 mg  diethylnitrosamine  per kg  of body weight;  (2) two weeks later the
 animals were  started  for 2 weeks  on the test  chemical  in the diet or
 drinking water;  (3) after  1  week  of administration  of  the  test chemical
 partial hepatectomy was performed; and  (4)  one week later  the rats were
 killed. Various control groups were  established.   The end points of the
 experiment  were  the number of liver hyperplastic  nodules and the histo-
 chemical observation  of yGT, acid phosphatase (acid-Pase),  glucose-6-
 phosphatase (G-6-Pase), and  adenosine triphosphatase (ATPase) activity.
 The chemicals tested  included N-2-fluorenylacetamide,  3'-methyl-4-
 (dimethylamino)azobenzene, dimethylnitrosamine, diethylnitrosamine, DL-
 ethionine,  quinoline,  5,7-dibromo-8-hydroxy-quinoline,  8-hydroxyquinoline,
 or 8-nitroquinoline.   The  first six chemicals, known carcinogens, in
 combination with diethylnitrosamine and partial hepatectomy, induced
 statistically significantly  more  hyperplastic liver nodules than did
 diethylnitrosamine and partial hepatectomy  alone.   y-GT activity, not
 demonstrated  in  normal hepatocytes or after partial hepatectomy, was
 seen  in hyperplastic  nodules and  in the epithelium  of  bile ducts and
 ductules in the  portal area.  Acid-Pase and G-6-Pase were  usually
 decreased,  and ATPase was  usually absent in hyperplastic areas.  Thus,
 the presence  of  y-GT  and absence  of ATPase  are good markers for detec-
 tion  of hyperplastic  nodules.
      Tatematsu et al.  suggested further evaluation  of  the  system, par-
 ticularly to  eliminate the possibility of a noncarcinogen  acting as a
 selective growth inhibitor.  They suggested that  it may be useful as an
 in vivo short-term test in conjunction with in vitro assay systems and
 together these tests  would provide a  secondary screening for chemicals
 to be studied  in long-term in vivo studies.   Laishes,  Fink, and Carr
 (1980)  have recently  demonstrated that three  noncarcinogenic chemothera-
 peutic  agents  are selectively cytocidal to  normal cells in vitro.  (See
 above.)

 5.7.4  Alkaline  Elution
     Petzold and  Swenberg  (1978)  described  a  procedure  for detecting
 single  strand breaks  in DNA of tissues taken  from a wide range of organs
of rats following in vivo  exposure to known carcinogens of several
classes.

-------
                                   361

     Newborn rats were injected intraperitoneally with (3H)dThd (triti-
ated deoxythymidine) for 3 weeks.   The animals were exposed to chemicals
for various periods of time, and tissues were then removed and homoge-
nized.  The (3H)DNA elution was carried out as described by Swenberg,
Petzold, and Harbach (1976).  Increased alkaline elution was expressed
as the increase in elution of treated rats over that of vehicle control
rats.
     Twenty-three chemicals were tested and the following organs were
evaluated for DNA damage:  liver,  lung, kidney, brain, thymus, duodenum
stomach, bone marrow, and mammary gland.  The tissues tested were from
target and nontarget organs, and there was a high degree of correlation
between tissues with increased elution after in vivo exposure to carcino-
gens and target organs for tumor susceptibility.  For example, the
hepatocarcinogens dimethylnitrosamine, diethylnitrosamine, 2-acetyl-
aminofluorene, and #-hydroxyacetylaminofluorene caused the greatest
elution in liver preparations.  Less correlation was seen with chemicals
not requiring metabolic activation.  The alkaline elution assay is rapid
(not as time-consuming as the alkaline sucrose gradient technique) and
is reproducible.
     Petzold and Swenberg (1978) recommended the alkaline elution test
as a confirmation of in vitro tests when the carcinogenic potential  of a
compound is unknown.  It can help identify target organs for procarcino-
gens and can be utilized to assess DNA damage in organs that are subject
to toxic changes.

5.7.5  q-Fetoprotein in Serum
     a-Fetoprotein has been found in animals bearing  chemically induced
liver tumors.  Watabe (1971) described  the appearance of a-fetoprotein
in the serum of rats after  the start of  feeding of  4-dimethylaminoazo-
benzene.  The protein was detected in  the sera  of approximately 20%  of
41 rats by as early as 3 weeks and in  76% of the rats by  6 weeks.  The
serum protein disappeared at 11 to 12  weeks and reappeared  at 13 weeks
in 27 of 33 rats; 26 of  these 27 rats  developed hepatomas.   Ninety-one
percent of the rats that developed "early" a-fetoprotein  levels had
tumors after 19 weeks.

-------
                                   362

      Serum levels  of the protein increased from 2  to  4 mg/dL  in the
 early stage up to  60 to 100 mg/dL in the later stage.   Watabe suggested
 that  the  early protein peak could be attributed to chemical toxicity  not
 related to carcinogenicity.
      In a later study Kroes,  Williams,  and Weisburger (1973)  tested the
 effects of different dose levels of  several hepatotoxins and  hepatocar-
 cinogens  on the early appearance of  a-fetoprotein.  They found that high
 levels of liver carcinogen were required to induce the "early" peak at 2
 to  5  weeks.   The protein was not detected in the sera of rats treated
 with  hepatotoxins.   Kroes suggested  possibly using the a-fetoprotein
 assay as  a short-term test to distinguish between  hepatotoxins and
 hepatocarcinogens.

 5.7.6 Strain Susceptibility
      Differences in the responses of various strains  of animals to
 chemical  carcinogenesis are discussed in Sect.  5.2.2.   Selection of
 sensitive strains  of test animals can shorten tumor induction time
 considerably.   The following are examples of such  instances.
      5.7.6.1  Hamster Fibrosarcoma System — Homburger and Hsuch (1970)
 tested the susceptibility of inbred  strains of hamsters to tumor induc-
 tion  with 7,12-dimethylbenz(a)anthracene.   A dose  of  500 pg of the
 chemical,  injected subcutaneously, gave rise to subcutaneous  fibrosarcomas
 in  a  relatively short time.   The 15.16  line was the most sensitive, with
males and females  developing tumors  having average induction  times of 10
and 9 weeks,  respectively.   The 82.73 line was the least sensitive, with
males and females  developing tumors  having average induction  times of 18
and 15 weeks,  respectively.
      5.7.6.2   Rat  Mammary Tumor System  — Huggins,  Grand,  and  Brillantes
 (1961) reported preferential mammary tumor induction  in female Sprague-
Dawley rats  following single oral doses of polynuclear hydrocarbons.
      All  rats  injected with 20  mg or 15 mg of 7,12-dimethylbenzanthracene
developed mammary  tumors;  the tumor  induction time and the number of
active centers  were  dose  related.  Doses of 2 to 100  mg of 3-methylcho-
lanthrene induced  tumors  in  10% to 100% of the animals,  and the effect

-------
                                   363
was dose related; induction times and the numbers of active centers were
not.  Doses of 100 mg and 500 mg of 2-acetylaminofluorene produced
tumors in 30% of the animals, and only the induction times were dose
related.
     Hormonal treatments suppressed the development of mammary tumors
caused by 3-methylcholanthrene.  7,12-Dimethylbenzanthracene was the
most effective compound for the induction of mammary tumors (100% tumors
in 43 days).
     In a later study, Huggins, Grand,  and Fukunishi (1964) demonstrated
in the mammary tumor system that intravenous injection of a compound in
three equal doses was more effective in inducing tumors than the sum of
the doses given as a single injection.   The latent period was shorter,
and more active centers per animal were produced, although in both cases
100% of the rats developed tumors.
     Griswold et al. (1968) tested 35 compounds using the mammary tumor
induction system.  Ten doses of the maximum tolerated dose were adminis-
tered to each animal, and the experiment was terminated 9 months later.
(The duration was increased to 9 months for added sensitivity.)   Multiple
doses increased the number of tumor-bearing animals and the number of
tumors per rat; however, multiple doses were no more effective than the
single dose for determining general carcinogenicity of a compound.  The
mammary tumor assay is a sensitive test for detection of polynuclear
aromatic hydrocarbons, polycyclic nitro and amino derivatives, and certain
heterocyclic compounds.  The investigators recommend the test  for rapid
screening of certain potential carcinogens.
     5.7.6.3  Mouse Lung Adenoma  System — The lung  adenoma system was
first utilized as a bioassay of chemicals for carcinogenic potential  by
Andervont and Shimkin  (1940).  In an exhaustive  review of  the  lung
adenoma bioassay system, Shimkin  and Stoner  (1975)  outlined the  now
standardized procedure:
 1.  A  susceptible  inbred  strain  of mouse is selected, usually strain A.
     Weanlings are  generally used, and both  sexes should be included in
     initial bioassays.

-------
                                   364
  2.   The maximum tolerated dose of the chemical must be determined,
      and that dose is administered three times per week for 8 weeks.
      The second dose level is one-half the first and that of the third
      group is one-fifth the first.  Chemicals are generally injected
      intraperitoneally, but other routes can be used more effectively
      with certain chemicals.
  3.   Animals can be sacrificed in 12 to 24 weeks.  The nodules appearing
      on the surface of the lung are counted and are usually statistically
      evaluated; however,  most tests are considered positive if the
      number of lung tumors per mouse is increased by one or more,  if
      there is a dose-response relationship, and if the mean number of
      lung tumors in the vehicle and untreated controls is that usually
      found.
      The advantages of the system are that it is a rapid, convenient,
 and  quantitative procedure.   The criticisms are that the adenoma does
 not  have a counterpart in the human and that the tumors represent  an
 increase in incidences of spontaneous tumors instead of an inductive
 process.

 5.7.7  The Sebaceous Gland Test
      The sebaceous gland suppression test has been suggested as a  possible
 screening test for skin carcinogens (Brune, 1977; National Academy of
 Sciences,  1975).   Exposure of the skin to polycyclic hydrocarbons  leads
 to thickened epidermis and atrophy of the sebaceous glands in a matter
 of days.   However,  false  positive results have been obtained with  the
 test  (Weisburger,  1976),  making it subject to criticism.

 5.7.8  Host-Mediated In Vivo-In Vitro Assay
      DiPaolo et al.  (1973)  described a combination in vivo-in vitro
 bioassay  for carcinogenicity  utilizing transplacental exposure of  Syrian
 golden  hamster embryos.   Pregnant females were injected intraperitoneally
with  test  chemicals  at 10 to  11 days gestation.   The embryos were  excised
at day  13  of  gestation, and cells from the embryos were cultured in
Dulbeccos1 modified  Eagle's or  F-12  medium supplemented with 10% fetal

-------
                                  365
bovine serum.  Cells were passed every 4 to 6 days from secondary cultures.
Colonies, formed by cells transferred to feeder layers, were scored for
morphologic transformation and were further checked for tumorigenicity
by animal innoculation.  The cells were harvested and were injected
subcutaneously into hamsters, which were watched closely for subsequent
tumor development.  Sarcomas were produced in 3 to 16 weeks.  Twelve
known chemical carcinogens, of which several were known to be inactive
in in vitro tests, were tested in this system, and all produced neoplas-
tic transformation; five noncarcinogens did not.  The results were
reproducible, the assay seems to eliminate false negative results that
occur because of the requirement for metabolic activation, and spontaneous
transformation does not occur.

5.7.9  Conclusions
     Short-term tests for carcinogenicity are not intended to replace
the conventional lifetime studies.  However, these types of assays could
be valuable tests in the screening of chemicals for carcinogenicity or,
more precisely, for lack of carcinogenic potential.  A battery of short-
term tests could be designed to identify safe compounds so that they can
be eliminated from further testing.
     The procedures described above can be completed in from 3 weeks
(appearance of a-fetoprotein in the serum) to 3 to 4 months (the time
required for the detection of actual tumors, as in the lung adenoma
assay and the embryo homograft technique).  Because exposure of cells  to
chemicals occurs in vivo, these particular assays have the additional
advantage of allowing for metabolic activation.  However, in order  to
determine the best application of in vivo short-term assays to the
screening of chemicals for carcinogenicity, further evaluation of such
tests is necessary, and their limits of sensitivity must  be established.

-------
                                   366


                       REFERENCES  FOR SECTION 5
Abrahms, W., G. Zbinden, and R. Bagdon.   1965.   Investigative Methods in
  Clinical  Toxicology.  J. New Drugs  5:199-207.

Adamson, R. H., R. W. Cooper, and R.  W.  O'Gara.   1970.  Carcinogen-Induced
  Tumors in Primitive Primates.  J. Natl. Cancer Inst. 45:555-560.

Adamson, R. H., P. Correa, and D. W.  Dalgard.  1973.  Occurrence of a
  Primary Liver Carcinoma in a Rhesus Monkey Fed Aflatoxin BI-  J. Natl.
  Cancer Inst. 50(2):549-553.

Agthe, C.,  H.  Garcia, P. Shubik, L. Tomatis, and E. Wenyon.  1970.  Study
  of the Potential Carcinogenicity of DDT in the Syrian Golden Hamster.
  Proc. Soc. Exp. Biol. Med. 134:113-116.

Akamatsu, Y.,  and B. Barton.  1974.   Neoplasms and Amyloidosis in Strains
  of Mice Treated with 3-Methylcholanthrene.  J.  Natl. Cancer Inst.
  52:377-385.

Alfred, L.  J., A. Globerson, Y. Berwald,  and R.  T. Phren.  1964.  Differ-
  ential Toxicity Response of Normal  and Neoplastic Cells In Vitro to
  3,4-Benzopyrene and 3-Methylcholanthrene.  Br.  J. Cancer 18:159-164.

Allen, M. J.,  E. Boyland, C. E. Dukes, E. S. Horning, and J. G. Watson.
  1957.  Cancer of the Urinary Bladder Induced in Mice with Metabolites
  of Aromatic  Amines and Tryptophan.  Br. J. Cancer 11:212-230.

Andervont,  H.  B.  1950.  Induction of Hemangio-endotheliomas and Sarcomas
  in Mice with 0-Aminoazotoluene.  J. Natl. Cancer Inst. 10:927-941.

Andervont,  H.  B.  1958.  Induction of Hepatomas  in Strain C3H Mice with
  4-0-Tolylazo-O-Toluidine and Carbon Tetrachloride.  J. Natl. Cancer
  Inst. 20:431-438.

Andervont,  H.  B., and T. B. Dunn.  1962.  Occurrence of Tumors in Wild
  House Mice.  J. Natl. Cancer Inst.  28:1153-1163.

Andervont,  H.  B., H. G. Grady, and J. E.  Edwards.  1942.  Induction of
  Hepatic Lesions, Hepatomas, Pulmonary  Tumors and Hemangio-endotheliomas
  in Mice with 0-Aminoazotoluene.  J. Natl. Cancer Inst. 3:131-153.

Andervont,  H.  B., and M. B. Shimkin.  1940.  Biologic Testing of Carcino-
  gens.  II.   Pulmonary-Tumor-Induction  Technique.  J. Natl. Cancer Inst.
  1:225-239.

Andrianova, M.  1971.  Transplacental Action of  3-Methylcholanthrene and
  Benz(a)pyrene on Four Generations of Mice.  Bull. Exp. Biol. Med.  (USSR)
  71(1):677-680.

-------
                                  367

Argus, M. F., and C. Hoch-Ligeti.  1963.   Induction of Malignant Tumors
  in the Guinea Pig by Oral Administration of Diethylnitrosamine.   J.
  Natl. Cancer Inst. 30:533-551.

Arnold, D., S. Charbonneau, Z. Zawidzka,  and H.  Grice.  1977.   Monitoring
  Animal Health During Chronic Toxicity Studies.   J.  Environ.  Pathol.
  Toxicol. 1:227-239.

Axelrad, A., and H. van der Gaag.  1962.   Susceptibility to Lymphoma
  Induction by Gross1 Passage A Virus in C3Hf/Bi Mice of Different Ages:
  Relation to Thymic Cell Multiplication and Differentiation.   J.  Natl.
  Cancer Inst. 28:1065-1094.

Baba, T., and S. Takayama.  1961.  Influence of p-Dimethylamine-Azobenzene
  (DAB) Pretreatment in Neonatal Stage on the DAB Heptacarcinogenesis in
  Adult Rats, with Special Reference to Sex Difference in the Carcino-
  genesis.  Gann 52:73-82.

Barnes, J., and F. Denz.  1954.  Experimental Methods in Determining
  Chronic Toxicity.  Pharmacol. Rev. 6:191-242.

Beckers, A., and H. Bazin.  1978.  Incidence of Spontaneous Ileocecal
  Immunocytomas in Hybrids of LOU/C Rats and Rat Strains with Low Spon-
  taneous Tumor Incidence  (brief communication).  J. Natl. Cancer Inst.
  60(6):1505-1508.

Benitz, K. F.  1970.  Measurement of Chronic Toxicity.  In:  Methods in
  Toxicology, G. E. Paget, ed.  F. A. Davis Company, Philadelphia.
  pp. 82-131.

Berenblum, I., L. Boiato, and N. Trainin.  1966.  On  the Mechanism of
  Urethane Leukemogenesis in Newborn C57BL Mice.  II.  Influence of Thy-
  mectomy and of Subsequent Thymus Reimplantation. Cancer Res.  26:361-363.

Bhisey,  R. A., and  S. M.  Sirsat.  1975.  Histological Changes  in the Mouse
  Skin After One Subcutaneous Injection of 20-Methylcholanthrene.  Indian
  J. Cancer 12:430-438.

Bielschowsky, F., and W.  S. Bullough.  1949.  Epidermal Mitotic Activity
  and  the Induction of  Skin Tumors  in Mice. Br. J. Cancer  3:282-285.

Bonser,  G. M., E. Boyland, E. R. Busby, D. B. Clayson, P.  L. Grover, and
  J. W.  Jull.  1963.  A Further  Study of Bladder  Implantation  in  the Mouse
  as the Means of Detecting Carcinogenic Activity:   Use  of Crushed Paraf-
  fin Wax or  Stearic Acid as  the Vehicle.  Br. J.  Cancer  17:127-136.

Bonser,  G. M., D. B. Clayson, J. W.  Jull,  and L.  N.  Pyrah.   1952.  The
  Carcinogenic Properties of  2-Amino-l-Naphthol  Hydrochloride  and Its
  Parent Amine  2-Naphthylamine.  Br. J.  Cancer  6:412-424.

Boorman,  G. A.,  and C.  F.  Hollander.   1974.   High Incidence of Spontane-
  ous  Urinary Bladder and Ureter Tumors  in the  Brown Norway Rat (brief
  communication).   J. Natl. Cancer  Inst.  52(3):1005-1008.

-------
                                   368

 Boyd,  E.   1968.   Predictive Drug  Toxicity:  Assessment of Drug Safety
   Before  Human Use.   Can.  Med. Assoc.  J.  98:278-293.

 Braun,  W.,  L.  Sung,  D.  Keyes, and R.  Kociba.   1977.   Pharmacokinetic and
   Toxicological  Evaluation of Dogs Fed 1,2,4,5-Tetrachlorobenzene  in the
   Diet for Two Years.   Toxicol. Appl.  Pharmacol.  41:218-219.

 Bridges,  B.  A.   1976.   Short Term Screening Tests for Carcinogens.  Nature
   261:195-200.

 Brune,  H.F.K.  1977.   Experimental Results with Percutaneous Applications
   of Automobile  Exhaust Condensates in Mice.   In:  Air Pollution and Cancer
   in Man,  U. Mohr, D.  Schmall, and L.  Tomatis, eds.   International Agency
   for  Research on Cancer,  Lyon, France,   pp.  41-47.

 Brusick,  D.  J.   1978.   The Role of Short-Term Testing in Carcinogen Detec-
   tion.   Chemosphere No.  5:403-417.

 Burek,  J.  D.,  and C.  F.  Hollander.   1977.  Incidence  Patterns of Sponta-
   neous Tumors in BN/Bi Rats.  J.  Natl. Cancer Inst.  58(1):99-105.

 Butler, W.  H.  1979.   Report of the Workshop  on Carcinogenicity.  J.
   Toxicol.  Environ.  Health 5:161-162.

 Cameron,  G.  R.,  and  K.  K.  Cheng.   1951.   Failure  of Oral DDT to Induce
   Toxic Changes  in Rats.   Br. Med.  J.  2:819-821.

 Campbell,  J. S.,  Y.  H.  Yang, and  J.  D.  Bolton.  1965.  Murine Lung Ade-
   nomas Induced  by Intravaginal Carcinogen.   Arch. Path. 79:588-594.

 Cardesa, A., P.  Pour,  J. Althoff,  and  U.  Mohr.  1974.  Effects of Intra-
   peritoneal Injections of Dimethyl- and  Diethylnitrosamine, Alone or
   Simultaneously on  Swiss  Mice.   Z.  Krebscorsch.  82:233-238.

 Chieco-Bianchi,  L.,  G.  De  Benedictis,  G.  Tridente, and L. Fiore-Donati.
   1963.  Influence of Age  on Susceptibility to Liver  Carcinogenesis and
   Skin  Initiating Action by Urethane in Swiss Mice.   Br. J. Cancer 17:
   672-680.

 Clapp,  N. K., R.  L.  Tyndall, and  J. A.  Otten.  1971.  Differences in
   Tumor Types and Organ Susceptibility in BALB/c  and  RF Mice Following
   Dimethylnitrosamine and  Diethylnitrosamine.  Cancer Res. 31:196-198.

 Clark, D.   1977.  Long-Term Inhalation Toxicology Studies.  In:  Current
  Approaches in  Toxicology, B. Ballantyne, ed.  John  Wright and Sons,
   Bristol,  pp.  105-114.

Crocker, T. T., J. E. Chase, S. A. Wells, and L.  L. Nunes.  1970. >Prelim-
   inary Report on Experimental Squamous Carcinoma  of  the Lung in Hamsters
  and in a Primate.   In:  Morphology of Experimental  Respiratory Carcino-
  genesis, P. Nettesheim, M. G. Hanna, Jr., and J. W. Deatheridge, Jr.,
  eds.   Oak Ridge, Tenn., USAEC Division  of Technical Information.
  pp.  317-328.

-------
                                  369

Dale, M. M.,  G. C. Easty, R. Tchao,  H.  Desai,  and M.  Andjargholi.   1973.
  The Induction of Tumors in the Guinea Pig with Methylcholanthrene and
  Diethylnitrosamine and Their Propogation In  Vivo and In Vitro.   Br.  J.
  Cancer 27:445-450.

Davies, R. F., I. R. Major,  and E.  R.  Aberdeen.   1970.  Pulmonary Adeno-
  mata Induced by Carcinogen Treatment in Organ Culture.   Influence of
  Increasing Amounts of Carcinogen.   Br.  J. Cancer 24:785-787.

Davies, R. F., I. R. Major,  and E.  R.  Aberdeen.   1971.  The Induction of
  Adenomata in Mouse Lung Homografts by Chemical Carcinogens.  Br.  J.
  Cancer 25:565-567.

Davis, B. R., J. K. Whitehead, M. E. Gill, P.  N. Lee, A.  D. Butterworth,
  and F.J.R.  Roe.  1975.  Response of Rat Lung to 3,4-Benzpyrene Adminis-
  tered by Intratracheal Instillation in Infusine With or Without Carbon
  Black.  Br. J. Cancer 31:443-452.

De Benedictis, G., G. Maiorano, L.  Chieco-Bianchi, and L. Fiore-Donati.
  1962.  Lung Carcinogenesis by Urethane in Newborn,  Suckling,  and Adult
  Swiss Mice.  Br. J. Cancer 16:686-689.

Deichmann, W. B., M. Keplinger, F.  Sala,  and E.  Glass.  1967.   Synergism
  Among Oral Carcinogens.  IV.  The Simultaneous Feeding of Four Tumori-
  gens to Rats.  Toxicol. Appl. Pharmacol. 11:88-103."

Deichmann, W. B., and J. L.  Radomski.  1963.  The Carcinogenicity and
  Metabolism of  2-Naphthylamine and Related Compounds.  Ind. Med. Surg.
  32:161.

Deichmann, W. B., J. L. Radomski, E. Glass, W.A.D. Anderson, M. M. Coplan,
  and F. M. Woods.  1965.  Synergism Among Oral Carcinogens. III.  Simul-
  taneous Feeding of Four Bladder Carcinogens to Dogs.  Ind. Med. Surg.
  34:640-649.

Delia Porta, G.  1968.  Use of Newborn and Infant Animals  in Carcinogenic-
  ity Testing.   Food Cosmet. Toxicol. 6:243-252.

Delia Porta, G., L. Kolb, and P. Shubik.   1958.  Induction of Tracheobron-
  chial Carcinomas  in the Syrian Golden Hamster.  Cancer Res. 18:592-597.

Delia Porta, G., M. Rappaport, U. Saffiotti, and P.  Shubik.  1956.  Induc-
  tion of Melanotic Lesions During  Skin Carcinogenesis in  Hamsters.   Arch.
  Pathol. 61:303-313.

Delia Porta, G., and B. Terracini.  1969.  Chemical  Carcinogenesis  in
  Infant Animals.   Prog. Exp. Tumor Res.  11:334-363.

DiPaolo, J. A.,  R.  L. Nelson, P. J. Donovan, and  C.  H. Evans.   1973.   Host
  Mediated In Vivo  - In Vitro Assay for Chemical  Carcinogenesis.   Arch.
  Pathol. 95:380-385.

-------
                                    370

Diwan, B. A., and H. Meier.  1974.  Strain- and Age-Dependent Transplacental
  Carcinogenesis by 1-Ethyl-l-Nitrosourea in Inbred Strains of Mice.
  Cancer Res. 34:764-770.

Doell, R., and W. Carnes.   1962.  Urethane Induction of Thymic Lymphoma
  in C57BL Mice.  Nature 194:588-589.

Durham, W. F., P. Ortega, and W. J. Hays, Jr.  1963.  The Effect of Various
  Dietary Levels of DDT on  Liver Function, Cell Morphology and DDT Storage
  in the Rhesus Monkey.  Arch. Int. Pharmacodyn. 141:111-129.

Dyer, H., M. Kelly, and R.  O'Gara.  1966.  Lack of Carcinogenic Activity
  and Metabolic Fate of Fluorenylacetamide in Monkeys.  J. Natl. Cancer
  Inst. 36:305-322.

Epstein, S., J. Andrea, and H. Jaffe.   1967.  Carcinogenicity of the
  Herbicide Maleic Hydrazide.  Nature (London) 215:1388-1390.

Fancher, 0.  1978.  Species Selection and Animal Models for Toxicologic
  Studies.  Clin. Toxicol.  12(2):239-247.

Farber, E.  1963.  Ethionine Carcinogenesis.  Adv. Cancer Res. 7:383-474.

Fare, G.  1966.  Rat Skin Carcinogenesis by Topical Applications of Some
  Azo Dyes.  Cancer Res. 26:2406-2408.

Federal Register.  1978.  Proposed  Guidelines for Registering Pesticides
  in the U.S. Hazard Evaluation:  Humans and Domestic Animals. 43FR(163):
  37336-37403.

Federal Register.  1979.  Proposed  Health Effects Tests Standards for
  Toxic Substances Control  Act Test Rules.  44FR(91):27334-27375.

Flaks, A.  1965.  The Effect of 9,10-Dimethyl-l,2-Benzanthracene on Young
  Mice of Low and High Cancer Strain.   Br. J. Cancer 19:547-550.

Flaks, A.  1966.  Test for  Carcinogenesis of Cigarette Tobacco Smoke
  Condensate Using Young Strong A and C57BL Mice.  Br. J. Cancer
  20:145-147.

Food Safety Council.  1978.  Proposed System for Food Safety Assessment.
  Food Cosmet. Toxicol. 16, Suppl.  2:1-136.

Fox, J. G.   1977.  Clinical Assessment  of Laboratory Rodents on Long-Term
  Bioassay Studies.  J. Environ. Path,  and Toxicol. 1:199-226.

Friedman, L.  1973.  Problems of Evaluating the Health Significance of the
  Chemicals Present in Foods.  In:  Pharmacology and the Future of Man,
  Proc. 5th Int. Congr. Pharmacology, San Francisco, 1972, Vol. 2.  Karger,
  New York.  pp. 30-41.

-------
                                   371

Fugmann, R. A., J. C.  Anderson, R.  L.  Stolfi,  and D.  S.  Martin.   1977.
  Comparison of Adjuvant Chemotherapeutic Activity Against Primary and
  Metastatic Spontaneous Murine Tumors.   Cancer Res.  37:496-500.

Gak, J. C., C. Graillot, and R. Truhaut.   1976.  Use  of  the Golden Hamster
  in Toxicology (Part II of two parts).   Lab.  Anim. Sci.  26(2):274-280.

Gargus, J., 0. Paynter, and W. Reese,  Jr.  1969.   Utilization of Newborn
  Mice in the Bioassay of Chemical  Carcinogens.  Toxicol.  Appl.  Pharmacol.
  15:552-559.

Gilbert, C., J. Gillman, P. Loustalot, and W.  Lutz.   1958.  The Modifying
  Influence of Diet and the Physical Environment on Spontaneous Tumor
  Frequency in Rats.  Br. J. Cancer 12:565-593.

Glucksmann, A., and C. Cherry.  1971.   Sex Difference and Carcinogenic
  Dosage in the Induction of Neoplasms in the  Salivary Glands of Rats.
  Br. J. Cancer 25:212-224.

Goerttler, K., and H.  Lohrke.  1977.  Diaplacental Carcinogenesis:  Tumor
  Localization and Tumor Incidence  in NMRI Mice After Diaplacental Initia-
  tion with DMBA and Urethane and Postnatal Promotion with the Phorbol
  Ester TPA in a Modified 2-Stage Berenblum/Mottram Experiment.  Virchos
  Arch. A. Path. Anat. and Histol.  376:117-132.

Golberg, L., ed.  1974.  Carcinogenesis Testing of Chemicals.  The Chemical
  Rubber Company, Cleveland, Ohio.   144 pp.

Golberg, L.  1975.  Safety Evaluation Concepts.  J. Assoc. Off. Anal. Chem.
  58(4)-.635-644.

Goldenthal, E.  1968.  Current Views on Safety Evaluation of Drugs.  U.S.
  Food and Drug Administration Papers May:13-18.

Goldwater, L. J., A. J. Rosso, and M. Kleinfeld.  1965.   Bladder Tumors
  in a Coal Tar Dye Plant.  Arch. Environ. Health 11:814-817.

Gorrod, J., R. Carter, and F. Roe.   1968.  Induction of Hepatomas by  4-
  Aminobiphenyl and Three of  Its Hydroxylated Derivatives Administered
  to Newborn Mice.  J. Natl.  Cancer Inst. 41:403-410.

Graffi, A., F. Hoffman, and M. Schutt.   1967.   tf-Methyl-tf-Nitrosourea
  As a Strong Topical Carcinogen When Painted  on  the Skin of  Rodents.
  Nature  (London) 214:611.

Grahn, D., and K. F. Hamilton.  1964.  Influence  of  Sex,  Environment,
  and Radiation Factors on Life Shortening and Tumor  Incidence  in C3H
  Mice.  Radiat. Res.  22:191.

Grasso, P.  1976.  Review  of  Tests  for Carcinogenicity  and  Their Signif-
  icance  to Man.  Clin. Toxicol. 9:745-760.

-------
                                    372

 Greenman,  D.  L.,  and R.  R.  Delongchamp.   1979.   Variability  of  Response
   to  Diethylstilbesterol:   A Comparison  of  Inbred  with Hybrid Mice.   J.
   Toxicol.  and  Environ.  Health 5:131-143.

 Greenman,  D., R.  Delongchamp,  and B.  Highman.   1979.   Variability  of
   Response to Diethylstilbestrol:  A Comparison of Inbred  with  Hybrid
   Mice.  J.  Toxicol.  Environ.  Health 5:131-143.

 Greenwald,  P.,  J.  Barlow,  P.  Nasca,  and  W.  Burnett.   1971.   Vaginal Cancer
   After Maternal  Treatment with Synthetic Estrogens.   New  Engl.  J. Med.
   285:390-392.

 Griesemer,  R. A.,  P.  Nettesheim,  D.  H. Martin,  and J.  E. Caton,  Jr.   1977.
   Quantitative  Exposure  of Grafted Rat Tracheas to 7,12-Dimethylbenz(a)-
   anthracene.   Cancer Res.  37:1266-1271.

 Griswold,  D.  P.,  A.  E. Casey,  E.  K.  Weisburger,  and J.  H.  Weisburger.
   1968.  The Carcinogenicity of Multiple Intragastric  Doses  of  Aromatic
   and Heterocyclic Nitro or Amino Derivatives  in Young Sprague-Dawley
   Rats.  Cancer Res.  28:924-933.

 Gruenstein,  M., D. R.  Meranze,  and M. B.  Shimkin.   1966.   Mammary, Seba-
   ceous, and Cutaneous Neoplasms  and Leukemia  in Male  Wistar Rats  Receiv-
   ing Repeated  Gastric Instillations of  3-Methylcholanthrene.   Cancer Res.
   26:2202-2205.

 Hayes, W.   1967.   Toxicity of  Pesticides to Man:   Risks from Present
   Levels.   Proc.  R.  Soc. London Ser.  B.  167:101-127.

 Herbst, A.,  L.  Ulfelder,  and D. Poscanzer.   1971.   Adenocarcinoma  of  the
   Vagina.   Association of  Maternal Stilbestrol  Therapy with  Tumor  Appear-
   ance in  Young Women.   New Engl.  J.  Med. 284:878-881.

 Herrold, K.  M.  1964a.   Induction of Olfactory  Neuroepithelial  Tumors in
   Syrian Hamsters  by Diethylnitrosamine.  Cancer 17:114-121.

 Herrold, K.   19642?.   Effect of  Route of  Administration on  the Carcinogenic
  Action of  Diethylnitrosamine.   Br.  J.  Cancer  18:763-767.

 Herrold, K.   1969.  Aflatoxin  Induced Lesions  in Syrian Hamsters.  Br. J.
  Cancer 23:655-660.

 Herrold, K. M., and L. J.  Dunham.   1963.  Induction of Tumors in the
  Syrian Hamster with Diethylnitrosamine (tf-Nitrosodiethylamine).
  Cancer Res. 23:773-777.

Heston, W. E.   1958.  Mammary Tumors  in  Agent-Free Mice.   Ann.  N.Y.
  Acad. Sci. 71:931-942.

Heston, W.  E., and G. Vlahakis.   1966.   Factors  in the Causation of
  Spontaneous Heptaomas  in  Mice.   J. Natl.  Cancer  Inst. 37:839-843.

-------
                                   373

Heston, W. E., and G. Vlahakis.   1968.  C3H-AVy - A High Hepatoma and
  High Mammary Tumor Strain of Mice.   J.  Natl.  Cancer Inst.  40:1161-1166.

Heston, W. E., G. Vlahakis, and M.  K.  Deringer.  1960.   High Incidence
  of Spontaneous Hepatomas and the Increase of  This Incidence with
  Urethane in C3H, C3Hf, and C3He Male Mice.  J. Natl.  Cancer Inst.
  24:425-435.

Heuper, W. C., and H. D. Wolf.  1937.   Experimental Production of Aniline
  Tumors of Dogs in the Bladder.  Amer.  J.  Pathol.  13:656-657.

Hiaro, F., T. Fujisawa, E. Tsubura, K. Akamatsu, Y. Yamamura.  1968.
  Experimental Lung Cancer in Rabbits  Induced by Chemical Carcinogens.
  Gann 59:497-505.

Hoag, W. G.  1963.  Spontaneous Cancer in Mice.  Ann. N.Y. Acad.  Sci.
  108:805-831.

Hodge, H., A. Boyce, W. Deichmann,  and H. Kraybill.  1967.  Toxicology
  and No-Effect Levels of Aldrin and Dieldrin.   Toxicol. Appl. Pharmacol.
  10:613-675.

Holland, J. M., D. G. Gosslee, and N.  J.  Williams.   1979.  Epidermal
  Carcinogenicity of Bis(2,3-epoxycyclopentyl)ether, 2,2-Bis(p-glyci-
  dyloxyphenyl) propane and m-Phenylenediamine  in Male and Female C3H
  and C57 BL/6 Mice.  Cancer Res. 39:1714-1721.

Homburger, F., and J. R. Baker.   1969.  Accelerated Carcinogen Testing.
  Progr. Exp. Tumor Res., Vol. 11.   Karger, New York.  pp. 384-394.

Homburger, F., and S. S. Hsuch.   1970.  Rapid Induction of Subcutaneous
  Fibrosarcoma by 7,12-DimethyIbenz(a)anthracene in an Inbred Line of
  Syrian Hamsters.  Cancer Res.  30:1449-1452.

Homburger, F., S. Hsuch, C. S. Kerr, and A. B.  Russfield.  1972.  Inher-
  ited Susceptibility of Inbred Strains of  Syrian Hamsters to Induction
  of Subcutaneous Sarcomas and Mammary and  Gastrointestinal Carcinomas
  by Subcutaneous and Gastric Administration of Polynuclear Hydrocarbons.
  Cancer Res. 32:360-366.

Homburger, F., A. B. Russfield.  1970.  An Inbred Line  of Syrian Hamsters
  with Frequent Spontaneous Adrenal Tumors.  Cancer  Res.  30:305-308.

Homburger, F., and A. Tregier.  1960.   Modifying Factors  in  Carcinogen-
  esis.  Prog. Exptl. Tumor Res. 1:311.

Howell, J.   1963.  The Experimental Production  of  Vascular Tumors in the
  Rat.  Br.  J. Cancer 17:663-671.

Huggins, C., L. C. Grand, and F. P. Brillantes.  1961.   Mammary  Cancer
  Induced by a Single Feeding of Polynuclear Hydrocarbons and Its Sup-
  pression.  Nature 189:204-207.

-------
                                   374

Huggins, C., L. Grand, and R. Fukunishi.  1964.  Aromatic Influences on
  the Yields of Mammary Cancers Following Administration of 7,12-Dimethyl-
  benz(a)anthracene.  Proc. Natl. Acad. Sci., Vol. 51.  pp. 737-742.

Ihle, J. N., L. 0. Arthur, and D. L.  Fine.  1976.  Autogenous Immunity
  to Mouse Mammary Tumor Virus in Mouse Strains of High and Low Mammary
  Tumor Incidence.  Cancer Res. 36:2840-2844.

Innes, J.R.M., B. M. Ulland, M. G. Valeric, L. Petrucelli, L. Fishbein,
  E. R. Hart, A. J. Pallotta, R. R. Bates, H. L. Falk, T. J. Gart, M.
  Klein, I. Mitchell, and J. Peters.   1969.  Bioassay of Pesticides and
  Industrial Chemicals for Tumorigenicity in Mice.  J. Natl. Cancer Inst.
  42:1101-1114.

Ivankovic, S.  1973.  Experimental Prenatal Carcinogenesis.  In:  Trans-
  placental Carcinogenesis, L. Tomatis and U. Mohr, eds.  Scientific
  Publication No. 4, International Agency for Research on Cancer, Lyon,
  France,  pp. 92-99.

JaLanko, H., I. Virtanen, E. Engvall,  and E. Ruoslahti.  1978.  Early
  Increase of Serum Alpha-Fetoprotein in Spontaneous Hepatocarcinogenesis
  in Mice.  Int. J. Cancer 21(4):453-459.

Jull, J. W.  1951.  The Induction of  Tumors of the Bladder Epithelium in
  Mice by the Direct Application of a Carcinogen.  Br. J. Cancer 5:328-330.

Jungherr, E.  1963.  Spontaneous Cancer in Zoo and Laboratory Animals.
  Part II.  Tumors and Tumor-Like Conditions in Monkeys.  Ann. N.Y. Acad.
  Sci. 108:777-792.

Kaplan, A., and H. Sherman.  1977.  Toxicity Studies with Methyl-tf-
  [(Methyllamine-carbonyl)oxyl]-ethanimidothioate.  Toxicol. Appl.
  Pharmacol. 40:1-17.

Kaye, A., and N. Trainin.  1966.  Urethan Carcinogenesis and Nucleic
  Acid Metabolism:  Factors Influencing Lung Adenoma Induction.  Cancer
  Res. 26:2206-2212.

Kelly, M., and R. O'Gara.  1961.  Induction of Tumors in Newborn Mice
  with Dibenz(a)anthracene and 3-Methylcholanthrene.  J. Natl. Cancer
  Inst. 26:651-679.

Kelly, M. G., R. W. O'Gara, R. H. Adamson, K. Gadekar, C. C. Botkin,
  W. H. Reese, Jr., and W. T. Kerber.  1966.  Induction of Hepatic Cell
  Carcinomas in Monkeys with tf-Nitrosodiethylamine.  J. Natl. Cancer
  Inst. 36:323-351.

Ketkar, M., G. Reznik, P. Schneider,  and U. Mohr.  1978.  Investigations
  on the Carcinogenic Burden by Air Pollution in Man.  Intratracheal
  Instillation Studies with Benzo(a)pyrene in Bovine Serum Albumin in
  Syrian Hamsters.  Cancer Lett. 4:235-239.

-------
                                   375

Klein, M.  1951.  The Action of Croton Oil in the Induction of Sarcomas
  in Mice.  J. Natl. Cancer Inst.  11:843-848.

Klein, M.  1952.  The Action of Methylcholanthrene,  Croton Oil, and 1,2-
  Benzanthracene in the Induction of Skin Tumors in Strain DBA Mice.  J.
  Natl. Cancer Inst. 12:735-742.

Klein, M.  1953.  Effects of Croton Oil on Induction of Tumors by 1,2-
  Benzanthracene, Deoxycholic Acid, or Low Doses of 20-MCA in Mice.
  J. Natl. Cancer Inst. 13:333.

Klein, M.  1963.  Susceptibility of Strain BLoAFi/J Hybrid Infant Mice
  to Tumorigenesis with 1,2-Benzanthracene, Deoxycholic Acid, and 3-
  Methylcholanthrene.  Cancer Res. 23(6):1701-1707.

Krasovskii, G.  1976.  Extrapolation of Experimental Data from Animals
  to Man.  Environ. Health Perspect. 13:51-58.

Kroes, R., G. M. Williams, and J.  H. Weisburger.  1973.  Early Appearance
  of Serum a-Fetoprotein as a Function of Dosage of Various Hepatocarci-
  nogens.  Cancer Res. 33:613-617.

Kuschner, M., S. Laskin, E. Cristofano, and N. Nelson.  1957.  Experi-
  mental Carcinoma of the Lung.  In:  Proceedings of the Third National
  Cancer Conference.  Lippincott,  Philadelphia,  pp. 485-495.

Laishes, B., and E. Farber.  1978.  Transfer of Viable Putative Preneo-
  plastic Hepatocytes to the Livers of Syngenic Host Rats.  J. Natl.
  Cancer  Inst. 61(2):507-512.

Laishes, B. A., L. Fink, and B. I. Carr.  1980.  A Liver Colony Assay  for
  a New Hepatocyte Phenotype as a Step Towards Purifying New Cellular
  Phenotypes That Arise During Hepatocarcinogenesis.  Ann. N.Y. Acad.
  Sci. (in press).

Lawrence, W.  1976.  Systemic Toxicity.  Acute vs. Subchronic  and  Chronic.
  Drug. Cosmet. Ind. November 1976:40-114.

Laws, J. 0., and A. Flaks.  1966.  Pulmonary Adenomata Induced by  Carci-
  nogen Treatment in Organ Culture.  Br. J.  Cancer 20:550-554.

Lee, C-C., J. Q. Russell, and J. L. Minor.   1978.  Oral Toxicity of Fer-
  ric Dimethyl-Dithiocarbamate  (Ferbam) and  Tetramethylthiuram Disulfide
  (Thiram) in Rodents.  J. Toxicol. Environ. Health  4:93-106.

Lee, K.,  B. Toth, and P. Shubik.   1963.  Carcinogenic  Response of  the
  Syrian  Golden Hamster Treated at Birth with  7,12-Dimethylbenz(a)-
  anthracene.  Proc. Soc. Exptl.  Biol. Med.  114:579-582.

Levy, B.  M.   1963.   Induction  of  Fibrosarcoma  in the Primate,  Tamarinus
  nigrioolUs.  Nature 200:182-183.

-------
                                   376

 Liebelt,  R.,  A.  Liebelt,  and M.  Lane.   1964.   Hormonal Influences  on
   Urethan Carcinogenesis  in C3H/f Mice.   Cancer Res.  24:1869-1879.

 Liebelt,  R. ,  R.  Yoshida,  and G.  Gray.   1961.   Enhancement  of Liver Tumor-
   igenesis in Zb Mice Injected with  Urethan at Newborn Age.   Proc.  Assoc.
   Cancer  Res.  3:245.

 Lijinsky,  W.,  A.  Ferrero,  R.  Montesano,  and C.E.M.  Wenyon.   1970.   Tumor-
   igenicity  of Cyclic Nitrosamines in  Syrian Golden Hamsters.   Z.
   Krebsforsch.  74:185-189.

 Lijinsky,  W.,  K.  Lee, Y.  Tomatis, and  W.  H.  Butler.   1967.   Nitrosoazeti-
   dine —  a Potent Carcinogen of  Low  Toxicity.   Naturwissenschaften 54:518.

 Lijinsky,  W.,  and H.  W. Taylor.   1978.   Relative Carcinogenic Effective-
   ness of Derivatives of  Nitrosodiethylamine in Rats.   Cancer Res.  38:
   2391-2394.

 Loomis, T.   1974.   Essentials of Toxicology,  2nd ed.,  Lea  and Febiger,
   Philadelphia,   pp.  189-191.

 Luz, A.   1977.   The Range of Incidence of Spontaneous  Neoplastic and Non-
   Neoplastic  Lesions  of the Laboratory Mouse.   Z. Versuchstierk. 19(6):
   342-343.

 Maekawa,  A.,  and S. Odashima.  1975.   Spontaneous Tumors in  AC1/N  Rats.
   J. Natl. Cancer Inst. 55:1437-1445.

 Magee, P.  1970.   Tests for Carcinogenic  Potential.   In:   Methods  in Tox-
   icology, G.  Paget,  ed.   F.  A.  Davis  Company, Philadelphia,   pp.  158-196.

 McNamara,  B.   1976.   Concepts in Health Evaluation  of  Commercial and
   Industrial  Chemicals.   In:   Advances in Modern Toxicology,  Part  1,  Vol.
   1, M. Mehlman,  R. Shapiro,  and H.  Blumenthal,  eds.   John Wiley and
   Sons, New York.   pp. 61-140.

 Milievskaja,  I.  L., and N.  S.  Kiseleva.   1976.   Comparative  Study  of the
   Carcinogenic Activities  of  NAS and Some Chemical  Carcinogens When Intro-
   duced into  the Buccal Pouch of the Syrian Hamster.   Bull.  World  Health
   Organ.  54:607-614.

 Miller, E. C., J.  A.  Miller,  and M.  Enomota.   1964.  The Comparative Car-
   cinogenicities  of 2-Acetylaminofluorene and  Its /V-Hydroxy  Metabolite  in
  Mice, Hamsters  and  Guinea Pigs.  Cancer Res.  24:2018-2031.

Ministry of Health and Welfare Canada.   1975.   The  Testing of Chemicals
   for Carcinogenicity, Mutagenicity, and  Teratogenicity.   Ministry of
  Health and Welfare, Ottawa.  183 pp.

Mohr, U.   1973.   Effects of Diethylnitrosamine on Fetal and  Suckling Syrian
  Golden Hamsters.  In:  Transplacental Carcinogenesis, L. Tomatis and  U.
  Mohr, eds.  Scientific Publication No.  4,  International  Agency for Research
  on Cancer, Lyon, France,  pp.  65-70.

-------
                                  377

Mohr, U., G. Reznik, and P. Pour.   1977.   Carcinogenic Effects of Diisopro-
  panolnitrosamine in Sprague-Dawley Rats.   J.  Natl.  Cancer Inst. 58:361-366.

Morris, H. P., and W. H. Eyestone.   1953.   Tumors of  the Liver and Urinary
  Bladder of Dogs After Ingestion of 2-Acetylaminofluorene.   J.  Natl.  Cancer
  Inst. 13:1139-1165.

Morris, H. P., and H. I. Firminger.   1956.   Influence of Sex and Sex Hor-
  mones on Development of Hepatomas and Other Hepatic Lesions in Strain
  AXC Rats Ingesting 2-Diacetylaminofluorene.   J. Natl.  Cancer Inst.
  16:927-949.

Mulay, A. S., and R. W. O'Gara.   1959.   Incidence of  Liver Tumors in Male
  and Female Rats Fed Carcinogenic Azo  Dyes.   Proc.  Soc. Exptl.  Biol.  Med.
  100:320-322.

Munro, I.  1977.  Considerations in Chronic Toxicity  Testing:  The Chemical,
  the Dose, the Design.  J. Environ. Pathol.  Toxicol. 1:183-197.

Nagasawa, H. , R. Yanai, and I. Azuma.  1978.   Suppression by Nocardia rubra
  Cell Wall Skeleton of Mammary DNA Synthesis,  Plasma Prolactin Level, and
  Spontaneous Tumorigenesis in Mice.  Cancer Res. 38:2160-2162.

National Academy of Sciences.   1975.  Principles for Evaluating Chemicals
  in the Environment.  National Academy of Sciences,  Washington, D.C.
  pp. 115-153.

National Academy of Sciences.   1977.  Principles and Procedures for Evalu-
  ating the Toxicity of Household Substances.  National Academy of Sciences,
  Washington, D.C.  130 pp.

National Institutes of Health.  1968-1973.  Survey of Compounds Which
  Have Been Tested for Carcinogen Activity.  Public Health Service Publi-
  cation No. 149, Government Printing Office, Washington, D.C.

Neiman, J. M.  1968.  The Sensitizing Carcinogenic Effect of Small Doses
  of Carcinogen.  Europ. J. Cancer 4:537-545.

Nelson, A. A., 0. G. Fitzhugh, and H. 0. Calvery.  1943.  Liver Tumors
  Following Cirrhosis Caused by Selenium in Rats.  Cancer Res.  3:230-236.

Nelson, A. A., and G. Woodard.  1953.  Tumors of the Urinary Bladder, Gall
  Bladder and Liver in Dogs Fed 0-Aminoazotoluene or p-Dimethylaminoazo-
  benzene.  J. Natl. Cancer Inst. 13:1497-1509.

Nettesheim, P., and R. A. Griesemer.  1978.  Experimental Models  for
  Studies of Respiratory Tract Carcinogenesis.   In:  Pathogenesis and
  Therapy of Lung Cancer, Lung Biology in Health and Disease 10,  C. Harris,
  ed.  New York.  pp. 75-188.

-------
                                   378


Nettesheim, P.,  and A.  S. Hammons.   1971.   Induction  of  Squamous Cell
  Carcinoma in  the Respiratory  Tract of Mice.  J. Natl.  Cancer  Inst.
  47:697-701.

Newberne, P. M.,  and A. Rogers.   1972.  Vitamin A, Liver, Colon Carcinoma
  in Rats Fed Low Levels of Aflatoxin.  Toxicol. Appl. Pharmacol.  22:280.

Nishizuka, Y.,  K. Nakakuki, and T.  Sakakura.   1964.   Induction  of  Pulmonary
  Tumors and Leukemia by a Single Injection of 4-Nitroquinoline 1-Oxide  to
  Newborn and Infant Mice.  Gann 55:495-508.

Noble, R., B. C.  Hochachka, and D.  King.   1975.  Spontaneous and Estrogen-
  Produced Tumors in Nb Rats  and Their Behavior After Transplantation.
  Cancer Res. 35:766-780.

Noyes, W. F.  1968.  Carcinogen Induced Sarcoma in the Primitive Primate,
  Tupaia glis.   Proc. Soc. Exp.  Biol. Med.  127:594-596.

Noyes, W. F.  1969.  Carcinogen-Induced Neoplasia with Metastasis  in a
  South American Primate, Saquinus  oedipus.  Proc. Soc.  Expt. Biol. Med.
  131:223-225.

O'Gara, R. W.,  and M. G. Kelly.   1965.  Pulmonary Arteriolitis  in  Infant
  Monkeys Produced by Subcutaneous  Injections  of Polycyclic Hydrocarbons.
  Arch. Path. (Chicago) 79:475-483.

Oppenheimer, B.  S., E.  T. Oppenheimer, and A.  P. Stout.   1948.  Sarcomas
  Induced by Rats by Implanting Cellophane.  Proc. Soc.  Expt. Biol. Med.
  67:33-34.

Outzen, H. C.,  R. Custer, R.  Phillips, G.  J. Eaton, and  R. T. Prehn.  1975.
  Spontaneous and Induced Tumor Incidence  in Germ Free Nude Rats.  J.
  Reticuloendothel. Soc. 17(1):l-9.

Page, N. P.  1977a.  Concepts of a  Bioassay Program in Environmental
  Carcinogenesis.  In:  Advances in Modern Toxicology, Vol. 3:  Environ-
  mental Cancer,  H. Kraybill  and M.  Mehlman, eds.  Hemisphere Publishing
  Corp., Washington, D.C.  pp.  87-171.

Page, N.  19772?.  Chronic Toxicity  and Carcinogenicity Guidelines.  J.
  Environ.  Pathol. Toxicol. 1:161-182.

Peacock, P- M.   1962.  A Short-Term Test for Carcinogenicity (The  Effects
  of Certain Closely-Related Polycyclic Aromatic Hydrocarbons on Embryo
  Tissue Homografts in BALB/c Strain Mice).  Br. J. Cancer 16:701-706.

Peacock, P. M.,  and E. Dick.  1963.   A Short-Term Test for Carcinogenicity
  (Mouse Embryo Tissue Homografts in BALB/c Strain Mice).  Br.  J.  Cancer
  17:59-61.

-------
                                  379

Peck, H.  1968.  An Appraisal of Drug Safety Evaluation in Animals and
  the Extrapolation of Results to Man.   In:   Importance of Fundamental
  Principles in Drug Evaluation, D.  Tedeschi and R.  Tedeschi, eds.
  Raven Press, New York.  pp. 449-471.

Peck, H.  1974.  Design of Experiments to Detect Carcinogenic Effects of
  Drugs.  In:  Carcinogenesis Testing of Chemicals,  L.  Goldberg, ed.  The
  Chemical Rubber Company, Cleveland, Ohio.   pp. 1-14.

Peraino, C., R.J.M. Fry, and E. Staffeldt.   1973.  Enhancement of Sponta-
  neous Hepatic Tumorigenesis in C3H Mice by Dietary Phenobarbital (brief
  communication).  J. Natl. Cancer Inst. 51:1349-1350.

Percy, D. H., and A. M. Jonas.  1971.  Incidence of Spontaneous Tumors
  in CD11-! H3M/ICR Mice.  J. Natl. Cancer Inst.  46:1045-1053.

Petzold, G. L. and J. A. Swenberg.  1978.  Detection of DNA Damage Induced
  In Vivo Following Exposure of Rats to Carcinogens.  Cancer Res. 38:
  1589-1594.

Pietra, G., H. Rappaport, and P. Shubik.  1961.   The Effects of Carcino-
  genic Chemicals in Newborn Mice.  Cancer 14:308-317.

Pietra, G., K. Spencer, and P. Shubik.   1959.  Response of Newly Born Mice
  to a Chemical Carcinogen.  Nature 183:1689.

Pollard, M. , and M. Kajima.  1970.  Lesions in Aged Germ Free Wistar Rats.
  Amer. J. Pathol. 61:25-32.

Pott, F., A. Brockhaus, and F. Huth.  1973.   Tests on the Production of
  Tumors in Animal Experiments with Polycyclic Aromatic Hydrocarbons.
  Zbl. Bakt. Hyg., I. Abt. Orig. B 157:34-43.

Pott, F., R. Tomingas, and J. Misfeld.  1977.  Tumors in Mice After  Subcu-
  taneous Injection of Automobile Exhaust Condensates.  In:  Air  Pollution
  and Cancer in Man, Mohr, Sahmahl, and Tomatis, eds.  International Agency
  for Research on Cancer,  pp.  79-87.

Pour, P., F. W. Kruger, and J. Althoff.  1974.   Cancer of the Pancreas
  Induced in the Syrian Golden Hamster.  Am. J.  Pathol. 76:349-358.

Pour, P., F. W. Kruger, and J. Althoff.  1975.   Effect of Beta-oxidized
  Nitrosamines on Syrian Hamsters.   III.  2,2-Dihydroxy-di-n-propylnitro-
  samine.  J. Natl. Cancer Inst.  54:141-145.

Pour, P., N. Kmoch, E. Greiser, U. Mohr, J.  Althoff, and A.  Cardesa.   1976.
  Spontaneous Tumors and Common Diseases in  Two  Colonies of  Syrian Hamsters.
  J. Natl. Cancer Inst. 56(6):931-935.

Rail, D.   1969.  Difficulties  in  Extrapolating  the  Results  of Toxicity
  Studies  in Laboratory Animals to Man.  Environ. Res.  2:360-367.

-------
                                   380

Reeves, A. L., H. E. Puro,  and R. G.  Smith.   1974.   Inhalation Carcinogen-
  esis from Various Forms of Asbestos.   Environ.  Res.  8:178-202.

Reuber, M. D.  1976.  Effect of Age and  Sex  on Lesions of  the Esophagus  in
  Buffalo Strain Rats Ingesting Diethylnitrosamine.  Exp. Cell Biol. 44:65-72.

Reuber, M. D., and E. L. Glover.  1967.  Hyperplastic  and  Early Neoplastic
  Lesions of  the Liver  in Buffalo Strain Rats of  Various Ages Given Sub-
  cutaneous Carbon Tetrachloride.  J. Natl.  Cancer  Inst. 38:891-899.

Reznik, G.  1975.  The  Carcinogenic Effect of (DMN)  on the Chinese Hamster
  (Cricetulus Griseus).  Cancer Lett. 1:25-28.

Rice, J.  1976.  Carcinogenesis:  A Late Effect of  Irreversible Toxic
  Damage During Development.  Environ. Health Perspect. 18:133-139.

Rigdon, R. H., M. C. Benge, H. Kirchoff, J.  Mack, and  J. Neal.  1969.
  Leukemia in Mice Fed  Benzo(a)pyrene:   A Clinical,  Pathologic and Hema-
  tologic Study.  Tex.  Rep. Biol. Med. 27:803-820.

Rigdon, R. H., and J. Neal.  1966.  Gastric  Carcinomas and Pulmonary Ade-
  nomas in Mice Fed Benzo(a)pyrene.   Tex. Rep. Biol. Med.  24:195-207.

Riley, V.  1975.  Mouse Mammary Tumors:  Alteration  of Incidence  as
  Apparent Function of  Stress.  Science  189:465-467.

Roe, F.  1975.  Neonatal Induction of Hepatic and Other Tumors.   In:
  Mouse Hepatic Neoplasia,  W. Butler  and P.  M. Newbern, eds.  Elsevier
  Scientific Publishing Co., New York.   Chapter 7.

Roe, F., R. Carter, and S.  Adamthwaite.  1969.  Induction  of Liver and
  Lung Tumors in Mice by 6-Aminochrysene Administered  During the  First
  Three Days of Life.   Nature (London) 221:1063-1064.

Roe, F., B. Mitchley, and M. Walters.  1963.  Tests  for Carcinogenesis
  Using Newborn Mice:   1,2-Benzanthracene, 2-Naphthylamine, 2-Naphthyl-
  hydroxylamine and Ethyl Methane Sulphonate.  Br. J.  Cancer 17:255-260.

Roe, F., K. Rowsen, and M.  Salaman.   1961.   Tumours  of Many Sites Induced
  by Injection of Chemical  Carcinogenesis into Newborn Mice, A Sensitive
  Test for Carcinogenesis:  The Implication  for Certain Immunological
  Theories.  Br. J. Cancer  15:515-530.

Rosenkrantz, H., R. Sprague, R. Fleischman,  and M. Braude.  1975.  Oral
  A9-Tetrahydrocannabinol Toxicity in Rats Treated  for Periods up to
  6 Months.  Toxicol. Appl. Pharmacol. 32:399-417.

Rygaard, J., and C. 0.  Polvson.  1974.  The  Mouse Mutant Nude Does Not
  Develop Spontaneous Tumors.  Acta.  Pathol.  Microbiol. Scand. 82:
  99-106.

-------
                                  381

Sabine, J. R.,  B.  J.  Norton,  and M.  B.  Wicks.   1973.   Spontaneous Tumors
  in C3H-AVy and C3H-AVyfB Mice:  High  Incidence in the United States and
  Low Incidence in Australia.   J.  Natl.  Cancer Inst.  50:1237-1242.

Saffiotti, U.,  F.  Cefis, R. Montesano,  and A.  R. Sellakumar.   1966.   In-
  duction of Bladder Cancer in Hamsters Fed Aromatic Amines.   Ind.  Med.
  Surg. 35:564.

Sass, B., L. S. Rabstein, R.  Madison, R.  M.  Nims,  R.  L.  Peters,  and G. J.
  Kelloff.  1975.   Incidence  of Spontaneous Neoplasms in F344 Rats Through-
  out the Natural Life-Span.   J. Natl.  Cancer  Inst.  54:1449-1456.

Schepers, G.W.H.  1971.   Lung Tumors of Primates and Rodents.  Part II.
  Inhalation Experiments.  Ind. Med. May:23-31.

Schmahl, D., and H. Osswald.   1967.   Carcinogenesis in Different Animal
  Species by Diethylnitrosamine.  Experentia 23:497-498.

Schrauzer, G.  N.,  D.  A.  White, J.  E. McGinness,  and C. J. Schneider.  1978.
  Arsenic and Cancer:  Effects of Joint Administration of Arsenite and
  Selenite on the Genesis of  Mammary Adenocarcinoma in Inbred Female
  C3H/St Mice.   Bioinorg. Chem. 9:245-253.

Schreiber, H.,  K.  Schreiber,  and D.  H.  Martin.  1975.  Experimental Tumor
  Induction in a Circumscribed Area of  the Hamster Trachea:  Correlation
  of Histology and Exfoliative Cytology.  J. Natl.  Cancer Inst. 54:187-197.

Shimkin, M. B.   1940.  Induced Pulmonary Tumors in Mice.  II.  Reaction
  of Lungs of Strain A Mice to Carcinogenic Hydrocarbons.  Arch. Path.
  29:239-255.

Shimkin, M. B., and G. D. Stoner.   1975.   Lung Tumors in Mice:  Applica-
  tion to Carcinogenesis Bioassay.  Adv.  Cancer Res. 21:1-58.

Shirasu, Y.  1965.  Comparative Carcinogenicity of 4-Nitroquinoline 1-
  Oxide and 4-Hydroxyaminoquinoline 1-Oxide in Three Strains of Mice.
  Proc. Soc. Exptl. Biol. Med. 118:812-814.

Shubik, P.  1950.   The Growth Potentialities of Induced Skin Tumors in
  Mice.  The Effects of Different Methods of Chemical Carcinogenesis.
  Cancer Res.  10:713-717.

Shubik, P.  1972.   The Use of the Syrian Golden Hamster in Chronic Tox-
  icity Testing.  Prog.  Exp.  Tumor Res. 16:176-184.

Silverstone, H., and A.  Tannenbaum.  1951.  Proportion  of Dietary Protein
  and the Formation of Spontaneous Hepatomas.  in the Mouse.   Cancer Res.
  11:442-446.

Snell, K. C.  1965.  Spontaneous Lesions of the Rat.   In:  The  Pathology
  of Laboratory Animals, W. E. Ribelin and J. R. McCoy,  eds.  Charles C.
  Thomas, Publisher, Springfield, Illinois.

-------
                                   382

Solt, D. B., A. Medline, and E. Farber.  1977.  Rapid Emergence of
  Carcinogen-Induced Hyperplastic Lesions in a New Model for the Sequen-
  tial Analysis of Liver Carcinogenesis.  Am. J. Path. 88:595-618.

Sontag, J. M., N. P. Page, and U. Saffiotti.  1976.  Guidelines for Car-
  cinogen Bioassay in Small Rodents.  NCI-CG-TR-1, February.  Bethesda,
  Maryland.

Spatz, M. , and G. Laqueur.  1967.  Transplacental Induction of Tumors in
  Sprague-Dawley Rats with Crude Cyad Material.  J. Natl. Cancer Inst.
  38:233-245.

Spitz, S., W. H. Maguigan, and K. Dobriner.  1950.  The Carcinogenic
  Action of Benzidine.  Cancer 3:789-804.

Steinmuller, D., L. A. Dillingham, and R. T. Prehn.  1969.  Lack of Car-
  cinogenic Activity of 3-Methylcholanthrene in the Squirrel Monkey.  J.
  Natl. Cancer Inst. 43:1175-1180.

Stevens, L. C.  1973.  A New Inbred Subline of Mice (129/terSv) with a
  High Incidence of Spontaneous Congenital Testicular Teratomas.  J.
  Natl. Cancer Inst. 50:235-242.

Stevenson, D.  1979.  Current Problems in the Choice of Animals for Tox-
  icity Testing.  J. Toxicol. Environ. Health 5:9-15.

Stoltz, D. R., L. A. Poirer, C. C. Irving, H. F. Stich, J. H. Weisburger,
  and H. C. Price.  1974.  Evaluation of Short-Term Tests for Carcino-
  genicity.  Tox. Appl. Pharmacol. 29:157-180.

Street, A.  1970.  Biochemical Tests in Toxicology.  In:  Methods in
  Toxicology, G. Paget, ed.  F. A. Davis Company, Philadelphia.
  pp. 313-337.

Strong, L. C., and H. Matsunaga.  1975.  An Inverse Correlation (Dose-
  Response) Between 5-Methyl-Cyildine and the Fate of Spontaneous Tumors
  in Mice.  Cytobios 12:13-18.

Sugiura, K., W. E. Smith, and D. A. Sunderland.  1956.  Experimental Pro-
  duction of Carcinoma in Rhesus Monkeys.  Cancer Res. 16:951.

Sumi, N., D. Stavrou, H. Frohberg, and G. Jochmann.  1976.  The Incidence
  of Spontaneous Tumors of the Central Nervous System of Wistar Rats.
  Arch. Toxicol. 35:1-13.

Sunderman, F. W., Jr.  1971.  Metal Carcinogenesis in Experimental Animals.
  Food Cosmet. Toxicol. 9:105-120.

Svenberg, J., A. Kostner, W. Wechsler, and R. Denlinger.  1972.  Quanti-
  tative Aspects of Transplacental Tumor Induction with Ethylnitrosourea
  in Rats.  Cancer Res. 32:2656-2660.

-------
                                  383

Swenberg, J.  A.,  G.  L.  Petzold,  and P.  R.  Harbach.   1976.   In Vitro DNA
  Damage/Alkaline Elution Assay  for Predicting Carcinogenic Potential.
  Biochem. Biophys.  Res.  Comm.  72:732-738.

Syndor, K. L.  1973.  A Preliminary Investigation of the Susceptibility
  to Sarcoma Induction by Low Doses of  B(a)P in Nine Strains of Inbred
  Male Rats.   Proceedings Tobacco and Health Workshop Conf., 4th University
  of Kentucky Tobacco and Health Research Institute.

Syndor, K. L., 0. Butenandt, F.  P. Brillantes, and C. Huggins.  1962.
  Race - Strain Factor Related to Hydrocarbon-Induced Mammary Cancer in
  Rats.  J. Natl. Cancer Inst.  29:805-814.

Tannenbaum, A., and H.  Silverstone.  1949.   The Influence of the Degree
  of Caloric Restriction on the  Formation of Skin Tumors and Hepatomas
  in Mice.  Cancer Res. 9:724-727.

Tatematsu, M., T. Shirai, H. Tsuda, Y.  Miyata, Y. Shinohara, and N. Ito.
  1977.  Rapid Production of Hyperplastic Liver Nodules in Rats Treated
  with Carcinogenic Chemicals:   A New Approach for an In Vivo Short-Term
  Screening Test for Hepatocarcinogens.  Gann 68:499-507.

Terracini, B., P. N. Magee, and  J. M. Barnes.  1967.  Hepatic Pathology
  in Rats on Low Dietary Levels  of Dimethylnitrosamine.  Br. J. Cancer
  21:559-565.

Terracini, B., G. Palestro, M.  Gigliardi, and R. Montesano.  1966.  Car-
  cinogenicity of Dimethylnitrosamine in Swiss Mice.  Br. J. Cancer 20:
  871-876.

Terracini, B., M. C. Testa, J. R. Cabral, and N. Day.  1973.  The Effects
  of Long-Term Feeding of DDT to BALB/c Mice.  Int. J. Cancer 11:747-764.

Thiery, M.,  and M. van Gijsegem.   1965.  Species and Strain Differences
  in the  Susceptibility of  the Cervico-Vaginal Squamous Epithel.  to 3,4-
  Benzo(a)pyrene.  Br. J. Cancer 19:418-429.

Tomatis,  L., C. Agthe, H. Bartsch, J. Huff, R. Montesano,  R.  Saracci,
  E. Walker, and J. Wilbourn.  1978.  Evaluation of  the Carcinogenicty
  of Chemicals:  A Review of the Monograph Program  of  the  International
  Agency  for Research  on Cancer  (1971  to 1977).  Cancer Res.  38:877-885.

Tomatis,  L., G. Delia  Porta, and P.  Shubik.   1961.   Urinary Bladder and
  Liver Cell Tumors Induced in Hamsters with  0-Aminoazotoluene.   Cancer
  Res. 21:1513-1517.

Tomatis,  L.,  and V. Turusov.  1975.  Studies  on  the Carcinogenicity of
  DDT.  Gann Monograph on Cancer  Res.  17:219-241.

Tomatis,  L.,  V.  Turusov, D. Guibbert,  B. Duperray,  C.  Malaveille, and
  H. Pacheco.  1971.   Transplacental Carcinogenic  Effect of 3-Methyl-
  cholanthrene in Mice and  Its  Quantitation in Fetal Tissues.  J. Natl.
  Cancer  Inst. 47:645-651.

-------
                                   384

 Toth,  B.   1968.   A Critical Review of Experiments in Chemical  Carcino-
   genesis Using  Newborn Animals.   Cancer Res.  28:727-738.

 Toth,  B.,  P.  Magee,  and P.  Shubik.   1964.   Carcinogenesis  Study  with
   Dimethylnitrosamine Administered Orally to Adult and Subcutaneously
   to Newborn  BALB/c  Mice.   Cancer Res.  24:1712-1721.

 Toth,  B.,  H.  Rappaport, and P.  Shubik.   1963.   Influence of Dose and Age
   on the  Induction of Malignant Lymphomas and  Other Tumors by  7,12-Dimethyl-
   benz(a)anthracene  in Swiss Mice.   J.  Natl. Cancer Inst.  30:723-741.

 Toth,  B.,  and P.  Shubik.   1963.   Carcinogenesis in Lewis Rats  Injected
   at Birth with  7,12-Dimethylbenz(a)anthracene.   Br.  J.  Cancer 17:540-545.

 Turner, F.  C.  1941.   Sarcomas  at the Site of  Subcutaneously Implanted
   Bakelite Discs in  Rats.   J. Natl.  Cancer Inst.  2:81.

 Turusov,  V.,  L.  Tomatis, D.  Guibbert, B.  Duperray,  and H.  Pacheco.   1973.
   The  Effect  of  Prenatal Exposure of  Mice to Methylcholanthrene  Combined
   with the Neonatal  Administration of Diethylnitrosamine.   In:   Trans-
   placental Carcinogenesis,  L.  Tomatis and U.  Mohr,  eds.   Scientific
   Publication No.  4,  International Agency for  Research on  Cancer, Lyon,
   France,   pp. 84-91.

 U.S. Food  and Drug Administration.   1971.   Advisory Committee  on Protocols
   for  Safety  Evaluation, Panel  on Carcinogenesis  Report  on Cancer Testing
   in the  Safety  Evaluation  of Food Additives and  Pesticides.   Toxicol.
   Appl. Pharmacol. 20:419-438.

 Verschuuren,  H.,  R.,  Kroes,  and G.  Van Esch.   1973.   Toxicity  Studies on
   Tetrasul.   I.   Acute, Long-Term,  and Reproduction Studies.   Toxicology
   1:63-78.

 Vesselinovitch,  S.   1973.   Comparative Studies on Perinatal Carcinogenesis.
   In:  Transplacental Carcinogenesis,  L.  Tomatis  and  U.  Mohr,  eds.   Sci-
   entific  Publication No. 4,  International Agency for Research on Cancer,
   Lyon, France,   pp.  14-22.

 Vesselinovitch,  S. D.,  A. P.  Kyriazis,  N.  Mihailovich,  and K.V.N. Rao.
   1975.  Conditions Modifying Development of Tumors  in Mice at Various
   Sites by  Benzo(a)pyrene.   Cancer  Res.  35:3948-2953.

 Vesselinovitch,  S., and N. Mihailovich.   1966.  Significance of  Newborn
  Age  and Dose of  Urethan in Leukemogenesis.   Cancer  Res.  26:1633-1637.

Vesselinovitch,  S, N.  Mihailovitch, and  L.  Itze.   1970.  Comparative
   Studies on  the Kinetics of  the  Neoplastic Competence in  Mice.   Cancer
  Res.  30:2548-2551.

Vesselinovitch, S., N.  Mihailovich, G. Wogan,  L.  Lombard,  and  K.  Rao.
  1972.  Aflatoxin Bj,  a Hepatocarcinogen  in the  Infant  Mouse.   Cancer
  Res.  32:2289-2291.

-------
                                  385

Vesselinovitch, S., K. Rao, and N.  Mihailovich.   1975.   Factors Modu-
  lating Benzidine Carcinogenicity  Bioassay.   Cancer Res.  35:2814-2819.
                                                                 w
Vlahakis, G.,  W. E. Heston, and G.  H.  Smith.   1970.   Strain C3H-A J fZ
  Mice:  Ninety Percent Incidence of Mammary Tumors  Transmitted by
  Either Parent.  Science 170:185-187.

Walpole, A. L., M.H.C. Williams, and D. C.  Roberts.   1952.  The Carcino-
  genic Action of 4-Aminodiphenyl and 3:2'Dimethyl-4-Aminodiphenyl.  Br.
  J. Ind. Med. 9:255-263.

Walpole, A. L., M.H.C. Williams, and D. C.  Roberts.   1954.  Tumors of
  the Urinary Bladder in Dogs After Ingestion of 4-Aminodiphenyl.  Br.
  J. Ind. Med. 11:105-109.

Watabe, H.  1971.  Early Appearance of Embryonic a-Globulin in Rat Serum
  During Carcinogenesis with 4-Dimethylaminobenzene.  Cancer Res. 31:
  1192-1194.

Watanabe, P.,  J. Young, and P. Gehring.  1977.  The  Importance of Non-
  Linear (Dose-Dependent) Pharmacokinetics in Hazard Assessment.  J.
  Environ. Pathol. Toxicol. 1:147-159.

Weil, C.  1962.  Applications of Methods of Statistical Analysis to
  Efficient Repeated-Dose Toxicological Tests:  1.  General Considera-
  tions and Problems  Involved.  Sex Differences in Rat Liver and Kidney
  Weights.  Toxicol. Appl. Pharmacol. 4:561-571.

Weil, C.  1972.  Guidelines for Experiments to Predict the Degree of
  Safety of a Material for Man.  Toxicol. Appl. Pharmacol. 21:194-199.

Weil, C.  1973.  Experimental Design and Interpretation of Data  from
  Prolonged Toxicity  Studies.  Pharmacology and the Future of Man.
  Proc. 5th Int. Congr. Pharmacol., San Francisco, 1972, Vol. 2:
  Toxicological Problems, Karger, New York.  pp. 4-12.

Weil, C., and C. Carpenter.   1969.   Abnormal Values in Control Groups
  During Repeated-Dose Toxicologic  Studies.  Toxicol. Appl. Pharmacol.
  14:335-339.

Weil, C., and D. McCollister.   1963.   Safety Evaluation of Chemicals.
  Relationships Between Short-  and  Long-Term Feeding Studies  in  Designing
  and Effective Toxicity Test.  Agric. Food Chem. 11:486-491.

Weil, C., M. Woodside, H.  Smyth, Jr.,  and C. Carpenter.   1971.   Results
  of Feeding Propylene Glycol in the Diet of Dogs for Two Years.  Food
  Cosmet. Toxicol.  9:479-490.

Weisburger, E. K.   1977.   Carcinogenicity Studies on Halogenated Hydro-
  carbons.  Environ.  Health Perspect.  21:7-16.

-------
                                   386

Weisburger, J. H.   1976.  Bioassays  and Tests  for  Chemical Carcinogens.
  In:  Chemical Carcinogens, American Chemical Society, Monograph 173,
  C. E.  Searle, ed. Washington,  D.C.  pp.  1-23.

Weisburger, J., M.  Klein, E. Weisburger,  R.  Glass,  G. Woodard,  and M.
  Gronin.   1970.  Comparison of  the  Effect of  the  Carcinogen tf-Hydroxyl-
  tf-2-Fluorenylacetamide  in Infant and Weanling Rats.  J. Natl.  Cancer
  Inst.  45:29-35.

Weisburger, J. H.,  and  E. K. Weisburger.   1967.  Tests for Chemical Car-
  cinogens.   From:  Methods in Cancer Res.,  Vol. I., H. Busch,  ed.
  Academic  Press, New York.  pp.  307-398.

Welsch,  C.  W.  1976.  Interaction of Estrogen  and  Prolactin in  Spontaneous
  Mammary Tumorigenesis of  the Mouse.  J.  Toxicol.  Environ. Health Suppl.
  1:161-175.

Wigley,  C.  B., J. Amos, and P. Brookes.   1978.   Different Tumours Induced
  by Benzo(a)pyrene and Its 7,8-Dihydrodiol Injected into Adult Mouse
  Salivary  Gland.   Br.  J. Cancer 37:657-661.

Winstead, J.  1978.   What Kinds  of Tests  Are Prerequisite to Labeling a
  Compound  as Toxic?  Int.  J.  Occup. Health Saf. 47(3):26-28.

Wislocki, P.  G., R. L.  Chang,  A.  W.  Wood,  W. Levin, H. Yagi, 0.  Hernandez,
  H. D.  Mah,  P. M.  Dansette, D.  M. Jerina, and A.  H. Conney.  1977.  High
  Carcinogenicity of  2-Hydroxybenzo(a)pyrene on Mouse Skin.  Cancer Res.
  37:2608-2611.

Wogan, G. N., and R.  C. Shank.   1971.  Toxicity and Carcinogenicity of
  Aflatoxins.  In:  Advances in  Environmental  Science and Technology,
  J. N.  Pitts and R.  L. Metcalf,  eds., John Wiley  & Sons, Inc.,  New York.
  pp. 321-350.

Womack,  J.  1979.   Genetic  Constitution and Response to Toxic Chemicals —
  An Overview.  J.  Toxicol. Environ. Health 5:49-51.

World Health  Organization.  1978.  Principles  and  Methods for Evaluating
  the Toxicity of Chemicals.   Part 1.  Geneva.   274 pp.

Yoshida, T.   1932.  Uber die Experimantelle Erzeugung von Hepatom durch
  die Futterung mit o-Amido-azotoluol.  Proc.  Imp.  Acad. Japan  8:464-467.

Zabezhinskii, M. A.   1970.  Effectiveness of Inhalation as a Method of
  Administration of Atomized Carcinogens.   Bull. Exptl. Biol. and Med.
  69:68-70.

Zackheim, H.  S.  1964.  Comparative  Cutaneous  Carcinogenesis in the Rat.
  Oncologia 17:236-246.

Zbinden, G.    1973.  Formal  Toxicology.  In:  Progress in Toxicology,
  Special Topics, Vol.  1.   Springer-Verlag,  New York.  pp. 4-27.

-------
                                  387

Zoller, M.,  S. Matzku, and K. Goerttler.   1978.   High Incidence of Spon-
  taneous Transplantable Tumours in BDX Rats.   Brit. J.  Cancer 37:61-66.

-------
                                   388


                       BIBLIOGRAPHY FOR SECTION 5
 Abrahms,  W.,  G.  Zbinden,  and R.  Bagdon.   1965.   Investigative Methods in
   Clinical Toxicology.   J.  New Drugs 5:199-207.

 Adamson,  R.  H.,  R.  W.  Cooper, and R. W.  O'Gara.   1970.   Carcinogen-Induced
   Tumors  in Primitive  Primates.   J.  Natl.  Cancer Inst.  45:555-560.

 Adamson,  R.  H.,  P.  Correa,  and D. W. Dalgard.   1973.   Occurrence of a
   Primary Liver  Carcinoma in a Rhesus Monkey Fed Aflatoxin Bj.  J.  Natl.
   Cancer  Inst. 50(2):549-553.

 Agthe,  C., H.  Garcia,  P.  Shubik, L.  Tomatis, and E. Wenyon.   1970.   Study
   of the  Potential  Carcinogenicity of DDT in the Syrian Golden Hamster.
   Proc. Soc.  Exp. Biol.  Med.  134:113-116.

 Akamatsu,  Y.,  and B. Barton.   1974.   Neoplasms  and Amyloidosis in Strains
   of Mice Treated with 3-Methylcholanthrene. J. Natl.  Cancer Inst.
   52:377-385.

 Alfred, L. J., A. Globerson,  Y.  Berwald,  and R.  T. Phren.   1964.   Differ-
   ential  Toxicity Response  of Normal and Neoplastic Cells  In Vitro to
   3,4-Benzopyrene and  3-Methylcholanthrene.   Br. J. Cancer 18:159-164.

 Allen,  M.  J.,  E. Boyland, C.  E.  Dukes,  E.  S. Horning,  and  J.  G.  Watson.
   1957.   Cancer  of  the Urinary Bladder Induced  in Mice with Metabolites
   of Aromatic  Amines and  Tryptophan.  Br.  J. Cancer 11:212-230.

 Andervont, H.  B.  1950.   Induction of Hemangio-endotheliomas and Sarcomas
   in Mice  with 0-Aminoazotoluene.  J.  Natl.  Cancer Inst.  10:927-941.

 Andervont, H.  B.  1958.   Induction of Hepatomas  in Strain  C3H Mice with
   4-0-Tolylazo-O-Toluidine  and Carbon Tetrachloride.   J. Natl. Cancer
   Inst. 20:431-438.

 Andervont, H. B., and  T.  B.  Dunn.  1962.   Occurrence  of Tumors in Wild
   House Mice.  J. Natl. Cancer Inst.  28:1153-1163.

 Andervont, H. B., H. G. Grady,  and J.  E.  Edwards.  1942.   Induction of
   Hepatic  Lesions, Hepatomas,  Pulmonary  Tumors and Hemangio-endotheliomas
   in Mice  with 0-Aminoazotoluene.  J.  Natl.  Cancer Inst.  3:131-153.

 Andervont, H. B., and M.  B.  Shimkin.   1940.  Biologic  Testing of Carcino-
   gens.  II.  Pulmonary-Tumor-Induction  Technique.  J.  Natl.  Cancer Inst.
   1:225-239.

Andreescheva, N.  1969.  Characteristics  and Criteria  of  the Toxic
  Effects  of Certain Nitro  and Amino Derivatives of Benzene.   Hyg.
  Sanit. 35(4):51-56.

-------
                                  389

Andrianova, M.  1971.  Transplacental Action of 3-Methylcholanthrene and
  Benz(a)pyrene on Four Generations of Mice.  Bull.  Exp.  Biol. Med. (USSR)
  71(1):677-680.

Argus, M. F., and C. Hoch-Ligeti.  1963.   Induction of Malignant Tumors
  in the Guinea Pig by Oral Administration of Diethylnitrosamine.  J.
  Natl. Cancer Inst. 30:533-551.

Arnold, D., S. Charbonneau, Z. Zawidzka,  and H. Grice.  1977.  Monitoring
  Animal Health During Chronic Toxicity Studies.  J. Environ. Pathol.
  Toxicol. 1:227-239.

Axelrad, A., and H. van der Gaag.  1962.   Susceptibility to Lymphoma
  Induction by Gross1 Passage A Virus in C3Hf/Bi Mice of Different Ages:
  Relation to Thymic Cell Multiplication and Differentiation.  J. Natl.
  Cancer Inst. 28:1065-1094.

Baba, T., and S. Takayama.  1961.  Influence of p-Dimethylamine-Azobenzene
  (DAB) Pretreatment in Neonatal Stage on the DAB Heptacarcinogenesis in
  Adult Rats, with Special Reference to Sex Difference in the Carcino-
  genesis.  Gann 52:73-82.

Barnes, J., and F. Denz.  1954.  Experimental Methods in Determining
  Chronic Toxicity.  Pharmacol. Rev. 6:191-242.

Beckers, A., and H. Bazin.  1978.  Incidence of Spontaneous Ileocecal
  Immunocytomas in Hybrids of LOU/C Rats and Rat Strains with Low Spon-
  taneous Tumor Incidence (brief communication).  J. Natl. Cancer Inst.
  60(6):1505-1508.

Benitz, K. F.  1970.  Measurement of Chronic Toxicity.  In:  Methods in
  Toxicology, G. E. Paget, ed.  F. A. Davis Company, Philadelphia.
  pp. 82-131.

Berenblum, I., L. Boiato, and N. Trainin.  1966.  On  the Mechanism of
  Urethane Leukemogenesis in Newborn C57BL Mice.  II.  Influence of Thy-
  mectomy and of Subsequent Thymus Reimplantation. Cancer Res. 26:361-363.

Bhisey, R. A., and S. M. Sirsat.  1975.  Histological Changes in the Mouse
  Skin After One Subcutaneous Injection of 20-Methylcholanthrene.  Indian
  J. Cancer 12:430-438.

Bielschowsky, F., and W. S. Bullough.  1949.  Epidermal Mitotic Activity
  and the Induction of Skin Tumors in Mice. Br. J. Cancer 3:282-285.

Bonser, G. M., E. Boyland, E. R. Busby, D. B. Clayson, P. L.  Grover, and
  J. W. Jull.  1963.  A Further  Study of Bladder  Implantation in  the Mouse
  as the Means of Detecting Carcinogenic Activity:  Use of Crushed Paraf-
  fin Wax or  Stearic Acid as  the Vehicle.  Br.  J. Cancer  17:127-136.

Bonser, G. M., D. B. Clayson, J. W. Jull, and L.  N. Pyrah.   1952.  The
  Carcinogenic Properties of  2-Amino-l-Naphthol Hydrochloride and Its
  Parent Amine 2-Naphthylamine.  Br. J. Cancer  6:412-424.

-------
                                   390

Boorman, G. A., and C. F. Hollander.  1974.  High Incidence of Spontane-
  ous Urinary Bladder and Ureter Tumors in the Brown Norway Rat  (brief
  communication).  J. Natl. Cancer Inst. 52(3):1005-1008.

Boyd, E.   1968.  Predictive Drug Toxicity:  Assessment of Drug Safety
  Before Human Use.  Can. Med. Assoc. J. 98:278-293.

Braun, W., L. Sung, D. Keyes, and R. Kociba.  1977.  Pharmacokinetic and
  Toxicological Evaluation of Dogs Fed 1,2,4,5-Tetrachlorobenzene in the
  Diet for Two Years.  Toxicol. Appl. Pharmacol. 41:218-219.

Bridges, B. A.  1976.  Short Term Screening Tests for Carcinogens.  Nature
  261:195-200.

Brune, H.F.K.  1977.  Experimental Results with Percutaneous Applications
  of Automobile Exhaust Condensates  in Mice.  In:  Air Pollution and Cancer
  in Man,  U. Mohr, D. Schmall, and L. Tomatis, eds.  International Agency
  for Research on Cancer, Lyon, France,  pp. 41-47.

Brusick, D. J.  1978.  The Role of Short-Term Testing in Carcinogen Detec-
  tion.  Chemosphere No. 5:403-417.

Burek, J.  D., and C. F. Hollander.   1977.  Incidence Patterns of Sponta-
  neous Tumors in BN/Bi Rats.  J. Natl. Cancer Inst. 58(1):99-105.

Butler, W. H.  1979.  Report of the  Workshop on Carcinogenicity.  J.
  Toxicol. Environ. Health 5:161-162.

Cameron, G. R., and K. K. Cheng.  1951.  Failure of Oral DDT to  Induce
  Toxic Changes in Rats.  Br. Med. J. 2:819-821.

Campbell,  J. S., Y. H. Yang, and J.  D. Bolton.  1965.  Murine Lung Ade-
  nomas Induced by Intravaginal Carcinogen.  Arch. Path. 79:588-594.

Cardesa, A., P. Pour, J. Althoff, and U. Mohr.  1974.  Effects of Intra-
  peritoneal Injections of Dimethyl- and Diethylnitrosamine, Alone or
  Simultaneously on Swiss Mice.  Z.  Krebscorsch. 82:233-238.

Carpanini, F., K. Butterworth, I. Gaunt, I. Kiss, P. Grasso, and S.
  Gangoli.  1978.  Long-Term Toxicity Studies on Chocolate Brown HT in
  Rats.  Toxicology 11:303-307.

Case, M., J. Smith, and R. Nelson.   1978.  Acute Mouse and Chronic Dog
  Toxicity Studies in Danthron Dioctyl Sodium Sulfosuccinate, Poloxalkol
  and Combinations.  Drug. Chem. Toxicol. 1(1):89-101.

Chieco-Bianchi, L., G. De Benedictis, G. Tridente, and L. Fiore-Donati.
  1963.  Influence of Age on Susceptibility to Liver Carcinogenesis and
  Skin Initiating Action by Urethane in Swiss Mice.  Br. J. Cancer 17:
  672-680.

-------
                                   391

Clapp, N. K.,  R. L.  Tyndall,  and J.  A. Otten.   1971.   Differences in
  Tumor Types and Organ Susceptibility in BALB/c and RF Mice Following
  Dimethylnitrosamine and Diethylnitrosamine.   Cancer Res.  31:196-198.

Clark, D.  1977.  Long-Term Inhalation Toxicology Studies.   In:  Current
  Approaches in Toxicology, B. Ballantyne, ed.   John Wright and Sons,
  Bristol,  pp. 105-114.

Coate, W., W.  Schoenfisch, T. Lewis, and W.  Busey.   1977.   Chronic Inhala-
  tion Exposure of Rats, Rabbits, and Monkeys  to 1,2,4-Trichlorobenzene.
  Arch. Environ. Health 32(6):249-255.

Crocker, T. T., J. E. Chase,  S. A. Wells, and  L. L.  Nunes.   1970.  Prelim-
  inary Report on Experimental Squamous Carcinoma of the Lung in Hamsters
  and in a Primate.   In:  Morphology of Experimental Respiratory Carcino-
  genesis, P.  Nettesheim, M.  G. Hanna, Jr.,  and J.  W. Deatheridge, Jr.,
  eds.  Oak Ridge, Tenn., USAEC Division of Technical Information.
  pp. 317-328.

Dale, M. M. , G. C. Easty, R.  Tchao,  H. Desai,  and M.  Andjargholi.  1973.
  The Induction of Tumors in the Guinea Pig with Methylcholanthrene and
  Diethylnitrosamine and Their Propogation In  Vivo and In Vitro.  Br. J.
  Cancer 27:445-450.

Davies, R. F., I. R. Major, and E. R. Aberdeen.  1970.  Pulmonary Adeno-
  mata Induced by Carcinogen Treatment in Organ Culture.  Influence of
  Increasing Amounts of Carcinogen.   Br. J.  Cancer 24:785-787.

Davies, R. F., I. R. Major, and E. R. Aberdeen.  1971.  The Induction of
  Adenomata in Mouse Lung Homografts by Chemical Carcinogens. Br. J.
  Cancer 25:565-567.

Davis, B. R.,  J. K.  Whitehead, M. E. Gill, P.  N. Lee, A. D. Butterworth,
  and F.J.R. Roe.  1975.  Response of Rat Lung to 3,4-Benzpyrene Adminis-
  tered by Intratracheal Instillation in Infusine With or Without Carbon
  Black.  Br.  J. Cancer 31:443-452.

Davis, K., A.  Nelson, R. Zwickey, W. Hansen, and 0. Fitzhugh.   1966.
  Chronic Toxicity of Ponceau SX to Rats, Mice, and Dogs.  Toxicol.
  Appl. Pharmacol. 8:306-317.

De Benedictis, G., G. Maiorano, L. Chieco-Bianchi, and L. Fiore-Donati.
  1962.  Lung Carcinogenesis by Urethane in Newborn,  Suckling,  and Adult
  Swiss Mice.   Br. J. Cancer 16:686-689.

Deichmann, W.  B., M. Keplinger, F. Sala, and E. Glass.  1967.   Synergism
  Among Oral Carcinogens.  IV.  The Simultaneous Feeding of  Four Tumori-
  gens to Rats.  Toxicol. Appl. Pharmacol. 11:88-103.

Deichmann, W.  B., and J. L. Radomski.   1963.   The Carcinogenicity and
  Metabolism of  2-Naphthylamine and Related Compounds.  Ind.  Med.  Surg.
  32:161.

-------
                                    392

 Deichmann,  W.  B.,  J.  L.  Radomski,  E.  Glass,  W.A.D.  Anderson,  M.  M.  Coplan,
   and F.  M.  Woods.   1965.   Synergism Among Oral Carcinogens.  III.   Simul-
   taneous Feeding  of Four Bladder  Carcinogens to Dogs.   Ind.  Med.  Surg.
   34:640-649.

 Delia Porta, G.   1968.   Use of Newborn and Infant Animals in  Carcinogenic-
   ity Testing.   Food Cosmet.  Toxicol.  6:243-252.

 Delia Porta, G., L.  Kolb,  and P.  Shubik.   1958.  Induction of Tracheobron-
   chial Carcinomas in the Syrian Golden Hamster.  Cancer Res. 18:592-597.

 Delia Porta, G., M.  Rappaport, U.  Saffiotti, and P.  Shubik.   1956.   Induc-
   tion of Melanotic  Lesions During Skin Carcinogenesis  in Hamsters.   Arch.
   Pathol.  61:303-313.

 Delia Porta, G., and B.  Terracini.   1969.   Chemical Carcinogenesis  in
   Infant  Animals.  Prog.  Exp.  Tumor Res.  11:334-363.

 DiPaolo,  J.  A.,  R. L.  Nelson,  P. J.  Donovan, and C.  H.  Evans.  1973.   Host
   Mediated In Vivo - In  Vitro Assay for Chemical Carcinogenesis.  Arch.
   Pathol.  95:380-385.

 Diwan,  B.  A.,  and  H.  Meier.   1974.   Strain- and Age-Dependent Transplacental
   Carcinogenesis by  1-Ethyl-l-Nitrosourea in Inbred Strains of Mice.
   Cancer  Res.  34:764-770.

 Doell,  R. ,  and W.  Games.   1962.   Urethane Induction of Thymic Lymphoma
   in  C57BL Mice.   Nature 194:588-589.

 Drozdz, M.,  E. Kucharz,  and M. Kaminski.   1977.  Histopathological  and
   Histochemical  Studies  of the Skin of Guinea Pigs  After Long-Term  Expo-
   sure  to  Nitrogen Dioxide.   Exp.  Pathol.  Board 14:162-170.

 Durham, W.  F., P.  Ortega,  and  W. J.  Hays,  Jr.   1963.  The Effect  of Various
   Dietary  Levels of  DDT  on Liver Function, Cell Morphology and DDT  Storage
   in  the Rhesus Monkey.  Arch.  Int.  Pharmacodyn.  141:111-129.

 Dyer, H., M. Kelly,  and  R.  O'Gara.   1966.   Lack of  Carcinogenic Activity
   and Metabolic Fate  of  Fluorenylacetamide in Monkeys.   J. Natl.  Cancer
   Inst. 36:305-322.

 Epstein, S., J. Andrea,  and H.  Jaffe.   1967.   Carcinogenicity of  the
  Herbicide  Maleic Hydrazide.   Nature  (London)  215:1388-1390.

 Fancher, 0.  1978.   Species Selection  and  Animal Models for Toxicologic
  Studies.   Clin. Toxicol.  12(2):239-247.

Farber, E.   1963.  Ethionine Carcinogenesis.   Adv.  Cancer Res.  7:383-474.

Fare, G.  1966.  Rat Skin  Carcinogenesis by  Topical  Applications  of Some
  Azo Dyes.  Cancer Res. 26:2406-2408.

-------
                                  393

Federal Register.  1978.   Proposed Guidelines for Registering Pesticides
  in the U.S. Hazard Evaluation:   Humans and Domestic Animals.  43FR(163):
  37336-37403.

Federal Register.  1979.   Proposed Health Effects Tests Standards for
  Toxic Substances Control Act Test Rules.   44FR(91):27334-27375.

Flaks, A.  1965.  The Effect of 9,10-Dimethyl-l,2-Benzanthracene on Young
  Mice of Low and High Cancer Strain.   Br.  J. Cancer  19:547-550.

Flaks, A.  1966.  Test for Carcinogenesis of Cigarette Tobacco Smoke
  Condensate Using Young Strong A and C57BL Mice.  Br. J.  Cancer
  20:145-147.

Food Safety Council.  1978.  Proposed System for Food Safety Assessment.
  Food Cosmet. Toxicol. 16, Suppl. 2:1-136.

Fox, J. G.  1977.  Clinical Assessment of Laboratory  Rodents on Long-Term
  Bioassay Studies.  J. Environ.  Path, and Toxicol. 1:199-226.

Frankel, H., R. Yamamoto, E. Weisburger, and J.  Weisburger.   1970.  Chronic
  Toxicity of Azathioprine and the Effect of this Immuno-suppressant on
  Liver Tumor Induction by the Carcinogen tf-Hydroxy-tf-2-Fluorenylacetamide.
  Toxicol. Appl. Pharmacol. 17:462-480.

Friedman, L.  1973.  Problems of Evaluating the Health Significance of the
  Chemicals Present in Foods.  In:  Pharmacology and  the Future of Man,
  Proc. 5th Int. Congr. Pharmacology,  San Francisco,  1972, Vol. 2.  Karger,
  New York.  pp. 30-41.

Fugmann, R. A., J. C. Anderson, R. L.  Stolfi, and D.  S. Martin.  1977.
  Comparison of Adjuvant Chemotherapeutic Activity Against Primary and
  Metastatic Spontaneous Murine Tumors.  Cancer Res.  37:496-500.

Futami, T., Y. Samata, T. Tsuda, and F. Masuda.   1977.  Toxicity Study on
  Sodium Polyalkylarylsulfonate (Na-PAAS) in Rats:  II.  Chronic Toxicity
  Study.  Oyo Yakuri 13(1):147-159.

Gage, J.  1970.  Experimental Inhalation Toxicity.  In:  Methods in Toxi-
  cology, G. Paget, ed.  F. A. Davis Company, Philadelphia,  pp. 258-277.

Gak, J. C., C. Graillot, and R. Truhaut.  1976.   Use of the Golden Hamster
  in Toxicology  (Part II of two parts).  Lab. Anim. Sci. 26(2):274-280.

Gargus, J., 0. Paynter, and W. Reese, Jr.  1969.  Utilization  of Newborn
  Mice in the Bioassay of Chemical Carcinogens.  Toxicol. Appl. Pharmacol.
  15:552-559.

Gilbert, C., J. Gillman, P. Loustalot, and W. Lutz.   1958.  The Modifying
  Influence of Diet and the Physical Environment on  Spontaneous Tumor
  Frequency in Rats.  Br. J. Cancer 12:565-593.

-------
                                   394

Giurgea, M.   1977.  Peracetam:   Toxicity  and  Reproduction  Studies.  Farmaco
  Ed. Prat.  32:48-52.

Glucksmann,  A.,  and C.  Cherry.   1971.   Sex  Difference  and  Carcinogenic
  Dosage in  the  Induction  of Neoplasms  in the Salivary Glands of Rats.
  Br. J. Cancer  25:212-224.

Goerttler, K., and H. Lohrke.   1977.  Diaplacental  Carcinogenesis:  Tumor
  Localization and Tumor Incidence  in NMRI  Mice After  Diaplacental  Initia-
  tion with  DMBA and Urethane and Postnatal Promotion  with the  Phorbol
  Ester TPA  in a Modified  2-Stage Berenblum/Mottram Experiment.  Virchos
  Arch. A. Path.  Anat.  and Histol.  376:117-132.

Golberg, L.,  ed.  1974.  Carcinogenesis Testing of  Chemicals.   The  Chemical
  Rubber Company, Cleveland, Ohio.   144 pp.

Golberg, L.   1975.  Safety Evaluation Concepts.  J.  Assoc.  Off. Anal. Chem.
  58(4):635-644.

Goldenthal,  E.   1968.   Current  Views on Safety Evaluation  of Drugs.  U.S.
  Food and Drug  Administration  Papers May:13-18.

Goldwater, L. J., A. J. Rosso,  and  M. Kleinfeld.  1965.  Bladder Tumors
  in a Coal  Tar  Dye Plant.  Arch. Environ.  Health 11:814-817.

Gorrod, J.,  R. Carter,  and F. Roe.   1968.   Induction of Hepatomas by 4-
  Aminobiphenyl  and Three  of Its Hydroxylated Derivatives  Administered
  to Newborn Mice.  J.  Natl. Cancer Inst. 41:403-410.

Graffi, A.,  F. Hoffman, and M.  Schutt.  1967. tf-Methyl-tf-Nitrosourea
  As a Strong Topical Carcinogen When Painted on the Skin  of Rodents.
  Nature (London) 214:611.

Grahn, D., and K. F. Hamilton.   1964.   Influence of Sex, Environment,
  and Radiation  Factors on Life Shortening  and Tumor Incidence  in C3H
  Mice.  Radiat.  Res. 22:191.

Grasso, P.   1976.  Review  of Tests  for  Carcinogenicity and Their Signif-
  icance to  Man.  Clin. Toxicol. 9:745-760.

Gray, J., R.  Weaver, J. Bollert, and E. Feenstra.   1972.   The Oral  Toxicity
  of Clindamycln in Laboratory  Animals.   Technol. Appl. Pharmacol.
  21:516-531.

Greenman, D.   L.,  and R. R. Delongchamp.   1979.  Variability of  Response
  to Diethylstilbesterol:  A Comparison of  Inbred with Hybrid Mice.  J.
  Toxicol. and Environ. Health  5:131-143.

Greenman, D., R. Delongchamp, and B.  Highman.  1979.   Variability of
  Response to Diethylstilbestrol:   A Comparison of  Inbred  with  Hybrid
  Mice.   J.  Toxicol. Environ. Health 5:131-143.

-------
                                   395

Greenwald, P., J. Barlow, P. Nasca, and W.  Burnett.   1971.  Vaginal Cancer
  After Maternal Treatment with Synthetic Estrogens.   New Engl. J. Med.
  285:390-392.

Griesemer, R. A., P. Nettesheim, D. H.  Martin, and J. E. Caton, Jr.  1977.
  Quantitative Exposure of Grafted Rat  Tracheas to 7,12-Dimethylbenz(a)-
  anthracene.  Cancer Res. 37:1266-1271.

Griswold, D. P., A. E. Casey, E. K. Weisburger, and J. H. Weisburger.
  1968.  The Carcinogenicity of Multiple Intragastric Doses of Aromatic
  and Heterocyclic Nitro or Amino Derivatives in Young Sprague-Dawley
  Rats.  Cancer Res. 28:924-933.

Gruenstein, M., D. R. Meranze, and M. B. Shimkin.  1966.  Mammary, Seba-
  ceous, and Cutaneous Neoplasms and Leukemia in Male Wistar Rats Receiv-
  ing Repeated Gastric Instillations of 3-Methylcholanthrene.   Cancer Res.
  26:2202-2205.

Haag, H., J. Finnegan, P. Larson, M. Dreyfuss, R. Main, and W. Riese.
  1948.  Comparative Chronic Toxicity for Warm-Blooded Animals of 2,3-
  bis-(p-chlorophenyl)-l,l,l-trichloroethane  (DDT).   Ind. Med. 17:477-484.

Haun, C.  1975.  Toxicity of High Density Jet Fuel Components.  AMRL-TR-
  75-125, Aerospace Medical Division, Wright-Patterson Air Force Base,
  Ohio.

Hayes, W.  1967.  Toxicity of Pesticides to Man:  Risks from Present
  Levels.  Proc. R. Soc. London Ser. B. 167:101-127.

Herbst, A., L. Ulfelder, and D. Poscanzer.  1971.  Adenocarcinoma of the
  Vagina.  Association of Maternal Stilbestrol Therapy with Tumor Appear-
  ance in Young Women.  New Engl. J. Med. 284:878-881.

Herrold, K. M.  1964<2.  Induction of Olfactory Neuroepithelial Tumors  in
  Syrian Hamsters by Diethylnitrosamine.  Cancer 17:114-121.

Herrold, K.  1964£>.  Effect of Route of Administration  on  the  Carcinogenic
  Action of Diethylnitrosamine.  Br. J. Cancer 18:763-767.

Herrold, K.  1969.  Aflatoxin Induced Lesions  in Syrian Hamsters.   Br.  J.
  Cancer 23:655-660.

Herrold, K. M., and L. J. Dunham.  1963.  Induction  of  Tumors  in  the
  Syrian Hamster with Diethylnitrosamine  (#-Nitrosodiethylamine).
  Cancer Res. 23:773-777.

Heston, W. E.   1958.  Mammary Tumors in Agent-Free Mice.   Ann. N.Y.
  Acad.  Sci.  71:931-942.

Heston, W. E.,  and  G. Vlahakis.  1966.  Factors  in  the Causation of
  Spontaneous Heptaomas  in Mice.   J. Natl.  Cancer  Inst. 37:839-843.

-------
                                   396

Heston, W.  E.,  and G.  Vlahakis.   1968.   CSH-A^ -  A High Hepatoma and
  High Mammary  Tumor Strain of Mice.   J.  Natl.  Cancer  Inst.  40:1161-1166.

Heston, W.  E.,  G.  Vlahakis,  and  M.  K.  Deringer.  1960.   High Incidence
  of  Spontaneous Hepatomas and the  Increase of  This Incidence with
  Urethane  in C3H,  C3Hf,  and C3He Male Mice.  J. Natl.  Cancer Inst.
  24:425-435.

Heuper, W.  C.,  and H.  D.  Wolf.   1937.   Experimental Production of Aniline
  Tumors  of Dogs in the Bladder.  Amer.  J.  Pathol.  13:656-657.

Hiaro, F.,  T. Fujisawa, E.  Tsubura,  K.  Akamatsu, Y. Yamamura.   1968.
  Experimental  Lung Cancer in Rabbits  Induced by Chemical Carcinogens.
  Gann 59:497-505.

Hoag, W.  G.   1963.   Spontaneous  Cancer in Mice.  Ann.  N.Y. Acad.  Sci.
  108:805-831.

Hodge, H.,  A. Boyce,  W. Deichmann,  and H. Kraybill. 1967.   Toxicology
  and No-Effect Levels of Aldrin and Dieldrin.   Toxicol.  Appl.  Pharmacol.
  10:613-675.

Holland,  J.  M., D.  G.  Gosslee, and  N.  J.  Williams.   1979.  Epidermal
  Carcinogenicity  of  Bis(2,3-epoxycyclopentyl)ether, 2,2-Bis(p-glyci-
  dyloxyphenyl) propane and m-Phenylenediamine  in  Male and Female C3H
  and C57 BL/6  Mice.   Cancer Res. 39:1714-1721.

Homburger,  F.,  and J.  R.  Baker.   1969.   Accelerated Carcinogen Testing.
  Progr.  Exp. Tumor Res.,  Vol. 11.   Karger,  New York.   pp. 384-394.

Homburger,  F.,  and S.  S.  Hsuch.   1970.   Rapid Induction of Subcutaneous
  Fibrosarcoma  by  7,12-Dimethylbenz(a)anthracene in an Inbred Line of
  Syrian Hamsters.   Cancer Res.  30:1449-1452.

Homburger,  F.,  S.  Hsuch,  C.  S. Kerr, and A.  B.  Russfield.  1972.   Inher-
  ited Susceptibility of  Inbred  Strains  of  Syrian  Hamsters to Induction
  of Subcutaneous  Sarcomas and Mammary and  Gastrointestinal  Carcinomas
  by Subcutaneous  and Gastric Administration of  Polynuclear  Hydrocarbons.
  Cancer Res. 32:360-366.

Homburger,  F., A.  B.  Russfield.   1970.   An  Inbred  Line of Syrian  Hamsters
  with Frequent Spontaneous  Adrenal  Tumors.   Cancer Res.  30:305-308.

Homburger,  F.,  and  A.  Tregier.   1960.  Modifying Factors  in  Carcinogen-
  esis.  Prog.  Exptl.  Tumor  Res.  1:311.

Howell, J.   1963.   The Experimental  Production  of  Vascular Tumors in the
  Rat.  Br.  J. Cancer  17:663-671.

Huggins,  C., L.  C.   Grand,  and F.  P.  Brillantes.  1961.  Mammary Cancer
  Induced by a Single  Feeding of  Polynuclear  Hydrocarbons and Its Sup-
  pression.  Nature 189:204-207.

-------
                                  397

Huggins, C.,  L. Grand, and R. Fukunishi.   1964.   Aromatic Influences on
  the Yields of Mammary Cancers Following Administration of 7,12-Dimethyl-
  benz(a)anthracene.  Proc. Natl.  Acad.  Sci.,  Vol.  51.   pp. 737-742.

Ihle, J. N.,  L. 0. Arthur, and D.  L. Fine.  1976.  Autogenous Immunity
  to Mouse Mammary Tumor Virus in Mouse Strains of High and Low Mammary
  Tumor Incidence.  Cancer Res. 36:2840-2844.

Innes, J.R.M., B. M. Ulland, M. G. Valeric, L. Petrucelli, L. Fishbein,
  E. R. Hart, A. J. Pallotta, R. R.  Bates, H.  L. Falk,  T. J. Gart, M.
  Klein, I. Mitchell, and J. Peters.  1969.  Bioassay of Pesticides and
  Industrial Chemicals for Tumorigenicity in Mice.   J.  Natl. Cancer Inst.
  42:1101-1114.

Ivankovic, S.  1973.  Experimental Prenatal Carcinogenesis.  In:  Trans-
  pi -icental Carcinogenesis, L. Tomatis and U.  Mohr, eds.  Scientific
  Publication No. 4, International Agency for Research on Cancer, Lyon,
  France,  pp. 92-99.

Jalanko, H.,  I. Virtanen, E. Engvall, and E.  Ruoslahti.  1978.   Early
  Increase of Serum Alpha-Fetoprotein in Spontaneous Hepatocarcinogenesis
  in Mice.  Int. J. Cancer 21(4):453-459.

Jull, J. W.  1951.  The Induction of Tumors of the Bladder Epithelium in
  Mice by the Direct Application of a Carcinogen.  Br.  J. Cancer 5:328-330.

Jungherr, E.   1963.  Spontaneous Cancer in Zoo and Laboratory Animals.
  Part II.  Tumors and Tumor-Like Conditions in Monkeys.  Ann.  N.Y. Acad.
  Sci. 108:777-792.

Kadota, T., H. Kohda, and J. Miyamoto.  1975.   Subchronic Toxicity  Studies
  of Sumithion, Sumioxon, and p-Nitrocresol in Rats and 92-Week Feeding
  Study of Sumithion with Special Reference to Change of Cholinesterase
  Activity.  Botyu-Kagaku 40:38-48.

Kaiser, J.  1964.  A One-Year Study of the Toxicity of Ethambutol in Dogs:
  Results During Life.  Toxicol. Appl. Pharmacol. 6:577-567.

Kaplan, A., and H. Sherman.  1977.  Toxicity Studies with Methyl-ff-
  [(Methyllamine-carbonyl)oxyl]-ethanimidothioate.  Toxicol. Appl.
  Pharmacol.  40:1-17.

Kaye, A., and N. Trainin.  1966.  Urethan Carcinogenesis  and Nucleic
  Acid Metabolism:  Factors  Influencing Lung Adenoma Induction.   Cancer
  Res. 26:2206-2212.

Kello, D., and K. Kostial.   1973.   The Effect of Milk  Diets  on  Lead Metab-
  olism in Rats.  Environ. Res. 6:355-360.

Kelly, M., and R. O'Gara.  1961.  Induction of  Tumors  in Newborn Mice
  with Dibenz(a)anthracene and  3-Methylcholanthrene.   J.  Natl.  Cancer
  Inst. 26:651-679.

-------
                                   398

Kelly, M. G., R. W. O'Gara,  R.  H. Adamson,  K.  Gadekar,  C.  C. Botkin,
  W. H. Reese, Jr., and W. T. Kerber.   1966.   Induction of Hepatic  Cell
  Carcinomas  in Monkeys with tf-Nitrosodiethylamine.   J.  Natl. Cancer
  Inst. 36:323-351.

Ketkar, M., G. Reznik, P.  Schneider,  and  U. Mohr.   1978.   Investigations
  on the Carcinogenic Burden by Air Pollution  in Man.   Intratracheal
  Instillation Studies with  Benzo(a)pyrene  in  Bovine  Serum Albumin  in
  Syrian Hamsters.  Cancer Lett.  4:235-239.

Kitagawa, K., M. Wakakura, and  S. Ishikawa.  1977.  Light  Microscopic
  Study of Endocrine Organs  of  Rats Treated by Carbamate Pesticide.  J.
  Toxicol. Sci. 2:53-60.

Klein, M.  1951.  The Action of Croton  Oil  in  the  Induction of  Sarcomas
  in Mice.  J. Natl. Cancer  Inst. 11:843-848.

Klein, M.  1952.  The Action of Methylcholanthrene, Croton Oil,  and 1,2-
  Benzanthracene in the Induction of  Skin Tumors in Strain DBA  Mice.  J.
  Natl. Cancer Inst. 12:735-742.

Klein, M.  1953.  Effects  of Croton Oil on  Induction  of Tumors  by 1,2-
  Benzanthracene, Deoxycholic Acid, or  Low  Doses of 20-MCA in Mice.
  J. Natl. Cancer Inst. 13:333.

Klein, M.  1963.  Susceptibility  of Strain  BLoAFi/J Hybrid Infant Mice
  to Tumorigenesis with 1,2-Benzanthracene, Deoxycholic Acid, and 3-
  Methylcholanthrene.  Cancer Res. 23(6):1701-1707.

Kociba, R., S. McCollister,  C.  Park,  T. Torkelson,  and  P.  Gehring.  1974.
  1,4-Dioxane Toxicity as  Determined  by a Two-Year Dose-Response Study
  in Rats.  Toxicol. Appl. Pharmacol. 29:86.

Kociba, R., B. Schwetz, D. Keyes, G.  Jersey, J. Ballard, D. Dittenber,
  J. Quast, C. Wade, and C.  Humiston.   1977.   Chronic Toxicity  and  Repro-
  duction Studies of Hexachlorobutodiene  in Rats.   Environ. Health
  Perspect. 21:49-53.

Koelle, G., and A. Gilman.   1946.  The  Chronic Toxicity of Diisopropyl
  Fluorophosphate (DPF) in Dogs,  Monkeys, and  Rats.   J.  Pharmacol.  Exp.
  Ther. 87:435-448.

Koss, G., S. Seubert, A. Seubert, W.  Koransky,  and H. Ippen.  1978.
  Studies on the Toxicology  of  Hexachlorobenzene.   III.  Observations
  in a Long-Term Experiment.  Arch. Toxicol. 40:285-294.

Krasovskii, G.  1976.  Extrapolation  of Experimental  Data  from  Animals
  to Man.  Environ. Health Perspect.  13:51-58.

Kroes, R., G. M. Williams, and  J. H.  Weisburger.   1973.  Early  Appearance
  of Serum a-Fetoprotein as  a Function  of Dosage of Various Hepatocarci-
  nogens.  Cancer Res. 33:613-617.

-------
                                  399

Kuschner, M., S. Laskin, E. Cristofano,  and N.  Nelson.   1957.  Experi-
  mental Carcinoma of the Lung.   In:  Proceedings of the Third National
  Cancer Conference.  Lippincott, Philadelphia,  pp. 485-495.

Laishes, B., and E. Farber.  1978.  Transfer of Viable Putative Preneo-
  plastic Hepatocytes to the Livers of Syngenic Host Rats.   J. Natl.
  Cancer Inst. 61(2):507-512.

Laishes, B. A., L. Fink, and B.  I. Carr.  1980.  A Liver Colony Assay for
  a New Hepatocyte Phenotype as a Step Towards  Purifying New Cellular
  Phenotypes That Arise During Hepatocarcinogenesis.  Ann.  N.Y. Acad.
  Sci. (in press).

Lawrence, W.  1976.  Systemic Toxicity.   Acute  vs. Subchronic and Chronic.
  Drug. Cosraet. Ind. November 1976:40-114.

Laws, J. 0., and A. Flaks.  1966.  Pulmonary Adenomata Induced by Carci-
  nogen Treatment in Organ Culture.  Br. J. Cancer 20:550-554.

Lee, C., J. Bhandari, J. Winston, W. House., P. Peters, R.  Dixon, and
  J. Woods.  1977.  Inhalation Toxicity of Vinyl Chloride and Vinylidene
  Chloride.  Environ. Health Perspect. 21:25-32.

Lee, C-C., J. Q. Russell, and J. L. Minor.  1978.  Oral Toxicity of Fer-
  ric Dimethyl-Dithiocarbamate (Ferbam)  and Tetramethylthiuram Disulfide
  (Thiram) in Rodents.  J. Toxicol. Environ. Health 4:93-106.

Lee, K., B. Toth, and P. Shubik.  1963.   Carcinogenic Response of the
  Syrian Golden Hamster Treated at Birth with 7,12-Dimethylbenz(a)-
  anthracene.  Proc. Soc. Exptl. Biol. Med. 114:579-582.

Levinsky, H., H. MacFarland, B. Procter, G. Rona, and A. Blair.  1978.
  A Study of the Chronic Toxicity of Inhaled Disodium Cromoglycate  in
  the Squirrel Monkey.  Toxicol. Appl. Pharmacol. 45:141-153.

Levy, B. M.  1963.  Induction of Fibrosarcoma  in  the Primate,  Tamarinus
  nigvioollis.  Nature  200:182-183.

Liebelt, R., A. Liebelt, and M. Lane.   1964.  Hormonal Influences on
  Urethan Carcinogenesis in C3H/f Mice.  Cancer Res. 24:1869-1879.

Liebelt, R. , R. Yoshida, and G. Gray.   1961.   Enhancement of  Liver  Tumor-
  igenesis  in Zb Mice Injected with Urethan at Newborn Age.   Proc.  Assoc.
  Cancer Res. 3:245.

Lijinsky, W., A.  Ferrero, R. Montesano, and C.E.M.  Wenyon.   1970.   Tumor-
  igenicity  of  Cyclic Nitrosamines  in Syrian Golden Hamsters.   Z.
  Krebsforsch.  74:185-189.

Lijinsky, W., K.  Lee, Y. Tomatis,  and W.  H. Butler.   1967.   Nitrosoazeti-
  dine — a  Potent Carcinogen of Low Toxicity.   Naturwissenschaften  54:518.

-------
                                   400

 Lijinsky, W.,  and H.  W.  Taylor.   1978.   Relative Carcinogenic Effective-
   ness of Derivatives of Nitrosodiethylamine in Rats.   Cancer Res.  38:
   2391-2394.

 Littlefield,  N.,  and R.  Kodell.   1979.   Influence of Genetic Population
   Structure on the Results of Chronic Toxicity Studies.   J.  Toxicol.
   Environ. Health 5:121-129.

 Loomis,  T.  1974.  Essentials of Toxicology, 2nd ed.,  Lea and Febiger,
   Philadelphia,   pp.  189-191.

 Luz,  A.   1977.  The Range of  Incidence of Spontaneous Neoplastic and  Non-
   Neoplastic Lesions of  the Laboratory Mouse.   Z. Versuchstierk.  19(6):
   342-343.

 Maekawa, A.,  and S. Odashima.  1975.   Spontaneous Tumors in  AC1/N Rats.
   J.  Natl. Cancer Inst.  55:1437-1445.

 Magee, P.  1970.   Tests  for Carcinogenic Potential.   In:  Methods in  Tox-
   icology, G.  Paget,  ed.  F.  A.  Davis Company, Philadelphia,  pp. 158-196.

 McCollister,  S.,  R. Kociba, C.  Humiston, D.  McCollister, and P.  Gehring.
   1974.   Studies  of the  Acute and Long-Term Oral Toxicity of Chlorpyrifos
   (p,o-Diethyl-o-(3,5,6-Trichloro-2-Pyridy1)Phosphorothioate).   Food
   Cosmet. Toxicol.  12:45-61.

 McNamara, B.   1976.  Concepts in Health Evaluation of Commercial and
   Industrial  Chemicals.   In:   Advances in Modern Toxicology, Part 1,  Vol.
   1,  M.  Mehlman,  R. Shapiro,  and H. Blumenthal, eds.   John Wiley and
   Sons,  New York.  pp. 61-140.

 Milievskaja,  I. L., and  N.  S. Kiseleva.   1976.  Comparative  Study of  the
   Carcinogenic Activities of  NAS and  Some Chemical Carcinogens When Intro-
   duced  into  the  Buccal  Pouch of the  Syrian Hamster.   Bull.  World Health
   Organ.  54:607-614.

 Miller,  E.  C., J. A.  Miller,  and M. Enomota.  1964.   The Comparative  Car-
   cinogenicities  of 2-Acetylaminofluorene and  Its #-Hydroxy  Metabolite  in
   Mice,  Hamsters  and  Guinea Pigs.  Cancer Res. 24:2018-2031.

 Ministry of Health  and Welfare Canada.   1975.   The Testing of Chemicals
   for Carcinogenicity, Mutagenicity,  and Teratogenicity.   Ministry of
   Health and Welfare, Ottawa.  183 pp.

 Misra, A.,  and S. Mule.   1977.   Severe  Toxicity and  Lethality in Some
  Monkeys Following Chronic Administration of  Z-a-Acetylmethadol (LAAM).
  Am.  J.  Drug. Alcohol Abuse  4(3):431-440.

Miyoshi,  K., and  S. Takauchi.  1977.  Chronic  Tellurium Intoxication  in
  Rats.   Folia Psychiatr. Neurol.  Jpn.  31(1):111-118.

-------
                                   401

Mohr, U.  1973.  Effects of Diethylnitrosamine on Fetal and Suckling Syrian
  Golden Hamsters.  In:  Transplacental Carcinogenesis, L. Tomatis and U.
  Mohr, eds.  Scientific Publication No. 4, International Agency for Research
  on Cancer, Lyon, France,  pp. 65-70.

Mohr, U., G. Reznik, and P. Pour.  1977.  Carcinogenic Effects of Diisopro-
  panolnitrosamine in Sprague-Dawley Rats.  J. Natl.  Cancer Inst. 58:361-366.

Molello, J., C. Gerbig, and V. Robinson.  1973.  Toxicity of [4,4'-(Iso-
  propylidenedithio)bis(2,6-di-t-butylphenol)], Probucol, in Mice, Rats,
  Dogs, and Monkeys:  Demonstration of a Species-Specific Phenomenon.
  Toxicol. Appl. Pharmacol. 24:590-593.

Morris, H. P., and W. H. Eyestone.  1953.  Tumors of the Liver and Urinary
  Bladder of Dogs After Ingestion of 2-Acetylaminofluorene.  J. Natl. Cancer
  Inst. 13:1139-1165.

Morris, H. P., and H. I. Firminger.  1956.  Influence of Sex and Sex Hor-
  mones on Development of Hepatomas and Other Hepatic Lesions in Strain
  AXC Rats Ingesting 2-Diacetylaminofluorene.  J. Natl. Cancer Inst.
  16:927-949.

Mulay, A. S., and R. W. O'Gara.  1959.   Incidence of Liver Tumors in Male
  and Female Rats Fed Carcinogenic Azo Dyes.  Proc. Soc. Exptl. Biol. Med.
  100:320-322.

Munro, I.  1977.  Considerations in Chronic Toxicity Testing:  The Chemical,
  the Dose, the Design.  J. Environ. Pathol. Toxicol. 1:183-197.

Nagasawa, H., R. Yanai, and I. Azuma.  1978.  Suppression by Noaardia rubra
  Cell Wall Skeleton of Mammary DNA Synthesis, Plasma Prolactin Level, and
  Spontaneous Tumorigenesis in Mice.  Cancer Res. 38:2160-2162.

National Academy of Sciences.  1975.  Principles for Evaluating Chemicals
  in the Environment.  National Academy of Sciences, Washington, D.C.
  pp. 115-153.

National Academy of Sciences.  1977.  Principles and Procedures for  Evalu-
  ating the Toxicity of Household Substances.  National Academy of Sciences,
  Washington, D.C.  130 pp.

National Institutes of Health.  1968-1973.   Survey of Compounds Which
  Have Been Tested for Carcinogen Activity.  Public Health Service Publi-
  cation No. 149, Government Printing Office, Washington,  D.C.

Neiman, J. M.  1968.  The  Sensitizing Carcinogenic Effect  of  Small Doses
  of Carcinogen.  Europ. J. Cancer 4:537-545.

Nelson, A. A., 0. G. Fitzhugh, and H. 0.  Calvery.  1943.   Liver  Tumors
  Following Cirrhosis Caused by  Selenium in  Rats.  Cancer  Res.  3:230-236.

-------
                                   402

Nelson,  A.  A.,  and G.  Woodard.   1953.   Tumors  of  the  Urinary  Bladder, Gall
   Bladder  and Liver in Dogs  Fed 0-Aminoazotoluene or  p-Dimethylaminoazo-
   benzene.   J.  Natl.  Cancer  Inst.  13:1497-1509.

Nettesheim,  P.,  and R.  A.  Griesemer.   1978.  Experimental Models  for
   Studies  of Respiratory Tract  Carcinogenesis.  In:   Pathogenesis and
   Therapy  of Lung Cancer,  Lung  Biology in  Health  and  Disease  10,  C. Harris,
   ed.  New York.   pp.  75-188.

Nettesheim,  P.,  and A.  S.  Hammons.   1971.   Induction  of  Squamous  Cell
   Carcinoma in  the Respiratory  Tract  of Mice.  J.  Natl.  Cancer  Inst.
   47:697-701.

Newberne,  P. M.,  and A.  Rogers.   1972.  Vitamin A, Liver, Colon Carcinoma
   in Rats  Fed Low Levels of  Aflatoxin.  Toxicol.  Appl. Pharmacol.  22:280.

Nishizuka,  Y.,  K.  Nakakuki,  and T.  Sakakura.   1964.   Induction  of Pulmonary
   Tumors and Leukemia by a Single  Injection of 4-Nitroquinoline 1-Oxide to
   Newborn  and Infant Mice.   Gann 55:495-508.

Noble, R.,  B. C.  Hochachka,  and D.  King.   1975.   Spontaneous  and  Estrogen-
   Produced  Tumors in Nb Rats and Their Behavior After Transplantation.
   Cancer Res. 35:766-780.

Norris,  J.,  R.  Kociba,  B.  Schwetz,  J.  Rose,  C. Humiston, L. Jewett, P.
   Gehring,  and  J.  Mailhes.   1975.   Toxicology  of  Octabromobiphenyl and
   Decabromodiphenyl Oxide.   Environ.  Health Perspect. 11:153-161.

Noyes, W. F.  1968.   Carcinogen Induced Sarcoma in the Primitive  Primate,
   Tupaia glis.   Proc.  Soc. Exp.  Biol.  Med.  127:594-596.

Noyes, W. F.  1969.   Carcinogen-Induced Neoplasia with Metastasis in a
   South  American Primate,  Saquinus  oedipus.  Proc. Soc.  Expt. Biol. Med.
   131:223-225.

O'Gara,  R. W.,  and M.  G. Kelly.   1965.  Pulmonary Arteriolitis  in Infant
  Monkeys Produced by  Subcutaneous  Injections  of  Polycyclic Hydrocarbons.
  Arch.  Path. (Chicago)  79:475-483.

Oppenheimer, B.  S.,  E.  T.  Oppenheimer,  and A.  P-  Stout.  1948.  Sarcomas
   Induced by Rats  by  Implanting  Cellophane.  Proc. Soc.  Expt. Biol. Med.
  67:33-34.

Outzen,  H.  C., R.  Custer,  R. Phillips,  G.  J. Eaton, and  R. T. Prehn.  1975.
  Spontaneous and  Induced  Tumor  Incidence  in Germ Free Nude Rats.  J.
  Reticuloendothel.  Soc. 17(1):l-9.

Page, N.  P.  1977a.  Concepts of a  Bioassay Program in Environmental
  Carcinogenesis.   In:  Advances in Modern Toxicology, Vol. 3:  Environ-
  mental Cancer, H. Kraybill and M. Mehlman, eds.  Hemisphere Publishing
  Corp.,  Washington, D.C.  pp. 87-171.

-------
                                  403

Page, N.  1977&.  Chronic Toxicity and Carcinogenicity Guidelines.  J.
  Environ. Pathol. Toxicol. 1:161-182.

Peacock, P. M.  1962.  A Short-Term Test for Carcinogenicity (The Effects
  of Certain Closely-Related Polycyclic Aromatic Hydrocarbons on Embryo
  Tissue Homografts in BALB/c Strain Mice).   Br. J.  Cancer 16:701-706.

Peacock, P. M., and E. Dick.  1963.   A Short-Term Test for Carcinogenicity
  (Mouse Embryo Tissue Homografts in BALB/c  Strain Mice).  Br.  J. Cancer
  17:59-61.

Peck, H.  1968.  An Appraisal of Drug Safety Evaluation in Animals and
  the Extrapolation of Results to Man.  In:   Importance of Fundamental
  Principles in Drug Evaluation, D.  Tedeschi and R.  Tedeschi, eds.
  Raven Press, New York.  pp. 449-471.

Peck, H.  1974.  Design of Experiments to Detect Carcinogenic Effects of
  Drugs.  In:  Carcinogenesis Testing of Chemicals,  L. Goldberg, ed.  The
  Chemical Rubber Company, Cleveland, Ohio.   pp. 1-14.

Peirson, M., P. Cheeke, and E. Dickinson.  1977.  Resistance of the Rabbit
  to Dietary Pyrrolizidine (Senecio) Alkaloid.  Res. Commun. Chem. Pathol.
  Pharmacol. 16(3)-.561-564.

Peraino, C., R.J.M. Fry, and E. Staffeldt.  1973.  Enhancement of Sponta-
  neous Hepatic Tumorigenesis in C3H Mice by Dietary Phenobarbital  (brief
  communication).  J. Natl. Cancer Inst. 51:1349-1350.

Percy, D. H., and A. M. Jonas.  1971.  Incidence of Spontaneous Tumors
  in CDR-1 HSM/ICR Mice.  J. Natl. Cancer Inst. 46:1045-1053.

Petzold, G. L. and J. A. Swenberg.  1978.  Detection of DNA Damage  Induced
  In Vivo Following Exposure of Rats to Carcinogens.  Cancer Res. 38:
  1589-1594.

Pietra, G., H. Rappaport, and P. Shubik.  1961.  The Effects of Carcino-
  genic Chemicals in Newborn Mice.  Cancer 14:308-317.

Pietra, G., K. Spencer, and P. Shubik.  1959.  Response  of Newly  Born Mice
  to a Chemical Carcinogen.  Nature 183:1689.

Pollard, M., and M. Kajima.  1970.  Lesions in Aged Germ Free Wistar  Rats.
  Amer. J. Pathol. 61:25-32.

Port, C.   1976.  Animal Model:  Lead Nephropathy in Gerbils  Following
  Short- and Long-Term Administration  of Lead Acetate.   Am.  J.  Pathol.
  85:519-522.

Pott, F., A. Brockhaus, and F. Huth.   1973.   Tests  on the  Production  of
  Tumors in Animal Experiments with Polycyclic  Aromatic  Hydrocarbons.
  Zbl. Bakt. Hyg., I. Abt. Orig. B 157:34-43.

-------
                                   404

 Pott,  F.,  R.  Tomlngas, and J.  Misfeld.   1977.   Tumors in Mice After Subcu-
   taneous  Injection of Automobile Exhaust Condensates.   In:   Air Pollution
   and Cancer in Man, Mohr, Sahmahl,  and Tomatis, eds.  International Agency
   for Research on Cancer,   pp. 79-87.

 Pour,  P.,  F.  W. Kruger, and J. Althoff.  1974.   Cancer of the Pancreas
   Induced  in the Syrian Golden Hamster.  Am.  J. Pathoi.  76:349-358.

 Pour,  P.,  F.  W. Kruger, and J. Althoff.  1975.   Effect of Beta-oxidized
   Nitrosamines on Syrian Hamsters.  III.  2,2-Dihydroxy-di-n-propylnitro-
   samine.   J. Natl. Cancer Inst.  54:141-145.

 Pour,  P.,  N.  Kmoch, E. Greiser, U. Mohr, J.  Althoff, and A.  Cardesa.  1976.
   Spontaneous Tumors and Common Diseases in Two Colonies of  Syrian Hamsters.
   J.  Natl. Cancer Inst. 56(6):931-935.

 Purchase,  I., and J. van der Watt.  1970.  The  Acute and Chronic Toxicity
   of  Sterigmatocystin.  In: Proceedings of the Symposium on Mycotoxins
   in  Human Health, Pretoria, September 2-4,  1970, I. Purchase, ed.
   MacMillan,  New York.  pp. 209-213.

 Rail,  D.   1969.  Difficulties  in Extrapolating  the Results of Toxicity
   Studies  in Laboratory Animals to Man.  Environ. Res.  2:360-367.

 Reeves, A. L.,  H. E. Puro, and R. G.  Smith.   1974.  Inhalation Carcinogen-
   esis from Various Forms  of Asbestos.   Environ. Res. 8:178-202.

 Reuber, M. D.  1976.  Effect of Age  and Sex on  Lesions of the Esophagus in
   Buffalo  Strain Rats Ingesting Diethylnitrosamine. Exp. Cell Biol. 44:65-72.

 Reuber, M. D.,  and E.  L. Glover.   1967.  Hyperplastic and Early Neoplastic
   Lesions  of  the Liver in  Buffalo Strain Rats of Various Ages Given Sub-
   cutaneous Carbon Tetrachloride. J.  Natl.  Cancer Inst. 38:891-899.

 Reznik, G. 1975.  The Carcinogenic  Effect of  (DMN) on the Chinese Hamster
   (Cricetulus Griseus). Cancer Lett.  1:25-28.

 Rice,  J.   1976.   Carcinogenesis:   A  Late Effect of Irreversible Toxic
   Damage During Development.   Environ.  Health Perspect.  18:133-139.

 Rigdon, R.  H.,  M. C.  Benge, H.  Kirchoff, J.  Mack, and J. Neal.  1969.
   Leukemia in Mice Fed Benzo(a)pyrene:   A Clinical, Pathologic and Hema-
   tologic  Study.   Tex.  Rep. Biol.  Med.  27:803-820.

Rigdon, R.  H.,  and J.  Neal. 1966.   Gastric  Carcinomas  and Pulmonary Ade-
   nomas in Mice Fed Benzo(a)pyrene.  Tex. Rep.  Biol. Med.  24:195-207.

Riley, V.   1975.   Mouse Mammary Tumors:  Alteration of  Incidence as
  Apparent Function of  Stress.  Science 189:465-467.

Roe, F.  1975.  Neonatal Induction of Hepatic and Other  Tumors.   In:
  Mouse Hepatic Neoplasia,  W.  Butler and P. M.  Newbern,  eds.   Elsevier
  Scientific  Publishing Co., New  York.   Chapter 7. .

-------
                                   405

Roe, F., R. Carter, and S. Adamthwaite.   1969.  Induction of Liver and
  Lung Tumors in Mice by 6-Aminochrysene Administered During the First
  Three Days of Life.  Nature (London)  221:1063-1064.

Roe, F., B. Mitchley, and M. Walters.   1963.   Tests for Carcinogenesis
  Using Newborn Mice:  1,2-Benzanthracene,  2-Naphthylamine,  2-Naphthyl-
  hydroxylamine and Ethyl Methane Sulphonate.   Br.  J. Cancer 17:255-260.

Roe, F., K. Rowsen, and M. Salaman.  1961.   Tumours of Many Sites Induced
  by Injection of Chemical Carcinogenesis into Newborn Mice, A Sensitive
  Test for Carcinogenesis:  The Implication for Certain Immunological
  Theories.  Br. J. Cancer 15:515-530.

Rosenkrantz, H., R. Sprague, R. Fleischman, and M.  Braude.  1975.  Oral
  A9-Tetrahydrocannabinol Toxicity in Rats  Treated  for Periods up to
  6 Months.  Toxicol. Appl. Pharmacol.  32:399-417,

Rygaard, J., and C. 0. Polvson.  1974.   The Mouse Mutant Nude Does Not
  Develop Spontaneous Tumors.  Acta. Pathol.  Microbiol. Scand. 82:
  99-106.

Sabine, J. R., B. J. Horton, and M. B.  Wicks.   1973.  Spontaneous Tumors
  in CSH-A^ and C3H-AVyfB Mice:  High Incidence in the United States and
  Low Incidence in Australia.  J. Natl.  Cancer Inst. 50:1237-1242.

Saffiotti, U., F. Cefis, R. Montesano,  and  A.  R. Sellakumar.  1966.  In-
  duction of Bladder Cancer in Hamsters Fed Aromatic Amines.  Ind. Med.
  Surg. 35:564.

Sass, B., L. S. Rabstein, R. Madison,  R. M. Nims, R. L. Peters, and G. J.
  Kelloff.  1975.  Incidence of Spontaneous Neoplasms in F344 Rats Through-
  out the Natural Life-Span.  J. Natl.  Cancer Inst. 54:1449-1456.

Schepers, G.W.H.  1971.  Lung Tumors of Primates and Rodents.  Part II.
  Inhalation Experiments.  Ind. Med. May:23-31.

Schmahl, D., and H. Osswald.  1967.  Carcinogenesis in Different Animal
  Species by Diethylnitrosamine.  Experentia 23:497-498.

Schrauzer, G. N., D. A. White, J. E. McGinness, and C. J. Schneider.  1978.
  Arsenic and Cancer:  Effects of Joint Administration of Arsenite and
  Selenite on the Genesis of Mammary Adenocarcinoma in Inbred Female
  C3H/St Mice.  Bioinorg. Chem. 9:245-253.

Schreiber, H., K. Schreiber, and D. H.  Martin.  1975.  Experimental Tumor
  Induction in a Circumscribed Area of the Hamster Trachea:   Correlation
  of Histology and Exfoliative Cytology. J. Natl. Cancer  Inst. 54:187-197.

Sherman, H., and A. Kaplan.  1975.  Toxicity  Studies with 5-Bromo-3-sec-
  Butyl-6-Methyluracil.  Toxicol. Appl. Pharmacol.  34:189-196.

-------
                                   406

Shimkin, M. B.  1940.   Induced Pulmonary Tumors  in Mice.  II.  Reaction
  of Lungs of  Strain A  Mice  to Carcinogenic Hydrocarbons.  Arch. Path.
  29:239-255.

Shimkin, M. B. , and G.  D.  Stoner.  1975.  Lung Tumors  in Mice:  Applica-
  tion  to Carcinogenesis Bioassay.  Adv. Cancer  Res. 21:1-58.

Shirasu, Y.  1965.  Comparative  Carcinogenicity  of 4-Nitroquinoline 1-
  Oxide and 4-Hydroxyaminoquinoline 1-Oxide in Three Strains of Mice.
  Proc. Soc. Exptl. Biol.  Med. 118:812-814.

Shubik, P.  1950.  The  Growth Potentialities of  Induced Skin Tumors in
  Mice.  The Effects of Different Methods of Chemical  Carcinogenesis.
  Cancer Res.  10:713-717.

Shubik, P.  1972.  The  Use of the Syrian Golden  Hamster in Chronic Tox-
  icity Testing.  Prog. Exp. Tumor Res. 16:176-184.

Silverstone, H., and A. Tannenbaum.   1951.  Proportion of Dietary Protein
  and the Formation of  Spontaneous Hepatomas in  the Mouse.  Cancer Res.
  11:442-446.

Smyth,  H., C.  Carpenter, C.  Weil, and J. King.   1970.  Experimental Tox-
  icity of Sodium 2-Ethylhexyl Sulfate.  Toxicol. Appl. Pharmacol. 17:53-59.

Snell,  K. C.   1965.  Spontaneous Lesions of the  Rat.   In:  The Pathology
  of Laboratory Animals, W.  E. Ribelin and J. R. McCoy, eds.  Charles C.
  Thomas, Publisher, Springfield, Illinois.

Solt, D. B., A. Medline, and E.  Farber.  1977.   Rapid  Emergence of
  Carcinogen-Induced Hyperplastic Lesions in a New Model for the Sequen-
  tial  Analysis of Liver Carcinogenesis.  Am. J. Path. 88:595-618.

Sontag, J. M., N. P. Page, and U. Saffiotti.  1976.  Guidelines for Car-
  cinogen Bioassay in Small  Rodents.  NCI-CG-TR-1, February.  Bethesda,
  Maryland.

Spatz, M., and G. Laqueur.   1967.  Transplacental Induction of Tumors in
  Sprague-Dawley Rats with Crude Cyad Material.  J. Natl. Cancer Inst.
  38:233-245.

Spicer, S., L. Chakrin, and  J. Wardell, Jr.  1974.  Effect of Chronic
  Sulfur Dioxide Inhalation  on the Carbohydrate  Histochemistry and Histol-
  ogy of the Canine Respiratory Tract.  Am. Rev. Respir. Dis. 110:13-24.

Spitz, S.,  W. H. Maguigan, and K. Dobriner.  1950.  The Carcinogenic
  Action of Benzidine.   Cancer 3:789-804.

Steinmuller,  D., L.  A.  Dillingham, and R. T. Prehn.  1969.  Lack of Car-
  cinogenic Activity of 3-Methylcholanthrene in  the Squirrel Monkey.  J.
  Natl.  Cancer Inst.  43:1175-1180.

-------
                                  407

Stevens, L.  C.  1973.  A New Inbred Subline of Mice (129/terSv) with a
  High Incidence of Spontaneous Congenital Testicular Teratomas.  J.
  Natl. Cancer Inst. 50:235-242.

Stevenson, D.  1979.  Current Problems in the Choice of Animals for Tox-
  icity Testing.  J. Toxicol. Environ. Health 5:9-15.

Stoltz, D. R., L. A. Poirer, C. C.  Irving, H. F.  Stich, J. H. Weisburger,
  and H. C.  Frice.  1974.  Evaluation of Short-Term Tests for Carcino-
  genicity.   Tox. Appl. Pharmacol.  29:157-180.

Street, A.  1970.  Biochemical Tests in Toxicology.  In:  Methods in
  Toxicology, G. Paget, ed.  F. A.  Davis Company,  Philadelphia.
  pp. 313-337.

Strong, L. C., and H. Matsunaga.  1975.  An Inverse Correlation (Dose-
  Response)  Between 5-Methyl-Cyiidine and the Fate of Spontaneous Tumors
  in Mice.  Cytobios 12:13-18.

Sugiura, K., W. E. Smith, and D. A. Sunderland.  1956.  Experimental Pro-
  duction of Carcinoma in Rhesus Monkeys.  Cancer Res. 16:951.

Sumi, N., D. Stavrou, H. Frohberg,  and G. Jochmann.  1976.  The Incidence
  of Spontaneous Tumors of the Central Nervous System of Wistar Rats.
  Arch. Toxicol. 35:1-13.

Sunderman, F. W., Jr.  1971.  Metal Carcinogenesis in Experimental Animals.
  Food Cosmet. Toxicol. 9:105-120.

Svenberg, J., A. Kostner, W. Wechsler, and R. Denlinger.  1972.  Quanti-
  tative Aspects of Transplacental Tumor Induction with Ethylnitrosourea
  in Rats.  Cancer Res. 32:2656-2660.

Swenberg, J. A., G. L. Petzold, and P. R. Harbach.  1976.  In Vitro DNA
  Damage/Alkaline Elution Assay for Predicting Carcinogenic Potential.
  Biochem. Biophys. Res. Comm. 72:732-738.

Syndor, K. L.  1973.  A Preliminary Investigation of the Susceptibility
  to Sarcoma Induction by Low Doses of B(a)P in Nine Strains of Inbred
  Male Rats.  Proceedings Tobacco and Health Workshop Conf., 4th University
  of Kentucky Tobacco and Health Research Institute.

Syndor, K. L., 0. Butenandt, F. P.  Brillantes, and C. Huggins.  1962.
  Race - Strain Factor Related to Hydrocarbon-Induced Mammary  Cancer in
  Rats.  J.  Natl. Cancer Inst. 29:805-814.

Tannenbaum,  A., and H. Silverstone.  1949.  The Influence of  the Degree
  of Caloric Restriction on  the Formation of Skin Tumors and Hepatomas
  in Mice.  Cancer Res. 9:724-727.

Tatematsu, M., T. Shirai, H. Tsuda, Y. Miyata, Y.  Shinohara,  and N.  Ito.
  1977.  Rapid Production of Hyperplastic Liver Nodules  in Rats Treated
  with Carcinogenic Chemicals:  A New Approach for an In Vivo  Short-Term
  Screening Test for Hepatocarcinogens.  Gann  68:499-507.

-------
                                   408

 Terracini,  B.,  P.  N.  Magee,  and J.  M.  Barnes.   1967.   Hepatic Pathology
   in Rats on Low Dietary Levels of  Dimethylnitrosamine.   Br.  J.  Cancer
   21:559-565.

 Terracini,  B.,  G.  Palestro,  M.  Gigliardi,  and  R.  Montesano.   1966.   Car-
   cinogenicity  of Dimethylnitrosamine  in Swiss Mice.   Br.  J.  Cancer 20:
   871-876.

 Terracini,  B.,  M.  C.  Testa,  J.  R. Cabral,  and  N.  Day.   1973.   The Effects
   of Long-Term  Feeding of DDT to BALB/c Mice.   Int.  J.  Cancer 11:747-764.

 Thiery,  M., and M.  van Gijsegem.  1965.  Species  and Strain Differences
   in the Susceptibility of the Cervico-Vaginal Squamous  Epithel.  to 3,4-
   Benzo(a)pyrene.   Br.  J. Cancer 19:418-429.

 Tomatis, L.,  C.  Agthe,  H. Bartsch,  J.  Huff, R. Montesano,  R.  Saracci,
   E.  Walker,  and J. Wilbourn.   1978.   Evaluation  of  the  Carcinogenicty
   of Chemicals:   A Review of the Monograph Program of the International
   Agency for Research on Cancer (1971  to 1977).  Cancer  Res.  38:877-885.

 Tomatis, L.,  G.  Delia Porta, and P.  Shubik.  1961.   Urinary Bladder and
   Liver  Cell Tumors Induced  in Hamsters with 0-Aminoazotoluene.   Cancer
   Res. 21:1513-1517.

 Tomatis, L.,  and V. Turusov.  1975.  Studies on the  Carcinogenicity of
   DDT.   Gann Monograph on Cancer Res.  17:219-241.

 Tomatis, L.,  V.  Turusov,  D.  Guibbert,  B.  Duperray, C. Malaveille,  and
   H.  Pacheco.   1971.   Transplacental Carcinogenic Effect of 3-Methyl-
   cholanthrene  in Mice and Its Quantitation in Fetal Tissues.  J.  Natl.
   Cancer Inst.  47:645-651.

 Torkelson,  T.,  B. Leong,  R.  Kociba,  W.  Richter, and  P. Gehring.   1974.
   Lack of Manifestation of Toxicity in Rats Inhaling 111 ppm  1,4-Dioxane
   for Two Years.  Toxicol. Appl.  Pharmacol. 29:86.

 Toth, B.  1968.  A  Critical  Review  of  Experiments in Chemical Carcino-
   genesis Using  Newborn Animals.  Cancer Res.  28:727-738.

 Toth, B., P. Magee, and P. Shubik.   1964.   Carcinogenesis  Study with
   Dimethylnitrosamine Administered  Orally  to Adult and Subcutaneously
   to  Newborn BALB/c Mice.  Cancer Res.  24:1712-1721.

 Toth, B., H. Rappaport, and  P.  Shubik.   1963.   Influence of Dose  and Age
  on  the Induction of Malignant Lymphomas  and  Other  Tumors by 7,12-Dimethyl-
  benz(a)anthracene in  Swiss Mice.   J.  Natl. Cancer  Inst.  30:723-741.

Toth, B., and P. Shubik.   1963.  Carcinogenesis in Lewis Rats Injected
  at Birth  with  7,12-DimethyIbenz(a)anthracene.   Br.  J.  Cancer 17:540-545.

-------
                                  409

Turner, F. C.  1941.  Sarcomas at the Site of Subcutaneously Implanted
  Bakelite Discs in Rats.  J. Natl.  Cancer Inst.  2:81.

Turusov, V., L. Tomatis, D. Guibbert, B.  Duperray, and H. Pacheco.  1973.
  The Effect of Prenatal Exposure of Mice to Methylcholanthrene Combined
  with the Neonatal Administration of Diethylnitrosamine.  In:  Trans-
  placental Carcinogenesis, L. Tomatis and U. Mohr, eds.  Scientific
  Publication No. 4, International Agency for Research on Cancer, Lyon,
  France,  pp. 84-91.

U.S. Food and Drug Administration.  1971.  Advisory Committee on Protocols
  for Safety Evaluation, Panel on Carcinogenesis  Report on Cancer Testing
  in the Safety Evaluation of Food Additives and  Pesticides.  Toxicol.
  Appl. Pharmacol. 20:419-438.

Verschuuren, H., R. Kroes, E. Dentonkelaar, J. Berkvens, P. Hellman, and
  G. Van Esch.  1975.  Long-Term Toxicity and Reproduction Studies with
  Metaldehyde in Rats.  Toxicology 4:97-115.

Verschuuren, H., R., Kroes, and G. Van Esch.  1973.  Toxicity Studies on
  Tetrasul.  I.  Acute, Long-Term, and Reproduction Studies.  Toxicology
  1:63-78.

Vesselinovitch, S.  1973.  Comparative Studies on Perinatal Carcinogenesis.
  In:  Transplacental Carcinogenesis, L.  Tomatis  and U. Mohr, eds.  Sci-
  entific Publication No. 4, International Agency for Research on Cancer,
  Lyon, France,  pp. 14-22.

Vesselinovitch, S. D., A. P. Kyriazis, N. Mihailovich, and K.V.N. Rao.
  1975.  Conditions Modifying Development of Tumors in Mice at Various
  Sites by Benzo(a)pyrene.  Cancer Res. 35:3948-2953.

Vesselinovitch, S., and N. Mihailovich.  1966.  Significance of Newborn
  Age and Dose of Urethan in Leukemogenesis.  Cancer Res. 26:1633-1637.

Vesselinovitch, S, N. Mihailovitch, and L. Itze.   1970.  Comparative
  Studies on the Kinetics of the Neoplastic Competence in Mice.  Cancer
  Res. 30:2548-2551.

Vesselinovitch, S., N. Mihailovich, G. Wogan, L.  Lombard, and K. Rao.
  1972.  Aflatoxin Bj, a Hepatocarcinogen in the Infant Mouse.  Cancer
  Res. 32:2289-2291.

Vesselinovitch, S., K. Rao, and N. Mihailovich.  1975.  Factors Modu-
  lating Benzidine Carcinogenicity Bioassay.  Cancer Res.  35:2814-2819.

Vlahakis, G., W. E. Heston, and G. H.  Smith.  1970.  Strain CSH-A^fB
  Mice:  Ninety Percent  Incidence of Mammary Tumors Transmitted by
  Either Parent.  Science 170:185-187.

Walpole, A. L., M.H.C. Williams,  and D.  C. Roberts.   1952.  The  Carcino-
  genic Action of 4-Aminodiphenyl and  3:2'Dimethyl-4-Aminodiphenyl.   Br.
  J. Ind. Med. 9:255-263.

-------
                                   410

 Walpole,  A.  L.,  M.H.C.  Williams,  and D.  C.  Roberts.   1954.   Tumors  of
   the Urinary Bladder in Dogs After Ingestion of 4-Aminodiphenyl.   Br.
   J.  Ind. Med.  11:105-109.

 Watabe,  H.  1971.   Early Appearance of Embryonic a-Globulin in Rat  Serum
   During Carcinogenesis with 4-Dimethylaminobenzene.   Cancer Res.  31:
   1192-1194.

 Watanabe, P., J. Young, and P- Gehring.   1977.   The  Importance of Non-
   Linear (Dose-Dependent) Pharmacokinetics  in Hazard Assessment.  J.
   Environ. Pathol.  Toxicol.  1:147-159.

 Weil, C.   1962.  Applications of  Methods of Statistical Analysis  to
   Efficient Repeated-Dose Toxicological Tests:   1.   General Considera-
   tions  and Problems Involved.  Sex Differences in Rat Liver and Kidney
   Weights.  Toxicol. Appl.  Pharmacol.  4:561-571.

 Weil, C.   1972.  Guidelines for Experiments to  Predict the  Degree of
   Safety of a Material  for Man.   Toxicol. Appl.  Pharmacol.  21:194-199.

 Weil, C.   1973.  Experimental Design and Interpretation of  Data from
   Prolonged Toxicity Studies.  Pharmacology and the  Future  of Man.
   Proc.  5th Int. Congr. Pharmacol., San Francisco, 1972,  Vol.  2:
   Toxicological  Problems, Karger,  New York.   pp. 4-12.

 Weil, C., and C. Carpenter.   1969.   Abnormal Values  in Control Groups
   During Repeated-Dose  Toxicologic Studies.   Toxicol.  Appl.  Pharmacol.
   14:335-339.

 Weil, C., and D. McCollister.  1963.   Safety Evaluation of  Chemicals.
   Relationships  Between Short- and Long-Term Feeding Studies in Designing
   and Effective  Toxicity Test. Agric.  Food Chem. 11:486-491.

 Weil, C., M.  Woodside,  H. Smyth,  Jr.,  and C.  Carpenter.   1971.  Results
   of  Feeding Propylene  Glycol in  the Diet of Dogs for Two Years.  Food
   Cosmet. Toxicol.  9:479-490.

 Weisburger,  E. K.   1977.   Carcinogenicity Studies on Halogenated Hydro-
   carbons.   Environ.  Health Perspect.  21:7-16.

 Weisburger,  J. H.   1976.   Bioassays and  Tests for Chemical  Carcinogens.
   In:  Chemical  Carcinogens,  American  Chemical  Society,  Monograph 173,
   C.  E. Searle,  ed.  Washington, D.C.   pp. 1-23.

 Weisburger,  J.,  M.  Klein, E.  Weisburger, R.  Glass, G.  Woodard,  and  M.
   Gronin.  1970.  Comparison  of the Effect  of the Carcinogen tf-Hydroxyl-
  #-2-Fluorenylacetamide  in  Infant  and Weanling Rats.   J. Natl. Cancer
   Inst. 45:29-35.

Weisburger, J. H.,  and  E. K.  Weisburger.  1967.   Tests  for  Chemical Car-
   cinogens.  From:   Methods in Cancer Res.,  Vol.  I.,  H.  Busch,  ed.
  Academic Press, New York.   pp. 307-398.

-------
                                   411

Welsch, C. W.  1976.  Interaction of Estrogen and Prolactin in Spontaneous
  Mammary Tumorigenesis of the Mouse.  J. Toxicol. Environ. Health Suppl.
  1:161-175.

Wigley, C. B., J. Amos, and P. Brookes.  1978.  Different Tumours Induced
  by Benzo(a)pyrene and Its 7,8-Dihydrodiol Injected into Adult Mouse
  Salivary Gland.  Br. J. Cancer 37:657-661.

Winstead, J.  1978.  What Kinds of Tests Are Prerequisite to Labeling a
  Compound as Toxic?  Int. J. Occup. Health Saf. 47(3):26-28.

Wislocki, P. G., R. L. Chang, A. W. Wood, W. Levin, H. Yagi, 0. Hernandez,
  H. D. Mah, P. M. Dansette, D. M. Jerina, and A. H. Conney.  1977.  High
  Carcinogenicity of 2-Hydroxybenzo(a)pyrene on Mouse Skin.  Cancer Res.
  37:2608-2611.

Wogan, G. N., and R. C. Shank.  1971.  Toxicity and Carcinogenicity of
  Aflatoxins.  In:  Advances in Environmental Science and Technology,
  J. N. Pitts and R. L. Metcalf, eds., John Wiley & Sons, Inc., New York.
  pp. 321-350.

Wold, J., J. Welles, N. Owen, W. Gibson, and D. Morton.  1978.  Toxico-
  logic Evaluation of Cefamandole Nafate in Laboratory Animals.  J.
  Infect. Dis. 137:551-559.

Womack, J.   1979.  Genetic Constitution and Response  to Toxic Chemicals —
  An Overview.  J. Toxicol. Environ. Health 5:49-51.

Woodard, G., M. Woodard, W. McNeely, P. Kovacs, and M. Cronin.  1973.
  Xanthan Gum:  Safety Evaluation by Two-Year Feeding Studies  in Rats
  and Dogs,  and a Three-Generation Reproductive Study in Rats.  Toxicol.
  Appl. Pharmacol. 24:30-36.

Worden, A.,  K. Rivett, D. Edwards, A.  Street, and A.  Newman.   1975.  Long-
  Term Feeding Study on Disodium 5'-Ribonucleotide  in Dogs.  Toxicology
  3:344-347.

World Health Organization.  1978.  Principles and Methods  for  Evaluating
  the Toxicity of Chemicals.  Part 1.  Geneva.  274 pp.

Wurzner, H., E. Lindstrom, L. Vualaz,  and H. Luginbuhl.  1977.  A  2-Year
  Feeding Study of Instant Coffees in  Rats.   I.  Body Weight,  Food Con-
  sumption,  Haematological Parameters  and Plasma Chemistry.  Food  Cosmet.
  Toxicol.  15:7-16.

Yoshida, T.  1932.  Uber die  Experimantelle Erzeugung von  Hepatom  durch
  die Futterung mit o-Amido-azotoluol.  Proc. Imp.  Acad. Japan 8:464-467.

Zabezhinskii, M. A.   1970.  Effectiveness of  Inhalation  as a Method  of
  Administration of Atomized  Carcinogens.  Bull. Exptl.  Biol.  and  Med.
  69:68-70.

-------
                                  412

Zackheim, H. S.  1964.  Comparative Cutaneous Carcinogenesis in the Rat.
  Oncologia 17:236-246.

Zbinden, G.  1973.  Formal Toxicology.  In:  Progress in Toxicology,
  Special Topics, Vol. 1.  Springer-Verlag, New York.  pp. 4-27.

Zoller, M., S. Matzku, and K. Goerttler.  1978.  High Incidence of Spon-
  taneous Transplantable Tumours in BDX Rats.  Brit. J. Cancer 37:61-66.

-------
                                              413 	

                                    TECHNICAL REPORT DATA
                              i-ase read fasfructions on the reverse before completing)
 1. REPORT NO.
 EPA-560/1-80-001
                              2.
4. TITLE AND SUBTITLE
 Scientific  Rationale for the  Selection of Toxicity
 Testing Methods:   Human Health Assessment
              5. REPORT DATE
              Publication date-December 1980
             6. PERFORMING ORGANIZATION CODE
                                                            3. RECIPIENT'S ACCESSIOWNO.
 7. AUTHOR(S)
 R. H. Ross,  M.  G.  Ryon, M. W.  Daugherty, J. S.  Drury,
 J. T. Ensminger,  and M. V. Cone
             8. PERFORMING ORGANIZATION REPORT NO.

             ORNL/EIS-151
9. PERFORMING ORGANIZATION NAME AND ADDRESS
 Health and  Environmental Studies  Program
 Oak Ridge National Laboratory
 P.O. Box X
 Oak Ridge,  Tennessee 37830
                                                            10. PROGRAM ELEMENT NO.
              11. CONTRACT/GRANT NO.

              80-D-X0856
 12. SPONSORING AGENCY NAME AND ADDRESS
 Office of Pesticides and Toxic Substances
 U.S. Environmental Protection Agency
 401 M St. SW
 Washington, D.C.  20460
              13. TYPE OF REPORT AND PERIOD COVERED
              Final                    	
              14. SPONSORING AGENCY CODE
 15. SUPPLEMENTARY NOTES
 16. ABSTRACT
      This document  is the first of a  two-part literature  analysis of  parameters
 associated with  the various toxicity  testing methods  (test animal selection, pathology
 requirements, etc.)   Acute, subchronic,  chronic, and  carcinogenic testing methods are
 covered; a discussion of some basic experimental considerations is also  included.
 This report was  prepared for the purpose of assisting and supporting  the U.S. Environ-
 mental Protection Agency in its efforts  to develop guidelines for more efficient and
 economical testing  procedures.
17.
                                KEY WORDS AND DOCUMENT ANALYSIS
                  DESCRIPTORS
b.lDENTIFIERS/OPEN ENDED TERMS
                                                                             COSATI Field/Group
 Reviews
 Toxicity
 Tests
Toxicity Testing
   Methodology
   06
13. DISTRIBUTION STATEMENT

Release unlimited
19. SECURITY CLASS (ThisReport)
Unclassified
21. NO. OF PAGES
  436
                                               20. SECURITY CLASS {This page)
                                               Unclassified
                            22. PRICE
EPA Form 2220-1 (9-73)
                                                             OU.S. GOVERNMENT PRINTING OFFICE: 1980*40-245/405

-------