PB86-122702
Health Assessment  Document for
1,2-Dichloroethane (Ethylene Dichloride)
Final Report
(U.S.) Environmental Protection Agency
Research Triangle  Park,  NC
Sep 85
                     U.S. DEPARTMENT OF COMMERCE
                  National Technical Information Service

-------
&EPA
            United States,
            Environmental Protection
            Agency
             Office of Health and
             Environmental Assessment
             Washington DC 20460
EPA 600/8-84, 006F
September 1 985
Final Report
            Research and Development
                           PE86-122702
Health Assessment
Document for
1,2-Dichloroethane
(Ethylene Dichloride)

-------
                                  • ICHNICAL REPORT DATA
                           Plfssc read ln^r.n -ions on ;he re\ er'.c it tore comr!:tnif.
  EPA/600/8-84/Q06F
                                                          3 lECIP'ENT'S ACCESSIO"»NO
                                                                    1227C? ,'3T
- TITLE
  HEALTH ASSESSMENT  DOCUMENT FOR 1,2-OICHLOROETHANE
  (Ethylene Dichloride):  Final  Report
                                                           REPORT DATE
                                                           September 1985
                                                          6 PERFORMING ORGANIZATION CODE
  AL7HCR.3)
                                                          i PERFORMING ORGANIZATION REPORT NO
.  See  List of  Consultants,  Reviewers, and Authors

9>ERFORM~lNG ORGANISATION NAME AND ADDRESS
                                                          10 PROGRAM ELEMENT NO
                                                          11 CONTRACT,GRANT NO
    Same as box 1 2.
12 SPONSORING AGENCY NAME AND ADDRESS
 U.S. Environmental Protection Agency
 Office of Health & Environmental Assessment
 Environmental Criteria & Assessment Office  (MD-52)
 Research Trianole Park. NC  27711	
                                                          13 TVPE OF REPORT AND PERIOD COVERED
                                                                Final           	
                                                          14 SPONSORING AGENCY CODE

                                                               EPA/600/23
IS SUPPLEMENTARY NOTES
16 ABSTRACT
 Ethylene Dichloride (EDC), a clear,  colorless  volatile liquid, is principally emitted
 to the environment during manufacturing.   Monitoring data, including ambient urban
 areas, indicate a concentration of equal  to or less than 0.5 ppb for most locations.
 EDC is rapidly absorbed, metabolized,  and eliminated.  Unmetabolized EDC is eliminated
 almost exclusively via the lungs.  In  humans,  the symptoms of acute toxicity from
 repeated exposures exceeding 60 ppm  are  irritation of the respiratory tract and  eyes
 and CNS depression.  According to available evidence EDC does not adversely affect  the
 reproductive or development process  in animals except at maternally toxic levels.
 Additional human epidemiologic studies are needed to establish conclusively that EDC
 is not a teratogen and does not cause  adverse  reproductive effects.  Positive response;
 in different text systems  indicate that  EDC is a weak, direct-acting mutagen; however,
 several of its metabolites, formed in  animals, are more potent mutagens than EDC.
 As a carcinogen, EDC induces tumors  in rats and mice by various routes of exposure
 (gavage, intraperitoneally, dermally).  However, lifetime inhalation exposure
 conditions did not produce tumors  in rats or mice.  Results from animal carcinogen
 studies, when considered with  the  positive evidence of mutagenicity and the presence
 of reactive metabolites and covalent bonding to DNA, suggest that EDC is a potential
 human carcinogen.
                               
-------
                                   DISCLAIMER



     This document  has  been reviewed in accordance with the U.S. Environmental



Protection  Agency's peer  and  administrative review policies and  approved  for




publication.  Mention of trade names or commercial products does not constitute




endorsement or recommendation for  use.
                                       ii

-------
                                    PREFACE



     The Office of Health and Environmental Assessment has prepared this health



assessment to serve as a "source document" for EPA use.   The  health assessment



document was originally developed for use by the Office of Air Quality Planning



and  Standards to  support  decision-making regarding  possible  regulation  of



ethylene dichloride as  a hazardous  air pollutant.  However,  the  scope of this



document has since been expanded to  address multimedia aspects.



     In the  development of the assessment document,  the  scientific literature



has been inventoried, key  studies have been evaluated  and  summary/conclusions



have been prepared so that  the  chemical's  toxicity and related characteristics



are qualitatively identified.  Observed effect levels and other measures of dose-



response relationships are discussed, where appropriate,  so that  the nature of



the  adverse   health   responses  is  placed   in  perspective  with  observed



environmental levels.



     Any information regarding  sources,  emissions, ambient  air concentrations,



and public  exposure  has been included  only to give  the  reader  a  preliminary



indication of the potential presence  of this substance in the ambient air.  While



the  available information is  presented  as  accurately  as   possible,  it  is



acknowledged to be limited and dependent in many instances on  assumption rather



than specific data. This information is not intended, nor should it be used, to



support any conclusions regarding risks to public health.



     If a  review of the health information indicates  that  the  Agency should



consider regulatory action for this substance,  a considerable effort  will be



undertaken to obtain appropriate information regarding sources,  emissions,  and



ambient air concentrations.  Such data will provide  additional information for



drawing regulatory conclusions  regarding the extent  and significance of public



exposure to this substance.
                                      iii

-------
                             AUTHORS AND REVIEWERS

AUTHORS

     The  EPA's  Office  of  Health  and  Environmental  Assessment  (OHEA)   is

responsible for the  preparation of this health assessment document.   The  OHEA

Environmental   Criteria   and   Assessment   Office   (ECAO-RTP)   had   overall

responsibility  for  coordination and direction  of the document  (Dr.  Robert  M.

Bruce,  Project  Manager).    The  chapters   addressing physical  and  chemical

properties, sampling and analysis and  toxic effects were either  rewritten  or

revised  by Syracuse  Research  Corporation.   The pharmacokinetics  chapter  was

written by Dr. I.W.F. Davidson of the Bowman  Gray School of Medicine, Wake Forest

University  and   Dr.  Jean  C.  Parker,  Carcinogen  Assessment  Group,   U.S.

Environmental Protection  Agency.  The  air quality chapters addressing sources,

emissions  and  ambient  concentrations  were  originally  written  by  Syracuse

Research Corporation and revised by Radian Corporation under a contract with the

Office of Air Quality Planning  and Standards.

     The  principal  authors  of the  chapters  prepared  by  Syracuse  Research

Corporation are:


             Stephen Bosch
             Life and Environmental Sciences Division
             Syracuse Research  Corporation
             Syracuse, New  York 13210-4080

             D. Anthony Gray, Ph.D.
             Life and Environmental Sciences Division
             Syracuse Research  Corporation
             Syracuse, New  York 13210-4080

             Joseph  Santodonato, Ph.D., C.I.H.
             Life and Environmental Sciences Division
             Syracuse Research  Corporation
             Syracuse, New  York 13210-4080
                                       iv

-------
     The OHEA Carcinogen Assessment Group (CAG) was responsible for preparation

of the sections on carcinogenicity.  Participating members of the CAG are listed

below (principal authors of present carcinogenicity materials are designated by

an asterisk [*]).



             Roy Albert, M.D. (Chairman)
             Elizabeth L. Anderson, Ph.D.
             Steven Bayard, Ph.D.
             David L. Bayliss, M.S.
             Robert P. Beliles, Ph.D.
             Chao W. Chen, Ph.D.*
             Margaret M.L. Chu, Ph.D.
             I.W.F. Davidson (consultant)
             Herman J. Gibb, M.S., M.P.H.
             Bernard H. Haberman, D.V.M., M.S.
             Charalingayya B. Hiremath, Ph.D.*
             Robert E. McGaughy, Ph.D.
             Jean C. Parker, Ph.D.*
             Charles H. Ris, M.S., P.E.
             Dharm V. Singh, D.V.M., Ph.D.
             Todd W. Thorslund, Sc.D.


     The OHEA  Reproductive  Effects  Assessment Group (REAG)  was responsible for

the preparation  of sections on  mutagenicity, teratogenicity  and reproductive

effects.  Participating members  of  REAG are listed below (principal authors of

present sections are indicated by an  asterisk [*]).   The Environmental Mutagen

Information Center (EMIC), in Oak Ridge, TN,  identified literature bearing on the

mutagenicity of EDC.


             Eric D. Clegg, Ph.D.
             John R. Fowle III, Ph.D.*
             David Jacobson-Kram, Ph.D.
             K.S. Lavappa, Ph.D.
             Sheila L. Rosenthal, Ph.D.
             Carol N. Sakai, Ph.D.*
             Lawrence R. Valcovic, Ph.D.
             Vicki L. Vaughan-Dellarco, Ph.D.
             Peter E. Voytek, Ph.D. (Director)

-------
REVIEWERS

     The following individuals provided peer  review of this draft and/or earlier

drafts of this document:


U.S. Environmental Protection Agency

             Karen Blanchard
             Office of Air, Noise and Radiation
             Office of Air Quality Planning and Standards
             Research Triangle Park, NC

             Robert M. Bruce, Ph.D.
             Office of Health and Environmental Assessment
             Environmental Criteria and Assessment Office
             Research Triangle Park, NC

             Larry T. Cuppitt, Ph.D.
             Office of Research and Development
             Environmental Sciences Research Laboratory
             Research Triangle Park, NC

             James W. Falco, Ph.D.
             Office of Health and Environmental Assessment
             Exposure Assessment Group
             Washington, DC

             Lester D. Grant, Ph.D.
             Office of Health and Environmental Assessment
             Environmental Criteria and Assessment Office
             Research Triangle Park, NC

             Thomas G. McLaughlin, Ph.D.
             Office of Health and Environmental Assessment
             Exposure Assessment Group
             Washington, DC

             David Patrick
             Office of Air,  Noise and  Radiation
             Office of Air Quality Planning  and Standards
             Research Triangle Park, NC

             David J. Reisman
             Office of Health and Environmental Assessment
             Environmental Criteria and Assessment Office
             Cincinnati, OH

             Jerry F. Stara,  D.V.M.
             Office of Health and Environmental Assessment
             Environmental Criteria and Assessment Office
             Cincinnati, OH
                                       vi

-------
CONSULTANTS,  REVIEWERS AND CONTRIBUTING AUTHORS:

             Joseph F. Borzelleca, Ph.D.
             Virginia Commonwealth University
             Richmond, VA

             Mildred Christian,  Ph.D.
             Argus Research Laboratories,  Inc.
             Perkasia, PA

             Herbert H.  Cornish, Ph.D.
             The University of Michigan
             Ypsilanti,  MI

             I.W.F. Davidson,  Ph.D.
             Bowman Gray School  of Medicine
             Wake Forest University
             Winston Salem,  NC

             Larry Fishbein,  Ph.D.
             National Center for Toxicological Research
             Jefferson,  AR

             Mark M.  Greenberg
             Office of Health  and Environmental Assessment
             Environmental Criteria  and Assessment Office
             Research Triangle Park,  NC

             Derek Hodgson,  Ph.D.
             University  of North Carolina
             Chapel Hill,  NC

             George R. Hoffman,  Ph.D.
             Holy Cross  College
             Worcester,  MA

             Rudolph  J.  Jaeger,  Ph.D.
             Consultant  Toxicologist
             7 Bolgert Place
             Westwood, NJ

             Marshall  Johnson, Ph.D.
             Thomas Jefferson  Medical College
             Philadelphia, PA

             Bruce  Kleinstein, Ph.D.
             1500 Locust Street, Suite  3504
             Philadelphia, PA

             John L. Laseter,  Ph.D.
             Environmental Affairs, Inc.
             New Orleans, LA
                                     vii

-------
Edmond J. LaVoie, Ph.D.
American Health Foundation
Valhalla, NY

Marvin S. Legator, Ph.D.
University of Texas Medical Branch
Calveston, TX

P.D. Lotilaker, Ph.D.
Pels Research Institute
Temple University Medical Center
Philadelphia, PA

Bernard Schwetz, Ph.D.
National Institute of Environmental Health Sciences
Research Triangle Park, NC

Sam Shibko, Ph.D.
Health and Human Services
Division of Toxicology
Washington, DC

Charles M. Sparacino, Ph.D.
Research Triangle Institute
Research Triangle Park, NC

Daniel S. Straus, Ph.D.
University of California
Riverside, CA

Darrell D. Sumner, Ph.D.
CIBA GEIGY Corporation
High Point, NC

Robert Tardiff, Ph.D.
1i»23 Trapline Court
Vienna, VA

Norman M. Trieff, Ph.D.
University of Texas Medical Branch
Department of Pathology, UTMB
Galveston, TX

Benjamin Van Duuren, Ph.D.
New York University Medical Center
550 First Avenue
New York, NY

James Withey, Ph.D.
Room 315C
Environmental Health Center
Division of Environmental and Occupational Toxicology
DeLa Colonbine
Tunney's Pasture
Ottawa, Ontario
CANADA K1A OL2
                         viii

-------
                            SCIENCE ADVISORY BOARD
                        ENVIRONMENTAL HEALTH COMMITTEE
                       CHLORINATED ORGANICS SUBCOMMITTEE


     The content of this health assessment document  on  ethylene dichloride was
independently  peer-reviewed  in  public  session  by   the  Environmental  Health
Committee of the Environmental  Protection Agency's Science Advisory Board.


                  ACTING CHAIRMAN.  ENVIRONMENTAL HEALTH COMMITTEE

Dr.  Seymour Abrahamson,  Professor  of  Zoology  and Genetics,  Department  of
     Zoology, University of Wisconsin,  Madison,  Wisconsin  53706.


                    EXECUTIVE SECRETARY.  SCIENCE ADVISORY BOARD

Dr. Daniel Byrd III,  Executive  Secretary, Science Advisory Board, A-101 F, U.S.
     Environmental Protection Agency,  Washington, DC  20460.


                                      MEMBERS

Dr.  Ahmed  E.   Ahmed,   Associate  Professor  of  Pathology,   Pharmacology  and
     Toxicology, the University of Texas  Medical Branch at Galveston.

Dr.  George T.  Bryan,   Professor  of  Human  Oncology,  K4/528  C.S.C.  Clinical
     Sciences,  University of Wisconsin,  500 Highland Avenue,  Madison, Wisconsin
     53792.

Dr. Ronald  D.  Hood,  Professor  and Coordinator,   Cell  and  Developmental Biology
     Section, Department of Biology, The University of Alabama,  and Principal
     Associate, R.D. Hood  and  Associates,  Consulting  Toxicologists,  P.O.  Box
     1927, University,  Alabama  35486.

Dr. K.  Roger. Hornbrook, Department of Pharmacology, P.O.  Box 26901, University of
     Oklahoma,  Oklahoma City, Oklahoma   73190.
                                        ix

-------
                               TABLE OF CONTENTS
DISCLAIMER	      ii

PREFACE	     iii

AUTHORS AND REVIEWERS	      iv

LIST OF TABLES	     xii

LIST OF FIGURES	    xvii

1.   SUMMARY AND CONCLUSIONS.. ^	     T'1

2.   INTRODUCTION	     2~1

3.   PHYSICAL AND CHEMICAL PROPERTIES	     3-1

     3.1. NAME	•.	     3-1
     3.2. CAS REGISTRY, RTECS,  AND STORET NUMBERS	     3-1
     3.3. DESCRIPTION	     3-1
     3.4. STRUCTURE	     3-1
     3.5. PHYSICAL PROPERTIES  OF PURE  ETHYLENE  DICHLORIDE	     3-1

4.   SAMPLING AND ANALYSIS OF  ETHYLENE DICHLORIDE	     1-1

     1.1. SAMPLING	     1*-1
     1.2. ANALYSIS	     4-1

          4.2.1.  Ethylene Dichloride  in Air	     4-1
          4.2.2.  Ethylene Dichloride  in Water	     1-2
          1.2.3.  Ethylene Dichloride  in Solid  Samples	     1-3
          4.2.4.  Ethylene Dichloride  in Blood	     4-3
          4.2.5.  Ethylene Dichloride  in Urine	     4-3
          4.2.6.  Ethylene Dichloride  in Tissue	     4-4

5.   SOURCES IN  THE  ENVIRONMENT	     5-1

     5.1. PRODUCTION PROCESSES	     5-1
     5.2. ETHYLENE DICHLORIDE  PRODUCERS	     5-2
     5.3. ETHYLENE DICHLORIDE  PRODUCTION AND TRENDS	    5-2
     5.4. ETHYLENE DICHLORIDE  USES	    5-5
     5.5. SOURCES  OF EMISSIONS	    5-9

          5.5.1.   Production and Related Facilities	    5-12
          5.5.2.   Dispersive Uses	    5-13
          5.5.3.   Conclusions	    5-14

-------
                           TABLE OF CONTENTS (cont.)
                                                                           Page

6.   TRANSPORT AND FATE IN THE ENVIRONMENT	    6-1

     6.1.  ATMOSPHERE	    6-1
     6.2.  AQUATIC MEDIA	    6-7
     6.3-  SOIL	    6-9
     6.4.  SUMMARY	   6-10

7.   ENVIRONMENTAL LEVELS AND EXPOSURE	    7-1

     7.1.  ENVIRONMENTAL LEVELS	    7-1

          7.1.1.   Atmospheric Levels	    7-1
          7.1.2.   Ground and Surface Water Levels	    7-9
          7.1.3.   Soil and Sediment Levels	   7-14

     7.2.  ENVIRONMENTAL EXPOSURE	   7-14

          7.2.1.   Exposure from Air	   7-18
          7.2.2.   Exposure from Water	   7-20
          7.2.3*   Exposure from Food	   7-20

     7.3.  CONCLUSIONS	   7-20

8.   ECOLOGICAL EFFECTS	    8-1

9.   BIOLOGICAL EFFECTS IN MAN AND EXPERIMENTAL ANIMALS	    9-1

     9.1.  PHARMACOKINETICS	    9-1

          9.1.1.   Absorption and Distribution	    9-1
          9.1.2.   Excretion	   9-18
          9.1.3.   Metabolism	   9-32
          9.1.4.   Summary and Conclusions	   9-55

     9.2.  ACUTE,  SUBCHRONIC AND CHRONIC TOXICITY	   9-57

          9.2.1.   Effects in Humans	   9-57
          9.2.2.   Effects in Animals	   9-86
          9.2.3.   Summary of Acute, Subchronic and Chronic Toxicity	  9-115

     9.3-  REPRODUCTIVE AND TERATOGENIC EFFECTS	  9-122

          9.3.1.   Summary and Conclusions	  9-129

     9.4.  MUTAGENICITY	  9-131

          9.4.1.   Gene Mutation Studies	  9-131
          9.4.2.   Chromosome Aberration Studies	  9-152
          9.4.3.   Other Evidence of DNA Damage	  9-157
          9.4.4.   Summary and Conclusions	  9-162
                                      XI

-------
                            TABLE OF CONTENTS (cont.)
                                                                            Page

      9.5. CARCINOGENICITY	   9-164

           9.5.1.  Animal Studies	   9-164
           9.5.2.  Epidemiologic Studies	   9-204
           9.5.3.  Risk Estimation from Animal Data	   9-205
           9.5.4.  Summary	   9-257
           9.5.5.  Conclusions	   9-260

 Appendix A.  Comparison Among Different Extrapolation Models	   9-263
 Appendix B.  Risk Calculation Based on Time-to-Event Data	   9-265

10.   REFERENCES 	    10-1
                                        xii

-------
                                LIST OF TABLES

Table                                                                      Page

5-1     Major Manufacturers of Ethylene Bichloride	     5-3

5-2     U.S. Production and Sales of Ethylene Dichloride	     5-*4

5-3     Ethylene Dichloride Uses	     5-6

5-H     Consumption of Ethylene Dichloride in 1979 and 197U	   •  5-7

5-5     Uses of Ethylene Dichloride for Intermediate Purposes	     5-8

5-6     Estimated Environmental Releases of Ethylene
        Dichloride in 1979	    5-10

6-1     Rate Constants for a Few Chlorinated Ethanes at
        Room Temperature	     6-3

7-1     Ambient Atmospheric Levels of Ethylene Dichloride	     7-2

7-2     Ambient Concentrations of Ethylene Dichloride in Urban Areas...     7-8

7-3     Reported Occurrence of Ethylene Dichloride in Groundwater
        Systems — Combined Federal Data (NORS,  NOMS, NSP,  CWSS, GWSS).    7-15

7-1     Reported Occurrence of Ethylene Dichloride in Surface Water
        Systems — Combined Federal Data (NORS,  NOMS, NSP,  CWSS)	    7-16

7-5     State Data on Ethylene Dichloride in Groundwater	    7-17

7-6     Estimated Respiratory Intake of Ethylene Dichloride by
        Adults and Infants	    7-19

7-7     Maximum Concentration Level in the Vicinity of Various
        Emission Sources	    7-19

7-8     Total Estimated Population (in Thousands) Exposed to
        Ethylene Dichloride in Drinking Water at the Indicated
        Concentration Ranges	    7-21

7-9     Estimated Drinking Water Intake of Ethylene Dichloride
        by Adults and Infants	    7-21

9-1     Physical Properties of Ethylene Dichloride and
        Other Chloroethanes	     9-3

9-2     Partition Coefficients for Ethylene Dichloride	     9-4


9-3     Fate of 14C-EDC in Rats 18 Hours After Oral (150 mg/kg)
        or Inhalation (150 ppm, 6-hr) Exposure	    9-6
                                     xiii

-------
                             LIST OF TABLES (cont.)

Table                                                                      Page

9-1     Peak Blood and Tissue Levels After Single Oral Dosage
        of EDC in Male Rats	     9-8

9-5     The Absorption Rate  Constants (k  ) and Area Under Curve
        (AUC) for Rats After Single Oral Doses of EDC in Oil
        and in Water as Vehicle  and After Intravenous
        Administration	•     9-9

9-6     EDC Tissue Levels After  50 ppm Inhalation Exposure	    9-14

9-7     EDC Tissue Levels After  250 ppm Inhalation Exposure	    9-15

9-8     Blood Tissue Levels  of EDC in Male Rats 2 Hours After
        Single Oral Doses in Corn Oil	    9-17

9-9     Percent Distribution of  Radioactivity Excreted (48-hr) by
                                           ill
        Mice Receiving 1,2-Dichloroethane-  C	    9-20

9-10    Pharmacokinetic Parameters of EDC Administered as Single Bolus
        Intravenous Injections in Saline  to Sprague-Dawley Male Rats.
        Parameters Calculated from a 2-Compartment Open Model	  9-24

9-11    Pharmacokinetic Parameters of EDC Administered as Single Oral
        Doses in Corn Oil and Water to Sprague-Dawley Male Rats.
        Parameters Calculated from a 2-Compartment Open Model	  9-26

9-12    Pharmacokinetic Parameters of EDC Following Termination of
        Steady State Inhalation  Conditions (5-hr Exposure) of 50
        and 250 ppm to Male  Sprague-Dawley Rats	  9-29

9-13    Pharmacokinetic Parameters of EDC Following Termination of
        Steady State Inhalation  Conditions; 6-Hr Exposure, 150 ppm
        to Male Osborne-Mendel Rats.  Parameters Calculated from a
        Two-Compartment Model	  9-30

9-14    Identified Metabolites of Ethylene Dichloride and
        Ethylene Bromide	    9-34

9-15    Percent Distribution of  Radioactivity Excreted (48-hr) as^
        Urinary Metabolites  by Mice Receiving 1,2-Dichloroethane-  C

        or 2-Chloroacetic Acid-^C	    9-37

9-16    Total Macromolecular Binding and  DNA Binding in Selected
                                           14
        Tissues of Rats After Exposure to  C-EDC by Oral or
        Inhalation Routes	    9-52
                                       xiv

-------
LIST OF TABLES (cont.)
Table
9-17


9-18

9-19

9-20

9-21

9-22

9-23
9-24
9-25
9-26
9-27

9-28
9-29

9-30
9-31

9-32
9-33

9-31

9-35


14
Effect of Dietary Disulfiram Upon the C Content of
Liver Nuclei Isolated 21 or 48 Hours After Administration
of a Single Oral Dose of 15 mg/kg [U-14C]EDB 	
Effects Associated with Acute Lethal Oral Doses of

Effects of Acute Oral Ingestion of 1 ,2-Dichloroethane

Effect of Ethylene Dichloride Exposure on Eye

Morbidity and Lost Workdays of Aircraft Industry Gluers

Concentrations of Ethylene Dichloride in Oil Refinery
Mineral Oil Purification Process Air 	
Effects Observed in Polish Oil Refinery Workers 	

Effects of Ethylene Dichloride on the Cornea 	

Summary of Mutagenicity Testing of EDC: Gene Mutations


Summary of Mutagenicity Testing of EDC: Gene Mutation
Tests in Insects 	
Summary of Mutagenicity Testing of EDC: Mammalian Systems....
Summary of Mutagenicity Testing of EDC: Chromosomal


Summary of Mutagenicity Testing of EDC: DMA Binding
Studies 	
Design Summary for 1 ,2-Dichloroethane (EDC) Gavage

Terminal Survival of Osborne-Mendel Rats Treated With

Page


9-54

9-58

9-65

9-72

9-78

9-80
9-81
9-87
9-95
9-102

9-132
9-143

. 9-145
9-151

9-153
9-158

9-161

9-166

9-170
          XV

-------
                             LIST OF TABLES (cont.)

Table                                                                      Page

9-36    Squamous Cell Carcinomas of  the Forestomach in Osborne-
        Mendel Rats Treated With 1,2-Dichloroethane (EDC)	    9-171

9-37    Hemangiosarcomas in Osborne-Mendel Rats Treated With
        1,2-Dichloroethane (EDC)	    9-172

9-38    Adenocarcinomas of the Mammary Gland in Female Osborne-
        Mendel Rats Treated With 1,2-Dichloroethane (EDC)	    9-173

9-39    Design Summary for 1,2-Dichloroethane (EDC) Gavage
        Experiment in B6C3F1 Mice	    9-175

9-10    Terminal Survival of B6C3F1  Mice Treated With
        1,2-Dichloroethane (EDC)	    9-176

9-11    Hepatocellular Carcinomas  in B6C3F1 Mice Treated With
        1,2-Dichloroethane (EDC)	    9-179

9-12    Alveolar/Bronchiolar Adenomas in B6C3F1 Mice Treated
        With 1,2-Dichloroethane  (EDC)	    9-180

9-13    Squamous Cell Carcinomas of  the Forestomach in B6C3F1 Mice
9-11
9-15
9-16
9-17
9-18
9-19
9-50
9-51
Adenocarcinomas of the Mammary Gland in Female B6C3F1 Mice
Treated With 1 , 2-Dichloroethane (EDC) 	
Endometrial Polyp or Endometrial Stromal Sarcomas in Female
B6C3F1 Mice Treated With 1 , 2-Dichloroethane (EDC) 	 ,
Characterization of 1 , 2-Dichloroethane (EDC) Inhalation
Design Summary for 1, 2-Dichloroethane (EDC) Experiment
Survival of Sprague-Dawley Rats Exposed to EDC at 52 and
1 01 Weeks 	 ,
Tumor Incidence in Sprague-Dawley Rats Exposed to
Mammary Tumors in Sprague-Dawley Rats Exposed to
Design Summary for 1 ,2-Dichloroethane (EDC) Experiment
9-182
9-183
9-181
9-185
9-186
9-188
9-192
9-195
                                      xv i

-------
                            LIST OF TABLES (cont.)



Table                                                                     Page
9-52

9-53

9-51

9-55

9-56


9-57

9-58


9-59

9-60
9-61

9-62

9-63

9-61

9-65

9-66


Survival of Swiss Mice Exposed to 1 , 2-Dichloroethane (EDC)
at 52 and 78 Weeks 	
Tumor Incidence in Swiss Mice Exposed to 1 , 2-Dichloroethane
(EDO 	
Pulmonary Tumor Response in Strain A/st Mice Injected
with 1 , 2-Dichloroethane (EDC) 	
Mouse Skin Bioassay of 1, 2-Dichloroethane (EDC) and
Chloroacetaldehyde 	
Incidence Rates of Hemangiosarcomas in the Circulatory
Systems of Male Osborne-Mendel Rats and of Hepatocellular

Disposition of 1 C-EDC in Rats After Single Administrations
of 150 mg/kg Orally and 150 ppm for 6 Hours by Inhalation 	
Comparison of the Metabolism and Pharmacokinetics of the
"High" Doses in Rats of the Maltoni et al. Inhalation and

Evidence of Nonlinear 'Kinetics for EDC in Rats After

1U
DNA Covalent Binding in Rats Exposed to C-EDC 	
Lifetime Average Human Equivalent Doses Based on the NCI

Lifetime Average Human Equivalent Doses Based on the NCI
Rat Bioassay 	
Incidence Rates of Hemangiosarcomas in the Circulatory
System of Male Osborne-Mendel Rats 	
Incidence Rates of Hepatocellular Carcinomas in Male
B6C3F1 Mice 	
The 95$ Upper-Bound Estimate of Risk at 1 mg/kg/day Using

Relative Carcinogenic Potencies Among 51 Chemicals
Evaluated by the Carcinogen Assessment Group as Suspect
Human Carcinogens 	

9-196

9-197

9-201

9-203


9-207

9-210


9-211

9-219
9-227

9-233

9-235

9-215

9-215

9-216


9-253
                                     xvii

-------
                                LIST OF FIGURES


Figure                                                                     I2E§

9-1     EDC levels after single oral administration in rats	    9-7

9-2     Top:  Blood levels of EDC observed during and following a
        6-hour inhalation exposure to  150 ppm EDC.  Bottom:  Semi-
        logarithmic plot of EDC blood  levels vs. time after
        exposure termination	   9-12

9-3     Blood levels of EDC in rats after i.v. administration	   9-23

9-1     Levels of EDC  in rats after inhalation exposure to 50 ppm
        (top) and 250  ppm  (bottom).  Levels were measured at
        termination of 5-hour exposure period	   9-27

9-5     Postulated microsomal oxidative metabolism of 1,2-dihaloethanes.   9-39

9-6     Further metabolism of 2-chloroacetaldehyde and 1-chloroso-
        2-chloroethane from miorosomal oxidation	   9-11

9-7     Postulated cytosolic metabolism of ethylene dichloride	   9-14

9-8     Growth curves  for male and female Osborne-Mendel rats
        administered EDG by gavage	  9-168

9-9     Survival comparisons  for  male  and female Osborne-Mendel rats
        administered EDC by gavage	  9-169

9-10    Growth curves  for male and female B6C3F1 mice administered
        EDC by gavage	  9-177

9-11    Survival comparisons  for  male  and female B6C3F1 mice
        administered EDC by gavage	  9-178

9-12    Relationship in mice  between  dose of EDC  (mg/kg) and the  amount
        of EDC exhaled unchanged  and  the amount  of  the dose metabolized
        to urinary metabolites  plus  C02	    9-211

9-13    Relationship  in Sprague-Dawley rats  between  the area under  the
        blood EDC concentration-time  curve  (AUC)  and  the gavage dose
        of EDC in corn oil (mg/kg) or inhaled doses  of EDC (ppm for
        6 hours)	  9-218

9-lU    Linear relationship for  EDC  between  the  logarithm  of the  area
        under the blood  C,t curve (AUC)  and  the  logarithm  of gavage
        dose  in  corn  oil  given to Sprague-Dawley rats	   9-221

9-15    Relationship  in  Sprague-Dawley rats  between gavage doses  of EDC
        in corn  oil  and  the ratio from the  area  under the  blood EDC
        concentration-time curve (AUC) divided by the administered  dose.   9-223
                                      xviii

-------
                                LIST OF FIGURES
Figure                                                                     Page

9-16    Blood levels of EDC following exposure to EDC by inhalation
        (150 ppm,  6 hours)  or gavage (150 mg/kg)	  9-225

9-17    Histogram  representing the frequency distribution of the
        potency indices of  the 54 suspect carcinogens evaluated by
        the Carcinogen Assessment Group	  9-252
                                     xix

-------
                          1.   SUMMARY AND CONCLUSIONS



     Cthylene dichoride  (EDC)  is a  clear,  colorless, volatile  liquid with  a



pleasant odor.   EDC has a molecular weight of 98.96, a boiling point  of 83-7°C



and  a  vapor pressure  of  64 torr  at 20°C.   Unlike  more highly  chlorinated



hydrocarbons, EDC has a flashpoint (17°C closed cup),  an  autoignition tempera-



ture (413°C) and explosive limits (6.2-15.6$ by volume in  air).   It has a water



solubility of 8820  mg/2. and  a log octanol/water partition coefficient of 1.48.



     EDC is analyzed best  by  gas  chromatography using either an electron capture



or halogen  specific  (microcoulometric or electrolytic conductivity)  detector.



Alternatively,  a gas chromatograph/mass spectrometer may  be used.   EPA methods



502.1,  601, 624 and 8010 detail the analysis of EDC  from  water,  wastewater  and



solid samples.   Variations  in  these methods  provide for analysis of EDC  in



biological media (e.g., blood, urine  and tissue).   EDC in  air is .analyzed  by



condensing the volatiles  in  a  previously collected air sample in  a GC column,



followed by temperature programming  the  column.



     EDC is produced commercially by  the direct chlorination or oxychlorination



of ethylene. Most  of the production capacity, which exceeds nine  million metric



tons annually,  is located  in  Texas and Louisiana.  Roughly 90$  of  the production



is captively consumed by the  producers.   Production during 1980,  1981  and 1982



totalled 5.037,  1.523  and 3-155  million metric tons, respectively.    A  major



portion (84$) of  the United  States EDC  production is consumed  to make  vinyl



chloride monomer.   It is  also  used  to  make chlorinated solvents, vinylidene



chloride and ethyleneamines.



     The major  source  of  EDC  emissions to  the environment  is  from   vent  gas



streams during  the manufacture of EDC  and its derivatives. Significant amounts



are also emitted to the environment from dispersive uses in gasoline, paints  and
                                      1-1

-------
cleaning agents.  In 1979, an estimated 6696 metric tons  of  EDC were released to



the  atmosphere from EDC and derivative production, while WW metric tons were



released from dispersive  uses.



     The most likely removal mechanism for EDC from  the atmosphere is reaction



with hydroxyl  free radicals.   Based  on available  kinetic data  and  average



tropospheric  hydroxyl  free radical concentrations, the half-life of EDC has been



estimated  to  range  from 36-127 days.   Due  to different  factors that may cause



variations in *OH concentrations in the troposphere, the persistence of EDC could



vary somewhat from  the estimated value.  Chloroacetyl chloride is probably the



principal  product resulting  from the reaction of  EDC  with >OH radicals.



     EDC  is   not  expected to play  a  significant role  in  stratospheric  ozone



destruction   reactions  due  to  its  relatively  short  tropospheric  half-life.



However, Chloroacetyl  chloride  may have sufficient stability to diffuse to the



stratosphere  and may participate in  UV reactions,  producing  chlorine atoms.



     In  the   aquatic   environment,   volatilization   appears to   be  the  most



significant removal mechanism.  The half-life for  this process has been estimated



to be -4 hours.



     From  the little information that is  available regarding the fate of EDC in



soil, it can be surmised that both volatilization  from and leaching through soil



may  be two significant  removal  mechanisms for EDC.



     The highest  atmospheric concentrations  that have been monitored  for EDC



have been  detected  near production and use facilities.  Mean levels as high as



27.5 ppb have been monitored near production-use facilities  in Lake Charles, LA.



However, atmospheric exposure  appears to  vary   greatly  from one  location  to



another.   The  available monitoring data,  which  includes   levels  detected  in



general ambient urban areas, indicate that most locations have concentrations of



<0.5 ppb.   The  data are  not  sufficient to  determine regional variations  in
                                       1-2

-------
exposure levels.  Using  Federal  Reporting  Data System information, it has  been



projected  that  EDC  levels in all groundwater  and surface water systems in  the



United  States  fall  below 10 ug/S,, and  that most  are  <1.0 ug/fc.   No data  are



available  indicating the presence of EDC in finished foods.



     Pharmacokinetic  studies  in animals  indicate  that  EDC  is   rapidly   and



extensively absorbed following oral and inhalation exposure.  Dermal absorption



is  negligible  in most vapor  exposure  situations, although  absorption by  this



route  may  be  significant  with  direct  liquid  contact.    Complete  tissue



distribution of EDC  is  consistent with  its  lipophilic nature;  the chemical



crosses the blood/brain and placental barriers and concentrates in breast milk.



Up to 90? of low oral  or  inhalation doses are metabolized by rats and mice, with



biotransformation  occurring  by  multiple  pathways;  EDC  is  metabolized  to



2-chloroacetaldehyde,    S-(2-chloroethyl)-glutathione,   and   other   putative



reactive metabolites  capable of  covalent  binding to  cellular macromolecules.



Metabolism  is  dose-related; as  the dose  increases,  the percent  metabolized



decreases,  although  the  absolute amount  of  EDC metabolized  increases  until



saturation of metabolic pathways  occur.  Elimination of unmetabolized EDC occurs



almost  exclusively  via the  lung,  with a  greater percentage being  exhaled at



higher  doses.    Total body  elimination of EDC  is  relatively rapid and  is



consistent  with  a   two-compartment   system  and   Michalis-Menten  kinetics.



Significant bioaccuraulation  of EDC in blood  and  tissues is not expected to occur.



     The effects of acute inhalation exposure  to  EDC  are similar in humans and



animals. Immediate symptoms of toxicity are CNS depression and irritation of the



respiratory tract  and eyes.   Death was  usually  ascribed to  respiratory  and



circulatory failure, and  pathologic  examinations  typically revealed congestion,



degeneration,  necrosis and  hemorrhagic  lesions of most  internal organs  (e.g.,



liver,  kidneys,  spleen, lungs,  respiratory  tract,  gastrointestinal  tract).
                                      1-3

-------
     Several  papers  describing human health  surveys  appear  in the literature;



adverse  effects  are largely  associated with the  gastrointestinal  and  nervous



systems.  The exposure information  in these studies is not well documented, but



taken together indicate  that  adverse effects have likely occurred in humans at



EDO  levels below  100 ppm,  although probably  not as low as  10 ppm.   Subtle



neurological  effects  (e.g.,  fatigue, irritability, sleeplessness)  may  be more



prevalent than overt symptoms of  CNS toxicity at  lower concentrations.  Studies



with multiple species of animals  have shown that  subchronic or chronic exposure



to EDC  at vapor concentrations of £100  ppm  did not produce treatment-related



adverse  effects on  survival,  growth,  hematology,  clinical  chemistry,  organ



weight or  histology.   Toxic  effects were apparent at higher concentrations and



were  exposure-related;   exposure  to 400-500  ppm  produced  high  mortality and



histopathological alterations in  rodents  within a few exposures.



     Limited  data  indicate that  the toxic  response to acute  oral  exposure is



similar to that of inhalation exposure in  humans and animals.  Subchronic/chronic



oral  administration  of  EDC  at  daily dosages of  =200 mg/kg and  higher caused



decreased growth rate and  mortality in  mice,  which may have been tumor-related.



Chronic  oral  administration of lower  dosages  of EDC (=34 mg/kg/day)  to rats



produced mortality that  appeared to be due to  non-neoplastic lesions including



bronchiopneumonia and endocardial thrombosis.



     The  available evidence  suggests  that EDC does not adversely affect the



reproductive  or development  process in laboratory animals except at maternally



toxic levels.  However, additional laboratory testing is needed,  as  well as



epidemiological  studies,  to  conclusively establish  that EDC  is  not  a  human



teratogen and does not cause  adverse reproductive effects.



     Positive responses  in different test systems representing a wide range of



organisms indicate that  EPC  is  capable  of causing gene mutations in prokaryotes
                                       1-14

-------
and eukaryotes.  Several of its  putative  metabolites,  thought to  be  formed in



rats and mice,  are judged to be more  potent mutagens  than EDO.  EDC has not been



adequately tested for its ability to  cause chromosomal  aberrations or heritable



effects.  Further testing is needed to assess its ability to  cause these effects.



     EDC was shown  to  be carcinogenic  in a  lifetime gavage bioassay  that was



conducted  by  the  National  Cancer  Institute,  producing  tumors  in both  rats



(forestomach  carcinomas,  cirulatory  system  hemangiosarcomas,   subcutaneous



fibromas, mammary  gland  adenocarcinomas)  and mice  (hepatocellular carcinomas,



alveolar/bronchiolar adenomas,  mammary  carcinomas,  endometrial  tumors).   EDC



produced  an elevated  but  not  a  statistically  significant increase   in  the



incidence of lung adenomas in strain A mice when administered intraperitoneally.



EDC induced statistically significant benign  lung  tumors in mice when applied to



the skin,  but  statistically significant  skin tumors  were  not  observed.   No



statistically significant increases in tumors occurred in rats or mice following



lifetime  inhalation exposure.     No  case  reports  or  epidemiologic  studies



concerning EDC were available in the  published literature for analysis.



     From a weight-of-evidenee approach,  the  direct and supporting evidence for



carcinogenicity includes:   1} multiple tumor types in an oral rat bioassay and an



oral mouse  bioassay,  2} suggestive  evidence in  two  other animal  bioassays,



3) demonstrated evidence of reactive metabolites  of  EDC  and formation  of a DNA



adduct, and 4)  evidence that EDC is also  a mutagen.



     The U.S. Environmental Protection Agency is  using,  on an  interim basis, a



proposed  classification  scheme  for  evaluating  the weight of  evidence  for



carcinogenicity.  Using this classification, the  positive  findings in  the oral



rat and  mouse  studies  would be  considered  a sufficient level  of  evidence in



experimental animals.  The sufficient level of animal evidence, together with an



absence  of epidemiologic  data,  provides  a  basis for  an  overall  weight-of-
                                      1-5

-------
evidence  ranking  of Group  B2,  meaning that EDC  is a "probable"  carcinogen in


humans.


     Applying   the  International   Agency  for  Research   on   Cancer  (IARC)


classification scheme, which  EPA  has used in the past, the positive finding in


the experimental animals and the absence of epidemiologic data provide an overall


weight-of-evidence  ranking  of  Group 2B, meaning  that  EDC is  a  "probable"


carcinogen in humans.


     While  the  "probable"  carcinogenicity  conclusion  obviously  applies  to


ingestion exposure since the positive evidence comes from gavage experiments, it


is likely that EDC is a "probable" carcinogen for humans via inhalation exposure.


In view of the  evidence,  it is prudent to consider EDC to be carcinogenic via


inhalation exposure although the potency may vary from that estimated from gavage


studies.


     Assuming that EDC is carcinogenic for humans, data from gavage studies using


both rats and  mice can be  used to  estimate the  carcinogenic potency  and unit


risks.  The development of the potency and unit risk values is for the purpose of


providing an estimator to  assess the possible magnitude  of the  public health


impact if EDC is carcinogenic  for humans. The carcinogenic  potencies estimated


on the basis of hemangiosarcomas in rats  and  hepatocellular carcinomas in mice


are comparable  when the linearized  multistage model is used.  The upper-bound

                                     _2
estimate  of  EDC potency  is 9.1 x 10  /mg/kg/day based  on the  hemangiosarcoma


response  in rats  using a time-to-death adjustment  and an  adjusted dose derived


from  the  metabolism/kinetic  evaluation.   The  upper-bound estimate of  the


incremental cancer risk due  to 1 ng/i of EDC  in drinking water  is 2.6 x 10~  .


Two upper-bound  estimates  of the incremental  cancer risk for the inhalation of


1 jig/nr of  EDC are calculated:   2.6 x 10   on the basis of the  gavage potency


value,  and  1.0 x  10~  on  the  basis  of a  negative  inhalation  study.    The
                                       1-6

-------
inhalation  unit risks  are  presented  as  two  values  in  recognition of  the



uncertainties associated  with the  different data  sets  used,  there  being  no



certain reasoning that one value  has more scientific merit than the other value.



The upper-bound nature of the unit-risk estimates is such that the true risk is



not likely to exceed these values and may be lower.



     Expressed in terms of relative  potency,  EDC ranks in  the  fourth quartile



among 51 carcinogens evaluated by EPA's Carcinogen Assessment Group.
                                      1-7

-------
                               2.  INTRODUCTION



     EPA's Office of Research and Development has  prepared  this health assess-



ment to serve as a "source document"  for Agency use.  This health assessment was



originally developed for use by the Office of Air Quality Planning and Standards



to support decision-making regarding  possible regulations of ethylene dichloride



under Section 112 of the Clean  Air Act.  However, based on the expressed interest



of  other  agency offices,  the  scope  of this document  was expanded  to address



ethylene dichloride in relation to sectors  of the environment outside of air.  It



is  fully  expected  that  this document will  serve the information needs of many



government agencies and private groups that may be involved  in decision-making



activities related to ethylene dichloride.



     In the development of the assessment  document, existing scientific litera-



ture has been surveyed in detail.  Key studies have been  evaluated and summary



and conclusions have been prepared so that  the chemical's toxicity and related



characteristics are qualitatively identified.



     The  document  considers  all sources  of ethylene  dichloride in  the  envi-



ronment, the likelihood for its exposure to humans, and the  possible effect on



man and lower organisms from absorption.   The  information  found in the document



is  integrated  into a format  designed as  the  basis for  performing  unit  risk



assessments.  When  appropriate,  the  authors of the document  have attempted to



identify gaps in current knowledge that  limit  risk  evaluation capabilities.



     The  basic  literature search for this  document  is complete  through  1983;



however,  selected  publications  have  been   included  in  the   sections  on



mutagenicity and carcinogenicity through  198H.
                                      2-1

-------
                     3.   PHYSICAL AND CHEMICAL PROPERTIES
3.1. NAME
     Ethylene dichloride (EDC)  is the common  name for  1,2-dichloroethane.   It is
also commonly  referred  to as  ethylene  chloride and  S-dichloroethane (Archer,
1979).
3.2. CAS REGISTRY, RTECS, AND STORET NUMBERS
     CAS Registry:  107-06-2
     RTECS:  KI05250000
     STORET:  3^531
3.3. DESCRIPTION
     Ethylene dichloride is a clear, colorless, volatile liquid with a pleasant
odor, and is stable at ordinary temperatures (Archer,  1979).
3.U. STRUCTURE
          C1CH2CH2C1
3.5. PHYSICAL PROPERTIES OF PURE ETHYLENE DICHLORIDE
     The physical properties of pure ethylene dichloride were taken from Archer
(1979) and Weast (1980), unless otherwise stated.
     Molecular weight:      98.96
     Melting point:         -35.3°C
     Boiling point:         83.7°C
     Density at 20°C:       1.2529 g/mfi.
     Viscosity:             1.U*»51 m Pa S (cP)
     Flashpoint:
       Closed cup           17°C
       Open cup             21°C
     Explosive limits in
     air at 25°C:           6.2-15.6* by volume
                                      3-1

-------
Autoignition tempera-
ture in air:
Vapor pressure:
413°C
^C
10
20
30
kPa
 5.3
 8.5
13-3
Torr
 40
 64
100
Solubility in water:   0.0891 M (Valvani et al., 1981)
                       8,820 mg/JZ,
Log Octanol/Water
Partition Coefficient: 1.48 (Valvani et al., 1981)
Henry's Law Constant             a
atm mj mol  :          9.14 x 10   (Mabey et al., 1981)
Blood/Air Partition
Coefficient (37°C):    19.5 + 0.5 (Sato and Nakajima, 1979)
                                 3-2

-------
               4.  SAMPLING AND ANALYSIS OF ETHYLENE DICHLORIDE



4.1. SAMPLING



     Talcing environmental samples representative  of the area in  question  is a



complex task beyond the scope of this  document.   A number of documents either



devoted exclusively to sampling or  containing considerable information relating



to  the  subject  are listed below and  should be  consulted prior  to  initiating



environmental sampling.  For air, Singh et al. (1979, 1983b),  Pellizzari (1978),



and Pellizzari et al.  (1979)  have detailed sampling methodologies that describe



and/or show equipment  and strategies.  Water and wastewater sampling is described



by Berg (1982) and solids sampling  by  U.S.  EPA (19&2b).



4.2. ANALYSIS



4.2.1.  Ethylene Bichloride in Air.   Ethylene dichloride in air  can be analyzed



by a number of methods; however, the method of Singh et  al. (1980) appears to be



substantially free of  artifact  problems  and completely quantitative.   In  this



method, an air sample  in a stainless steel canister at 32 psig is connected to a



preconcentration  trap consisting  of  a  4"  x 1/16"  ID  stainless steel  tube



containing glass  beads,  glass wool, or  3% SE-30 on acid washed  100/120  mesh



Chromosorb W.  The sampling line and trap,  maintained at 90°C,  are flushed with



air from the canister; then the trap is immersed  in liquid 0_ and air is passed



through the trap, the  initial  and final pressure being noted (usually between 30



and 20 psig) on a high-precision pressure gauge.  The ideal gas law can be used to



estimate the volume of air passed through the trap.  The contents of the trap are



desorbed onto a  chromatography column by backflushing it with an inert gas while



holding the trap at boiling water temperature.  Suitable columns include 10$ SP-



1000 on  Supelcoport  (100/120  mesh,  15'  x  1/8" stainless  steel) and  0.2$ CW-

-------
1500 on Carbopack C (80/100 mesh,  10' x 1/8" Ni).  Both columns can be operated at



45°C with a carrier gas flow of 25 mil/minute on the former column and 10 mil/min-



ute on the latter.  An electron capture detector  operating at 330°C was found to



be optimum.  It should be noted that the above authors found Tenax to be unsuit-



able for air analyses because  of  the presence  of artifacts in the spectrum from



oxidation of the  Tenax monomer.  In addition, when Tenax is used as a sorbent,



safe sampling volumes (i.e., that volume of air which,  if sampled over a variety



of circumstances, will not cause significant breakthrough) should be used.  Brown



and Purnell (1979). determined  the safe volume for ethylene dichloride per gram



Tenax  to  be  27  I  (flow rate  5-600  mi/minute;  ethylene dichloride  cone.



<250 mg/nr; temp,  up  to 20°C)  with  a safe  desorption  temperature of 90°C.



     The detection limits of this method were not specified and  are dependent on



the volume of air sampled.  Analyses  as low as 33 ppt have been reported using



this method (Singh et al.,  1980).



4.2.2.   Ethylene Dichloride  in  Hater.   Ethylene dichloride  in water  can be



analyzed  by the  purge-and-trap  method (Method 502.1)  as  recommended  by the



Environmental Monitoring and Support Laboratory  of the U.S. EPA (1981).  In this



method, an inert  gas  is bubbled through 5  ml of  water at a rate of 40 mil/minute



for  11  minutes,  allowing the  purgable organic compounds  to partition into the



gas.   The gas is  passed  through a column  containing  3% OV-1  on Chromosorb W,



Tenax  GC silica  gel,  and  coconut  charcoal at  22°C, which traps  most of the



organics removed from the water.  The adsorption column is then heated rapidly to



1dO°C  and backflushed with  helium (20-60  mi/minute,  4  minutes) to desorb the



trapped organics.   The  effluent of the column is  passed  into an analytical gas



chromatography  column  packed  with  1$  SP-1000  on  Carbopack-B  (60/80  mesh,



8' x 0.1"  ID)  maintained at 40°C.   The column  is then  temperature programmed



starting  at 45°C for 3  minutes and  increasing  at  8°C/minute until  220°C is
                                       4-2

-------
reached; it is then held there for  15 minutes or until all compounds have eluted.



A halogen-specific detector (or GC-MS)  having a sensitivity of 0.10 \ig/l with a



relative standard deviation of <10$ must be used.  This method is similar to EPA



method 601,621 for use with wastewater  (Longbottom,  1982).



U.2.3.   Ethylene Dichloride in Solid  Samples.   Ethylene  dichloride  in solid



samples may be analyzed by  EPA method 8010 "Halogenated Volatile Organ!cs" (U.S.



EPA,  1982b).   With this method, a portion  of the sample is  dispersed  in poly-



ethylene glycol or methanol.  The  dispersion  is then mixed with water and purged



in a manner identical to the analysis of EDC in water.  The trap contains 3% OV-1



on Chromosorb W (60/80 mesh), Tenax  GC  (60/80 mesh),  Grade  15 silica gel (35/60



mesh),  and activated  coconut charcoal  (6/10  mesh);  desorption  is  performed at



180°C for 1 minutes with a gas flow  of  20 to  60 mi/minute.   Gas chromatographic



conditions are identical to the analysis of ethylene dichloride in water.



1.2.1.   Ethylene Dichloride  in Blood.   Ethylene dichloride  in blood  can be



analyzed by using  a  modified purge-and-trap  method (Pellizzari  et  al.,  1979).



This method involves diluting an aliquot of whole blood (with anticoagulant) to



=50 mil with prepurged, distilled water.   The mixture  is placed in a 100 mi 3-neck



round bottom flask along with a Teflon-lined magnetic stirring bar.   The necks of



the flask are equipped with a helium  inlet,  a  Tenax trap, and  a thermometer.  The



Tenax trap is a 10 cm x 1.5 cm ID glass tube  containing pre-extracted (soxhlet,



methanol, 21 hrs) and conditioned (270°C, 30  mil/minute helium flow, 20 minutes)



35/60 mesh Tenax  (=1.6 g,  6 cm).  The  sample is then heated to 50°C and purged



with a helium flow rate of 25 mi/min for 90 minutes.  Analysis  can be  performed as



indicated in Section 1.2.2.



1.2.5.   Ethylene Dichloride  in Urine.   Ethylene dichloride  in urine  can be



analyzed by using an apparatus identical to the one described in Section 1.2.1,



using 25 mH of urine, diluted to 50  ml, instead of blood.
                                      4-3

-------
U.2.6.   Ethylene Dichloride  in  Tissue.  Ethylene dichloride  in  tissue  can be



analyzed by using an apparatus identical to the one described in Section U.2.U,



using 5 g of tissue, diluted  to  50 md,  instead of blood and macerated in an ice



bath.  The purge time is reduced  to 30  minutes.

-------
                         5.   SOURCES  IN  THE  ENVIRONMENT

5.1. PRODUCTION PROCESSES

     Two  commercial  processes account  for  virtually all  of the  EDC produced

currently.  One is the direct chlorination of ethylene with chlorine.  The other

is an oxychlorination process in which ethylene, hydrogen chloride and  oxygen are

reacted to form EDC.   Currently, most production  centers around large manufac-

turing plants employing a balanced combination of both  of these two  processes.

Over  80$  of  the  EDC produced  is  used  to make  vinyl  chloride  monomer  via

dehydrohalogenation  of EDC.   Most  EDC producers have  nearby  vinyl chloride

production facilities.  The balanced plants use the hydrogen chloride recovered

when  the  EDC  is   dehydrohalogenated   to  vinyl  chloride  as  feed  to  the

oxychlorination reactor.   This requires roughly  a 50-50 split  between direct

chlorination and  oxychlorination  for a  completely balanced operation assuming

all EDC is cycled to the vinyl chloride  facilities.  In this manner,  there will

be no net production of hydrogen chloride.   As of 1971,  =58? of the  total EDC

production capacity was based upon direct chlorination and  =42$ was based upon

oxychlorination (Pervier et  al.,  1974).  Current capacities are  judged  to be

roughly the same.

     In the direct chlorination process,  EDC is produced by the catalytic vapor-

or liquid-phase chlorination of ethylene as  follows (Archer, 1979):


                                         FeCl.
                CH,  =  CH_ +  Cl- (5$ air)	^	» C1CH,CH-C1
                              *          40-50°C         *   2


Most liquid-phase processes use ferric  chloride  as the catalyst.   The  chlori-

nation is  carried out  at  40-50°C with  5%  air added  to prevent  substitution

chlorination of the product.
                                      5-1

-------
     The  oxychlorination  process is  usually  incorporated into  an integrated

vinyl chloride plant in which hydrogen  chloride (which is recovered from cracking

the EDC to vinyl chloride) is recycled  to an oxychlorination unit (Archer, 1979).

The hydrogen chloride by-product  is used as the chlorine source  in the chlorina-

tion  of  ethylene  in the  presence of  oxygen  and copper  chloride  catalyst as

follows:
                                        CuCl
               2CH-  = CH_  +  4HC1  + 0-	=-» 2C1CH_CH_C1 + H,0
                  22           2   270oc         22      2


5.2. ETHYLENE DICHLORIDE PRODUCERS

     The  producers of EDC in the United States  are listed in Table 5-1 along with

their respective production capacities.  As can be noted from Table 5-1, most of

the production  capacity is  located  in  Texas  and Louisiana.   These production

facilities also captively  consume the  major  portion of the EDC produced, as in

the  manufacture  of  vinyl  chloride   and,  to  a  much lesser  extent,   in  the

manufacture of vinylidene chloride and various chlorinated  solvents.  From Table

5-2, it can  be  noted that only =10$ of  the EDC  production has  been sold on the

open market in most  recent years.

5.3* ETHYLENE DICHLORIDE PRODUCTION AND TRENDS

     U.S. production volumes and sales  of EDC are listed in  Table 5-2.  Blackford

(1974) and the  U.S.  International Trade Commission  (1974)  have stated that the

production volumes as listed  may  be somewhat smaller than the actual production

because some EDC may be produced, but  not  separated  or  accurately measured (and

therefore not reported) by some producers.  Blackford (1971) has  estimated actual

productions on the order of  10$ higher than the  reported productions.

     The  demand  for  EDC for  the  years 1982  and  1983 has  been  estimated to be

4.127 and 4.716 million metric  tons, respectively, while the demand  for the year

1987 is expected to be 5.487 million metric tons  (CMR,  1983).  Production of EDC
                                      5-2

-------
                                   TABLE b-T
                  Major Manufacturers of Ethylene Dichloride"
Manufacture
Plant Sites
     Annual Capacity
(Millions of Metric Tons)
Atlantic Richfield (ARCO)

Borden, Inc.

Dow Chemical USA



E.I. duPont (Conoco)
Port Arthur, TX

Geismar, LA

Freeport, TX
Oyster Creek, TX
Plaquemine, LA

Lake Charles, LA
         0.200

         0.230

         0.725
         0.500
         0.840

         0.525
Ethyl Corp.
Formosa Plastics Corp.
Georgia-Pacific Corp.
B.F. Goodrich Co.
PPG Industries
Shell Chem. Co.
Union Carbide Corp.
Vulcan Materials Co.

Baton Rouge, LA
Pasadena, TX
Baton Rouge, LA
Point Comfort, TX
Plaquemine, LA
Deer Park, TX
La Porte, TX
Calvert City, KY
Convent , LA
Lake Charles, LA
Deer Park. TX
Norco, LA
Taft, LAd
Texas City, TX
Geismar, LA

0.320
0.110
0.240
0.385
0.735
0.110
0.720
O.J»50
0.360
1.230
0.620
0.540
0.070
0.070
0.160
Total 9.110
Note:  Capacities are flexible depending on finishing capacities for
       vinyl chloride and chlorinated solvents.
a
 Source:  SRI, 1983; CMR,  1983

 Operated under a toll agreement with Diamond Shamrock Corp.
«i
"Closed for an indefinite  time period because of the temporary closing
 of Shell's vinyl chloride monomer plant located there.

 Captive use only.
                                          5-3

-------
                                   TABLE 5-2
               U.S.  Production and Sales of Ethylene Dichloride3
Year
1982
1981
1980
1979
1978
1977
1976
1975
1974
1973
1972
1971
1970
1965
1960
1950
Millions of Metric
Production
3- ^55
4.523
5.037
5.349
4.989
4.987
3.617
3.617
4.156
14.214
3.541
3.428
3-383
1.113
0.575
0.138
Tons
Sales
0.640
0.401
0.510
0.633
0.469
0.692
0.617
0.345
0.596
0.613
0.656
0.595
0.596
0.140
0.198
0.021
aSource:  USITC, USTC,  1952-1982
 Production totals may  be understated  in  some years  because some
 ethylene dichloride is produced  but not  separated or accurately
 measured (and therefore not  reported)  by some  producers.
                                        5-4

-------
in 1982 was  sharply  lower than in preceding  years  (see Table 5-2).   This was



largely due  to  a  recession in  the vinyl  chloride market  and  a  decision of



producers to use their EDC inventories during the winter  of  1982 (Chemical and



Engineering News, 1982, 1983).   Production of EDC is expected to rebound during



1983.  Historically,  the demand for EDC grew at a  rate of =1% per year during the



period from  1973-1982.  Demand is  anticipated to grow  at  a rate of 1$ per year



through 1987 (CMR,  1983).



5.4. ETHYLENE DICHLORIDE USES



     A major portion (84J) of U.S.  EDC production is converted to vinyl chloride



monomer.  The major uses of EDC  for the years 1983, 1980, 1977 and 1974 are given



in Table 5-3*  Consumption of EDC for specific end uses is given in Table 5-4.



     In addition to  vinyl chloride, EDC  is  used  as  a starting material  in the



production of  chlorinated solvents  such as  1,1,1-trichloroethane,  trichloro-



ethylene  and  perchloroethylene  and  as  intermediate  in  the  production  of



ethyleneamines and vinylidene chloride (Archer,  1979; Blackford,  1974).   Table



5-5 lists  the users of EDC for these purposes.  In general, these users are also



producers  of EDC.



     EDC is  used as  an additive  (lead scavenger) in tetraethyl  lead antiknock



mixtures for  gasolines.  Scavenging agents are used in gasolines to transform the



combustion products  of lead alkyls  to forms  that  are more likely to be vaporized



from the  engine  surfaces.  The  most  important   lead scavengers  used  for this



purpose are EDC and ethylene dibromide (McCormack  et al., 1981; Blackford, 1974).



The sale of  tetraalkyl  lead  compounds  is always  in  admixture with  EDC  and/or



ethylene dibromide.   Conventional motor-mix formulations contain 1 raol of EDC and



1/2 mol of ethylene  dibromide  per mol  of  tetraethyl  lead  (McCormack et al.,



1981).   This use of EDC  is expected  to decline  in  future  years  due  to EPA



regulations concerning the use of leaded gasolines.  CMR (1982) projects that the
                                      5-5

-------
                                   TABLE 5-3
                           Ethylene Bichloride Uses
                                                   a
Percent of Total
Use
Vinyl chloride monomer
Chlorinated solvents
Vinylidene chloride
Exports
Lead scavenger
Amines
Miscellaneous
1983
en
4
2
9
—
—
1b
1980
84
7
2
5
—
--
2b
1977
80
10
—
—
•3
—
7°
1974
78
8
—
~
3
2
9°
aSource:  CMR, 1974, 1977, 1980, 1983
 Includes lead scavenger
 Includes exports
                                           5-6

-------
                                  TABLE 5-t






             Consumption of Ethylene Dichloride in 1979 and 1974a
Consumption (Thousands of Metric Tons)

Vinyl chloride monomer
1,1,1 -Trichloroe thane
Trichloroethylene
Perchloroe thy lene
Vinylidene chloride
Ethyleneamines
Lead scavenger
Exports
Preparation of pclysulfides
Paints, coatings and adhesives
Extraction solvents
Cleaning of fabric and polyvinyl
chloride equipment
Grain fumigation
Other uses
Total
1979 • %
4420 85.0
—
89.1 1.7
—
118.7 2.3
168.1 3.2
81.7 1.6
179.0 3.4
0.5
1.36
1.05
0.91
0.46
0.46
5199. W 99.8
1974
3891.2
153-3
132.8
124.7
97.0
131.5
97.0
167.3
—
—
—
—
—
6.8
14804.4
%
81.0
3.2
2.8
2.6
2.0
2.7
2.0
3.5
—
—
—
—
—
0.1
99.9
aSource:  Seufert et al.,  1980;  Blackford,  1974
                                         5-7

-------
                                                           TABLE 5-5
                                    Users of Ethylene Dichloride for Intermediate Purposes
                                                                                          a
i
00
                                                                                                            Vinylidene
                               1,1,1-Trichloroethane  Trichloroethylene  Perchloroethylene  Ethyleneamines  Chloride
Dow Chem. USA
Freeport, TX
Plaqueraine, LA

X
X

X X
X

X X
	 X
PPG Industries

     Lake Charles, LA

Union Carbide Co.

     Taft, LA
     Texas City, TX

Diamond Shamrock Corp.

     Deer Park, TX

E.I. duPont

     Corpus Christ!, TX
                                                                                                                X
                                                                                                                X
      aSource:  SRI, 1983; Blackford,  1971
      X = produces this chemical from  ethylene dichloride

-------
demand for lead  in  gasoline  will decrease at a rate  of  11$  per year (average)



through 1986.



     Exports of EDO have been increasing in recent years  (see Table 5-6) and may



grow by as much as 8$ per year through the mid 1980s (CMR,  1983)*  The Japanese



are expected  to  become major importers  of  EDC as they  close  down significant



amounts of  chlorine capacity (mercury  cell method)  because  of environmental



regulations  (some  of the chlorine made  by  this method  is used to  make EDC).



However, part of that demand will be  filled by  Shell's one billion pounds/year



EDC plant in Jubail, Saudi Arabia,  which is  due on stream in 1985.



     EDC has a variety of relatively small miscellaneous uses.   It is used as a



solvent in applications that  include textile and PVC cleaning, metal degreasing,



extractions,  and use in paints, coatings and adhesives.  As an intermediate, EDC



is used to  produce polysulfide  elastomers  and  ethylenimine (aziridine).   Dow



chemical captively  consumes  several  million pounds  per year  of EDC  at their



Freeport,  TX, facility to produce ethylenimine.  EDC is used as  a grain fumigant;



Auerbach Associates (1978) estimated grain fumigant uses;  Metcalf  (1981) lists



EDC as  a fumigant  for  use  in   household  and soil applications.    Other  uses



mentioned for EDC  include  varnish  and  finish  removers,   soaps and  scouring



compounds,  wetting and penetrating  agents and  ore flotation (Hawley,  1981).



5.5.  SOURCES OF EMISSIONS



     EDC  can  enter  the  environment  through  atmospheric  emissions,  waste



effluents to waterways, and  land disposals of liquid  and solid wastes.   EDC in



liquid and  solid waters  evaporates  rapidly  because   of  its  high  volatility;



consequently, releases to land and  water can be expected to enter predominantly



into the atmosphere.  The sources of  EDC  emissions  are during EDC manufacture,



intermediate use of EDC in production of other chemicals, and dispersive uses of



EDC in end-point product applications.
                                      5-9

-------
                                    TABLE 5-6






        Estimated Environmental Releases of Ethylene Dichloride in 1979a

Application
Production of 1 ,2-dichloroethane
Indirect production
Feedstock uses
Trichloroethylene
Tetrachloroethylene
1 , 1-Dichloroethylene
Ethyleneamines
Preparation of polysulfides
Vinyl chloride
Exports
Dispersive uses
Lead scavenging
Fabric and PVC equipment cleaning
Paints, coatings, adhesives
Extraction solvents
Grain fumigation
Miscellaneous uses
Total
Releases (metric
Air Water
6,151 61
65 191

73
138
86
104 	
5
136
180

956
864
1,364
1,000
460
300 	
11,885 252
tons)
Solid waste
	
	

	
	
	
	
5
	
	

	
46
	
50
	
	
101
aSource:  Seufert et al.,  1980
                                            5-10

-------
     A number of estimates have  been generated that predict  the  amount  of EDO



that ±a released to the environment.   These estimates are based,  in large part,



on a sampling of monitoring data and a variety of engineering estimates.  In many



instances, the impact of current control technology may not have been adequately



assessed.  Therefore,  the estimates  that have been  generated for EDC releases



should not be interpreted as  exact measurements, but are best regarded as order



of magnitude estimates.  Estimates of releases are in Table 5-6.



     Table 5-6  lists sources of  EDC emissions  in  1979  and  estimates  of the



amounts released in  air,  water,  and solid waste.   Total  1979 emissions  to the



atmosphere, water,  and solid  waste amounted to 12,238 metric tons.  The data in



Table 5-6 indicate that gaseous emissions from EDC production amounted to 52.3$



of  total  atmospheric releases,  while feedstock uses,  dispersive applications,



and exports  accounted for 12.6,  33-6, and  1.5$,  respectively (Seufert et al.,



1980).



     Utilizing  data  from  EDC  producers,   state  and  local  emissions  control



agencies and the open literature, Hobbs and Key (1978) estimated  1978 emissions



of  EDC at =11,000  metric tons.   The degree  of  current  emissions control on



domestic  processes  involved  in  the primary  production of EDC was assessed by



Hobbs and Key (1978).



     Estimates  by  SRI  International  (U.S.  EPA,  1979a)  indicated  that  total



domestic  emissions  of EDC from primary production,  fugitive  sources, and tank



storage are  44,000 metric tons  or =0.8$  of the amount produced.   The impact of



current control technology was not addressed in the  report.



     Eimutis  and  Quill (1977) estimated process  emissions of EDC for 1977 at



50,000 metric tons.  Presumably,  this estimate  did  not  take into account current



control technology.
                                      5-11

-------
     For  197*4,  Patterson et al.  (1975)  estimated total  domestic  emissions at




71,000 metric tons.  Total  domestic  emissions for 1973 were reported at 54,000



metric tons by Sharael et al. (1975).  The sources of EDC releases are discussed



below.



5.5.1. Production and Related  Facilities.   In general, EDC is produced commer-



cially at  an integrated manufacturing  facility that  uses  some .or all  of the




produced EDC to make vinyl chloride and, in some cases, other  derivatives.  In a




typical vinyl  chloride  manufacturing facility, waste  streams  are  generated by




the three distinct processes:  direct chlorination of  ethylene, oxychlorination




of  ethylene,  and dehydrochlorination  to vinyl  chloride;   these are  typically



combined at a given facility for recovery,  treatment  and disposal.  The specific




number of  point sources of releases at  a  manufacturing site  is a  function of



plant design.  Point sources of EDC loss from an integrated  vinyl chloride plant



include direct and oxychlorination reactor  vent streams, light-ends distillation



column vent,  heavy-ends from  the EDC  recovery tower, wastewater  from drying




columns and scrubbers,  and  fugitive  emissions from storage, pumps,  seals, etc.



(Catalytic, 1979; Drury and Mammons,  ?979).   Releases from these manufacturing



sites are mostly to the  atmosphere and arise largely from vent  gas streams (Drury




and Hammons, 1979).  Control devices used to  limit EDC escape  to the atmosphere




include thermal oxidizers,  catalytic oxidizers, vent condensers, scrubbers, and



vent gas post-reactors  (Hobbs and Key, 1978).




     Combined  wastewaters  that are  generated  from  EDC manufacture  (vent gas




scrubbers, water produced during oxychlorination, and  washwater) are treated in




several ways depending upon  the plant (Catalytic, 1979). Treatments include pre-



treatment and steam  stripping  prior  to  biological treatment,  incineration of a




portion of  the  waste stream, neutralization  and  chemical treatment,  secondary



treatments and  final discharges  to  surface  waters,  to public owned treatment
                                      5-12

-------
works, or to deep-well injection.  Estimates of  the  amounts  of EDC released to



the  surface  waters are  dependent upon  engineering estimates  of  the  overall



success of treatments and the volume of wastewater flow.



     Solid wastes generated  at  an integrated vinyl  chloride  plant are usually



treated to recover organic compounds present.  Wastes are subsequently disposed



of  in a  landfill or  incinerated,  recovering  chlorine  as   hydrogen  chloride



(McPherson et al., 1979).  Some solid wastes, possibly tars and heavy-ends, may



be  a  suitable  feedstock  for  tetrachloroethylene/carbon  tetrachloride  via  a



chlorination process.



5.5.2.  Dispersive Uses.



     5.5.2.1.  LEAD SCAVENGING — Seufert et al.  (1980)  estimated that 956 metric



tons  of  EDC were released  to  the  environment  in  1979  from  lead scavenging



applications.   Releases  occur during  blending  of the gasolines,  refueling of



automoblies, filling  and-evaporation  from gasoline storage tanks, and combustion



of  the gasoline.   Approximately  1Jt of the EDC used  for lead  scavenging is



estimated to be  released into the environment;  the remainder  is  converted to



hydrogen chloride and then to lead chloride during combustion.



     5.5.2.2.  PAINTS, COATINGS, ADHESIVES — In  this  application, EDC is used as



a solvent that is allowed to evaporate. Therefore,  all of  the EDC  is emitted to



the environment.



     5.5.2.3.  GRAIN FUMIGATION — Fumigants are defined as gaseous pesticides.



They must remain in the gas or vapor  state and in sufficient concentration to be



lethal to  the  target pest species.   Therefore, all of  the  EDC  used  for this



purpose will be vented to the atmosphere,  as there are no control methods used.



     5.5.2.4.  FABRIC AND PVC CLEANING — It is presumed that most EDC used for



cleaning purposes will eventually be emitted to  the atmosphere.  Some cleaning



wastes from PVC reactors may be drummed for landfill disposal.
                                      5-13

-------
     5.5.2.5.  OTHER  DISPERSIVE USES —  In  other dispersive uses,  it has been



assumed that most EDC will eventually be emitted  to  the atmosphere.  Some may be



landfill, as with extraction solvents,  and some may be incinerated.



5.5.3.   Conclusions.    The major  sources of EDC  emissions are  from  vent gas



streams released during the manufacture of EDC and subsequent integrated produc-



tion of vinyl chloride monomer, and  other EDC derivatives.  Significant amounts



of EDC are  also  released to the environment from dispersive uses.  In 1979, an



estimated 6,696  metric tons of EDC were released  to the  atmosphere  from EDC



production and feedstock uses,  while an  estimated 4,944 metric tons were released



to the atmosphere from dispersive  uses  (see  Table 5-6).
                                      5-14

-------
                   6.  TRANSPORT AND FATE IN THE ENVIRONMENT



     The transport and fate of EDC  in  the environment depend on  the  medium in



which it is present.   The fate and transport of EDC in three environmental media



(atmosphere, water and soil) are discussed below.  Since a large percent of EDC



emitted into the environment  is  in  the  atmospheric  medium (see Chapter 5), the



fate and transport of the chemical  in this medium deserve special attention.



6.1. ATMOSPHERE



     The fate  of  EDC in the  atmosphere is dictated by its ability to undergo



chemical and  physical removal processes  in this  medium.   Reaction  with *OH



radicals is  the principal  chemical  process by  which many organic compounds,



including EDC,  are removed from the atmosphere (Crutzen and Fishman,  1977; Singh,



1977; Altshuller,  1979;  Cupitt, 1980).    Photolysis  of 0  in  the troposphere



produces singlet atomic oxygen [01Q]  that then reacts with water vapor to produce



•OH radicals.  The tropospheric  half-life (t1/?)  of a compound is related to the



•OH radical concentration according to the  expression t 1/2 = [0.693] [K(-OH)]~1



where K is the  rate  constant  of  the  reaction and [«OH]  is the concentration of



•OH radicals.



     It is obvious  from the above equation that  an estimation of the tropospheric



half-life for EDC due to  -OH radical reaction requires that the values of both K



and  [-OH]  be  known.   There  are only  two measurements  of EDC  reaction  rate



constant with -OH radicals.  The absolute rate constant for EDC reaction with -OH



radicals was determined by Howard and Evenson (1976) in a conventional discharge



flow system.   The value of the rate constant obtained in the system at pressures



ranging from  0.7  to  7 mm Hg  and at a  temperature of  23°C was 22  +  5 x  10~14



(standard deviation of average)  cm   molecule'   sec~  .
                                      6-1

-------
     The rate constant for the reaction of «OH radicals with EDC in the presence



of 02  and  Ng was determined  by  Butler et al.  (1978).   At  29.5°C and  a total



pressure of 400  mm Hg,  the  reaction rate  constant  was determined  to  have  a



probable value of 6.5 x 10~14 cm3 molecule'1  sec~1, with an  upper limit value of



29 x 10~11*  cm3 molecule'1 sec~1.   Although Butler et al. (1978) argued that the



presence of oxygen  and nitrogen at a pressure of 400  mm Hg was more represen-



tative  of  actual tropospheric  conditions,  the presence of 02 and N? actually



complicated the reaction scheme through side reactions and made the extraction of



kinetic data more difficult.  It is for this reason that  Butler et al. (1978)



failed  to  determine  the  precision  of  the  determined  rate  constant  value.

                                                              _1H   3         _!
Therefore,  the measured  rate constant value of 22.0+5 x  10   cm  molecule



sec'1 for EDC reaction with -OH radicals as reported by Howard and Evenson (1976)



appears to  be more  accurate than  the value of 6.5 x  10~    cm molecule   sec



given by Butler  et  al.  (1978).



     In  Table 6-1,  the rate constants  for  a number  of  chlorinated ethanes,



including EDC, at room temperature have been  shown.   It  seems clear from this



table  that  a value  of 22.0 x  10~   cm molecule"  sec~ is  more consistent with


                                                                            -11
the measured K values for other  chlorinated  ethanes  than a  value of 6.5 x 10



cm  molecule  sec



     On the basis of the above discussion, it  is reasonable to accept  a value of



22 x 10~    cm  molecule'  sec'  as the rate constant  value  for EDC reaction with



•OH radicals at  296°K (room  temperature).



     It should be recognized that the value of K is  dependent on temperature.



The  temperature  in  the troposphere  is dependent  on latitude,  altitude  and



seasonal variations.   The average annual tropospheric temperature weighted over
                                       6-2

-------
                                   TABLE 6-1

                 Rate Constants for a Few Chlorinated Ethanes
                              at Room Temperature
Compound
                  1U
Rate Constant x 10

cnr molecule*"  sec"
                                                           Reference
CH.CH2C1
CH2C1CH2C1
     39, W



       26


     22, 6.5



       33
Howard and Evenson,
1976; Butler et al.,
1978

Howard and Evenson,
1976

Howard and Evenson,
1976; Butler et al.,
1978

Singh et al.,  19&1
                                        6-3

-------
those variables is close  to  265°K,  or -8°C (Altshuller,  1979).   Therefore,  the


evaluation  of  reaction  rate  at  this  temperature  representing  an  average


tropospheric  reaction  rate must incorporate the  rate  constant  value  at 265°K.


When the rate constant is  not known,  Altshuller (1979) estimated it by dividing



the  K  value  at 298°K  by  1.75.    This  is  an empirical  relationship and  is
applicable only to saturated organic compounds.  Therefore, the rate constant for


                                                     x 1
                                                     i—

                                                    1 7"5
                                                         _
                                                  22 x 10   ^         -1    -1
EDC reaction with -OH at 265°K can be estimated as - — i— ^ cm^ molecule   sec  ,
or 12.6 x  10    car molecule'   sec"  "


     The second factor required for the evaluation of tropospheric half-life for



EDC  reaction with  «OH radicals  is  the  average  -OH concentration.    Besides



hemispheric difference, the  concentration of  -OH  radicals in the troposphere is



dependent  on  latitude,  altitude and seasonal variations.   The concentration of



tropospheric  »OH radicals in the Northern Hemisphere across  the latitudes of the



continental United States has been estimated by different  investigators to range



from 0.2 to  0.9  x  10  molecules cm"3 annually  (Logan et al.f 1981; Crutzen and



Fishman, 1977;  Neely and  Plonka,  1978).   Singh  et al.  (1983a)  have recently



estimated  a mean hydroxyl  radical  concentration of 0.1-0.6 x 10  molecules cm"



over the troposphere of the continental United States.  Therefore, a  value of 0.5



x  10   molecules  cm   is a good estimate for the  average tropospheric hydroxyl



radical  concentration  over  the  continental  United  States.    It  should  be



emphasized that  the «OH concentration can  vary substantially from the average



value of 0.5 x 10  molecules  cm  .   Calvert  (1976) estimated the average ambient



level  of »OH  radicals  in  the morning hours in  Los Angeles  to be =2.6 + 2 x 10



molecules  cm   .   Assuming equal periods of daylight and  darkness,  the average

                                                  fi             T
concentration  for a 2^-hour  day should be =1  x  10  molecules cm" .



     The  calculations  of  tropospheric  half-life  for  EDC  reaction   with  -OH



radical  can  be made under two scenarios.   In  the first  case,  a rate constant



value  at  ambient  temperature  (22 x  10~    cm   molecule"   sec" )  and  an  «OH
                                       6-4

-------
 concentration of 1 x 10  molecule cm~^ can  be used  to derive the half-life  value
 of 36  days.  In the second case, a rate constant value  of 265°K corresponding  to
 annual average tropospheric temperature over the continental United States  (12.6
 x  10~    cm3 molecule'   sec   )  and a  value  of -OH concentration of 0.5 x 10
 molecules cm"^ corresponding to the annual  average  tropospheric concentration  of
 •OH radical over the continental United States can be used to  derive  a half-life
 value  of 127 days.  Therefore, the tropospheric half-life  for EDC reaction with
 •OH may vary from 36 to  127 days.
     The reactivity of EDC with  chlorine atoms was studied by Spence and Hanst
 (1978).  These  investigators  found that the chlorine-sensitized photooxidation
 of 10  ppm  EDC  with  4  ppm C12  at 22.5°C  and irradiated by UV lamp  (365   nm
 wavelength max.)  and  sun lamp  (310  nm wavelength  max.)  produced a number   of
 reaction  products.    Although  hydrochloric acid   (6.5  ppm), carbon  monoxide
 (1  ppm), formyl chloride (3.5 ppm), C02 (2 ppm) and chloroacetyl chloride (0.5
 ppm) were formed,  no phosgene could be detected as a reaction product.  EDC was
 found  to have  a relatively low  reactivity toward  Cl  atoms  compared  to other
 chlorinated ethanes.
     That  a chlorine-sensitized  photooxidation  is representative  of  actual
 tropospheric conditions has been contested  by  Singh (1978).   This investigator
 estimated that chlorine atom concentrations in  simulation  chambers can be 10^  to
 10  times larger than in the troposphere. Thus,  the  role of chlorine atoms would
 be  minimal  under  actual   tropospheric  conditions  where  -OH  radicals  are
 predominant.
     The role of halocarbons  in the catalytic destruction of  ozone (0,) is well
documented  (Altshuller,  1979;  Altshuller  and  Hanst,   1975).   Any  halogenated
compound with a sufficiently long lifetime may be transported to the stratosphere
where  photolytic  mechanisms  may release halogen atoms  to participate  in the
                                      6-5

-------
destruction of ozone.  The  rate  of reaction of -OH radical with halocarbons is


one mechanism that competes with  stratospheric photolysis as a halocarbon sink.


     Since  transport to  the stratosphere  is a  relatively slow  process,  the


reported half-life  for EDC  suggests that only a small fraction released in the


troposphere will  reach the  stratosphere.  Thus, emissions  of  EDC  would not be


expected  to play  a significant  role  in  the catalytic destruction  of ozone.


However, in the view of Altshuller  and  Hanst  (1975), EDC should be considered a


threat  in  this regard since  the  production volume of EDC  is  so  large.  These


investigators cite chloroacetyl chloride as  the sole product of  EDC  reaction with


•OH.  The possibility that this reaction product may have sufficient stability to


travel to the stratosphere was raised.   If formed in the troposphere, most of the


chloroacetyl chloride probably will be  washed out by rain.   If  it travels to the


stratosphere, however, it may undergo  photodissociation to form  atomic  chlorine.


     The three most likely physical processes  that may participate  in the removal


of EDC from the atmosphere were studied by Cupitt  (1980).  The  half-life for EDC


removal by rain droplets  was  estimated  to be  390 years.  EDC in the vapor phase


may be adsorbed on  aerosol  particles and removed  from the atmosphere with the


aerosol.  However, the half-life  of this removal process was estimated to be =13


years (Cupitt, 1980). It  has  been estimated by Cupitt (1980) that compounds with

                                     _7
saturation  vapor  pressures  of  <10     mm  Hg  are  likely to  be adsorbed  on


atmospheric aerosols.  Since  the vapor pressure of EDC  is much  higher, it is not


expected to be adsorbed  by  aerosol, and the probability of its removal by this


process seems to  be  remote.


     The  third physical  removal mechanism,  dry deposition of EDC, was  also


studied by Cupitt (1980).   He estimated a half-life value  of 25 years  for this


process.  Therefore, it is not expected to be a significant  EDC  removal mechanism


from the atmosphere.
                                       6-6

-------
     It is apparent  from  the above discussion that  physical  processes are not



significant  for  the  removal of  EDO from  the atmosphere.    Of  the  chemical



processes, the most significant reaction for removal of EDO from the atmosphere



is its reaction with «OH  radicals.  The  half-life  for this reaction  has been



estimated to  be -36 to 127  days.   Therefore,  it is anticipated  that  EDC will



persist for a sufficiently long time in the atmosphere that it may participate in



intramedia transport from its source of emissions, but not sufficiently long to



allow for transport to the stratosphere.



6.2. AQUATIC MEDIA



     The  fate of  EDC in aquatic  media  is  expected  to  be determined  by its



chemical, physical and microbiological removal mechanisms.  The fate of EDC with



respect to its possible chemical reactions has been reported by Callahan et al.



(1979).  Both photolysis and hydrolysis of EDC in aquatic media are expected to



be insignificant fate processes (Callahan et  al.,  1979).  Based on the oxidation



rate constant data given by  Mabey  et al.  (1981)  and the  concentration of free



radicals in aquatic media given by  Mill et al.  (1982), the oxidation of EDC by



singlet  oxygen  (K«360 M~   hr~ )  and  peroxy  radicals  (1  M~   hr~ )  can  be



predicted to be environmentally insignificant processes.



     The degradation  possibilities  of  EDC by  microorganisms  in  aquatic media



have been studied  by several authors.  From the results of the laboratory tests



conducted by several  investigators with activated  sludge or  sewage  seed  as



microbial inoculum in aquatic media, it  can be concluded that EDC may biodegrade



slowly under these conditions (Price et al.,  1974; Tabak  et al.,  1981; Ludzack



and Etlinger, 1960;  Henckelikian  and Rand,   1955; Stover  and  Kincannon, 1983).



Under methanogenic  conditions,  Bouwer   and  McCarty  (1983)  observed up  to 50$



degradation of EDC to C02 over a 25 week period.  In a  river die-away test, Mudder



et al. (1982) reported no  degradation  of EDC at  concentrations of =1  ppm and
                                      6-7

-------
above.   Stuck!  et al.  (1981,  1983)  isolated an organism from  soil  capable of




utilizing EDC as  a sole source of carbon.  The bacterium could not grow on solid




media but grew in liquid culture.  No degradation studies were performed in soil



so that  no assessment can  be made of the ability of this organism to degrade EDC




in  the  environment.    It  can be   concluded  from   these  discussions  that




biodegradation will  be  an  insignificant  fate process for EDC in ambient aquatic




media.



     The two likely  physical processes that may remove EDC from aquatic media are




sorption and volatilization. The removal of EDC by sorption  on sediments and the



subsequent sedimentation is probably not a significant  process (Callahan et al.,




1979; Mabey et al.,  1981).  A  calculation  based  on  EXAMS modeling by the method




of Burns et  al.  (1982)  with the input parameters given  in  Section 3 also shows



that EDC is  not  likely to be  significantly  sorbed onto  particulate  matter in




water.



     The evaporation half-life  for  EDC  from water  at a  depth  of  1.5  cm,  a




concentration of  2 ppra, and a  temperature  of 24°C was estimated to be 8 minutes




under stirring (Chiou et al.,  1980).   Under somewhat more realistic conditions of




a wind speed of 3  m/sec, water current of 1  m/sec, and a water depth of  1 m, Lyman



et al.   (1982) calculated  the  evaporation  half-life of  EDC  as U hours.  Both of




these methods,  however, neglect a number  of important environmental variables




such as  sorption, advection, and diffusion.  These are  particularly important in



bodies of water deeper  than one  mile.




     The bioconcentration factor   (BCF)  for  EDC  in  aquatic organisms  was




estimated  as 6.03  by  Kenaga  (1980)  from  the  water  solubility and  a linear



regression  equation.   In  a  14-day  exposure  experiment  with  bluegill sunfish



(Lepomis macrochirus).  Barrows et al. (1980)  determined a  BCF  of  2.0 for  this



compound.  Therefore, EDC  is not likely  to bioconcentrate in aquatic organisms.
                                       6-8

-------
     It  can  be concluded  from the discussion  above that  the  primary removal
mechanism of EDC from aquatic  media is  probably volatilization.  The half-life
for volatilization is such that the compound may persist in aquatic media for a
reasonable period and may participate in transport in aquatic bodies.
6.3- SOIL
     Data  pertaining  to  the  fate of  EDC in  soil are very  limited.   If one
considers the chemical reactivity of EDC in aquatic media (see Section 6.2), it
is possible  to  speculate that chemical reactions  of  EDC  may  be faster in soil
than in  water,  particularly with  soil  acclimatized with  EDC.   However,  Wilson
et al. (1981) found virtually  no biodegradation  of  EDC with soils collected near
Ada, OK, that contained an average sand of 92$ and organic carbon of 0.09$.
     The sorption constant (K   ) of EDC on soils was predicted by Kenaga (1980)
                             oc
to be  43 (standardized  with respect to  soil organic content)  from regression
equation and solubility  of this chemical in water.  Chiou et al.  (1979) estimated
a value of 19 for K   with Willamette  silt loam containing 1.6$ organic matter,
26$ clay, 3-3$ sand and 69$ silt.   Therefore,  the compound is not expected to be
sorbed  strongly onto soils,  particularly onto soils  containing  low  organic
matter, and may percolate through  the soil column.  This  has  been confirmed by
Wilson et al. (1980),  who found *50$  percolation of EDC through a column of 140
cm depth containing the previously described  Oklahoma soil.   The fact that Page
(1981) reported the detection of  EDC in  10$  groundwater samples  in New Jersey
also indicates that the compound may  transport downwards through some soils.
     The volatilization of EDC from soil may  be an important removal mechanism.
However, very few investigative data are available on this  subject.  Again, if
one considers the investigation of Wilson et al.  (1981) that reported the loss of
50$ EDC  from the  soil   column  through  volatilization, one  can  conclude  that
volatilization may  be one of the most significant physical removal processes for
EDC from soil.

                                     6-9

-------
6.4. SUMMARY
     The most likely  removal mechanism for EDC from the atmosphere is reaction
with  hydroxyl  free  radicals.   Based on  available kinetic  data  and  average
tropospheric hydroxyl free radical concentrations, the  half-life of  EDC has been
estimated to range from 36-127  days.
     EDC  is not  expected  to play a  significant role  in  stratospheric ozone
destruction  reactions  due  to  its  relatively  short   tropospheric half-life.
However, chloroacetyl chloride  may have  suffficient  stability to diffuse to the
stratosphere and may  participate  in UV reactions  producing chlorine atoms.
     In the aquatic environment,  the  most  significant  removal mechanism appears
to be its volatilization.   The  half-life for  this process has been  estimated to
be =4 hours.   Therefore,  EDC may persist  in  the  aquatic media for  a reasonable
period and may participate  in its transport in water bodies.
     From the little  information  that is available regarding the fate of EDC in
soil, it has been  concluded that both volatilization  from and leaching through
soil are the two significant removal  mechanisms for EDC.
                                      6-10

-------
                     7.   ENVIRONMENTAL LEVELS AND EXPOSURE



7.1. ENVIRONMENTAL LEVELS



     EDC contamination of the environment can  result  from the manufacture,  use



and disposal of the chemical.  Sources of environmental release and quantitative



estimates of releases are discussed in Section 5 of this report.  This section is



concerned with the levels of EDC  that  have  been detected in  various  areas  and



monitoring sites.  Sizable quantities of EDC were  judged to be released to the



air during manufacture and conversion to vinyl chloride at an integrated manufac-



turing site.  This judgment is borne  out  by the high ambient concentrations of



EDC found near several of these sites.



7.1.1.   Atmospheric  Levels.   In  addition  to atmospheric releases  of  EDC from



manufacture and feedstock uses,  EDC can be  released  to  the  air from dispersive



uses such as  grain fumigation,  solvent uses in  paints,  coating and adhesives,



cleaning applications, and gasoline uses related to lead scavenging.



     Recent air  monitoring  data  for EDC are  listed  in Table 7-1.   These data



indicate that EDC is  often present in ambient air in urban and industrial areas,



especially near plants manufacturing or using the chemical. Measurements made at



sites in three geographical areas central to EDC production and user facilities



indicated EDC concentrations as  high  as 184 ppb  (Elfers,  1979).   These levels



were  associated  with  industrial  sources  and  occurred  during  atmospheric



conditions of calm or low wind  speeds.   Measurements were  made during 10- to 13-



day periods at Lake Charles and New Orleans, LA, and  at  Calvert City,  KY.  The



highest concentrations were reported for the Lake Charles  area, with the highest



single concentration  being 184  ppb  and the  average detectable  concentration



being 27.5  ppb.   In  Calvert City, ambient  concentrations ranged  from <0.12 to
                                      7-1

-------
                   TABLE 7-1



Ambient Atmospheric Levels of Ethylene Dichlonde
Location
ALABAMA
Birmingham

Hirmingham
ARIZONA
Phoenix
Grand Canyon
CALIFORNIA
borcinguez

Loa Angelea
Oakland
71 Ri'/erside
w Upland

Upland
COLORADO
Denver
ILLINOIS
Ch i cago
KENTUCKY
Calvert City

LOUISIANA
Baton Rouge
Baton Rouge
GeLsmar
[berville Pariah
Lake Charles
Type or Si'e

urban

urban

urban
rural

urban

urban
urban
urban
urban

urban

urban

urban

12 cities near
chera. plant

urban
urban
Industrial
urban
urban
Date

April, 1977

April, 1977

April-Hay, 1979
Nov-Dec, 1977

May, 1976

April, 1979
June-July, 1979
July, 1980


Aug-Sept, 1977

June, 19BO

April, 1981

Aug-Sept, 1978


March. 1977
March, 1977
Feb. Mar, 1977
Jan, Feb. 1977
June-Oct, 1978
Detection
Method

GC/MS

GC/MS

GC/ECD


GC/MS

CC/ECD
CC/ECD
GC/ECD
CC/MS

CC/MS

GC/ECD

GC/ECD

GC/MS


GC/MS
GC/MS
GC/MS
GC/MS
GC/MS
Number of
Samples

2

7 (1 ND)

NR
7 (7 ND)

1



102
B

16 (11 ND)

95

NR

88


25 (9 ND)
18 (2 ND)
10 (1 ND)
11
98
Concentration (ppt. v/v)
Max. Mln. Avg.

98.6

99

1451.1
—

—

1351.8
803. 0
580
212.9

210

480

2820

17800


630
2556.5
2551.5
11614.2
61000

50.7

0

38.8
—

—

173.0
38.3
89
61.8

0

100

22

<120


0
19.3
21.7
. 2.2
150

7U.7

35

216.3
—

3662.3

519.2
82.5
360
110.3

27

210

195

5000


270
129.1
760.1
320.9
27000
Reference

Pellizzan and
Bunch, 1979
Pelllzzan, 1979a

Singh et al., I960
Pellizzan, 1979a

Pellizzari and
Bunch, 1979
Singh et al., 1980
Singh et al., I960
Singh et al., 1982
Pellizzan and
Bunch, 1979
Pellizzari, 1979a

Singh et al., 1980

Singh et al., 1982

Elfers, 1979


Pellizzari et al . , 1979
Pellizzari, 1978a
Pellizzari, 1978a
PeHlzzan, 1978a
Pellizzari, 1979b

-------
TABLE 7-1 (cont.)
Location
LOUISIANA (cont.)
Lake Charles

Lake Charles

New Orleans

Plaquemine

MISSOURI
St. Louis
NEW JERSEY
^ Bats to

<•»» Bound Brook

Bridgeport

Bridgewater
Burlington

Caraden

Carlstadt
Clifton

Deepuater

East Brunswick

Edison

Edison

Type of Site

urban

12 sites near
chera. plant
near chem.
plant
urban


urban

rural

off highway






urban

urban
urban

near chem.
plant
marina

urban

near waste
disposal area
Date

June, 1978

Sept-Oct, 1978

October, 1978

Jan-Feb, 1977


May- June, 1980

Feb-Dec, 1979

March, 1976

September, 1977

July-Aug, 1978
September, 1977

Apr-Oct, 1979

September, 1979
March, 1976

June, 1977

July, 1976

Mar-July, 1976

Mar-July, 1976

Detection
Method

GC/MS

GC/MS

GC/MS

GC/MS


GC/ECD

GC/FID-ECD-MS

GC/MS

GC/MS

NR
GC/MS

GC/FID-EDC-MS

GC/MS
GC/MS

GC/MS

GC/MS

GC/MS

GC/MS

Number of
Samples

6

110

91

11


90

42 (40 ND)

1

2

22 (18 ND)
1

23 (21 ND)

16 (7 ND)
1

6 (3 traces)

2

33 (20 ND)

16 (1 trace)

Concentration (ppt. v/v)a
Max. Min. Avg.

307.3

184000

Hi 700

921.1


260

—

—

—

380
—

—

998.0
—

13.1

85.8

3400

14091.5


5.2

<120

<120

2.2


65

—

—

—

0
—

	

33-4
—

5.9

37.1

8.6

53.6


86.5

27500

2900

337.2


120

Trace
(2 samples)
Trace

Trace

170
Trace

Trace
(2 samples)
537.2
15950

7.4

61.6

1600

2712

Reference

Pellizzarl and
Bunch, 1979
Elfers, 1979

Elfers, 1979

Pellizzari and
Bunch, 1979

Singh et al.,

Bozzelli et al

Pellizzarl and
Bunch, 1979
Pellizzari and
Bunch, 1979
Bozzelli et al
Pellizzarl and
Bunch, 1979
Bozzelli et al












1980

., 1980





., 1979


., 1980

Pellizzarl, 1978b
Pellizzari and
Bunch, 1979
Pellizzari and
Bunch, 1979
Pellizzari and
Bunch, 1979
Pellizzari and
Bunch, 1979
Pellizzari and
Bunch, 1979











-------
TABLE 7-1 (cent.)
Location
NEW JERSEY Ccont.]
Elizabeth
Elizabeth

Fords

Hoboken

Linden

Linden
Middlesex
Newark
Newark

Newark



Hdssaic

Haterson

Rahuay
Rutherford

Rutherford
Sayreville

Somerset
South Amboy
New YORK
Niagara Falls

Staten Island

Staten Island
Type of Site

urban
near indus-
trial plant
urban

urban

Industrial

urban
suburban
urban
urban

urban








suburban

suburban



industrial

residential
(Love Canal)
urban

urban
Date

Sept 1978-Dec, 1979
Jan-Dec, 1979

March, 1976

March, 1976

June, Nov. 1977

June, 1977
July, 1978 '
Jan -Dec, 1979
March, 1976

Mar, 1976-Dec, 1979



March, 1976

March, 1976

September, 1978
May, 1978-Dec, 1979

May, 1979
July, 1976

July, 1978
Jan-Dec, 1979

February, 1978

November, 1976

Mar-Apr, 1981
Detection
Method

GC/FID-ECD-MS
GC/FID-ECD-MS

CC/MS

GC/MS

GC/MS

GC/MS
NR
GC/FID-ECD-MS
GC/MS

GC/FID-ECD-MS



GC/MS

GC/MS

GC/MS
GC/FID-ECD-MS

GC/FID-ECD-MS
GC/MS

NR
GC/FID-ECD-MS

GC/MS

GC/MS

GC/ECD
Number of
Samples

71 (53 ND)
54 (7 trace, 16 ND)

1

2 (2 ND)
%
16 (1 trace)

11 (1 ND)
IB (13 ND)
37 (9 trace, 28 ND)
1

160 (112 ND)



1

1

16
196 (141 ND)

46 (3 trace, 42 ND)
1

29 (17 ND)
48 <4 trace, 44 ND]

9 (2 trace, 7 ND)

3

NR
Concentration (ppt, v/v)a
Max. Mln. Avg.

2200
—

—

0

48.2

1.9
290
—
—

5800



—

—

77-3.5
3200

—
—

1800
—

—

50.7

4312

0
—

—

0

2.0

1.9
28
—
—

0



—

—

165.4
0

—
—

0
	

—

46.0

55

220
2202.7

Trace

0

11.3

1.9
110
Trace
Trace

450



Trace

Trace

525. B
370

593-3
9372.8

490
Trace

Trace

48.2

256
Reference

Bozzelli et al., 1980
Bozzelli et al., I960

Pellizzari and
Bunch, 1979
Pellizzari, )977a;
Pellizzari et al., 1979
Pellizzari and
Bunch, 1979
Pellizzari, 1978c
Bozzelli et al., 1979
Bozzelli et al., 1980
Pellizzari and
Bunch, 1979
Bozzelli and
Kebbekus, 1979;
Bozzelli et al., I960;
Pellizzari, 1977
Pellizzari and
Bunch, 1979
Pellizzari and
Bunch, 1979
Pellizzari, 197Bb,d
Bozzelli et al.,
1979, 1980
Bozzelli et al., 1980
Pellizzari and
Bunch, 1979
Bozzelli et al., 1979
Bozzelli et al., 1980

Pellizzari, 1978c,d

Pellizzari and
Bunch, 1979
Singh et al., 1982

-------
TABLE 7-1 (cont.)
Location
NORTH CAROLINA
Chapel Hill
OKLAHOMA
Liberty Mound
Tulsa
Vera
PENNSYLVANIA
Bristol

Marcus Hook

N. Philadelphia

Pittsburgh
TEXAS
Aldine
Beaumont
Deer Park

El Paso
Freeport

Houston

Houston
Houston

La Porte

Pasadena
UTAH
Magna
Type of Site



rur.il
urban




urban

urban

urban


urban
industrial

urban
industrial

urban

urban
streets,
parks, rural
highway

urban

rural
Detection
Date Method

June, I960

July-Sept, 1977
July-Sept, 1977
July, 1977

August, 1977

August, 1977

August, 1977

April, 1981

June-Oct, 1977
March, 1980
August, 1977

Apr-May, 1978
August, 1977

July 1976-May, 1980

NR
July, 1976; June-
July, 1978
August, 1976

July, 1976

Oct-Nov, 1977

NR

GC/MS
GC/MS
GC/MS

GC/KS

GC/MS

GC/MS

GC/ECD

GC/MS
NR
GC/MS

CC/MS
GC/MS

CC/ECD

GC/MS
GC/MS

GC/MS

GC/HS

GC/HS
Number of
Samples

6

2 (2 ND)
2 (2 ND)
1 (1 ND)

2

2

4

NR

3 (3 ND)
11
6 (3 trace)

22 (19 ND)
2

99 (5 NO)

30
10 (1 trace)

1

1

9 (9 ND)
Concentration (ppt. v/v)a
Max. Kin. Hvg.

110

0
0
0

—

—

238.6

237

0
670
16390.6

29
1112.5

3000

16390.6
109.8

—

• 39

0

110

0
0
0

—

—

11.3

66

0
670
1002.5

0
815.8

50

73.1
30.1

—

39

0

no

0
0
0

63.8

IB. 2

139.2

121

0
670
6353.5

1.3
961.2

1300

893-0
63.0

192.3

39

0
Reference

Wallace, 1981

Pellizzari, 197Se
Pellizzari, 1978e
Pellizzari, 1978e

Pellizzarl and
Bunch, 1979
Pellizzarl and
Bunch, 1979
Pellizzarl and
Bunch, 1979
Singh et al., 1982

Pellizzari et al., 1979
Hal lace, 1981
Pellizzari and
Bunch, 1979
Pellizzarl, 1979a
Pellizzari and
Bunch, 1979
PeUlzzarl et al., 1979
Singh et al., 1980
Pellizzari, 1978b
Pellizzari and
Bunch, 1979
Pellizzari and
Bunch, 1979
Pellizzari et al., 1979

Pellizzari, 1979a

-------
                                                                    TABLE 7-1 (cent.)
Location
VIRGINIA
Front Royal

WEST VIRGINIA
Charleston

Charleston
Institute
Institute

Nitro

Nitro
St. Albans

71 St. Albans
O* S. Charleston

5. Charleston

W. Belle
W. Belle

WASHINGTON
Pullman

Pullman
Type of Site

industrlrl


industrial

industrial
industrial
industrial

Industrial

industrial
industrial

industrial
industrial

industrial

industrial
industrial


rural

rural
Detection
Date Method

Oct-Nov, 1977


Sept, Nov, 1977

Sept-Nov, 1977
November, 1977
November, 1977

Oct. Nov, 1977

Sept-Oct, 1977
October, 1977

Sept-Nov, 1977
Har, Sept-Nov, 1977

Mar-Nov, 1977

Sept-Nov, 1977
Sept-Nov, 1977


Dec, 1974-Feb, 1975

November, 1975

GC/MS


GC/MS

GC/MS
GC/MS
GC/MS

GC/MS

GC/MS
GC/MS

GC/MS
GC/MS

GC/MS

GC/MS
GC/MS


GC/MS

GC/ECD
Number of
Samples

16


3

M (4 ND)
2 (2 ND)
3

M

6 (6 ND)
1

M (4 ND)
6

16 (15 ND)

6 (6 ND)
4


NR

1
Concentration (ppt. v/v)a
Max. Min. Avg.

86.0


	

0
0
—

63.8

0
—

0
63.8

37

0
61.8


...

——-

37.3


—

0
0
—

37.3

0
—

0
37.3

0

0
37.3


<5

10

48.2


57.1

0
0
37.3

46.7

0
48.2

0
52.4

2.3

0
47.0




™-
Reference

Pellizzari and
Bunch, 1979

Pellizzari and
Bunch, 1979
Pellizzari, 1978e
Pellizzari, 1978e
Pellizzari and
Bunch, 1979
Pellizzari and
Bunch, 1979
Pellizzari, 197Be
Pellizzari and
Bunch, 1979
Pellizzari, 1978e
Pellizzari and
Bunch, 1979
Pellizzari, 1979a;
Pellizzari, 1978e
Pellizzari, 1978e
Pellizzari and
Bunch, 1979

Grimsrud and
Rassmusaen, 1975
Harsch et al., 1979
 Assume ambient temperature of 25°C and atmospheric pressure of 760 mmHg.
ND = Not detected
NR = Not reported
ECD : Electron capture detection
FID - Flame lonization detection
GC = Gas chromatography
MS = Mass spectronetry

-------
17.8 ppb.  Levels recorded in the New  Orleans  study area ranged  from  <0.12 to



Ul.7 ppb.



     Analyses done by Elfers  (1979)  were performed by collecting ambient air on



charcoal tubes followed by desorption by carbon disulfide.  Detection and quanti-



tation was made by gas-chromatograph-mass spectroraetry.   Quality control check



sample analysis indicated that  recovery of  standard concentrations  was highly



variable.  The detection  limit of the gas chromatographic method was reported as



1 jig.  The relative standard deviation of replicate standard solutions was found



to be 3$.  The precision  of the  analytical and sampling methods together was 6%.



The accuracy of the analytical  method varied between 72 and 97$.



     The  highest  reported  reading  for  a  single sample listed in  Table 7-1 is



191.5 ppb, which was recorded in Lake Charles, LA,  the site for several EDC-vinyl



chloride  manufacturing facilities.   Concentrations  in  excess of  15 ppb for a



single sample were also reported near manufacturing facilities in Calvert City,



KY; New Orleans,  LA;  Deer Park  and  Houston,  TX.



     Recent studies have  measured general urban  ambient concentrations of EDC in




ten cities (Singh et al., 1980, 1981,  1982).   The results of these studies are



summarized in Table 7-2.  The averaging time  for  the mean concentrations  in these



studies  was  2 weeks.    Electron  capture detector-gas  chromatography  was  the



primary means of analysis.



     Low  levels of EDC were  recorded  in a year-long monitoring program of  five



cities in industrial northern New Jersey (Bozzelli et al., 1980).  Samples  were




collected  at  the  five sites regularly throughout 1979.   Only two  of  the  208



samples analyzed had EDC concentrations higher than trace (*I1 ng/nr or  10 ppt).



     In  a survey  of air  contaminants  in  the  rural  northwest,  Grimsrud  and



Rasmussen (1975) found EDC concentration of <5 ppt.
                                      7-7

-------
                                   TABLE  7-2






        Ambient Concentrations of Ethylene Dichloride  in Urban Areas
City
Los Angeles, CA
Phoenix, AZ .
Oakland , CA
Houston, TX
St. Louis, MO
Denver , CO
Riverside, CA
Staten Island, NY
Pittsburgh, PA
Chicago , IL
Ethylene
Mean
519
216
83
1512
124
241
357
256
121
195
Dichloride Concentrations (ppt)
Maximum
1353
1450
842
7300
607
2089
2505
4312
237
2820
Minimum
173
39
38
50
45
56
63
55
66
22
^Source:   Singh et al., 1980, 1981, 1982
                                        7-8

-------
7.1.2.   Ground  and Surface Water  Levels.   EDC  can be  released to  the  water



environment via wastewaters generated during  production  of ethylene dichloride



and its derivatives.  It is also possible that EDC may be  inadvertently produced



in the water environment due to chlorination processes of public water supplies



or chlorination of sewage or wastewaters (Versar,  1975;  Seufert et  al., 1980).




One chemical reaction that  could lead to EDC production during water treatment is



the  reaction  of  alkenes  with  hypochlorite.    However,   it is expected  that




industrial discharges  to  surface  water and  leaching from  solid waste are the



primary causes of EDC contamination in drinking water (Letkiewicz et al., 1982).




Most discharges of EDC  are judged  to  ultimately reach the atmosphere because of



its high volatility (Letkiewicz et  al.,  1982).  The identification and levels of



EDC that have been found in the ground and surface water of the United States are



discussed below.



     EDC was reported to be a principal  contaminant in  finished  water in 1975 by



Dowty et al.  (1975).  Because the gas chromatographic  peak could  not be resolved,




it was not possible to  quantitate EDC.  It was indicated that EDC levels could be



increased as a result of the water  treatment processes and could pass the treat-




ment plant without removal.  The water analyzed in this study was obtained from



the Mississippi River.



     Deinzer et al. (1978), citing  the results of a 1975 national drinking water



survey, reported EDC concentrations ranging from 0 to 6 ppb.



     While sampling for pollutants in 14 heavily-industrialized river basins in



the United States,  Ewing et al. (1977)  found  that EDC was present in 53 of the 204




samples purged for volatile organic analysis.   The majority of  the 81 purgeable



organic compounds detected  were C  to  C,  halogenated  hydrocarbons.  Only chloro-



form,  trichloroethylene  and  tetrachloroethylene  were  found  with  greater
                                      7-9

-------
frequencies.  Identification was made by a  GC-MS procedure.  The limit of sensi-




tivity was <1 ppb.



     In a compilation of pollutants found in water, Shackleford and Keith (1976)



identified EDC as having been found in industrial effluents, in finished drinking




water, and in river waters.



     Letkiewicz  et al.  (1982),  in a recent study for the U.S. EPA, discuss the



results of six federal  surveys  in which a  number  of public water supplies were



selected for analysis of chemical contaminants, including EDC.  The six Federal



drinking  water  surveys  providing data on  EDC  include the  National  Organics



Reconnaissance Survey  (NORS),   the  National Organics Monitoring Survey (NOMS),



the National Screening  Program for Organics  in Drinking  Water  (NSP),  the 1978



Community Water  Supply  Survey  (CWSS),  the  Groundwater Supply Survey (GWSS), and



the Rural Water Survey  (RWS).  All surveys sampled  both ground and surface waters



except for the GWSS.  The scope and methodology of each survey is outlined below,



along with the identification  of EDC  presence.



     The National Organics Reconnaissance Survey (NORS)  was conducted in 1975 to



determine  the  extent of  the  presence  of  EDC, carbon  tetrachloride,  and four



trihalomethanes  in drinking water  supplies from 80  cities  across the country



(Symons et al.,  1975).   Another stated objective  of the study was to determine




the effect of raw water source  and treatment practices on  the  formation of these



compounds.  The  water samples  were collected in  50 ml sealed vials, with vola-



tilization  prevented  by the absence  of head space.   Samples were subsequently



shipped on  ice to the  EPA  Water Supply Research  Laboratory  in Cincinnati for




analysis.  Analyses were  performed  by purge-and-trap gas  chromatography with an



electrolytic conductivity detector.   A population base of 36  million  from 80



cities across the  country was  covered during the  study.
                                      7-10

-------
     In the NORS study,  16 groundwater systems were analyzed for EDC contamina-



tion.  None of the 16 systems  contain detectable levels.  Of the 64 surface water



samples analyzed for CDC,  six were found to contain quantifiable  levels of EDC at



0.2-6.0 |ig/&.



     The National Organics Monitoring Survey  (MOMS) was  instituted to identify



contaminant  sources,  to  determine  the  frequency  of occurrence  of  specific



drinking water contaminants, and to provide data for the establishment of maximum



contaminant levels (MCL's) for various organic  compounds in drinking water (U.S.



EPA, 1977).  The NOMS was conducted  in three phases, March-April 1976, May-July



1976, and November 1976-January  1977.   Drinking water samples from 113 communi-



ties were analyzed  for  21 different  compounds  by  purge-and-trap  gas chromato-



graphy with electrolytic conductivity detectors.  During Phase I the samples were



collected in 25- or 40-mi open-top screw cap septum vials with no head space, a



feature  that  prevented  volatilization  of  the compounds of  interest.    These



samples were then stored at 2-8°C  for  1-2 weeks prior  to analysis.  Samples were



collected in a similar fashion for Phases II and III,  but were held at 20°C for



periods up to 6 weeks.  Sodium thiosulfate  was  added to a number of samples from



Phases II and III to reduce any  residual chlorine present.



     Of 18 groundwater systems analyzed  for EDC during Phase I of the NOMS study



(March to April 1976), none contained quantifiable  levels.   When these  systems



were sampled again during Phase II (May  to July 1976),  one  system was found to



contain EDC,  at 0.02 |jg/
-------
During  the  third phase of  the  NOMS (November  1976  to January 1977), analyses




revealed EDC contamination  in one  system,  at 1.25  ng/i.



     In  the National Screening Program for  Organics  in Drinking Water  (NSP),



conducted  from June  1977  to March  1981,  both raw and  finished drinking water



samples from 169 water systems in 33  states were analyzed for 51  organic chemical



contaminants (SRI,  1981).   Analyses were carried out by  gas chroraatography with



various  detection methods,  including  the  mass spectrometer,  halogen-specific




detector, and  flame  ionization  detector.



     Thirteen  groundwater  supplies were tested for EDC contamination  during  the



NSP  study.   Of  these systems,  one  was found  to be  contaminated  with  EDC at




0.2  ug/£.   Surface water samples for 107 drinking water systems were analyzed  for



EDC.   Of  these, only  one  system  was  found  to  be contaminated  with  EDC,  at



3.8  ug/fc.



     In  the Community  Water  Supply  Survey  (CWSS),  carried out  in  1978,  110



surface  water  and 330  groundwater supplies were examined  for  contamination by




volatile  organic  chemicals  (U.S.  EPA,  198la).    Fourteen purgeable organic



compounds were analyzed, and total organic carbon levels were determined.   At  the



time of analysis, the samples  were  1  to 2 years old.  Long storage periods  may



have resulted  in the loss of some aromatics  and' unsaturated halocarbons  due to




biological  action.   The analytical method of choice for  the CWSS  was  purge-and-



trap gas chromatography with an  electrolytic  conductivity detector  for  halo-



carbons  and a  flame ionization detector for aromatic analysis.



     The CWSS  survey provided information on EDC levels  in finished groundwater




supplies  from 312  systems.   Of  the  samples  taken from these systems,  three



contained  detectable quantities of EDC  with  levels  of 0.57,  1.06  and 1.1  jig/*.-




Of  the 110 surface  water  systems tested, only one  contained quantifiable EDC,



with a concentration of 0.62
                                       7-12

-------
     The Groundwater  Supply  Survey of  1980  (GWSS)  was initiated  to provide a



clearer  picture  of the  extent  of contamination  of groundwater  supplies with



volatile organic  compounds  (U.S.  EPA,  1982a).   A  total of  945  systems  was



sampled, of which 466 were chosen at random  and 479  were  picked from locations



near potential sources of contamination.   Samples were collected at points close



to the actual distribution source, with mercuric chloride added as a preservative



to prevent biodegradation of  aromatics.  Sodium thiosulfate was also added  to the



vials to reduce any residual  chlorine,  thus preventing its reaction with organic



matter.  Analyses were made for 37 volatile organic compounds by purge-and-trap



gas  chromatography with  electrolytic  conductivity  detectors.     Of  the  166



randomly chosen water systems, seven were contaminated with  EDO,  at concentra-



tions ranging from 0.29-0.57  ug/i.  Of the seven positive samples,  six were from



systems  serving  populations  in excess of 10,000  people.   The  average  for  all



randomly chosen systems was 0.5 ug/H.  Of the  479  nonrandom  locations sampled,



nine were contaminated with EDC, at concentrations between  0.33-9.8 |ag/i.   Of the



nine positive samples, four were from  systems  serving populations in excess of



10,000 people.  The average EDC level for  the positive nonrandom systems was 2.7



ug/l.



     The Rural Water Survey (RWS),  conducted in 1978 (Brass,  1981), was carried



out in response to Section 3 of the Safe Drinking Water Act, which mandated that



EPA "conduct a survey  of the quantity, quality and availability of rural drinking



water supplies."  A total of 800 of  the  2655 samples collected in  the  RWS  was



analyzed for  volatile organic chemicals.    Three  hundred of the  samples were



selected by a random generation procedure and 500  were picked randomly by state



(10 per state).  There were 633 samples from groundwater, 47 samples from surface



water, and 81 samples  from other water  source categories.




     Of 633 groundwater samples, five  were found to have  EDC  present (minimum



quantification limit generally 0.5-1.0 \ig/l).  The range  of  concentrations  was






                                     7-13

-------
0.5-18 jig/A; the mean and median  concentrations of  the positive values were 5.6



and 1.7 ug/2., respectively.  Both the mean and median values of all samples were



<0.5 ug/fc.  Of the 47 surface water samples,  only one (2.1?) was found to have EDC




present (minimum quantification limit generally 0.5-1.0 iig/J,).   The concentra-



tion of the one positive sample was  19  ug/fc.  The mean and  median values of all




samples were both <0.5 |ig/&.



     The  combined  EDC groundwater data and surface  water  data  from the above



Federal studies are summarized in Tables  7-3  and  7-1,  respectively.   The RWS




study was deleted  from these summaries because the  RWS data  were recorded by a



number of service connections  rather than population  served.   From  the data




presented in Tables 7-3 and 7-4, Letkiewicz et al. (1982) have projected that EDC



levels in all groundwater  and  surface water systems in  the United States fall




below 10 ug/&, and that most are  below  1.0  ug/Jl.



     In addition  to  the  six Federal  surveys discussed  by  Letkiewicz  et al.



(1982), six  states  (California,  Connecticut, Deleware, Indiana, Massachussetts



and New Jersey)  provided the U.S.  EPA with information concerning EDC contamina-




tion in groundwater supplies.  These data are listed in Table  7-5.   The only



state-supplied surface water information on EDC was from New York;  one sample




from Poughkeepsie assayed positive at 5.9 iig/&   (Letkiewicz et al., 1982).



7.1.3.  Soil and Sediment Levels.  No data were available  on monitoring of EDC




concentrations in soils, sediments or solid wastes  in the sources consulted for



this report.



7.2. ENVIRONMENTAL EXPOSURE




     The  general  population can be exposed  to EDC from  three  sources,  air



emissions, drinking water and consumed foods.  Some individuals may  be exposed to




EDC from  sources other than the  three considered here, such  as in occupational



settings  and  in the  use  of consumer products  containing  EDC.   However,  this

-------
                                                     TABLE 7-3

                       Reported Occurrence of Ethylene Dichloride in Groundwater Systems
                               Combined Federal Data  (NORS, NOMS, NSP, CWSS, GWSS)a

System size
(population
served )
<100
101-500
501-1,000
i
^ 1,001-2,500
2,501-3,300
3,301-5,000
5,001-10,000
10,001-50,000
50,001-75,000
75,001-100,000
>100,000

Number of
systems
in U.S.
19,632
15,634
4,909
1,331
881
1,065
1,159
1,101
68
16
58
Number of
positive
systems
Number of
systems
sampled
1/175
0/220
0/114
3/151
0/40
2/79
1/114
11/296
1/37
0/12
1/43

Positive
systems
(*)
0.6
0.0
0.0
2.0
0.0
2.5
0.9
3.7
2.7
0.0
2.3

Number
undetected
174
220
114
148
40
77
113
285
36
12
42




Number of systems with
measured concentration
(ug/Jl) of:
<1.0
1
0
e
1
0
1
0
6
1
0
1
1.0-5
0
0
0
2
0
1
1
4
0
0
0
>5-10
0
0
0
0
0
0
0
1
0
0
0
>,0
0
0
0
0
0
0
0
0
0
0
0
 Source:   Letkiewicz et al., 1982
^Positive systems are those with quantified levels of ethylene dichloride.

-------
                                                     TABLE 7-4

                      Reported Occurrence of Ethylene Dichloride in Surface Water Systems —
                                   Combined  Federal  Data  (NORS,  NOMS,  NSP,  CWSS)a
System size
(population
served)
<100
101-500
501 -1 , 000
1 ,001-2,500
2,501-3,300
3,301-5,000
5,001-13,000
10,001-50,000
50,001 -75,000
75,001-100,000
MOO.OOO
Number of
systems
in U.S.
1,412
2,383
1,341
1,911
51 4
720
912
1,306
156
85
21 8
Number of
positive
systems
Number of
systems
sampled
0/4
0/19
0/13
1/22
0/6
1/15
0/10
1/38
2/22
0/14
5/102
Positive
systems
(*)
0.0
0.0
0.0
4.5
0.0
6.7
0.0
2.6
9.0
0.0
4.9
Number
undetected
<1 . 0 jig/A
4
19
13
21
6
14
10
37
20
14
97
Number of systems with
measured concentration
( u«/£) of:
0.0
0
0
o
1
0
0
0
1
1
0
3
1.0-5
0
0
0
D
0
1
0
0
1
0
1
>S-10
0
0
0
0
0
0
0
0
0
0
1
>,0
0
0
0
0
0
0
0
0
0
0
0
 Four systems reported as undetected with the unusually high detection limit of 2.0 Mg/? were deleted.

 Positive systems are those with quantified levels of ethylene dichloride.
Source: Letkiewicz et al., 1932

-------
                                   TABLE 7-5

               State Data on Ethylene Dichloride in Groundwaterc
Location
CALIFORNIA
Baldwin Park
Morada
Unspecified
Unspecified
CONNECTICUT
Colchester
Danbury
DELAWARE
Collins Park
Midvale
Newark
(North and South)
INDIANA
Elkhart
Granger
MASSACHUSETTS
Acton
Belchertown
Dartmouth
Rowley
NEW JERSEY
Bergen County
Essex County
Fair Lawn
Morris County
Passaic County
9 counties
12 counties



Water
type

N/S
N/S
N/S
N/S

D
D

F
F
F


F, N/S
N/S

F
F
F
F

N/S
N/S
F, N/S
N/S
N/S
N/S
N/S
N/S
N/S
N/S
Mean
(ug/Jl)

21
12
1.2


ND
7.8

ND
ND
ND


111


ND
14.5

ND

ND
ND
1.3

ND
ND
ND



Number
Range of
(ug/&) samples

1
1
2 (1 ND)
1.2, 5.3 2

1
1

1
1
2


30-2,100 11 (4 ND)
19, 160 15 (13 ND)

4
10.1-19.1 10 (7 ND)
11.6, 18.2 2
1

4
7
1.1-1.9 15
2.0-2.1 13 (11 ND)
1
154
228
0.1-0.9 4
1-10 4
10-100 1
 Source:  Letkiewicz et al.,  1982
ND = Not detected;  N/S = Not  specified;  F
= Finished; D r Distribution
                                     7.17

-------
section is limited to air, drinking water and food  since these are considered to
be general sources common to most individuals.  It  must be noted that individual
exposure will vary  widely depending upon  factors  such as  where the individual
lives, works or  travels,  or what the individual may eat or drink.  Individuals
living  in  the   same neighborhood  can  experience  vastly  different  exposure
patterns.
     Unfortunately,  methods  for  estimating the  exposure of identifiable popula-
tion  subgroups   from all sources  simultaneously  have not  yet  been  developed
(Letkiewicz et al.,  1982).   Exposure from the  three sources considered here is
discussed below.
7.2.1.  Exposure from Air.   Atmospheric  exposure to EDC appears to vary greatly
from  one location  to another.   Mean  levels  as  high as  27.5 ppb  have  been
monitored near production and use facilities in Lake Charles, LA (Elfers, 1979).
However,  the  available  monitoring  data  (see  Section 7.1) indicate.that  most
locations  have  EDC  concentrations  of  0.5 ppb or less.   The  monitoring  data
presented are not sufficient to determine regional  variations in exposure levels
for EDC.   The  majority  of  high values  reported  were for samples  taken  near
production and use facilities.
     Letkiewicz  et   al.  (1982)  have  estimated respiratory intake  of ethylene
dichloride by adults and infants;  these estimates  are summarized in Table 7-6.
From available  monitoring data,  Letkiewicz et al.  (1982)  estimated  that  low,
intermediate and high exposure levels  for EDC in air were  0.25, 2.5 and 25 ug/m  ,
respectively.  Daily intake of EDC for adults in intermediate exposure  levels was
estimated at 0.82 [xg/kg.
     Dispersion  models  have also been used to  estimate ambient concentrations.
Based on modeled results  for individual  plants, maximum concentration levels in
the   vicinity   of   various   emission   sources   have   been   calculated   (SRI
                                      7-18

-------
                                   TABLE 7-6

              Estimated  Respiratory Intake of Ethylene Dichloride
                            by Adults and Infants3


Low:
Intermediate:
High:
Exposure
Hg/m3
0.25
2.5
25
Level
(ppb)
(0.062)
(0.62)
(6.20)
Intake
Adult
0.08
0.82
8.21
(ug/kg/day)
Infant
0.06
0.57
5.71
Assumptions:  70 kg=man,  3.5 kg-infant,  20 m  of air inhaled/day (man),
             0.8 nr of air inhaled/day  (infant).

aSource:  Letkiewicz et al.,  1982
                                   TABLE 7-7

                Maximum Concentration Level in  the Vicinity  of
                           Various Emission Sources3
        Source
Maximum Annual Average EDO
 Concentration Level (ppb)
EDC Production Facilities

End Use Production Facilities

Gasoline Service Stations

Automobile Emissions

Automobile Refueling
      0.60 - 0.99

      0.01 - 0.029

      0.01 - 0.029

          <0.01
 Source:  SRI International,  1979

 An accurate estimate cannot  be determined from this reference, but it is
 thought to be in the range of 10-15 ppb.
                                     7-19

-------
International,  1979).    Table  7-7  shows  a  summary  of  estimated  maximum



concentrations that people may be exposed to in the vicinity of specific emission




sources.



7.2.2.  Exposure  from Water.  Letkiewicz et al. (1982) have projected that EDC



levels in all  groundwater and surface water systems  in  the United States fall



below 10 ug/Jl, and that most are below  1.0 \i&/H (see  Section 7.1.2).  The total



estimated population exposed to EDC from  both ground and surface water sources is



shown in Table 7-8.   The  values in the table were obtained by use  of the Federal



Reporting Data System data on populations served by primary water  supply systems




and data  on the estimated number  of these water systems that contains a given




level of EDC (see Section 7.1.2).



     Letkiewicz  et  al.  (1982)  have  also estimated  daily  intakes  of  EDC from



drinking water.  These estimates are  given in Table 7-9.



7.2.3.  Exposure from Food.   EDC is used as a fumigant for  grain, so contamina-



tion  of  flour and bread  is  possible.   Several studies on this  question have




indicated that EDC dissipates when the  bagged flour is exposed to air, and that



no detectable EDC remains after baking (U.S. EPA, 198lb).   No further information




on the presence of EDC in food  was uncovered in the literature search.




7.3- CONCLUSIONS



     Most EDC  exposures result  from the  production  and use of the  chemical.  The



highest atmospheric  concentrations  monitored  for  EDC have  been  detected near




production  and use facilities.  Therefore, it appears that  the risk for popula-




tion exposure  to EDC  is greatest in the  vicinity  of these  sources.
                                      7-20

-------
                                   TABLE  7-8

         Total  Estimated  Population (in Thousands)  Exposed to Ethylene
      Dichloride  in  Drinking  Water  at  the Indicated Concentration Ranges'
System type
Groundwater
Surface water
TOTAL
(% of total)
Total
served
in U.S.
(thousands)
69,239
126,356
195,595
(100*)
Population (thousands) exposed
to concentrations (ug/S,) of:
<1.0 to 10
69,239
126,356
195,595
(100*)
>10
0.0
0.0
0.0
(0.0*)
 Source:   Letkiewicz et al.f  1982
                                   TABLE 7-9
                 Estimated Drinking Water Intake of Ethvlene
                       Dichloride by Adults  and Infants
Exposure Level
1.0
5
10
100
Intake ( ug/kg/day )
Adult Infant
0.029
o.m
0.29
2.9
0.24
1.2
2.4
24.0
Assumptions: 70 kg-man,  3-5 kg-infant, 2 8, of water/day (man),
             0.85 I of water/day (infant).

aSource:  Letkiewicz et al., 1982
                                        7-21

-------
                            8.   ECOLOGICAL EFFECTS



     A variety of studies have  demonstrated that EDC,  in the role of a fumigant,



is toxic to insects infesting stored grains  (Wadhi  and  Soares,  1964;  Ellis and



Morrison,  1967;  Vincent  and Lindgren,  1965;  Snapp,  1958; Krohne  and Lingren,



1958; Finnegan and Stewart, 1962; Lindgren et  al.,  1954;  and  Bang and Telford,




1966).  Little is known,  however, of the effects of EDC to soil micoflora.



     EDC has been reported to be non-toxic to many economically important plant



species when directly applied to growing plants (Cast  and Early, 1956).  LeBlanc



(1980) reported  the  24-hour LC5Q for  Daphnia magna  to  be 250 mg/fc   (nominal



concentration) and the 48-hour  LC Q  to  be 220 rag/5, (nominal concentration).  A no



discernible effect concentration was estimated to be  <68 mg/S,.



     Toxicity of EDC  to  barnacles (Barnacle  nauplii)  and  unicellular algae has




been reported (Pearson and McConnell, 1975).  The LC_0 for Barnacle nauplii was



reported  to be  186  mg/£.   The effective concentration  needed to  reduce the



photosynthetic ability of unicellular algae by 50$ was 340 ppm (340 mg/S,).



     A 24-hour  median tolerance limit  of  320 ppm  (320 rag/2,)  was reported for



brine shrimp under static test  conditions (Price and  Conway,  1974).



     Static  24-hour   and  96-hour LC5_  concentrations  of >600  and  430  mg/S,,



respectively,  have   been  determined  for  the  freshwater  bluegill  (Lepomis



macrochirus)  (Buccafusco  et al., 1981).   Garrett  (1957a,b) reported that the



concentration of  EDC  needed to  produce  >50$ mortality in the  marine pinperch



(Lagodon rhomeboides) under static conditions was 175 mg/i.



     An  acute LC,-0 of 115 mg/S,  was  reported for the marine  flatfish (Limanda



limanda)  (Pearson and McConnell,  1975).   In  acute  tests with  another  marine



species, static  24-,  48-, 72-  and  96-hour LC5_s in the range of  130-230 mg/S,
                                      8-1

-------
were determined  for sheepshead minnows  (Cyprinodon variegatus)  (Heitmuller et



al., 1981);  the  observed no-effect  concentration  was reported to be 130 mg/fc.



The  estimated acceptable  concentration of  EDC  in  a 32-day  early  life stage



toxicity  test with  freshwater  fathead  minnows  (Pimephales promelas) lies in the




range of  29-59 mg/Jl (Benoit  et al.,  1982).
                                       8-2

-------
             9.   BIOLOGICAL EFFECTS IN MAN AND EXPERIMENTAL ANIMALS



9.1. PHARMACOKINETICS



9.1.1.    Absorption and Distribution.  Ethylene dichloride  (EDC;  1,2-dichloro-



ethane) is a colorless, oily liquid with a sweet taste and with a chloroform-like




odor detectable  over  a range of 6-40  ppra.   It is appreciably soluble in water



(0.869 g/100 mi) (Von Oettingen,  1964), with a vapor  pressure of 64  torr at room




temperature  (20°C).    Consequently,  EDC  is  rapidly and  extensively absorbed



through  the lungs in  its  vapor form and  from  the gastrointestinal  tract  in



solution.  Inhalation is considered the primary route of entrance into man from




occupational exposure and air pollution.  Absorption after oral ingestion is of



particular interest for EDC as a contaminating component  of drinking water and



foodstuffs.   Skin absorption is negligible in most  industrial  vapor exposure



situations,  although  absorption  may be  significant  by this route  with direct



liquid contact, as evidenced by toxic  symptomatology in man (Section 9.2),  and



also quantitative animal measurements.




     9.1.1.1.  DERMAL ABSORPTION — Tsuruta  (1975,  1977)  studied  the percuta-



neous absorption  of  a series of chlorinated organic solvents  (including EDC)



applied to a standard area  of shaved abdominal mouse  skin for 15 minute periods.



Absorption was quantified by the presence of the compound  in  total mouse body




plus expired air, as determined by  gas chromatography (GC).  For all solvents,



percutaneous absorption linearly increased  with time and  the rate  was directly




related to water solubility.  For EDC,  the absorption  rate was 479 nmoles/min/cm2



skin,  second highest of 8 solvents measured.   Tsuruta  concluded that skin absorp-



tion from  liquid  contact  could be  a  significant  route for EDC entry into  the



body.
                                      9-1

-------
     Jakobson  et al.  (1983)  also carried  out dermal  absorption  studies with


guinea pigs  for  10  chlorinated organic solvents including EDC.  Liquid contact

                  o
(skin area,  3.1  cm  ) was maintained for up to 12 hours and solvent concentration


monitored  in blood during  and  following dermal  application.  Dermal absorption,


as reflected by  blood concentration profile, was again observed to be related to


the water solubility of the solvent.  For solvents like EDC, which are relatively


hydrophilic  [300-900 mg/dl],  the blood concentration increased steadily during


the entire exposure.  EDC  reached 20  ug/m& blood in  12 hours.   With extended


dermal exposure,  the blood  concentration  eventually  became fatal.  This pattern


of steady blood accumulation indicates that dermal absorption occurs faster than


body elimination by  metabolism or pulmonary excretion.  Following dermal expo-


sure, the EDC concentration in blood  declined in a manner consistent with a two-


compartment  kinetic model,  i.e.,  the sum  of two exponential terms.


     9.1*1.2.  ORAL ABSORPTION — In man,  gastrointestinal absorption of EDC has


not been specifically studied, although the absorption rate after oral ingestion


has been determined  in animal studies.  As expected  from the neutral and lipo-


philic properties of EDC  (Tables 9-1  and 9-2), transmucosal  diffusive passage


occurs readily.  There are numerous reports of poisoning in humans as a result of


accidental or suicidal ingestion  of EDC and the peroral LD5Q approximates 0.2-1


g/kg (NIOSH, 1976).


     In  animals, extensive  gastrointestinal absorption  of  EDC has  also been


clearly demonstrated by  the biological effects produced by peroral administra-


tion of a wide range of dosages and dosing schedules  in toxicity studies in rats,


nice, guinea pigs and dogs  (Section 9.2) and in metabolism  studies in rats (Reitz


et al.,  1980,  1982;  Spreafico et al.,  1978,  1979, 1980).  Reitz et al.  (I960,


1982) found  that   C-EDC in corn oil  given perorally  to rats  (150  rag/kg)  was


completely absorbed by virtue of a complete recovery  of radioactivity in exhaled
                                       9-2

-------
                               TABLE  9-1

               Physical Properties of Ethylene Bichloride
                        and Other Chloroethanes3
                       Vapor  Pressure
                       at  25°C,  torr.
                                           Ostwald  Solubility  Coeff..  37°C
Water/
 Air
Blood/
 Air
Olive Oil/
   Air
1,2-Dichloroethane           80

1,1-Dichloroethane          250

1,1,1-Trichloroethane       125
11.3

 2.7

 0.93
19.5
 3.3
   187

   356
 Source:   Sato and Nakajima,  1979
                                         9-3

-------
                                   TABLE  9-2

                 Partition Coefficients for Ethylene Dichloride
MAN

  Blood/air, 37°C

  Adipose tissue/blood, 25°C
                19.5

                56.7
Sato and Nakajima,  1979

Bonitenko et al.,  1977
RAT. 250 ppra inhalation exposure

  Blood/air, 37°C, 250 ppm
             37°C, 150 ppm

  Adipose tissue/blood

  Lung tissue/blood

  Liver tissue/blood
                                a
                30.3
                11.9

                 8.7b

                 O.U51

                 0.72*
Spreafico et al.,  1980
Reitz et al.,  1980

Reitz et al.,  1980

Reitz et al.,  1980

Reitz et al.,  1980
 Conversion factors:
At 25°C/760 torr, 1 ppm in air = 1.05 mg/m3
 1 mg/liter air = 247 ppm
 Dose-dependent

-------
air, urine and carcass (Table 9-3).   Spreafico et al.  (1978, 1979,  1980)  found



that 25,  50  and  1 50  rag/kg administered orally to rats in corn oil  was  rapidly



absorbed with peak blood levels occurring within  20  minutes.  Their  data,  given



in  Figure  9-1,  show  the time-course of  blood,  liver,  lung and adipose  tissue



following oral administration of the three dose  levels.  The  peak  blood  levels,



which, according to linear kinetics,  should be proportional to the amount of EDC



absorbed into the rat body, are summarized in Table 9-4.  Although the peak blood



levels in this study do not deviate  drastically  from linearity,  the  peak tissue



levels are not  linearly  proportional to  the three  oral  doses.   These  results



strongly suggest a passive transport across the gastrointestinal tract of EDC and



the occurrence of nonlinear elimination kinetics for  EDC, i.e., a nonlinear dose-



dependency related to saturation of liver metabolism of EDC.  An influence of the



dose on kinetic parameters has also been found for halogenated  ethylene compounds



(e.g., vinyl chloride, vinylidene chloride) by other investigators  (Gehring et



al., 1976, 1977,  1978;  Bolt, 1978; McKenna et al., 1978; Reichert and Henschler,



1978; Filser and Bolt, 1979).



     Table 9-5 gives  the  absorption rate constants (k )  and areas under the blood
                                                    Si


concentration curves  (AUC)  found by Spreafico et al.  (1973,  1979, 1980)  for rats



given a  single  oral  dose of EDC.   The rate  constant  for absorption was  dose-



dependent and was  probably  influenced  by liver  metabolism.   A markedly  lower



value was observed for the highest dose (150 mg/kg) at which saturation of liver



metabolism probably occurs.  Nonetheless, these data show that EDC  is  absorbed



from the gastrointestinal tract rapidly,  with  one-half  the  dose  absorbed within



3.3 minutes for  the lowest dose given in corn oil  (25 mg/kg) and  6.4  minutes for



the  highest  dose  in  corn oil  (150 rag/kg).   As expected,  absorption of  EDC



occurred significantly faster with an oral dose  in water  than in oil [k  ,  0.299
                                      9-5

-------
                                      TABLE 9-3
                      11
              Fate of 1HC-EDC in Rats 48 Hours After Oral (150 mg/kg)  or
                         Inhalation (150 ppm, 6-hr) Exposure3
                                 Oral
                    umole/kg
metabolites
                          Inhalation
                                                    iimole/kg
metabolites
Body burden
 (total radio-
  activity)

Charcoal trap
 (B)
                     1539 ± 391
                     447  +   60
                 512 + 135
                 9.4 ± 0.4
                 9.4 + 0.4
Total metabolites
(A-B)
Urine
CQy trap
Total carcass
(48 hr after
exposure)
Feces
Cage wash
(1092)

926 + 348
83.1 + 11.9
46.9 ± 14.7


26.3 + 14.7
12.5 ± 6.37
(100)

85.7
7.7
4.3


2.1
1.1
(503)

432 + 121
36.1 + 6.89
22.7 ± 3.38


8.90 + 2.84
3.34 ± 1.34
(100)

84.4
7.0
4.4


1.7
0.7
Values are mean ± S.D., with  n  =  4  for each route,.of exposure and are jimole equiva-
lents of EDC, based on  the  specific activity of  C-EDC (3.2 mCi/mM).

aSource:  Reitz et al., 1980
                                          9-6

-------
             Ill
I
 5
o
o
   0.1
                    2h
6h
       Figure 9-1. EDC levels after single oral administration in rats:
       Top panel, 25 mg/ kg; middle, 50 mg/kg; bottom, 150 mg/kg
       dose. Adipose tissue ( • !, blood ( o ), liver ( • ), lung ( A }.

       Source: Spreafico et al. (1978, 1979, 1980).
                                   9-7

-------
                                  TABLE 9-4
             Peak Blood and Tissue Levels After Single Oral Dosage
                              of EDC  in Male Rats3
Dose
ig/kg
25
50
150
Blood
13.29
31. 94
66.78
Adipose tissue
ug/ml or |ig/g
110.67
148.92
259.88
Lung
2.92
7.20
8.31
Liver
30.02
55.00
92.10
aSource:  Spreafico et al., 1980
                                           9-8

-------
                                  TABLE 9-5
            The Absorption Rate Constants (k )  and Area Under Curve
             (AUC) for Rats After Single OraT. Doses of EDC in Oil
         and in Water as  Vehicle and After Intravenous Administration'
V
Dose a
(mg/kg) min~
Oral 25 0.299 (water)
0.209 (oil)
50 0.185 (oil, male)
0.181 (oil, female)
AUC (blood)
jig x rain x ml"
466
466
1700
1685
                       150
              0.109 (oil)
7297
Intravenous (water)
 1

 5

25
   9

  5U

 595
*Source:   Spreafico,  1978,  1979,  1980

-------
     Comparison  of the  area under  the  blood concentration curve  (AUC)  after



intravenous administration with AUC  after  the same oral dose provides a measure



of the extent or completeness of oral  absorption.  Table 9-5 shows that the AUC



was 595 >ig x min x mA~1 after a  25 mg/kg  intravenous dose and was 166 jig x min x




mi~  after  25 mg/kg oral dose.  These results indicate that absorption of the



oral dose was very  extensive,  averaging  78$ of complete  absorption,  or 100?



absorption if it is assumed that the  first-pass effects in liver  (metabolism) and



lung (elimination) after oral  absorption  decreased EDC  appearance  in systemic



blood (Reitz, 1980, 1982).  It is notable that the AUCs for differing intravenous



doses, where intestinal absorption is not a factor (Table 9-5), are not linearly



proportional,  providing further  evidence  of the occurence of Michaelis-Menten



elimination kinetics for EDC.



     Withey  et al.  (1982)   investigated the  effect  of the dosing  vehicle  on



intestinal  absorption  of EDC  in  fasting  rats  (100  g)  following intragastric



intubation of equivalent doses  (100 mg/kg)  in =1  mi of water or of corn oil.  The



post-absorptive  peak  blood  concentration  averaged  5  times  higher  for  water




vehicle   than  corn  oil  (81.6  vs   15.9   ug/mH);  moreover,  the  peak  blood



concentration was reached 3  times faster  for water solution than  for oil solution




(3*2 vs.  10.6 minutes).  The ratio  of the areas under the blood concentration



curves for 5 hours after dosing  (AUC, 5 hr) was 3; water:corn oil.  These results



are  in  general  agreement with those of  Spreafico  et al.  (1978,  1979,  1980)



(Tables 9-1 and  9-5),  who found  that  intestinal absorption was faster with an



oral dose in water than  in oil.  The observations of  these  investigators stress



again the dependence of  oral absorptive  rate in  rats  not only on the dose, but



also  on  the  vehicle.    While  these  factors  are   unlikely   to  affect  the



pharmacokinetics of EDC  in man  in any practical  way,  they are of importance to




interpretation of  data from long-term carcinogenicity  tests  of EDC in rodents
                                      9-10

-------
 where  the modes of  intragastric  dosing employed may  differ by  both dose  and



 vehicle.




     9.1.1.3.  PULMONARY ABSORPTION — EDC has a moderately high vapor pressure



 (80  torr at 25°C; Table 9-1)  and,  in man, a high blood/air partition coefficient




 compared  to chloroethanes (19.5 at 27°C;  Table 9-2).  Hence, its vapor  in ambient



 air  is a  primary mode of exposure, and the lungs are a principal route of entry




 into the body.  The total amount absorbed via the lungs (as for all vapors) can be



 expected  theoretically to be directly  proportional to:  (1) the concentration  of



 the  inspired air;  (2) the duration of exposure; (3)  the blood/air Ostwald solu-



 bility coefficient;  (4)  the solubility  in  the various  body  tissues;  and  (5)




 physical activity which increases  pulmonary  ventilation rate and cardiac output.



 Hence, the basic  kinetic  parameters  of pulmonary  absorption  of EDC  and   its




 equilibrium in the body are as  valid for  the very low concentrations expected  in



 urban ambient air as for  the  higher vapor concentrations found in the industrial



 environment and the  workplace.  However, these  parameters  of pulmonary absorp-




 tion have  not been studied  in  any detail in man,  although some information  is



 available  from pharmacokinetic  studies in rats.




     Urosova (1953) reported  that  women exposed during a normal day in the work-




 place to =15.5 ppm EDC in air  accumulated  the chemical in breast milk and that



 initial concentrations in exhaled air following daily exposure  were  14.5  ppm.




 These observations indicate that the women  absorbed  EDC through their lungs and



 reached blood and total body equilibrium with inspired air concentration within




 the daily work period.  The deficiencies  in the reporting of this work makes the



 results only suggestive.




     Spreafico et  al.  (1978,  1979, 1980)  and Reitz  et  al.  (1980,  1982)  have



studied the kinetics of pulmonary  absorption of EDC in rats.   Figure  9-2  shows




 the time-course of blood concentration of EDC, observed by Reitz et al.  (1980,
                                     9-11

-------
                                  i	1	1	1—i	r
                   i     i     t     i    i    i
                 100
200       900        400      600

     TIME (mlnutM)
    10.0
O
O
O
(A

K
O
2
O
i
                           40                80

                               TIME (minutes)
      Figure 9-2. Top: Blood levels of EDC observed during and following
      a 6-hour inhalation exposure to 150 ppm EDC.  Data from four male
      Osborne-Mendel rats were fitted to a two-compartment open model
      as described by Gehring et at. The computer plot is shown.  The
      arrow indicates exposure termination. Bottom: Semi-logarithmic
      plot of EDC blood levels vs. time after exposure termination.
      Source: Reitz et al. (1980. 1982).

-------
 19&2)  during a 6-hour  inhalation exposure  to  150  ppm EDC.   Blood  and  body



 equilibrium  with the  blood  concentration  maintained a plateau level of  9  ug/mil



 (blood/air partition coefficient, 14.9).  Spreafico et al. (1980) exposed  their



 rats to 50 and 250 ppm EDC for 6 hours.  Their data are given in Tables  9-6 and



 9-7.  Blood and body equilibrium (liver, lung and  adipose tissue) was established




 at  =2  hours  for  50 ppm exposure, and  at  3  hours for 250  ppm exposure.  During



 inhalation of EDC, when  equilibrium  is achieved, the arterial blood concentra-




 tion of EDC should theoretically always be directly proportional to inspired air



 concentration.   This fixed  relationship  is  defined by the  blood/air  Ostwald




 solubility coefficient for EDC.  As calculated from the data for 50 and  250 ppm




 exposures of Spreafico and co-workers, the blood/air partition coefficients for




 EDC are 6.3  and 30.3, respectively.   Including the data of Reitz et al. (1980)



 for 150 ppm  exposure  in  rats (Figure 9-2),  these  results  do  not  demonstrate a



 direct proportional  relationship between  inspired  air  concentration  and  blood



 concentration of EDC, i.e.,  a constant blood/air  partition  coefficient value.




 Furthermore,   linear   kinetics for  EDC  after  inhalation  exposure  cannot be



 presumed,  and indeed  these data  suggest otherwise, and are in  accord  with  the




 evidence of nonlinear kinetics after oral  and intravenous dosing as noted  above



 (Table 9-5).




     9-1.1.4.  TISSUE DISTRIBUTION — After  pulmonary or peroral absorption,  EDC



 is distributed into all  body  tissues.  As expected from its  general anesthetic




 properties in man  and in animals, EDC readily passes the  blood-brain barrier.



The compound  crosses the placental  barrier  and  has been found  in the  fetus




 (Vozovaya, 1975,  1976,  1977).   EDC  also  distributes into human  colostrum  ar.d



mature breast  milk.   Urusova (1953)  found  EDC concentrated  in breast   milk



 (5.4-6.4  mg/2,) in  the workplace.   The compound  remained in breast  milk for 18




hours  following workday  exposure, even though its  concentration  in  exhaled  air
                                     9-13

-------
                                     TABLE  9-6
                 EDC  Tissue  Levels After  50  ppm  Inhalation Exposure
Time of
Exposure
                                 EDC  ug/g  or  ng/ml + S.E.
   Blood
Liver
Lung
Adipose Tissue
30 minutes


1 hour


2 hours


4 hours


6 hours
0.48 + 0.05     0.32+0.02     0.14+0.02
0.92 + 0.09     0.67 + 0.04     0.27 ± 0.03
1.34 ± 0.09     0.84 + 0.09     0.34 + 0.03
1.31+0.11     1.14+0.17     0.42+0.05
                               2.91  + 0.22


                               7.19  + 0.60


                              10.31  + 0.94


                              11.08  + 0.77
1.37+0.11     1.02+0.10     0.38+0.02      10.19+1.00
 Source:  Spreafico et al.,  1980

-------
                                    TABLE 9-7
                EDC Tissue  Levels  After  250  ppra Inhalation Exposure6
Time of
Exposure
    Blood
                                EDC ug/g or (ig/ml + S.E.
Liver
Lung
                                                 Adipose Tissue
30 minutes
1 hour
3 hours


6 hours
 6.33 ±1.04     3.82 + 0.78     2.19 + 0.21      26.75 + 3.12


11.65+1.12     7.34+0.7U     6.40+0.20      82.64+2.19
2 hours         23.64+0.91    16.39+1.18    14.07+0.47     151.53+12.17
29.36+1.01    20.83+2.21    14.47+1.12     252.18+14.62


31.29+1.19    22.49+1.12    14.14+0.90     273-32+12.46
 Source:   Spreafico et al.,  1980
                                        9-15

-------
fell to  4  ppm.  The  lack  of detailed  reporting  of  this study precludes adequate



evaluation.   Sykes  and  Klein (1957) have  demonstrated  the  presence  of EDC in



cows' milk after  oral  administration.



     There is little information on  EDC  distribution and concentration in the




various  body  tissues of man after exposure,  and few  controlled exposure studies



in  animals investigating the distribution  of  EDC  in body tissues and defining



dose-dependent  tissue concentrations.    Spreafico et al.  (1978,  1979,  1980)



determined EDC  levels in rat blood,  liver, lung and epididymal adipose tissue



after single oral administration of  25, 50 and 150 mg/kg.   The time-course of EDC



concentrations are shown  in Figure  9-1.  This figure shows that tissue accumula-



tion occurs  most rapidly in the liver where  peak concentrations were reached



within  10  minutes of  administration.  Following  complete absorption and body



equilibration after  2  hours, the decay in tissue concentrations of liver, lung



and  adipose  tissue  parallels the  first-order  decline in blood concentration.



Table  9-8 summarizes  blood  and tissue  concentrations  determined at  2  hours



following  administration.   Blood levels and tissue levels in liver and adipose



tissues  were not  found  to be  linearly  proportional to dose,  but  increased



exponentially, providing  evidence of liver metabolism saturation and nonlinear



kinetics after oral administration.  At oral dose levels of 25, 50  and  150 mg/kg,



adipose  tissue  displayed   the  greatest  concentration   of  EDC  with  adipose



tissue/blood  partition coefficients  of  17, 9  and 7, respectively.   No  other



tissue, including liver, demonstrated a partition coefficient >1.0.  The  very low



concentration levels of EDC  in lung tissue may  occur  as a  result of EDC elimina-



tion by  this  route.




     Table 9-8 also gives blood concentrations after chronic oral administration



to rats (11 daily doses) of  50 mg EDC/kg.  Comparison of these tissue levels with



those following a single oral administration of 50 mg/kg EDC provides no evidence



of blood or tissue accumulation  with  chronic administration.






                                      9-16

-------
                                  TABLE 9-8

              Blood and Tissue Levels of EDO in Male Rats 2 Hours
                     After Single Oral  Doses in  Corn Oila
Tissue
( |ig/ml , or
Hg/g ± SE)
Blood
Liver
Kidney
Brain
Spleen
Lung
Adipose
Dose,


0.
0.



<0.
7.

25
46 + 0.04
32 + 0.01



05
80 + 0.72


5
3
3
2
1
1
55


.89
.81
.43
.96
.79
.44
.78

50
+
+
-»•
+
^
+
+
mg/kg


0
0
0
0
0
0
6


.41
.45
.26
.36
.35
.10
.88


26.
21.



1.
178.


60
62



62
50

150
•t- 1.
± 3.



-i- 0.
± 24


23
99



25
.66
         Blood and Tissue Levels of EDC in Male Rats 2 Hours After the
         Last of 11 Daily Oral Doses in Oil (50 mg/kg)

Blood                                  8.11 ± 0.23
Liver                                  3.95 + 0.48
Lung                                   2.24 ± 0.47
Adipose                               53-71 + 10.07
a
 Source:  Spreafico et al.,  1978,  1979,  1980
                                      9-1?

-------
      Sppeafico and his colleagues (1978,  1979,  1980) also investigated blood and



 tissue concentrations in rats after  inhalation  exposure to  50  and  250  ppm EDC.



 The results  of these experiments are given  in Tables 9-6 and 9-7.  Blood  and



 tissue equilibrium (liver, lung  and  adipose  tissue)  occurred at 2 hours  and  3




 hours, respectively,   for the  two inspired  air  concentrations.   In  general,  the



 blood and  tissue distribution of EDC at  body equilibrium  is  similar to  the



 results obtained with oral administration given in Table 9-8.  A very clear dose



 dependence in  tissue levels  of  EDC was  again found with  the two  inhalation



 concentrations, with  differences in EDC concentrations on  the order  of  20-30



 times when blood, liver, lung and adipose  tissue are considered.  The highest




 absolute levels of EDC were measured  again  in adipose tissue with concentrations



 that  were between 8 and 9 times  greater  than those measured in the blood.  As



 previously noted for  oral  administration  (Table 9-8),  liver  and  lung  concentra-



 tions  were less than  blood concentrations.




      A comparison of the absolute blood and  tissue  concentrations at body equili-



 brium  for oral  and inhalation  modes of administration  (Tables 9-6,  9-7  and 9-8)




 indicates that  50 ppm inhalation  roughly equates to 25 mg/kg oral, while 250  ppm



 inhalation provides blood, liver  and  adipose tissue levels  slightly  greater than



a 150  mg/kg oral dose.




9.1.2.     Excretion.   Elimination of EDC  from  the body is perforce the sum of



metabolism and  excretion  of  unchanged  EDC  via  pulmonary  and  other   routes.



Unmetabolized  EDC is  excreted almost exclusively through  the  lungs;  however,



metabolism of EDC  is extensive, with the proportion excreted  unchanged dependent



on body  doses.   While no controlled  experimental  studies have been made on  the



kinetics  of excretion  of  EDC in man,  recent  studies have  been  performed on



experimental animals.
                                      9-18

-------
     9.1.2.1.  PULMONARY EXCRETION — Urusova  (1953) reported that women exposed



to =15.5 ppm EDC in ambient air of industrial environs demonstrated initial EDO



concentrations in exhaled air of 11.5 ppm.  The breath concentration declined to



-3 ppm after 18 hours.  These values  lead to an approximation of 9 hours for the



half-time of pulmonary elimination of EDC in man.  Although this human data was



difficult to evaluate, similar observations have been made for animals (monkey,



dog, cat, rabbit, rat and guinea pig) in early investigations of the anesthetic



properties and toxicities of  EDC  (Heppel et al.,  1945; Kistler and Luckhardt,



1929; Lehman  and Schmidt-Kehl,  1936).   In controlled studies  in  mice,  Yllner



(1971a)  found  that  up to H5% of an  intraperitoneal  injected  dose of  EDC (170



mg/kg) was recoverable unchanged in exhaled air (Table 9-9).  The percentage of



EDC recovered  unchanged  in  exhaled air increased  exponentially  with  the dose,



indicating a  limited  capacity of  biotransformation,  i.e.,  non-linear kinetics.



Thus, in mice, pulmonary excretion  of EDC  is a  major  route  of  elimination,



increasing in importance with higher body doses.



     Similar observations have  been made  for  the rat by  Reitz et al.  (1980,


                                       14
1982).   In  a balance study  utilizing   C-EDC given orally (150 mg/kg)  and by



inhalation (150 ppm, 6 hours), these  investigators  found that for the oral dose,



29% was  recovered  unchanged in exhaled  air,  and  that only 1.8? of  the lower



inhalation dose was  excreted by the pulmonary route (see Table 9-3).  Sopikov and



Gorshunova (1979) observed that  after an  intraperitoneal 250 mg/kg dose to rats,



30$ of the EDC dose was eliminated in the exhaled air within 5 hours.



     9.1.2.2.  OTHER ROUTES OF EXCRETION — EDC is not ordinarily eliminated in



significant amounts from the body by  any route other  than  pulmonary.   However,



Shchepotin  and Bondarenko  (1978)  identified  EDC by  gas  chromatography  (GC)



methods in the urine of persons with severe symptoms of EDC poisoning.  Studies



of chlorinated compounds in the urine  after EDC  inhalation exposure or peroral
                                     9-19

-------
                                    TABLE 9-9

           Percent Distribution of Radioactivity Excreted fl(48-hr) by Mice
                         Receiving  1,2-Dichloroethane-  C
                                                Dose g/kg
                                 0.05      0.10      0.11      0.17
14C02 (exhaled air)                13         8         4         5

Dichloroethane (exhaled air)       11        21        U6        15

Urinary metabolites                73        70        18        50
aSource:  Yllner, 1971 a
                                        9-20

-------
dosage to experimental animals have failed to detect unchanged EDC (Spreafico et



al., 1980; Yllner, 1971 a).  Mutagenic metabolites  of  EDC  have been reported in



rat and mouse bile (Rannug and Beije, 1979; Rannug, 1980a),  but  EDC itself has



not been found.  Diffusion of volatile  haloalkane  anesthetic agents into bowel



space and through skin  is known to occur (Stoelting and Eger,  1969),  and these



routes of  excretion  may also be  significant  for EDC because  of similarity of



structure and comparable high lipid/blood partition coefficients.



     9.1.2.3.  KINETICS  OF EXCRETION  —  While the kinetic parameters of elimina-



tion of EDC for each of the various routes of excretion, i.e., pulmonary, metabo-



lism, etc., are not well defined for  either man or experimental animals, several



investigative groups have determined the kinetic parameters  for  whole-body and



tissue compartment excretion of EDC in the  rat  (Retiz  et al., 1980; Spreafico et



al., 1978, 1979, 1980;  Withey and  Collins,  1980).



     Withey  and Collins  (1980)  determined  the kinetics  of  distribution and



elimination of EDC from  blood of Wistar  rats after intravenous administration of



3, 6, 9, 12 or 15 mg/kg of EDC given in 1 mil water intrajugularly.  For the two



lower doses (3 and 6 mg/kg), the blood decay curves exhibited two components of



exponential disappearance  and  best fitted a first-order  two compartment model



with kinetic parameters  of:  ke>  0.24  min~ ; Vd, 43 ml;  k12  and k21, 0.02 and 0.04



min~ , respectively.  For higher doses  (9,  10 and 15 mg/kg),  these investigators



found  their  data  best   fitted  a  first-order,  three-compartment  model;  they



suggest  that the shift from two to three-compartment kinetics may have occurred



either because of a dose-related alteration in  the kinetic mechanisms of uptake,



distribution,  metabolism and elimination,  or that  three-compartment kinetics



were followed  at all dose levels but with lower doses;  the third exponential



component, representing the third compartment,  was  obscured by the  limit of



analytical sensitivity.   Average kinetic values for the three-compartment  model
                                      9-21

-------
were: ke, 0.094 min"1;  Vd, 67.9 ml; k12, k21, k13, k31; 0.06,  0.08, 0.01 and 0.01



min'1, respectively.   Withey and  Collins  state  that they found no evidence in



their kinetic analysis of the disposition of intravenous bolus injections of EDC



into  the rat of nonlinear or  dose-dependent Michaelis-Menten  kinetics.   These



workers  suggested that  a  dose  of 15 rag/kg  EDC in  the  rat is  below  hepatic



metabolism saturation.



     Spreafico et al. (1978, 1979, 1980) intravenously administered EDC in water



to Sprague-Dawley rats as bolus  injections of 1, 5 and 25 mg/kg and determined



the whole blood concentration  of EDC  as  it decayed with time.  Figure 9-3 shows



the semilog plot of the results of these experiments.  A biphasic decline of EDC



blood concentration was  evident  for all doses,  indicative of a two-compartment



system,  with a distributive  phase (a)  and  an excretion phase (B).  The data for



intravenous  treatment  were  analyzed  by computer fitting  to a two-compartment



open model with the results  shown in  Table 9-10.  Of immediate note is the fact



that the kinetic parameters  are not independent of the dose as required by linear



kinetics.  Thus  the  half-time (T..,-) of body elimination  from central or blood



compartment  increases  with  the  dose,  while whole-body clearance  (Cl)  and the



volume of distribution (V.)  decrease with an increase of the dose.  These results
                         d


indicate that  the rate of whole-body excretion of EDC is  largely determined by



the metabolism of EDC, a saturable process and a major route of elimination.



     Spreafico and his co-workers (1978,  1979,  1980) have  also  investigated the



whole-body kinetics  of  oral administration of  EDC  in rats.   Figure 9-1 shows



semilog  plots  of blood  and  tissue concentrations  during and  after absorption



(excretory phase)  following single oral doses of 25,  50 and 150 mg/kg given in



corn oil.  In general, the slopes of  the disappearance curves  (excretion compo-



nent) for adipose, liver and lung tissues  roughly parallel and  reflect the blood



decay curve.   The  data for  blood were computer-fitted to  the kinetic equations
                                      9-22

-------
                                 I   I   I  I   I   I   I   I   I   I   I  I   I
001
                     0.5 h
                                                             VBh
Figure 9-3. Blood levels of EDC m rats after i.v. administration.  ( • ) 25 mg/kg i.v.;
( O ) 5mg/kg i.v.; ( A) 1 mg/kg i.v.
Source: Spreafico et al. (1978. 1979, 1980).

-------
                                  TABLE 9-10

         Pharmacokinetic Parameters of EDC Administered as Single Bolus
         Intravenous  Injections  in Saline  to  Sprague-Dawley Male  Rat|.
            Parameters  Calculated from a Two-Compartment Open Model.
Parameter
Co, ng x ml'1
Kel> min'1
K-pi min~
K, 1 , min~
AUC, fig x min x
ml"1
T 1/2 B min
Clearance
ml x m~
Dose , mg/kg
1 5 25
1.50 8.00 38.12
0.277 0.142 0.078
0.121 0.063 0.040
0.158 0.140 0.118
9 54 595
7.30 9.49 14.07
21.98 17.46 7.98
Vol. distr., ml 231 239 162


« 	

K12

•^^^ ^
*21
              el  (inhalation)

                  or

             V / (Kffl + C) (oral)
Source:   Spreafico et al., 1979, 1980
                                        9-24

-------
for a two-compartment open model  for  oral absorption;  the results are given in
Table 9-11.  Like the kinetics observed  for  intravenous  dosing,  the values for
the  parameters,  T^gi  Cl and  V., are dose  dependent.    Furthermore,  the oral
dosing vehicle also affects the parameters.   Comparison  of the absorption rate
constants, K   half-times of elimination, T.,~ an<* volumes of distribution, Vrf,
for 15 mg/kg EDC in oil and 25 mg/kg EDC in water indicates a faster absorption of
EDC  in  water,  a shorter  half-time  of body  elimination  and a small  volume of
distribution.  Nonetheless,  the AUCs indicative of the total absorbed dose were
similar for oil and water vehicles,  as were the body  clearance  rates of EDC.  On
the other hand, no significant differences in the  kinetic parameters were seen
for male and female rats  given the  same  dose, nor were significant differences
observed between a  single dose and 11 daily administrations of the same dose (50
mg/kg) of EDC.
     Spreafico et  al.  (1980) included  in their  comprehensive studies  on the
pharmacokinetics of EDC  in the rat, experiments  to determine the kinetics of EDC
after inhalation dosing.   Rats  were  exposed to 50  and 250 ppm EDC for 5 hours, a
period  sufficiently  long to  establish  and  maintain  whole body  steady-state
conditions with  these  inspired  air concentrations  (Tables 9-6 and  9-7).   The
exposure was terminated,  and blood and other tissues were sampled  to determine
the decay of EDC  concentrations in these tissues with time.  The results of these
experiments are presented as semilog plots of tissue  concentrations versus time
in  Figure  9-1.   As for  oral  dosing, the  slopes of  the tissue decay  curves
parallel and reflect generally the blood  concentration curves.   In  contrast to
intravenous and  oral  dosing, which have  several components  to their  curves
representing absorption  and/or  distribution,  the  curves  after  steady-state
inhalation   are   monophasic,  representing    only   excretion.     Thus,   the
pharmacokinetic parameters calculated  from the blood  decay curves following the
                                     9-25

-------
                                    TABLE 9-H

           Pharmacokinetic  Parameters of EDC  Administered as Single Oral
              Doses in Corn Oil and Water to Sprague-Dawley Male Rats^
              Parameters  Calculated from a  2-Compartment Open Model.
Parameter
Oil Vehicle
Ka, mln~1
K , min"
AUC, fig x min x ml
T 1/2 B min
Clearance, ml x min"
Vol. Distr., ml.
Aqueous Vehicle
K , min"1
ci
K lf min"
AUC, fig x min x ml"
T 1/2 B min
Clearance, ml x min"
Vol. Distr., ml.
Dose, mg/kg
25 50
0.209 0.185
0.029 0.017
446 1700
24.62 44.07
10.64 5.58
367 328

0.299
0.046
446
14.12
10.20
221
150
0.109
0.010
7297
56.70
3.90
390






Source:   Spreafico et al., 1979, 1980
                                          9-26

-------
   1000
1
8

2
o
o
    100 —
| T
—
I I 1 I 11
i
               0.6h
   1000
    0.1
              0.5h
          Figure 9-4.  Levels of EDC in rats after inhalation exposure
          to 50 ppm (top) and 250 ppm (bottom).  Levels were mea-
          sured at termination of 5-hour exposure period.  Adipose
          tissue ( • ), blood ( o ), liver ( • ), lung ( A ).


          Source: Spreafico et al. (1978,  1979, 1980).

-------
t  i-dy-state  inhalation  conditions are essentially those  applicable to a single



c^apartment  open model.   The  kinetic  parameters are given in Table 9-12.   The



bcdy burden or "dose" of  EDO in the animals at termination  of inhalation exposure



is not known;  however, a comparison of the areas under the blood  decay curves, a



reflection of body  burden,  suggests that the body  burden  after 250 ppra exposure



was 40 times, not the 5  times expected of  the  50  ppm exposure.  Similarly, the



half-life  of  EDC  and the  zero-time  blood  concentrations  are  dose-dependent.



These  results from  inhalation exposure are  in  accord with the dose dependent



pharmacokinetics exhibited  after intravenous  and oral  dosing  of EDC.   They



strongly indicate that the  kinetics of absorption,  distribution  and elimination



of EDC are appropriately described by  nonlinear kinetics  that take into account



saturable  processes,  presumably  in  this  case the  metabolism  of EDC  by the



organism.



     The pharmacokinetics of  EDC after inhalation have  also  been explored by



Reitz  et  al.  (1980,  1982)  in male Osborne-Mendel rats.   These  investigators



determined the whole-blood levels of EDC during and following a 6-hour inhalation



exposure to   150  ppm EDC.   Figure  9-2 shows the  data  obtained  from  4 rats,



computer-fitted  to  the equations  of a two-compartment model and the idealized



computer-fit  plotted.   Within 2 hours  of  exposure,   the  animals  approached a



steady-state  blood  concentration (9 ug/m£)  with the inspired air concentration;



but, following termination of exposure  at 6 hours, the blood concentration of EDC



rapidly decayed  to  near  zero within  10 hours.  Figure 9-2  shows also the semilog



plot of computer-fitted  data of  EDC blood  concentration  levels  vs.  time after



exposure termination.  In contrast to the observation  of Spreafico et al. (Figure



9-4) that  only a raonophasic plot was obtained following inhalation exposure,



Reitz et al.  found a biphasic elimination with an initial rapid alpha phase and a



slower beta phase.   Table 9-13 gives  the pharmacokinetic parameters calculated



from a  two-compartment  open  model.   These results  may  be  compared  with  data



obtained by Spreafico et al.  for inhalation exposure  (Table  9-12).





                                       9-28

-------
                                    TABLE 9-12

               Pharmacokinetic  Parameters  of EDC  Following Termination
       of Steady State Inhalation Conditions (5-hr  Exposure)  of  50  and 250 ppm
                            to  Male  Sprague-Dawley  Rats.
Parameter

CQ, ug x ml~
i
K ., min
el
T 1/2, min
i
AUC, }ig x min x ml
Exposure
50
1.42

0.0561

12.69

26
Concentration, ppm
250
30.92

0.0313

22.13

1023
aSource:  Spreafico et al.,  1979,  1980

-------
                                  TABLE 9-13

            Pharmacokinetic Parameters of EDC Following Termination
     of Steady State Inhalation Conditions; 6-Hr Exposure, 150 ppm to Male
  Osborne-Mendel Rats.   Parameters Calculated from a Two-Compartment Model
             Parameter

             CQ, jig x ml~1                           9.0

             K, min"1                              0.092
             K21> min"1                              0.025"

             AUC, |ig x min  x  ml~1                 3018


             T1/2B min                              35
             Vol. Distr, ml.                       513
aSource:  Reitz et al.,  1980
                                        9-30

-------
     Reitz et al.  (1982) also determined in the  rat  the  pharmacokinetics after
single oral dosing (150 mg/kg in  corn oil).  In this case, a semilog plot of blood
EDC levels after absorption produced an elimination  curve typical  of nonlinear
kinetics.  These workers therefore computer-fitted the data to a two-compartment
nonlinear model and  calculated the rate of elimination from the central compart-
ment  (blood)  according to  Michaelis-Menten  kinetics.   Table  9-13  gives  the
phartnacokinetic parameters calculated  from the model.
     Several  differences   exist  between   the  observed   data  and  calculated
parameters of Reitz  et al. for inhalation vs. oral exposure (Table 9-13) as well
as those of Spreafico et al. (Table 9-12).  The EDC inhalation concentration used
by Reitz et al. (150 ppm)  was intermediate to those of Spreafico et al. (50 and
250 ppm).  Plateau blood concentrations in the three inhalation studies (1.1 at
50; 8.3 at 150; and  30.9 ug/m£ at 250 ppo), clearly demonstrate saturation of an
elimination process,  presumably metabolism.  Reitz et al. found peak blood levels
following oral dosing (30-44 ug/m£ at  150 mg/kg)  to be similar to those found by
Spreafico et al. after a 250 ppra inhalation exposure.  Reitz  et al.  also found a
biphasic elimination after both  oral and inhalation exposure with the second or
3 phase  essentially  equal  (T1/2  =  28  rain), and similar  to  the  monophasic
elimination calculated by Spreafico et al. for the 250 ppm  exposure (T1/2 = 22
min).  The relatively slow alpha elimination phase  (T1 ,~, 90 min) following oral
dosing of  150 mg/kg suggested to Reitz et  al.  a  saturable elimination process.
The  data  of  Spreafico and  co-workers are  generally in  agreement  with this
treatment; however,  several kinetic parameters  are  not in good agreement between
the  studies.   Of particular note  are the biphasic  and  monophasic elimination
curves and the large discrepancy of AUG.  The explanation for these differences
may  lie  in the different  kinetic models,   species  differences and  inability of
linear kinetics to accurately describe the kinetics of EDC body elimination.
                                      9-31

-------
     9.1.2.4.  BIOACCUMULATION —  There  is no definitive experimental evidence
in the  literature  concerning bioaccumulation in man after  chronic  or repeated
-:uj.ly exposure to EDC.  While EDC  has a moderately high fat tissue/blood parti-
_on coefficient (Table 9-2), its  half-time of elimination in man appears to be
=6-8 hours (Urusova,  1953).  In the rat,  Spreafico et al.  (1978,  1979, 1980) and
Reitz et  al.  (1980,  1982)  have  provided estimates of  half-time of whole-body
elimination of 25-57  minutes for acute oral dosing, and  13-35 minutes after 5-6
hour  inhalation exposures.   The. half-time  of  elimination  of  EDC from  the
epididymal adipose tissue  was essentially  the  same as whole-body elimination.
These  relatively  short  half-times  of  elimination  indicate  that the  risk of
significant bioaccumulation of EDC is small.  Based on  the pharmacokinetic data
from rats, these investigators predict essentially complete elimination of EDC
from the  body in 24 hours  following  acute  oral  exposure or  five  6-hour exposure
periods via  inhalation.   Furthermore,  Spreafico et al.  (1979,  1980) found no
significant  differences in the kinetic parameters of EDC elimination between  a
single  dose  and  multiple  daily  oral  administration of  EDC.
9.1.3.    Metabolism
     9.1.3.1.   KNOWN  METABOLITES  — Metabolism of EDC  in  man during or after
exposure has not been studied.   However,  one of the earliest  suggestive  reports
of an active mammalian biotransformation of EDC is related to human exposure. In
1945,  Bryzkin reported that  EDC underwent rapid  transformation  to  an  "organic
chloride" in patients who died  after ingesting  150-200  mi; EDC itself  was not
detectable  in tissues at autopsy.  Although Heppel and Porterfield  demonstrated
as early as  1948 that both ethylene dichloride and dibromide  were dehalogenated
by rat liver enzyme  preparations, current  knowledge of mammalian metabolism of
EDC derives  primarily from studies performed within the last  10 years.   Metabo-
lites  that  have been identified either in vivo in mice and rats or  in liver and
                                       9-32

-------
kidney tissue crude enzyme systems are listed in Table 9-11.  In addition to the



very  active  metabolite,  2-haloacetaldehyde, other possible  reactive  interme-



diates formed during the metabolism of 1,2-dihaloethylenes by glutathione-depen-



dent  reactions,  such  as  an episulfonium  ion,  may be involved  in  the  covalent



binding to  tissue  macromolecules leading to tissue damage  (Rannug  and  Beije,



1979; Hill et al.,  1978; McCann et al., 1975; Guengerich et al.,  1980; Anders and



Livesey, 1980).



     9.1.3.2.  MAGNITUDE   OF  EDC  METABOLISM  --  The  capacity  of  mammalian



organisms to metabolize EDC has been studied  only in the mouse  and rat.  In these



two  species,  the extent  of biotransformation  of single doses  of  EDC  has been


                                       11
estimated by balance studies utilizing   C-EDC.



     Yllner  (1971a)  administered intraperitoneally  doses of 50-170  mg/kg  of


11
  C-EDC to mice in metabolism cages and collected  the expired CO.  and  volatile



organic metabolites using  the  appropriate  solvent  traps.   Yllner  recovered



97-100$ of radioactivity  in exhaled air and  urine within 21 hours.   The results



of his balance studies are summarized in Table 9-9*  For the low dose of 50 mg/kg,



Yllner  found  that  EDC was  metabolized to  CO.  (13$)  and water  and  to  other



metabolites appearing in the urine  (73$).   Unchanged  EDC did not appear in the



urine, but 11$ of the dose was eliminated unchanged in exhaled air.   An increase



of the intraperitoneal dose resulted in a smaller  percentage of the d  se being



metabolized.  For the largest dose, 170 mg/kg,   55$  was  metabolized to  C02 (5$)



and urine metabolites  (50$).  These results indicate that extensive biotransfor-



mation (55-86$) occurs in the mouse.  Furthermore,  the  extent of metabolism is



dose dependent, suggesting a limited capacity for biotransformation with satura-



tion of the metabolizing system(s).  Saturation of metabolism to C0p and urinary



metabolites occurred at a dose level of between 100-110 mg/kg and 50-100 mg/kg,



respectively.
                                     9-33

-------
                                    TABLE 9-11

        Identified Metabolites  of Ethylene  Dichloride and Ethylene Bromide.
Metabolites
System
      Reference
Inorganic halide


co2


2-Chloroethanol



2-Chloroacetic acid

Bromoace taldehyde


Thiodiglycolic acid


Ethylene


S-(2-hydroxyethyl)-cysteine
N-acetyl-S-(2-hydroxyethyl)-
 cysteine
N-acetyl-S-(2-hydroxyethyl)-
 oxide

S-carboxymethyl cysteine

S-(2-hydroxyethyl)-glutathione
S-(2-hydroxyethyl)-S-oxide
rat liver
cytosol

mouse, rat
exhaled air

mouse, rat
urine, blood
mouse urine

rat liver
microsomes

mouse, rat
urine

rat liver
cytosol

rat liver
cytosol,
rat urine

rat urine

blood
mouse urine

rat liver
cytosol,
tissue
S,S'-ethylene-bis-glutathione     rat  liver
Heppel and Porterfield,  1918
Nachtorai,  1970

Yllner, 1971b
Reitz et al., I960

Yllner, 1971b
Kokarovtseva and
Kiseleva,  1978

Yllner, 1971b

Hill et al.,  1978
Yllner,  1971b
Spreafico et al.,  1979

Livesey and Anders,  1979
Nachtorai et al.,  1966

Edwards et al.,  1970

Edwards et al.,  1970




Yllner, 1971b

Nachtorai,  1970



Nachtomi,  1970
                                         9-31

-------
     Reitz et al. (1980, 1982) have found  that  EDC  is also extensively metabo-


                                                                     14
lized in the rat.  These investigators conducted balance studies with   C-EDC in



Osborne-Mendel rats  after  oral (150 rag/kg)  and inhalation (150 ppm for 6 hours)



exposure.  Their results are summarized in Table  9-3-  In the oral balance study,


             14
150 mg/kg of   C-EDC (or,  1520 ^moles/kg) were administered.  The sum of radio-



activity recovered in exhaled air,  urine and  feces, and that  remaining in the



carcass  was  101$,  or complete  recovery.    Some 29%  of  the dose  was excreted



unchanged in exhaled air;  5% was metabolized completely to   COp and 60% of the


                        14
dose was metabolized to   C-metabolites  appearing in  the  urine.   Less than 3%



radioactivity remained in  the  body  48 hours after dosing (probably as covalently



bound metabolites).   These results indicate that the extent of total metabolism



of the oral dose was =70% in these rats.  For the inhalation exposure study, the



absolute dose administered  by a 150 ppm,  6-hr  exposure was unknown.   From the



recovery after  termination  of  exposure  of   C-radioactivity  in  exhaled  air,



urine  and  feces,  and radioactivity remaining  in  the  carcass 48  hours after



exposure,  it was  estimated  that   the  inhalation  dose was  50.5  mg/kg  (512



^.moles/kg)  or  one-third  of  the  oral dose.    Of  this dose,  2%  was excreted



unchanged in exhaled air,  7%  was  metabolized  completely to    CO-,  and 84$ was


                14
metabolized  to    C-metabolites appearing  in  the urine.   Less than  5% of the



radioactivity was left in the  body  48 hours after inhalation exposure.  Ignoring



the pharmacokinetic differences  between  acute oral and  inhalation dosing, the



results of Reitz et al.  (1980)  demonstrate extensive  biotransformation  in  the rat



(70-91$) and also suggest a dose dependency with greater  metabolism for lower



doses, as Yllner (1971a) observed in the  mouse.



     The balance studies of  both Yllner (oral  dosing) and Reitz et al.  (oral and



inhalation dosing) demonstrate that the primary route of elimination  of EDC is by



metabolism.  Renal excretion  of  nonvolatile metabolites  predominates; however,
                                     9-35

-------
5-10? is metabolized completely to C02 and water.  It is of interest  to note that



a small amount  of EDC or its metabolites appear in  the  feces  (<2%),  even from



inhalation exposure.  Yllner (1971a) identified the  metabolites appearing in the


urine of  mice after dosing with  14C-EDC  (170  mg/kg)  and found  six l4C-metabo-



lites;  the  three  principal metabolites  were  S-carboxymethylcysteine  (455&),



thiodiacetic  acid  (33*)  and chloroacetic acid  (15*)  (Table 9-15).   However,


                                                           Ml
Spreafico et al.  (1979)  dosed rats with 50 and  150 mg/kg   C-EDC and found the



principal metabolite to be thiodiacetic acid;  chloroacetic  acid was not detected



and S-carboxymethylcysteine  was  found  only in  trace amounts.



     9.1.3.3.   PATHWAYS  OF METABOLISM — A number of studies have been carried



out describing  the metabolism  of 1,2-dihaloalkanes.  Two principal  pathways,



involving microsomal  and cystosolic enzymes,  respectively, have been proposed.



These pathways account for the formation of the metabolites  identified in both iji



vivo and in vitro studies listed in Table 9-14  and also suggest the nature of the


reactive   intermediates   involved  in   the   covalent  binding   to   cellular



macromolecules  (protein  and  DNA)  which has been demonstrated in both in vivo and



in vitro  studies  (Section 9.1.3-5).



     9.1.3.3.1.     Microsomal  Reactions — Yllner  (1971a) originally proposed



that  the  degradation of 1,2-dichloroethane  to 2-chloroacetic acid  involved a



primary  reaction in  which chlorine was  removed from one  of  the carbon atoms



(hydrolytic  dehalogenation)  to  yield 2-chloroethanol.   As evidence  for this



reaction, he  found chloroethanol to be a minor metabolite  in the urine of mice



injected  with EDC  (Table 9-15).   Following  2-chloroethanol  formation,  Yllner



proposed  that  alcohol  was  enzymatically converted to  2-chloroacetic  acid (a



major urinary metabolite) via 2-chloroacetaldehyde.  Kokarovtseva and Kiseleva



(1978)  have also identified chloroethanol  in the  blood  of rats  (T1/2  ca.  9



hours), and  in  rat liver tissue within 1 hour  and  for  24-48  hours after oral
                                      9-36

-------
                                   TABLE 9-15
         Percent Distribution of Radioactivity Excreted (18-hr) as Urinary
              Metabolites by Mice Receiving 1,2-Dichloroethane-  C or
                             2-Chloroacetic Acid-  C.
                                     After                   After
                                 dichloroethane           chloroacetate
Metabolite                        (0.17 g/kg)              (0.10 g/kg)
Chloroacetic acid                     16                      13

2-chloroethanol                        0.3

S-carboxymethylcysteine               45                      39

Conjugated S-carboxymethyl-            3                       3
 cysteine

Thiodiacetic acid                     33                      37

S,S-ethylene-bis-cysteine              0.9

Glycolic acid                         —                       4

Oxalic acid                           --                       0.2
aSource:  Yllner, 1971a,b
                                     9-37

-------
 administration of EDC (750 rag/kg).  Van Dyke and Wineman (1971), and subsequently



 Salmon et al.  (1978,  1981),  found that rat microsomal fractions, in the  presence



 of Qy  and NADPH, dechlorinated  a series  of haloalkanes including  EDC.   The



 reaction for EDC had a  Vfflax of  0.24  nraol/min/mg  protein  with a Km of 0.11 mM



 (Salmon et al.,  1981).  Thus, EDC and other haloalkanes interact with  cytochrome



 P-450 to give a Type I difference spectra associated with the metabolism of these



 substrates by direct C-hydroxylation  (Ivanetick et  al.,  1978).  Besides EDC,



 Ivanetick et al.  (1978)  observed that chloroacetaldehyde  and  chloroethanol also



 interacted with cytochrome P450 with K  values of 10.3, 30 and  =15,  respectively.
                                      s


 Hill   et  al.    (1978)  reported   that  1,2-dibromoethane   was activated  to  an



 irreversibly bound  species  by  microsomal  mixed-function oxidases,   and also



 provided evidence that 2-bromo-acetaldehyde was  formed in  such reactions.  More



 recently,  Guengerich et al.   (1980) showed that 2-chloroethanol was a  product of



 rat  microsomal mixed-function oxidation of  EDC,  requiring 02 and NADPH.  The



 reaction was blocked  by classic P450  inhibitors and  increased  by  pre-treatment



 with   phenobarbital.    Guengerich  et  al.  (1980)  proposed   that  part of the



 microsomal mixed-function  oxidative  metabolism  of EDC  proceeded from oxygen



 insertion into  a C-H  bond to form an  unstable chlorohydrin which  spontaneously



 dehydrohalogenates  to form  2-chloroacetaldehyde.    Figure 9-5 illustrates the



 proposed  reaction  and   pathway.     Chloroacetaldehyde  is   then  reduced  to



 chloroethanol by alcohol dehydrogenase in the  presence of NADH, or oxidized to 2-



 chloroacetic  acid by  aldehyde dehydrogenase.   Johnson  (1967)  previously had



observed that chloroethanol  was  readily dehydrogenated  to  chloroacetaldehyde by



alcohol  dehydrogenases  from  yeast or  horse  liver.   The  reaction,   therefore,



proceeds  in  either   direction  depending  on enzyme,  substrate  and cofactor



concentrations.   Williams  (1959) also  had  suggested that  chloracetic acid



appeared in vivo  via chloroacetaldehyde.
                                      9-38

-------
                  P450.0,  r  ii0-H  i
                  	-+• X-6-C-X
                  NADPH  L      M   J
                                    -HX
                                      Alco. D
                          -X-C-(
BINDING —I H'
PROTEIN. DNA 2 haloacat.   I
                                                    Hi
                 aldehyde  * Aid. D
                                             2 haloathanol
                         2-haloacatic acid
          I'i1    P450.0,  , F   5'1?'  4  "1
        -OH
                                                2 haloethanol
                        2 haloacataldahyda
                       T
                                Aid O
                             H.   ^)
                          CI-C— C-OH

                          2 haloaeatle acid
Figure 9-5 Postulated microsomal oxidative metabolism of 1 ,2-dihaloethanes.


Source:  Adapted from Guengench et al. (1980) and Anders and Livesey (1980).
                                  9-39

-------
      Further evidence for this  reaction sequence has been provided by Guengerich



et  al.  (1980).   These  workers observed  that covalent  binding to  microsomal



protein and DNA was  inhibited 30-40? by inclusion of alcohol or aldehyde dehydro-



genases in  the reaction mixture, strongly indicating that chloroacetaldehyde was




the reactive intermediate responsible  for this portion of the total  irreversible



binding of  EDO.  Chloroethanol  itself, when added to the microsomal  system, gave




only  a low  level  of  irreversible binding.  In order to account for the remainder



of  the  microsomal   mixed-function  oxidative  irreversible  binding  of  EDC,



Guengerich  et al.  (1980)  proposed  the oxidative formation of the reactive meta-



bolite 1-chloroso-2-chloroethane,   which  spontaneously  rearranges to  1-chloro-




acetaldehyde via a  hypochlorite.   The reaction  is illustrated in  Figure 9-5.



These workers noted  that  a reactive chloroso compound could  react directly with



raacromolecules,  or   hydrolyze   (with  release  of  hypochlorite  ion)  to  form



2-chloroethanol.



      As  a   consequence   of microsomal   P-450-mediated  oxidation   of EDC  as



illustrated in Figure  9-5, further  metabolism of  the oxidative metabolites,




2-chloroacetaldehyde  and   1-chloroso-2-chloroethane,    leads  either   to   a



"detoxification"   or  to  a  further   formation  of   reactive  intermediates,



respectively, as illustrated in Figure 9-6.




      Regarding detoxification reactions of chloroacetaldehyde  with glutathione,



Johnson  (1955,  1966a,b,  196?)  found that Chloroethanol, administered orally to



the rat,  caused rapid depletion of liver glutathione  (GSH) with a  concomitant



formation of S-carboxymethylglutathione.  In  vitro,  the reaction with a rat liver



cytosol  fraction  required stoichiometric  amounts  of  GSH  (1  mole)  and  NAD



(2 moles).    Since  pyruvate  was  also required  for  reaction,  Johnson (1967)



postulated   that  Chloroethanol was   converted  by alcohol   dehydrogenase  to



chloroacetaldehyde,    which     then    conjugated    with     GSH     to    give
                                      9-40

-------
       "OH                 T>H
2-cMoroacataldahyda   2-chioroacatata
        ,-cr
                     GSH
GT
GSH  1             7  -Ci-
        ty J
    GS-C-C-H
S-formylmathylglutathlona
        H.  *,
          DH
         i

cs-c-
-------
S-forraylmethylglutathione, and thence by an NAD requiring dehydrogenation to S-




carboxymethylglutathione.  Johnson (1966b) reported that chloroacetaldehyde also



rapidly conjugates with GSH in vitro by a non-enzymatic reaction at pH 7.0.  He



also has shown that S-carboxymethylglutathione is rapidly  degraded by rat kidney



homogenate to yield glycine, glutamic acid and S-carboxymethylcysteine, part of




which is further metabolized to  thiodiglycolic acid.  Yllner (1971a) found that



these  latter  two  metabolites  were  the  two  major  urinary   compounds  after



administering EDC to mice  (Table 9-15).  Since these two compounds were also the



major urinary  metabolites after administering 2-chloroacetate  to  mice (Table



9-15),  Yllner  (1971b)  proposed  that 2-chloroacetic  acid could also conjugate



with  GSH with  chloride excision  forming S-carboxymethylcysteine  and thereby




enter the pathway  (Figure  9-6).   Spreafico et al.  (1979)  found  that after rats



were given oral doses of 50 and 150 mg/kg EDC, 2-chloroacetic acid did not appear



in  the  urine,  but  thiodiglycolic acid did appear as  the  major  urinary product.



The difference between the urinary metabolites found in mice by  Yllner and  those




observed in  rats by Spreafico  et al. may be explained by  the availability of GSH




in  the  livers of the  animals  in  the  two  studies.



     Reactions of putative 1-chloroso-2-chloroethane: Guengerich et al. (1980)



noted that the chloroso compound that they proposed was formed by microsomal PU50




oxidative metabolism of EDC, would be extremely reactive and could be expected  to



either rearrange to 2-chloroacetaldehyde, hydrolyze to 2-chloroethanol,  or  react



directly  with microsomal  protein  (Figure 9-5).   In addition, these workers



postulated   that  the  chloroso  compound  may  react  with  GSH  to   form   S-(2-



chloroethyDglutathione,   a  half  sulfur  mustard,  which  could react  via  an



episulfonium ion  intermediate with macromolecules (Figure 9-6).  However,  in  an




Ames  test  with S.  typhimurium TA1535, EDC activated  with rat  liver microsomes,



NADPH,  0  and  added GSH,   did not enhance mutagenic activity.   In contrast,  the
                                       9-12

-------
use of 100,000 g liver supernatant containing GSH and GSH transferases markedly



increased the number of revertants per 1,2-dichloroethane plate.  These results



suggested that either the putative chloroso compound is not formed in the micro-



somal  system,  or that  differences in  the  subcellular  systems contribute  to



differences in covalent binding and rautagenicity.



     9.1.3.3.2.     Cytosolic  reactions  —  Heppel   and  Porterfield  (1948)



obtained an enzyme preparation  from rat liver capable of hydrolyzing the carbon-



halogen bonds of chloro derivatives of methane and ethane, including EDC.  Bray



et  al.   (1952)  also studied  the  dehalogenation of  dichloroethane  and other



halogenated   hydrocarbons    by   rabbit    liver   extracts   and   nonenzymatic



dechlorination by direct interaction with sulfhydryl groups of GSH and cysteine.



Naehtomi et  al.  (1966,  1970), found  that  an enzyme  system from  the soluble



supernatant fraction of rat  liver  catalyzed a reaction between EDC and GSH.  The



formation of inorganic halide  in  these studies could occur  as a consequence of



attack by GSH  resulting in  the excision  of halide,  and  the  formation of S-(2-



haloethyl)-GSH (Figure 9-7). This metabolite is  a reactive intermediate, a half



sulfur mustard,  which,  via its episulfonium ion and  further  reaction  with a



second GSH or with water,  may be expected to form S,S-ethylene-bis-GSH or S-(2-



hydroxyethyl)-GSH.   The S,S'-ethylene-bis-GSH may be  presumed to be subject to



degradation  to S,S-ethylene-bis-cysteine by  glutathione in  liver  and kidney.



Yllner (1971a,b) found small amounts of S,S'-ethylene-bis-cysteine in  the urine



of mice  injected with EDC  (Table  9-15).   Nachtorai et al. (1970) found that the



products of  the soluble fraction of  rat  liver  reaction with EDC  were S,S'-



ethylene-bis-glutathione and  S-(1-hydroxyethyl)glutathione.    Nachtorai  et al.



(1970) identified the same  two compounds  plus the sulfoxides  of the former in



urine of EDC- and dibroraoethane-treated rats.  Naehtomi et al.  (1966) have also



identified  N-acetyl-S-(2-hydroxyethyl)cysteine  in   the  urine  of  EDC-  and
                                      9-13

-------
   H,H,
ci-6-c-ci
     1
       CT
       GSH
GS-CH,-CH,-CI

S-C2-chloro«thylVGSH
         HOH
                         Ethylana
      
-------
1,2-dibromoethane-treated rats.   Jones and Edwards  (1968)  and Edwards  et  al.



(1970) confirmed the work of Nachtorai with dibromoethane, and moreover, isolated



the sulfoxides of both S-(2-hydroxyethyl)cysteine and its mercapturic acid from



the urine of dibromoethane-treated rats.



     The half  sulfur  mustard,  S-(2-chloroethyl)-GSH, and its  episulfonium  ion



formed by cytosolic GSH-dependent transferase metabolism  (Figure  9-7) has been



suggested  by  Rannug  et al.  (1978,   1979,  1980a)  to  be  responsible  for  the



mutagenic action  of  EDC in S.  typhimurium TA1535.   These  workers  found that



activation  of  EDC occurred  only  with  the  soluble  fraction  of   rat  liver



homogenates or  with  purified  GSH  transferase  enzyme in  the presence  of GSH.



Their findings  have been confirmed by Guengerich et  al.  (1980).   In addition,



Rannug and co-workers demonstrated that S-(1-chloroethyl)cysteine and N-acetyl-



S-(2-chloroethyl)-cysteine  produced  direct  mutagenic  effects  when  tested  on



Salmonella, whereas S-(2-hydroxethyl)cysteine showed  no  mutagenic effect.  These



results showed that the enzymatic degradation of the  GSH moiety does not abolish



the mutagenic properties of the conjugate,  whereas a substitution of the chlorine



with a hydroxyl group does.



     9.1.3.3.3.     Formation  of ethylene  —  Livesey  and  Anders  (1979) have



identified ethylene in rat liver and kidney cytosol incubated  with EDC.  Ethylene



was produced in only small amounts which were,  however, linearly independent on



the cytosolic protein  concentration.   The enzymatic  conversion was glutathione



dependent and  specific for this thiol.   Studies of substrate specificity with



1,2-dihaloethanes  showed  that  reactivity  following the halide order with C-Br



bond cleavage and elimination  occurring at a faster rate than C-C1  bond breakage.



Whereas EDC conversion was  primarily  an enzymatic reaction,  ethylene formation



occurred  equally well  from  1,2-dibromoethane  by a  nonenzymatic  reaction with



GSH.  The microsomal P-450 inhibitor, SKF 525-A,  had no effect on EDC metabolism,
                                      9.115

-------
but the reaction was inhibited by p-chloromercuribenzoic acid, methyl iodide and




diethylmaleate.  Livesey and Anders proposed  that the intermediate in the conver-



sion of EDC to ethylene was S-(2-chloroethyl)glutathione (Figure  9-7).  An analog



of this intermediate, S-(2-chloroethyl) cysteine, was nonenzymatically converted




to ethylene in the presence  of  glutathione and other  thiols.



     9.1.3.3.4.      Oxidation of EDC  to CO-  — Following oral administration of



14C-EDC to mice,  Yllner (1971a), and rat, Reitz et al. (1982),  found 5-10$ of the




dose administered  was metabolized  to CO..    Yllner  (1971b)  also observed that



after 2-chloroacetate administration, small amounts of glycolic  and oxalic acids



appeared in urine (Table 9-15).  Since these  acids  are known to be metabolized to




CO-, Yllner proposed that 2-chloroacetate,  arising from EDC metabolism (Figure



9-5) is enzymatically  hydrolyzed to glycolate by dehydrohalogenation, a portion



of which is further  oxidized to oxalic acid.



     9.1.3.3.5.      Comparison  of the Metabolism  of EDC  to Vinyl Chloride —



Rannug  and Beije  (1979)  have  drawn attention  to  the  similarities  in  the



biotransformation pathways of EDC and vinyl  chloride.   For both metabolisms, S-




(2-chloroethyDcysteine,  thiodiglycolic  acid,   S-(2-hydroxyethyl)cysteine  and



its  mercapturic  are  involved  as end  products.     Guengerich  et al.  (1980)




considered  the possibility  that reductive  dechlorination  of EDC would  yield




chloride  ion  plus  a  chloroethyl  radical,  which  could   either  react  with



macromolecular  targets,  or  lose  a  hydrogen  atom  to  form  vinyl  chloride.



Alternatively, some  other dehydrohalogenation mechanism (-HC1)  could give rise




to vinyl  chloride.   Vinyl chloride  could  then be microsomally  oxidized  to 1-



chloroethylene  oxide,  which  can  either  react  with  macromolecules,  or  be



rearranged  to 2-chloroacetaldehyde and proceed  through the  pathway  of Figure




9-6.  However, Guengerich et al.  found that reductive dechlorination of EDC to a



chloroethyl radical  was  an  unlikely reaction because metabolism and  binding of




EDC was dependent upon the  presence of 0_ in microsomal incubations containing






                                      9-46

-------
NADPH.  Guengerich  et al. calculated that the rate of EDC conversion to either a
total nonvolatile or to  an  irreversibly bound metabolite was  =25-50 times too
high  to  support an  obligatory role  for vinyl  chloride as  an  intermediate.
Moreover, they found that irreversible binding of  label  from  vinyl chloride to
microsomal protein  was  inhibited 95$ by either alcohol or aldehyde dehydrogenase
in microsomal incubations while these conditions produced only 30-10$ inhibition
of binding for EDC.  These  investigators concluded that the metabolism  of EDC
probably does not involve vinyl chloride as  an intermediate.
     9.1.3.1.  METABOLISM, TOXICITY AND MODIFIERS — Dichloroethane metabolites,
chloroacetaldehyde, chloroethanol  (oral  LD  Q  for  rats,  96 mg/kg),  and  chloro-
acetic acid (oral LD,.Q  for rats, 76 mg/kg) are several times more toxic than EDC
itself (oral LD5Q for rats,  770 mg/kg) (Heppel et  al.,  1945,  1946;  Woodward et
al., 1941; Ambrose, 1950; Hayes et al.,  1973).   Since the symptoms of poisoning
from  both accidental and occupational   exposures  of humans  and  experimental
exposures of animals to  these  metabolites are very  similar to those resulting
from EDC, it can be assumed that the toxicity of EDC for  both man and animals is
in large  part  the  result of biotransformation  to these metabolites.   Johnson
(1967) was the  first to suggest that chloroacetaldehyde may be  the premier toxic
metabolite, since this very reactive compound is capable of both enzymatic and
non-enzymatic  interaction  with  cellular  sulfhydryl   groups   (Figure   9-6).
However, Yllner (1971a,b) found that chloroacetic  acid also  reacted extensively
with sulfhydryl compounds in vivo.
     Heppel et al.  (1945, 1946, 1947)  found  a high  mortality (35$) in rats given
1.3 g/kg EDC orally.  Mortality was reduced by pre-  or  post-administration of
methionine, cysteine and  other sulfhydryl compounds.   Sulfur-containing amino
acids,  cysteine  and methionine,   also  protected  young rats   from  inhalation
                                     9-47

-------
exposure.  This protective effect  of sulfhydryl compounds is clearly related to



the marked  depletion of  glutathione  levels that occurs in  the  livers  of rats



given EDC, chloroethanol  or  chloroacetaldehyde (Johnson,  1965,  1966,  1967)  and



to the enzyme pathways of metabolism of EDC (Figures 9-5 to 9-7).  Johnson (1965,



1967)  has  noted  that   the  morbidity  and   mortality  of  young  rats  given



chloroethanol orally  was  reduced by concomitant administration of ethanol.   He



postulated that  the  protective  effect  of  ethanol was due  to  simple substrate



competition  for  alcohol dehydrogenase   which   catalyzes  the  conversion  of



chloroethanol to chloroacetaldehyde  (Figure 9-5).  Ethanol also inhibited early



effects of chloroethanol  on  liver  glutathione  depletion in these animals.  This



author  also suggested  that  the  minimal  toxicity observed  with  chronic  low



inhalation doses of  EDC  in different animal species may be simply explained by



the rapid replenishment of tissue  glutathione.



     Similar observations on the relation of the level of glutathione in liver



and other tissues, and toxicity of the chlorocarbons,  have been made by Jaeger



and his co-workers (1974, 1979).  Like EDC, vinylidene  chloride is detoxified by



glutathione-dependent pathways.  Jaeger et al.  (1974)  found in rats that an 18-



hour overnight fast decreased the LC,.- from a 4-hour  inhalation concentration of



vinylidene chloride  from  15,000  ppm  (fed)  to 600  ppm (fasted) and decreased the



concentration of vinylidene  chloride that produced a significant plasma eleva-



tion of a-ketoglutarate   transaminase, evidence of  liver  cytoxicity (2000 ppm,



fed; 150 ppm, fasted).  Increased  susceptibility  to hepatotoxicity was shown to



be related to a decreased hepatic  glutathione  concentration associated with the



fasting.  Jaeger (1979) also demonstrated  in rats a circadian rhythm for tissue



glutathione concentration of liver, blood,  lung and kidney.  For rats maintained



on a 12-hour light-dark cycle  (6 pm to 6 am, dark), hepatic glutathione content



was lowest at  6-10 pm and highest at  4-12 am; these periods correspond to the
                                      9-48

-------
greater  and  lesser  hepatotoxicity  respectively  associated  with  vinylidene
chloride exposure during these two periods.
     Nakajima  and  Sato  (1979)  studied  the  metabolism  of the  chlorocarbons
in vitro with microsomes from livers of fed  and fasted male rats and found that
the metabolism of EDC  increased  1.5-fold  as the  result  of  a 24-hour  fast,
although fasting produced no significant  increase in the microsomal protein and
cytochrome P-450 liver contents.   Microsomes  from  livers  of fasted female rats
displayed even more active metabolism  of EDC  (3.6-fold over fed rats).   These
observations suggest that the  increased  toxicity of EDC  that  occurs with food
deprivation may be  due not only to decreased liver glutathione content, but also
to a greater production  of toxic metabolites:  chloroacetaldehyde, chloroethanol
and chloroacetic acid.
     Sato et al.  (1980)  have studied the effect of chronic  ethanol consumption on
hepatic metabolism of chlorinated  hydrocarbons in rats to provide information on
the effect  of ethanol  consumption on  the  toxic effects  of chlorinated  hydro-
carbons in the work environment.  Male rats were maintained on a daily intake of
ethanol amounting to 30% of their total energy intake for 3 weeks.   The hepatic
microsomal metabolism of EDC was increased  5.5-fold over microsomes from livers
of ethanol-free rats, although ethanol feeding produced only a slight increase in
the microsomal P-450  content.  The  increase in enzyme metabolism of EDC occurred
only with  microsomal  fractions and not with  cytosolic fractions,  and one-day
withdrawal of ethanol feeding almost completely abolished the effect of chronic
alcohol consumption.
     Sato  et  al.  (1981) also  studied  the  effects  of  an  acute single dose of
ethanol (given as an aqueous solution by gastric intubation)  on the metabolism of
chlorinated hydrocarbons in  rats.   Hepatic metabolism of EDC (by a 10,000 g liver
fraction) added in  vitro was accelerated (up to 2.5-fold) by doses of ethanol up
                                     9-49

-------
to 14 mg/kg without causing  any  increase in P-U50 content.  The stimulation was



not seen and even suppression occurred  with higher doses of ethanol.  Increased



metabolism was  most  marked 16-18  hours after ethanol administration.   Ethanol



added  directly  to the  incubation mixture, however,  depressed EDC metabolism.



These investigators suggest that ethanol is capable of exerting a dual effect on



EDC-metabolizing enzymes, i.e., inhibition and stimulation,  depending on ethanol




concentration.



     9.1.3.5.  METABOLISM AND COVALENT  BINDING  — The  metabolism of EDC results



in the  production of reactive  metabolites of an  electrophilic  nature  such as




those proposed to be  formed in  the metabolic  schemes of Figures 9-5 to 9-7.  The



occurrence of these metabolites,  chloroacetaldehyde,  the putative  1-chloroso-2-




chloroethane, the half-mustard S-(2-chloroethyl)glutathione and its episulfonium



ion, provides a theoretical basis for the  covalent  binding observed with EDC to



cellular macromolecules  such  as protein and  DNA with  consequent possibility of



damage to cellular integrity and genetic apparatus.  Such  covalent  binding reac-




tions are known to be related to  teratogenesis,  mutagenesis and carcinogenesis,



although  chemical reactivity alone  is  not  evidence  of  genetic  damage (Lutz,



1979).  The amount of covalent binding,  and hence damage potential,  is related to



the  pharmacokinetics  of EDC  exposure.    At   least  in the rat,  there  is ample



evidence from pharmacokinetic studies of EDC  that the metabolic capacity  for EDC



metabolism is saturable  (Sections 9.1.2 and 9.1-3).   It may therefore be specu-



lated  that the organism when  confronted  with  low  quantities of  EDC utilizes



glutathione   conjugation    detoxification   reactions    (Figure    9-6)   and




nonelectrophilic  metabolites  are  primarily  produced, but when confronted with



higher  EDC doses,  increasing  amounts of electrophilic metabolites are produced



to a saturating and limiting  rate for their enzymatic pathways (Figures  9-6 and



9-7).   Reitz et al.   (1980, 1982) determined  total covalent binding and  binding
                                      9-50

-------
to DNA in rats exposed to 1l|C-EDC by  the  oral (150 mg/kg) and  inhalation (150
ppm, 6 hour)  routes.  The results of their  study with these presumably saturating
doses are given in Table 9-16.  These results show no striking difference between
the  two  routes of  exposure  in the  distribution of  covalent binding  and  DNA
binding among the various organ tissues,  although the tissue levels of covalent
binding  are  generally  higher  after inhalation  exposure for  total  binding  and
after gavage exposure for DNA binding.  The exposure doses used in these studies
of Reitz et al. are comparable to the oral doses of EDC use in the lifetime NCI
carcinogenicity  study  (NCI,   1978)  and  to  the   inhalation   doses  of  the
carcinogenicity studies in rats of Maltoni et  al.  (1980).  No excess tumors were
reported by Maltoni et al.,  while liver and forestomach were sites of malignant
tumors in the NCI study.  Recently,  Storer et al. (1982) determined DNA damage
produced in vivo in the livers  of male  B6C3F1 mice  following single oral doses of
EDC  (100 mg/kg).  DNA  damage was  evaluated by sedimentation of liver nuclei in
alkaline sucrose  density centrifugation,  and by DNA recovery, which  measure
single strand breaks in DNA or alkaline label sites.  DNA from EDC-treated mice
isolated U hours after dosing  sedimented more slowly and the total DNA recovery
was decreased 16£ from untreated mice.  However, dimethyl nitrosamine, used as a
positive control,  decreased recovery of DNA 61$.
     In contrast to the low covalent binding potential of EDC  in. vivo, inhalation
exposure of rats to only 20 ppm,  1,2-dibromoethane  (EDB;  7 hours/day, 5 days/week
for  18 months) produced tumors  in  the liver and kidney, sites  of covalent binding
(Hill et al.,  1978).  Furthermore,  Plotnick et  al.  (1980)  found that the carcino-
genicity of  EDB  was enhanced  by  the dietary addition  of  disulfiram (0.055S by
weight; an inhibitor of aldehyde dehydrogenase) which blocks the further oxida-
tion of broraoacetaldehyde formed in the metabolism of EDB (Figure 9-5) and which
presumably  results  in  increased  tissue  levels of  bromoacetaldehyde  with  an
                                     9-51

-------
                                     TABLE 9-16
         Total Macromolecular Binding and DNA Binding in Selected Tissues
          of Rats After Exposure  to   C-EDC by Oral or Inhalation Routes
                                    Nanomole equivalents EDC/g tissue
                                    oral                    inhalation
                                  (150 mg/kg)              (150 ppm 6 hr)
Total Binding (n=4)

Liverb                            175 + 24                  268 + 45
Kidney                            183 + 25                  263 + 48
Spleen                             65+21                  130 » 22
Lung                              106+34                  147+16
ForestomaGh                       160+19                   71 ± 19
Stomach                            90+2                  156+29

DNA Binding  (n=3)

Experiment 1
Liver"
Spleen
Kidney
Stomach
Experiment 2
Liver
Spleen
Kidney
Stomach
21.3 + 7.4
5.8 + 0.7
17-4 + 2.3
14.9

13.9 + 2.1
2.5 + 0.3
14.5 + 6.2
6.7
8.2 + 3-3
1.8 + 0.3
5.2 + 3.7
2.8

3.3 + 1.2
1.8 + 0.5
2.0 + 0.3
1.9
Results are reported as nanomole equivalents of EDC/g tissue + S.D.
Animals were sacrificed U hr after oral dosing or* immediately following a 6-hr
inhalation exposure.


 Source:  Reitz et al., 1982


 Site where malignant tumors were observed in the NCI study (1978) after EDC was
given by gavage, also forestomach.  No excess tumors were reported in the inhala-
tion carcinogenicity bioassay  (Maltoni et al., 1980).
                                        9-52

-------
increase of covalent binding.  Plotnick et al. (1980) showed that dietary disul-


                                            14
firam markedly increased covalent binding of   C-EDB in the nuclei of liver cells



in rats dosed with EDB (Table  9-17).



     Covalent binding of metabolites  from both oxidative rnicrosomal  and cyto-



solic metabolism of EDC  has been demonstrated in vitro by several investigative



groups.  Van Duuren and  his colleagues (Banerjee and Van Duuren, 1979; Banerjee



and Van Duuren,  1978,  Banerjee et al., 1979, 1980) observed microsomal-activated



covalent binding  of both EDC  and EDB to  native DMA from salmon sperm,  and to



liver and lung microsomal protein from rats  and mice.   Binding to DMA  did not



occur in the absence  of raicrosomes or in  the presence of denatured microsomes.



Cytosolic metabolism  produced insignificant  metabolic activation  and binding.



The  microsomal-activated   binding   was   enhanced  by   phenobarbital  and  3-



methylcholanthrene pretreatment,  whereas  the  addition  of  glutathione  to the



microsomal reaction reduced the  binding.   Similar results were obtained by Sipes



and Gandolfi (1980).   Van Duuren and his  co-workers suggested  that EDC and EDB



were biotransformed by microsomal oxidative reactions to the reactive transient



metabolites, 2-haloacetaldehyde,  2-haloethanol or  haloethylene oxide,  all of



which are electrophilic  in  nature.  Banerjee et al.  (1979) demonstrated that both



2-bromoacetaldehyde and  2-bromoethanol  bind  covalently  to   protein and  DNA


                                                                        14
without metabolic activation.  Guengerich et al. (1980) have found that   C-EDC



is metabolized  by both microsomes  and  cytosolic  systems to  metabolites that



covalently bind to protein and  calf  thymus DNA.  Cytosolic metabolism depended



upon the presence of  glutathione and  involved  glutathione  transferases (Figure



9-7), although glutathione  inhibited microsomal-activated binding to protein but



stimulated binding to DNA.   These investigators  also produced evidence that 2-



chloroacetaldehyde,   S-(2-chloroethyl)glutathione,   and   their   putative  1-



chloroso-2-chloroethane  are the  metabolites involved in the covalent  binding.
                                     9-53

-------
                                  TABLE 9-17


                                                 14
           Effect of Dietary Disulfiram Upon the   C Content  of Liver


           Nuclei Isolated 24 or  48 Hours After Administration of a


                    Single Oral Dose of 15 rag/kg [U-1 C]EDBa
Time Interval                Control                 Disulfiram




   24 hours                  687 ±  82b                1773 ± 314



   48 hours                  460 +  42                 1534 + 197
aSource:  Plotnick et al.,  1980



bResults are expressed as dpra/pellet  (mean  +  S.E.M.)  of duplicate determinations

 on 6 animals per group  at  24-hr  and  5  animals  per  group at 48 hr.
                                         9-54

-------
9.1.1.     Summary and Conclusions.   At ambient temperatures, EDC  is  a volatile


liquid  with appreciable solubility in water, and hence the  principal routes of


entry to the body are by pulmonary and oral absorption.  The pharmacokinetics of


absorption and excretion  of EDC have been studied extensively  in  the mouse and


rat. Absorption from the gastrointestinal tract is rapid and complete, occurring


by first order passive processes with a half-time of  <6 rain.   A dose-dependent


first-pass effect with pulmonary elimination of unchanged  EDC  occurs with oral


ingestion, thus decreasing  the amount reaching  the systemic circulation.  At the


low concentration of EDC  existent in food and water,  nearly complete extraction


by  the  liver is  expected.   With  inhalation  exposure,  a  blood/air partition


coefficient of  =20  at 37°C has  been  observed.  Tissue distribution of EDC is


consistent with its  lipophilic nature.  The chemical crosses the blood brain and


placental barriers and distributes  into  breast milk.   The adipose tissue/blood


partition coefficient varies from 7-57 depending on dose.


     Excretion  of  unmetabolized EDC  is  almost  exclusively  via  the lungs;


however, metabolism and excretion of the metabolites by other routes is extensive


and dose related. In the mouse and rat, after both oral and  inhalation exposure,


the half-life of EDC in blood increases with dose,  although it is <60  rain at high


doses.  The parameters of total body elimination are compatible  with a 2-corapart-


ment system and Michaelis-Menten kinetics.  The short half-life of EDC  suggests


that the risk of bioaccumulation from intermittent multiple oral and inhalation


exposures  is  small.   Daily oral  dosing of rats does  not  result in  significant


bioaccumulation in blood or other tissues.


     Pharmacokinetic  parameters  on  multiple  doses  of EDC are  very  limited.

                                          Ill
Treatment  of animals with  EDC  prior to    C-EDC  may  alter the detoxification


mechanisms,   thus   leading   to   enhanced   toxicity   and    carcinogenicity.


Biotrans format ion of EDC has been shown  to  occur  by  multiple pathways, in  both
                                      9-55

-------
liver microsomal and cytosol  fractions.  In  the mouse and rat, up to 90% of low



oral  or  inhalation  doses  (<50 mg/kg)  are  metabolized,   with  a  decreasing



percentage of the dose metabolized as the dose approaches and exceeds saturation



of metabolic  capacity.   Metabolism produced  2-chloroacetaldehyde, S-(2-chloro-



ethyDglutathione  (a  half sulfur mustard) and other putative  reactive metabo-



lites capable of covalent binding to cellular macromolecules, as well as nonreac-



tive glutathione conjugates  from "detoxification reactions."  The intensity of



covalent binding of reaction metabolites to proteins, lipids and DNA, however, is



considerably  less  than  that  observed  with  the carcinogenic bromine analog and




other known carcinogens.



     Exposure  to EDC  decreases  liver  levels of reduced glutathione in a dose-



dependent  manner.    Glutathione  plays an  important role  through  glutathione



conjugation detoxification mechanisms  in protecting against  binding and cellular



toxicity.    Covalent  binding,   hepatotoxicity  and  mortality  of experimental



animals are  all reduced by administration  of sulfhydryl  amino acids or gluta-



thione.  Conversely, toxicity is enhanced by low liver and tissue  concentrations



of glutathione.  Phenobarbital and other inducers of P-450 metabolism increase



the metabolism of EDC, the  toxicity and covalent binding. Fasting  reduces gluta-



thione  tissue content and  increases  both the  metabolism and  toxicity of EDC.



Ethanol, acutely or chronically  administered,  may  either enhance  or inhibit EDC



metabolism and  toxicity depending on  the tissue alcohol concentration.
                                      9-56

-------
9.2. ACUTE,  SUBCHRONIC AND CHRONIC TOXICITY



9.2.1.  Effects in Humans.  The preponderance of reports on the toxicity of EDC



to humans was published in the foreign literature (primarily German, Russian, and



Polish).  Many  of  these  reports involved exposure to high  levels of compound,



involved single cases or  small numbers  of individuals, were anecdotal in nature



and discussed observations that related to the overall toxicity of this chemical



in man,  and/or provided little detailed  correlation between toxic effects and the



amount or duration of exposure.   Since detailed reviews of the foreign studies



were available  from NIOSH (1976) and the U.S.  EPA (1979),  these sources were used



in part as a basis for the following discussion of human health effects.  Trans-



lations were obtained and evaluated independently for all  foreign studies that



provided dose-response data of relevance to  human risk  assessment.   Reports in



which exposures were not  primarily to EDC  (i.e., most reports  of mixed solvent



exposures) were not discussed.



     9.2.1.1.   ACUTE EXPOSURES



     9.2.1.1.1.     Case  Reports and Surveys



     9.2.1.1.1.1.    Oral  Exposure.  Information on the  effects of ingested EDC



is available solely from  clinical case  reports and  surveys involving accidental



and intentional exposures.  Many of the cases involved fatalities and described



symptoms  and  signs  of poisoning  that  preceded  death.    The progression  of



signs/symptoms and the outcome/findings  of cases  in which  the quantity of EDC



ingested leading to death was reported  are summarized in Table  9-18.  Ingestion



of  quantities  of EDC  estimated to range  from 8 to 200 mH ( = 143-3571  mg/kg,



assuming 70 kg body weight) were reported as lethal  to  adult males.  Following



ingestion there was often a latent period, generally 30 minutes  to 3 hours, prior



to the onset of symptoms.   Symptoms were indicative  of  CNS  effects  and gastro-
                                     9-57

-------
        4 males/20  to 29 years
        3 males/19  to  27  years
vC



00
               a
        Male/NS
        Hale/63 years
                                                                           TABLE 9-18

                                        Effects Associated with Acute Lethal Oral Doses  of Ethylene Dlchlorlde In  Humans
Patient8
Sex/Age
Amount
Ingested
Symptoms and Signs Outcome and Findings
Reference
150 to 200 mi
(»188 to 250.6 g)
70, 80 and 100 ml
(=67.7, 100.2 and
125.3 g)
=82 ml ("102.7 g)
mixed with coffee
and beer

2 oz. (»75.2 g)
mixed with orange
Juice and ginger ale
Symptoms were not specified,
but appeared after 3 to 1 hours.
Onset of symptoms within a
few minutes:  vomiting;
weakness; dizziness; lost
consciousness
Intoxication; vomiting;
diarrhea; unconsciousness;
dypsnea

Onset of symptoms after 2 hours:
nausea; faintness; vomiting.
Subsequently, cyanosis; dilated
pupils; coarse rales; weak
and rapid pulse; diarrhea;
increased cyanosis; absence
of pulse and heart sounds;
dypsnea
Deaths after 10, 15, 33 and     Bryzhin, 1915
35 hours.  Punctuate
hemorrhaging In the epi-
cardlum, pleura, and mucous
membranes of the stomach and
duodenum; varying degrees of
liver damage with focal
hemorrhaging in one case; yellow-
white flbrinous bundles of blood
In the heart cavities and lesser
circulatory vessels; hemolytic
jaundice of the endocardium,
aortal Intima, and dura mater;
evidence of decomposition of
circulating erythrocytes

Deaths after 5 to 8 hours       Kalra,  1966
Hyperemla and hemorrhagic
lesions  (see text);
evidence of overt bleeding
into the visceral organs

Death after 6 hours
Hyperemla and hemorrhagic
lesions  (see text)

Death after 22 hours
attributed to circulatory
failure.  Extensive
hemorrhagic colitis; nephrosls
with calcifications of  the
tubular  epithelium and  tubular
and vascular elastic membranes;
fatty degeneration of the liver
spleen and lungs; multiple
perivascular hemorrhages of
the brain
                                                                                                                               Noetzel,  1944
Hueper and Smith,
1935

-------
                                                                TABLE 9-18 (cont.)
       Patient8
       Sex/Age
    Amount
   Ingested
     Symptoms and Signs
    Outcome and Findings
   Reference
Male/16 years
Male/80 years
Male/18 years
Male/57 years
50 ml
(=62.7 g)
50 mi
(=62.7 g)
50 ml
(=62.7 g)
10 mSL
(= 50.1 g)
Vomiting; epigastric pain;
muscle spasms, hiccups, rapid
pulse and lack of eyelid
response to light on the 4th
day

Elevated serum enzymes:  LDH;
SCOT; SGPT; alkaline phosphatase
glutamlc dehydrogenase; RNAase.

Onset of symptoms after 1 hour:
somnolence and cyanosis.
Diarrhea after 4 hours.
Impaired blood coagulation
after 5.5 hours:  Increased
prothrombln time; decrease In
clotting factors II and V;
thrombocytopenla; no Increase
In fibrlnolysls

Somnolence; vomiting; sinus
tachycardia; ventricular
extrasystoles; regained
consciousness after 14 hours;
dypsnea; loss of blood
pressure; cardiac arrest.
Impaired blood coagulation
observed after 24 hours;
prolonged bleeding from
venlpunctures; reduction in
activity of clotting factors
II, V, VII and VIII; complete
defibrinatlon; thrombo-
cy topenla; Increased thrombin
time
Death after 91 hours
Death within a few hours
Death after 17 hours
attributed to circulatory
shock.  Intravascular
thrombosis were not found.
Roubal, 1947
Secchl et al., 1968°
Schonborn et al.,
1970°
Death after 24 hours.
Thrombi In the pulmonary
arterloles and capillaries;
hemorrhages into the mucosa
of the esophagus, stump of the
stomach, rectum and in the
subeplcardial, subendocardlal
and myocardial tissues.  The
coagulation disorder with
thrombocytopenla attributed
to disseminated intravascular
coagulopathy and hyper-
flbrlnolysis
Martin et al., 1969

-------
                                                                  TABLE  9-18  (cont.)
Patient3
Sex/Age
Hale/30 years
Amount
Ingested
40 ml
(» 50. < g)
Symptoms and Signs
Slight cough; reddened
conjunctlvae; shock; weak,
Outcome and Findings
Death after 28 hours
Hyperemia and hemorrhagic
Reference
Garrison and
Leadingham, 1954°
Hale/50 years
Hale/55 years
 (asthmatic)
Hale/NS
Hale/NS
=•30 ml
(=37.6 g)
20 ml
(=25.1 g)
=20 ml
(=25.1  g)
920 ml
(=25.1  g)
rapid pulse.  Regained
consciousness after
3 hours; hyperactlvlty
alternated with
semlconatose condition

Onset of symptoms after
30 minutes: unconsciousness;
vomiting; cyanosis;
dilated and fixed pupils;
pulmonary edema; extreme
dypsnea
Epigastric pain; extreme
dizziness; sleeplessness;
vomiting; slow pulse

Onset of symptoms after 1 hour:
repeated vomiting. Cyanosis
and dypsnea after 12 hours

Symptoms not reported
                                                                                       lesions (see text);  evidence
                                                                                       of overt bleeding into the
                                                                                       visceral organs
Death after 10 hours
Diffuse hemorrhagic
gastritis; bilateral
pulmonary congestion;
acute necrotizlng
bronohlolltls and
bronchitis; acute toxic
nephrosis; diffuse hepatic
necrosis; hyperemla of the
brain with scattered perl-
vascular hemorrhages In
the pons

Death after 24 hours
Death after 13 hours
                                                                                       Death within 12 hours
Lochhead and Close,
1951
Roubal, 1947b
Flowtow, 1952"
                                Flowtow, 1952

-------
                                                                                TABLE  9-18   (cont.)
                    Patient"
                   Sex/Age
 Amount
Ingested
   Symptoms and Signs
    Outcome and Findings
                                                                     Reference
               Male/11 years
15 mH
(» 18.8 g)
vO
               Male/32 years
8 ml
(=00 g)
               Male/32 years
glass
Onset of symptoms after 2 hours:
Progressive appearance of
severe headache; staggering;
lethargy; periodic vomiting;
decreased blood pressure;
ollgurla; dypsnea; somnolence;
Increased dypanea; and ollgurla,
hemorrhaglc nasogastrlc aspirate;
ecohymoses; sinus tachycardia;
cardiac arrest, pulmonary
edema; refractory hypotension.
Hypoglycemla on day 2 and
hypercalcemla on day 4.
Progressive decrease In blood
coagulation ability:  increased
prothrombln time; all clotting
factors except VIII were
markedly decreased on day 4.
Death after 6 days
Extensive mid-zonal liver
necrosis; renal tubular
necrosis; focal adrenal
degeneration and necrosis
Yodaiken and
Babcock, 1973
Burning sensation in mouth,
throat and stomach; drank milk
and vomited; weakness; speech
retardation; lethargy; asthenia;
cold sweat; muffled heart sound;
weak and rapid pulse.  22 hours
after ingestion:  excitation;
restlessness; delirium; flushed
face; systolic murmur; respiratory
depression; circulatory weakness;
anuria

Vomiting, weakness, stomach
pains.  On 3rd day: rest-
lessness, coated and dry
tongue; bloody diarrhea;
abdomen painful on palpitation;
enlarged liver; dry, moist rales;
rapid, weak pulse; anuria;
increasing cyanosis; un-
consciousness
                                                                                                      Death after 56 hours
                                Bogoyavlenski et al.,
                                19686
Death on the 3rd day
Hyperemia and hemorrhagio
lesions (see text)
                                                                                                                                      Agranovlch,  19*18

-------
                                                                                  TABLE 9-18  (cont.)
Patient3
Sex/Age
Amount
Ingested
Symptoms and Signs
Outcome and Findings
Reference
                Hale/27 years
half glass
                Hale/43 years
                (alcoholic)
4 drinks diluted
with orange Juice0
o
 i
ro
                Hale/43 years
                (alcoholic)
                Male/NS
4 drinks diluted
with orange juice0
                                             several mouthfuls
Onset of symptoms after
2.5 hours:  Unconsciousness
that was regained after
12 hours; vomiting
(dark vomitus); burning
sensation in the digestive
tract; dypanea; nausea;
cyanosis; depressed
respiratory rate; moist
rales; muffled heart sounds;
rapid pulse; extrasystoles;
anurla

Unconsciousness
Death after 19 hours
Hyperemia and henorrhagic
lesions (see text); evidence
of overt bleeding into the
visceral organs
Confusion; deep sleepiness;
unconsciousness; vomiting
with blood
                        Onset of symptoms after
                        30 min:  vertigo; nausea;
                        vomiting; slightly muffled
                        heart sound; tachycardia.
                        After 17.5 hours: headache;
                        substernal pain; cyanosis;
                        rapid, weak pulse; dry rales;
                        decreased blood pressure;
                        vomiting with bile; diarrhea;
                        subconjunctliral hemorrhage;
                        ollgurla; paranephric hemorrhage,
                        5th day pulmonary edema and
                        unconsciousness
Bogoyavlenskl et al.,
1968
Death after 8 hours
Hyperemia and hemorrhagic
lesions (see text);
evidence of overt bleeding
into the visceral organs
and lungs.  Death after
24 hours.  Hyperemia and
hemorrhagic lesions (see
text); evidence of overt
bleeding into the visceral
organs and lungs

Death after 24 hours
Hyperemia and hemorrhagic
lesions (see text); evidence
of overt bleeding into the
visceral organs and lungs

Death on the 5th day
Hulst et al., 1946
Hulst et al., 1946
                                                                  Horozov, 195B

-------
                                                                7ABLB 9-18  (cont.)
     Patient8
    Sex/Age
 Amount
Ingested
   Syoptons and Signs
    Outcome and Findings
   Reference
Hale/63 years
1 or 2 sips
Male/1 1/2 years
Hale/79 years
Hale/2 years
Hale/23 years
1 alp
1 sip
1 alp
1  alp
Unconsciousness shortly after
Ingestion, but soon
regained; strong vomiting;
period of improvement;
unconsciousness again
after 10.5 hours; falling
blood pressure

Extreme weakness; comatose;
vomiting
Vomiting; weakness; pale;
cyanosis; scarcely conscious;
vagueness; rapid, regular
pulse; blood pressure not
measurable

Vomiting; diarrhea; tonic
spasms; increasing loss of
consciousness; dypsnea;
impaired circulation

Onset of symptoms after 1 hour:
dizziness; nausea; unconscious-
ness; vomiting; cyanosis; no
pupil reaction; no corneal
reflex; dypsnea; strong motor
unrest
Death after 14 hours
attributed to circulatory
failure.  Hyperenia and
hemorrhagic lesions (see t«xt)
Death the next day
Hyperemia and heoorrhaglc
lesions (see text)

Death after 10 hours
attributed to heart and
circulatory failure.
Hyperenlc and hemorrhagic
lesions (see text)

Death after 20 hours
Hyperemia and hemorrhagic
lesions (see text)
Death after 8 hours
attributed to respiratory
and circulatory failure
Hyperemia and heoorrhaglc
lesions (see text); evidence
of overt bleeding Into the
visceral organs
Freundt et al.. 1963
Keyzer,
                                                                                                                       Weisa, 1957
                                                                                                                       Relnfried, 1958°
Relnfrled, 1954
"NS s Hot stated
bData compiled from NIOSH, 1976
cPresumably EDC was mixed instead of alcohol

-------
intestinal disturbances,  and frequently included  dizziness,  nausea,  headache,



periodic vomiting,  diarrhea,  epigastric pain or tenderness,  dilated pupils and



lack of corneal reflex,  rapid  and weak pulse rates,  progressive cyanosis, dyspnea



and unconsciousness.   These  symptoms  of toxicity are  characteristic  of  those




produced  by  many  of   the  chlorinated  aliphatic hydrocarbons.    Somnolence,



oliguria  and  anuria,  muffled  heart sounds  and  motor  unrest  have also  been




described in a number of  cases.



     The results  of several  health  surveys  of  large numbers of orally exposed




individuals have recently been reported by  Russian investigators, and summarized



by PEDCo Environmental,  Inc.  (1979)  (Akimov et al.,  1976,  1978; Shchepotin and




Bondarenko,  1978;  Bonitenko  etal.,   1974,  1977;   Luzhnikov  et  al.,  197**;



Luzhnikov   and  Savina,   1976)  and   Chemical   Abstracts   (Andriukin, 1979).



Information regarding the design  of  these  surveys is not available, and dosage



information  is either  not  available   or  not  adequately  reported  (i.e.,  not




correlated  with effects)  (Table  9-19).   The clinical  symptoms  and  signs  of



exposure recorded  in  the  surveys are consistent with those described in single




case reports  (see Table 9-18) (i.e., neurological  effects and evidence of liver




and kidney dysfunction),  but there appears to be  a  more frequent indication of



cardiovascular insufficiency.  The range of doses that elicited effects in the



surveys  (=12.5 to  250.6  g)  also  appear to  be  consistent  with  the single case



reports (see Table 9-18), but incidences of fatalities were not  presented in the




available summaries.  Although  none  of  the  effects reported  in  the  surveys were



correlated  with  specific doses,  several   of the studies  indicated  that the




severity of poisoning was related to blood levels of  EDC (Bonitenko,  197U,  1977;



Luzhnikov et  al.,  1971*,  1976).



     The clinical  syndrome of oral EDC  poisoning in  children  is similar to that




seen in adults, but the lethal dose range  is somewhat lower (0.3  to 0.9 g/kg)



(Hinkel, 1965).   This range of  doses did not always  cause death, and is derived






                                      9-61

-------
                                                                                TABLE 9-19


                                                   Effects of Acute Oral Ingestlon of 1,2-Dichloroethane (Survey Results)
                 Route      No.  of Subjects
                                                           Dose
                                                                 Principal Findings
                                                                                                                                          Reference
              Oral
                                 121
                20 to 200 ml
                (=25 to 250.6 g)
              Oral
              Inhalation
211
 37
NR
NR x 20 to 30 minutes
a\
              MRa
                               110
                NR
Mild to severe poisoning characterized by
pronounced DCE odor on breath (911),  dry akin
(759), hypotension (71J), extreme pupil dilation
(72*), mucosal cyanosis (67*), tachycardia (62*),
respiratory difficulty (99%)i muscular
hypotonla (46*), decrease in tendon reflexes (46»,
loss of consciousness (42*).  Neurological syn-
dromes noted in 118 aubjecta:  comatose (42J),
atactic (42*), asthenlo with autononio vascular
insufficiency (27*), extrapyranidal (23*),
convulsive (7.If), psychotic (51).  Lethality
not reported.

Aggregate findings for 218 total patients reported.
Neurological disorders (incl. unconsciousness,
respiratory depression) 000*), cardiovascular
insufficiency (incl., arrhythmias, hypotension,
reduced cardiac output, decreased peripheral
resistance) (60», liver dysfunction (inol.,
enlargement, hyperbilirubinemia, increased
serum albumin and asparagine transaminase (35*).
Nephropathology (ollgurla, proteinurla, azotemia,
acute renal failure with disturbed acid/base
balance) particularly associated with inhalation
exposures.  Gastroenteritis milder in subjects
exposed via inhalation.  No correlation between
severity of poisoning and the blood or urine
concentration or the amount inhaled.

Acute gastritis with vomiting (77»,  neurological
disorders including coma (81*), acute cardio-
vascular insufficiency (57$), hepatitis (56<)
with liver enlargement and dysfunction
(abnormal bromoaulfaleln, plasma billrubln,
plasma glutamlne - aparagine transaminase).
Clinical symptoms of poisoning were observed
at blood concentrations of 0.5 mg J,  and coma
developed at 5 to 7 mg J.
                                                                                        Aklmov et  al.,
                                                                                        1976,  19T8
Schepotln and
Bondarenko, 1978
                                                                                        Luzhnikov et al.,
                                                                                        1976, 1978

-------
                                                                             TABLE 9-19 (cont.)
                 Route      No. of Subjects
                                                           Dose
                                                                    Principal Findings
                                                                                                                                           Reference
              NSa
   160B
NA
o
 i
              Oral
              Inhalation
2H
 3
10 to 100 ai (=<12.5 to 125 g)
NR
               Oral
32
                                                 NR
Hemodynamlc shock due to myocardlal dysfunction.
Characterized by compensatory (increased
peripheral resistance, normal or slightly
Increased arterial blood pressure,  decreased
cardiac output and blood volume, decreased
cardiac isometric contraction, increased
ventricular expulsion time, asynchronous
contractions) and deoompensatory (pronounced
and progressive hypotension, decreased cardiac
output, normal or slightly decreased peripheral
resistance, decreased myocardlal contractile
force during ventricular systole, prolonged
periods of asynchronous contractions) phases.
EKG changes including arrhythmias observed in
both compensated and decompensated shock.
Pathologic changes in the myocardium charac-
terized by capillary endothellal cell edema,
capillary lumen stenosis, edema of the
myooardlal Interstices with leukocyte
accumulation and miorofocal hemorrhages,
decreased glycogen, degenerative changes, and
decreased mitoohondrlal enzyme activities.

17 patients exhibited manifestations of
light/mild poisoning:  headache, vertigo,
abdominal pain, nausea, vomiting, and signs of
cardiac Insufficiency (increased contraction
rate, decreased minute volume, decreased
circulating blood volume, decreased cardiac Index);
5 of 17 had hepatomegaly, Icteric skin, solera,
and Increased bilirubin levels.  The other 10
patients were comatose and  had more pronounced
symptoms of cardiac Insufficiency.

Gastroenteritis and protelnurla.  Elevated
leukocyte count and elevated  alanine/aapartate
amlnotransferase activities correlated with
severity of poisoning.  Coma  was associated with
blood concentrations  of 15  to 30 mg %, and
consciousness returned at  levels below 8  to
10 mg ).  Adipose  tissue and  blood levels of
68 and 1.2 mg t, respectively,  at autopsy.
Luzhnlkov et al.,
1576, 1978
                                                                                                                                        Andrlukin, 1979
                                                                                        Bonitenko et al.,
                                                                                        1971, 1977

-------
                                                                            TABLE 9-19  (cent.)
                 Route
No. of Subjects
                                                           Dose
               Principal Findings
                                                                                                                                          Reference
             Oral0
7 children
(1 to 6 years)
Clinical syndrome similar to that In adults:
severe and persistent vomiting (within 1  hour),
and subsequent (Immediate to 10 to 12 hours)
narcotic effects (e.g., somnolence, motor unrest,
reflex Increases, convulsions or coma).  Less
frequent Indications of circulatory failure,
kidney and liver functional disturbances, and
tachycardia.  No remarkable hematologic changes.
Typical pathological anatomical findings.
U>20 reportedly ranged from 0.3 to 0.9 8/kg.
                                                                                                            Hlnkel,  1965
             aRoute  is  probably oral,  but  not  specifically stated  In the  EPA (1979)  summary of this  study.

             bThe EPA summary  of this  study stated  that  "at least" 160 patients were studied.
             °Exposure  to  a  "nerve  balsam" medicine that was 75% 1,2-dlchlorethane  (other components not  stated  in EPA summary).
vO

 en

-------
from a review of seven cases of accidental poisoning in children that ranged in
age from 1 to 6 years.
     Foreign  reports  of individual non-fatal  cases  of EDC ingestion (lenistea
and Mezincesco,  1943;  Bloch,  1916; Stuhlert,  1949; Flowtow,  1952;  Kaira,  1966;
Rohmann  et  al.,  1969; Gikalov et  al.,  1969;  Pavlova et al.,  1965; Agranovich,
1948) were in most  instances not detailed in the NIOSH (1976) or U.S.  EPA (1979)
reviews, presumably because the effects were similar to those that preceded death
with lethal exposures, and because  the quantities ingested were not  known.  Bloch
(1946)  did  note bloody  diarrhea  and  evidence of adverse liver   (enlargement,
slightly increased serum bilirubin, urobilinogen,  abnormal galactose and alcohol
load test  results)  and   kidney  (oliguria,  albumin and   casts  in  the  urine,
temporary retention of nitrogenous substances) effects  in one man who claimed to
have swallowed "only  a small amount" of EDC.
     Death  was usually  ascribed to circulatory  and  respiratory  failure,  and
autopsies  revealed   tissue  congestion,  cellular  degeneration,   necrosis  and
hemorrhagic  lesions  of most organs, including the stomach,  intestines,  liver,
kidneys,  spleen,  heart,  lungs,  respiratory tract and  brain  (see  Table 9-18).
There  was occasionally evidence of gross  bleeding into the  visceral organs or
lungs.   Several  of  the more  recent  reports  (Martin et al.,  1969; Schonborn
et al.,  1970; Yodaiken and Babcock, 1973) indicated that the hemorrhagic effects
may  have been exacerbated  by  a condition  known  as disseminated  intravascular
coagulation  (DIG).  DIG is characterized by a depletion (consumption)  of clotting
factors, hypofibrinogenemia and  thrombocytopenia,  and  the resultant  effect  is  a
severe bleeding  tendency.
     The results of  the  studies  by Luzhnikov  and  coworkers (1976,  1978) suggest
that  EDC can exert a direct  toxic effect on  the  myocardium  (see Table 9-19).
Pathologic  examinations of EDC-exposed  individuals  (amount ingested and number
                                      9-68

-------
examined not available) revealed significant edema in the capillary endothelial



cells and stenosis of the capillary lumens, pronounced edema  of the myocardial



interstices with accumulation of polymorphonuclear leukocytes,  microfocal hemor-



rhages, diminished glycogen content,  and degenerative changes.   Mitochondrial



damage was indicated  by a decrease in  enzyme activities.   Myocardial functional



changes (e.g.,  decreased contractile force, asynchronous contractions) were also



observed prior  to death.  The cardiotoxic  effect of EDC apparently precipitated a



clinical state  of  shock; hemodynamic disorders  commonly observed  in  at  least



160 patients included increased peripheral resistance, decreased cardiac output,



hypotension and EKG changes  including  arrhythmias.




     9.2.1.1.1.2.    Inhalation Exposure.   Reports of  numerous cases  of  acute



occupational exposure to EDC  vapor have been published  (NIOSH,  1976).   These




acute exposures have  involved  both fatal  (27 cases) (Wendel,  1948; Brass,  1949;



Hadengue and Martin,   1953; Ollivier et  al.,  1954; Domenici,  1955;  Salvini  and



Mazzuchelli, 1958;  Guarino and Lioia, 1958; Troisi and Cavallazzi,  1961) and non-




fatal  (57 cases)   outcomes  (Wirtschafter  and  Schwartz,  1939;  Jordi,  1944;



Agronovich,  1918;  Baader,   1950;  Paparopoli and  Cali,  1956;   Menschick,  1957;



Smirnova and Granik,  1970), but none of the reports provided exposure concentra-




tions.  Furthermore,  although EDC was usually reported to be the primary vapor to



which  the  workers were  exposed,   the  preponderance  of  exposures  were  poorly



characterized mixtures of EDC  and  other solvents, or  EDC of unknown  purity.  The



occupational exposures were  generally associated with maintenance and  cleaning



operations or  the  use of EDC  as a paint thinner,  but many (39 cases)  involved



exposure to Granosan, a fumigant composed  of 30% carbon tetrachloride and 70% EDC




(Domenici,  1955?   Salvini and  Mazzucchelli,  1958;  Guarino  and  Lioia,  1958;



Paparopoli and  Cali,  1956).
                                     9-69

-------
     The  effects associated  with  the  above cited  fatal and  non-fatal  acute



inhalation exposures  were very similar  to  those found after ingestion (NIOSH,



1976).  In general, the initial symptoms appeared to be consistent with both CNS



effects  (headache,  dizziness, lethargy,  feelings  of drunkenness,  unconscious-



ness) and gastrointestional disturbances (nausea, vomiting,  diarrhea), which are



characteristic  of chlorinated aliphatic hydrocarbon toxicity.   In  some  cases



workers were asymptomatic and were not overcome during exposure,  but later became



unconscious.  Other signs and symptoms  of inhalation exposure commonly included



cyanosis,  epigastric   tenderness  and/or  pain,   hepatomegaly,  and  jaundice.



Laboratory  studies were consistent with these  observations and  indicative of



hepatic  dysfunction  (increased serum bilirubin  and  urobilinogen, hypoglycemia,



abnormal  function tests)  and  renal disturbances  (oliguria,  anuria,  abnormal



function  tests).   In  addition,  conjunctivitis,  respiratory  tract irritation and



inflammation, rales and leukocytosis were occasionally associated with the acute



vapor exposures.



     Death  from acute inhalation exposure was generally  attributed to respira-



tory and  circulatory failure (NIOSH, 1976),  although  it  is not certain if EDC



exerted  a direct toxic effect  on the cardiopulmonary system or precipitated a



shock-like  state  that could  have  elicited  many  of  the  changes  observed at



autopsy.  Autopsies  frequently revealed pulmonary  edema,  congestion of internal



organs  (liver,   kidneys,  spleen, lungs,  brain)  and  cellular  degeneration and



necrosis  (liver  and  kidneys).   Hyperemia  and hemorrhagic lesions  were  also



observed  in  the kidneys,  brain,  respiratory tract, lungs, and heart.



     9.2.1.1.1.3.   Dermal  Exposure.    Several  reports  described  instances (a



total  of 6  cases)  in which  skin contact with  EDC  occurred concurrently  with



inhalation   exposures  (Wirtschafter   and   Schwartz,   1939;  Anonymous,   1916;



Rosenbaum,  1917; Hadengue and Martin,  1953)*  Severe  dermatitis was a commonly
                                       9-70

-------
reported result of the dermal exposures, as  is  the case with many  chlorinated



hydrocarbons which produce  defatting of the skin.  Several of these reports also



suggest that dermal absorption may  significantly contribute to total  exposure



(characteristic symptoms of intoxication  were noted in workers who were wearing



EDC-soaked clothing).



     9.2.1.1.2.     Experimental  Investigations.



     9.2.1.1.2.1.   Physiologic  Effects.   Borisova (1957) examined  eye sensi-



tivity to light and plethysmographic and spirographic responses in a small number



of subjects  (3 to 4)  who were experimentally exposed  to  concentrations of EDC



that ranged from =1 to 12.4 ppm (4 to 50 mg/m3).   This range  of concentrations



represented subthreshold, threshold and  above threshold odor perception concen-



trations  (Section  7.1.2.2).   The concentration  of EDC  in  air was  determined



nephelometrically; the method is  sensitive  enough (0.001 mg/sample reported) to



monitor EDC at levels lower than  the lowest concentration tested (4 mg/nr).



     In  the  light sensitivity experiment,  baseline perception  thresholds were



determined prior to the exposures for each of 3  subjects  (17  to 24 years old),



and the  subjects  inhaled EDC vapor for  15 minutes  prior  to testing (Borisova,



1957).  When tested after  40 minutes of  dark adaptation,  the  threshold (inten-



sity) at which light was perceived was found to be higher during  exposure to EDC,



and the  threshold appeared to increase  with increasing concentrations of EDC



(Table 9-20).  No change in the light sensitivity of eyes was observed  in any of



the subjects at  1 ppm,  and the lowering of  eye  sensitivity to light  was most



marked at 12.4 ppm.



     The  effect  of low  concentrations  of EDC  on blood volume (vascular con-



striction)  and pulse  fluctuations  was  studied   with  a  finger plethysmograph



(Borisova, 1957).  Four subjects  were exposed to  vapor  concentrations of 1, 1.5,



3,  5.7 and  12.4  ppm  for 30 seconds or  15 minutes.   A 30-second  exposure at
                                      9-71

-------
                                  TABLE 9-20
      Effect of Ethylene Dichloride Exposure on Eye Sensitivity to Light6
Concentration
ppm (mg/nr)
1 (4)
1.5 (6)
2.2 (9)
3 (12)
4.3 (17.5)
5.7 (23.2)
6.2 (25)
7.4 (30)
12.4 (50)
Subject 1
no effect
-0.2
-0.25
-0.25
-0.3
-0.45
-0.45
-0.5
-1.2
Perception Threshold
(Units of Optical Density)
Subject 2
no effect
<0.05
-0.3
-0.25
-0.35
-0.4
-0.5
-0.5
-1.2
Subject 3
no effect
no effect
+0.15
no effect
-0.45
-0.25
-0.7
-0.75
-0.6
a
 Source:  Borisova, 1957

bEye sensitivity to light was determined after 40 minutes of adaptation in the
dark at different EDC concentrations.
                                        9-72

-------
1.5 ppm resulted in a  temporary  vasoconstriction in all 4  subjects.   Complete
data were not reported, but vasoconstriction appeared  to be more pronounced at
3 ppm and higher  concentrations.   Results of tests  in which EDC  exposure was
extended to  15  minutes reportedly showed that the above concentrations caused
"characteristic changes"  in  the  plethysmograms  of  all subjects that  included
increased pulse beat,  pulse waves,  and  pulse amplitude.  The observations at the
higher concentrations  in both the  30-second and  15-rainute  exposure experiments
also suggested  that the  degree  of response was proportional to  the  exposure
concentration.  Exposure  to  concentrations of  1.5,  3, 5.7,  and  12.4  ppm for
1 minute produced changes in depth of  breathing  in  4 subjects (as  indicated by
increased amplitude of respiratory waves on spirograms), but  concentrations of
1 ppm reportedly had no effect on respiration  (Borisova,  1957).
     9.2.1.1.2.2.    Odor  Detection and  Recognition  Thresholds.   Hellman and
Small (1973,  1974} determined an  absolute odor  threshold of 6.0  ppm and an odor
recognition threshold  of  40 ppm for  EDC.   A  trained odor  panel  was  used to
characterize the odor  properties  of this compound and 101 other  petrochemicals,
but the number of panelists was not stated.  The  absolute odor threshold is the
concentration at which 50$ of the odor panel observed an odor, and  the recogni-
tion threshold is the  concentration at which  100$ of the panel defined the odor
as representative of EDC.   Borisova (1957)  determined the odor threshold of EDC
with 20 subjects  and  a total of  1256  tests.   Thirteen subjects perceived EDC
vapor at a concentration of approximately 6 ppm (23.2 to 24.9 mg/m3),  6 subjects
perceived EDC at  4.5 ppm  (17.5 mg/m3),  and  1  subject  detected the compound at
3 ppm (12.2  mg/m3).
     When diluted in water,the odor recognition threshold of EDC  was  determined
to be 29 ppm (v/v water)  (Amoore  and Venstrom,  1966).  An odor panel  of 13 men
and 16 women was  used  for  the determination,  and the  threshold  defined by the
                                     9-73

-------
panelists was the concentration of EDC that yielded 70$ correct response (signi-



ficant at the "\% level) in a paired comparison against pure  water as a control.



     9.2.1.2.   REPEATED  AND CHRONIC EXPOSURES.



     9*2.1.2.1.      Case Reports  — Repeated  exposure to EDC vapor in the work-



place has resulted in effects that are consistent with those  resulting  from acute




exposures.   Symptoms and  signs  including anorexia, nausea, vomiting, weakness



and  fatigue,  nervousness,  epigastric  pain/discomfort and  irritation of  the



respiratory  tract and eyes  were described in numerous case reports of  industrial



exposures  (McNally  and Fostvedt,  194"i Siegel, 1947;  Rosenbaum,  1939; tfatrous,



1917; Rejsek and Rejskova, 1947;  Delplace et al.,  1962; Suveev and Babichenko,



1969).  In one study (Suveev and Babichenko, 1969), examination of 12 symptomatic



workers who  were brought to a clinic revealed paleness and cold sweat (12 of 12),



bradycardia  (9 of 12), systolic murmur (5 of  12),  diarrhea (5 of 12;  3  of 12 with



blood) and  enlarged livers that were soft and  tender to palpitation  (9 of 12);



muffled  heart  sounds,  increased  rate of  respiration,  rales,  coated and  dry




tongues were also observed but incidences of occurrence were not stated.  Signs



indicative  of nervous system  dysfunction have also  been reported  in cases of



chronically  exposed workers;  these include  nystagmus, fine tongue tremors and



sluggish  patellar  reflex  (MoNally  and Fostvedt,  1941),  encephalic disorders



(Delplace et al., 1962),  and decreased  muscle tone, loss of  reflexes,  a positive



Romberg's sign,  and deafness (Suveev and Babichenko,  1969).  Complaints of hand




and arm eczema that appeared within the first year of exposure were recorded in



11  of  16 cases  by  Delplace et al.  (1962).   It  should  be  noted that exposure



concentrations and  information on the types of exposures were not provided  in any




of  the above cited  reports.



     Rosenbaum (1947) discussed  experiences with cases of  EDC intoxication in



Russian industries  from  1931 to 1945.  He observed over a period of 10  years that
                                      9-74

-------
characteristic symptoms of acute poisoning could develop with repeated exposure



to concentrations of 75 to  125 ppm in air.  Fatalities have resulted when workers



experienced these symptoms  2 or more times in a period of 2 to  3 weeks.  There was



no mention, however, of the number of people affected or durations of exposure.



     Byers (1943) noted that a "number of persons" exposed to EDC complained of



delayed toxic effects.  The workers stated that the worst effects, which varied



from lassitude and malaise to nausea, vomiting and abdominal pain, occurred after



the evening meal. These symptoms were associated with exposure to concentrations



"only slightly in excess of 100 ppm for 7.5 hours daily", and were not completely



alleviated when ventilation procedures reduced  the EDC concentration to 70 ppm.



     Exposure conditions were  described in partial detail for 2 workers who were



chronically exposed  to  EDC for  7 and  9 months  during the manufacture of hexa-



chlorophene (Guerdjikoff, 1955).  CDC  was used as a  catalyst in  this process.



During one of the operations, which was repeated for 2 to 3 minutes several times



a day for  a total of about 30 minutes, exposures  were associated with adding EDC,




trichlorophenol and sulfuric acid to the reaction vat.  EDC  exposure concentra-



tions were not measured during the filling operations, but the  workers wore an




air-supplied respirator and were probably only exposed to EDC occasionally due to



improper fit.  During another operation which lasted 10 minutes and was repeated



3 to 4 times a day, the exposure concentration of EDC was about 120  ppra.  During a



final operation, the workers were exposed once  a day for =10  to  15 minutes when



an EDC pipe was cleaned; these exposure concentrations were not measured but were



considered by Guerdjikoff (1955) to be more than  120 ppm.  Both workers exhibited




similar symptoms,   typical  of EDC  exposures,  of  anorexia,  epigastric  pain,



fatigue, irritability, and  nervousness after about 3 weeks  of exposure.  Neuro-



logical effects  such  as  hand  tremors,  hyperhidrosis  and difficulty  in talking



were eventually experienced.
                                     9-75

-------
     9.2.1.2.2.     Health  Surveys —  Kozik  (1957)  reported the results  of  a




health and morbidity  survey of Russian aircraft industry workers.   All  of the



workers in the study group  (size  not stated) were employed in a shop where glue



that contained EDC as  a solvent was used to bond rubber sheets to metal forms in a



soft tank  fabrication process.   Most of these workers were gluers,  but a small



number worked inside the  completed tanks to disassemble  the  forms.  EDC was




emitted to the air during application  and  glue drying.



     About 500 atmospheric  measurements of EDC were  reported to have been taken




(Kozik, 1957).  Although the sampling and analytical methods  were not mentioned,



NIOSH  (1976)  felt that the data  were  presented  in  sufficient  detail to  permit




estimations  of TWA  exposures.    NIOSH estimated that  M  to 16$ of the total



exposure occurred during the gluing operations,  when the TWA  concentrations were



s28  ppm  during application  and  =1 6 ppm when the glue  was  drying.   When other



operations were performed  in the same  shop (during  the  second half of the work-



shift), the EDC TWA concentrations were ^11 ppm.  The TWA for the total shift was



estimated  to  be  15 ppm.   Concentrations ranged  from approximately U to  50 ppm;



concentrations in excess  of 20  ppm were associated only with  the  gluing and



drying operations and occurred about 15$ of the time.   NIOSH (1976) has noted,



however,  that the aforementioned TWA  concentrations may be an overestimate of



most of  the workers' exposures  for  several  reasons.  First, the tabulation of



measurements  in  the  glue application category also contained high values (45 to



 52 ppm)  that were experienced  only by an insignificant number  of workers who




disassembled  the molds within the finished tanks.  Second, the  measurements were



apparently not breathing zone measurements and third, the ventilation system was



designed  with the exhaust ducts on the floor.  In light of these  considerations,



 NIOSH concluded  that a more realistic  appraisal  of the TWA exposure  of  the




majority of the  workers is 10 to  15 ppm.
                                       9-76

-------
     Workers (total  number not stated) who were engaged  in the production of soft



tanks during the years  195"  to 1955  experienced  increased  morbidity and  los1:



workdays when compared  *rith  workers  in the entire factory (Table o_2i).   Disease



categories  examined  included acute  gastrointestinal  disorders,  neuritis  and



radiculitis, and other  diseases.  An in-depth analysis of the morbidity rate with



temporary disability for 195^ to 1955 reportedly showed high  rates  for  gastro-



intestinal diseases, liver and gall bladder diseases,  and diseases of the muscle,



tendons and  ganglion (Table 9-21).   The liver  and  gall bladder diseases  were



considered by Kozik (1957)  to be related to a specific  toxic effect  of  EDC (the



dyspeptic symptoms  it causes reportedly are  often diagnosed  as gastritis),  but



the diseases of the muscle,  tendon and ganglia were associated with the  numerous



repetitive  motions  the workers had to  make when applying  the glue.   Farther



examination of 83 of the gluers revealed diseases of the liver and bile ducts (19



of 83), neurotic conditions (13 of 83),  autonomic. dystonia (n of 83),  asthenia



conditions  (5 of 83), and goiter and hyperthyroidism (10 of 33).



     Visual-motor reactions were studied at  the  beginning and end of 11  workdays



in 1 7 of the gluers  and 10 control machinists (Kozik, 1957).  It was stated that a



device wag used to determine simple  and complex  reaction (color differentiation)



times, as well as reaction times in  a  modification of the complex reaction task;



a total of 3700 reaction tests were  conducted, but details of the tests were not



given.  A comparison of the mean rates for all three reactions show no signifi-



cant differences in the two groups either before or after work.  Nervous system



dysfunction  was suggested, however, by the inadequately reported results of the



complicated  reaction tests.   "Most" of the  gluers  made errors  in  the complex



reaction task, while the machinists did  not make any  errors  (additional data are



not  available).   In the modifier! complex reaction  test,  errors were committed
                                      9-77

-------
                                                                                TABLE  9-21
o
i
                                           Morbidity and Lost Workdays of Aircraft Industry Gluers  Exposed  to Ethylene Diohloride

                                                                             (Rates/100 Workers)
Year

1951
1952
1953
1951
1955
Total
Morbidity

Cases
Days
Cases
Days
Cases
Days
Cases
Days
Cases
Days
Plant
120.2
995.8
121.0
960.9
135.6
101(0.8
150.7
"75.9
127.6
978.1
Shop
159.8
1115.5
137.6
996.0
163.9
1236.5
191.8
1563.2
176.6
1162.1
Acute Gastro-
intestinal
Disorders
Plant
5.
19.
1.
15.
11.
15.
5.
19.
1
3
2
1
4
6
3
3
3.6
12.1
Shop
11.6
13.5
5.7
23.1
6.2
19.1
9.6
31.8
5.0
15.3
Neuritis
and
Radiculltis
Plant
5.2
59.9
5.0
11.8
7.5
67.3
7.9
73.8
5.9
51.1
Shop
13.0
127.0
9.7
91.5
16.5
116.0
16.7
182.8
10.3
90.2
Diseases of
the Muscles,
Other Liver and Call Acute Chronic Tendons,
Diseases Bladder Diseases Gastritis Gastritis and Ganglia
Plant
31.1
351.2
31.0
335.2
35.3
338.3
10.8
386.4
37.9
315.7
Shop
13.2
511.8
10.8
378.7
53.5
521.0
63.8 21
596.2 251.5
63.3 21
640.5 290

.
.
.
13 6 US
64.5 45 170
8 3 8
27 14 50
               ^Source:  Kozlk, 1957
                The Incidences are reportedly elevated,  but reference values were not given.

-------
both before and after work by 15 of 17 gluers; 4 of 10 machinists made errors, but



only at the  end of the workday.



     Cetnarowicz (1959) investigated the possibility of EDC poisoning in Polish



oil refinery workers.   The workers were employed  in  a  mineral  oil purification



(dewaxing) process that involved mixing oils  with a  solvent that contained 80$



EDC and 20%  benzene at 40°C; the mixture was  subsequently  cooled to -24°C, and



precipitated paraffin  was  separated by centrifugation.  Durations  of exposure



were not stated, but concentrations  of CDC ranged  from =10  to  200 ppm (0.04 to



0.8 mg/Jt),  with the  highest  excursions in  the centrifuge room (Table 9-22).



Benzene concentrations ranged from =3.1 to 7.8 ppm (0.01 to 0.025 mg/O, but were



not specifically reported for the different  work areas detailed in Table 9-22.



Nineteen workers (18 to 48 years old;  18 men, 1 woman) who had been employed for 2



to 8 months were  included in the initial  study group; 2 men and the woman were not



examined due  to medical history complications.  The results of these examinations



are summarized in Table 9-23 and  in  the text below.



     Ten  of  the 16 workers  were employed  in the centrifuge  room,  where EDC



concentrations ranged  from 62 to  200 ppm.   All 10  of these workers complained of



a burning sensation in the eyes  and  lacrimation, and 6 of  the 10 workers stated



that they had experienced dryness of the mouth, an unpleasant sweet aftertaste,



dizziness, fatigue, drowsiness,  nausea, occasional vomiting,  constipation, and



loss of appetite.  Three of these workers also complained of pain in the epigas-



trum.   All  of the above-mentioned  subjective complaints  disappeared  when the



workers changed  workplaces, but  reappeared upon reexposure.   Of the 6 workers



employed  in  the pump  and recrystallization rooms  (10   to   37  ppm),  only  1



complained of similar symptoms.   As  detailed in Table 9-23, clinical evaluation



of  the  16 workers indicated  the likelihood  of  liver  effects  (tenderness to



palpitation   with minimal enlargement,  epigastric pain, elevated urobilinogen
                                     9-79

-------
                            TABLE 9-22
      Concentrations of Ethylene Dichloride in Oil Refinery
              Mineral Oil Purification Process Air*
Location
Centrifuge room
Pump room 1
Pump room 2
Pump room 2
Crystallization room
(repeated
6U
16
25
30
30
ppm
measurements)
62
10
13
37
37

200
17
-
-
-
•Source:  Derived from Cetnarowicz,  1959 by NIOSH, 1976.
                                9-80

-------
                                                        TABLE 9-23

                                     Effects Observed in Polish Oil Refinery Workers*
      Concentration of  1,2-DCE
                      Effect
      62, 61 and 200 ppm
f     10 to 37 ppm
Co
      10 to 200 ppm
Dryness of the mouth; unpleasant sweet aftertaste;
  dizziness; lassitude; sleepiness; nausea;
  vomiting; poor appetite
Burning sensation of the eyes that disappeared with
  adaptation to the atmosphere
Epigastrium pain
Insignificant tenderness of the epigastrium
Livers tender to palpitation with minimal enlargement
Normal arterial blood pressure
Relaxed pulse (60 to 65/minutes)
Intensified reflexes and automic neuroses

Complaints similar to those associated with
  exposure to 62, 61 and 200 ppra

Sweet aftertaste; dizziness; nausea; vomiting; lack
  of appetite
Livers tender to palpitation with insignificant
  enlargement
Epigastrium pain
Emaciation (2 to 10 kg below expected weight)
Unremarkable opthalmologic examination
Unremarkable examination of upper respiratory tract,
  lungs and heart
Augmented reflexes and vegetative neurosis
Incidence
6/104
                                                                                                    10/10
3/10b
7/10°
4/10°.
10/10
6/10°
3/10b

1/6°
                                                                                                    3/42
                                                                                                    16/16
                                                                                                    16/16
                                                                                                    16/16

                                                                                                    3/16

-------
                                                        TABLE 9-23 (cont.)
           Concentration  of l,2-DCEa
          10  to  200  ppm  (cont.)
vO
                      Effect
Incidence
Icteric skin coloration                                      1/16
Elevated urobilinogen levels                                 "majority"
                                                               of 16
X-ray observable chronic catarrh of the stomach with         6/16
  atrophy of the mucous membrane
Periodic spasm of the pylorus                                3/16
Moderate hyperchromic anemia                                 1 /1 3e
  (3,430,000 erythrocytes/cu mm and 60H hemoglogin)
Slight reticulocytosis (0.1  to 0.3*)                         1/136
Diminished osmotic fragility of erythrocytes in NaCl         6/13
Slight leukocytes (11,200/cu ram)                             1/136
Low platelet count (10,000 to 55,000 cu mm)                  2/1 3e
Decreased number of neutrophils (10 to 5058)                  2/1 3e
Slightly increased number of neutrophils (70 to 78$)         6/13e
  with decreased number of lymphocytes (15 to 25%)
"Abnormal" distribution of white blood cells                 1/1 3e
Increased number of erythrocytes,  increased percentage       5/1 3e
  of polymorphonuclear neutrophils, mild stimulation
  of erythropoiesis with a less significant increase
  of leukopoiesis in the bone marrow
Elevated serum bilirubin (2.3 »g %)                          1/16

-------
                                                       TABLE 9-23 (cont.)
U)
          Concentration of 1,2-DCEE
          10 to 200 ppm (cont.)
                      Effect
Elevated blood nonprotein nitrogen (55 mg %)
Normal overall quantity of serum protein
Diminished serum albumin
Elevated serum globulin levels
Diminished blood fibrin content
Positive Takata-Ara liver function test
Borderline Takata-Ara liver function test
Positive Cadmium turbidity teat
Borderline Cadmium turbidity test
Negative glucose tolerance test
Decreased secretion of gastric hydrochloric acid
X-ray observable catarrhal changes of gastric mucosa
Periodic pyloric spasms
Incidence
1/16
16/16
6/16
8/16
3/16
1/16
5/16
6/16
5/16
8/16
12/16
6/16
3/6f
          Source:  Cetnarowicz, 1959 (derived from NIOSH, 1976)

          Concurrent exposure to =3 to 8$ benzene (see text)
           Workers employed in the centrifuge room (see Table 9-22)

          °Workers employed in the pump and crystallization rooms (see Table 9-22)
           12 workers initially examined; effects on these workers prompted further examinations of 16 workers from
           one shift (see below).

           Heraatological examinations were performed on 13 workers.

          f3 of the 6 with catarrh

-------
levels,  positive Takata-Ara  liver function  tests,  negative glucose tolerance



tests),  and changes  in the  gastrointestinal tract  (X-ray  observable  chronic



gastritis with mucosal  inflammation,  pyloric  spasms,  impaired hydrochloric acid



secretion).   The  distribution of  these findings by  room  (i.e.,  high  or low



exposure) was  not,  however,  given.  Other abnormal findings included blood and



bone  marrow changes  that  may  be attributable  to  the concurrent  exposure  to



benzene  (Table 9-23).



     Rozenbaum  09t7)  reported  the  results  of studies of  approximately 100



Russian  workers  from different industries  (not  specified)  who  had experienced



exposure  to EDC  at  levels  below  25 ppm (<0.1   ing/1)  in air  for  6 months  to



5 years.   "Many"  of these workers exhibited non-specific  functional  nervous



system    disorders   (e.g.,  >*ed  dermographisra,   muscular  torus,  bradycardia,



increased  perspiration),  and  "some"  of the  subjects complained  of fatigue,



irritability,  headaches and  insomnia.  Hematologic or functional organ changes



(not  elaborated) were  not,   however,  noted.   The  incidence  of  the observed



effects, average  exposure  concentrations, the range of exposure concentrations



to which the workers were exposed  and information on experimental design were not



reported.



     Brzozowski et al.  O951*) considered that absorption of  EDC  through the skin



was largely  responsible for adverse  health  effects  among certain agricultural



workers  in  Poland.   EDC  was  used as  a fumigant and  work practices involved



transport of the liquid to  fields  in barrels,   pouring  the EDC by  hand  into



buckets, carrying  the open buckets to the site of application,  and pouring the



EDC into a series of  holes in  the ground.  The workers were exposed to particu-



larly  high  concentrations of  EDC  vapor during  pouring.   Significant  dermal



exposure is indicted by  the frequent spilling of quantities of EDC on clothes and



sleeves during the  transport of  the  open buckets to the places of application
                                      9-84

-------
(resulting in soaked clothing that was  not  changed),  and by the  fact that the



workers used EDC to wash their skin.  A  single atmospheric sample, comprising 10



subsaraples collected  from locations representative  of  the  working  zone (the



collection apparatus was moved from place to place with  the workers), showed a



concentration of 4 ppm of EDC.   Because of the  practical  difficulties of sample



collection (the workers did not stay at one location sufficiently long  to collect



an  entire sample),  conditions  were  simulated  in  a laboratory to estimate



potential exposure concentrations better.  Analysis of laboratory samples showed



concentration of =14.5 to 15  ppra; a sample taken during the pouring of EDC into



buckets, considered to be the maximum exposure  of a worker,  contained 60 ppm.




     Medical examinations were  performed on  118 of  the agricultural  workers



(Brzozowski et al., 1954). Ninety had  positive findings  that included conjunc-



tival congestion (82/118), weakness (54/118), reddening of the pharynx (50/118),



bronchial  symptoms (43/118), metallic  taste in  the  mouth  (40/118),  headache



(39/118), dermatographism (37/118),  nausea (31/118),  cough (30/118),  liver pain




(29/118),  conjunctival  burning  sensation  (24/118),  tachycardia  (21/118),  and



dyspnea after effort  (21/118).   The  Quick Test  for hippuric acid was  used  to




evaluate liver function and was reported to be  positive  in  40  of 56  investiga-



tions.  Skin sensitization tests with pieces of gauze soaked with 0.1%  EDC  in




alcohol or 50%  EDC in soybean oil were reportedly negative when  scored after



40 hours of contact,  but the  number  of  workers  tested  was not stated.
                                     9-85

-------
9.2.2.  Effects  in Animals



     9.2.2.1.  ACUTE  EXPOSURE



     9.2.2.1.1.     Exposure  — The observed  toxic  effects  of ingested EDC in



laboratory animals  summarized in Table 9-21  tend  to confirm the findings from



cases involving  human oral  exposures.   The LD   for  a single oral exposure was



reported  to  be 680  mg/kg in  a study that employed 80 rats and 4 dosage levels



(McCollister et  al.,  1956) and 770 mg/kg in a range-finding study of rats (Smyth



et al., 1969).  These data suggest that EDC  is moderately  hazardous by the oral



route, and indicate  that the compound is more acutely toxic than 1,1-dichloro-



ethane,  1,1,1-trichloroethane,  pentachloroethane,  and hexaehloroethane (Patty,




1981).  According to a World Health Organization report (1970; cited by Torkelson



and Rowe, 1981) rabbits and mice are as susceptible  as rats to EDC. Heppel et al.



(1945) observed  60J mortality (6 of 10 treated) in mice 10 days after an acute



oral dose of 700 mg EDC/kg body weight, which was  administered as a 5% solution



in olive  oil.    Similar treatment with 900 mg/kg  EDC in  5%  olive  oil produced



mortality  in  10/10  mice, but  6/6 survived  a dose  of 500  mg/kg  given  as  a



10 percent solution in  olive  oil.  Munson et al.  (1982) recently determined that



the single oral dose LD_ 's of EDC for male and female CD-1 mice were 189 and 413
                       3U


mg/kg, respectively.  In these studies, EDC was apparently administered in water



solution, and all mice died over a 46-hour period.  Gross pathologic examination



indicated that the  lungs and liver appeared to be the target organs.  Dogs and



humans  may  tolerate  larger  doses.    Since  EDC  causes  vomiting,  the  higher



tolerance in dogs and  humans is likely attributed to the ability  to vomit in



these two species. Ristler and Luckhardt (1929) found that oral doses >500 mg/kg



tended to be vomited  by dogs.



     In 2 unanesthetized dogs (weighing 9  and 12.5  kg)  administration of 0.63



g/kg  EDC  by gavage  produced initial  excitement  (8 minutes  after  treatment)
                                      9-86

-------
                                                                             TABLE 9-21

                                                          Effects of Acute Exposure to Ethylene Dlchloride
Route
Oral
Oral
Oral
Oral
Oral
Species
rata
rats
alee
alee
(CD-I, male)
nice
(CD-I, female)
Number
BO
6/group
10
10
6
MS
NS
Doae
0.68 g/kg
0.77 g/kg
0.90 g/kg (in olive oil)
0.70 g/kg (In olive oil)
0.50 g/kg
0.489 g/kg (In water)
0.413 g/kg
Effect
LD50
"So
10/10 dead
6/10 dead
no deaths
LD50
LD50
Reference
McCol lister et
Smyth et al.,
Heppel et al.,
Nunson et al.,
Hunson et al . ,
al., 1956
1969
19»5
1982
1982
oo
         Inhalation
         Inhalation
rata
rats
(female)
6 male or female  1000 ppm for 4 h
Sherman Strain
(100-150 weight)


4-6/group         12,000 ppm for 0.1 h
                                 0.2 h
                    3000 ppm for 0.3 h
                                 0.5 h
                    1000 ppm for 1.5 h
                                 3.0 h
                     300 ppm for 3*0 h
                                 5.5 h
                     200 ppa for 7.0 h
2/6, 3/6 or 4/6 dead In
11 day observation period.
Ho adverae effecta
Adverse effect
Mo adverae effect
Adverae effect
No adverse effect
Adverse effect
No adverse effect
Adverse effect
No adverae effect
Carpenter et al., 1919
Spencer et al., 1951

-------
                                                                         TABLE 9-24  (cent.)
 i
0.1
co
Route Spec lea Number
Inhalation rats 20
22
40
DO
51
44
32
44
22
10
30
10
41
24
10
32
31
32
30
20
33
20
20
Inhalation rats 20
15
15
12
13
mice 22
20
23
guinea pigs 14
12
rabbits 16
cats 3
hogs 2
raccoons 2
Dose
3000 ppm for 6 h
3000 ppm for 5 h
3000 ppm for 4 h
3000 ppn for 3 h
3000 ppn for 2 h
3000 ppm for 1.6 h
3000 ppn for 1 .0 h
3000 ppn for 0.7 h
3000 ppm Tor 0.5 h
1 500 ppm for 8 h
1 500 ppm for 7 h
i 500 ppm for 6 h
1 500 ppm for 4 h
i 500 ppm for 3 h
1 500 ppm for 2 h
1000 ppm for 8 h
l 000 ppn for 7 h
1000 ppn for 6 h
800 ppn for 7 h
600 ppn for 8 h
600 ppn for 7 h
600 ppm for 5 h
300 ppn for 7 h
3000 ppm for 7 h
3000 ppn for 3>5 h
3000 ppn for 1.5 h
1 500 ppn for 7 h
1 500 ppm for 4 h
3000 ppn for 7 h
i 500 ppm for 7 h
1500 ppm for 2 h
3000 ppn for 7 h
i 500 ppn for 7 h
3000 ppm for 7 h
3000 ppn for 7 h
3000 ppm for 7 h
3000 ppn for 7 h
Effect Reference
20/20 dead Spencer et al., 1951
22/22 dead
38/40 dead
24/40 dead
6/51 dead
1/44 dead
1/32 dead
1/44 dead
0/22 dead
7/10 dead
24/30 dead
7/10 dead
2/41 dead
1/24 dead
0/10 dead
20/32 dead
17/31 dead
5/32 dead
10/30 dead
4/20 dead
3/33 dead
0/20 dead
0/20 dead
20/20 dead Heppel et al., 1945
1 5/1 5 dead
0/1 5 dead
4/20 dead
0/13 dead
22/22 dead
20/20 dead
1/23 dead
14/14 dead
6/12 dead
12/16 dead
0/3 dead
2/2 dead
0/2 dead

-------
                                                                          TABLE 9-2* (Cont.)
              Route
 Species
                                             Number
           Intraperitloneal  guinea pig    I/group
                              (male)
           Subcutaneous
dogs          6

oats          NS

rabbits       NS

albino rats   NS
vo
A,
VO
           Subcutaneous
           Subcutaneous
                             rats
mice
10/group
4/group at
the lowest dose

10/group
                                                                       Dose
                                150, 300 and 600 ng/kg
                                1  dose
                  0.94 or 1.26 g/kg body weight

                  0.94 or 1.26 g/kg body weight

                  0.9» or i .26 g/kg body weight

                  0.91 or 1.26 g/kg body weight
                                0.31, 0.5, 0.75, 1.0,
                                 1.25 g/kg (In olive oil)
0.25, 0.3U, 0.75 g/kg
 (In olive oil)
                                                                                                      Effect
                                                                                                                                    Reference
No effect on liver histology
 2*t hours after Injection.
No effect on serum ornlthlne
 carbamyl transferase (OCT)
 Activity 24 hours later at
 1 50 or- 300 rag/kg.
Increased OCT activity at
 GOO rag/kg; death of 1 animal.

Death within 21 hours at
 higher dose; histology:  mild
 perllobular fatty degeneration
 in liver, swelling of tubular
 cells and mild hemorrhage In
 the kidney, lung edema hls-
 tological changes more marked
 in dogs.
In dogs only: corneal opacity
 evident by 1 0 hours; necrosis
 of corneal endothelluo; corneas
 became clear by fifth day.

0.75/kg:  50J mortality;
0.38 g/kkg:  no deaths
                                                                                    Divlncenzo and Krasavage,
                                                                  Kuewabara et al., 1968
0.38 g/kg:
0.25 g/kg:
8Of mortality
no deaths
                                                                  Heppel  et al.,  1945
                                                                                                                               Heppel  et  al.,  1945
           h = hours

-------
followed by decreased excitement.  Progressive incoordination and salivation was
evident.  In 24 minutes, sleep (from which the dogs could  be aroused) supervened
for another 20 minutes,  at which  time depression was relieved.  Vomiting occurred
subsequently, after which signs of recovery were evident  (Kistler and Luckhardt,
1929).  Larger doses of 1.26 g/kg in 2 dogs,  1.89 g/kg in  one dog, and 2.51 g/kg
in- two  dogs produced similar symptoms of greater  severity.   Vomiting occurred
earlier after administration of  the  compound;  little food or water was ingested
and vomiting  persisted  in  one dog that received 1.26 g/kg EDO.  The corneas of
the eyes in all five animals were  opaque  within  24 hours.
     9.2.2.1.2.     Dermal  Exposure — Skin absorption  LD Q  values of 4.9 g/kg
(Torkelson  and  Rowe,  1981) and  2.8  g/kg  (Torkelson and  Rowe,  1981)  have been
determined with rabbits.  Strong erythema, edema, or slight necrosis was produced
upon application of 0.01 mil of undiluted EDO to the  uncovered rabbit belly (Smyth
et al., 1969).
     9.2.2.1.3*     Inhalation   Exposure  —  Sayers  et  al.   (1930,  cited  in
Browning,  1965  and NIOSH,  1976) reported  that  death  occurred  in  guinea pigs
within  a  few minutes of exposure  to 100,000 ppm EDC,  and on the day following
exposure to  10,000  ppm  EDC for  25  minutes.   Progressive signs of intoxication
were  observed in  groups  of  3  or  6 guinea pigs  that  were  exposed to  EDC at
concentrations ranging  from 600  to 60,000 ppm for durations up to 8 hours.  One-
third of each group was sacrificed immediately after exposure, another one-third
sacrificed after 4 days  of  exposure and the final one-third was sacrificed at the
end  of the  eighth post-exposure  day unless  death supervened.    The  signs of
intoxication  (squinting and lacrimation  of  the  eyes  and rubbing  of  the nose,
vertigo, static and motor ataxia, retching movements, unconsciousness, incoordi-
nation of extremities,  and  marked  changes in  respiration) appeared in less than
10 minutes, and death occurred in  30 minutes,  after exposure to 60,000 ppm EDC.
                                      9-90

-------
Exposure to  a lower  concentration  (10,000 ppm) for  15  to  20 minutes produced




these symptoms in 25 minutes and delayed death a day or more after exposure.  No



mortality or signs of intoxication occurred  in  guinea pigs  exposed  to 1200 ppra



for 8 hours.



     Congestion and edema of the lungs and generalized passive congestion of the



visceral organs were  observed  in animals that  died  during  exposure (Sayers et



al.,  1930,  cited  in  NIOSH,  1976).   In  animals sacrificed  immediately after



exposure, congestion  of the  liver,  spleen,  lungs,  and  kidneys  was observed.



Pulmonary congestion, pulmonary edema and  renal  hyperemia  were  seen  in those



animals that died  1 to 8 days after exposure,  and the renal and lung effects were




more pronounced in the animals  sacrificed 3 to 4 days post-exposure than in those



sacrificed immediately after exposure.   Partial resolution  of visceral effects



was observed  by  8 days after exposure.   In  these studies, the  severity of



pathological changes was dependent on duration and concentation of exposure.



     Heppel et al. (1945) reported that  a single exposure to 3000 ppm EDO (12.4



mg/i)  for 7 hours produced death in all of 14 treated guinea pigs within 3 days of




exposure.  The guinea pigs  were  inactive and breathing was  labored after expo-



sure.    Microscopic examination of  11 guinea pigs  revealed congestion  in  the



liver, lung and adrenal glands.   Focal necrosis of the adrenal cortex in 5 of the



guinea pigs, and slight  to moderate  fatty degeneration of renal tubular epithe-



lium was noted in 8 of the animals.   In another  group of 8 guinea pigs similarly



exposed to 3000 ppm EDC,  fatty infiltration of the myocardium was observed in all




of 7 animals that were examined histologically.  Changes  in the lung, kidney and



adrenal cortex, similar to those observed in  the  first group of animals were also



found.




     Keppel et al. (1945)  also found that  inhalation exposure to 3000 ppm EDC



(12.4  mg/£) for 7  hours  was fatal to rats,  mice  and rabbits within  3  days of
                                     9-91

-------
exposure.   Varying  degrees  of  narcosis  were observed  in the  animals during



exposure, and dyspnea and increasing weakness preceded death.  Pulmonary conges-



tion, mild  to moderate  degeneration of renal tubular epithelium and liver, and




occasional  necrosis  of the adrenal cortex were observed at autopsy;  congestion of



the  spleen  was  additionally  found  in  rats.   Inhalation of 3000 ppm  EDC  for 7




hours was fatal for 2 hogs but was not  fatal  for  2 raccoons or 3 cats.  Reduction



of dose by exposure to one-half the  concentration of EDC (1500  ppm)  for a similar



duration (7 hours) was lethal to 6 of 12 guinea pigs  and to less than one-fifth of



the  rats (4 of 20).  This lower dose was fatal, however,  to all of  a group of 20




mice.   Reduction  of the duration  of  exposure  in rats  to  4  hours and mice to




2 hours decreased  mortality  (Heppel et al.,  19*14).



     Spencer  et al.  (1951)  subjected  groups of 4  to 6  female rats to varying



concentrations  of  EDC (200, 300,  1000,  3000 or 12,000 ppm)  for varying durations



to determine  the  doses and durations  of  single  exposures that produced and did



not  produce adverse effects.   No-adverse effect exposures ranged from 200 ppm




(0.8 mg/i)  for  7  hours  to 12,000 ppra  (48.6 rag/i) for 0.1 hours.  Doubling the



duration of exposure at 12,000 ppm produced adverse  effects.   Exposure to 3000



ppm  EDC for 0.3 hours was without effect, but increasing the duration of  exposure



to 0.5 hours caused  adverse  effects.



     Spencer et al.  (1951) also sacrificed groups of  10-54 rats after inhalation



exposure to EDC at concentrations ranging from 300 to 20,000 ppra for durations of



8.0  to 0.1  hours.   A  decrease  in body weight, increase  in  liver and kidney



weight,  increase  of  blood  urine   nitrogen,  increase  of plasma prothrorabin



clotting time,  decrease in serum phosphatase, increase in liver lipids  and histo-



logical  changes in  the kidneys,  liver and adrenal  glands  were  observed.   The



kidney changes  consisted of tubular damage  that ranged  from slight parenchyma-



tous degeneration  of  the  epithelium   to  complete  necrosis   accompanied  by
                                      9-92

-------
interstitial  edema,  congestion and  hemorrhage.    Changes  ranging  from slight



congestion and slight parenchymatous  degeneration to marked hemorrhagic necrosis



were observed  in  the liver.   Some  parenchymatous degeneration of  the adrenal



cortex was found in the most severely affected animals.  Pulmonary congestion and



edema was observed at concentrations  above  3000 ppm.  Carpenter  et al. (1949)



reported an inhalation LC5_  in  rats to be 1000 ppm in air for  a U-hour exposure.



     9.2.2.1.3.1    Central Nervous System  and  Cardiovascular System.   Inhala-



tion of 3000  ppm  EDC  (12.4  mg/Jl)  for 7  hours in  five  species (rabbits, guinea



pigs, hogs, rats and mice) produced varying degrees of narcosis (Heppel et al.,



1945).  Spencer et al. (1951) partially attributed the inactivity or stupor and a



slower response  to handling in rats, at  vapor concentrations of 3000 ppm and



lower, to toxic injury to organs  other  than depression of the central nervous



system.  At concentrations of 12,000  ppm and lower, varying degrees of "drunken-



ness" were attributed to  depression of the central nervous system.   Considerable



depression of the central nervous system was produced at 22,000 ppm causing death



in rats within O.U hours.  Animals  that died  from  exposure to 20,000 ppm EDC were



in a  state of  deep  anesthesia during the  period of  exposure.   At all  vapor



concentrations a  large proportion of  the  rats died suddenly  after  leaving the



exposure chamber.   Marked cyanosis, reduced body temperature, stupor or coma, and



failing respiration were  observed.  Spencer et al. (1951) hypothesized that this



response is suggestive of cardiovascular collapse.  Other deaths, which occurred



over a period of two to seven days,  were accompanied by loss  in body weight and



other signs of toxicity;  the authors  attribute these deaths to renal injury.



     Inhalation of EDC  (0.1  to 5 cc, introduced by volatilization into a Jackson



Carbon Dioxide Absorption Apparatus)  produced  a drop in blood pressure, depres-



sion of the knee  jerk reflex and  stimulation  followed  by a short cessation  of



respiration in sodium barbital  anesthetized dogs (number unspecified)  (Kistler
                                     9-93

-------
and Luckhardt,  1929).   These dogs had been subjected  to  spinal cord transection
prior  to EDC treatment.  Similar effects on blood pressure and the knee jerk were
observed  in sodium barbital  anesthetized dogs  (11-20  kg)  after  intravenous
injection  of amounts  as small  as O.i  cc, while 0.3 cc caused  a cessation of
respiration  for a duration  of up to 40 seconds.
     9.2.2.1.3.2.    Eyes.   Inhalation of EDC  was observed to  produce a clouding
of  the cornea  in dogs and foxes,  but  not  in  rabbits,  guinea  pigs,  mice,  rats,
hogs,  raccoons,  chickens or  cats (Heppel et al.,  1944).  A single exposure to a
vapor  concentration of 1000 ppm for  7  hours  produced  turbidity  in  the eyes of
dogs  (Table  9-25).  A higher concentration of 3000 ppm  caused turbidity in the
cornea of  the  red  fox  after a single  exposure of  7  hours  duration.   Repeated
exposure interspersed  with  two days of no exposure at a lower concentration of
400 ppm produced clouding  in the cornea of six  dogs,  which cleared during the
days free of exposure.   Development of  resistance to toxic effects in the cornea
after  repeated  exposure was  observed.
     Corneal opacity was also observed  within 10 hours in 6  dogs  that received a
subcutaneous injection  of  0.94 g/kg EDC  (Kuwabara  et  al.,  1968) (Table 9-25).
Marked  swelling of most of  the  endothelial cells of the cornea  was seen  at 12
hours,  and  the  reparative  process was  found to occur  by  24 hours.   Similar
corneal changes were produced in dogs, rabbits and cats when  EDC (a 1$ suspension
dissolved in mineral oil and homogenized in 0.556  gelatin  solution) was instilled
directly into the  anterior  chamber of the eye.    The authors suggested that the
apparent vulnerability  of the dog  eye to EDC  by  routes other  than direct  local
application was due to "a greater amount of dichloroethane coming  in contact with
the dog endothelium  rather than  an unusual susceptibility of the eye itself."
                                      9-94

-------
Inhalation
Inhalation
                                                                    TABLE 9-25

                                                   Effects of Ethylene Dlchlorlde on  the Cornea






vO
1
\o

-------
                                                                              TABLE 9-25 (Cont.)
                    Route
 Species
Number of
 Animals
       Dose
       Effect
                                                                                                                                          Reference
               Inhalation
               Inhalation
               Inhalation
dog




guinea pigs

rata

rabbi ta

dog
  10




  US

  NS

  NS

   3
vD

vO
               Inhalation
                                 dog
                  10
               Inhalation
                                 eats              NS

                                 monkeys

                                 chickens

                                 rodents (species, NS)
1000 ppm for 7 h,
i exposure
                                                                 1500 ppm for 7 h,
                                                                 repeated exposure
1500 ppm for 7 h, repeated
exposure
                1000 ppm for 7 h,  5 d/uk,
                repeated exposure,
                length NS
                                1000 ppn for 7 h,  5 d/ufc,
                                repeated exposure,  length  NS
Symmetric turbidity in the
 corneas of 8; up to 3 weeks
 required for partial
 regression.

Death in aost animals
 after 1 exposures;
 no adverse effect on eyes.
                                                                                                  Intense bilateral corneal
                                                                                                   opacity in both eyes 48 h
                                                                                                   after first exposure; death
                                                                                                   after 5,  6 or 30 exposures.
                                                                                                  Turbidity In eyes retained
                                                                                                   throughout.
                                 Increasing turbidity of
                                  cornea during 5 exposure days,
                                  clearing during 2 non-exposure
                                  days;  increased tolerance with
                                  successive bouts of exposure;
                                  complete resistance of cornea
                                  finally.

                                 No adverse effects in eyes
Heppel et al., 1901
                                                                  Heppel et al., 1944
                                 Heppel et al. 1944
                                 Heppel et al., 1944
                                                                                  Heppel  et  al.,  1944

-------
                                                                             TABLE 9-25 (Cent.)
Route
Subcutaneous



Inhalation
Species
dogs
(puppies and
adults)
cats
rabbits
albino rats
dogs
Number of
Animals
6
NS
NS
NS
6

0.91
0.94
0.94
0.94
400
Dose
g/kg
g/kg
g/kg
g/kg
ppm for 7 h, 5 d/wk,
Effect
Corneal opacity evidence within
10 hours; necrosis of corneal
endotnelium; clearing by
fifth day.
No effect on cornea
No effect on cornea
No effect on cornea
Hild clouding during first
Reference
Kuuabara et al
Kuwabara et al
Kuwabara et al
Kuwabara et al
Heppel et al . ,
. . 1968
., 1968
. . 1968
.. 1966
1944
                                                                for 10 weeks
week, cleared during 2 non-
exposure days; 5th week of
exposure; faint opacity of
cornea 10th week of exposure:
no trace of turbidity.
vo
              h  = hour; d  = day; wk  = week

-------
     9.2.2.2.   SUBCHRONIC AND CHRONIC EXPOSURE




     9.2.2.2.1.      Oral  Exposure — EDC has been tested  for carcinogenicity in



an  NCI bioassay  with Osborne-Mendel  rats and B6C3F1  mice (NCI, 1978).   The



compound was administered at  time-weighted average doses of 95 and 47 mg/kg (rats




of both sexes), 195 and 97 mg/kg (male mice) and 299  and 119 rag/kg  (female mice)



for  a period of  78  weeks, according  to the regimen described in Section 9.5.



Fifty  animals of each sex were treated/dose level, and the animals were observed




for  an additional  28  weeks  (rats)  or 12-13 weeks  (mice) after  pretreatment.



     As  detailed  in Section  9.5,  no  distinct dose-related   mean body  weight



depression  was  apparent  in  either  male or  female rats  relative to  vehicle




controls.  Mortality was, however,  early and severe in many of the dosed rats,



particularly those given  the  highest dose.   Mean  survival was  =55 weeks for the



high-dose  males and  females.   Toxic  rather than carcinogenic  effects  of  EDC



appeared to be  responsible  for the  deaths.   No distinct, dose-related mean body



weight depression  was observed in male mice or in  low-dose female mice, but mean



body weight  depression for high-dose  female mice was apparent as early as the



1 5th week  of treatment.  A significant positive  association  between  increased



dosage and elevated  mortality was  found  for female mice;  72$  (36/50)  of  the



females died between weeks 60  and 80.  The presence of one or  more  tumors in these




mice  suggests  that  these deaths may  have  been  tumor  related.    There  was  no



statistically  significant  association  between dosage  and  mortality for  male



mice.




     Munson et  al. (1982) evaluated the effect of subchronic  1 M-day  and 90-day



oral EDC exposures on the immune  response of male CD-1  mice.   EDC was adminis-



tered daily by  gavage in  the 11-day study at  levels of U.9  and 19 mg/kg, which



represent 0.01  and O.i  times  the single dose  LD_0 determined  in a preliminary
                                      9-98

-------
study  (Section  9-2.2.1.1).   For the 90-day study,  EDC  was administered in  the



drinking  water  at  levels  intended to  be equivalent to  those administered by



gavage; in addition, a 10-fold  higher dose was added because the results of  the



11-day exposure  indicated  that  doses  >19 mg/kg could be tolerated.  The calcu-



lated  time-weighted average doses of EDC  delivered  in the 90-day study based on



actual fluid consumed were  reported  to  be  3, 21  and  189 mg/kg.   Results of



standard toxicological analyses showed that body weight  was  unaltered during  the



14-day study, but  that EDC did  elicit a dose-dependent decrease in growth rate



and fluid consumption when administered over 90 days.  Thirty-two mice per dose



were weighed in the 90-day study but the  numbers weighed in the 11-day study were



not stated.  EDC did  not alter  the weights of  selected organs (liver, spleen,



lungs,  thyraus, kidneys,  brain)  in  either of  the studies.  It was further found



that exposure to 49 mg/kg/day EDC  caused  a 30% decrease in leukocyte number in



the 11-day study, although the number of  leukocytes was normal after 90 days of



exposure.   Other hematologic parameters  (hematocrit and  hemoglobin  evaluated




after  11 or 90 days, erythrocytes and  platelets  evaluated after 90 days), coagu-



lation  values  (fibrinogen  and  prothrombin time evaluated after  11  days),   and



clinical  chemistry parameters  (lactic  dehydrogenase,   serum  glutamic-pyruvate



transaminase and blood urea nitrogen evaluated after 11 days) were unaltered by



EDC exposure.  The hematological/coagulation/clinical chemistry parameters were



assessed in  10-12  and  16 mice/dose in  the 11-day and  90-day  studies,  respec-



tively.




     The status  of the humoral  immune  system was  determined  by  measuring the



number of IgM spleen antibody-forming cells  (AFC)  to sheep erythrocytes (sRBC)



after  11 and 90  days,  the serum antibody  level to  sRBC  after  90  days,  and the



lymphocyte  response  to  the   B-cell  mitogen  LPS  (lipopolysaccharide  from



Salmonella typhosa  0901)  after 90 days  (Munson et al., 1982).  Results of assays
                                     9-99

-------
 with 10-12 mice (14 days) and 16 mice/dose showed that EDC produced a significant



 (P<0.05) reduction in  AFCs  at  4.9 and 49 mg/kg in the 14-day study (25 and 40$



 suppression, respectively), but that EDC exposure for 90 days caused no signifi-



 cant (P<0.05)  change  in the number  of  AFC  spleen cells  (although  there was an



 indication of suppression at 189  mg/kg/day).   There  also appeared  to  be an EDC



 dose-dependent reduction in hemagglutination titer after 90 days, although this



 response was not significant.at the p<0.05 level.  EDC did not alter the spleen



 cell response  to  three concentrations of  LPS.    Cell-mediated immunity  was



 assessed by measuring the delayed hypersensitivity response  (DTH)  to sRBC and the



 response to the T-lymphocyte mitogen, concanavalin A. EDC produced a slight but



 significant (P<0.05)  non-dose-dependent  inhibition at both  4.9 and  49  mg/kg in



 the  14-day  study, but  90-day  exposure  did  not  alter DTH response or  spleen



 lymphocyte response to concanavalin A.  EDC did  not alter the functional activity



 of the reticuloendothelial  system, as measured by the  vascular clearance rate and



 tissue  (i.e.,  liver,  sple.en, lungs, thymus or  kidneys) uptake of 51Cr  after 90



 days.    Dexamethasone  was  used as  a positive control  in  the  above  studies:



 Results  of similar tests with female mice were not presented, but reportedly were



 not  remarkably  different from the males.



     The results of the above experiments (Munson et al., 1982) indicate that EDC



 produced a  suppression  of both  humoral and cell-mediated immunity when  adminis-



 tered daily for 14 days by  stomach  tube,  but not  when consumed in the  drinking



 water for 90 days (although there were  trends  toward suppression  at  the  high



 exposure level).   Two  explanations were  offered for  these  results.  First,  the



 effective dose at the immunocompetent cell may be higher with the bolus presenta-



 tion than with the semi-continuous  self-administration  in  the drinking water.



Second,   EDC may induce its  own  metabolism over  the longer  exposure period,



 thereby  effectively reducing the amount  of chemical reaching the  immune cells.
                                     9-100

-------
     9.2.2.2.2.     Inhalation Exposure — Four animal species (15 rats of both



sexes, 8 guinea  pigs  of both sexes,  2  male and 1  female rabbits, and  2 male



monkeys) were exposed to EDC vapor at levels of 100 pptn (1.62 rag/1) and 100 ppm



(0.105 rng/5.) for 7 hours daily, 5 days/week for 6 months (Spencer et al.,  1951).



In addition, 1 5 rats of  both sexes and 8 guinea pigs of both sexes were similarly



exposed to 200 ppm EDC (0.8l mg/R.) for 151  and 180 7-hour periods,  respectively.



Repeated exposure at 100 ppm did  not produce adverse effects in any of the four



species as  determined by  general appearance  and behavior,  mortality,  growth,



body and organ weights,  and gross and microscopic examination of tissues  (Table



9-26).  No adverse effects were indicated  by periodic hematological examination



of the treated rats, guinea pigs, or  rabbits.   Exposure to 200 ppm EDC did not



produce any adverse effects in the  rats,  but did  elicit  slight  parenchymatous



degeneration of the liver  with a  few diffusely distributed fat vacuoles in half



of the guinea pigs.  A slight increase of total lipid, phospholipid, neutral fat



and free and esterified  chloresterol was also found in the guinea pigs exposed to



200 ppm EDC, and  the  final body weights of  the  treated male guinea  pigs were



significantly different  from control animals.  Severe toxic effects were found in



the rats,  guinea pigs, and monkeys that were exposed to 400 ppm EDC (1 .62 mg/£)



(see Table 9-26).  The  three  rabbits  that  were  exposed to 400 ppm EDC were not



adversely affected, although Heppel et al. (1946)  reported a high mortality in



rabbits exposed to this  concentration of EDC.



     Heppel et  al.  (1945) exposed  rats, mice, rabbits, guinea pigs,  hogs and dogs



to 1500 ppm  EDC  (6.4  mg/S,).   Almost  all of  these  animals died before 6  daily




exposures of 7 hours each were completed.   Hemorrhaging in  the lungs, gastro-



intestinal tract and adrenals; fatty  degeneration  of the  myocardium;  degenera-



tion within the renal  tubules; and congestion of the  liver and  intestines were



found at autopsy.
                                     9-101

-------
                                                                         TABLE 9-26

                                                    Effeota of Subohronlo Exposure to Ethylene Dlchlorlde
           Route
 Speolea
   Number
Dose
Effect
Deference
      Inhalation
rata
26                1000 ppm (3.0 mg/1) 7 h/d x 5 d/wk   20 dead within 15  exposures;     Heppel  et al.,  1946
(average weight:                                        progressive weakness culmln-
 19B g)                                                 atlng In Inability  to stand.
                                                       Histology of 4 rata:
                                                        degeneration and  proliferation
                                                        changes in renal  tubular
                                                        eplthellun;  chronic splenltls
                                                        In 20 rats;  pulmonary congestion
                                                        In 2 rats.
      Inhalation
      Inhalation
o
K)
rabbits




guinea pig


guinea pig


guinea pig
 6                1000 ppn (3.9 mg/l)  7 h/d x 5 d/wk  5 dead within 43  exposures
(average weight:                                        survivor exposed 64  days
 2480 g>
                                                              Heppel et al.,  1946
15                1000 ppn (3.9 mg/l) 7 h/d x 5 d/wk   10 dead within 2 exposures       Heppel  et  al.,  1946
(average weight:
 161 g)
10                1000 ppm (3.9 mg/l) 7 h/d x 5 d/wk   10 dead within 2 exposures       Heppel  et  al.,  1916
(average weight:
 558 g)
16                1000 ppm (3.9 mg/l) 7 h/d x 5 d/wk   16 dead within 4 exposures       Heppel  et  al.,  1946
(average weight:                                         lacrimatlon and marked
 900 g)                                                 inactivity during exposure;
                                                        congestion of lung,  liver,
                                                        heart, kidney,  adrenal gland
                                                        and spleens.
      Inhalation
                      dogs
              6 (7000-8300 g)   1000 ppm (3.9 mg/l 7 h/d x 5 d/wk
                                                       I  dead after 30 exposures
                                                       1  dead after 43 exposures
                                                              Heppel et al.,  1946

-------
                                                                           TABLE 9-26  (Cent.)
                  Route
 Species
Number
Doae
Effect
Reference
             Inhalation
                             cats
              6 (2660 g         1000 ppa (3.9  ng/l)  7 h/d x  5 d/wk   2 dead after «3 exposures
              average weight)                                        congestion and fatty meta-
                                                                     morphosis In liver of all.
                                                                                    Heppel et al., 1916
             Inhalation      monkeys       2 (1880 g         1000 ppa (3.9 ag/l) 7 h/d x 5 d/wk   1  dead after  2  exposures         Heppel «t al., I9t6
                                           average weight)                                        1  dead after  43 exposures;
                                                                                                   fatty degeneration of liver
                                                                                                   and slight fatty  changes In
                                                                                                   kidney;  focal  myocarditis  In
                                                                                                   1  Donkey.
             Inhalation
                             cats
                                500 ppa 6 h/d x 5 d/wk for 6 wks
                                                    No deaths;  dilated hearts;       Hoffmann et al., 1971
                                                     blood urea nitrogen Increased
                                                     to 11H og/100 mi.
o
10
             Inhalation
             Inhalation
rabbita
guinea pigs   10
               500 ppa 6 h/d x 5 d/wk for 6 wks
               500 ppa 6 h/d x 5 d/wfc for 6 wks
                              3/4 dead after 10-17 exposures;   Hoffman et  al.,  1971
                               dilated hearts.
                              9/10 dead after 4-11 exposures;   Hoffman et  al.,  1971
                               apathy and weight loss,  fatty
                               degeneration and necrosis-of
                               myocardium, liver, kidney and
                               adrenals.
             Inhalation
rata
              10
               500 ppm 6 h/d x 5 d/wk for 6 wks
                              All dead after 1-5 exposures;
                               hypereuia of lungs; fatty
                               degeneration and necrosis of
                               myocardium, liver, kidney,
                               and adrenals.
                         Hoffman et al., 1971

-------
                                                               TABLE 9-26 (Cont.)
     Route
 Speoiaa
   Number
                                                       Dose
                                                                                           Effect
                                                                                                            Reference
Inhalation
rata
Inhalation
guinea pigs
Inhalation
Inhalation
rabbits
dogs
15 male
1 female
(average weight:
 167 g)
                              23 controls
IB male
2 female
(average weight:
 303-737)
30 controls

2 male
3 female
(average weight:
 3110 g)
4 controls

3 male puppies
(average weight:
 1330 g)
6 female adult
(average weight:
 9100 g)
                              4 controls
MOO ppm (1.54 mg/l) for
 7 h/d s 5 d/wk for
 60 exposures
400 ppm (1.54 mg/l) for
 7 h/d x 5 d/wk
400 ppm (1.54 ag/l} for
 7 h/d x 5 d/wk
400 ppm (1.54 mg/l) for
 7 h/d x 5 d/wk for
 167-177 exposures
Total dead:  9
 (6 dead after 4 exposures)
Histology In 6 survivors:
 no changes in 5; diffuse
 myocarditis and fatty
 degeneration of liver,
 kidney and heart in 1.
1 dead.
Histology:  no change.

13 dead after 45 exposures
 1 dead after 65 exposures
Histology in 10:  fatty
 degeneration of liver and
 kidney In 4 dead; fatty
 degeneration of heart in
 2 dead; no histologlcal
 change in 6 survivors of
 exposure.
3 deaths.

All dead after 97 exposures.
No hematological changes.
No deaths.

No deaths; 6 adults: no
 effect on mean arterial
 pressure, Bromsulphaleln
 excretion rate, prothrombin
 time, plasma total protein,
 albumin, globulin, non-
 protein nitrogen, Icterus
 index, urine; slight fatty
 change In livers of 5 and
 kidney of 1.
No deaths.
Heppel et al., 1946
                                                                                                                     Heppel  et al.,  1946
                                                                                                                     Heppel et al.,  1946
                                                                                                                     Heppel et al.,  1946

-------
                                                                          TABLE 9-26  (Cont.)
                 Route
                 Specie?
                 Number
       Doae
                                                                            Effect
                                       Reference
            Inhalation
            Inhalation
                            rata
                            rata
                              15 male,
                              15 female
                              20 male,
                              20 female
                                400 ppo (1.62 mg/l)  7 h/d,
                                 5 d/wk
                                400 ppm (1.62 mg/l) 7 h/d,
                                 5 d/wk
i
o
Inhalation
            Inhalation
guinea pigs   8 male,
              8 female
                guinea pigs   2 male
400 ppo (1.62 ng/l) 7 h/d,
 5 d/ wk
                                400 ppffl (1.62 mg/l) 7 h/d,
                                 5 d/wk
Females died within 10
 exposures.
Kales died within 40
 exposures.

60ft mortality after 2 or 3
 exposures; rapid loss of
 body weight; slight Increase
 of liver and kidney weight;
 slight cloudy swelling of
 liver with a few large fat
 vacuoles.
No significant differences In
 blood urea nitrogen, non-
 protein nitrogen, serum
 phosphatase and plasma
 prothrombln olotting time.

Hales died within 10
 exposures.
Females died within 24
 exposures.
                                                                                                                                Spencer et al., 1951
                                                                                                                     Spenoer  et al., 1951
                                     Sacrificed after 1,  3,  4,  and   Spencer et al.,  1951
                                      10 exposures;  rapid loss  of
                                      body weight;  Increased liver
                                      and kidney weight;  slight to
                                      moderate central fatty
                                      degeneration  of liver; slight
                                      to moderate cloudy  swelling of
                                      renal tubular epithelium; no
                                      alteration in lungs,  heart,
                                      spleen and teates;  increased
                                      BUN and Increased blood non-
                                      protein nitrogen levels;  no
                                      change In serum phosphatase
                                      or plasma prothrombln clotting
                                      time.

-------
                                                                            TABLE 9-26 (Cont.)
Route
Inhalation
Species
rabbits
Number
2 male,
i female
Dose
400 ppn (1 .62 ng/fc) 7 h/d,
5 d/wk for 165 exposures
Effect
No effect on general
appearance, behavior;
Reference
Spencer et al., 1951
                                                                                                   mortality, body weight,
                                                                                                   histology of liver, kidney,
                                                                                                   lungs, heart, spleen or teatea;
                                                                                                   organ weights, blood non-
                                                                                                   protein nitrogen.   BUN,
                                                                                                   serjo phosphatase, plasma
                                                                                                   prothrombln clotting time.
             Inhalation
                             monkeys
2 males
100 ppm (1.62 mg/l) 7 h/d,
 5 d/wk
I
.-»
o
                                                       One sacrificed In moribund
                                                        condition after 8 exposures:
                                                        enlarged liver with In-
                                                        creased neutral fat and
                                                        esterlfled cholesterol
                                                        content, marked degeneration
                                                        and vacuolation of hepatic
                                                        cells;  moderate degeneration
                                                        of renal tubular epithelium
                                                        with cast formation;  In-
                                                        creased plasma prothrombin
                                                        clotting time.
                                                       Second sacrificed after 12
                                                        exposures:  similar,  but
                                                        milder, changes.
                                                       No significant hematologlcal
                                                        change.
Spencer et al., 1951
             Inhalation
                             rats
                                           12 male
                                            Osborne-Mendel
                                           (average weight:
                                            72 g)
                                           12 controls
                  200 ppm (0.73 mg/t,) 7 h/d x 5 d/wk   8  dead after 6 exposures
                                                                                                  No deaths.
                                                                     Heppel et al.,  1916

-------
                                                              TABLE 9-26  (Cont.)
     Route
                              Species
                                 Number
                                                                    Dose
                                                                                           Effect
                                                                                                                                         Reference
Inhalation
                rata
                                           1 male Wlatar
                                           11 female Ulstar
                                           (average weight:
                                            249 g)
                  200 ppa (0.73 og/1)  7 h/d  x 5 d/wk
                              1>I controls
                                     7 dead; 1 after  73 exposures,
                                      6 after  14 exposures.
                                     Symptoms:  weight loss;
                                      listless; crusting of eyes.
                                     Histology of  5 rats killed
                                      after 86 exposures:  fatty
                                      degeneration of kidney  In
                                      1  rat; no other changes
                                      noted in liver, heart,  lungs,
                                      kidney,  adrenals and spleen;
                                      no effect on red and white
                                      blood cell count, hemoglobin
                                      and differential counts
                                      (7 treated compared  with 6
                                      control  rats).
                                     1 dead.
                                                                                                                    Heppel et al., 1916
^
o
Inhalation


Inhalation
                             mice
                             guinea pigs
20
12 male,
2 female
(average weight:
 376 g)
                              18 controls
200 ppm (0.73 mg/l) 7 h/d x 5 d/uk   18  dead after 7 exposures.       Heppel  et  al., 1916
200 ppm (0.73 Bg/O 7 h/d v 5 d/wk
4 dead after 88 exposures;
 1 dead after 115 exposures.
Histology of 9 guinea pigs
 killed after 124 exposures:
 pulmonary congestion in 4,
 liver necrosis in 1,
 necrosis of adrenal cortex
 in 1.
1 dead.
Histology of 5 guinea pigs:
 fatty liver and myocardium
 of 2; no other changes noted
 in liver, heart, lung,
 kidneys, adrenals and spleen.
Heppel et al., 1946

-------
                                                                    TABLE 9-26 (Cont.)
         Route
 Species
                                     Number
                         Dose
                                                                                                Effect
                                                                                                            Reference
    Inhalation
rabbits
                                  (average weight:
                                   2720)
                                  1 controls
                  200 ppn (0.73 ng/A) 7 h/d x 5 d/wk
                                                                     No deaths.
                                                                     No hlatologlcal changes
                                                                      found; no effect on red
                                                                      and white blood cell
                                                                      count, hemoglobin, and
                                                                      differential counts
                                                                      compared with controls.
                                                                     No deaths.
                                                                                                                         Heppel et al., 1916
    Inhalation
monkeys
2 males
(average weight:
 8740 g)
                                  no controls
200 ppm (0.73 mg/l) 7 h/d x S d/wk
 for 125 exposures
                                                                                         No deaths.
                                                                                         Histology of both:  focal
                                                                                          calcification in adrenal
                                                                                          medulla of 1; fatty
                                                                                          droplets In liver and
                                                                                          myocardium of both.
Heppel et al., 1946
    Inhalation
rats
o
oc
15 male,
15 female
    Inhalation
guinea pigs   6 male,
              8 female
200 ppn (0.81 mg/4) 7h/d,
 5 d/wk for 151 exposures
                  200 ppm (0.61 ug/l) 7 h/d,
                   5 d/ wk for 180 exposures
                                                                     No adverse effects on
                                                                      general appearance, behavior,
                                                                      growth, mortality, final
                                                                      body and organ weights,
                                                                      hematology, gross and
                                                                      microscopic histology.
                                                                                                                         Spencer et al., 1951
                                                                                         Final body weight
                                                                                          significantly different
                                                                                          from controls in males, and
                                                                                          growth less than controls,
                                                                                          but not siglflcant In females.
                                                                                          Significant increase in male
                                                                                          liver weights only; slight
                                                                                          parenchyoatous hepatic
                                                                                          degeneration in half of
                                                                                          guinea pigs of both sexes;
                                                                                          no effect on hematology; slight
                                                                                          increase in trial lipid,
                                                                                          phospholipid, neutral fat,
                                                                                          free and eaterified cholesterol.
                                                                     Spencer et al., 1951

-------
                                                               TABLE  9-26  (Cont.)
     Route
 Species
                                 Number
                                                       Dose
                                                                            Effect
                                                                                                                            Reference
Inhalation
                rats
              15 male,          100 ppa (0.105 mg/i) 7 h/d,
              15 female          5 d/wk
                                 males:  151  exposures
                                 females:  112 exposures
                                                       No adverse effect on general
                                                        appearance, behavior,
                                                        mortality, growth,  final body
                                                        and organ weights,  hematology,
                                                        gross and microscopic histology,
                                                        BUN, blood non-protein nitrogen,
                                                        serum phosphatase,  plasma
                                                        prothroobin clotting time,
                                                        total levels llpld, phosptio-
                                                        llplds, neutral fat In liver,
                                                        free and esterlfled
                                                        cholesterol In liver.
                                                                     Spencer et al.,  1951
Inhalation
guinea pigs
8 male,
8 female
100 ppm (0.105 mg/JO 7 h/d,
 5 d/wk
 males:  121 exposures
 females:  162 exposures
Inhalation
rabbits
Inhalation
monkeys
2 male,
1 female
2 male
100 ppm (0.105 mg/ft) 7 h/d,
 5 d/wk for 178 exposures
100 ppm (0.105 me/£) 7 h/d,
 5 d/wk for 148 exposures
No adverse effect on
 mortality, growth,  final
 body and organ weights,
 BUN, blood non-protein
 nitrogen, serum phosphatase,
 plasma prothrombln clotting
 time, gross and microscopic
 histology, total liver llpld
 phosphollplds, neutral fat
 In liver, free and esterlfied
 cholesterol In liver.

No adverse effect on
 appearance, behavior,
 growth, final body and
 organ weights, gross and
 microscopic histology,
 hematology.

No adverse effect on
 appearance, behavior,
 growth, final body and
 organ weights, gross and
 microscopic histology,
 hematology.
Spencer et al., 1951
Spencer et al., 1951
Spencer et al., 1951

-------
                                                              TABLE  9-26  (Cent.)
     Route
                              Species
   Number
       Doae
                                                                                           Effect
                                       Reference
Inhalation
                rats
                                           16  female
                                           23  male
                  100 pom (0.12  mg/O  7 h/d  x  5 d/wk
                   for 1  months
Inhalation
 1^

o
                              guinea plga    10 male
                                             6 female
                              30 control
                  100 ppm (0.42 mg/l)  7 h/d  x  5 d/wk
                   for 4 months
Inhalation
                              nice
19 Juveniles
100 ppm (0.42 mg/H) 7 h/d x 5 d/wk
 for 19 exposures
No deaths, no effect on rate
 of growth in 15 males
 compared with 1 5 male
 controls.
i5 of 16 females became
 pregnant; rat pups were
 unaffected by exposure.
No hiatologleal effects in
 liver,  heart, lungs, kidney,
 adrenal glands and spleen of
 10 rats examined.

2 dead (disease of neck with
 enlarged oaaeous glands)
No histologlcal effects in
 liver,  heart, lungs, kidney,
 adrenal glands and spleen of
 10 animals examined.
3 dead (disease of neck with
 enlarged easeous glands).

No deaths; no effect on weight
 gain.
                                                                    Heppel et al., 1946
                                                                     Heppel  et  al.,  1946
Heppel et al., 1946
Inhalation
                              cats           4
                              rabbits        4
                              guinea pigs   10
                              rats          10
                  100 ppm 6 h/d x 5 d/wk for 1 7 wks
                                     No clinical symptoms
                                     No effect on serum levels of
                                      creatlnine, urea, SCOT,
                                      SGPT,  on body weight.
                                     No changes In liver, kidney,
                                      and other organs (not  specified).
                                Hoffman et al., 1971
h = hour; d = day; wk = week

BUN = Blood urea nitrogen

-------
     In  another  study,  Heppel  et  al. (1946)  found that repeated  exposure (7



hours/day, 5 days/week) to 1000  ppm  EDC  elicited death in most of  the exposed



rats,  rabbits,  guinea pigs,  dogs,  cats  and monkeys  (see Table 9-26).   Rats,



rabbits and guinea pigs appeared to  be more  susceptible  than  the  other species



tested.  Pathological changes, which were  varied,  included  congestion of lung,



liver, heart, kidney, adrenal  gland  and  spleen in guinea pigs; congestion and



fatty metamorphosis in the liver of  all  exposed  cats;  renal changes in exposed



monkeys and  rats;  and  pulmonary congestion in a  few  rats.    Chronic  splenitis



occurred in  all  rats exposed  to repeated  inhalation of  1000 ppm EDC.   Focal



myocarditis was found in one monkey.   Similar repeated exposures to 400 ppm EDC



produced high mortality in rabbits,  rats, and guinea pigs (see Table 9-26), but



mortality was not observed in 9 dogs  subjected to  from 167  to 1 77 exposures at



this concentration.  No changes were observed in a variety of clinical parameters



that were assessed in 6 adult dogs,  although slight fatty changes  were noted in



the  livers  of five  and  the kidney  of  one  of the dogs.   Rat and guinea pig



survivors of 400 ppm EDC exposure did not exhibit histological changes with the



exception of 1  of 6 rats that exhibited diffuse myocarditis  and fatty degenera-



tion of the liver, kidney and heart.   Repeated inhalation exposure to 200 ppm EDC



produced mortality in mice,  rats and  guinea pigs, but  not in rabbits or monkeys



(see Table 9-26).  Pathologic effects of  exposure to 200  ppm EDC included a few



cases  of  pulmonary congestion,  fatty degeneration in the  kidney of  one rat,



necrosis of the liver and adrenal cortex in  one  guinea pig, and fatty droplets in



the  liver  and  myocardium of  both monkeys.   No  clinical or  histopathological



effects were observed in 39  rats or  16 guinea pigs exposed to 100  ppm EDC for 4



months or in 19 juvenile mice exposed to  this level of EDC for 19  exposures.



     Hoffman et al. (1971) exposed groups of 1 cats, 1 rabbits, 10  guinea pigs



and  10  rats  to 500 or  100  ppm EDC  for  6  hours/day,  5 days/week for  6 weeks.
                                     9-111

-------
Mortality  from exposure  to  500 ppm EDC  was high (see Table  9-26).   Variable



changes  in  the heart,  lungs,  liver,  kidney,  and  adrenals were  found at autopsy.



No toxic effects  were  observed in  the  exposed  animals at  100 ppm.  An elevation



of  BUN  and  creatinine  was  noted  in  one  fourth  of the  rabbits,  but  the



significance  of  these  changes is uncertain.   The  exposed  cats did not grow as




well as  the control  cats.



     The  effect  of  EDC  on blood elements and on  renal and liver function was



investigated  using groups of 10 rabbits that were  exposed to 3000 ppm  for 4



hours,  or  to 3000  ppm  for  2 hours/day,  5  days/week   for  90  days (Lioia,



1959a,b,c,d).   The  only significant  changes  observed  after  the acute  1-hour




exposure were granulations in about  20$ of the granulocytes. Varying degrees of



anemia accompanied by  leukopenia and thrombocytopenia and  frequent hyperplasia



of granuloblastic  and erythroblastic parenchyma in the bone marrow were observed



after subchronic  exposure.   There was also  a  reduction in leukolipids,  but no



change in polysaccharides, peroxidase, or ribonucleic acid.  In tests for liver



function,   a  decrease  in  albumin-globulin  ratio,   a  slightly   elevated  BSP



retention,   slightly  elevated  values  in  colloidal   tests   (cadmium  and



cholesterol), and  normal Van den Berg (indicating an absence of  elevated serum



bilirubin)  and blood amino acid  levels were observed. Measurement of creatinine



clearance,  portal  blood  flow, and glomerular  filtration rate indicated altered




renal function.  Congestion, vascular degeneration and small necrotic areas were



found in livers and  kidneys.



     Dimitrieva and  Kuleshov  (1971)  examined the effect  of EDC  on  brain activity



in 18 albino rats exposed to  1235 ppm EDC  (5 mg/i) for 3-5  months.   The duration,



frequency and number of exposures were not reported.   Electroencephalograms were



recorded  before  exposure and at monthly intervals  thereafter  from  silver and




platinum electrodes  implanted in the brains of the  test animals.  The stimulus



was composed  of  an arhythmic photic stimulus of  constant intensity  and pulse






                                     9-112

-------
duration.  A  maintained  frequency of activity was  observed  in the  EEC  of the



treated rats.  A progressive diminution  of amplitude of vacillations occurred,



with the amplitude  of delta  rhythms reaching 50-70 MV (amplitude of delta rhythm



before exposures was not specified), while the amplitude of beta rhythm decreased



to 10 to 15 MV (amplitudes of pre-exposure beta rhythms were 30 to 80 MV).  A loss



of ability to assimilate an imposed rhythm was observed by the authors.



     The   results   of   an   inhalation   bioassay   for  carcinogenesis   with



Sprague-Dawley rats and Swiss mice have  been  published (Maltoni  et al.,  1981).



The experimental design and  results of these assays are detailed in Section 9>5,



but it should be  noted that  exposure to 250 to 150 ppm,  50, 10 or 5 ppm EDC for 7



hours/day, 5  days/week for 78 weeks elicited no treatment-related changes in body



weight  or survival ability in  rats  of  either  sex  or male  mice.   There  was,



however, decreased  survival  ability in  female mice exposed to the high level of



EDC.  The animals exposed to 250 ppm at the onset of the study exhibited signs of



toxicity  (i.e.,  ruffling  of hair,  hypomotility  and  loss   of  muscle  tone),



prompting dose decrease to  150 ppm after several  weeks.  Systemic pathological



examinations  were  performed on all  the  major  organs  of all animals with  or



without pathological  changes,  but the only non-neoplastic  histologic  effects



reported were some  "regressive" changes in the liver and adrenal glands that were



not dose-related (Cox, 1982).



     In a related study with Sprague-Dawley rats, Spreafico et al. (1981)  inves-



tigated the effect  of inhalation exposure  to identical levels  of EDC (250 to 150,



50, 10 and 5  ppm) on blood clinical chemistry parameters.  Histological examina-



tions were not performed  on  the  rats  exposed to  EDC in this study.   Eight to 10



animals  per  dose  level  were  sacrificed  after  3,  6, 12  or 18  months of  7



hours/day, 5  days/week exposure.   Rats  used for the  3,  6  and 18  month raeasure-
                                     9-113

-------
ments were  started  at  3 months of age, and the 12 month determination was made



with animals  that were exposed from 14 months  of  age.



     The results suggest that long-term inhalation exposure of one year or longer



to EDC at levels of 150 ppm does not produce marked toxicity in rats exposed from



3  months  of  age   (Spreafico  et  al.,  1981).    There  were  no  statistically



significant  changes   between  treated  and  control  animals  with  respect  to



circulating levels  of  red  blood  cells,  total white blood cell  numbers, platelet



numbers,  relative  percentages of lymphocytes, granulocytes and monocytes,  and



circulating protein levels.  Percent albumin values were significantly increased



in both sexes at 3  and 18  months but not at 6 months, but no clear  dose-response



relationship  was apparent.  Gamma globulin percentages were  significantly lower



at 3 months in the 150 ppm EDC  group.   Although  changes were  not  apparent at 6



months, at  18 months there was a significant decrease observed only  at 5 and 10



ppm.



     No  treatment  related effects were  found on levels of y-glutarayl  trans-



peptidase   (y-GT),   serum  glutaraic-oxalacetic  transaminase   (SCOT),   serum



glumatic-pyruvic transaminase  (SGPT),  serum alkaline phosphatase, bilirubin or



cholesterol (Spreafico et  al., 1981).  A slight, but not significant,  increase in



creatine phosphokinase (CPK) level was seen  at 18 months in males  exposed to 50



and  150  ppm  EDC.   Lactic acid  dehydrogenase  (LDH)  levels were  significantly



elevated after 3 months of  exposure in both treated male  and  female  groups, but a



dose-response relationship was apparent only in males.  At 6 months, there was an



insignificant increase in  LDH  levels.   At 18  months,  significantly higher  LDH



levels were observed only  in males exposed to 5, 50 and 150 ppm EDC without an



apparent dose-response relationship.   No clear  changes were  observed  for  BUN



levels, serum glucose  levels or  uric  acid  levels  (Spreafico et al.,  1981).



     No consistent and  significant differences  in  a battery of urinary tests (pH,



proteins, bilirubin, glucose,  hemoglobin, erythrocytes,  leukocytes, epithelial



                                      9-111

-------
cells, casts, cystals, mucus and microorganisms) were  observed  after 18 months




of inhalation exposure (Spreafico et al.,  1981).



     Changes  in  liver and  kidney  function  were  indicated,  however,   in  rats



exposed to EDC at 14 months of  age.  SGPT levels were significantly increased in



rats of both  sexes  exposed to  50 and 150 ppm.  y-GT  levels  were significantly



elevated in females  treated with  50 and 1 50 ppm.  SCOT levels  were significantly



elevated at the two lower  doses  (5  and  10  ppm), and significant decreases were



observed at the  2  higher  doses  (50 and 150 ppm).  Cholesterol  levels were signi-



ficantly lower at 50  and 150 ppm in both sexes, but  there were no significant



changes in bilirubin, CPK, alkaline phosphatase, LDH or glucose levels.  Signifi-



cantly higher levels of uric acid were found at 50  and 1 50 ppm and higher blood



glucose levels were  observed at 150 ppm.  No significant  changes  were  found in



blood elements or in  the battery of tests conducted on urine in  the rats that



were initially exposed at 1U months  of age  (Spreafico et al.,  1981).



9.2.3.  Summary of Acute, Subchronic and Chronic Toxicity.



     9.2.3.1.  INHALATION EXPOSURE — Information regarding the acute effects of



inhaled EDC in humans  is  available primarily from cases of  occupational exposure



(see Section 9.2.1.1). Although  the reported cases had both fatal and non-fatal



outcomes,  many of  the reports  were  foreign  and none  of  the reports  provided



quantitative exposure  data.  Further, although EDC was  usually reported to be the



primary vapor to which the workers were exposed, the preponderance of exposures



were to poorly characterized mixtures  of EDC and other solvents,  or to EDC of



unknown purity.   Symptoms and  signs  of acute  inhalation exposure  were  often



indicative of CNS and  gastrointestinal disturbances,  and  clinical  evidence of



liver and  kidney dysfunction, as well as irritation of the  respiratory tract and



eyes, have also been  observed.   Death was usually  ascribed  to  respiratory and



circulatory  failure,  and  autopsies frequently  revealed  pulmonary edema  and
                                     9-115

-------
 congestion,  cellular  degeneration,  necrosis  and  hemorrhagic  lesions of  most
 internal organs  (e.g.,  liver,  kidneys,  spleen,  lungs and respiratory  tract,
 brain,  stomach and intestines).
      The results  of acute  experimental  studies  with three  or four  subjects
 suggest that the threshold of light perception  (duration of exposure not stated),
 depth of breathing (1-minute  exposure)  and  vasoconstriction  (30-second and  15-
 minute  exposures)  increased  with exposure to EDC (Borisova, 1957).  Spirographic
 and plethysmographic  responses reportedly first differed from baseline values at
 1.5 ppo and  appeared  to be dose-related  to  12.4 ppm,  the  highest level tested;
 exposure to 1  ppm (the  lowest  level tested)  reportedly  had  no effect on  the
 physiologic  end points.  Although these Russian experiments appear to indicate a
 threshold of toxic action, the  reliability  of the data is uncertain due  to  the
 small number of subjects  tested  and an absence of reported  baseline  measurements
 for each subject.  Some  assurance of data  credibility can be gleaned from  the
 fact that the  range  of concentrations  tested (i.e.,  =1-12.4 ppm)  represented
 corroborated sub-threshold,   threshold   and  above  threshold  odor  perception
 concentrates.   Borisova (1957) determined the odor  perception threshold of  EDC to
 be  =6 ppm, and  this value is consistent with an absolute odor threshold of 6  ppm
 that was determined by  Hellman and Small (1973,  1974)  in a Union  Carbide study.
 Further,  the sensitivity  of  the  analytic method  used by Borisova  (nephelometry)
 appears  to be more  than adequate to determine EDC at  the  reported  experimental
 levels.   Hellman  and  Small  (1973,  1974) also  determined an odor  recognition
 threshold of 40 ppm for EDC.
      Acute inhalation studies with a variety of animal  species  indicate that  the
 effects  of single  EDC  exposures  are  similar  to  those in humans.   Immediate
symptoms of  toxicity are  related to CNS  depression (e.g.,  narcosis)  and delayed
histopathologic changes (e.g.,  congestion and degenerative  effects) have been
                                     9-116

-------
observed  primarily  in the  liver,  spleen,  kidneys,  lungs  and adrenals.    As



detailed in Section 9.2.2.1.3 and Table 9-21, the severity of effects are depen-



dent upon  duration  and  concentration of  exposure.   For rats  exposed  for  5-8



hours, it appears that adverse effects are  not  elicited  by  exposure at  200  ppm



(Spencer et al., 1951); signs of intoxication first appear at 300 ppm (Spencer et



al., 1951)  and  mortality at >600 ppra  (Heppel  et al.,  1974; Carpenter,  1949).



Concentrations as high as 12,000  ppm  were  inhaled by  rats for 0.1  hours  without



adverse effects (Spencer  et  al.,  1951).



     The dose-response relationships for  acute  inhalation  exposure are  not  as



thoroughly characterized  for other species.  Corneal opacity was observed  in dogs



exposed via inhalation to 1000  ppm for  7 hours  (Heppel  et al.,  1944).  Signs of



intoxication were not present in  guinea pigs that were exposed to  1200  ppm  EDC



for 8 hours (Sayers et al.,  1930), but exposure  to 1500 ppra for 7 hours produced



mortality in mice, guinea pigs  and rabbits  (Heppel et al.,  1945).   Histopatho-



logical  effects  (e.g.,   pulmonary congestion/edema,  alterations  to kidneys,



liver, spleen and adrenals)  were generally apparent in rats, mice,  guinea pigs



and rabbits at concentrations of >3000 ppm (Spencer et al.,  1951 ; Heppel  et al.,



1945; Sayers et al., 1930).  Spencer  et al.  (1930) felt  that narcosis that  was



elicited by concentrations <3000 ppra was attributable to organ injury rather than



depression of the CMS.



     Limited quantitative data  are available, primarily from the foreign  litera-



ture, regarding the effects of repeated EDC exposure on humans.   The studies that



are available are deficient because control data are lacking and  because dura-



tions of exposure  and  numbers of subjects were poorly  characterized.  The avail-



able data suggest that chronic  intermittent exposure in  the range  of 10-37  ppra



may represent a lowest-observed-adverse-effect  level  (LOAEL) but,  as summarized
                                     9-117

-------
 subsequently,  the data  do not  provide  a basis for  identifying a  no-observed-




 adverse-effect level (NOAEL) or a no-observed-effect-level  (NOEL) for humans.



      Case  reports   (Byers,  19**3;  Rosenbaum,  19^7;  Guerdjikoff,   1955)  and a



 foreign health survey (Cetnarowicz, 1959) of workers who were exposed repeatedly



 to  1 ,2-EDC vapor  indicate that  typical  symptoms  and signs of acute poisoning



 developed with exposure  to concentrations in the range of =60-200 ppm.   In  the



 Cetnarowicz (1959) survey,  16  workers  who  were exposed to  =10-200 ppm for  2-8



 months  were examined; 10 of 10 who  were exposed  to  60-200 ppra  complained of



 adaptable eye  irritation and 6  of  the 10 experienced  typical symptoms of expo-



 sure, but only 1  of  6 exposed to 10-37  ppm  were symptomatic.  Positive symptoms



 and signs of exposure, particularly those indicative  of mucous membrane irrita-



 tion, were also observed  in 90 of 118  agricultural  workers who were  concurrently



 exposed  to EDC  vapor  (=15-60  ppm)  and  liquid  (prolonged dermal  contact)



 (Brzozowski et al.,  1954).




     The results of  two other foreign health surveys of workers exposed to EDC in



 the  range  of  the Cetnarowicz  (1999)  low  exposure group  (10-37 ppm)  support



 Cetnarowicz's  apparent finding  that overt  symptoms may  not be the  predominant



 effect  of lower exposures;  there appears, instead,  to  be a prevalence of neuro-



 logic and  clinical  effects.   Rosenbaum (19**7) examined 100 workers who were



 exposed  to <25 ppm for 6  months to  5 years  and  found disturbances that included



 "heightened lability" of  the autonomic  nervous  system,  increased  hidrosis and



 frequent  complaints  of fatigue, irritability and sleeplessness,  but no  hemato-



 logic or functional  changes of  the  internal  organs.   In   the study by  Kozik



 (1957),   which  is  particularly notable for  its  reliable  exposure  data,  workers



exposed  to  time-weighted  average  concentrations  of  10-15 ppm  experienced



 increased  morbidity,  particularly  from  gastrointestinal,  liver  and bile  duct



disorders.  The number of workers surveyed  in this  study was not  clearly stated
                                     9-118

-------
in the available translation,  but it was >83.   The occurrence of overt symptoms



in these workers was not discussed,  but impaired performance on control reaction



time tests suggested a possible effect on the  nervous system.



     Case reports of workers who were repeatedly  exposed  to  unknown concentra-



tions of EDC also suggest that neurologic effects  such  as nervousness,  irrita-



bility, tremors and  loss  of reflexes (McNally and Fostvedt,  19^1 ;  Delplace et



al.,  1962;  Suveev  and  Babichenko,   1969)  and complaints  of  skin  and  mucous



membrane irritation (Suveev and Babichenko,  1969;  Rosenbaum,  1947;  Guerdjikoff,



1955) are more  prevalent in  chronically exposed workers than in workers who were



acutely exposed.



     Studies with several species of animals  provide  information regarding the



threshold region of  effects for  subchronic  and chronic inhalation  exposure to



EDC.  Three studies  in which rats,  guinea pigs, rabbits,  cats and  monkeys were



exposed to 100  ppm CDC for 7 hours/day, 5 days/week for 4-6 months have shown no



treatment-related adverse  effects  on  survival,  growth,  hematology,  clinical



chemistry, organ weights or histology (Heppel et al., 1946; Spencer et al., 1951 ;



Hoffman et al.,  1971).  Adverse effects were also not  noted in juvenile mice that



were exposed to  100 ppm EDC for  19 exposures  (Heppel et al., 1946).   The apparent



NOAEL of  100 ppm  is supported  by the results of  comprehensive chronic  studies



with rats in which similar weekly exposures to EDC  (7  hours/day, 5 days/week) at



concentrations  of 5, 10 or 50 ppra for 18-19 months had no significant treatment-



related adverse effects on histology (Maltoni et  al., 1981)  or blood or urine



clinical  chemistry  indices (Spreafico  et  al., 1981) in rats of   either  sex;



exposure to 150 ppm elicited decreased  survival in  female  rats,  but no clear



effects on clinical  chemistry parameters  in either sex.  There was an indication



of  renal  and  liver  damage (increased  SGPT,  increased  serum Y~GT»  decreased



cholesterol, increased uric acid,  increased  blood  glucose)  in mature (i.e., 14-
                                     9-119

-------
months-old) rats that were similarly exposed to 50 OP 150 ppm EDC for 12 months,



but hematologic  effects were not  found  and  histological examinations  were  not



conducted.



     Subchronic exposure to 200 ppm EDC (7 hours/day, 5 days/week) produced toxic



effects  in  several  species,  but the severity of  effects appeared  to  vary with



investigation.   Heppel et al.  0946)  observed mortality  among  rats,  mice  and



guinea pigs exposed to 200 ppm EDC, but not among  rabbits or monkeys, and histo-



logical  lesions were  not  found in any of these species.  Spencer et al. O951 )



did not  observe mortality in rats or guinea pigs  similarly exposed to 200  ppm



EDC, but other effects were noted  in the guinea pigs (decreased body weight gain



and increased liver weight in males,  slight hepatic  degeneration  in  males  and



females).   Rabbits and monkeys  were  not tested by Spencer et al.  (I95t)>   An



unequivocal frank-effect  level (FED of UOO  ppm is  representative for  EDC as



indicated by  production of high mortality and histopathological  alterations in



the liver and kidney  in rats and  guinea pigs after a few exposures  (Heppel et



al., 19^6; Spencer  et al., 1951).   Spencer et al.  (1951) reported that exposure



to 100 ppm EDC had  no effect  on  rabbits  (as reported  at  200 ppm), but Heppel et



al.   O9U6) observed  some  mortality at this  level.   Exposure to  500 ppm  EDC



produced high mortality in rats, guinea  pigs and  rabbits within a few exposures



(Hoffman et al., 1971).



     9.2.3-2.  ORAL EXPOSURE  —   Limited data  are available on  the effects of



oral exposure to EDC.   Human case reports of accidental or intentional ingestion



of EDC indicate  that  the  toxic response to oral exposure is similar to that of



inhalation exposure (Section 9.2.1.1 .1 .1 ).   Ingestion of quantities of EDC esti-



mated to have ranged from 8-200  ml have been  reported to be  lethal (see Table



9-18).  This is equivalent to =110-3570  mg/kg  if it is assumed  that an average



human weighs 70 kg.  Median lethal doses  in rats (McCollister et al., 1956; Smyth
                                     9-120

-------
et al.,  1969)  and mice (Heppel et al.,  1945)  have been reported in the range of



700 mg/kg.  Higher oral doses  {up to 2.5  g/lcg)  were tolerated by  dogs without



mortality (Kistler and Luckhardt, 1929).   The apparent higher  tolerance in dogs



and some humans may be related to the ability or these species to  vomit.   Dose



information was not reported  in  human  case reports of non-lethal  ingestion of




EDC.



     Administration of 189 mg/kg/day EDC in the drinking water  for 90 days caused



a decrease in growth rate in mice,  but  no  significant  effects  on  organ weights,



hematology indices,  clinical  chemistry indices, blood  coagulation or  immune



system function,  although a  trend towards immunosuppression was noted (Munson et



al., 1982).   Similar  administration of 24 or 3 mg/kg/day EDC in  the  drinking



water for 90 days had  no effect on mice.



     The only dose-response  data  that are available for chronic oral exposure to



EDC are the results of a 78-week NCI carcinogenesis bioassay with rats and mice



(NCI, 1978).   Administration of 95 mg/kg/day EDC  via gavage (5 days/week) caused



early and severe  mortality in  rats  of  both  sexes,  and  ^47  mg/kg/day  caused



decreased survival in male rats {after  90  days)  and female rats (throughout the



experiment).    Non-carcinogenic toxic effects appeared to  be  the  cause  of the



deaths,  as indicated by the presence of a  variety of lesions including broncho-



pneumonia and endocardial thrombosis.  Body weight depression was not  found in



any of the treated male or female rats.  Treatment-related weight depression and



mortality was found in female  mice that were similarly exposed  to 299 rag/kg/day,



but not  in females exposed  to 1^9 mg/kg/day  or  in males exposed  to 195 or 97



mg/kg/day.   The presence of  tumors in the  high  dose  female mice  suggested,



however, that the deaths may have been  tumor-related.
                                     9-121

-------
9.3- REPRODUCTIVE AND TERATOGENIC EFFECTS
     Murray et  al.  (1980,  unpublished) performed a subchronic inhalation study
to determine the effects of ethylene dichloride (1,2-dichloroethane, EDO on the
reproductive ability of rats and effects on their offspring.  Tne results of this
study were reported at  the fifth annual Banbury Conference (Rao et al.,  1980).
Twenty to  thirty  male  and  female Sprague-Dawley rats were initially exposed to
25, 75,  and 150  ppm  EDC  (lot  no.   TAC06228, analyzed  as 99.98* pure  by  Dow
Chemical Laboratories)  for 60  days  (6  hrs/day.  5  days/week for 12 weeks),  and
they  were  successively  bred  to produce  two  litters,  the F..  and  F1g.   The
breeding  protocol  consisted   of caging  one  male  and  one  female  rat  for  4
consecutive days,  and  remating any  females  that  had not mated (no  sperm  in
vaginal smear) with a different male after a 3 day rest period.  The results from
remated animals were combined  with those of the first breeding.  The females were
exposed  to EDC throughout  breeding, gestation and  lactation  (6 hrs/day,  7
days/week)  except  for day 21  of gestation to  day  U postpartura to  allow  for
delivery and rearing of the young.   A second mating, for the production of the F
generation, took place after  the F    litter was  sacrificed (day 21 of birth).
     During the seventh week  of this study,  both  control and treated animals
suffered from  symptoms of sialoacryoadenitis (red  crusty material around  the
eyes  and nose and conjunctivitis),  which was thought to  have resulted  from a
viral infection.  The authors  noted  that more  males than females contracted the
disease, but  suggested that this greater  incidence  was  related to the housing
accommodations  (males  were caged  away from  females) and  not to any greater
susceptibility  of  the  males for developing  infection after  EDC exposure.   All
                                     9-122

-------
 animals  (both control and experimental)  recovered one to  two weeks after  the



 onset of  the  disease with no recurrence for  the duration of the  study.



     Murray et al.  (1980) reported that the  physical well being  of  the  parental



 generation was not  significantly affected by EDC exposure.   However, during  the



 first and second weeks of exposure,  the females consumed significantly less food



 than the  controls,  although for the rest of the study,  there were no alterations



 in  food  consumptions.   In  the males,  food  consumption was both  sporadically



 increased and decreased  throughout the study with  a general  trend for increased



 food consumption in males exposed to the highest concentration of EDC (150 ppm)



 for longer durations of  exposure (weeks 15 to 29).




     There was a statistical increase in the absolute liver weight at the 150  ppm



 level, and a statistical linear trend for increased liver weights in adult male



 rats exposed to EDC (Murray et  al., 1980).  However, this trend was  not  observed



 for liver weights  in  relation to body weight.   All adult  males displayed some



 degree of chronic renal  disease.  However, the severity of  the disease did  not




 appear  to be  related to  treatment.   Both control  and experimental  females



 displayed signs of renal  disease.  There was  a greater incidence of  inflammation



 and edema of  the salivary glands  in males  exposed  to 150  ppm.   Three deaths



 occurred during the course of the study, but these deaths did  not appear to be



 related to EDC exposure.




     The  fertility and  survival  index  on days   1,  7,   14,  or  21   was  not



 statistically different at any dose level.  However, the greatest pup mortality



was observed in groups exposed  to 150 ppm between days 14 and 21.  The sex ratio,



average number  of  live  pups  per  litter  and  neonatal  body  weight  were  not



affected.    There   were  sporadic,  statistically  significant  differences  in
                                     9-123

-------
reproductive  indices of treated versus control groups.  However, these did not



appear to be attributable to EDC exposure.  A few neonates exhibited  external and



internal malformations.   However,  the incidences  of these  effects were sporadic



and  statistically  insignificant,  reflecting  natural variability  and  not the



result of EDC exposure (Murray et  al.,  1980).  Therefore,  it  was concluded that




EDC  exposure,  under these conditions, did not adversely affect reproduction in



rats or the development  of  their offspring.




      Lane  et  al.  (1982)  evaluated  the  effects  of  EDC  on  the  reproductive



capability of mice  using a  multigenerational reproductive  protocol  modified for



screening teratogenic  and dominant lethal effects.   Animals were exposed to EDC



for  35 days, then 10 male and 30 female mice (parental generation FO) were mated




to produce  the first  set offspring (FIA)«   After  the F.. were weaned,  the FO



adults were remated  to produce the second set offspring (P-m)*  * parental stock



was  chosen from  the  FIQ  litter  (30  females,  10  males)  to  produce  a  second



generation of offspring ^y^'  After weaning,  the FIQ adults were remated to




produce  offspring  (F2B^  for use  in  teratology and  dominant lethal screening



tests.




      In this  study,  the  EDC  (Aldrich  Chemical Co.,  99$ pure)  was dissolved in a



1$ solution of Emulphor  EL-620  (GAF Corp.,  Linden,  NJ)  and then further diluted



with  deionized water to  concentrations of 0.03, 0.09, and 0.29 mg/mJl for nominal



doses  of approximately 5,  15,  and  50  mg/kg/day.    The  test  animals  were



continuously  maintained  on  EDC solutions or control solution  (deionized water,



or water containing 0.17 mg/mZ p-dioxane dissolved in  1.0$ Emulphor solution).
                                     9-121

-------
Fresh drinking solutions were  prepared  twice weekly and placed  in amber glass



bottles with  cork stoppers and  stainless steel  drinking  tubes.   The authors



reported no decrease  in fluid consumptions  in either the  EDC or  the solvent



control groups.




     Lane et al.  (1982)  reported  that EDC  produced no treatment-related signs of



toxicity such  as  lowered  body weights  or gross pathological  changes  in major



organs.   There  were  no  significant  differences  in  the  fertility  index  or




gestation index observed in the FIA,  FIB  or F?A  generations.  There were sporadic



incidences of increased mortality throughout the generations but these were not



dose-related;  the reason for the deaths was not  apparent at necropsy.  There were




no differences in the  litter sizes at birth, pup body weight, or survival of pups



at days H and  21  of birth.  There was a decrease in the survival indices for the




F2A Seneration as compared to  values for  the F ,  and FIQ generation, but these



decreases were not dose related.   In the dominant lethal screening tests, there



were no statistically  significant dose-related  effects.  However, both increases



and decreases  were  observed  in  the  ratios of  dead to live  fetuses.    In  the




teratology screening,  continuous administration  of EDC  produced  no  apparent



adverse reproductive  effects  or  observable abnormalities.   It  should  be noted



that continuous exposure used  in this  study is not a  recommended protocol for



evaluating  teratogenic  effects.    In  teratology  testing,  the  duration  of



exposures is usually limited  to the  period of organogenesis.  Also in the study,



the F-c skeletal  specimens  were lost and,  therefore, these  effects could not be



evaluated.



     From this  study  (Lane  et al.,  1982),  it  is not  possible  to  determine



conclusively whether  EDC  has  the  potential to cause  adverse  reproductive  or



teratogenic  effects  because the doses were not high enough to produce any adverse
                                     9-125

-------
effects,  including  maternally toxic effects.  However, under the conditions of



the experiment,  it  appears that EDC given in drinking water (nominal doses of 5,



15, 50  mg/kg/day)  produced  no observable adverse reproductive effects in Swiss



ICR mice.



     Schlachter  et  al.  (1979)  conducted  teratology  testing  in 16 to 30 Sprague-



Dawley  rats and  19 to 21 New Zealand White rabbits.   The animals were exposed to



inhalation  to either  100 or  300  ppm EDC  (Lot  Nos. TAC201851L  and TA022&51L,



analyzed as >99.9$ pure by the Dow Chemical Laboratories).   The rats were exposed



7 hrs/day on days 6 through  15 of gestation.  Two thirds of the maternal rats died



when exposed to 300 ppm EDC in contrast to no deaths occurring in  the control and



100  ppm groups.    In  the 300 ppm  group,  only  one rat  in six  survivors had



implantation sites; all of these sites were resorbed.  Two  additional rats at the



300 ppm level  showed early  implantation sites when  the uterus was stained with



sodium  sulfide.   In the  100  ppm  group,  three rats  delivered early on  day 21;



however, there was  no  indication of embryo or fetotoxicity in the offspring.  In



addition, the rest  of  the rats at  the 100 ppm level delivered normally with no



alterations  in  litter  size,  number of  resorptions  or  fetal body measurements



(crown  to rump length).



     At the 300  ppm level, one of  the six surviving rats had a decrease in mean



body weight  gain,  while  in  the 100  ppm level,  there was  an  increase  in body



weight  on days  6,  8,  10, 16, and 21 of gestation.   At  the 300 ppm level, the



absolute  liver  weight was   decreased,  while  the   relative  liver weight  was



increased; at the  100  ppm level there was no difference.  In the 100 ppm group,



there was an increase in water consumption on days 15-17 and  18-20 of gestation,
                                      9-126

-------
but food consumption was not altered.  There was  no increase in the occurrence of



malformations with only isolated  sporadic  incidents observed within the range of




normal variability.



     In this teratology study (Schlachter et al.F 1979), four out of twenty-one



rabbits in the 100 ppm group, and three out of  nineteen rabbits  in the 300 ppm




group  died.   Three control  rabbits  delivered  early (days  15,  28, and  29 of



gestation) as did one  animal at  the 300 ppm level (day 28 of gestation).   Two



animals  that  died at  the 100  ppm  level  also  aborted on days  26 and  28 of



gestation.  There was no effect on mean litter size,  incidence of resorptions or



fetal  body measurements (crown to rump  length)  in animals delivering normally.




Staining of the uterus  revealed only one additional implantation site in control



animals.  The mean body weights of pregnant animals were generally comparable to



those of controls, but  there was  an  increase in mean body weight gains in the 300



ppm  group  on  days  19  through 28  of  gestation.    A slight,  statistically



insignificant increase  in  both absolute and relative liver weights were observed




in both the 100 and 300 ppm  groups.  There was no increase in the incidence of



malformations,   although  sporadic   occurrences  within  the  range  of  normal



variability were reported.  It was  concluded from this  study that EDC exposure



does not produce adverse effects on the developing conceptus.



     Several Russian  studies  by  Vozovaya (1971,  1975,  1976,  1977) have reported



that  EDC  produces a   number  of adverse  reproductive  outcomes  which  include



lengthening the total estrous cycle,  changes in the duration of various stages of



the  estrous  cycle, increases  in  the number  of perinatal  embryonic  deaths,




decreases in the weight of newborn  animals, and decreases in the weight gain of



offspring after birth.   In addition,  EDC was   reported  to  concentrate  in the
                                     9-127

-------
placenta,  amniotic fluid  and  fetal  tissue.  However, It Is difficult  to  evaluate



or  seriously consider these  reports since they  are presented with insufficient



detail  or critical scientific  review.   Inadequacies in  these reports  include:



lack  of original  data, various techniques and tests mentioned but  results not



presented,  no information on the source or purity  of  the chemical,  statistical



analysis mentioned, but the type of statistical  tests  not stated.



      The  above  comments  apply  similarly  to  the study  by  another  Russian



scientist,  Urusova  (1953),  who reported  that EDC  accumulated in  the  milk of



nursing mothers.  This is obviously an important observation.  However, it cannot



be  accepted as scientific evidence. This  report was presented  in a  subjective,



anecdotal  manner  with many  inadequacies  in reporting.    These inadequacies



include lack of  details concerning the numbers  of  women  involved in the study,



the women's  age  or physical status,  the  concentrations and  duration of EDC



exposure after dermal  exposure, the repeatability of this finding and number of



samples analyzed.



      Because of  the concern  for  EDC contamination  in mother's milk, Skyes and



Klein (1957)  conducted a  study to determine whether oral administration of EDC



might pass into  the milk of  cows.  Only five  cows  were used in this study; two



cows were exposed  to 100 ppm EDC for 22 days;  two cows were  administered 500 ppm



EDC for  10 days followed by 1000 ppm for 12 days;  one cow was used as the control.



Two different breeds of  cows,  Holstein  and Jersey, were used, but  it  was not



stated which  cows  received which dose.   Seven milk samples collected over a three



week  period  containing no more than 0.4 ppm EDC.   However, the authors stated




that  this  amount  was  too low  for  accurate detection  by the Volhard titration



method  used  in this study.   The samples collected  from  cows ingesting  EDC had



consistently  higher concentrations  of EDC  than the  controls, but because of the
                                      9-128

-------
 small sample size and the lack of sophisticated analytical procedures,  it  is not



 possible  to make firm  conclusions  as to the  amount of EDC  entering the milk



 supply.  More importantly, this study was not designed to evaluate the possible



 health consequences of EDC ingested in the milk of nursing offspring.  However,



 recent pharmacokinetic studies do indicate that EDC has a tendency to accumulate



 in  fatty tissues  (Spreafico et  al.,  1980) although the  reproductive   health



 effects of EDC in breast milk have not been evaluated.



 9.3.1.    Summary and Conclusions.  The available scientific information  on the



 potential of ethylene dichloride  (EDC) to affect  reproductive or developmental



 processes adversely  Includes a teratology  test  conducted in  rats  and rabbits



 (Schlachter et al.,  1980),  a single-generation reproductive  test  conducted in



 rats (Murray et al.,  1980),  a multigenerational  reproductive  test conducted in



 mice (Lane  et al.,  1981), several  reports  by  Russian scientists  describing a



 number of adverse  reproductive effects observed in  laboratory animals and man



 (Vozovaya 1971,  1975, 1976,  and  Urusova,  1953),  and  an investigation measuring



 the amount of EDC  found  in the milk of cows after ingesting EDC (Sykes and Klein,



 1953).



     The results  of  reproductive  and teratogenicity testing  (Murray et al.,



 1980, Schlachter  et  al., 1979; and  Lane et al.,  1982)  indicate that EDC has



 little potential  for producing adverse  reproductive affects  or  for  adversely



 affecting the developing conceptus, except when the  mother  is exposed to doses



 high enough  to  produce  maternal  toxicity.   The  Russian studies  describing a



 variety of  adverse reproductive  effects  associated   with EDC exposure  do not



 provide conclusive scientific evidence because  the   studies  were  inadequately



 performed or reported.   The study of EDC in the  milk of cows  is inadequate since



only a  few animals were  used  and the biological effects of EDC-contaminated milk



were not investigated.
                                     9-129

-------
     In conclusion, the available studies indicate that EDC has little ability to



affect  the  reproductive or  developmental  processes  adversely  in  laboratory



animals  except at maternally  toxic levels.   However,  it should be  noted that



these studies have inadequacies that weaken the strength of this  conclusion.  The



multigenerational  study  by  Lane et al., 1982 does not include a range of doses



that include maternally toxic doses.  The study by Murray  et al.  (1980) does not



evaluate reproductive effects  for more than a single generation.  The studies by



Sykes and Klein (1953) on the effects on lactation are inadequate.  In addition,



no properly conducted epidemiological study has been done to evaluate  the effects



of EDC on human reproduction.   Therefore,  although the studies to date indicate



that  EDC does not  pose a  specific hazard  to  reproductive  or developmental



systems, this cannot be conclusively established,  especially for humans, without



additional studies.  The chemical similarity of EDC to ethylene dibromide (EDB)



and  1,2-dibromochloropropane   (DBCP)  might suggest  that  additional  testing



related to testicular toxicity  would be appropriate.
                                     9-130

-------
9.1.  MUTAGENICm



     A review of the  literature indicates that ethylene dichloride (EDO has been



tested for mutagenic activity in  bacteria,  plants,  Drosophila,  mammalian cells



in  vitro,  and  intact  rodents.    These studies  are  discussed  below and  are



summarized in  Tables 9-27 to  9-32.    The  reader may also refer  to published



reviews of the mutagenic potential of EDC (e.g., Fishbein,  1976,  1979; Fabricant



and Chalmers, 1980; Rannug, 1980b; Simmon,  1980).



9.4.1.  Gene Mutation Studies



     9.1.1.1.   BACTERIAL TEST SYSTEMS —  Many investigators  have studied the



ability of EDC to cause gene mutations in bacteria (Table 9-27).  Most reported



marginal positive  responses,  approximately two-fold  above background,  without



metabolic activation and  stronger  positive  responses with exogenous  hepatic



metabolic activation, indicating  that EDC is mutagenic to bacteria  by itself but



that  metabolites,  such  as  S-(2-chloroethyl)-L-cysteine,  are  more   potent



mutagens.



     Ethylene dichloride has  been  reported positive in four Salmonella/microsome



plate incorporation assays (McCann et al.,  1975; Rannug 1976;  Rannug and Ramel,



1977; Rannug et al.,  1978), in assays testing the mutagenicity of bile obtained



from EDC-perfused rat livers or livers from EDC-treated mice (Rannug and  Beije,



1979), and in  two  Salmonella spot tests.  In one of the  Salmonella spot tests



(Brera et  al., 1971), an analysis of duplicate experiments carried  out on at least



three different occasions  revealed a twofold increase in  revertant counts  for



strains TA1530  and  TA1535  (mean values  of 50 and 51 revertants  on treated  plates



versus 23 and  26 in control plates  for TA1530 and TA1535, respectively).   No



difference  in revertant counts was noted for  strain TA1538.  This response is



consistent  with that  expected for  an  alkylating agent.  The authors stated that
                                     9-131

-------
                                           Summary or Kutagenlcity Testing of EDC:   Gene Mutations  in  Baateria
Reference
Bren et al.,
1971
Teat
System
Salmonella
(spot teat)
Strain
TA1530
TA1535
TA153B
Activation
System
Hone
Chemical
Information
10 |imol on filter disk
Source: Not given
Purity: Not given
Results Comments
Weak positive 1. Could not perform
plate incorporation
testa because of
volatility.
Principe et
al., 1981
                                                                                        Salmonella revertants
Salmonella/
mammalian
mlcrosooe
assay (spot
teat)
                   S.. coellcolor
                   forward mutation
                   assay to Str
                   A. nldulans
                   forward mutation
                   to 8-AGr
                                          TA1535
                                          TA1537
                                          TA1538
                                          TA98
                                          TA100

                                          p<0.018





-59
0
39
17
19
82
188

EDC
Water
Chloramphenicol
Dose u£/plate
+S9
100 0
16 33
8 10
12 21
83 77
188 171
Dose iit/plate Survival t
0
2
10
20
100

0
250
500
100
100
100
100
100

100
100
42
TA1530 Ml 535
50 51
.23 26
20 31


100
103*
a
27
81
169
Strr /plate
2.5 + 0.6
0.2 7 0.2
0.7 T O.U
0.7 * 0.1
1.0 * 0.6
8-AG/plate
2.5 i- 0.9
2.0 + 1.1
1.0 7 0.7
TA153B
19
19
14

















1.  Positive controls
    indicated system
    working properly.

2.  Positive results In
    Salmonella in spot
    test with TA1535.

3.  Mo precautions taken
    to prevent excessive
    evaportlon of EDC
    and ensure adequate
    exposure.

1.  Toxicity results
    indicate exposure
    was minimal.

-------
                                                                   TABLE 9-27  (cent.)
Reference
McCann et al.,
1975









Test
System
Salmonella/
nlcrosome assay
(plate teat)








Activation
Strain System
TA100 PCB-induced
rat liver S9
mix








Chemical
Information
Concentration Tested:
1.3 * 104 Mg/plate
(13 umol)

Source: Aldrlch
ChBDlcal Co.

Purity: Not given,
but stated
to be highest
purity
Results
Negative or at
best only marginal
positive response.
Induced 25
colonies plate
above background.
(0.19 revertants/
linol) In TA100.



Comnents
1. Non-mutagenlc In
this study.
Reproducible dose-
response curves not
obtained.

2. Metabolic activation
did not increase
positive response.

3. Chloroethanol and
Rannug,  1976
Salmonella/
nlcrosome assay
(plate teat)
TA1535    Liver fractions
          from Sprague-
          Dauley or R
          strain Wlstar
          rats Induced
          with phenobar-
          bltal with and
          without NADPH-
          generatlng syste
          and with and
          without gluta-
          thlone S-trans-
          f erases A, B
          and C.
Concentration tested:
Up to 60 umol/plate

Source:  BDH Chemicals, Ltd.

Purity:  Not given but
         reported to be
         checked by glass
         capillary column
         chromatography
         using a flame
         loniiatlon detector
Marginally
positive
without activation
(two-fold In-
creases positive
response with
activation (ten-
fold increases).
Spontaneous back-
ground 8-14
revertants/plate.
    chloroacetaldehyde
    (two putative
    intermediates in the
    metabolism of EDC In
    mammals) tested
    positive (i.e., 0.06
    and 716 revertanta/
    umol, respectively).

1.  EDC activated by the
    liver cytosol
    fraction; mixed-
    function oxygenases
    not Involved.

2.  NADPH-independent GSH
    S-transferase dependent
    activation.

3.  Strain differences noted
    In ability to metabolize
 •   EDC.

M.  Thought that mutagenlclty
    of EDC after activation
    caused by formation of
    highly reactive half
    sulfur mustard,
    S-(2-chloroethyl)-L-
    cystelne.

-------
                                                                            TABLE 9-27 (oont.)
Reference
Rannug and Ranel,
1977








Teat
System
Salmonella/
Blcroaoue assay
(plate teat)







Activation
Strain System
TA1535 S9 nix from
livers of un-
induced male
R strain
Wlstar rata
plua NADPH-
generatlng
system


Chemical
Information
Concentration tested:
Up to 45 |imol/plate

Source: BDH Chemicals, Ltd.

Purity: Not given




Results
Positive reaponae
(two- fold Increaae
without activation;
nearly ten-fold
Increase with
activation.)
Negative control a
yielded roughly
15 revertanta/
plates.
Comments
1. Compared mutagenlcity
of EDC tar with EDC.
The level of EDC
present at the highest
dose tested for EDC
tar would only exert
a weak mutagenlc
effect, yet a strong
reaponae was
observed .
         Rannug et al.,
         1978
Salmonella/
mlcrosome assay
(plate teat)
TA1535
ui
         Rannug and Belje,
         1979
Salmonella/
Isolated
perfused
rat liver
TA1530
TA1535
Rat liver
mlcrosone
system with
and without
NADP
Isolated
perfused liver
from male R
strain Wlstar
rata
Concentration tested:
15 umol/plate

Source:  EDC given BDH
         Chemicals, Ltd.

Purity:  Not given for
         EDC, but checked
         using glass
         capillary chroma-
         tography using
         a flame ioniza-
         zation detector

Concentration tested:
0.1 ml (1.3 mM)
for up to 4 hours.
Marginal positive
response without
activation (two-fold
increase 24.8 +
3.06 at 15 umol
vs. 13.0 + 1.76
control);
positive reaponae
with activation
(ten-fold Increase)
Independent of
presence of NADP.
Positive.  Highest   1.
response 15-60
minutes after
addition of EDC
(45-60 revertanta
compared to 7-10
in controls).
Activation of EDC
tar dependent on
NADPH.  EDC activa-
tion Independent
of NADPH.

EDC major component
of EDC tar.
Hutageniclcity of
tar Increased with
metabolic activation
only with addition
of NADP.

Mutagenlcity of EDC
tar not solely due
to EDC.
Positive
responses consis-
tent with conju-
gation of EDC
with glutathlone.
         Rannug and Beije,
         1979
Salmonella/
(plate test)
TA1535
Bile from male
CBA mice
80 mg/kg EDC l.p.;
removal of liver and
collection of bile 30
and 60 minutes later.
Positive. Greater
than two-fold
increases with bile
from liver removed
30 minutes after
addition of EDC
(28.8 + 27 rever-
tanta compared to
11.3 + l.D.

-------
                                                                            TABLE  9-27  (cent.)
Reference
King et al.,
1979







Test
System
Salmonella/
nlcroaooe assay
(plate test)

E. coll K 12/
3"»3/ll3
(suspension test
and intrasangulneous
host-mediated assay)
Strain
TA1535
TA100
TA1537
TA153B
TA98




Activation
System
PCB-lnduced
rat liver S9







Chemical
Information
Concentration bested:
36 umol/plate


10 mM (suspension assay)

2 mmol/kg l.p. injection
female NMfll mice

Results
Negative



Negative

Negative


Comments
1 . Standard plate
incorporation test
was conducted. No
precautions were
taken to prevent
excessive evapo-
ration of EDC.


         Nestnan et al.,
         1980
vO
 i
Salmonella/            TA1535    PCB-lnduced
nicroaone assay        TA100     rat liver S9
(plate test and        TA1537    mix.
and desiccator         TA1536
exposure).             TA98
Source:  Herck Co.
         Darmstadt, FRG

Purity:  Not given.
         Stated that samples
         had correct melting
         point and elemental
         analysis.

Concentration tested:
Up to 9 mg/plate (91
IIDOI) In desiccators.
10 mg/plate In plate tests.

Source: Chem Service

Purity:  Not given

Solvent:  DHSO
Negative in
standard test.
Positive in
desiccator testing
in strain TA1535.
1.  Stated that
    maximum yield with
    TA100 is 20
    revertants above
    background (i.e.,
    negative).  For
    TA1535 a doubling
    of mutant colonies
    observed.  No
    other data pre-
    sented.

2.  Cannot adequately
    evaluate results.

-------
TABLE 9-27 (cont.)
Teat
Reference System
Stolzenberg and Salmonella/
Hlne, 1980 micro3onie
assay (plate test)






Activation
Strain System
TA100 PCS- induced
rat liver S9
nix (2 ng
protein/0.5 ml)





Chemical
Information
Concentration tested:
Up to 10 |imol/plate

Source: Aldrich
Chemical Co.

Purity: 99% pure


Results
Negative








Comments
1. All compounds
tested In trip-
licate with and
without S9 mix.

2. Experimental
values minus
background
revertants.
Revertants





Barber et al . , Salmonella/
1981 oicrosone assay
(vapor exposure)








TA1535 PCB-induced
TA100 rat liver S9
TA1538 nix
TA98







Concentration tested:
Up to 231.8 (imol/plate
as determined by GLC
analysis of distilled
water samples.

umol/plate -S9 +S9
10"1 0 0
1 0 0
10 15 No
Toxic
Negative in
standard plate.
test. Positive
in desiccator
testing in strains
TA1535 and TA100.



growth

1 . Bacteria exposed in
gas tight exposure
chambers .

2. Plastic plates
round to absorb
   Source:   Eastman Kodak Co.
   Purity:   99.98*
    dibromomethane
    In parallel
    experiment.  Thus,
    glass plates used
    Tor all other*
    testing.

3.  Weak positive
    result.  0.002
    revertanta/nmol
    in TA1535 with or
    without activation.
    0.001 revertants/
    nmol in TA100 with or
    without activation.

4.  Revertants selected
    from each experiment
    and tested to ensure
    that they were
    actually his .

-------
plate incorporation tests could not be performed because of the volatility of the



test  agent.    Positive and  negative  control  tests  were  conducted  for  these



experiments and indicated the systems  were working properly.   Principe et al.



(1981) conducted  a spot test using  strains TA1535,  TA1537,  TA1538,  TA98,  and



TA100.   A  positive response  was observed  for  TA1535 when a  triangular shaped



paper disc soaked with  100 u£ EDC  was  placed on the agar in the presence of S9



from Aroclor 1254-induced rat liver  (i.e.,  103  revertants on  the treated plate



vs. 33 revertants  for  the negative control).   Negative  responses were obtained



with  the other  strains.    A negative response  was  also  obtained  in  plate



incorporation tests conducted with TA100 and TA1535  at doses up to 100 [ill/plate.



A standard assay was conducted;  no precautions  were taken to  prevent excessive



evaporation of  the EDC.   Similarly in  forward mutation tests  conducted  with



Streptomyces  coelicolor  and Aspergillus  nidulans  negative  responses  were



obtained at doses up to 100 and  500 uH/plate in plate  incorporation and  spot



tests.  There was  a 100$ survival in the test conducted with  ^. coelicolor and a



60$ reduction  in cell survival at the highest dose with A. nidulans.  Thus, these



tests are  judged  to provide less  than adequate  conditions for  assessing  the



mutagenicity of EDC because insufficient exposure  to the  test organisms may have



occurred.



     McCann et al. (1975)  exposed Salmonella strains TA1525, TA100,  and TA98 to



EDC (Aldrich, stated to be highest purity available)  concentrations as high as 13



rag/plate (131  (imol/plate).   An  equivocal  response was observed  in  TA100 (0.19



revertants/umol).   However,  reproducible dose-related response  curves were not



obtained.  The presence of an exogenous S9 mix metabolic activation system (from



rat livers induced with either phenobarbital or Aroclor  1254)  did not increase



the response.   Chloroacetic acid,  which is a known mammalian metabolite of EDC,



and the putative intermediates  (i.e., chloroethanol and  chloroacetaldehyde;  see
                                     9-137

-------
section 9.1.3.3A), were also tested for their tnutagenic potential.  Chloroacetic



acid  was  negative,  but  chloroethanol  yielded  a  marginally  positive  and



chloroacetaldehyde  a strong positive  result  (0.06  and  7^6  revertants/nraol,



respectively, in TA100).  The authors  speculated that the mutagenic response of



EDC  may  be  due   to relatively  inefficient  conversion  of chloroethanol  to



chloroacetaldehyde in the in vitro  system.



     Two   reports   (Rannug,   1976;  Rannug   and   Ramel,   1977)   compared  the



mutagenicity  of EDC  with that of EDC  tar, a  complex  mixture formed  during the



manufacture of vinyl chloride from either acetylene,  ethylene,  or a mixture of



the  two.   The mixture  is called  CDC  tar  because EDC  is  a major  component



comprising about 30% of  the  mass.  The sample of EDC tested in these studies was



from British  Drug  House (BDH) Chemicals, Ltd.  (purity not given).   Salmonella



tester strain TA1535 was dosed up to 45 ^mol/plate in  the presence or absence of



an exogenous  metabolic  activation system (from livers of  male  strain  R Wistar



rats)  with  and without NADP.    Positive  (twofold) increases  in  revertant



frequencies over background  (13*0 +  1.76) were  observed in the plates receiving



the highest dose (21.8 + 3*06) in tests conducted without metabolic activation.



However,  with activation,   a  stronger positive  response  was  observed.    The



revertant count was elevated tenfold over  the spontaneous level (132.8 + 8.35 vs.



15.5 + 1.21).  The response  to EDC  was independent of the  presence of NADP; in



contrast, the mutagenicity of EDC tars was increased with metabolic activation



only in the presence of NADP.  It  was concluded  that the mutagenicity of EDC tar



cannot be  ascribed  primarily to EDC,  because  EDC and EDC  tar  have  different



requirements  for metabolic  activation.  In addition,  the  concentration of EDC



present  in EDC tar  at  the  highest  dose tested  would  yield only a  marginal



positive response if tested  alone, while in fact a strong response was observed



for the EDC tar.
                                     9-138

-------
     Following these observations, Rannug et al.  (1978)  tested Salmonella strain



 TA1535 with EDC  (BDH Chemicals  Ltd.;  although  the purity was not reported, the



 material was checked by glass capillary chromatography using a flame ionization



 detector) at doses up to 60 iiraol/plate.  Tests were conducted  with and without an



 exogenous  rat  liver   metabolic  activation  system.    The  components of the



 activation system were varied in  order  to study  the mechanism of activation of



 EDC.   Phenobarbital-induced  liver fractions  from Sprague-Dawley or  R strain



 Wistar rats were  prepared with  and without an  NADPH-generating system and with



 and without glutathione transferases  A,  B, and C.  It was observed that EDC is



 activated  by  the cytosol fraction of liver homogenates;  thus,  mixed-function



 oxygenases are not involved.  The activation was NADPH-independent but required



 glutathione (GSH) A or  C S-transferase enzyme activity.  The extent of activation



 was found to be dependent on the rat strain used (liver fractions from R strain



 rats were more effective than those from Sprague-Dawley rats)  and the handling of



 the extract (e.g., storage on ice or  freezing  reduced the effectiveness of the



 activation system).  The report by McCann et al.  (1975)  (described  earlier in



 this section)  that EDC  was not metabolized  to a mutagenic intermediate may be due



 to  differences  in the exogenous  activation system used  (S9  mix rather  than



 cytosol fraction) in the two studies.  Rannug et al. (1978) hypothesized that the



 mutagenicity of EDC after metabolic activation was caused by the formation of a



 highly reactive half sulfur mustard,  S-(2-chloroethyl)-L-cysteine (see section



 9.1.3-3.B). This compound (>99? purity) was found to be more strongly mutagenic



 in TA1535 than is EDC.  At 0.2 umol/plate,  12.8 + 1.5 and 176.8 + 11.1 revertants



 were  observed   per plate  after   treatment  with  EDC  and  S-(2-chloroethyl)-



 L-cysteine, respectively.   At  5.0 |imol/plate the observed numbers of revertants



 per plate were 13.0 + 1.1 and 1951  (only one plate tested),  respectively.   Thus,



S-(2-chloroethyl)-L-cysteine gives a strong mutagenic effect at low doses where



no effect can  be seen  for  EDC.
                                     9-139

-------
     In intact mammals most GSH conjugates are normally excreted in bile.  Rannug



and Bieje (1979) reasoned that if EDC were raetabolically activated by conjugation



with glutathione  to  form a half sulfur mustard, rautagenic products in the bile



would  be produced in EDC-treated mammals  or  perfused  livers.   To  test  this



hypothesis, an  EDC concentration of  0.1  mi (1.3 mmol)  were perfused through R



strain Wistar  rat livers  for up to  1  hours.   Bile was collected right after



addition of EDC and  at 15 and 30 minutes,  as well as 1 „  1.5,  1.75, 2, 3, and 4



hours later.  The bile was then added directly to top agar or diluted about 5 to



10  times in sterile  water and added  to  top agar containing Salmonella strain



TA1535 for plate  incorporation tests.  Positive responses  were obtained.   The



greatest  response (15-60  revertants per  plate,  compared to  7-10 in negative



controls) was  reached about 15 minutes to  1 hour after  addition  of  EDC.   In a



second experiment,  80 mg/kg  EDC was given to male CBA  mice intraperitoneally.



The animals were  sacrificed,  their livers removed, and bile collected 30 and 60



minutes later for mutagenicity testing with TA1535 in plate  incorporation tests.



An increase in revertants  (greater than twofold) was  observed for bile collected



30 minutes after treatment compared to bile from negative control animals  (28.8 +



27 revertants and  11.3 ± 1.1  revertants,  respectively).   The positive responses



obtained  with bile  from  the  perfused  rat livers and intact  mouse livers are



consistent with  the  hypothesis that  EDC is activated by  conjugation with gluta-



thione.



     Four studies have been reported in which EDC was found to be negative in the



standard  Salmonella/microsome  assay plate  incorporation tests  (King  et al.,



1979;  Nestmann  et al., 1980; Stolzenberg and Mine,  1980; Barber  et al.,  1981).



The maximum  doses employed  in  the  first  three  studies  were  36 umol/plate,  91



umol/plate, and  10 ^mol/plate, respectively; the fourth study did not  report the



doses  used.   The doses  reported  in these  studies therefore  encompass a  range

-------
at which positive responses  have been reported in other studies.  Except for the



report  by   Stolzenberg   and  Hine  (1980),  the   negative   Salmonella  plate



incorporation studies were conducted with appropriate  positive controls.   Each



test was conducted with and without PCB-induced rat liver S9 mix.  It should be




pointed out, however,  that the positive controls, which require activation, were



not structually similar to EDC.   Therefore,  these  positive controls may not be



able to determine the effectiveness of the components in the S9 mix necessary for




EDC activation.   King  et al.  (1979) also reported negative responses when E. coli



K12/3^3/113 was  tested in  either a liquid suspension test or an intrasanguineous



host-mediated assay.



     Two of  the  negative  studies  (i.e.,  Nestmann  et al.,  1980;  Barber et al.,



1981)  reported  that although EDC  did  not induce  mutations in  standard  plate



incorporation tests,  positive responses  were obtained  when the studies were



conducted in air  tight  exposure   chambers.   Nestmann  et  al.  (1980)  exposed



Salmonella  strains  TA1535 and TA100 to  doses from 3  to 9 rag/plate  (30  to 91



umol/plate)  in desiccators.  It was reported  that this treatment yielded positive



results, at  least for TA1535 in  which  there was  a doubling  in  the  number of



mutant colonies  over the control.   It was stated that concentrations  were tested



up to levels where cell-killing was observed,  but no data are given and insuffi-



cient detail is  provided  to  allow  the results to be adequately evaluated.



     Barber   et al.  (1981) exposed Salmonella tester strains TA1535,  TA98,  and



TA100 to four levels of EDC  vapors (Eastman  Kodak  Co.,  99.98$ pure)  in a 3.1-



liter airtight exposure chamber.   These  exposures  resulted  in estimated  plate



concentrations ranging from  31.8 to 231.8 umol/plate as determined by gas liquid



chromatography  analysis  of  distilled water  samples  placed  in the  exposure



chamber.  Linear, dose-related increases in revertant  counts were observed for



TA1535 and  TA100.   The mutagenicity of EDC in these two strains was  0.002 and

-------
0.001  revertants/nmol,  respectively.   No  difference  in  revertant  counts  or
potency of EDC was noted in comparisons of tests done with and without metabolic
activation.  The  positive  results were obtained by  Nestmann  et  al.  (1980) and
Barber  et  al.  (1981)  only  when  tests  were  conducted  in  airtight exposure
chambers.  This suggests that  standard mutagenicity  testing of EDC (bp 83°-84°)
may  not  provide  an  adequate  assessment of  its mutagenic  potential due  to
excessive evaporation.
     In  summary,  the positive  responses  obtained without metabolic activation
indicate that EDC is a mutagen in bacteria.  The positive responses obtained with
metabolic activation indicate that one or more  of its metabolites  are more potent
mutagens.  The negative findings  reported in bacterial  tests are not considered
to contradict  the reported positive  results,  because the  negative  results may
have  been due  to excessive  evaporation of  EDC or to inadequacy  of the  S9
activation system.
     9.1.1.2.   EUCARYOTIC TEST SYSTEMS — In addition to causing gene mutations
in bacteria, EDC  also has been shown  to cause  gene mutations in eukaryotes.
     9.4.1.2.1. Higher  Plants —  Two  reports were  evaluated  concerning the
ability of EDC to induce mutations  in higher plants  (Table 9-28).  Ehrenberg et
al. (197*0 treated barley seeds (variety Bonus) with  30.3 mmol  EDC (Merck, purity
not  given)   for   24  hours  and scored  for sterile   spikelets at  maturity  or
chlorophyll mutations in about 600 spike progenies in the subsequent generation.
The dose  level tested  corresponds to  the LDg_.   EDC treated kernels  had an
increased  incidence  of  chlorophyll  mutations  (6.8$)  compared   to  untreated
controls (0.06$).
     Kirichek  (1974) exposed  eight  varieties of pea seeds (100 each) to gaseous
EDC (purity and concentration not given) for 4 hours.   The germination of treated
                                      9-142

-------
                                                                                TABLE 9-28

                                                          Summary of Hutagenielty Testing of EDC:  Higher Plants
Reference
Ehrenberg et al . ,
1974
Teat
System
Segregating
chlorophyll
(gene) mutations
in barley
Chemical
Information
Concentration tested:
200 seeds treated
with 30.3 0001/21
hours (LD,.) in a
closed vessel.
Results Comments
Positive response. 1. About 600 spike progeny were tested.
(6-8J mutants from
treated progeny vs.
0.06) mutants from
control progeny).
                                                                                  Source:  Nerck Co.
                                                                                           Darmstadt, PRG

                                                                                  Purity:  Not given
         Kiricheck, 1974
\o
 i
Visible mutations
In peas, 8 varieties
Concentration tested:
EDC (concentration not
reported) or water
(negative controls)
vapors for 4 hours.

Source: Not reported

Purity: Not reported
Reported postive.
5.42-28.13* of
seeds reported
to be mutated.
1.  English translation of Russian article.

2.  Germination of the treated seed varied
    with the variety tested from IS) to 58*
    compared to 100J germination of the
    control seeds.

3.  Control mutation frequency not given.

1.  Putative mutations not characterized.

5.  Not possible to adequately evaluate
    results.

-------
seeds  differed  with  the  variety  tested  but was  reduced  (15-50$ germination)



compared  to negative  control  seeds exposed  to water vapor for  4  hours (100$



germination).  From 5.4$ to 28.13$ of the  seeds  were  reported to be mutated.   It



is reported that two times as many morphological mutants  were induced as chloro-




phyll mutations.  The  author considered this  a  positive  response, but the muta-



tion  frequency  for  negative  controls was  not  given.   This limitation  of  the



report plus a lack of information concerning the characterization of the putative



mutations precludes  an adequate evaluation of the  results.



     9.4.1.2.2. Insects — Four studies were evaluated concerning  the ability of



EDC to cause mutations in  Drosophila melanogaster (Table 9-29).   These studies



demonstrated the  ability of EDC to cause sex-linked recessive  lethal mutations



(Shakarnis, 1969, 1970; King et  al.,  1979).   The  fourth study  demonstrated the



induction of somatic  cell mutations by EDC  (Nylander et  al., 1979).



     Shakarnis  (1969i  1970)  performed two experiments to assess  the ability of



EDC  (purity  not  given)  to  cause  sex-linked   recessive  lethal mutations  in



Drosophila.  In the  first  study (Shakarnis  1969), adult females  from  a radio-



sensitive strain (Canton S) were exposed to 0.07$ EDC gas  for 4 or 8 hours at 24°-



25°C.  Immediately after treatment the females were mated to Muller-5 or In wa B



males.  Fertility of  treated females was reduced 47$  by the 1-hour treatment and



91$ by the 8-hour treatment. The F_ progeny  were scored  for lethality (measured



as the absence  of wild-type males).   A statistically significant (p<0.05) time-



related increase in  the incidence  of  sex-linked recessive lethals was observed.



The  frequencies  of   lethals  were   0.3$   (negative  controls),  3*2$  (4-hour



treatment) and  5-9%  (8-hour treatment).   In his second study,  Shakarnis (1970)



exposed females from a radiostable strain (D-32)  of Drosophila melanogaster to



0.07$ EDC vapors (source and purity not reported) for 1 or 6 hours.  Immediately
                                      9-144

-------
                                                                                  TABLE 9-29

                                                    Summary of Mutagenlcity Testing of EDC:   Gene Mutation Teats in Insects
Reference
Shakarnia, 1969
Test
System
Drosophlla
nelanogaster
Chemical
Information Results
Concentration tested: Postive response
virgin 3-day -old
Comments
1. Huller-5 or In wa B males.
                                rsex-linked
                                 recessive lethal
                                 test
Canton S females
exposed to 0.079
EDC gas for M or B
hours at 2U-25°C.

Source:  Not given

Purity:  Not given
2.  Fertility of treated females
    reduced significantly (V7) after
    4 hour treatment and 91J after
    6 hour treatment).

3.  Canton S strain reported to be
    sensitive.
\o
 i
XT

-------
                                                                              TABLE 9-29 (cont.)
VO
 I
Test Chemical
Reference System Information Results
Shakarnls, 1970 Prosophliia Concentration tested: Positive response 1.
melanogaster virgin 3-day-old
sex-linked radios table D-32 2.
recessive lethal strain females
test exposed to 0.07* EDC
gas for 4 or 6 hours
at 24-25°C. 3.

Source: Not given

Purity: Not given
Duration of
Treatment
(h) with Number of
0.07* EDC Chromosomes Scored
Control 1904
4 2205
6 2362

Comments
Nuller-5 males.

Experiment conducted



twice for
4 hour exposure and three times
for 6 hour exposure.

4 hour treatment did
significantly reduce
but 6 hour treatment
by 50*.



Lethal Nutations
No. *
1 0.05
46 2.00
7B 3.30
(p<0.05)


not
fertility
reduced It











-------
                                                                              TABLE 9-29 (cont.)
so
 I
Test Chemical
Reference System Information Results
King et al., Drosophlla Concentration tested: Positive response 1.
1979 melanogaster 50 mM solution of EDC
sex-linked in 5% sucrose fed to 2.
recessive lethal 1- to 2-day-old Berlin K
test males for 3 days (near
Source: Merck Co.
Darmstadt, PRO
Purity: Not given but
stated to have
correct melting
point (SIC) and
elemental
analysis
Cone. Days after Number of
(mM) Brood Treatment Chromosomes Scored
0 I-III 0-9 22,048
50 I 0-3 1,185
II 4-6 1,179
III 7-9 156
I-III 0-9 2,520
Comments
Base females.
Lethal frequency Increased for all
broods. Greatest effect in brood
II, which corresponds to the
speraatid stage of spermatogenesis.
Lethal Mutations
No. \ p
47 0.21
6 0.51
41 3.48 <0.01
2 1.28
49 1.94 <0.01

-------
                                                                              TABLE 9-29 (cont.)
Reference
Nylander et al.,
1976

Teat
System
Drosophlla
melanogaater
somatic cell
mutations so
- |j Soie 19
stocks.
Chemical
Information Results
Concentration tested: Positive response
0.1J and 0.5* EDC In
food at 25°C and 75*
humidity during larval
development.
Source: Fisher
Scientific Co.
Comments
1. Mutations at £ locus scored in
files of stable and unstable
genotype. Both have same phenotype
Instability thought to be caused
by transposable genetic element.
2. Survival of treated flies reduced
significantly In both genetically
                                                      Purity:   Not  given
stable (342 reduction for 0.If and
77S reduction for 0.5% dose) and
genetically unstable stocks (86J
reduction for 0.51 dose).
I

oo
Treatment
Control
Stable
Unstable
0.01}
Stable
Unstable
0.05$
Stable
Unstable
Number
Hales
Scored

M441
5363

6260
2889

610
201
Number
with
Sectors

2
I

263
271

44
50
J

0.045
0.075

4.20
9.2B

7.21
24. 8B
t Value Differences
Within Genotypes

—
—

18.748
24.31a

11.463
13. 12a
t Value Differences
Between Genotypes


0.60


9.l6a


5.66a
            p<0.001

-------
after treatment they were mated to Muller 5 males and the F2 progeny scored for



sex-linked recessive lethals.   The  4-hour treatment did not significantly reduce



fertility but the 6-hour treatment reduced it by 50%.  As in the first study, a



statistically significant (p<0.05)  time-related  increase in sex-linked recessive




lethals was observed.  A  frequency  of 0.05% lethals was observed in the negative



controls, and frequencies of 2.00% and 3-30? lethals were observed in offspring



from the 4-hour and 6-hour  treatments,  respectively.  This  corresponds  to the



mutagenic effect induced  by  an  acute dose of two to three  thousand  (2000 to 3000)



rad of x-rays.



     King  et  al.  (1979) also  studied the ability  of EDC to  cause sex-linked



recessive lethals  in Drosophila.  One to two day  old  Berlin K  males were fed a 50



mM solution of EDC (Merck and Co.,  purity not given but correct melting point and



elemental analysis reported) in 5% sucrose for  3  days.   This dose approximated



the LD,-Q.  The males were immediately mated to virgin Base females every 3 days



for a  total of 9 days  to  determine if  EDC preferentially  damaged particular



stages  in  spermatogenesis.    Progeny of  the  F_ generation  were scored  for



lethality.  The lethal frequency was found to be  elevated for  all three broods



(0.51$, 3.48?, and  1.28$, respectively, for  broods  I,  II, and III) compared to



the negative controls  (0.21$).  The highest lethal  frequency (3.48$) was found in



brood  II  (p<0.01),  which  corresponds  primarily to  the spermatid  stage  of



spermatogenesis.




     Mylander et al.  (1979)  raised flies on food containing 0.1? and 0.5$ EDC



(Fisher Scientific Co., purity not given) during  larval development and scored F



progeny for  the induction of somatic cell sex-linked mutations in  the  eye.   A



genetically   unstable   stock   (so z w+)   and  a  genetically   stable   stock




(^ Dp w+  e  ) both having  the same phenotype  were used in these  studies and



mated to attached  X  females.  Mutations result in the expression of normal (dark



red) eye pigment in adult treated males.  The genetic instability of 50 £ w+ is






                                     9-149

-------
caused by the insertion of a transposable genetic element.  The survival of flies



raised on the EDC-treated food was significantly reduced compared to the negative



control values (e.g., 77$ reduction for the stable stock and 86% reduction of the



unstable stock at 0.5% EDO).  Statistically significant increases in somatic cell




mutations occurring  in both stocks were observed  at both  the  low and high doses



(p<0.001).



     The positive responses in Drosophila indicate that EDO is capable of causing



both somatic cell and heritable germinal mutations in a raulticellular eukaryote.



     9.1.1.2.3.  Mammals



     9.4.1.2.3.1. In Vitro.   Tan  and Hsie  (1981) found  that  EDC (Matheson,



Coleman and Bell, purity  not given)  caused  a dose-related  increase  in HGPRT



mutations in cultured Chinese hamster ovary  (CHO) cells (Table 9-30). CHO-K.-BH^



cells  were  exposed  in suspension  culture  for  a  period  of 5  hours  to  EDC



concentrations  ranging up  to 3 mM (30$ survival)  in  tests with exogenous rat



liver S9 mix for metabolic  activation and concentrations ranging up  to 50 mM (50%



survival) in tests  conducted without metabolic activation.   Positive responses



were  observed  both  in  tests  with   (5  mutants/10   cells/mmol)   and  without



(1 mutant/10  cells/mmol)  metabolic activation (p<0.01).  EDC was detected as a



mutagen in studies not requiring metabolic activation only at high doses, but the



induced mutation frequency was increased about tenfold over control values.  Only



about a fourfold additional  increase  in  mutagenicity  was noted with metabolic



activation  but  excessive  toxicity precluded  testing at concentrations greater



than 3mM (i.e.,  S9 mix  increased mutagenic activity by approximately  fourfold and



cytotoxic activity from 5  to 25-fold).  The  metabolic activation system was only



effective in the presence  of NADP.  This contrasts  with  Rannug (1976) who found



metabolic activation of EDC to  be NADP-independent.
                                      9-150

-------
                                                                                 TABLE  9-30

                                                         Summary  of Hutagenlclty Testing of  EDC:  Mammalian Syaterns
Reference
Tan and Hsie,
1981
Test
System
Chinese hamster
ovary cell HGPRT
gene mutation
assay
Chemical
Information
Concentration tested:
Up to 3 mM in tests
with rat liver S9 mix
and up to 50 mH in
Results
Positive response ,
60 vs. 3 mutants/10
clonable cells for
50 and 0 mM EDC
Comments
1. Mutagenic activity of EDC without and
with metabolic activation calculated
to be 1 and 5 mutants/ 10 cells/
mmol, respectively.
         Gocke et al., 1983
House Spot Test
C57Bl/6JHan (F)
     X
T-stock (M)
                                                    tests without
                                                    metabolic activation.
                                                    LD5Q is 1 mH with
                                                    activation and 6 mH
                                                    without activation.
One l.p. injection 10
days after conception
of 300 mg/kg EDC in
olive oil.
                                                    Source:
                              Merck Co.
                              Darmstadt, FRG
                                               without activation.>
                                               28 vs. 3 mutants/ID
                                               clonable cells for
                                               1.5 and 0 nM EDC
                                               with activation.
7/110H offspring
had spots compared
to 2/812 In the oil
controls and 3/2161
in the cumulated
controls.
2.  S9 nix increases mutagenlcity by
    about fourfold and cytotoxiclty
    5- to 25-fold.

3.  NADP required in S9 mix for
    metabolic activation.

1.  Data suggestive of a positive response.
ui
                                                    Purity:  Not given

-------
     9.4.1.2.3.2. In Vivo.   Gocke et al. (1983) conducted a mouse spot test to



evaluate the ability of EDC to induce somatic cell mutations in developing mice.



One  300  mg/kg intraperitoneal  injection of EDC  (Merck,  purity  not  given)  was



administered  to  C57  Bl/6JHan females  10 days  after mating with  T-stock males.



Seven out of the resulting 1104 offspring had mutant coat color spots compared to



2/812 in the  controls and  3/2161  in the combined untreated controls.   The data



show  a  significant  (p=0.03)  effect  against  the  cumulated  controls, but  no



significance  (p=0.l8) against  the oil control.  These data are suggestive of a




positive response.



     The consistency of positive  results obtained in  higher plants, Drosophila,



and  cultured  mammalian cells  demonstrates  that  EDC causes  gene  mutations  in




higher eukaryotes.



9.4.2.  Chromosome  Aberration Studies.  Five studies were evaluated  regarding



the  ability of EDC to cause chromosome aberrations (Table 9-31).  One was  an



abstract of testing  EDC  for its ability to  cause chromosome breakage  in Allium



root  tips  and  cultured  human  lymphocytes  (Kristoffersson,  1974).    Two were



Drosophila  melanogaster  X  chromosome  nondisjunction tests  (Shakarnis,  1969,



1970), and two  were  raicronucleus tests  (King  et  al.,  1979;  Jenssen  and Ramel,




1980).



     In  the paper  by  Kristoffersson (1974)  EDC (source,  purity,  and concentra-



tion not given) was reported to cause C-raitoses in Allium  root  tip cells but not



to  cause chromosome  breaks  in  Allium root  tip  cells   or  human lymphocytes.



Because  the report was an abstract,  insufficient  information was available to



substantiate  the reported results.  However,  the induction of C-mitoses  in Allium



root  tip cells  suggests  that  EDC  can  interfere  with  the mitotic spindle



apparatus.
                                      9-152

-------
                                                            TABLE  9-31

                               Summary  of Mutagenicity  Testing  of EDO:   Chromosomal  Aberrations Tests
Reference
Test
System
Chemical
Information Results
Comments
     Shakarnis, 1969
Drosophila
melanogaster
X chromosome
nondisjunction.
Canton S (F)
     X
Muller-5 (M)
Concentration tested:
virgin Canton S females
exposed to 0.07$ EDC
in 1.5 & desiccator
for 1 and 8 hours at
24-25°C.

Source:  Not given

Purity:  Not given
vo
.^
ui
Statistically
significantly
(p<0.05) increases
in exceptional
female progeny reported
for 4-hour exposure
and male and female
progeny for 8-hour
exposure.  However,
test Judged to be
invalid.
The study was poorly
designed in that the
Canton S stock did not
possess appropriate
genetic markers to
rule out non-virginity
as the cause for the
"exceptional" progeny.

The control rate was
much lower than the
historical background
rate.
0.07$ EDC
Duration of
Treatment
(h)
Control
4
8


Normal
No. Females
5,848
24,125
8,437


Progeny
No. Males
5,727
21,297
7,773

Exceptional
Females
No. $
0 0
8 0.033
15 O.l8a

Progeny
Males
No. $
0 0
3 0.01
7 0.093
      p<0.05

-------
                                                       TABLE 9-31 (cont.)
Reference
Test
System
Chemical
Information Results
Comments
    Shakarnis,  1970
vO
i
VJl
Drosophila
melanogaster
X chromosome
nondisjunction.
Canton S (F)
     X
Muller -5 (M)
Concentration tested:
3-day-old females
from radiostable D-32
strain exposed to 0.0751
EDC in a 1.5 £
desiccator for 4 or 6
hours at 24-25°C.

Source:  Not given

Purity:  Not given
Incidence of nondis-
junction greater in
treated group than
in control.  Increase
not statistically
significant.  Test
judged to be invalid.
See comments for
previous study.
0.07} EDC
Duration of
Treatment
(h)
Control
4
6
Exceptional Progeny
Normal Progeny
No. Females
2172
3584
4034
No. Males
2205
3401
4090
Females
No. %
0
1
4
0
0.03
0.10
Males
No. %
1
1
3
0.03
0.03
0.07

-------
                                                         TABLE 9-31 (cont.)
Reference
King et al. ,
1979
Test
System
Micronucleus test:
NMRI mice
Chemical
Information Results
Concentration tested: Negative
2 i.p. injections of
4 mmol/kg (400 mg/kg)
Comments
1 . 1 , 000 polychromatic
erythrocytes
analyzed/animal .
VJ1
                                                 given 2<4 hours apart.
                                                 4 animals/dose
                                                 sacrificed after
                                                 second  injection.
                                                Source:
                              Merck Co.
                              Darmstadt, FRG
                     Purity:  Not specified,
                              but melting
                              point and ele-
                              mental analysis
                              were correct
                                                                     2.  Frequency of micro-
                                                                         nuclei not given.
     Jenssen and Ramel,
     1980
Micronucleus test:
CBA mice
Concentration tested:
single i.p. injection
100 mg/kg.
                                                Source:
                              BDH Chemicals,
                              Ltd.
                                                 Purity:  Not given
Negative.
(0.15 + 0.11) poly-
cromatic erythrocytes
with micronuclei in
controls vs. 0.17 ±
0.10 in treated
animals).

-------
     Data perhaps consistent with this possibility, but not conclusively showing



non-disjunction, are found in studies by Shakarnis (1969,  1970) using Drosophila



melanogaster.   Females  were  exposed to 0.07$ EDC  vapors  (source and purity not



given).  In the first study (1969) a statistically significant (p<0.05) increase



in  exceptional  F..  progeny,  indicative  of meiotic nondisjunction,  was reported



after a 1-hour treatment (0.03$ exceptional females) and 8-hour treatment (0.18$



exceptional  females  and  0.09$  exceptional  males)  compared  to  the  negative



control  value  (0$; 0/11575  progeny).   In  the  second  study, the  incidence of



exceptional progeny was elevated  in the treated groups but not significantly so.



However, both studies were flawed by an experimental design which did not allow



discrimination  between  progeny resulting from nondisjunction compared to those



from non-virgin  females.



     Micronucleus  tests were  performed by King  et  al. (1979)  and Jenssen and



Ramel  (1980).    Both  studies  reported  negative  results.   King et  al.  (1979)



injected NMRI mice with  two  intraperitoneal injections of 4 mmol/kg  (100 mg/kg)



EDC  (Merck  Co.,  purity not  specified  but  melting point and chemical analysis



reported to be correct).   The  authors state this corresponds to an "approximate



lethal  dose"  (the LD,   for  intraperitoneal injections  of  EDC  in mice  is 250
                     lo


mg/kg).   The  injections were  given 24-hours apart;  the animals were killed 6-



hours after the second injection,  and bone marrow smears were made.  One thousand



polychromatic  erythrocytes  (PCEs)  were  analyzed per  animal.   Frequencies of



micronuclei were not given, but the results were evaluated to be negative by the



authors.



     Jenssen and Ramel  (1980)  also reported negative  results in a micronucleus



test.    CBA  mice  were  given  a  single  intraperitoneal injection of  EDC   (BDH



Chemicals  Ltds., England,  purity  not  given)  at  a  dosage  of  100  mg/kg.   The



animals were sacrificed the next  day and PCEs (number not given) were scored for
                                      9-156

-------
micronuclei.  The frequencies of PCEs with micronuclei  were  0.15 +. 0.14 in the



controls and 0.17 +_ 0.10 in treated animals.



     The equivocal X-chromosome  loss  test in Drosophila  (Shakarnis,  1969) may



suggest  that  EDC can  cause  meiotic  nondisjunction.     A  properly  designed




experiment  should be  conducted  to  determine  this.    The  negative  responses



obtained  in  the raicronucleus  tests  may  indicate  that  EDC  does not  cause



chromosomal damage in mice.   However, because  EDC has not been adequately tested



for  its  ability to  cause  structural  chomosomal  aberrations,  it  would  be



appropriate to perform mammalian in vitro and iji  vivo  cytogenetic tests.   Such



testing is required before a judgment can be made  on  the ability of EDC to cause



chromosomal aberrations.



9.^.3*  Other Evidence of DNA Damage.   Three other tests have been conducted on



the genotoxicity of EDC.  These tests do not measure mutagenic events per se in



that  they do  not demonstrate  the induction of  heritable  (i.e., somatic  or



germinal)  genetic alterations,  but  positive results  in  these  test  systems



suggest that DNA has  been damaged.  Such test systems provide supporting evidence



useful for assessing genetic risk.



     9.4.3.1.   BACTERIAL TEST SYSTEMS



     9.4.3.1.1. PolA Assay — Ethylene dichloride has been reported positive in



the polA assay which measures toxicity associated with unrepaired damage in DNA



(Table 9-32).   Brem  et al.  (1974) soaked  sterile filter disks  with  10  \ii (80



jijnol) EDC.  These were centered on the agar surface of petri dishes covered with



bacteria  (one  set with  polA* strain,  the other  with polA~  strain).   After



incubation the  plates were scored  for differential  inhibition of growth.  An 8-mm



zone of growth  inhibition  was  observed in the polA* strain  compared  to  a 9-mm



zone of  inhibition  for the  polA~  strain.    These  responses were said   to  be
                                     9-157

-------
                                                                               TABLE 9-32

                                                           Summary of Mutagenicity Testing of EDC:   PolA Assay
Reference
Bren et al . ,
1974
Test
System
polA differential
cell killing
assay
Strain
E. coll
polA
polA*
Activation
System
None
Chemical
Information
Concentration tested:
10 \it on filter disk
Results
Questionable
positive
Comments
1. All assays carried out In
duplicate on at least
three different occasions.
                                                                                                                              Only a small difference
                                                                                                                              in diameter (i.e.,  1  mm)
                                                                                                                              was noted between the
                                                                                                                              zones of killing in the
                                                                                                                              two strains.  Thus, the
                                                                                                                              A*/A~ ratio may be
                                                                                                                              misleading.
                                                                                               Pol A Zone of Inhibition
vO
CD


EDC
HHS
Chloramphenicol
A*
no
8
"45
28
A"
nun
9
51
28
A*/A~

1.26
1.44
1.00

-------
reproducible.  The ratio between the zones of inhibition  (polA XpolA") was  1.26



which  is interpreted by the  authors to  be  a positive response.   However,  it



should  be noted  that  1 mm is  not a  big difference,  and  the  ratio  may  be



misleading.



     9.U.3.2.   EUKARYOTIC TEST SYSTEMS



     9.1.3.2.1. Unscheduled  DNA Synthesis  — One  test has  been  performed  to



assess  the ability of EDC to cause  unscheduled DNA synthesis  (Perocco and Prodi



1981).   Although demonstration that a chemical causes unscheduled DNA synthesis



does not provide a measurement of its mutagenicity. per se, it  does indicate  that



the material damages DNA.  Perocco and  Prodi  (1981)  collected  blood samples  from



healthy humans, separated the lymphocytes, and cultured 5 x 10   of them in 0.2  m£



medium  for 4 hours at 37°C  in the presence or absence of EDC (Carlo Erban, Milan,



Italy  or Merck-Schuchardt,  Darmstadt,   FRG,  97%-99%  pure).    The  tests   were



conducted both  in  the presence and in  the absence  of PCB-induced  rat liver  S9



mix.  A comparison was made between treated and untreated cells for scheduled DNA



synthesis (i.e., DNA replication) and unscheduled DNA synthesis.  No difference



was noted between the groups with respect to scheduled DNA synthesis measured  as



dpm of  [3H]  deoxythymidylic acid (TdR) after U hours of culture (2661  + 57 dpm  in



untreated cells compared to 2287 + 60 dpm in cells treated  with 5 [ii/mSL [0.06 mM]



EDC).  Subsequently 2.5,  5,  and  10 \ii/mi  (0.03, 0.06 and 0.1 mM) EDC was added  to



cells  which were  cultured in  10  mM  hydroxyurea  to  suppress scheduled  DNA



synthesis.  The amount  of  unscheduled DNA synthesis was estimated by measuring



dpm from incorporated [3H]TdR  U  hours later.  At  10 pi/ml EDC 483 + 37 and 532 +



21  dpm  were   counted  without  and  with   exogenous   metabolic   activation,



respectively.  Both values were lower than corresponding  negative controls of 715



i 24 and 612 +_ 26 dpm, respectively.  No positive controls were  run to ensure  that



the system was  working  properly although testing of  chloromethyl  methyl ether
                                     9-159

-------
(CMME) with  activation resulted  in  a doubling of  dpras  over the corresponding



negative control  values  (1320 + 57 at 5 ul/ml CMME versus 612 + 26 untreated).



The authors calculated an effective DMA repair value (r)  for  each chemical based



on the  control and experimental  values  with and without metabolic activation.



Ethylene dichloride  was  evaluated by the authors as  positive  in the test,  but



they did not state their criteria for classifying a chemical as positive.  It is



important to note that none of the experimental values from cells treated with



EDC  without metabolic activation had  higher  dpm  values   than  the controls.



Furthermore, although  two out of  three experimental values were greater than the



controls with metabolic activation (673 ± 45 at 5 uS,/m£ and 630 +3*4 at 2.5 ufc/mJl



compared  to control value  of 612 +  26) the increases  were not statistically



significant.   The positive  finding reported in  this work  is  therefore judged to



be inconclusive.



     9.4.3.2.2. Detection  of DNA Adducts  — Reitz et al.  (1982)  compared the



pharmacokinetics  of EDC administered  to Osborne-Mendel rats  after inhalation and



after exposure by gavage.   (See section  9.1.3,  especially 9.1.3.5.)   As part of



their   study,   DNA  alkylation   was   measured   in  bacteria  at  EDC  cytosol



concentrations corresponding to those used in the DNA binding study (Table 9-33).


                                                          14
Two gram aliquots of TA1535 were  incubated with 7.06 umol [   C] EDC/mfc (sp. act.



= 3.2 mCi/mmol) and varying amounts of cytosol.   DNA alkylation values at cytosol



concentrations of 2.2,  7.8,  27,  and 71$  were  8.65,  27,  107,  and 137 dpra/mg



purified DNA,  respectively.   This corresponds to  4, 12.5, 49, and 64 alkylations



x 10   DNA nucleotides.  The corresponding reversion frequencies were 4.6 + 0.82,



23.5  +  3.0, 80.2 ±  9.6,  and 111  +  2.6, respectively (n =  3 in each case).   A



direct  correlation between the degree of alkylation and  an  increase in mutation



frequency was  indicated  by linear regression analysis (r =  0.9976).   In the DNA



alkylation   studies  with   rats   [  C]   EDC  (sp.  act.  =  0.32   mCi/mmol)  was



administered  to groups    of    three   animals   by  gavage   (150   mg/kg)  or





                                      9-160

-------
                                                                               TABLE 9-33


                                                      Summary of Mutagenicity Testing of EDC:  DMA Binding Studies
vo
 i
Activation
Reference Test System Strains System
Reitzetal., DNA alkylation TA1535 Phenobarbital-
1982 and mutagenesis induced rat > Cytosol
in Salmonella liver cytoaol 2.2
7.8
27
71
DNA alkylation Osborne- NA
in rats Mendel rat Route of
Exposure
Gavage
150 mg/kg


Inhalation
150 ppm,
6 hours

Chemical Information
Revertants
4.6 + 0.82
23.5 + 3.0
80.2 7 9.6
111 * 2.6
Tissue
Liver
Spleen
Kidney
Stomach
Liver
Spleen
Kidney
Stomach
Alkylation x 10~6
Nucleotides
4
12.5
49
64
Alkylation x 10~6
Nucleotides (means
from 2 experiments
21.3
5.8
17.4
14.9
8.2
1.8
5.2
2.8
13.9
2.5
14.5
6.7
3.3
1.8
2.0
1.9
Comments
1. Bacteria incubated with
7.06 |iool EDC/md.
2. Significant correlation
between degree of alkylation
and increased reversion
frequency (r = 0.9976).
3. Rats sacrificed 4 hours post
gavage or immediately after
Inhalation.

4. Sp. act. 3.2 mCi/mmol for
bacteria; 0.32 mCi/mmol
for rats.





-------
inhalation (150 ppm, 6 hours).  The animals were subsequently  sacrificed and DMA



was extracted from the liver, spleen, kidney,  and stomach for  measurement of DNA



alkylation.  Overall, three  to  five  times  more  DNA alkylation was present after



gavage than  after  inhalation.   The values ranged from 5.8 *  0.7  to  23.1  ± 7.1



alkylation/10   nucleotides   for  gavage  versus  1.8  +  0.3  to  8.2  +  3-3



alkylation/106 nucleotides for  inhalation.  Under the conditions of test used in



the experiments by Reitz et  al.  (1982) EDC exposure resulted in a similar degree



of adduct formation in  both rats  and bacteria.   Because DNA is  the genetic



material  in both  bacteria  and rats,  these  data predict that  mutations were



induced in the rat at the exposures  used.   This is in keeping with the positive



responses in other eukaryotes including Drosophila and cultured CHO cells.  DNA



alkylation was not measured  in rat gonads so it  is not possible to estimate what



the heritable genetic risk might be.



9.4.1.  Summary and  Conclusions.  Ethylene dichloride  (EDC)  has  been shown to



cause  gene  mutations  in bacteria,  plants,  Drosophila,  and  cultured Chinese



hamster ovary (CHO) cells.  Positive responses were observed in bacterial and CHO



cells  in  tests  that did not require an exogenous metabolic  activation system.



Stronger  positive responses  were found when hepatic metabolic  activation systems



were incorporated.   Based  on these positive findings in different test systems



representing a wide  range  of organisms, EDC is  judged to  be  capable  of causing



gene mutations.



     EDC  has  been  reported to cause  meiotic  chromosomal  nondisjunction in



Drosophila.  The induction of meiotic nondisjunction is a  significant genotoxic



effect; however, insufficient data are provided  to be able to  evaluate this  test



adequately.  A positive response in a properly conducted test is needed to  permit



a judgment on the  validity of this report.  With respect to its ability to cause



structural   chromosomal  aberrations,  sufficient  testing  has also   not  been
                                      9-162

-------
performed.   There is  only  one study on the ability of EDC  to  cause structural



chromosomal aberrations (i.e., in Allium root  tip cells  and  human lymphocytes).



Because this  study  was reported in  an  abstract, the author's  conclusions are



unsubstantiated.   Even though negative  results are reported from micronucleus



tests, additional information  is  needed to draw conclusions on the  ability of EDC



to cause chromosomal aberrations.  For example, it would  be  appropriate to test



EDC in ^n  vitro  and iri vivo  mammalian cytogenetic assays.  A  sister chromatid



exchange assay would  also  be  useful in assessing the ability  of EDC  to  cause



chromosome damage.



     There are no available data  on the ability of EDC  to damage DNA in mammalian



germ cells.  Thus, studies  on  the ability of EDC to reach germinal tissue would



be appropriate to determine  whether EDC  has  the potential to  cause heritable



mutations which may contribute to the genetic  disease  burden.   The finding that



EDC causes heritable mutations in Drosophila and alkylates  DNA in several somatic



tissues in the rat reinforces  the need for further germ cell  studies in mammals.



     Based on the weight-of-evidence, EDC  is judged to be  a mutagen that may have



the potential to cause adverse effects in humans.
                                     9-163

-------
9.5.  CARCINOGENICITY




     The purpose  of  this  section is to  provide  an  evaluation of the likelihood




that ethylene dichloride  (EDC)  (1,2-dichloroethane)  is  a human carcinogen and,




on  the assumption that  it is  a  human carcinogen, to  provide a basis for estima-




ting its public health  impact,  including  a  potency evaluation in relation to




other carcinogens.   The evaluation of carcinogenicity depends heavily on animal




bloassays and epidemiologic evidence.   However, other factors, including muta-




genicity, metabolism (particularly in relation  to  interaction with DNA), and




pharmacoki.netic behavior,  have  an  important bearing  on  both the qualitative and




quantitative assessment of carcinogenicity.  The available information on these




subjects is reviewed in other sections  of this  document and used in this evalu-




ation as appropriate.   This section presents an evaluation of the animal bio-




assays, the human epidemiologic evidence, the quantitative aspects of assessment,




and finally, a summary  and conclusions  dealing  with  all of the relevant aspects




of the carcinogenicity  of EDC.




9.5.1.  Animal Studies.   The  carcinogenic potential  of EDC has been investigat-




ed in a number of studies in  which EDC  was  administered to rats and mice via




various routes of administration.   The  following studies will be discussed:




one gavage study  performed by the  National  Cancer  Institute (1978) in which




EDC was administered to rats  and mice;  two  inhalation studies, including one




by Spencer et al.  (1951)  using  rats,  and  one bioassay by Maltoni et al. (1980)




in which EDC was  administered to rats and mice; one  intraperitoneal study by




Theiss et al. (1977) in which EDC  was administered to mice; and one skin-




painting study by Van Duuren  et al.  ( 1979)  in which  EDC was administered to




mice.




     9.5.1.1.  NATIONAL CANCER  INSTITUTE  (1978) RAT  STUDY ~ Hazleton Labora-




tories America Inc., Vienna,  Virginia,  under the sponsorship of the NCI, con-






                                      9-164

-------
ducted a bioassay of 1,2-dichloroethane (EDC) using Osborne-Mendel rats.  The




NCI published a final report of the bioassay in 1978.  Their results were also




reported by Weisburger (1977), the International Agency for Research on Cancer




(IARC, 1979), and Ward (1980).




     Technical-grade EDC, obtained for this study from the Dow Chemical Company,




Midland, Michigan, was tested for its purity.  Gas-liquid chromatography re-




vealed a purity of 98% to 99% EDC with 4 to 10 minor peaks (Ward, 1980).  How-




ever, in the NCI broassay a purity of greater than 90% was reported, with only




two unidentified minor contaminants.   Recently this discrepancy was resolved




when the original sample of EDC was reanalyzed, at which time the EDC was




found to be greater than 99% pure, with several unidentified contaminants.




This analysis was also performed by the National Institute for Occupational




Safety and Health (NIOSH) after completion of the bioassay; the sample contained




about 99% EDC, with chloroform as the major contaminant (Hooper et al., 1980).




     Solutions of EDC were prepared in corn oil and administered by oral




intubation to 200 Osborne-Mendel rats starting at 8 weeks of age.  The design




summary of this experiment is given in Table 9-34.  On the basis of a sub-




chronic EDC study, 50 rats of each sex were used for each of two dose levels




in the chronic study.  The maximum tolerated dose (MTD) was determined to be 95




mg/kg/day for both males and females; the second dose was one-half the MTD




(47 mg/kg/day).




     Twenty animals of each sex served as untreated controls; an equal number




were given the vehicle (corn oil) by  gavage.  Because of the inadequacies of the




subchronic studies,  the initial doses administered to the test animals were




found to be inappropriate.   Early signs of toxicity necessitated several changes




in the dosages (Table 9-34).  Anitnal  weight and food consumption per cage were




obtained weekly for the first LO weeks and monthly thereafter.   Animals were






                                     9-165

-------
        TABLE 9-34.
DESIGN SUMMARY FOR 1,2-DICHLOROETHANE  (EDC) GAVAGE
  EXPERIMENT IN OSBORNE-MENDEL RATS



Group
Males
Untreated control
Vehicle-control
Lou-dose




High-dosea




Females
Untreated control
Vehicle-control
Low-dose




High-dosea





Initial
number of
animals

20
20
50




50





20
20
50




50





1,2-dichloro-
e thane
dosage3

	
0
50
75
50
5QC
0
100
150
100
10QC
0

	
0
50
75
50
50=
0
100
150
100
100C
0


Observation period
Treated
(weeks)

	
78
7
10
18
34
_•_
7
10
18
34
—

	
78
7
10
18
34
—
7
10
18
34
— f *
Untreated
(weeks)

106
32
—
	
	
9
32
	
—
	
9
23

106
32
	
	
	
9
32
	
—
	
9
15
Time-weighted
average dosage
over a 78-week
periodb

	
0
47
	
	
	
™"™
95
— —
	
	
_•«

	
0
47
	
	
	
~ ~~
95
— — —
	
	
^^^
aDosage, given in mg/kg body weight, was administered by gavage five consecutive
 days per week.
^Time-weighted average dosage  =  (dosage x weeks  received)/78 weeks.
cThese dosages were cyclically administered with a pattern of one dosage-free
 week followed by 4 weeks  (5 days  per week) of dosage at the level indicated.
"All animals in this group died  before the bioassay was terminated.
SOURCE:  NCI, 1978.
                                      9-166

-------
checked daily for mortality.  Weight depression was observed in both groups




exposed to EDC.  By SO weeks,  the weight depression averaged 12% in high-dose




rats (Figure 9-3).  Mortality  was early and severe in dosed animals, especial-




ly those given the highest doses.  The mean survival was  approximately 55 weeks




on test for high-dose males and females (Figure 9-9 and Table 9-35).  The




early deaths were usually not  due to cancer; rather, the  toxic effects of EDC




appeared to be responsible for these deaths.  Rats dying  early had a variety of




lesions, including bronchopneumonia and endocardial thrombosis, which may have




contributed to early death. The pneumonia may have been  the result of a viral,




bacterial, or mycoplasmal infection, and the exposure to  the chemical may have




increased the tendency to develop severe pulmonary lesions,  which would lead to




death.  For the high-dose male rats, 50% (25/50) were alive at week 55 and 16%




(8/50) were alive at week 75.   Survival was higher in the other groups; 52%




(26/50) of the rats in the low-dose group lived at least  82 weeks, and 50%




(10/20) in the vehicle-control group lived at least 72 weeks.  In the high-dose




female rats, 50% (25/50) were  alive at week 57; 20% (10/50)  of the rats in the




low-dose group were alive at week 85.   Despite the sacrifice of five females at




week 57, 65% (13/22) of the untreated control group survived until the end of




the study.




     A gross necropsy was performed on each animal that died during the experi-




ment or was killed at the end.  A total of 28 organs, plus any tissues contain-




ing visible lesions, were fixed in 10% buffered formalin,  embedded in paraplast,




and sectioned at 5 y for slides.   Hematoxylin and eosin stain were used rou-




tinely, and other stains were  employed when necessary. Diagnoses of tumors




and other lesions were coded according to the Systematized Nomenclature of




Pathology (SNOP) of the College of American Pathologists  (1965).
                                     9-167

-------
   750
   600-
2
I  450-
g
uu

>  300-
o
CD
z
   150-
          MALE RATS
                                    Untreated Control
                                    Vehicle-Control
                                    Low-Dose
                                    High-Dose
                                                     •750
                                                    -600
                                                    -450
                                                    -300
                                                                         -150
        I
15     30      45      60      75

               TIMEONTEST(Weeks)
                                                      90
                                         105
                                                                       120
   750
|  600-
03
w
(S

I  450-1
O
2
   300-
uj  150-
2
                                                     750
          FEMALE RATS
                                                          Untreated Control
                               	Vehicle-Control
                               	Low-Dose
                               	High-Dose
                                                   - 600
                                                   -450
                                                   -300
                                                                         -150
                                       \
                          \
              15
30
                             45       60      75

                             TIME ON TEST (Weeks)
                                        90
 I
105
120
    Figure 9-S.  Growch curves for male and female Osborne-Mendel  rats
    administered EDC by gavage.

    SOURCE:  MCI, 1973.
                                  9-168

-------
1 n

-J 0.8-
C
^<
>
QC
—5
w 0.6-
u.
0

>
j 0.4-
CQ

CQ
0
£ 0.2-
-
0.0 -
MALE RATS

\_ \ ''--,_, L|

'~"L. '--l 	 1
1 	 1 1^1
i '• — . _ V

1 	 , • 	 j
• * • "" "L S
U^L ! 	 '-
<-, i
\ i '--,
\ L. ';
— • — « n '
L— '"'
	 Untreated Control V "'-•._ i-._.
	 Vehicle-Control i "'-,
	 	 Low-Dose | "l;
	 High-Dose "L--L_ '--,_
' l ' l ' i ' i ' i ' i ' '
- 1.0
-0.8


-


-0.6




- 0.4



-0.2
-
0.0
0 15 30 45 60 75 90 105 120
TIME ON TEST (Weeks)



_J
<
>
>
cc
Z)
to
u.
0
>
^
_i
CO
<

0
cc
Q.




1 n


0.8-


•

0.6 — '

-

0.4-




0.2-




rrM AI i n ATr>
--J=o_ _ ~ 	 '~I I 	 1
"Li~, \. h
~I 	 ^ ' 	 ! ' 	 .
— ^ __ ~ ~ ~"»_ '
L_^___ -, !
i_
*^» ' — i
i V
T. [^ | 	

i ~ "i ! __
"'•i ~!
T
-I
I -I
	 Untreated Control "^
	 Vehicle-Control 1,
•-i -i_
	 Low-Dose "*•— •
L, ^
	 High-Dose ~" 	 ^
' '< ' : ' ! ' 1 ' I '



-0.8


-

-0.6

-

-0.4




-0.2

~


         15     30      45       60      75
                        TIMEONTEST(Weeks)
                        90
105    120
Figure 9-9.   Survival  comparisons  for
racs administered  EDC  by  -savage.

30URCE:   :;CI,  19 73.
             T.aie and  female Osborne-Mendel

-------
             TABLE 9-35.  TERMINAL  SURVIVAL OF OSBORNE-MENDEL RATS
                     TREATED WITH  1,2-DICHLOROETHANE (EDC)
                            Males                          Females

                  Weeks  in      Animals alive      Weeks in     Animals alive
Group               study      at end of study       study     at end of study


Untreated control     106        4/20a  (20%)           106       13/20a (65%)

Vehicle-control       110        4/20   (20%)           110        8/20  (40%)

Low-dose              110         1/50   (2%)            101        1/50   (2%)

High-dose             101        0/50b  (0%)            93        0/50*>  (0%)


aFive rats were sacrificed  at  75 weeks.
^All animals in this  group  died before the bioassay was terminated.

SOURCE:  Adapted from NCI,  1978.



     Squamous cell  carcinomas  of  the forestomach  occurred in 3/50 (6%) low-dose

males, 9/50  (18%) high-dose males,  0/50  (0%) high-dose females, and 1/49 (2%)

low-dose females (Table  9-36). None of  these  tumors  occurred in the controls.

For male rats, the  Cochran-Armitage Trend Test  indicated a significant (p =

0.01) positive association  between  dosage and  the incidence of squamous cell

carcinoma of the stomach.   The Fisher  Exact Test  confirmed the significance

of these results with p  values of 0.039 and 0.001 when comparisons were made

with the matched vehicle-control  group and the  pooled vehicle-control* group,

respectively.

     Squamous cell  carcinomas  of  the forestomach  were first observed in high-

dose male rats at 51  weeks  after  oral  intubation  of EDC.  These lesions were
*The pooled vehicle-control group combined  the  vehicle-controls from the
 studies of 1,2-dichloroethane,  1,1,2-trichloroethane, and  trichloroethane.
                                      9-170

-------
characterized microscopically by acanthosis and hyperkeratosis in the super-

ficial area.  The basal epithelial layer contained papillary cords and nests of

anaplastic squaraous epithelium supported by a dense band of fibrous connective

tissue.  These carcinomas extended through the muscularis mucosa, submucosa,

muscular layers, and serosa, and in one high-dose male, raetastasized to adja-

cent tissues.
   TABLE 9-36.  SQUAMOUS CELL CARCINOMAS OF THE FORESTOMACH IN OSBORNE-MENDEL
                   RATS TREATED WITH 1,2-DlCHLOROETHANE (EDC)
Group
Untreated control
Matched vehicle-
control
Pooled vehicle-
control
Low-dose
High-dose
Males p value3
0/20 (0%)
0/20 (0%)
0/60 (0%)
3/50 (6%) NS
9/50 (18%)b 0.001
Females p value3
0/20 (0%)
0/20 (0%)
0/59 (0%)
1/49 (2%) NS
0/50 (0%) NS
ap values calculated using the Fisher Exact Test.   Treated versus pooled
 vehicle-control.
t"A squaraous cell carcinoma of the forestoraach metastasized in one male in this
 group.
NS = not significant when p values are greater than 0.05.

SOURCE:  Adapted from NCI, 1978.
     Hemangiosarcomas were noted in some treated rats, as described in Table

9-37, but not in any of the controls.   Low-dose males and females showed higher

incidences of hemangiosarcoma than high-dose animals.  Tumors were observed in

several sites, including spleen, liver, adrenal glands, pancreas, large intes-

tine, subcutaneous tissue, and abdominal cavity.  The Cochran-Armitage Trend

                                     9-171

-------
Test indicated a significant  (p  =  0.021) positive association between dosage

and the incidence of hemangiosarcomas  in male  rats as compared to the pooled

vehicle-control rats;  the  trend  was  also positive in female rats (p = 0.042).

The Fisher Exact Test  confirmed  these  findings, with statistically significant

p values  for high-dose males  versus  pooled  vehicle-controls (p = 0.016), for

low-dose  males versus  pooled  vehicle-controls  (p = 0.003), and for both low-

and high-dose females  versus  pooled  vehicle-controls (p = 0.041).
       TABLE 9-37.   HEMANGIOSARCOMAS  IN  OSBORNE-MENDEL RATS TREATED WITH
                             1,2-DICHLOROETHANE  (EDC)
Group
Untreated control
Matched vehicle-
control
Pooled vehicle-
control
Low-dose**
High-dosec
Males p value3
0/20 (0%)
0/20 (0%)
1/60 (2%)
9/50 (18%) 0.003
7/50 (14%)b 0.016
Females
0/20 (0%)
0/20 (0%)
0/59 (0%)
4/50 (8%)
4/50 (8%)
p value3



0.041
0.041
ap values calculated  using the Fisher Exact Test (one-tailed).  Treated versus
 pooled  vehicle-control.
bonly  48 animals  were examined for hemangiosarcomas of  the  large  intestine.
cOnly  49 animals  were examined for hemangiosarcomas of  the  spleen and adrenals
 and 48  for  hemangiosarcomas of the pancreas.

SOURCE:  Adapted  from NCI, 1978.
      In  addition to stomach carcinomas and hemangiosarcomas,  EDC-treated  female

rats  showed  significant increases in the incidence of mammary adenocarcinomas

(Table 9-38).   Tumors were observed in the high-dose group as early  as  20 weeks

after treatment.  The Cochran-Armitage Trend Test detected a  significant

                                      9-172

-------
(p < 0.001) positive association betwen the dosage and the incidence of mammary

adenocarcinomas when compared with either control group.   This tumor incidence

was significant when the high-dose group was compared with either the matched

vehicle-control group (p < 0.001) or the pooled vehicle-control group (p =

0.002) using the Fisher Exact Test.  Historically, this tumor was observed in

4/200 (2%) of the vehicle-control females.
   TABLE 9-38.  ADENOCARCINOMAS OF THE MAMMARY GLAND IN FEMALE OSBORNE-MENDEL
                   RATS TREATED WITH 1,2-DICHLOROETHANE (EDC)
                                Adenocarcinoma of
Group                           the mammary gland                 p value3


Untreated control                    2/20 (10%).

Matched vehicle-control              0/20 (0%)

Pooled vehicle-control               1/59 (2%)

Low-dose                             1/50 (2%)                      NS

High-dose                           18/50 (36%)                     0.0008


ap values calculated using the Fisher Exact Test (one-tailed).  Treated versus
 pooled vehicle-control.
NS = not significant.

SOURCE:   Adapted from NCI,  1978.



     An increased incidence of fibromas of the subcutaneous tissue was reported

in both high-dose (p = 0.007) and low-dose (p = 0.017) male rats when compared

to the pooled vehicle-control group.

     In summary, a statistically significant increase in the incidence of squa-

mous cell carcinomas of the forestomach, hemangiosarcomas of the circulatory

system, and fibromas of the subcutaneous tissue occurred in male rats.  There


                                     9-173

-------
was also a statistically  significant  increase  in  the  incidence of adenocarci-




nomas of the mammary  gland and hemangiosarcomas of  the circulatory system in




female  rats.



     9.5.1.2.   NATIONAL CANCER INSTITUTE  (1978) MOUSE STUDY — Hazleton Labora-




tories  America  Inc.,  under the sponsorship of  the NCI, conducted a bioassay of




1,2-dichloroethane  (EDC)  using B6C3F1 mice.  The  results  of this study were




published by the  NCI  (1978) and were  also reported  by Weisburger (1977), the




International Agency  for  Research em  Cancer (IARC,  1979),  and Ward (1980).




     Technical-grade  EDC  (with the same purity as that described in the NCI




rat study) was  administered to 200 B6C3F1 mice starting at 5 weeks of age.  The




design  summary  for  this experiment is given in Table  9-39. On the basis of




results of  the  subchronic studies, 50 mice of  each  sex were used at each of the




two dose levels for the chronic study.  The MTD was determined to be 195 mg/kg/




day for male mice and 299 mg/kg/day for female mice.  The second dose, which




was one-half the  MTD, was determined  to be 97  mg/kg/day for male mice and 149




mg/kg/day  for  female  mice.



     Twenty mice  of each sex served as untreated  controls, and an equal number




were given  the  vehicle (corn oil) by  gavage.  Because of  inadequacies in the




subchronic  studies, the initial doses administered  to the test animals were




found  to be inappropriate.  Signs of  toxicity  in  these animals early in the




study  led  to  changes  of the dosages several times (Table  9-40).  Animal weights




and food consumption were recorded for the first  10 weeks and monthly thereaf-




ter.   Animals  were  checked daily for mortality and  signs  of toxic effects.  No




dose-related  mean body weigtit depression was observed in  male mice or low-dose




female  mice (Figure 9-10).  A depression in the mean  body weight of  the high-




dose female mice  was  apparent as early as week 15.   The estimated probabilities




of  survival for male and female mice in the control and EDC-dosed groups are






                                      9-174

-------
Group
        TABLE 9-39.   DESIGN SUMMARY FOR 1,2-DICHLOROETHANE  (EDC)  GAVAGE
                           EXPERIMENT  IN B6C3F1  MICE
 Initial   1,2-dichloro-  Observation period
number of     ethane      Treated  Untreated   Time-weighted
 animals      dosage3 •    (weeks)   (weeks)    average dosage'1
Males
Untreated control
Vehicle-control
Low-dose


High-dose

Females
Untreated control
Vehicle-control
Low-dose



High-dose


a Dosage, given in

20
20 0
50 75
100
0
50 150
200
0
20 	
20 0
50 125
200
150
0
50 250
400
300
0
rag/kg body weight, was
days per week.
^T iine-weigh ted average dosage = (dosage

	
78
8
70
_«_
8
70
™»™

78
8
3
67
-»*—
8
3
67

administered by
x weeks received)

90
12
	
	
12
__—
13
91
32
	
	
	
13
.^—
	
13
gavage


	
0
97
	
™-"—
195
	 *

0
149
	
	
— —
299
	
~ ~ -~
five consecutive

                                 weeks receiving chemical
SOURCE:  Adapted from NCI, 1978.
                                     9-175

-------
shown in Figure 9-11.  Terminal  survival of  treated and control mice is shown

in Table 9-40.  For male mice, no  statistically significant association between

dosage and mortality was observed.   In  the high-dose group, 50% (25/50) of the

mice were alive at 84 weeks and  42%  (21/50)  survived until the end of the

study.  In the low-dose group, however-, survival was low.  By 24 weeks, 52%

(26/50) of the low-dose group and  55%  (11/20) of the untreated control group

had died.  In the high-dose group, 72%  (36/50) of the animals died between

weeks 60 and 80.  These deaths may have been tumor-related, since 69% (25/36)

had one or more tumors.  Survival  was high in the other groups; 68% (35/50) of

the low-dose and 80% (16/20) of  the  untreated control groups survived until the

end of the study.
           TABLE 9-40.  TERMINAL  SURVIVAL OF B6C3F1 MICE TREATED WITH
                             1,2-DICHLOROETHANE  (EDC)
Males
Group
Untreated control
Vehicle-control
Low— dose
High-dose
Weeks in
study
90
90
90
91
Animals alive
at end of study
7/20 (35%)
11/20 (55%)
11/50 (22%)
21/50 (42%)
Females
Weeks in
study
90
90
91
91
Animals alive
at end of study
16/20 (80%)
16/20 (80%)
34/50 (68%)
1/50 (2%)
SOURCE:  Adapted  from NCI,  1978.
     A gross necropsy was  performed on  each  animal  that died during the experi-

ment or was killed at the  end.   Twenty-eight organs  in all, plus any tissues

containing visible lesions,  were fixed  in  10% buffered formalin, embedded in


                                      9-176

-------
DU —
1 40-
2
5

t-
0 30-
UJ
§
I 20-
00
z
^
^ 10^




(

-<:-^.
—*?'/ *^^ *^**^>^r-^
^^ ^^ -^X^X^ ^x * ^"^^r ** "^^"^

-^^" N 	 * 	 ^^
^/if
rT
f
1

	 Untreated Control
	 Vehicle-Control
P.
-— — — -— Low-uose
	 High-Dose MALE MICE

I I I I ' 1 i 1 i 1 ' 1 1
) 15 30 45 60 75 90 105 1:
-40

_


-30

~
-20



-10


- 0

10
                             TIME ON TEST (Weeks)
   50
   40-
   30-
LU


Q  20-
O
GO
10-
             Untreated Control
             Vehicle-Control
             Low-Dose
             High-Dose
                                                       FEMALE MICE
-\	1	1	1	1	1	1	r-
 15       30      45      60      75
                 TIME ON TEST (Weeks)
                                                      90
                                                           T~
                                                           105
                                                                         •50
                                                                        -40
                                                                        -30
                                                                     -20
                                                                     -10
                                                                          0
120
          Figure  9-10.   Growth  curves for male and female 86C3F1 mice
          administered  EDC  by gavage.

          SOURCE:   Adapted  from NCI,  1978.
                                9-177

-------



<
>
>
cr

t/5
LL.
0
H;
m
CD
O




1 n


0.8 -

_


0.6 -

0.4 -

-
0.2-




'- 	 |~~!_ ~| _
L- -, 	 i rt Is j
*~ 	 *• 	 L "*""' 1
1 ~~nVi L'
i '-^S I
1 K '., '.
*! • •— •- i
|__ ls T"L

iy-k V'
s t"
1
1
hi
— — Untreated Control
	 Vehicle-Control
	 Low-Dose
	 High-Dose MALE MICE
1 i 1 i 1 i 1 'I1!1!1



-0.8

__'


-0.6

-0.4

m
-0.2
_
0 0

0 15 30 45 60 75 90 105 120
                     TIME ON TEST (Weeks)

.0

^ 0.8-
^
>
tr
w 0.6-
ij
0
5 0.4-
co
CD
o
oc 0.2 J
a.



(



^^— 't
"" ^-^— i
1 '-,
1,
1
i
^
•"i
\

	 Untreated Control
i
	 Vehicle-Control 1
	 Low-Dose \
	 H,gh-Dose 'n^ FEMALE MICE
^™* ""
1 1 ' 1 ' 1 1 1 1
) 15 30 45 60 75 90 105 1
— 1 n
1 .w

-0.8

—

-06

-
-0.4



-0.2



20
                     TIME ON TEST (Weeks)
Figure 9-11.  Survival comparisons for male and female  36C3F1  mice
administered EDC by gavage.

SOURCE:  Adapted ;ron NCI, 1973.

                          9-178

-------
paraplast, and sectioned at 5 p for slides.  Hematoxylin and eosin stain were

used routinely, with other stains employed as necessary.  Diagnoses of tumors

and other lesions were coded according to the Systematized Nomenclature of

Pathology (SNOP) of the College of American Pathologists.

     The histopathologic findings of the study concerning hepatocellular

carcinomas in mice are tabulated in Table 9-41.  Hepatocellular carcinomas

occurred in 2/17 (12%) untreated control males, 1/19 (5%) low-dose males, and

12/48 (25%) high-dose males.  A significant number of hepatocellular carcinomas

were observed in high-dose males.  The Cochran-Armitage Trend Test indicated

a positive dose-response association when the high-dose group was compared with

either the matched vehicle-controls (p = 0.025) or the pooled vehicle-controls

(p = 0.006).  The Fisher Exact Test supported this finding with a significant

(p = 0.009) comparison of the high-dose to the pooled control group.
         TABLE 9-41.  HEPATOCELLULAR CARCINOMAS IN B6C3F1  MICE TREATED
                         WITH 1,2-DICHLOROETHANE (EDC)
Group
Untreated controls
Matched vehicle-
controls
Pooled vehicle-
controls
Low-dose
High-dose
Males p value3
2/17 (12%)
1/19 (5%)
4/59 (7%)
6/47 (13%) NS
12/48 (25%) 0.009
Females
0/19 (0%)
1/20 (5%)

0/50 (0%)
1/47 (2%)
p value3



NS
NS
ap values calculated using the  Fisher Exact Test (one-tailed).   Treated versus
 pooled vehicle-control.
NS = not significant.

SOURCE:  Adnpted from NCI,  1978.

                                     c>-179

-------
     A  large  number of  alveolar/bronchiolar  adenomas were found in mice

 treated with  EDC  (Table 9-42);  31% in both male  (15/48) and female (15/48)

 high-dose  mice.   These  adenomas were  not  observed  in untreated or vehicle-

 control males.  For both sexes  the Cochran-Armitage Trend Test showed a sig-

 nificant (p = 0.005)  positive  dose-response  association when the dosed groups

 were compared to  either control group.  The  Fisher Exact Test also indicated

 that the high-dose  group for both sexes had  significantly higher (p = 0.016)

 incidence  rates than  either  of  the two control groups.  For female mice, the

 Fisher  Exact  Test comparing  the low-dose  group to  the pooled vehicle-control

 group was  also statistically significant  (p  = 0.046).
       TABLE 9-42.   ALVEOLAR/BRONCHIOLAR  ADENOMAS  IN B6C3F1 MICE TREATED
                          WITH  1,2-DICHLOROETHANE (EDC)
Group
Untreated controls
Matched vehicle-
controls
Pooled vehicle-
controls
Low-dose
High-doseb
Males p value3
0/20 (0%)
0/19 (0%)
0/59 (3%)
1/47 (2%) NS
15/48 (31%) 0.0025
Females
1/19 (5%)
1/20 (5%)
2/60 (3%)
7/50 (14%)
15/48 (31%)
p value3



0.046
0.0201
ap values calculated using  the  Fisher Exact Test  (one-tailed).  Treated versus
 pooled vehicle-control.
°In addition, one high-dose  female mouse had an alveolar/bronchiolar carcinoma.
NS = not significant.

SOURCE:  Adapted from NCI,  1978.
                                     9-180

-------
     Squamous cell carcinomas of the forestomach occurred in 1/19 (5%) vehicle-

control males, 1/46 (2%) low-dose females,  and 5/48 (10%) high-dose females

(Table 9-43).  For female mice,  the Cochran-Armitage Trend Test indicated a

significant (p = 0.035) positive association between dosage and the incidence

of squamous cell carcinomas of the forestomach when comparing the dosed groups

to the pooled vehicle-controls.   The Fisher Exact Test, however,  was not

significant.  The microscopic appearance of the stomach of mice was comparable

to that described for rats.
            TABLE 9-43.   SQUAMOUS CELL CARCINOMAS OF THE FORESTOMACH
              IN B6C3F1  MICE TREATED WITH 1,2-DICHLOROETHANE (EDC)
Group
Untreated controls
Matched vehicle-
controls
Pooled vehicle-
controls
Low-dose
High-dose
Males p value3
0/20 (0%)
1/19 (5%)
1/59 (2%)
1/46 (2%) NS
2/50 (4%) NS
Females
0/20 (0%)
1/20 (5%)
1/60 (2%)
2/50 (4%)
5/48 (10%)
p value3



NS
NS
ap values calculated using the Fisher Exact Test (one-tailed).   Treated versus
 pooled vehicle-control.
NS = not significant.

SOURCE:  Adapted from NCI, 1978.
     The incidence of adenocarcinomas of the mammary gland in female mice

treated with EDC is presented in Table 9-44.  The Cochran-Armitage Trend Test

indicated a significant (p = 0.007)  positive association between dosage and
                                     9-181

-------
the  Incidence  of  adonocarcinomas of  the mammary  gland when compared to the

pooled vehicle-controls.   The Fisher Exact  Test  confirmed these results with a

significant  (p £ 0.003)  comparison of both  high-dosu (7/48) and low-dose (9/50)

groups to  the  pooled  vehicle-control groups (0/60).
       TABLE  9-44.   ADENOCARCINOMAS OF THE  MAMMARY  GLAND  [N FEMALE B6C3F1
                    MICE  TREATED WITH I,2-IHCHLOROETHANE (EDC)
                                   Adenocarcinoma  of
Group                              the  mammary  gland                 p value"1


Pooled vehicle-controls                0/60  (0%)

Matched vehicle-controls               0/20  (0%)

Low-dose                               9/50  (18%)                    0.0005

High-dose                              7/48  (15%)                    0.0026


ap values calculated using  the  Fisher  Exact  Test  (one-tailed).  Treated versus
 pooled vehicle-control.

SOURCE:  Adapted  from  NCI,  1978.



     Endometrial  tumors observed  in  female  mice are described in Table 9-45.

The Cochran-Armitage Trend  Test indicated a  significant  (p = 0.017) positive

association between dosage  iml  combined  incidence.  The  Fisher Exact Test

showed a statistically significant (p  =  0.014) incidence In the high-dose

(5/47) group.

     In summary,  the NC [  study  in B6C3F1 mice  demonstrated a statistically

significant increase i.i incidences of  hepatocellular carcinomas .ind alveolar/

bronchiolar adenomas in male  mice and  a  statistically significant increase in

incidences of alveolar/bronchlolar adenomas, mammary carcinomas, and endometrial

tumors in female  mii-f.

                                     9-182

-------
       TABLE 9-45.  ENDOMETRIAL POLYP OR ENDOMETRIAL STROMAL SARCOMAS IN
            FEMALE B6C3F1 MICE TREATED WITH 1,2-DICHLOROETHANE (EDC)
Group
    Endometrial polyp or
endometrial stromal sarcomas
p valuea
Fooled vehicle-controls

Matched vehicle-controls

Low-dose

High-dose
        0/60 (0.0%)

        0/20 (0.0%)

        5/49 (10.0%)

        5/47 (11.0%)
0.016

0.014
ap values calculated using the Fisher Exact Test (one-tailed).   Treated versus
 pooled vehicle-control.

SOURCE:  Adapted from NCI, 1978.
     9.5.1.3.  SPENCER ET AL.  (1951) RAT STUDY — An early  inhalation study

conducted by Spencer et al.  (1951) found no evidence of carcinogenic activity

when 15 male and 15 female Wistar rats were exposed 151 times during a 212-day

period to EDC at 200 ppra for 7 hours per exposure.

     9.5.1.4.  MALTONI ET AL.  (1980) RAT STUDY — A more recent inhalation study

conducted by Maltoni et al.  (1980) in Sprague-Dawley rats provided no evidence

of carcinogenicity after lifetime exposure to EDC.  The EDC used in this study

was supplied by Montedison,  and had a purity of 99.82%, with five contaminants

(Table 9-46).  The design of the experiment is given in Table 9-47.

     Maltoni et al. exposed  four groups of 12-week-old Sprague-Dawley rats

(each group consisting of 180  rats of both sexes) to EDC concentrations of 250-

150 ppm, 50 ppm, 10 ppm, and 5 ppm, respectively, 7 hours per day, 5 days per

week, for 78 weeks.  After several days of 250 ppm exposure,  the rats began to

exhibit severe toxic effects,  and the concentration was reduced to 150 ppm.  Two
                                     9-183

-------
      TABLE 9-46.  CHARACTERIZATION  OF  1,2-DICHLOROETHANE  (EDC) INHALATION
                        EXPERIMENT IN SPRAGUE-DAWLEY  RATS
Component                                                 Purity


1,2-Dichloroethane                                        99.82%

1,1-Dichloroethane                                         0.02%

Carbon tetrachloride                                       0.02%

Trichloroethylene                                          0.02%

Perchloroethylene                                          0.03%

Benzene                                                    0.09%


SOURCE:  Maltoni et al. ,  1980.



groups, composed of 180 rats  per group,  served as  controls.  One of the two

control groups was kept in an exposure  chamber under  the same conditions and

for the same length of  time as  the  exposed  rats.   At  the end of the treatment

period, the animals were  allowed to live until spontaneous death.  Animals

were weighed every 2 weeks during the treatment period and every 8 weeks there-

after.  All detectable gross  pathologic  changes were  recorded.  A complete

autopsy was performed on  each animal, with  histopathologic examinations conduc-

ted on the following:  brain, Zymbal glands,  retrobulbar glands, tntersr.apular

brown fat, salivary gland,  tongue,  lungs, thymus,  diaphragm, liver, pancreas,

kidneys, spleen, stomach,  various segments  of the  intestine, bladder, gonads,

lymph nodes (axillary, inguinal,  and mesenteric),  and any other organ with

pathologic lesions.

     The extent of mortality  varied with the  different groups, but there appears

to be no direct relationship  between mortality and exposure to EDC (Table 9-A8).
                                      9-184

-------
            TABLE 9-47.  DESIGN SUMMARY FOR 1,2-DICHLOROETHANE (EDC)
                       EXPERIMENT IN SPRAGUE-DAWLEY RATS3
Group
I

II

III

IV

V

VI

Concentration
250-150 pprac

50 ppra

10 ppm

5 ppm

Controls in chambers

Controls

Sex
M
F
M
F
M
F
M
F
M
F
M
F
Animals'3
Number
90
90
90
90
90
90
90
90
90
90
90
90
aExposed 7 hours/day, 5 days/week, for 78 weeks.
bSprague-Dawley rats, 12 weeks old at start.
cAfter a few weeks the dose was reduced to 150 ppm, because of high toxicity
 at the 250 ppm level.

SOURCE:  Maltoni et al., 1980.
                                     9-185

-------
                        TABLE 9-48.  SURVIVAL OF SPRAGUE-DAWLEY RATS EXPOSED TO EDC AT 52 AND 104 WEEKS3
oo
Dose
Group (ppm)
I 250-15Qb

II 50

III 10

IV 5

V Controls
in chambers
VI Controls

Sex
M
F
M
F
M
F
M
F
M
F
M
F
Initial
numbers
90
90
90
90
90
90
90
90
90
90
90
90
Survivors at
52 weeks of age
Number
79
84
87
87
81
87
89
90
80
79
83
88
Percent
87.8
93.3
96.7
96.7
90.0
96.7
98.9
100.0
88.9
87.8
92.2
97.8
Survivors at
104 weeks of age
Number
10
21
17
29
13
26
45
48
12
22
16
36
Percent
11.1
23.3
18.9
32.2
14.4
28.9
50.0
53.3
13.3
24.4
17.8
40.0
Survivors
52 weeks
start of
Number
67
79
70
84
70
81
75
85
64
73
72
84
after
from
study
Percent
74.4
87.8
77.8
93.3
77.8
90.0
83.3
94.4
71.1
81.1
80.0
93.3
       aRats were 12 weeks old at the start of the experiment.
       bAfter a few weeks the dose was reduced to 150 ppm because of high toxicity at the 250 ppm level.

       SOURCE:  Maltoni et al., 1980.

-------
The highest survival rate was observed in both males and females of the group




exposed to EDC at 5 ppm.   In females the highest mortality rate was observed in




the control group in the  chamber and in the group exposed to EDC at 250-150




ppm.  The survival rates  at 52 and 104 weeks of age are reported in Table 9-48.




At these ages the overall survival rates were 93.9% and 27.3%,  respectively.




The survival rates after  52 weeks from the start of the experiment are also




given in Table 9-48.  The overall survival rate was 83.7%.




     The results of histopathologir analysis are shown in Table 9-49.   No




statistically significant increase in the incidence of any specific type of




tumor was found in the treated rats when compared with controls.  There was,




however, an increased incidence of mammary tumors in some rats  in Group IV,




particularly when compared with the control group in the chamber (Table 9-50).




     It appears that the  increase in mammary tumors in some of  the treated




groups, particularly when compared to controls in the chamber,  was not due to




malignant tumors, but to  fibromas and fibroadenomas.  The increased incidence




of mammary fibromas and fibroadenomas is statistically significant in  groups




exposed to EDC at 250-150 ppm, 50 ppm, and 5 ppm, when compared to controls




in the chamber, but not significant when compared to controls outside  of the




chamber.  Although the author stated that the onset of fibromas and fibroade-




nomas was age-correlated, the incidences of these tumors observed in the vari-




ous groups were probably  due instead to the differences in survival rates




within the groups.  The highest difference in incidence was found between the




control groups in the chamber, which had low survival rates, and the groups




treated with EDC at 5 ppm, which had high survival rates.  The  difference in




survival rates is thus seen to be related to the differences in incidence in




the two control groups.
                                     9-1S7

-------
IABI>  9-49.  rilMDR INCIUhNO  IN SPKAGMt-DAULKY KATS fcXPIISHI TO  1,2-DICHLOROF.THANK (KIIC)
Animals with




Mammary tin
Anlnvilh"


(•r.inp (rfnii i nt t ill. in s.'\
1 'SO-ISH p ,.,„<• M
).
M iii.l 1
1 1 Ml [ipm "I
h
M .in.) 1
VO
,L III Ml ppra M
00 H
OO M .iinl Y
IV S |.pm H
}
N and !•
V (.mil nil* In H
i li iinhi'r* !•
H .mil !•
VI (nnt ml* M
t
M anil t
1.H II

Niimhi-l
11 I ill
'III
'lit
IH.I
'in
'lit
IHll

til
111
IHll
411
911
IHll
9(1
9l>
IH'l
90
911
180
10811



( nt ri'i toil Tol il

84
40
174
40
Oil
IHll

X9
40
1/4
411
41)
|8ll
90
90
IHll
40
90
180
107)1
immhrr
II
'ii
hi
III
58
hH

S
ll
iK
II
h5
76
8
18
!th
S
52
">'

Percent™
12.1
57.7
JS.2
II. 1
64.4
17.8

5.h
'•7.8
2h.H
12.2
72.2
42.2
8.9
42.2
25.5
5.5
57.8
31.7

nors
Average
latency
time

92.9
78.6
81.1
H8.8
78.5
80.1)

60.8
79.4
77.5
110.2
81.2
87.1
85.5
81.1
81.6
42.1)
85.5
86.1

/ymhal gland carcinomas


Total
number
II
2
2
II
1
1

0
II
(I
0
2
2
,
1)
1
1
1)
1




Percent*1
	
2.2
I.I
	
1 . 1
0.6

—
	
	
	
2.2
I.I
I.I
	
0.6
I.I
	
0.6

Avrrngp
lal'-nry
time

	
78.0
78.0
	
86.0
86.1)

	
	
	
	
47.1)
47.0
78.0
	
78.0
74.0
	
74.0



Total
numhe r
0
0
0
1
2
1

4
0
4
2
A
8
1
3
6
0
3
1

tumorn

Leukemlas



Percent"
_.__
	
—
I.I
2.2
1.7

4.6
	
2.2
2.2
6.7
4.4
I.I
3.1
2.2
	
3.3
1.7

Avernge
latency
time

..__
	
	
fld. (1
74.5
76.3

74.0
	
74.0
111. 5
81.5
96.11
17.0
M.I
59.5
	
79.3
79.3



Nephroblastnmas


Total
numbe r
0
0
»
0
0
0

0
0
0
1
(I
1
0
0
0
(1
0
0

Average
latency
tine
Percent" 
-------
TABIF 4-49.  (rnnllnnnH)


Animals with tumr
Anglnsarcomas




Group Cnn« 4*«ll rat l»n SPK
I JSO-IVI ffaf H
F
•< ,v»d F
1 1 Sn rpm M
K
^l" H and F
00 III in ppn M
*£> t
M and 1
IV S ppn H
F
N and f
V GiiUrnll (n M
chamber*. F
M and F
VI Cnnlrnls 1
r
H anri F
l.lver
Aver-ir.-
Intent i
Tut il I INK*
nnmher Pen em n (wk)
-------
TABLE 1-49.  (continued)
Animals with tumors



l.nmp Cum iMit r.il Inn Sc<
1 2SII-IMI |ipnp M
F
H .ind >•
I 1 Sll ppm H
F
M and F
VO
1 III Id ppm H
rx *
vD
O H .ind F
IV •) ppm M
|.
H and V
V GmirnK In H
iliamhrrs F
H and t
VI Cnntrols H
F
M and F
Hepatomaa
Average
latency
Tiu.il lime
number IVrc«nth (wk)"1
II 	 —
II
II ... —
o — —
II
II — —

0
I) 	 	
II
0
o

0
0
0
o
0
0
Foreatomach epithelial

Total
number
0
0
0
1
0
1

0
1
1
1
0
1
2
1
1
2
1
3


Percent b
._ .
—
—
I.I
—
O.n

—
I.I
O.b
I.I
	
0.6
2.2
1.1
1.7
2.2
I.I
1.7
Average
latencv
time

...
...
	
60. 1)
	
60. 0

	
78.0
78.0
101. 1)
...
101.0
78.)
61.0
71.1
110.0
102.0
107.3
Skin carcinomas
Average
latency
Tut a 1 tine
number Percent11 (wk)d
0
1)
0
1 I.I th.O
0
1 0.6 36.0

1 I.I 94.0
1) 	 	
1 0.6 94.0
,1
o
„
0
0
0
1,
0
0
Subcutaneous aarcoaan

Total
number Percent1"
0
0
0
0
0
0

0
0
0
1 I.I
0
1 0.6
0
1 I.I
1 0.6
0
0
0
Average
latency
time
(„.)<
	
...
.__
—
	
_-.

	
—
...
30.0
	
10.0
—
7H.O
78.0
	
	
	

-------
                                                                                  TABLE 9-49.   (continued)



Anlnu
ils with tumors
Kncephallc tunorB



(•rmip f'onrenlrHl Ion Sex
1 2VI-I5II ppme M
F
H and f
II 'ill i ppm H
F
H and f
V Controls In N
ih.imhi'rs F
M and >
V\ l.nntrul* N
t
H and F
Neuruhl iqtom.is
Avur.iRe
Inleiif y
Toi.il time
numhvr Percent11 (wk)d
1) 	 	
0 	 	
o
o
0
0 	 	
o
O 	 	
o
II — —
I)
0,
o
o — —
n
o — —
o
o
Other sites


Total
number
0
0
0
1
1
2
0
2
2
0
0
0
1
|
2
0
3
3


Percent"
_—_
	
	
1.1
I.I
I.I
1.1
	
2.2
	
	
	
I.I
I.I
I.I
	
1.3
1.7


Total
number
9
II
20
16
6
20
4
4
8
Ih
9
25
8
4
12
7
8
IS
Others


Benign
numbe r
7
5
12
14
}
17
2
3
5
15
5
20
7
2
9
7
5
12


Malignant
numbe r
2
6
8
0
3
3
2
1
3
1
4
5
1
2
3
0
3
3


Number
15
54
69
20
56
76
13
43
56
30
65
95
17
38
55
14
56
70


Perr«ntn
16.9
60.1)
38.5
22.2
62.2
42.2
14.6
47.8
31.3
33.3
72.2
52.8
18.9
42.2
30.6
15.6
62.2
38.9
Totals'
Number ol
HI ffprrnt
tumiirs/tufliiir-
beiirliiK nnfmals
1.2
2.0
1.9
1.6
1.7
1.7
I.I
l.n
1.5
1.4
1.8
1.7
1.3
1.9
1.7
1.1
1.7
1.6
Fxpnsiin  by Inh.iI it Ion  to Flic  In air nt 250-151) ppm.  5U  ppm,  10 ppm, and 5 ppm, 7 hours/day,  5 days/week,
 fur  78  upi-ks.   K.-sults after  148 week's (end of experiment).
"Spr.iK'ie-ltawluy r.its, 12 weeks  old at stnrt.
blhe  pere i'iil.ices rufer  to the  corrected numbers.
cAv<*r.i>>e •tf.f - onset of the first mammary tumor  per .mlmal detected at the periodic  control or at autopsy.
dAver.iKi' time from the  itnrt nf  the experiment to detection at  the periodic control oc  at autopsy.
eAfter A frw week's I he  dr>
-------
                                                      TABLE 9-50.  MAMMARY TUMORS IN SPRAGUB-DAWLEY RATS

                                                             EXPOSED TO 1,2-DICHLOROETHANE (EDC)
o
r-
Mammary tumors


Croup Concentration
I 250-150 ppod


II 50 ppm


III 10 ppm


IV 5 ppm


V Controls In
chambers

VI Controls


Total


Sex
M
F
M and F
M
F
M and F
M
F
M and F
M
F
M and F
M
f
M and F
M
F
M and F

Animals
Number
at start
90
90
180
90
90
180
90
90
180
90
90
180
90
90
180
90
90
180
1080

Corrected
number8
89
90
179
90
90
180
89
90
179
90
90
180
90
90
180
90
90
180
1078

Total
number
11
52
63
10
58
68
5
43
48
11
65
76
8
38
46
5
52
57



Percent b
12.3
57.7
35.2
11.1
64.4
37.8
5.6
47.8
26.8
12.2
72.2
42.2
8.9
42.2
25.5
5.5
57.8
31.7

Average
latency
time
("k>c
92.9
78.6
81.1
88.8
78.5
80.0
60.8
79.4
77.5
110.2
83.2
87.1
85.5
83.3
83.6
92.0
85.5
86.1

Number of
tumora/ tumor-
bearing animals
1.1
1.9
1.7
1.4
1.5
1.5
1.0
1.4
1.4
1.4
1.6
1.5
1.0
1.8
1.6
1.0
1.5
1.5

                                                                                                  (continued on the following page)

-------
                                                                               TAKI.F  9-SII.   (conHini
 I
vD
Mammary tnmorse
Hlstologlcal evaluation
Haimary tumors

C.ritiip Concent r.il Inn Sex
1 2 SO- ISO pin1' M
F
H .mil F
1 1 SO ppm M
r
M .111.1 F
III II) ppm M
1-
H .mil F
IV S ppm M
F
M riiiil F
V Com nils In M
chambers r
M *nd h
VI Controls M
r
H and r
Tnr.il
number
9
SO
S9
<,
SI
h2
S
IH
41
II
61)
/I
H
(S
4)
S
49
S4

Percent'
HI. 8
96. 1
91.6
til.H
Ol.'i
41.2
10(1. (1
MH.4
Hl.h
1llll.ll
92. 1
91.4
IIIU.O
9J.1
MI.S
Mill. II
94.2
9'.. 7

Nit m her
,
47
54
7
'.9
S6
1
11
)6
1 1
Sh
67
7
27
34
•j
',}
so

Per.entB
77.8
94.0
91. S
77. H
92.4
111. )
1,11.0
Hh.U
HI. 7
IIHI.II
91. 1
94.4
87. S
II. \
79.1
(,0.0
9S.9
92.6
Average
time
nsiiru  hy  Inlinl.illnn Hi I.IK  In .ilr .it  2SO-ISIJ ppm, SO  ppm. HI ppm,  anil "> |>pm.  7  hours/d.iy,  S  tl.iys/uoi'
             for  /H ui'fks.  Ki'sulls nfli-r  I4H wcok«  (enil ol .-tpHr Ini'nt).
            s  illvc afli-r  12 wucks,  uhun the  first  tnnnr (a uiminnry crfrrlnnim) w.is observed.
            ''Tlii1  pt-ri unt.*Kt"«  refer to C h<»  currecLeil  nunhfrs.
            < Avur.u'e  i^c >IL the onset of lliv flrht mammary tiimur per .inlnal iletc* ted .it the  periodic (nntrul  or nt
            rfAflfr .1  few wi-vks the dose unadi>nnm.is,  c.ir. Inoraas. sarcomas, cdrLlnoS'irconus) may
             hi' pn-sunt  In the same .inlm.il.
            'The  per i i-nin^i".  refer to lulal number of  animals hr.irlni; mammary tumors.
            Erin-  PITI i-nt.i|>,i's  refer to total numher nt  .inlm.iK ht-arlni; rn.imn.iry mmors, hi<:tulat;lcal ly examined.

            SlUiKCr    Haltonl  et dl.. 1VHII.

-------
     9.5.1.5.  MALTONI  ET  AL.  (1980) MOUSE  STUDY — Four groups of Swiss mice




(180 mice of both  sexes per  group) were  exposed to EDC, at the level of purity



previously  described  for the rat inhalation study, in concentrations of 250-150




ppm, 50 ppm, 10  ppra,  and 5 ppra,  respectively,  7 hours per day, 5 days per week,




for 78 weeks.  After  several days of 250 ppm exposure, the mire began to exhibit



severe toxic effects, and  the  concentration was reduced to 150 ppm.  One group




of 249 mice served as controls.   The design of the experiment is given in




Table 9-51.



     At the end  of the  treatment period,  the animals were allowed to live un-




til spontaneous  death.   The  remainder of  the procedure was the same as that



described previously  for the Maltoni et  al.  (1980) rat study.




     The survival  rates for  female mice  were slightly lower in the group treated




with EDC at 250-150 ppm.   The  survival rates for mice at 52 and 78 weeks of age



are shown in Table 9-52.   At 52  and 78 weeks,  the overall survival rates were



82.4% and 45.9%, respectively.   The survival rates after 52 weeks from the




start of the experiment are  also given in Table 9-52.  The overall survival



rate was 67.8%.



     The results of histopathologic analysis of various tumors are shown in




Table 9-53.  These results do  not indicate  a statistically significant increase



in the incidence of any specific type of  tumor in the treated mice as compared



with controls.




     In conclusion, although Maltoni et  al.  conducted extensive carcinogenicity




studies in  rats and mice,  no significant  incidences of tumors were seen in any




of the target organs.   Several factors may  have contributed to these findings,




such as differences in  the strains of rats  used, and pharmacokinetic differences




in rates of formation and  retention of reactive metabolites in the target organs



between gavage and inhalation  routes of  administration.






                                     9-194

-------
            TABLE 9-51.  DESIGN SUMMARY FOR 1,2-DICHLOROETHANE (EDC)
                            EXPERIMENT IN SWISS MICE
Animals3
Group Concentration
I 250-150 ppmb

II 50 ppm

III 10 ppm

IV 5 ppm

V Control

Sex
M
F
M
F
M
F
M
F
M
F
Number
90
90
90
90
90
90
90
90
115
134
aSwiss mice, 11 weeks old at start.  Exposed 7 hours/day, 5 days/week, for 78
 weeks.
^After a few weeks the dose was reduced to 150 ppm,  because of high toxicity
 at the 250 ppm level.

SOURCE:  Maltoni et al., 1980.
                                     9-195

-------
            TABLE 9-52.  SURVIVAL OF SWISS MICE EXPOSED TO 1,2-DICHLOROETHANE (BOC) AT 52 and 78 WEEKS8
Survivors at
52 weeks

Croup
I

II

III

IV

V

Dose
(ppm) Sex
250-150 ppmb H
F
50 H
F
10 M
F
5 M
F
Control H
F
Initial
numbers
90
90
90
90
90
90
90
90
115
134
of
Number
56
75
68
83
74
86
54
84
91
127
age
Percent
62.2
83.3
75.6
92.2
82.2
95.6
60.0
93.3
79.1
94.8
Survivors at
78 weeks
of
Number
26
44
30
49
34
50
26
68
42
76
age
Percent
28.9
48.9
33.3
54.4
37.8
55.6
28.9
75.6
36.6
56.8
Survivors after
52 weeks from
start
Number
39
58
46
73
59
72
42
84
72
112
of study
Percent
43.3
64.4
51.1
81.1
65.6
80.0
46.7
93.3
62.6
83.6
"Mice were 11 weeks old at the start of the experiment.
bAfter a few weeks the dose was reduced to ISO ppm, because of high toxlclty at the 250 ppm level.

SOURCE:  Maltont et al., 1980.

-------
TARI.F 9-%
              TtfHIlK INCNIFNCF IN SWISS HICK
                                                     HI I ,2-niCIIUIROHHANF  (KnC)






Mammary tumors
Anlnals"
Croup Cunrent mt 1 »n leu
I 2SH-\3« pprf M
F
H and ^
1 1 30 ppn H
F
H and F
III ID ppn H
F
H and t
IV 5 npm H
F
H and F
V Control;, M
F
M and F
Total
Number
at st.irt
90
911
180
911
9J
Ihl)
91)
90
18(1
9I>
9(1
ISO
115
134
249
969
Cnrrei tt
-------
                                                            TABLE 9-51.  (continued)
 I
I—1

00





Croup Concentration Sex
1 250-150 ppaf H
r
H and F
11 V) ppm H
F
N and F
III 10 ppm M
F
H and F
IV i ppm H
F
M and F
V Controls In H
chambera F
H and F
VI Controls N
F
H and F


Nephroblastomas
Average
latency
Total tine
number Percent (wk)'1
n ___ ___
0 	
I)
n __. __«
0 	 	
0
0
n — — — ___
o
n 	
0
o
o
o
0 	 	
0 	
0
U ™ ™
Animals with tumors

Kidney adenocarclnomas
Averse
latency
Total time
number Percent0 (wk)H
0 —
o
0
0 	 	
0 	 	
o
0
0 — -—
o
n _-. ___
1 I.I 62.0
1 0.6 62.0
n — — _—
o
Q __— ___
n ___ ___
o
n __._ «__


Liver
Average
latency
Total time
number Percent0 («k)d
O — — — —•
o — —
o
0 ___ __«
0 	 	
o
o
fl — — — — 1_
0 ™ —
o
f) .•__ ___
fl — — — — - -
o
n — — «»
o
0 	 	
0 	 	
o




Total
number
1
0
1
0
0
0
0
0
0
0
0
0
0
0
0
1
0
'


Other sit


Percent0
1.2
—
0.6

	
	
	

	
	
	
	
	
	
	
0.9
—
0.4


es
Average
latency
tine

40.0
	
40.0
	
	
	
	
	
	
121.0
131.0
1^8.0
	
	
	
69.U
	
69.11
                                                                                                                                                (continued on the following page)

-------
                                                           TAK1F. 9-53.   (continued)
vO
vO







AnRlomas

Animals
«lth tumors



and f IbroanKlomas
Liver
Crimp CnnciMiir.il Ion Sex
1 JSO-ISO ppm'' M
F
M and F
1 1 VI ppm N
F
H nnil K
III to ppm H
»
H and f
IV S pna H
K
H and F
V Cnntrnll In H
chambers F
H And F
Tut a I
number
(1
11
1)
II
1
1
0
O
0
0
0
II
n
0
0
Per<.entc
	
	
I.I
H.6
	
	
	
	

	
0.9
	
11.4
Average
latpnLy
time
—
	
R3.0
S3.0
	
	
	
	
__-
	
69. n
—
69.0
Total
number
0
0
0
|
1
i
0
1
1
0
0
n
,
0
1
Other sites
Percent0
	
I.I
I.I
1.1
	
1.1
0.6
	
...
	
0.9
	
0.4

Average
latency
time
	
76.0
83. 0
79. i
93.0
93.0
	
	
_..
	
76.0
	
76.0
Hepatomas
Average
latency
Total time
number Percent' (wk)''
0
o
o
0
o
0
0
o
o
o -— ---
o
4 3.5 82.0
A _ 	 ___
4 1.6 82. 0
Forestnnurh vulthHIal
tunnrs
Total
number
0
0
0
0
0
0
0
0
0
0
0
0
0
0
0
Average
latency
tine
Percent."1 (uk)d
	
	 	
	 	
	 	
	 	
	 	
	 	
	 	
...
	 	
	 	
	 	

                                                                                                                                               (continued on the follm*Ing page)

-------
                                                                                                  TABI-F. 9-51. (continued)
O
O




Croup ronrenlr.it Ion Sex
1 2511-150 ppra? M
F
H and t
1 1 5ll ppm H
F
H and F
III III ppm M
K
H and F
IV 5 ppm M
F
N and F
V rVintrnls In N
chambers F
M and F

Skin

Total
number
II
II
I)
I)
1

0
I)
0
0
1
"
1
1
2

epithelial tumors
Average
latency
time
l'ercentc (wk)d
___ _ 	
	 —
	
	 	 ___
I.I 101.0
ll.h 11)1. 0
___
	 	
—
	 	
I.I 79.0
ll.h 79.0
0.9 109.0
0.7 77.0
O.R 91. 1)

Animals
with tumors
Subcutaneous sarcomas

Total
number Percent0
0
0 	
11
0
0 	
0
1 I.I
1 I.I
2 I.I
0
0
(1
1 0.9
O 	
0 (1.4
Average
latency
lime
(wk)11
___
	
—
___.
	
—
77.0
25.0
51.0
	
	
	
lOh.O
	
Illh.O

Total
number
0
2
2
1
4
s
,
2
1
1
4
5
1
4
5
Others

Benign
number
0
2
2
0
1
1
,
1
2
0
3
1
1
2
1
Total'

Malignant
number
0
0
0
1
1
2
0
1
1
1
1
2
0
2
2


Number
2
II
11
9
15
24
12
17
29
4
19
23
14
27
41


Percent1"
2.4
12.9
7.7
III. A
17.2
14.0
11.5
19.1
Ih.l
5.7
21.1
14.5
12.4
20.1
In. ft
Number of
different
tumors/tumnr-
hearlng animals
1.0
1.0
1.0
1.2
I.I
1.2
I.I
1.0
I.I
1.0
1.0
1.0
1.4
1. 1
1.2
       hxposun-  hv lnli.Tl.it Ion to H)f  In  all  Hi J 511- 150 ppm, 50  ppm,  K) ppm, and  5 ppm, 7 houri/day, 5 ddys/wcrk,
        fur  ?H wei-k-.  Krsuli-  liter  119 weeks (cml  ul  exprr linrnt ).
       (1swlsK rnlif,  II wi>i*ks *ilil at start.
       "AnlmiK  illvo  ilti-r  l'> wi-i-ks,  wliun tht* flr>.l  I umor (n  Ivnki-ml.i) w.i<. ohst-rvcd.
       1 llu>  pi'i 1 1 nl.ij't's  refrr to llif  t MTIIM tfil numlitTs.
         time t rum  tin- st.irt of  the experiment  to detection nt  tin- periodic control  or at autnpuy.
       eAfti'r  i  I iiw wfeks  tin1 dose was  n ducpcl to  1 5
-------
     9.5.1.6.  THEISS ET AL. (1977) MOUSE STUDY — Theiss et al. (1977) con-

ducted a pulmonary tumor bloassay with EDC and other organic, contaminants of

drinking water in the United States.  The compounds were injected intraperito-

neally into 6- to 8-week-old mice of the A/st strain.  Each dose of reagent-

grade EDC, with tricaprylin as the vehicle, was injected into mice in groups

of 20, three times a week, for a total of 24 injections per mouse.  Dose levels

of 20, 40, and 100 mg/kg (the maximum tolerated dose) were used.

     The mice were sacrificed 24 weeks after the first injection, and their

lungs were placed in Tellyesniczky*s fluid.  After 48 hours, the lungs were ex-

amined microscopically for surface adenomas, and the frequency of lung tumors

in each group was compared with that in a vehicle-treated control group by means

of the Student's t Test.  The incidence of lung tumors increased with dose, but

but none of the groups had pulmonary adenoma responses that were significantly

greater (p > 0.05) than that of the vehicle-treated control mice (Table 9-54).

Because they found a nonsignificant elevation in pulmonary tumor response, the

authors suggested further investigation of the carcinogenic potential of EDC.
    TABLE 9-54.  PULMONARY TUMOR RESPONSE IN STRAIN A/st MICE INJECTED WITH
                            1,2-DICHLOROETHANE (EDC)
Dose/ Inject ion
(mg/kg)
Qa
20
40
100
Number of
injections
24
24
24
24
Fraction
surviving
46/50 (92%)
14/20 (70%)
16/20 (80%)
20/20 (100%)
Number of
tumors /mouse
0.39 + 0.06
0.21 + 0.06
0.44 + 0.11
0.75 + 0.17
aTricaprylin vehicle.

SOURCE:  Theiss et al. ,  1977.
                                     9-201

-------
     9.5.1.7.  VAN DUUREN ET  AL.  (1979) MOUSE STUDY — Van Duuren et al. (1979)




conducted a bioassay of EDC and  of  a suspected metabolite, chloroacetatdehyde,




as initiators, promoters, and complete carcinogens using two-stage skin tests.




Female non-inbred ICR/Ha Swiss mice were  used, with treatment beginning at 6 to




8 weeks of age.  The experimental group consisted of  30 animals, exclusive of




the no-treatment groups or those receiving  repeated application of phorbol



rayristate acetate (PMA).  The results of  this study are given in Table 9-55.




The compounds, in O.I raL and  0.2 mL acetone, were applied three times a week to




the dorsal skin.  Skin  lesions were diagnosed as papillomas when they reached




approximately  1 mm and  persisted for 30 days or more.




     All animals were examined daily and  weighed monthly; findings were recor-




ded monthly.   Animals in poor health or with large tumor masses were killed.




Animals were completely autopsied at the  termination  of the experiment or at




death.  Tissue sections were  fixed  in 10% formalin, processed, blocked in par-




affin, and stained with heraatoxylin and eosin for pathologic diagnosis.  The




results indicate that neither EDC nor chloroacetaldehyde induced a statistical-




ly significant increase in the incidence  of carcinomas of the skin.  This




result was probably due to the low  dose levels used in the study.  EDC was



found to induce a statistically  significant increase  in the incidence of benign




lung papillomas.




     9.5.1.8.  SUMMARY  — The NCI study (1978), as well as the studies by




Theiss et al.  (1977) and Van  Duuren et al.  (1979), show that, under appropriate




conditions, EDC can increase  the incidence  of tumors.  The data obtained in




mutagenicity studies have also shown that,  under appropriate experimental




conditions, EDC can cause DNA damage (Storer et al.,  1982) and mutations




(Guengerich et al., 1980).   In contrast,  the lifetime inhalation study conducted




by Maltoni et  al.   (1980) showed no increased incidence of tumors.






                                     9-202

-------
                                                         1A»I>  1-SS.
                                                                              SKIN HIilASSAY  lit I ,^-IMl HLIIKOI- CIIANI-  (tl)C) AND (JILOROAO rALnKIIYIIK
N>
O
In 1 1 1 il Inn-pr-Jimtt Ion '
Dnsc Mivs tit Mice with
mj*/.ippl h it 1 mi/ 1 i rst papl 1 lom.is*'/
Compiii in*1 nmiKf* t nmir tnt.i I p»ipt 1 Inmds
MIC i "«.i> is; 1/1


Chloroiri-t ildphyeli' 1." IV 1/1(1)
HHA mm roll
(1211 mltH) II.IUI2S 141 I/ H> (1)
(/' (2)
ArPtniip (II. 1 mL) — — 	

No i rp.ilnpnt
(MID mli-e)

Kcpciti'd iippl i rat ton*1
Iliisv Days to Nice with Nil. ol mire
mi;/appllr.itlnn/ first fapl 1 lnnsc/ with distant
nniioi- tumil total pnplllnm.ii tnmnrs11
I2h.ll 	 0 2h IIIIIK
1 stnnvnh
•'iZ.lt 	 II 17 InilK
1 s t otlil t h
l.ll 	 (1 14 lung
	
	
II. 1 mL 	 II II lung
i il iimn ill
0 III I" OR
*t itom.lrli
                        "Ml appllcHtlons  wr« to tlw  ilorial -skin  hy  mlrmplppl f i-.   HIT. was  .idmt nlntfivil onrc only In O. i  nl. nrptune,
                         fnllnwrd 14 days  later by *> UK of I'MA In  <>..! ml. .icPt.ino  three Hmei wci-kly.  Chlnroan'trtlriohydp u.iv ndmlnls-
                         tervd  unrc only  In O.I ml niPtunr, fcillnuril  14 it.iy.*.  liter  hy 2.1) UK nf  WA In O.I  ml. ncetont- three times
                         weekly.
                        hKIT w,i-  .id ml nlnli-rcd In II.j ml. Hrotnm*.   (.Ill urn.K ctalripliydi* was .idrnl nl if ircrt three t Impo weekly
                         In IV. I nl. acetone.
                        rNnnihprs  nf rolri" with •sqiMmiHif coll .-.ir.-lnniws .ire' Klven  In pirenthcispn.
                        <*A1 I I"")! lnnnr  Klvon only  where  siRnlflcant,  I.e., p <  O.!)1).

                        SOIIKCh-   Adapted frnn V.in I	n-n ct al., l*)7<>.

-------
     The apparent discrepancies  between  inhalation and other routes of exposure




in the oncogenicity studies  have led  to  considerable discussion of the conduct




and results of these studies (Maltoni  et  al.,  1980; Hooper et al., 1980; Reitz




et al., 1982) (see discussion in section  9.5.3.).  The NCI Clearing House




Committee review of the NCI  bioassay  report  (1978) acknowledged several design




features which may have adversely affected the outcome of the experiment.  For




example, the studies were  conducted in rooms housing animals dosed with other




volatile carcinogens.  It  was concluded,  however, that these shortcomings were




not significant enough to  invalidate  the  findings.  Other investigators (Hooper




et al., 1980) have concluded that unless  unusual and unexpected synergisms




occurred, the presence of  other  animals  in the test rooms would not have affected




the results.




     Regardless of the explanations for  the  observed differences  (Hooper et




al., 1980; Reitz et al., 1982),  the data  show  both an ability of  EDC to produce




neoplasms and an inhalation  level at  which no  tumors are formed.  This may be




due to the difference in rates of formation  of reactive metabolites, rates of




formation of DNA-adduct, and rates of  deactivation between the oral and inhala-




tion routes.  Thus, the results  indicate  that  EDC is carcinogenic in rats and




mice, and is also a mutagen.




9.5.2.  Epidemiologic Studies.   No epidemiologic studies have been published




regarding the carcinogenicity of EDC.




9.5.3.  Risk Estimation from Animal Data.  Evidence for the carcinogenicity




of EDC consists principally  of positive  long-term gavage studies  in mice and




rats (NCI, 1978), a suggestive positive  pulmonary tumor bioassay  in mice (Theiss




et al. , 1977), and a suggestive  positive  two-stage mouse skin painting bioassay




(Van Duuren et al. , 1979).   The  NCI bioassay reported excess incidences of




forestoraach squamous cell  carcinomas,  circulatory system heraangiosarcomas, and




subcutaneous tissue fibroma^ in  male  rats, and in fi'nwlr> r^ts, mammary inland




                                      9-204

-------
adenocarcinomas and hemangiosarcomas.  In male mice, an excess Incidence of




hepatocellular carcinomas and bronchiolar adenomas were found, and in female



mice, bronchiolar adenomas, mammary carcinomas, and endometrial tumors.




Theiss et al. reported that their pulmonary tumor bioassay resulted in a dose-




related increased frequency of lung adenomas in mice, while Van Duuren et




al. found an increased incidence of benign lung papillomas in mice treated




with EDO by skin painting.  In contrast to these positive reports, lifetime




inhalation studies in mice and rats by MaItonI et al. (1980) showed no excess




incidence of tumors.  Part of the explanation for these differing results




may be the differences in the pharraacokinetics and metabolism of EDC for the




two routes of administration, as discussed below.




     It is generally assumed that the carcinogenic potential of EDC is dependent




on the metabolism of the parent compound to nucleophilic metabolites (such as




chloroacetaldehyde, the half-sulfur mustard S-(2-chloroethyl)-GSH, and its



episulfonium ion) that extensively covalently bind to cellular lipids and




proteins with organ localization and binding intensity paralleling sites of




tumor formation in liver, lung, spleen, and forestomach (Reitz et al., 1982).




DNA adduct formation with EDC metabolites has also been demonstrated both in




vitro (Banerjee and Van Duuren, 1978, 1979; Banerjee et al. , 1980; Guengerich




et al., 1980) and in vivo (although at a low level) (Reitz et al., 1982; Storer



et al., 1982).




     9.5.3.1.  GENERAL OBSERVATIONS — For EDC, the positive results of the




NCI (1978) oral gavage lifetime studies in male Osborne-Mendel rats and male




B6C3F1  mice showing a statistically significant dose-related excess of tumors




(hemangiosarcomas in rats and hepatocellular carcinomas in mice) were select-




ed for risk assessment.   Tumors were found at both "high" (maximum tolerated




dose)  and  "low" doses (one-half maximum tolerated dose), as  summarized in






                                     9-205

-------
Table 9-56.




     The chronic oral  dosage  used in  the  NCI rat  bioassay (95 and 47 mg/kg/




day) was well below  (8-  to  16-fold) the acute  LD5Q  for rats (680 to 770 mg/kg;




McCollister et al. ,  1956; Smyth,  1956; Heppel  et  al., 1945).  Similarly, the




chronic oral dosage  in the  NCI  mouse  bioassay  (195  and 97 mg/kg/day) was well




below the acute LD5Q report for mice  (489 and  413 mg/kg male and female,




respectively; Munson et  al. ,  1982).   Nonetheless, signs of toxicity from




chronic administration of CDC (less weight gain,  shortened life span) were




evident in the NCI bioassay for both  rats and  mice.   Presumably EDC is sub-




jected to elimination  processes during the 24-hour  interval between doses (and




on weekends), and  hence  the chronic dosage of  the NCI assays may not be




cumulative.  EDC has a short  half-life in rodents after acute oral administra-




tion (in corn oil) of  1 hour for a 150 mg/kg  dose  (Spreafico et al. , 1979,




1980); hence, the  conditions  of the NCI study  could  be represented kinetically




as repeated (multiple) acute  exposures rather  than  chronic exposure in which




the administration is  repeated  at intervals  (24 hours in this case) that are




shorter than the time  required  to clear all EDC from the body.  Thus the




toxicity of EDC evident  in  the  NCI gavage study of  rats and mice may not be




due to the intact  EDC  molecule  per se but to cumulative cellular damage from




covalent binding of  reactive  metabolites  from  EDC metabolism.  It is of in-




terest that the Indices  of  toxicity (weight gain, depression, mortality) in




the NCI gavage study were not more severe for  mice  than for rats, although the




maximum and half-maximum "tolerated"  time-weighted  average daily dose was




twofold greater for  mice than for rats on a mg/kg basis (Table 9-56).  How-




ever, on a proportional  surface area  basis for the  two species (rat, 500 g;




mouse, 33 g)''^ =6.1, the  doses  given to rats in terms of mg/antmal were




actually slightly greater than  those  given to  mice  (ratio, ratrmouse, 7.4;






                                      9-206

-------
                          TABLE 9-56.   INCIDENCE RATES OF HEMANGIOSARCOMAS IN THE CIRCULATORY SYSTEMS OF MALE
                               OSBORNE-MENDEL RATS AND OF HEPATOCELLULAR CARCINOMAS IN MALE B6C3F1 MICE
XO


O
Average
terminal
Species Sex weight (g)
Rats M 500


Mire M 33


Time-weighted average
gavage dosea
mg/kg/day
0
47
95b
0
97
195
mg/animal/day
0
23.5
47.5
0
3.2
6.4
Incidence
0/40
9/48
7/27
1/19
6/47
12/48
(0.0%)
(19%)
(26%)
(5%)
(13X)
(25%)
          aGavage dose  in  corn  oil  administered  5 days/week with  a  time-weighted average dose of: dosage x weeks received
           divided  by all  weeks receiving EDC.   Mg/animal  based on  average  terminal weights of  rats (500 g) and mice  (33 g).
          bAll animals  In  this  group  died before the  bioassay was terminated.

          SOURCE:   NCI,  1978.

-------
Table 9-56).



     The  fact  that  the NCI  and  Maltoni et  al.  carcinogenicity bioassays were




conducted using  two different routes  of EDC  administration (oral and inhala-




tion, respectively),  presents substantial  interpretive difficulties, pharmaco-




kinetic and otherwise, for  the  evaluation  of the  positive results of the NCI




study and the  negative findings of  the Maltoni et al. study.  A major reason




for these difficulties is evident  from a consideration of the dosages used in




the two bioassays.  The  time-weighted average  dose levels administered by




gavage per day to Osborne-Mendel rats in the NCI  study were 47 and 95 mg/kg




(5 days/week for 78 weeks);  the inhalation doses  for  rats (S-D) in the Maltoni




study were 150,  50, 10,  and 5 ppm,  7  hours/day, 5 days/week, for 78 weeks.  In




both assays higher  doses (NCI,  150  mg/kg;  Maltoni,  250 ppm, 6 hours) were




initiated but  were  reduced  after a  few weeks of treatment.  The data of Reitz




et al. (1982)  provide an approximate  comparison of these doses.  These inves-




tigators determined the  assimilated dose of  Osborne-Mendel rats given a single




oral administration of EDC  (150 mg/kg) and a single inhalation exposure (150




ppm, 6 hours).   These doses represent the  highest levels of EDC administered




for any significant length  of time  in the  two  bioassays.  The assimilated EDC




from the  150 rag/kg  gavage dose  was  observed  to be completely absorbed (150




mg/kg), while  the inhalation exposure was  estimated to represent an average




assimilated dose of 50.7 mg/kg, i.e., one-third of the oral dosage (Table




9-57).  Furthermore,  the assimilated  dose  from 150 ppm inhalation exposure to




rats (the highest dose level of the 150, 50, 10,  and  5 ppm Maltoni et al.




bioassay), 50.7  mg/kg, is approximately equal  to  the  lowest time-weighted dose




of the NCI bioassay of rats but is  less than the  lowest dose given to mice




(Table 9-56).  Therefore, in respect  to dosage levels of the NCI and Maltoni et




al. bioassays, it appears that  the  animals in  the Maltoni study were subjected






                                      9-208

-------
                  TABLE 9-57.   DISPOSITION OF 14C-EDC IN RATS3
                AFTER SINGLE ADMINISTRATIONS OF 150 MG/KG ORALLY
                     AND 150 PPM FOR 6 HOURS BY INHALATION
                              Oral                       Inhalation
                             mg/kg                         mg/kg
Assimilated doseb          152.3 ± 38.6                  50.7 ± 13.4

Exhaled air                 44.2 t  5.9  (29.0%)          0.9 ±  0.04  (1.8%)
  (unchanged EDC)
Metabolized
C02
Urine
Feces
Carcass
Cage wash


8.2 ±
91.6 ±
2.6 t
4.6 ±
1.2 ±
108.2

1.2
34.4
1.5
1.5
0.6
(71.0%)

3.6 ±
42.8 ±
0.9 ±
2.2 ±
2.2 ±
49.8

0.7
11.9
0.3
0.3
0.3
(98.2%)
aOsborne-Mendel rats, 150 to 250 g.
^Radioactivity recovery in exhaled air, urine and feces, 48 hours after admin-
 istration.  The values given are means ± SD for 4 rats for each route of
 exposure and are milligram equivalents of ll*C-EDC radioactivity.

SOURCE:  Adapted from Reitz et al., 1982.
                                     9-209

-------
to lower effective dose  levels  than  those  in  the NCI mouse study and were, at




most, comparable  to  the  low-dose  group  in  the NCI  rat study.  Other evidence of




the effective dosage difference between the two bioassays is summarized in




Table 9-58 and discussed in  section  9.5.3.2.5.




     9.5.3.2.  INTERSPECIES  DOSE  CONVERSION




     9.5.3.2.1.   General Considerations — This section discusses the metabolism,




pharmacokinetics, covalent binding,  and toxicologic findings that are relevant




to the carcinogen risk assessment of EDC.  Information on the metabolism




and kinetics is used to  the  extent possible for calculating the carcinogenic




risk for EDC.



     Scaling of toxicologic  effects, including carcinogenicity, among species




has been elaborated  in several  published papers (Freireich et al., 1966; Gillette,




1976; Krasovski,  1976; Dedrick, 1973; Dedrick and  Bischoff, 1980;' and Gehring




et al., 1980).  The  major components requiring consideration in determining an




appropriate extrapolation base  for scaling carcinogenicity data from laboratory




animals to man are:   1)  toxicologic  data,  2)  metabolism and kinetics, and 3)




covalent binding.  The published  literature concerning the biological basis




for extrapolation of the dose-carcinogenic response relationship of laboratory




animals to man has been  reviewed  in  a paper prepared by Davidson (1984) for




the Carcinogen Assessment Group and  presented by Parker and Davidson (1984).




     9.5.3.2.2.   Toxicologic Data — Section  9.2 of this document outlines the




general features  of  the  acute,  subchronic, and chronic toxicities of EDC in




man and animals.  However, comparative  quantitative data across species on




specific indices  of  EDC  toxicity  are experimentally lacking.  Gross measures




such as acute oral LD5Q  are  similar  in  rats and mice (680 to 770 mg/kg, rats;




413 to 489 mg/kg, mice)  and  reflect  overwhelming CNS and cardiac depression




effects rather than  specific organ toxicities (McCollister et al., 1956; Smyth






                                      9-210

-------
         TABLF. 9-58.  COMPARISON OF THt: METABOLISM AND PMARMACOKINETICS
          OK THE "HIGH" DOSKS IN RATS OF THE MALTONI ET AL.  INHALATION
                            AND NCL (JAVAGK IUOASSAYS
    Observation
Maltonl et al,
 inhalation
   150 ppm,
   7 hours
 5 days/week
    NCI
   gavage
 150 mg/kga
5 days/week
Assimilated dose

 ADC, pg/mL x min
   Spreafico et al.
   Re it 7. et al.
     350b
    2910
  7297
  4500
 Balance study with
   1"C-EUC, mg/kg
   Keitz et al.

 Fraction metabolized,
   mg/kg
   Reicz et al.

Peak blood levels, \tg/mL
      50.7
      49.8
   150
   108.2
Spreafico et al.
Reitz et al.
Time to peak blood level, hr
Spreafico et al.
Reitz et al.
Blood clearance time, hr
Spreafico et al.
Reitz et al.
Hepatic DNA covalent binding
gmole equiv E DC/mole DNA
Reitz et al.

lib
9.4
4 to 6
2.5 to 6
l.5b
1.5C

8.2d
3.3
66.8
30 to 40
0.6
0.25
5.6
5

21.3
L3.9
aTime-weinhted average for 78-weok treatment period was 95 mg/kg/day.
blnterpolation from the data of Spre-ifico et al. (1978, 1979, 1980), Table 9-56.
cRstimate from data of Keitz et al.  (1982), Figure 9-16.
"Determined 4 hours after gavaye .me' immediately at termination of inhalation
                                     9-2 1 I

-------
1956; Munson et al.,  1982).   Other  toxicologic end points have been measured,




but not across species  in  comparative  dose-related comprehensive studies.




Differences in toxicity according to species, strain, or sex have not been




reported for EDC.




     9.5.3.2.3.  Metabolism  and  Kinetics — Few data are available on the




metabolism and kinetics of CDC in man,  but these kinetics have been studied in




mice and rats.




     9.5.3.2.3.1.   Metabolism.   In  mice and rats, EDC is extensively metabolized




after both oral and inhalation exposures.  After oral and inhalation exposures




in rats, at 150 rag/kg and  150 ppra for  6 hours ("high" doses in the NCI and




Maltoni et al. carcinogenicity bioassays), 71% oE the oral dose and 98% of the




inhalation dose was metabolized  (Table 9-57).  However, as outlined below,




metabolism is dose-dependent, with  proportionately greater percentages of the




dose metabolized at low doses and less at higher doses.  Metabolism occurs by




multiple pathways  in liver,  microsomal, and cytosol functions.  Microsomal P450




oxidation produces  2-chloroacetaldehyde, while cystosolic reactions of EDC with




glutathione are postulated to produce  the half-sulfur mustard S-(2-chloro-




ethyl)-GSH and its  episulfonium  ion.   Both chloroacetaldehyde and the half-




sulfur mustard are  reactive  metabolites that covalently bind to cellular




macroraolecules, a mechanism  that is presumed to underlie the liver and other




organ tissue cytopathology observed from EDC exposure.




     The cytosolic  interaction of EDC  with glutathione results in an activation




to a highly reactive half-mustard metabolite as well as to nonreactive gluta-




thione conjugates  from  "detoxification reactions."  Johnson (1965) acutely




administered AO mg/kg EDC  in corn oil  to rats by gavage and observed an average




of 52% depletion of hepatic  GSH  within 2 hours (peak EDC blood concentration




after oral dosing of 150 mg/mL in corn oil, 40 minutes; Table 9-56).  Reitz et






                                     9-212

-------
al. (1982) assayed GSH levels in rats after a 150 mg/kg oral dose in corn oil.




After termination of a 150 ppra,  6-hour inhalation exposure, 4 hours after




exposure, hepatic GSH was depleted by 77% and 74%, respectively.  These obser-




vations demonstrate the involvement and importance of cellular levels of GSH to




the cytotoxic potential of EDC,  both for activation and for detoxification.  It




would appear that the "high" doses in the carcinogenicity bioassays do not




completely overwhelm the availability of cellular GSH.




     Species Comparison.  The metabolism of EDC has been studied in only two




species, the rat and mouse.  There is no evidence that the metabolic pathways




in these two species differ qualitatively, since urinary end-metabolites are




similar.  In these two species,  metabolism has been found to be capacity-




limited, with dose-dependent kinetics.  There are, however, no direct compara-




tive balance studies (same laboratory, same time period) of the extent of




metabolism in these species, as  are available, for example, for certain of the




halogenated ethylene compounds.




     For the mouse, Yllner (1971b) provided data on the extent of metabolism




in this species (Figure 9-12);  10 years later, for the rat, Reitz et al. (1982)




found that for a gavage dose of  150 mg/kg (a dose approaching "saturation" for




rat and mouse), 108 rag/kg was metabolized (Table 9-57).  Thus, for a 200 g rat,




21.6 rag of a 30-mg dose was metabolized.  For a comparable dose (150 mg/kg),




Yllner found that 2.25 tng was metabolized by a 25 mg mouse.  The ratio of




metabolism, rat:mouse, for this  particular dose (150 mg/kg) is 9.6, while the




ratio of the surface areas is (200/25)^/3 = 4.0.  Comparison of these data




suggests that metabolism of EDC  in the rat and mouse is more associated with




the relative body weights of these species than with surface area.  However,




for the reasons given, direct comparison of data from these two diverse studies




cannot be fully justified on experimental grounds.






                                     9-213

-------
                                                                       B
\0
to
                             25  50 75  100 125 150 175 200
                                  DOSE
0  25  50  75 tOO 125 150 175 200
       DOSE
               Figure 9-12.  Relationship  in  mice  between dose of  EDC (nig/kg) and the amount of EDC exhaled
               unchanged (Panel A) and  the amount  of  the dose metabolized to urinary metabolites plus CO2
               (Panel B).  The female NMRl mice weighed  20-30 g.
               SOURCR:  Adapted  from Yllner,  I97lb.

-------
     9.5.3.2.3.2.  Kinetics.  Conclusive evidence exists that the pharmacoki-




netics of EDC in rats and mice are nonlinear.  To detect and establish the




nonlinear nature of the pharmacokinetics of a compound, at least two, and




preferably more, dosage levels must be studied (Wagner, 1975).  For EDC, the




studies of Yllner (1971b) in mice, and of Spreafico et al. (1978, 1979, 1980)




in rats meet this essential requirement, while the single-dose studies of




Reitz et al. (1982) do not.




     EDC is completely and rapidly absorbed after oral administration (Yllner,




1971b; Spreafico et al., 1978, 1979, 1980; Reitz et al., 1982) with a first-




order constant, k , for an oral dose of 150 mg/kg of 0.11 minutes  , indicating




95% absorption within 30 minutes.  Yllner was the first to demonstrate both the




nonlinear nature of the pharmacokinetics of EDC after absorption and the sub-




stantial first-pass effect with EDC administration.  Yllner administered ^C-




EDC intraperitoneally in olive oil to NMRI mice at four dose levels, 50, 100,




140,  and 170 mg/kg.  This dosage range approximated that of the NCI bioassay




in mice.  Yllner quantitated ^C-radioactivity in urine and feces (as EDC




metabolites), as well as   CO? (EDC metabolite) and unchanged   C-EDC in ex-




haled air for 72 hours after administration.   Total recovery of ^C-EDC




radioactivity ranged from 97% to 102% of the  dose.  The results are shown




plotted in Figure 9-12.  With increased dose, the portion of the dose exhaled




as unchanged EDC (first-pass effect) increased in an exponential manner, so




that at a dose of 170 mg/kg,  45% of the dose  was excreted as EDC.  The remain-




ing portions of the doses were metabolized.   Figure 9-12B shows the nonlinear




relationship between dose and metabolism,  indicative of nonlinear Michaelis-




Menten kinetics for the metabolism of EDC, with "saturation" of metabolism




occurring in the mouse between 150 to 200 mg/kg administered dose.   These




data can he used to estimate  the fraction of  the assimilated dose metabolized

-------
and  contributing  to tumorigenesis for the two  dosage  levels  of  the NCI mouse




bioassay.




      Spreafico et al,  (1978,  1979, 1980)  investigated the  pharraacokinetics




of CDC  in  rats after oral dosing, inhalation exposure,  and intravenous bolus




administration at two  (inhalation) or three dose  levels (oral and intravenous




administration).   These workers determined the blood  EDC concentration-time




curves  (C,t  curve)  after the  various administrations  and fitted  the C,t data




to a  linear  two-compartment open kinetic  model,  thereby testing  the linearity




of EDC  pharraacokinetics.   According to basic assumptions and definitions,




linear  kinetics require that  kinetic parameters  (T  1/2,  AUC, Vj, clearance,




slopes  of  semilog plots of blood concentration decay  curves, etc.) be indepen-




dent  of dose.   Hence,  these kinetic parameters, determined at two or more




dose  levels  in accordance to  a linear kinetic  model,  provide a rigorous test




for nonlinearity  (Wagner,  1975).  If the  magnitude  of some or all of the




parameters changes  with dose  (and especially if there are  dose-related trends),




then  nonlinear kinetics are highly probable.   If  Michaelis-Menten kinetics




are operative,  then one expects (I) that  the percentage  metabolism by the




Michaelis-Menten  path  will decrease with  increase of  dose  (as in the data of




Yllner  above),  (2)  that the area under the blood  C,t  curve will  increase more




than  proportionately with increase in dose,  and (3) that semilog plots of




blood levels will curve inwardly with pseudolinear  terminal portions, and




slopes  of  the  pseudolinear terminal portion will  decrease  with increase in




dose.




     Figure 9-13  presents  the  semilog plots of blood  C,t curves obtained by




Spreafico et al.  (1980) after  oral administration of  three dose  levels of EDC




to rats.  Also shown are  adipose tissue,  lung, and  liver C,t curves.  In each




case, the slope of  the  apparently linear  decline  curves  decreases markedly






                                      9-216

-------
vO
ro
               o
                 7J
               o
               c
               E
               X
               L
               X
               U
                 2.S-
                              1W

                              100

                              00

                              •0
   00
E  so
x
                              30
                              20
                              H>
                                        25
                                                56
1  I  • '  •  • I
75       tea
  DOSE lag/kg)
                                                                         125
                                                                                       ORAL
                                                                                       INHALAflON
                                                                                  •I
                                                                                  159
                                                               175
                                                                        1 I
                                                                       2B0
                                     ' I ' '  ' ' I
                                     25     58
' I  ' ' '  ' I '
75     188
                                    125
                                          i-r-r-r-
                                          158
                    175
                          1 ' I  ' ' '  ' I
                          2BB    225
258
     i i i  | i i
      275
                                                            DOSE Ippn 6 hr. >
               Figure  9-13.  Relationship in Sprague-Dawley rats  between the area under the blood EDC
               concentration-time  curve (AUC) and  the gavage dose of EDC in corn oil (rag/kg) or  Inhaled
               doses of  EDC (ppm for 6 hours).
               SOURCE:   Adapted from Spreafico et  al., 1980.

-------
with increase in dose,  in  a  manner  inconsistent with  first-order linear

kinetics but providing  an  excellent  indication of  the nonlinear dose-dependent

nature of the kinetics  of  EDC  in rats.   Immediately apparent also from Figure

9-13 is that, as a  consequence of nonlinear  kinetics, the higher the dose the

longer and more disproportionate is  the  time of clearance of EDC from the

blood and, therefore, from the body.   Definitive evidence of the dose-depen-

dency of EDC kinetics is given by the  linear kinetic  parameters obtained by

Spreafico et al. by fitting  their data to  a  linear two-compartment model, as

shown in Table 9-59.  Both experimentally  observed measures (peak blood

concentration, time to  peak, areas  under C,t curve) and calculated kinetic

parameters (half-life,  volume  distribution,  clearance) are not independent of

dose, as required by linear  kinetics,  but  rather increase with increase in

dose.  Furthermore,  the nonlinear dose-dependent nature of EDC pharmacoki-

netics is evident for all  three routes of  administration:  oral, inhalation,

and intravenous.  Spreafico  et al.  did not further analyze their data, al-

though it would have been  appropriate  for  them to have fitted their data to a

first-order absorption  model with parallel Michaelis-Menten kinetics, as well

as to a first-order elimination model.

     The area under the blood  C,t curve  (AUC) is an experimental measure inde-

pendent of its assumption  of any kinetic model.  The AUC can be represented by

the definite integral

                                       t
                                  AUC = /  C  dt
                                       o


The AUC is related  to the  amount of  compound absorbed and, under certain

conditions, can be  used to estimate  the  total internal dose.  The AUCs deter-

mined from oral administration of EDC  in corn oil vehicle and from inhalation
                                      9-218

-------
 I
CO
vO
                           TABLE 9-59.  EVIDENCE OF NONLINEAR KINETICS FOR EDC IN RATS AFTEK GAVAGE, INTRAVENOUS,

                                                           AND INHALATION DOSAGE
                                                         Oral
                               Inhalation
            Kinetic parameter
     mg/kg dose3


25       50      150
ppra, 6-hr dose


 50        250
                            Intravenous
     mg/kg dose


25      50      150
Experimental determination'*
Peak blood cone. , pg/mL
Time to peak, hr
AUC, yg x mL~l x mln
Calculated from kinetic model**
Tl/2> mln
Vd, mL
Clearance, mL x min~l

13.2
-0.1
446

24.6
367
10.6

31.9
-0.2
1700

44.1
328
5.6

66.8
-0.6
7297

56.7
390
3.9

1.3 31.3 1.5 8.0
4 6 zero time
26 1023 9 54

12.7 22.1 7.3 9.5
231 239
22.0 17.5

38.1

59

14.1
162
8.0
            aCavage dose administered In corn oil.

            bThese parameters were experimental observations  Independent of any kinetic model.  The calculated parameters

             were obtained by fitting the blood EDC concentration-time data to a linear two-compartment open model.
            SOURCE:  Adapted from Spreaflco et al.,  1978,  1979,  1980.

-------
exposure at two or  three  dose  Levels, respectively,  to rats are shown plotted




against administered dose in Figure  9-L3.  As expected from the dose-dependent




nature of EDC kinetics, the AUCs  increase  disproportionately with the dose




for both oral and inhalation exposures.




     Consistent also with Michaelis-Menten dose-dependent kinetics for the




metabolism of EDC is the  fact  that when  the  logarithm of the AUCs is plotted




against the logarithm  of  the oral dose,  a  linear  relationship is obtained as




shown In Figure 9-14.  Since the  AUCs represent the  portion of the EDC dose




that reaches the systemic circulation, in  the case of oral administration the




AUCs principally represent the  absorbed  dose (assumed to be complete) minus




the first-pass effect  (primarily  elimination of unchanged EDC via the lungs).




Comparison of AUCs  for oral and intravenous  doses  (Table 9-59), a method used




to estimate first-pass effects,  shows that for a  25  rag/kg oral dose of EDC,




the AUC (446 ug x mL~l x  rain)  is  25% less  than for an identical intravenous




dose (595 ug x mL"1 x  min).  Hence,  the  AUCs after oral administration, like




those after intravenous administration,  approximate  the internal dose subject




to metabolism.  Of  interest, therefore,  is a comparison of the AUCs (or inter-




nal dose) after oral and  inhalation  exposures to  EDC, which were the condi-




tions of the NCI and Maltoni carcinogenicity bioassays.  From Figure 9-13, it




is readily apparent that  the AUCs for an oral dose of EDC of 100 mg/kg (the




"high" dose of the  NCI study)  and 150 ppm  for 6 hours (the highest dose of the




Maltoni study) are  approximately  10:1.




     The disproportionate increase of AUCs,  or internal dose subject to metabo-




lism, relative to linear  increases in oral dosages of EDC, can be more clearly




seen If the ratio of the  AUC to dose (AUC  per unit dose) is plotted against




the administered dose, as shown in Figure  9-15.   As  the oral dose increases,




the internal dose approaches an asymptotic limit  similar to the Michaelis-






                                     9-220

-------
                      K>
                      8
                  x  6H
                      5-
                  CD

                 O
to
ts>
                      2-

                       1-

                      0
                            tf = 0.989
                             M=1.54
                             B=t25
                         1
  3
In DOSE
               Figure 9-14.   Linear  relationship for EDC between  the  logarithm of the area under  the blood
               C,t curve (AUC)  and the logarithm of gavage dose in  corn oil given to Sprague-Dawley rats.
               The coefficient  of regression, r2, is 0.989.
               SOURCE:   Adapted  from Spreafico et al. , 1978,  1979,  1980.

-------
vO

M
NJ
M
                              20
40
60      80     100     120     140
     DOSE (mg/Ug oral)
160
200
               Figure  9-15.   Relationship in Sprague-Dawley  rats between gavage doses of  EDC  in corn oil
               and  the ratio  from the area under the blood EDC concentration-time curve (ADC) divided by
               the  administered dose.  The dashed line represents the limit approached in change of AUC
               per  unit dose  as the administered dose is  increased.
               SOURCE:   Adapted  from Spreaflco et al.,  1978,  1979,  1980.

-------
Menten expression.  Assuming that the internal dose closely approximates or




is proportional to the portion of the administered dose metabolized, then




EUC metabolism in the rat approaches saturation at about 150 to 200 mg/kg,




as in the mouse.  This relationship can be used to estimate the internal dose




and metabolism of EDC for the administered oral doses used in the NCI bioassay.




     Reitz et al. (1980, 1982) also carried out an investigation of the phar-




macokinetics of EDC.  As stated in Reitz et al. (1982), one objective was to




determine the absorbed dose of EDC in rats (Osborne-Mendel) and to characterize




its elimination after single exposures at a single dose level from inhalation




(150 ppm, 6 hours) and oral administration in corn oil (ISO mg/kg).  These




doses were used because they represented the "highest levels of EDC adminis-




tered for any significant length of time in the two bioassays" (NCI and




Maltoni bioassays).   Actually, the oral dose exceeds the time-weighted average




"high" dose for Osborne-Mendel rats of the NCI study (95 mg/kg, Table 9-56).




As shown by the data of Spreafico et al. (see above), the oral dose (150




mg/kg) is close to a saturating dose,  and in comparison, the inhalation dose




(150 ppm, 6 hours) is considerably less (~L/10; Figure 9-13).   Because




Reitz et al. did not examine the kinetics of multiple dose levels, they were




unable to determine  the dose-dependent nonlinear nature of EDC kinetics.




However, these investigators fitted their blood C,t data to kinetic models




empirically.  They found that their blood C,t curve, after inhalation expo-




sure, could be fitted to a linear two-compartment model with first-order elim-




ination, while the hlood C,t data after oral administration was "complex" and




was fitted to a two-compartment,  first-order absorption model  with Michaelis-




Menten elimination kinetics.  Their blood C,t data are shown in Figure 9-16.




The Reitz et al.  observations, because of their internal inconsistency, are




of limited interest  and use; there is  no reason to believe that the elimina-






                                     9-221

-------
                                 20 30  40  50  60  70  80
                       0.2
                                                                    so
                                                                    41-1
                                                                    31
                                                                  O
                                                                 CD  11
                                                                 B
                                                                  O)
                                     MINUTES
                                                   60   120   180  240  300  360
                                                          MINUTES
vO
I
ro
ro
                Figure 9-16.  Blood  levels  of EDC  following  exposure  to F.DC  by (A)  Inhalation (150 ppm, 6
                hours) or (B) gavage (150 mg/kg).   The  Inhalation  C,t data were fitted to a linear two-com-
                partment kinetic  model,  and the  gavage  C,t data were  fitted  to a nonlinear two-compartment
                Nichaells-Nenten  model.  The solid line is the  predicted blood concentration from the model
                and the data point means of 4 rats ±  SD.  A  goodness-of-fit  was not reported for either
                model.  The calculated  kinetic parameters are:
        Inhalation
       Linear model

C0 (peak blood level), 9.4 yg/mL
AUC, 2910 gg/mL x min
    0.091 min
       7.6 min
Vd, 425 mL/kg
                       T1/26
                       V   4
                                                                                    Gavage
                                                                               Nonlinear model
Peak blood level, 30-40
AUC, 4500 pg/mL x min
Vm, 0.166 pg/mL x min
1C,,,, 1.96 ug/mL
Vd, 3400 mL/kg
                                                                                              ug/mL
                SOURCE:  Adapted  from  Reltz  et  al.,  1982.

-------
tion kinetics of EDC differs with respect to the two routes of administration,




inhalation and oral.  The data of Spreafico et al. show that the elimination




kinetics of EDC for all three routes of administration (inhalation, oral, and




intravenous), are dose-dependent, nonlinear kinetics.




     Reitz et al. (1982), in balance studies in rats, compared the assimilated




or "absorbed" dose of EDC after oral (150 mg/kg in corn oil) or inhalation




(150 ppm, 6 hours) exposure to ^C-EDC.  Radioactivity in exhaled air, urine,




feces, and carcass was determined for 48 hours after exposure.  Total recovery




of EDC radioactivity averaged 101% for the oral dose; it was not possible to




determine recovery after inhalation exposure.  The results are given in Table




9-57.  The assimilated dose (152 mg/kg) found after oral administration repre-




sented complete absorption of EDC from the GI tract; the assimilated dose




(50.7 mg/kg) following inhalation exposure proved to be one-third of the oral




dose (Table 9-57).  As found by Yllner (1981b) in mice and by Spreafico et al.




(1978, 1979, 1980) in rats, the data of Reitz et al. also indicate that the




percentage of the administered dose excreted by pulmonary excretion increased




with the dose and that the remaining percentage (metabolized) decreased,




suggesting dose-dependent Michaelis-Menten kinetics for metabolism.  Reitz et




al. found that the major urinary metabolites of EDC in the rat appeared to be




identical in the same relative amounts, i.e., independent of the route of




exposure.




     9.5.3.2.4.  Covalent Binding — Covalent binding from both oxidative




microsomal and cytosolic metabolism of EDC has been demonstrated jln vitro




(Banerjee and Van Duuren,  1978,  1979; Banerjee et al., 1980; Sipes and Gan-




dolfi, 1980; Guengerich et al.,  1980).   Metabolites of EDC covalently bind




in vitro to added protein and DMA;  binding from cytosolic metabolism depends




on the presence of GSH and requires GSH transferases.






                                     9-225

-------
      In vivo covalent  binding from EDC  metabolism  has also been demonstrated




in Che rat.  However,  neither a dose-binding  relationship nor turnover studies




are available  for  EDC;  the studies conducted  have  only been at single, fixed-




dose  levels.   Theoretically,  covalent binding might  be expected to relate to




the dose-dependent metabolism of EDC.




      Reitz et  al.  (1982)  determined total  tissue levels of covalent binding




and specific binding to DMA in rats exposed to 14C-EDC by the oral (150 mg/




kg) and Inhalation (150 ppra,  6 hours) routes,  i.e.,  "high" doses of the




carcinogenicity  bioassays.   No striking differences  between the two routes




were  observed  in the distribution of covalent binding and DNA binding among




the various organ  tissues,  although the tissue levels of covalent binding were




generally higher after  inhalation exposure for total binding and after gavage




exposure for DNA binding.   DNA isolated from  the organ tissues was assessed




for covalent binding, and  in  general, higher  levels  of DNA alkylation (three-




to fivefold) were  observed after gavage exposure,  with highest values for




liver and kidney (Table 9-60).   These DNA  covalent binding studies are in good




accord with the  differences in metabolism  and pharmacokinetics (Table 9-58)




noted for the  NCI  gavage  bioassay in rats  (150 mg/kg "high" dose) and the




Maltoni et al. Inhalation bioassay (150 ppm,  7 hours "high" dose).




      For mice, Storer et  al.  (1982) reported  that  DNA damage was produced in




vivo  in the livers of male B6C3F1 mice  following single oral doses of EDC at




200 mg/kg (the "high" dose of the NCI mouse bioassay).  DNA was evaluated by




differental sedimentation  of  liver nuclei  in  high-density sucrose gradient.




Nuclei from EDC-treated mice  isolated A hours  after  dosing sedimented more




slowly, and total  DNA recovery  from the nuclei was decreased slightly (16%)




as compared with untreated  mice.




     9.5.3.2.5.  Comparison of  Maltoni  et  al.  and  NCI Studies — The NCI (1978)






                                     9-226

-------
          TABLE 9-60.  DNA COVALENT BINDING IN RATS EXPOSED TO 14C-EDC



                                  Gavage                 Inhalation

                                 150 ing/ kg             150 ppro,  6 hours

                                  pmole equvalents bounds/mole DNA
                                         Mean ± SD (n = 3)
Experiment 1
Liver
Spleen
Kidney
Stomach

21.
5.
17.

3
8
4

±
±
+
14.

7.
0.
2.
9

4
7
3

8.
1.
5.

2
8
2

+
±
±
2.

3.
0.
3.
8

3
7
7
Experiment 2

  Liver                          13.9 ±  2.1               3.3 ± 1.2
  Spleen                          2.5+0.3               1.8 ± 0.5
  Kidney                         14.5 ±6.2               2.0 ±0.3
  Stomach                             6.7                     1.9


aAnimals were sacrificed 4 hours post gavage or immediately  following inhalation
 exposure.

SOURCE:  Reitz et al. ,  1982.
                                     9-227

-------
lifetime gavage studies  in rats and  mice  produced  excess  tumors, while the




Maltoni et al.  (1980)  lifetime  inhalation study  did  not.  The kinetic studies




of EDC disposition  in  rats after gavage and  inhalation exposures, and the




metabolism studies  of  Reitz et  al.  (1982) provide  partial explanations for the




contrasting  results of the two  carcinogenicity bioassays.  The two EDC studies




differ in (1) assimilated  or effective internal  dose, (2) fraction of assimilated




dose metabolized  to nucleophilic metabolites, and  (3) peak blood levels and




duration of  metabolism from a single daily dose.




     Table 9-58 contrasts  some  metabolism and kinetic parameters of the "high"




doses of the Maltoni inhalation (150 ppm, 7  hours) and NCI gavage (150 rag/kg)




rat bioassays available  from the studies  of  Spreafico et  al. (1978, 1979, 1980)




and Reitz et al.  (1982).   While the  data  from these  two Investigative groups




are from differing  study designs and differ  in some  measures, they are never-




theless in surprisingly good agreement.   Table 9-58  shows that the internal




dose assimilated  by rats exposed to  150 ppm  EDC  is one-tenth to one-third less




than that from a  150 mg/kg gavage dose, and  that it  produces less than one-half




the metabolites of  a 150 mg/kg  gavage dose.  Furthermore, peak blood levels




of EDC from  a 150 mg/kg gavage  dose  are three- to  fivefold greater, and are




achieved within one-half hour,  as compared to 3  to 6 hours from inhalation




exposure.  Since  the rate  of metabolism is proportional to blood level, these




observations suggest that  the intensity of covalent  binding may be expected




to be greater with  the gavage dose,  and this has been found to he the case;




covalent binding  to DNA in vivo is three- to fivefold higher after gavage




exposure than after inhalation  exposure.




     Additionally,  it  can  be noted that,  contrary  to what might be assumed,  an




EDC inhalation exposure of 6 to 7 hours (150 ppm) does not produce a signifi-




cantly longer presence of  EDC in the blood available for continuing liver and






                                      9-228

-------
other organ metabolism than an oral gavage dose of 150 mg/kg.   In both in-




stances, blood EDC is essentially eliminated (cleared) within 6 to 10 hours,




indicating that the "chronic" dosage in these bioassays is better described




as multiple single dose administration, since a given day's dose of CDC may




be expected to be eliminated from the body before the next day's dose.




However, the residual DNA covalent binding and general cellular covalent




binding, greater for the NCI gavage doses, may be cumulative and may exceed




the capacity of repair mechanisms with chronic EDC administration.  In total,




these observations provide cogent reasons to conclude that the dose range of




the Maltoni inhalation study (5 to 150 ppm) may be insufficient to produce




measurable tumorigenesis.  However, as will be discussed later in section




9.5.3.4.6, reasons other than those indicated here cannot be excluded.




     9.5.3.2.6.  Calculation of Human Equivalent Doses — The NCI carcinogen-




icity studies of EDC in mice and rats were performed by daily (5 days/week)




gavage dosing of EDC in corn oil.  Experimental metabolism and pharmacokinetic




observations pertinent to the conditions of the NCI mouse bioassay are those




of Yllner (1971b), who administered acute doses of EDC in corn oil to mice by




gavage and determined at several dosage levels the fraction of the assimilated




dose metabolized.   Pertinent studies in relation to the NCI rat bioassay are




the experimental kinetic observations of Spreafico et al. (1978, 1979, 1980)




who administered acute doses of EDC in corn oil to rats at several dosage




levels.   The studies of both Yllner and Spreafico et al. included dose ranges




comparable to those of the NCI bioassays.  However, the observations were not




made on "chronic"  dosed animals, as in the NCI bioassays, although the experi-




mental evidence indicates that body clearance of EDC from rodents after a




single daily dose, while dose-dependent, is less than 12 hours for the highest




assay dose, i.e.,  less than the 24-hour dosing interval of the NCI bioassay.






                                     9-229

-------
     While there is  little  information on  the metabolism or pharmacokinetics

of EDC in man, the pertinent  experimental  observations in animals may be

summarized as follows:

     1.  It is assumed  that biochemical  lesions  precede the toxic effects of

EDC.  Formation of reactive metabolites  (P450 oxidation, chloroacetaldehyde;

GSH-mediated cytosolic  reaction,  S-(2-chloroethyl)-GSH) is followed by irre-

versible binding to  proteins,  lipids, and  nucleic  acids critical to cell func-

tion and resulting in toxic,  rautagenic,  or carcinogenic events.

     2.  After gavage administration in  corn oil to mice or rats, EDC is

rapidly and completely  absorbed.

     3.  After gavage administration, a  portion  of the administered dose

(increasing with the dose)  is excreted via the lungs unchanged.  Metabolism of

EDC is less than one-half saturation at  the time-weighted average dose levels
                                            •
of the NCI rat bioassay (47 and  95 mg/kg), but is  at or near saturation for the

highest dose of the  NCI mouse bioassay (195 mg/kg).

     4.  EDC is cleared from  the body well within  the bioassay dosing interval,

even at the highest  bioassay  doses.  The clearance is by first-order pulmonary

excretion of unchanged  EDC  and by parallel dose-dependent Mlchaelis-Menten

metabolism kinetics.  The end-metabolites  (urinary metabolites) are more slowly

cleared than EDC itself.  The rapid clearance of EDC, representing a fraction

of the 24-hour bioassay dosage interval, indicates that accumulation with

multiple doses would not occur;  there is time between doses for repair of cellu-

lar damage from EDC  metabolism,  and also for repletion of cellular glutathione.

     5.  There is no evidence to suggest qualitative differences in the meta-

bolic pathways of EDC in the  two species,  mouse  and rat, and no difference

resulting from the route of exposure (oral or inhalation) in the rat.

     6.  Covalent binding to  cellular protein and  lipids occurs in vivo after


                                     9-230

-------
oral administration of EDC to the rat.   DNA alkylation occurs not only at the

organ sites of tumors found in the NCI bioassay but also at other sites.  There

is no information available on the turnover time or repair time of DNA adducts,

but DNA binding was evident 4 hours after oral dosing.  DNA alkylation after

gavage is estimated at only 2 to 20 / 10& nucleotides.

     Using this metabolic and pharmacokinetic information, the lifetime average

human equivalent doses for the NCI rat and mouse carcinogenicity bioassays of

EDC have been calculated as follows.

     9.5.3.2.6.1.  NCI Bioassay (Mouse):   Liver Carcinomas.  In this study, the

time-weighted average gavage dose levels  of CDC were 195 and 97 mg/kg/day, or,

based on the average terminal weight  of the male mice (33 g), 3.2 and 6.4

mg/male mouse/day (Table 9-56).  Doses in this range given in corn oil have

been found to be completely absorbed  by rodents, although a dose-related,

first-pass effect has been observed,  leading to a portion of the dose being

excreted in exhaled air unchanged.  From the data of Yllner (1971b), a dose-

metabolism curve for EDC administered to mice is available (Figure 9-12).

The metabolism of EUC in mice shows dose-dependent kinetics with the "high"

dose of the NCI bioassay (195 mg/kg)  at saturation of metabolism (Figure

9-12); at this dose level, the amount of  metabolites contributing to tumori-

genesis is 95 mg/kg.  At the "low" dose level of the NCI bioassay (97 mg/kg),

the amount metabolized is 73 mg/kg.  Hence, the fractions of the daily assim-

ilated dose metabolized by the mice (33 g) are:


                Male mice
            Assay dose level
          time-weighted average                    Amount metabolized (AM)
                mg/kg                                mg/kg      mg/animal

                195                                    95        3.14

                 97                                    73        2.41
                                     9-23L

-------
The  lifetime average daily exposure (LAE)  of the mice is  given  by:



                   weeks/90 weeks) x (5 days/7 days)  x AM,  mg/animal/day
where AM is  the amount of dose metabolized.   For humans,  the LAE  is given

by:
                                  LAEM x scaling factor
The  interspecies  scaling is assumed to be on a surface  area  basis (W?/3).

The  scaling  factor  for  mouse to humans is (70/0. 033)2/3 =  165.  Table 9-61

presents  the lifetime average human equivalent doses  for the NCI mouse car-

cinogenicity bioassay of EDC, expressed in units of mg/day, mg/m^ surface

area/day, and mg/ kg/day.
              TABLE  9-61.   LIFETIME  AVERAGE  HUMAN EQUIVALENT DOSES
                         BASED ON THE NCI MOUSE  BIOASSAY
                                         Equivalent  Lifetime average
         Male mice                         human  exposure dosage3	
        gavage dose
      (time-weighted)
          rag/kg                      mg/day    (mg/m2)/day    (mg/kg)/day
           97                        246.2         133.1         3.52

          195                        320.7         173.4         4.58


aWhere the terminal average weight of  the male mouse  is 0.033 kg; the average
 weight for a human is assumed  to be 70 kg with  1.85  m  surface area.
                                      9-232

-------
     9.5.3.2.6.2.  NCI ttioassay (Rat):  Angiosarcomas.  For this study, the

time-weighted average gavage doses ("high" and "low" doses) given to male

Osborne-Mendel rats in corn oil were as follows:
            Gavage dose
           male 0-M rat
              mg/kg
          (time-weighted)                       mg/animal/day

               47                                   23.5

               95                                   47.5
and the corresponding dose per single animal/day is given as calculated from

the average terminal weight of 500 g.  Spreafico et al. (1978, 1979, 1980) and

Reitz et al. (1982) found that gavage doses in corn oil spanning this dosage

range were rapidly and completely absorbed, although a portion, depending on

dose, effects a first-pass of the liver and is excreted in exhaled air un-

changed.  The area under the EDC blood C,t curve (AUC) represents EDC that

has reached the systemic circulation after oral absorption and first-pass pul-

monary excretion.  The AUC represents the approximate internal dose available

for metabolism, and thus is equivalent (or nearly so) to the fraction of the

administered dose metabolized.

     Data from Spreafico et al.  (see Figure 9-15) and Reitz et al. (1982) can

be used to determine the fraction of the dose metabolized for the NCI bioassay

on rats.  On the basis of Figure 9-15, the rate of change (i.e., the slope to

the curve) at dose levels 47, 95, and 150 mg/kg can be estimated to be 0.36,

0.17, and 0.094, respectively.  Since the rate of change decreases as the dose

increases, it is reasonable to assume that the relative amount of EDC exhaled

unchanged is inversely proportional to these rates.  Now, on the basis of

Reitz et al. (1982), 29% of EDC was exhaled unchanged after an administered

                                     9-233

-------
dose of  150 mg/kg.  Thus,  the percentage  unchanged  at NCI doses, 47 and 95

rag/kg, are, respectively,  0.29 x 0.094/0.36  =  0.08,  or 8%; and 0.029 x

0.094/0.17 =  0.16, or  16%.   Therefore,  the fractions of EUC metabolized

for the  NCI study on rats  are 92% at  the  dose  47 mg/kg and 84% at the dose

95 mg/kg.  The  amounts metabolized for  the NCI EDO  bioassay are as follows:



                Male rats
            Assay  dose  level
           time-weighted average                    Amount metabolized (AM)
                mg/kg                                mg/kg      rag/animal


                 47                                  43.24       21.62

                 95                                  79.80       39.90



The lifetime average daily  exposure  (LAE) of  the  rats is given by:



       LAER =  (78  weeks/104 weeks) x (5 days/7 days) x AM, mg/animal/day


where  104 weeks is assumed  to be the average  life span for rats, and AM is the

amount of dose metabolized.   For humans, the  LAE  is given by:
                                = LAEg  x  scaling  factor
The interspecies  scaling  is  assumed  to  be  on  a  surface area basis

The scaling factor  for  rats  to  humans is (70/0. 5)2/3 = 26.96.  Table 9-62

presents the  lifetime average human  equivalent  doses for the NCI rat bioassay,

expressed in  units  of mg/day, mg/ia2  surface area/day, and rag/kg/day.
                                      9-234

-------
              TABLE 9-62.  LIFETIME AVERAGE HUMAN EQUIVALENT IJOSCS
                         BASED OM THE NCI RAT BEOASSAY
                                         Equivalent lifetime average
        Gavage dose                 	human exposure dosage3	
      (time-weighted)
          mg/kg                     mg/day    (mg/m2)/day    (mg/kg)/day


           47                       312.25       168.78        4.46

           95                       576.27       311.50        8.23


aWhere the terminal average weight of the male rat is 0.5 kg; the average
 weight for a human is assumed to be 70 kg with 1.85 m2 surface area.
     9.5.3.3.  CHOICE OF RISK MODEL

     9.5.3.3.1.  General Considerations — The unit risk estimate for EDC

represents an extrapolation below the dose range of experimental data.  There

is currently no solid scientific basis for any mathematical extrapolation model

that relates exposure to cancer risk at the extremely low concentrations,

including the unit concentration given above, that must be dealt with in eval-

uating environmental hazards.  For practical reasons the correspondingly low

levels of risk cannot be measured directly either by animal experiments or by

epidemiologic studies.  Low-dose extrapolation must, therefore, be based on

current understanding of the mechanisms of carcinogenesis.  At the present time

the dominant view of the carcinogenic process involves the concept that most

cancer-causing agents also cause irreversible damage to DMA.  This position is

based in part on the fact that a very large proportion of agents that cause

cancer are also mutagenic.   There is reason to expect that the quantal response

that is characteristic of mutagenesis is associated with a linear (at low

doses) nonthreshold dose-response relationship.   Indeed, there is substantial
                                     9-235

-------
evidence  from mutagenicity studies with both ionizing radiation  and a wide




variety of  chemicals  that this type oE dose-response model  is  the appropriate




one  to use.   This is  particularly true at the lower end  of  the dose-response




curve; at high doses,  there can be an upward curvature,  probably reflecting the




effects of  multistage  processes on the mutagenic response.   The  low-dose linear




nonthreshold  dose-response relationship is also consistent  with  the relatively




few  epidemiologic studies of cancer responses to specific agents that contain




enough information to  make the evaluation possible (e.g., radiation-induced




leukemia, breast and  thyroid cancer,  skin cancer induced by arsenic in drinking




water, liver  cancer induced by aflatoxins in the diet).  Some  supporting evi-




dence also  exists from animal experiments (e.g., the initiation  stage of the




two-stage carcinogenesis  model in rat liver and mouse skin).




     Because  its scientific basis, although limited,  is  the best of any of the




current mathematical extrapolation models, the nonthreshold model, which is




linear at low doses, has  been adopted as the primary basis  for risk extrapola-




tion to low levels of  the dose-response relationship.  The  risk  estimates made




with such a model should  be regarded as conservative,  representing a plausible




upper limit for the risk; i.e., the true risk is not likely to be higher than




the  estimate,  but it could be lower.




     For several reasons, the risk estimate based on animal bioassays is only




an approximate  indication of the absolute risk in populations  exposed to known




carcinogen  concentrations.   First, there are important species differences in




uptake, metabolism, and organ distribution of carcinogens,  as  well as species




differences in  target  site susceptibility, immunological responses, hormone




function, dietary factors,  and disease.   Second,  the  concept of  equivalent




doses for humans compared to animals  on a mg/surface area basis  is virtually




without experimental verification as  regards carcinogenic response.  Finally,






                                      9-236

-------
human populations are variable with respect to genetic constitution and diet,

living environment, activity patterns, and other cultural factors.

     The risk estimate can give a rough indication of the relative potency of

a given agent as compared with other carcinogens.  Such estimates are, of

course, more reliable when the comparisons are based on studies in which the

test species, strain, sex, and routes of exposure are similar.

     9.5.3.3.2.  Mathematical Description of the Low-Dose Extrapolation Model -

The mathematical formulation- chosen to describe the linear (at low doses) non-

threshold dose-response relationship is the linearized multistage model.  This

model employs enough arbitrary constants to be able to fit almost any mono-

tonically increasing dose-response data, and it incorporates a procedure for

estimating the largest possible linear slope (in the 95% confidence limit

sense) at low extrapolated doses that is consistent with the data at all dose

levels of the experiment.

     Let P(d) represent the lifetime risk (probability) of cancer at dose d.

The multistage model has the form
                P(d) = 1 - exp [-(q0
where
                          q^ > 0, i =



Equivalently,



                  Pt(d) = 1 - exp [-<



where
                              P (d) = P(d) - P(0)
                               c        1 - P(0)
                                     9-237

-------
is Che extra  risk  over  background rate  at  dose  d  or  the effect of treatment.




     The point estimate of  the coefficients q^, 1 =  0,  1, 2, ..., k, and con-




sequently, the extra risk function,  Pt(d), at any given dose d, is calculated




by maximizing the  likelihood function of the data.




     The point estimate and the 95%  upper  confidence limit of the extra risk,




Pt(d), are calculated by using the computer program,  GLOBAL83, developed by




Howe (1983).  At low doses,  upper 95% confidence  limits on the extra risk and



lower 95% confidence limits  on the dose producing a  given risk are determined




from a 95% upper confidence limit, q^,  on  parameter  q^.  Whenever q^ > 0, at




low doses the extra  risk Pt(d) has approximately  the  form Pc(d) = qi x d.



Therefore, q^ x d  is a  95%  upper confidence limit on  the extra risk and R/q?




Is a 95% lower confidence limit on the  dose, producing an extra risk of R.  Let




LQ be the maximum  value of  the log-likelihood function.  The upper-limit, q^,




is calculated by increasing q^ to a  value  q^ such that when the log-likelihood




is remaximized subject  to this fixed value, qj, for  the linear coefficient,




the resulting maximum value  of the log-likelihood LI  satisfies the equation






                               2(L0 - LI) = 2.70554






where 2.70554 is the cumulative 90%  point  of the  chi-square distribution with




one degree of freedom,  which corresponds to a 95% upper-limit (one-sided).




This approach of computing  the upper confidence limit for the extra risk Pt(d)




is an improvement  on the Crump et al. (1977) model.  The upper confidence limit



for the extra risk calculated at low doses is always  linear.  This is conceptu-




ally consistent with the linear nonthreshold concept discussed earlier.  The




slope, q^, is taken  as  an upper bound of the potency of the chemical in




inducing cancer at low  doses.   (In the  section calculating the risk estimates,




Pt(d) will be abbreviated as P.)






                                     9-238

-------
     In fitting the dose-response model, the number of terms in the  polynomial

is chosen equal to (h-1), where h is the number of dose groups in the experi-

ment, including the control group.

     Whenever the multistage model does not fit the data sufficiently well,

data at the highest dose is deleted, and the model is refit to the rest of the

data.  This is continued until an acceptable fit to the data is obtained.  To

determine whether or not a fit is acceptable, the chi-square statistic
                               2
                              X
is calculated where N^ is the number of animals in the i   dose group, X( is

the number of animals in the i   dose group with a tumor response, P^ is the

probability of a response in the ith dose group estimated by fitting the multi-

stage model to the data, and h is the number of remaining groups.  The fit is

determined to be unacceptable whenever X^ is larger than the cumulative 99%

point of the chi-square distribution with f degrees of freedom, where f equals

the number of dose groups minus the number of non-zero multistage coefficients.

     9.5.3.3.3.   Adjustments for Less Than Lifespan Duration of Experiment —

If the duration of experiment Le is less than the natural life span of the test

animal L, the slope qi, or more generally the exponent g(d), is increased by

multiplying a factor (L/Lg) .  We assume that if the average dose d is continued,

the age-specific rate of cancer will continue to increase as a constant func-

tion of the background rate.  The age-specific rates for humans increase at

least by the third power of the age and often by a considerably higher power,

as demonstrated by Doll (1971).  Thus, it is expected that the cumulative tumor

rate would increase by at least the third power of age.  Using this fact, it is

assumed that the slope qp or more generally the exponent g(d), would also

                                     9-239

-------
increase by at  least  the  third power  of  age.   As  a  result, if the slope qj



[or g(d)] is calculated at  age Le,  it is expected that  if the experiment had



been continued  for  the full life span L  at  the given average exposure, the


       *                                                       *)
slope qj [or g(d) ]  would  have  been  increased  by at  least (L/Lg) .



     This adjustment  is conceptually  consistent with the proportional hazard



model proposed  by Cox (1972) and the  time-to-tumor  model considered by Daffer



et al. (1980),  where  the  probability  of  cancer by age t and at dose d is


given by



                        P(d,t) = 1  -  exp [-f(t) x g(d)].






     9.5.3.4.   CALCULATION  OF  UNIT  RISK



     9.5.3.4.1.  Definition of Unit Risk — This  section deals with the unit



risk for EDC in air and water  and the potency of  EDC relative to other carcino-



gens that the CAG has evaluated.   The unit  risk estimate for an air or water



pollutant is defined  as the incremental  lifetime  cancer risk occurring in a



hypothetical population in  which all  Individuals  are exposed continuously from


birth throughout their lifetimes to a concentration of  1 yg/m-* of the



agent in the air they breathe, or to  1 yg/L i° tne  water they drink.  This



calculation is  done to estimate in  quantitative terms the impact of the agent



as a carcinogen.  Unit risk estimates are used for  two  purposes:  (1) to com-



pare the carcinogenic potency  of several agents with each other, and (2) to



give a crude indication of  the population risk that might be associated with



air or water exposure to  these agents, if the actual exposures are known.



     9.5.3.4.2.  Unit Risk  Estimates  and Interpretation — The unit risk



estimate based  on animal  bioassays  is only  an approximate indication of the



absolute risk in populations exposed  to  known carcinogen concentrations.



There may be important differences  in target  site susceptibility, immunological




                                      9-240

-------
responses,  hormone function,  dietary factors,  and disease.   In addition,




human populations are variable with respect to genetic constitution and diet,




living environment, activity patterns, and other cultural factors.




     The unit risk estimate can give a rough indication of  the relative potency




of a given agent compared with other carcinogens.  The comparative potency of




different agents is more reliable when the comparison is based on studies in




the same test species, strain, and sex.




     The quantitative aspect of carcinogen risk assessment  is prepared because




of its possible value in estimating the potential magnitude of the public health




hazard.  However, it should be recognized that the estimation of cancer risks to




humans at low levels of exposure is uncertain.  At best, the linear extrapola-




tion model used here provides a rough but plausible estimate of the upper-limit




of risk; i.e., it is not likely that the true risk would be much more than the




estimated risk, but it could very well be considerably lower.  The risk esti-




mates presented in subsequent sections should not be regarded as immutable




representations of the true cancer risks; however, the estimates presented may




be used to the extent that the concept of upper-risk limits is found to be useful.




     9.5.3.A.3.  Alternative Low-Dose Extrapolation Models — In addition to




the multistage model currently used by the CAG for low-dose extrapolation, two




more models, the probit and the Weibull, are also employed for purposes of




comparison.  These models cover almost the entire spectrum of risk estimates




that could be generated from existing mathematical extrapolation models.




Generally statistical in character, these models are not derived  from biolog-




ical arguments, except for Llie inul" ista^K model, which has been used to support




the somatic mutation hypothesis of carcinogenesis (Armitage and Doll,  1954;




Whittemore,  1978; Whittemore and Keller,  1978).  The main difference among




these models  is  the  rate ^t which the  response function  P(d) approaches ^e






                                      9-241

-------
or P(0) as dose d  decreases.   For instance,  the  probit model would usually




predict a smaller  risk  at  low doses  than  the multistage model because of the




difference of  the  decreasing  rate in the  low-dose  region.  However, it should




be noted that  one  could always artificially  give the  multistage model the same




(or even greater)  rate  of  decrease as the probit model by making some dose




transformation and/or by assuming that some  of  the parameters in the multistage




model are zero.  This,  of  course, is not  reasonable without knowing, a priori,




what the carcinogenic process for the agent  is.




     9.5.3.4.4.  Calculation  of the  EDC Carcinogenic  Potency (Slope) — The




multistage model is  used in connection with  the  most  sensitive tumor sites,




and for purposes of  comparison, other extrapolation models and data sets are




also used to estimate the  carcinogenic potency  of  EDC.




     Data from both  rats and  mice (NCI, 1978) are  used to estimate the carcino-




genic potency  of EDC.   For rats, hemangiosarcomas  in  the circulatory system of




male rats were selected, because these tumors are  the most sensitive and were




not located at the site of direct contact with  the agent.  Because of the high




mortality rate In  the high-dose group, the cancer  risk is calculated by using




two types of data:




     1.  Dichotomous data. The number of animals  that survived at least 50




         weeks was used as the denominator of the  incidence rate  (Table 9-63).




         This  data set  serves as a basis  for comparing multistage models with




         the other two  extrapolation models.




     2.  Time-to-death  data (Table B-l, Appendix B).  This data set is more




         appropriate for use  in the calculation of the carcinogenic potency




         of EDC.   In this  calculation, it is assumed  that animals with heraan-




         giosarcomas were  killed by the tumors.   This assumption  seems reason-




         able  since  almost all of the animals with hemangiosarcomas died




         before  terminal sacrifice.




                                      9-242

-------
     For mice, hepatocellular carcinomas In male mice (Table 9-64) were used.



Again, this tumor site was selected because it is the most sensitive in mice.



Using the incidence data in Tables 9-63 and 9-64 and the corresponding human



equivalent doses, the maximum likelihood estimates of the parameters in each



of three extrapolation models are calculated and presented in Table A-l of



Appendix A.  These models ca.n be used to calculate point estimates of risk at



given doses or point estimates of doses for given levels of risk.  The 95%



upper-bound estimates of the risk at 1 mg/kg/day, calculated by various



models using different data sets, are presented in Table 9-65.  From this



table, it is observed that the multistage model predicts a comparable risk on



the basis of either hemangiosarcomas in male rats or liver carcinomas in male



mice, while the probit and Wei bull models are extremely unstable and predict a



wide range of risk, depending on the data base used.



     The slope, qp which is the 95% upper-bound estimate of the linear



coefficient in the multistage model, is also comparable when different data



sets are used.  The values are presented in Table 9-65.


                                                 *            7
     The CAG recommends that the slope estimate q^ = 9. L x 10  /mg/kg/day



be used to represent the carcinogenic potency of EDC by oral exposure.   This



value is calculated on the basis of hemangiosarcomas in male rats using the



multistage model with the time-to-death factor.  This value will be used to



estimate the unit risk of EDC by inhalation and drinking water.



     9.5.3.4.5.  Risk Associated with 1 ug/L of EDC in Drinking Water — It



is assumed that 100% of EDC in drinking water can be absorbed, and that the




water intake is 2 L/day.   Under these assumptions, the daily dose from con-



sumption of water containing 1 ng/L (I ppb) of EUC is
        d = I  pg/L x 2 L/day x 10~3 mg/ug x 1/70 kg = 2.9 x 10-5 mg/kg/day
                                     9-243

-------
      TABLE  9-63.   INCIDENCE  RATES OF HEMANGIOSARCOMAS  IN THE CIRCULATORY
                        SYSTEM OF MALE OSBORNE-MENDEL  RATS
Experimental
dose
(mg/kg/day)
0
47
95
Human
equivalent
metabolized
dose
(mg/kg/day)3
0
4.46
8.23
Tumor
incidence
rates'3
0/40
9/48
7/27
aHuman equivalent  doses  are  taken from Table 9-62.
''The number of animals that  survived  at  least 50 weeks is used as the
 denominator  for the EDC-treated group.

SOURCE:  NCI, 1978.
          TABLE 9-64.   INCIDENCE  RATES OF HEPATOCELLULAR CARCINOMAS
                              IN MALE B6C3F1 MICE
Experimental
dose
(mg/kg/day)
0
97
195
Human
equivalent
metabolized
dose
(mg/kg/day)a
0
3.52
4.58
Tumor
incidence
rates
1/19
6/47
12/48
aHuraan equivalent doses are  taken  from Table 9-61,

SOURCE:  NCI, 1978.


                                     9-244

-------
        TABLE 9-65.  THE 95% UPPER-BOUND ESTIMATE OF RISK AT I  MG/KG/DAY
                         USING THREE DIFFERENT MODELS3
                             Multistage
Data base                 (with time factor)*5     Probit            Weibull


Hemangiosarcomas             6.0 x 10~2         2.81 x KT1        2.70 x 10~l
  in male rats              (8.7 x 10~2)


Hepatocellular carcinomas
  in male mice               6.1 x 10~2         1.87 x 10~4        5.04 x 10~3
alt is assumed that, at low doses,  a given dose is totally metabolized.   There-
 fore, the risk estimates presented here are corresponding to either adminis-
 tered or metabolized dose.
''The corresponding 95% upper-bound  estimates of the linear component in the
 multistage model are:


                         q* = 6.1 x 10~2/mg/kg/day


 on the basis of dichotomous data (hemangiosarcoma);


                         qj = 9.1 x 10~2/mg/kg/day


 on the basis of time-to-death data (hemangiosarcoma);


                         q* = 6.2 x 10-2/mg/kg/day


 on the basis of hepatocellular carcinomas.
                                     9-245

-------
Therefore, the upper-bound  estimate  of  the  incremental  risk associated with




1 ug/L of EDC in drinking water  is






                     P  =  9.1  x  10-2 x 2.9  x  10-5  = 2.6 x 1(T6






     9.5.3.4.6.  Risk  Associated with 1 ng/m3 of EDC in Air — The only




available inhalation study  (Maltoni  et  al.,  1980) did not  indicate any signif-




icant carcinogenic  effects  in  either rats or mice.  This seems contradictory




to the result of the NCI gavage  study,  in wh'ich  EDC was shown to be




carcinogenic to both rats and  mice.   The  seemingly discrepant results between




the two exposure routes  might  be ascribed to one or more of the following




causes:



     1.  The strains of  test animals differ in responsiveness;




     2.  There is a difference in duration  of exposure  In  the peak blood levels




         of EDC, and therefore the target tissue levels of its intermediate




         reactive metabolites  are different for  the doses  in the two studies;




     3.  An artifact has been  introduced  by the  intercurrent mortality.




     One issue that is of interest to the risk assessment  is to determine




whether or not the  dose  used in the  inhalation study is too low to induce a




detectable tumor response in the number of  animals tested, in view of the




fact that EDC is carcinogenic  by oral route of exposure.  Reitz et al. (1982)




indicates that rats exposed to EDC in air at 150 ppm for 6 hours had an assim-




ilated dose of 50.7 mg/kg and  a metabolized dose of 49.8 ing/kg.  On this basis,




the high-dose group (150 ppra to 250 ppm,  7  hours/day)  in the Maltont et al.




study on rats should have assimilated and metabolized  doses greater than 50.7




mg/kg/day and 49.8  mg/kg/day,  respectively.  Therefore, the Maltoni et al.




high-dose group should be at least as responsive as  the NCI low-dose (47




mg/kg/day) group of rats in which 19% (9/48) of  the male rats developed heman-






                                      9-246

-------
giosarcomas.  If the dose available to animals (assimilated or metabolized)




is the only factor that determines tumor response, one would expect to see at




least one tumor type in the Maltoni et al. study based on what was observed in




the NCI study.  It is unlikely that 19% (9/48) of hemangiosarcomas would be




observed in one study and none in the other in which"more animals (79 male




rats survived at 52 weeks) were used in the experiment.  The probability of




observing such an event is very small (p < 0.0001) if the total dose available




to animals is indeed the only factor that determines the tumor response.




Therefore, the discrepant results between the two exposure routes cannot be




totally explained by the difference in dose available to animals.




     The observation of greater (three- to fivefold) peak blood concentrations




in the gavage study when compared to the inhalation study, together with dif-




ferences in the rates of formation, deactivation, and binding to macromolecules




between the two routes of exposure, may explain the differing response pattern.




     The implication of the considerations just discussed is that, in addition




to the EDC kinetic information (oral vs. inhalation) on assimilated and metabo-




lized dose, it is important to determine whether strains of test animals,




duration of exposure, and/or route of exposure make significant differences in




the tumorigenic responses.   If the strains of animals differ in responsiveness,




then the most sensitive animal strain (i.e.,  the one from the NCI gavage study)




should be used for potency estimation via inhalation.   On the other hand, if




the route of exposure makes a difference in the tumorigenic response, it would




be more appropriate to use the inhalation study rather than the gavage study




to estimate the risk by inhalation.  Since the reason for the discrepant results




between the two exposure routes is not known, both the NCI gavage study and




the negative inhalation study by Maltoni et al. (1980) are used to calculate




the risk for EDC by inhalation.   The unit risk estimates calculated on the






                                     9-247

-------
basis of these two data  sets  provide  a  range of  risk estimates that reflect




several types of uncertainty,  including strains  and routes of exposure used in




the bioassays.



     9.5.3.A.6.1.  Unit  Risk  for Air  Calculated  on  the Basis of Rat Gavage Data.




The daily dose due to  1  pg/m3 of EDC  in air is






                  d  =  1  ug/m3 x 20 m3/day  x 10~3 mg/kg/70 kg




                     =  2.86 x  10~4 rag/kg/day






where 20 m3  is assumed to be  the volume of daily air intake by a 70 kg person.




In this calculation, the EDC  in air breathed  is  assumed  to be 100% absorbed.




It is reasonable to  assume 100% absorption and metabolism at low doses.




Thus, the upper-bound  estimate of the incremental risk due to 1 yg/ra3 of EDC




in air is






                     P  = 9.1 x ID'2 x 2.9 x 10"4  = 2.6 x  lO'5






     The information on EDC kinetics is not used in this derivation of unit




risk.  Although  Reitz  et al.  (1982) provides  some information on the compara-




tive metabolism  orally (at 150 mg/kg) and by  inhalation  (at  150 ppra, for 6




hours), this information cannot be appropriately used for route-to-route extrap-




olation because  the  relative  amounts metabolized between the  two routes of




exposure do  not  appear to be  linearly related to the administered  dose, as




evidenced  by the data  of Spreafico et al.  on  the blood  concentration-time  curve




(AUC).  Using the  Spreafico et al. data in Table 9-59 and the assumption that




the  amount metabolized is linearly related to AUC, as  the administered EDC dose




decreases,  the amount  metabolized, when administered by  inhalation, decreases  in




a  greater  proportion than the amount of EDC metabolized  when administered  by  a




single  bolus by  the  oral route.  Therefore,  if the comparative  metabolism






                                      9-248

-------
observed at high doses in Reitz et al.  (1982) is used for route-to-route extrap-




olation, the inhalation risk at low doses (e.g., 1  yg/m^) would he over-




estimated.




     9.5.3.4.6.2.  Unit Risk for Air Calculated on  the Basis of a Negative Rat




Inhalation Study.  A negative inhalation study by Maltoni et al. (1980) can be




used to calculate an upper-bound estimate of risk.   The data from male rats is




used to calculate an upper-bound estimate of risk because it was found to be




most sensitive in the NCI study.  The smallest upper-bound estimate is calcu-




lated by using the information from the high-dose group in which 79 male rats




survived at 52 weeks.  These animals were exposed to EDO in air at 150 ppm,




7 hours/day, 5 days/week, for 78 weeks.  The lifetime average exposure is cal-




culated to be






    d = (150 ppm x 7 hours/24 hours) x  (5 days/7 days) x 78 weeks/104 weeks




      = 23.44 ppm






or equivalently,




                        d = 23.44 ppra x 4,121 (pg/m3)/ppm




                          = 96,596.24 ug/m3






Assuming no "tumor" is observed among the 79 animals, the 95% upper limit




of the tumor probability is R = 0.037,  calculated by solving the equation




(1 R)79 = 0.05.  By using the one-hit model, R = 1  - exp(-b x d), the




slope, b, Is calculated as






                               b = [ -  ln(l-R)]/d




                                 = 3.9  x 10-7/(ug/ra3)






Thus, for male rats, the risk due to 1  pg/m3 of EDC in air is 3.9 x 10~7.




To convert this animal risk estimate into human risk, it is assumed that body




                                     9-249

-------
burden per surface area  is  equivalent  among  species.   For  L yg/m3 of EDC in

air, Che total EDC uptake (body  burden)  of a rat weighing  0.35 kg is


                               1  ug/m3  x  0.224 m3/day


where 0.224 m3/day is  the daily  air intake for  a 0.35  kg rat.  The concentra-

tion for humans, C(ug/m3),   which is equivalent to  1 jig/m3 for rats, is

calculated to be C = 0.38 ug/m3, by solving  for C from the equation
                    20  m3/day x C(ug/m3)  = 1  ug/m3  x 0.224 m3/day
                          (70 kg)2/3          (0.35 kg)2/3
where 20 m3/day  is  assumed to be the volume  of  air intake  by a 70 kg person.

Therefore,  the upper-bound estimate of incremental human cancer risk due to

1 ug/m3 of  EDC in air is


                               P = 3.9 x 10-7/0.38

                                 = 1.0 x 10~6


This risk estimate  is 26 times smaller than  that calculated on the basis of

the gavage  study.   As discussed previously,  this upper-bound risk estimate  is

appropriate only if the discrepant results in tumor response between the oral

and inhalation  routes is due to the possible difference in the rates of forma-

tion or rates of deactivation or rates of covalent bond formation and  if, in

combination, these  results differ between oral  versus inhalation routes of

exposure, but are  not due to the strains of  test animals.

     9.5.3.5.  COMPARISON OF POTENCY WITH OTHER COMPOUNDS  — One of  the uses

of quantitative  potency estimates is to compare the potencies  of various car-

cinogens.   Figure  9-17 is a histogram representing the frequency distribution

of potency  indices  of 54 suspect carcinogens evaluated by  the  CAG.   The data

                                      9-250

-------
4th
QUARTILE
1 x
3rd
QUARTILE
10'1 4>
2nd
QUARTILE
I
MO*2 2 x
1s:
QUARTILE
1C'3
 -1
1234      56

LOG  OF POTENCY  INDEX
Figure 9-17.   Histogram representing  the  frequency distribution
of the potency indices  of  the  5A  suspect  carcinogens evaluated
by the Carcinogen Assessment Group.
                             9-251

-------
summarized  by  Che  histogram  are presented  in  Table  9-66.  The potency index



is derived  from q^,  the  95%  upper  bound  of  the  linear component in  the



multistage  model,  and  is expressed in terms of  (mmol/kg/day)"1.  Where human



data were available  for  an agent,  they have been  used to calculate  the Index.



Where no human data  were available,  animal  oral studies have been used in



preference  to  animal inhalation studies, since  oral  studies constitute the



majority of animal studies.



     Based  on  available  data concerning  hemangiosarcomas in male rats (NCI,



1978), the  potency index for EDC has  been calculated as 9 x 10°.  This figure


                                      fc           9
is derived  by  multiplying the  slope q^ = 9.1  x  10~^/mg/kg/day and the



molecular weight of  EDC,  98.9.   This  places the potency index for EDC in the



fourth quartile of the 54 suspect  carcinogens evaluated by the CAG.



     The ranking of  relative potency  indices  is subject to the uncertainties



involved in comparing  a  number of  potency estimates  for different chemicals



based on varying routes  of exposure  in different  species, by means of data from



studies whose  quality  varies widely.   All of  the  indices presented are based on



estimates of low-dose  risk,  using  a  low-dose  linear extrapolation model.  These



indices may not be appropriate for the comparison of potencies if linearity



does not exist at  the  low-dose range,  or if comparison is to be made at the



high-dose range.   If the latter is the case,  then an index other than the one



calculated above may be  more appropriate.



9.5.4.  Summary



     9.5.4.1.  QUALITATIVE — Although seven  cancer  bioassay studies of EDC



have been reported,  only the NCI bioassay in  which EDC was administered to



rats and mice  by gavage  produced a clear positive tumorigenic response.



     In the NCI (1978) rat study,  EDC  produced  a  statistically significant



increase in the incidence of squamous  cell carcinomas of the forestomach,





                                      9-252

-------
                                                                                                                    July  1985
             TABLE 9-66.  RELATIVE CARCINOGENIC POTENCIES AMONG 54 CHEMICALS EVALUATED BY THE CARCINOGEN ASSESSMENT GROUP
                                                     AS SUSPECT HUMAN CARCINOGENS
vO
I
Level
of evidence3
Compounds
Acrylonitrile
Aflatoxin B.
Aldrln
Allyl chloride
Arsenic
B[a]P
Benzene
Benzidene
Beryllium
1,3-Butadiene
Cadmium
Carbon tetrachloride
Chlordane
CAS Number
107-13-1
1162-65-8
309-00-2
107-05-1
7440-38-2
50-32-8
71-43-2
92-87-5
7440-41-7
106-99-0
7440-43-9
56-23-5
57-74-9
Humans
L
L
I

S
I
S
S
L
I
L
I
I
Animals
S
S
L

I
S
S
S
S
S
S
S
L
Grouping
based on
IARC
criteria
2A
2A
3

1
2B
1
1
2A
2B
2A
2B
3
Slopeb
(mg/kg/day)-l
0.24(W)
2900
11.4
1.19x10-2
15(H)
11.5
2.9xlO-2(W)
234(W)
2.6
l.OxHT'U)
6.1(U)
1. 30x10-!
1.61
Molecular
weight
53.1
312.3
369.4
76.5
149.8
252.3
78
184.2
9
54.1
112.4
153.8
409.8
Potency
lndexc
1x10+1
9xlO+5
4x10+3
9x10-'
2x10+3
3x10+3
2x10°
4xlO+4
2xlO+1
5x10°
7x10+2
2x10+'
7x10+2
Order of
magnitude

-------
                                                       TABLE 9-66.  (continued)
I
NJ
Level
of evidence8
Compounds
Chlorinated ethanes
1 , 2-Dichloroethane
hexachloroethane
CAS Number Humans
107-06-2
67-72-1
1,1,2,2-Tetrachloroethane 79-34-5
1,1,2-Trichloroethane 79-00-5
Chloroform
Chromium VI
DDT
Dichlorobenzidine
1 , 1-Dichloroethylene
(Vinylidene chloride)
Dichloromethane
(Methylene chloride)
DieLdrln
2,4-Dinitrotoluene
Diphenylhydrazine
Epichlorohydrin
Bis(2-chloroethyl)ether
67-66-3
7440-47-3
50-29-3
91-94-1
75-35-4

75-09-2

60-57-1
121-14-2
122-66-7
106-89-8
111-44-4
I
I
I
I
I
S
I
I
I

I

I
I
I
I
I
Animals
S
L
L
L
S
S
S
S
L

L

S
S
S
S
S
Grouping
based on
1ARC
criteria
2B
3
3
3
2B
1
2B
2B
3

3

2B
2B
2B
2B
2B
Slopeb
(mg/kg/day)-1
9.1x10-2
1.42x10-2
0.20
5.73x10-2
7x10-2
41(W)
0.34
1.69
1.16(1)

6.3xlO-A(I)

30.4
0.31
0.77
9.9xlO-3
1.14
Molecular
weight
98.9
236.7
167.9
133.4
119.4
100
354.5
253.1
97

84.9

380.9
182
180
92.5
143
Potency
indexc
9x10°
3x10°
3xlO+1
8x10°
8x10°
4xlO+3
lxlO+2
4xlO+2
1x10+2

5x10-2

1x10**
6xlO+1
lxlO+2
9x10-!
2xlO+2
Order of
magnitude
(lOglQ
index)
•H
0
+1
+1
+ 1
+*
+2
+3
+2

-1

+4
+2
+2
0
+2

-------
                                                        TABLE 9-66.  (continued)
 I
tv.
Compounds
Bis(chloromethyl)ether
F.thylene dibromvde (EDB)
Elhylcne oxide
Keptrichlor
Hexachlorobenzene
HexachLorohutadlene
Hexachlorocyclohexane
technical grade
alpha isomer
beta tsomer
gamma isomur
Hexdchlorodlbenzodioxin
\ 1 l_k-' 1 ft! 1 1 ML' 1 " illlSt
(Nil 1 -.,ih-.'i i i ulii)
Li m ii. •
1)1 ,V 1 II ' 1 11111 Hi-
ll. !.l. • ,1110
1), < . \' • 1 • .1 IK-
N- 1. 1 r osii|", . !il I no
N-'i i : i>-..i-V i.ivlurc.i
CAS Number
542-88-1
106-93-4
75-21-8
76-44-8
118-74-1
87-68-3


319-84-6
319-85-7
58-89-9
34465-46-8



h2- *.-l(
.<>- ,- ,
4.''i •- >
'( - .
/ .'J- .- '
Level
of evidence3
Humans Animals
S S
I S
L S
I S
I S
1 L


I S
1 L
T L
1 S
S S


1 S
1 S
1 S
!
1 S
Grouping
based on
IARC
criteria
1
2B
2A
2B
2B
3


2B
3
3
2B
1


2H
2B
>B
in
in
Slopeb
(mg/kg/day)~l
9300(1)
41
3.5xlO-1U)
3.37
1.67
7.75xlO~2

4.75
11.12
1.84
1.33
6.2xlO+3
1.05(W)


2S.9(not b>
4 }.5(noL bv
ri.43
J.I3
i '.9
Molecular
weight
115
187.9
44.1
373.3
284.4
261

290.9
290.9
290.9
290.9
391
240.2


qf> 74.1
qf) 102.1
ISH.J
100.2
H7.I
Potency
index0
lxlO+6
8xlO+3
2xlO+1
lx!0+3
5xLO+-
2xlO+1

lxlO+3
3xlO+3
5xlO+2
4xlO+2
2xlO+6
2.5X1Q-1--2


2xlH+3
'ixl'if<
'i \ 1 1 1 ' -'
Mi 'J
txl"'(
Order of
magnitude
(logl()
Index)
+6
+4
+ 1
+3
+3
+ 1

+3
+ )
+ j
+ 3
+6
+2


+ 3
KI
r \
• :
' •
                                                                                               (rontinumt on t ho 11> I lowing

-------
                                                       TABLE 9-66.  (continued)
vO

to
in
Level
of evidence3
Compounds CAS Number
N-ni t roso-N-me t hylu rea
N-nitroso-diphenylamine
PCBs
Phenols
2 , 4, 6-Trichlorophenol
Tetrachlorodibenzo-
p-dioxin (TCDD)
Tetrachloroethylene
Toxaphene
Trichloroethylene
Vinyl chloride
684-93-5
86-30-6
1336-36-3
88-06-2
1746-01-6
127-18-4
8001-35-2
79-01-6
75-01-4
Humans
I
I
I
I
I
I
1
I
S
Animals
S
S
S
S
S
L
S
L/S
S
Grouping
based on
IARC
criteria
2B
2B
2B
2B
2B
3
2B
3/2B
1
Slopeb
(mg/kg/day)-l
302.6
4.92xlO-3
4.34
1.99x10-2
1.56x10+5
5.1x10-2
1.13
l.lxlO-2
1.75x10-2(1)
Molecular
weight
103.1
198
324
197.4
322
165.8
414
131.4
62.5
Potency
indexc
3x1 0+*
1x10°
1X10+3
4x1 0°
5x1 0+7
8x10°
5xlO+2
1x10°
1x10°
Order of
magnitude
Uog1Q
index)
+4
0
+3
+1
+8
+ 1
+3
0
0
       aS = Sufficient evidence; L ° Limited evidence; I = Inadequate evidence.
       bAnimal slopes are 95% upper-bound slopes based on the linearized multistage model.   They are calculated based on
        animal oral studies,  except for those Indicated by I (animal inhalation), U (human occupational exposure), and H
        (human drinking water exposure).   Human slopes are point estimates based on the linear nonthreshold model.  Not all
        of the carcinogenic potencies presented in this table represent the same degree of certainty.  All are subject to
        change as new evidence becomes available.   The slope value is an upper bound in the sense that the true value (which
        is unknown) is not likely to exceed the upper bound and may be much lower, with a lower bound approaching zero.
        Thus,  the use of the  slope estimate in risk evaluations requires an appreciation for the implication of the upper
        bound  concept as well as the "weight of evidence" for the likelihood that the substance is a human carcinogen.
       cThe potency index is  a rounded-off slope in (mmol/kg/day)-1 and is calculated by multiplying the slopes in
        (mg/kg/day)-l by the  molecular weight of the compound.

-------
hemangiosarcomas of the circulatory system, and fibromas of subcutaneous tissue




in male rats.  There was also a statistically significant increased incidence




of adenocarcinoraas of the mammary gland and hemangiosarcomas of the circulatory




system in female rats.



     In the NCI (L978) mouse study, EDC produced a statistically significant




increased incidence of hepatocellular carcinomas and alveolar/bronchiolar




adenomas in male mice and a statistically significant increased incidence of




alveolar/bronchiolar adenomas, mammary carcinomas, and endometrial tumors in




female mice.



     Two inhalation studies of EDC were conducted.  The study by Spencer et




al. (1951) in Wistar rats showed no evidence of a positive response.   However,




this study was inadequate to assess the carcinogenicity of EDC because the




experiment was conducted in a small number of animals for 212 days with L51




exposure days at 200 ppm.  A second study by Maltoni et al. (1980), conducted




in both rats and mice, did not produce a statistically significant increase in




tumor incidences in any of the organ sites as compared to control animals.




     The study by Theiss et al. (1977), a pulmonary tumor bioassay in which EDC




was administered intraperitoneally to strain A mice, produced an elevated, but




not a statistically significant, increase in the incidence of lung tumors in




treated animals.




     In a study by Van Duuren et al. (1979), EDC was applied to the skin of




ICR/Ha mice to assess its carcinogenic potential as a skin turaorigen.  This




study did not show a statistically significant increase in skin carcinomas;




however, an increased incidence of benign lung tumors in mice treated with the




high dose (126 mg/mouse) was reported.




     No case reports or epidemiologic studies concerning EDC are available in




the published literature.






                                     9-257

-------
     9.5.4.2.   QUANTITATIVE  — Data from gavage  studies  using  both rats and


mice have been  used  to  estimate the carcinogenic potency of EDC.  An evaluation


of the metabolism and kinetic data shows that there is no evidence to suggest


qualitative differences in the metabolic pathways of  EOC in the mouse and


rat, and no difference  resulting from the route  of exposure (oral or inhalation)


in the rat.


     Lifetime average human  equivalent doses for the  NCI rat and mouse carcino-


genicity bioassay can be calculated and are used in estimation of the cancer


potency.  For rats,  data on  hemangiosarcoraas in  the circulatory system are used.


For mice, data  on hepatocellular carcinomas are  used.  The carcinogenic poten-


cies estimated  on the basis  of these two data sets are comparable when the


linearized multistage model  is used.   The upper-bound estimate of EDC potency

            «t            —O                   jt
(slope) is qi = 9.1  x 10  /mg/kg/day.  This qi is calculated on the basis


of hetnangiosarcomas  using the time-to-death data and  an  adjusted dose derived


from the metabolism/kinetic  evaluation.


     The upper-bound estimate of the Incremental cancer  risk due to 1 yg/L


of EDC in drinking water is  2.6 x 10~6.


     Because of the  discrepant tumor responsiveness between oral and inhala-


tion assays, two upper-bound estimates of the incremental cancer risk due to


1 ug/m3 of EDC  in air are calculated:  2.6 x 10~5 on  the basis of the NCI


gavage study, and 1.0 x 10~6 on the basis of a negative  inhalation study by


Maltoni et al.  (1980).   These two estimates reflect the  various uncertainties


associated with the  data used, including the uncertainties due to the strains


of test animals and  routes of exposure used in the bioassays.  If the strains


of test animals differ  in responsiveness, the unit risk  estimate, 2.6 x 10~5,


calculated from the  positive response of the most sensitive strain of animals,


should be used.  On  the other hand, if the discrepant results  between the



                                      9-258

-------
gavage and inhalation studies are due solely to the difference in route of




exposure, than it would be more appropriate to use che unit risk estimate,




1.0 x 10~6, calculated on the basis of an inhalation study, even if it is a




negative study.  Given the evidence currently available,  there is no certain




scientific reason to explain why animals responded differently in the NCI




gavage study compared with the Maltoni et al. inhalation study.




9.5.5.  Conclusions.  There is evidence that EDC is a probable human carcino-




gen.  This conclusion is based on:  (1) multiple tumor types in both an oral




rat bioassay and an oral mouse bioassay; (2) suggestive evidence in two other




bioassays, one which showed an elevated increase in lung tumors with intraperi-




toneal injection and a second study which showed a significantly increased




Incidence of benign lung tumors with skin application of EDC; (3) demonstrated




evidence of reactive metabolites of EDC and formation of DNA adduct; and (4)




evidence that EDC is also a mutagen.




     There is direct animal evidence for carcinogenicity via the oral (gavage)




route of exposure, but the only specific bioassay data for inhalation exposure




are a negative rat and mouse study.  Because of the presence of distant site




tumors in the oral studies, it Is likely that EDC is also carcinogenic via




inhalation exposure.  The negative findings in the rat and mouse inhalation




studies do not contradict the indirect, suggestive, and ancillary evidence for




the likelihood of EDC being carcinogenic via inhalation.




     The U.S. Environmental Protection Agency is using, on an interim basis,




a proposed classification scheme for evaluating the weight-of-evidence for




carcinogenicity (U.S. EPA, 1984).  Using this classification, the positive




findings in the oral rat and mouse studies would be considered as a sufficient




level of evidence in experimental animals.  The sufficient level of animal




evidence, together with an absence of epidemiologic data, provides a basis for






                                     9-259

-------
an overall welght-of-evidence ranking of  Group  B2, meaning  that EDC is


"probably" carcinogenic  in  humans.


     Applying  the  International Agency for  Research  on Cancer (IARC) classifi-


cation scheme, which  EPA has  used  in  the  past,  the positive  findings in the


experimental animals  and the  absence  of epideraiologic data  provide an overall


weight-of-evidence ranking  of Group 2TJ, meaning that EDC is  "probably" carci-


nogenic  in humans.


     While the "probable" carclnogenicity conclusion obviously applies to


ingestion exposure since the  positive evidence  comes from gavage experiments,


it is likely that  EDC is a  probable carcinogen  for humans via inhalation expo-


sure.  In view of  the evidence, it  is prudent to consider EDC to be carcinogenic


via inhalation exposure, although  the potency may vary from that estimated from


gavage.  Research  investigations to verify  this assessment  conclusion should be


implemented.


     Assuming  that EDC is carcinogenic for  humans, upper-bound incremental unit


risks have been  estimated for both ingestion and Inhalation  exposure.  The


development of these  unit risk estimates  is for the  purpose  of evaluating the


"what if" question:   If  EDC is carcinogenic for humans, what is the possible


magnitude of the public health impact? The upper-bound estimate of EDC's car-

                       JL            —O
cinogenic potency  is  q,  = 9.1 x 10  /rag/kg/day  based on the  occurrence of


heraangiosarcomas in the NCI gavage rat study.   The upper-bound estimate of


the incremental  cancer risk due to  1  pg/L of EDC in  drinking water is 2.6 x


10~6, based on the previously mentioned potency value.  Two  upper-bound


estimates of the incremental cancer risk  due  to 1 [jg/ra-^ of  EDC in air are


calculated:  2.6 x 10~5, on the basis of  the gavage  potency  value and 1.0 x


10~6, on the basis of a negative inhalation study by Maltoni et al. (1980).


The inhalation unit risks are presented as  two  values in recognition of the



                                      9-260

-------
uncertainties associated with the differing data that were used to estimate




the values.  The upper-bound nature of these estimates is such that the true




risk is not likely to be exceeded and may be lower.




     The potency index for EDC, defined as qj x molecular weight, lies in




the fourth quartile among 54 carcinogens evaluated by the Carcinogen Assessment




Group.
                                     9-261

-------
                                   APPENDIX A

                COMPARISON AMONG  DIFFERENT EXTRAPOLATION MODELS




     Three models used  for low-dose  extrapolation, assuming the independent


background, are:
Multistage:                  P(d)  - 1  - exp  (-(q±d + . . . +


where q^ are non-negative  parameters;


Probit:                                A + Bln(d)
                             P(d)  =    /  f(x) dx
                                     — OB

where f(.) is  the  standard normal probability density function;


Weibull:                     P(d)  = 1  - exp  [-bdk]


where b and k  are  non-negative  parameters.

     The maximum  likelihood estimates (MLE) of  the parameters in the multi-

stage model are calculated by means of the  program GLOBAL83, which was developed

by Howe (1983).   The MLE estimates of the parameters  in the probit and Weibull

models are calculated by means  of the program RISKS 1, which was developed by

Kovar and Krewski  (1981).

     Table A-l presents the MLE of parameters in  each of  the three models

that are applicable to a data set.
                                      9-262

-------
                    TABLE A-l.  MAXIMUM LIKELIHOOD ESTIMATES
                           OF THE PARAMETERS FOR EACH
                       OF THE THREE EXTRAPOLATION MODELS
                         BASED ON DIFFERENT DATA BASES
Data base
Multistage
ProbiC
                                                                 Welbull
Hemanglosarcomas
 in male rats
Hepatocellular
 carcinomas
 in male mice
q2
= 4.15 x 10

= 0

= 0.0

= 1.00 x 10
                                      -2
              -2
A = -1.48

B =  0.39

A = -4.27

B =  2.27
b = 8.46 x 10~2

k = 0.60

b = 5.70 x 10~4

k = 3.95
                                     9-263

-------
                                   APPENDIX B




                  RISK CALCULATION BASED ON TIME-TO-EVENT DATA









     Because of the high mortality rate of rats in the high-dose group of




the NCI (1978) gavage study, it  is more appropriate to use time-to-event




data (Table B-l) to calculate  the potency of EDC.  The probability of




cancer by time t at dose d is  given by:






                        P(d,t) = 1 - exp [-f(t) x g(d)]






where g(d) is a polynomial in  dose d, and f(t) is a function of time t.  The




maximum likelihood estimate of the parameters and the asymptotic properties




of the incremental risk estimate were Investigated by Daffer et al. (1980).




Their approach to estimating the parameters resembles Cox's regression-life-




table approach (Cox 1972), in  which the time function f(t) need not be




specified.




     Using the data in Table B-l, the lifetime cancer risk is estimated to be
                           q*  =  9.1  x  lO'Vmg/kg/day






This value  is  the 95%  upper  bound of the  risk  calculated at t = 90 weeks.




Since the model  fits the  data  very well up  to  90 weeks but poorly beyond




90 weeks (overestimates),  P(d,  90) is  used  to  approximate the lifetime risk.




This seems  reasonable  because  the median  life  span  for control animals is also




less than 90 weeks  (approximately 70%  of  the control  animals died before 90




weeks).
                                      9-264

-------
            TABLE B-l.   TIME-TO-DEATH IN WEEKS  FROM HEMANGEOSARCOMAS
                 IN MALE OSBORNE-MENDEL RATS  FED EDC BY  GAVAGE
     Control (vehicle and untreated):  dose  =  0

          2B,  37,  50, 50, 53,  53,  53,  54,  57,  57,  57,  57,  57,  61,  61,
          69,  71,  76, 78, 80,  80,  83,  85,  86,  87,  89,  90,  101,  101,
          104, 106,  106,  106,  106,  106,  108,  110,  110,  110,  110.

     Low-dose  group:  dose = 4.46 mg/kg/day

          30,  34,  36, 51, 52,  52,  55,  55,  59,  61,  63,  65,  69,  73(H)a,
          74(H),  75,  75,  76, 77, 77,  77(H), 77,  80,  81,  82,  82,  82,  84,
          87(H),  89(H),  89,  89,  89, 90,  92, 92,  93(H),  95(H),  96,  97,
          98,  99,  99, 102{H),  103,  103,  104,  104,  109(H),  110.

     High-dose group: dose = 8.23 lag/kg/day

          3, 8, 9, 9, 10, 10,  10,  10,  10,  11,  15,  15,  16,  20,  22,  33,
          33,  33,  34, 37, 41,  48,  51,  52,  54,  57,  58,  59,  60,  61(H),
          62,  63,  63, 68(H), 68(H), 71,  72, 73,  74,  74(H),  74,  74, 76(H),
          76,  78(H),  83(H),  84,  89, 101.
aH indicates death from hemangiosarcoma.
                                     9-265

-------
                                10. REFERENCES



Agranovich,  B.Y.A.  (1948)  Clinical treatment  and  pathology of  toxicologic-



ehemical injuries of the liver in the case of industrial poisoning. Moscow, USSR:




Academy of Medical  Science;  pp.  132-113.








Akimo, G.A.; Buchko,  V.M.;  lolesnichenko, I.P.  (1976)  Changes in  the nervous




system in acute diohloroethane poisoning.  Voen.-Med.  Zh.  5:  35-37.








Akimo, G.A.; Buchko,  V.M.;  Kolesnichenko,  I.P.  (1978) Neurological  disorders in



acute dichloroethane poisoning. Zh. Kevropatol. Psikhiatr.  in.  S.S. Korsakova.




78: 687-692.








Allport, J.; Casey,  S.; Cook,  J.; Hall,  P.;  Helmes, C.T.  (1977) A  study of



industrial   data   on   candidate   chemicals   for  testing.   EPA   report   no.



EPA-560/5-77-006.  Washington, DC:  U.S. Environmental Protection Agency, Office



of Toxic Substances;  Available from: NTIS, Springfield,  VA;  PB-27126U.








Altshuller,  A.P.; Hanst, P.L.  (1975)  Report on the  problem of halogenated air



pollutants   and  stratospheric   ozone.   Research   Triangle  Park,   NC:  U.S.



Environmental Protection Agency, Office of Research and Development; EPA report



no. EPA-600/9-75-008.  Available from:  NTIS, Springfield, VA; PB-247-699.








Altshuller,  A.P. (1979)  Lifetimes of  organic molecules  in  the troposphere and



lower  stratosphere. Advances  in Environmental Science and  Technology  10:   181-




219.
                                      10-1

-------
Ambrose,  A.  (1950)  lexicological studies  of  compounds  investigated  for  use as



inhibitors  of  biological  processes.  II.  Toxicity of  ethylene  chlorohydrin.



Arch. Ind. Hyg. Occup. Med.  2:  591-597.







Araoore, J.E.; Venstrom, D.  (1966) Sensory analysis  of odor qualities in term of



the stereochemical  theory.  J. Food  Sci.  31: 118.








Anders, M.W.;  Livesey, J.C.  (1980)  Metabolism of  1,2-dihaloethanes.  In:  Ames,



B.N.; Infante, P.; Reitz, R., eds. Ethylene dichloride: a potential health risk?



Cold  Spring  Harbor, NY:  Cold Spring Harbor  Laboratory; pp.  331-343.  (Banbury



report no. 5).








Andriukin,  A.A.  (1979)  Toxic effects of  dichloroethane on  the cardiovascular



system. Klin. Med.  57: U3-47.







Anonymous. (1946) Ethylene  dichloride. In: Annual report of the Chief Inspector



of Factories for the year 19*15.  London,  United Kingdom: His Majesty's Stationery



Office; p. 77.







Appleby,   A.   (1976)  Atmospheric   freons  and  halogenated  compounds.   U.S.



Environmental Protection  Agency;  Office  of Research and Development, EPA report



no. EPA-600/3-76-108.








Archer, W.L. (1979) Other chloroethanes.  In: Grayson, M.; Eckroth,  D., eds. Kirk-



Othmer encyclopedia of chemical  technology, 3rd ed., New York,  NY:  John Wiley and



Sons, Inc. v. 5, pp. 722-712.
                                      10-2

-------
Armitage, P.; Doll, R. (1954) The  age distribution of cancer  and  a multistage



theory of carcinogenesis. Br. J. Cancer 8:  1-12.








Auerbach  Associates.  (1978)  Miscellaneous  and  small  volume  consumption  of



ethylene dichloride. Philadelphia,  PA: Unpublished report prepared for the U.S.



Environmental  Protection  Agency  under  contract  number  EPA-68-01-3899  and



Auerbach Associates, Inc., number AAI-2431-104-TN-1,  p.  7.








Baader, E.W. (1950) Multiple poisonings from a floor cleaner because of failure



to observe safety laws.  Arch. Hyg.  Bakteriol. 132: 219-226.








Banerjee,  S.;  Van Duuren,  B.L. (1978)  Interaction  of activated  carcinogenic



intermediates of ethylene dihalides with protein and DMA in mice and rat tissue



in vitro. Proc. Amer. Assoc. Cancer Res. 19: 67.








Banerjee,  S.;   Van Duuren,  B.L.  (1979)  Binding of carcinogenic  halogenate



hydrocarbons to cellular macromolecules. J. Natl. Cancer Inst. 63: 707.








Banerjee,  S.;  Van Duuren,  B.L.;  Kline, S.A.  (1979) Interaction  of potential



metabolites of the carcinogen ethylene dibromide with protein and DNA in vitro.



Biochem. Biophys. Res. Comm. 90: 1214-1220.








Banerjee, S.; Van Duuren, B.L.;  Oruambo,  F.I.  (1980) Microsome-mediated covalent



binding  of 1,2-dichloroethane to lung microsomal protein and salmon sperm DNA.




Cancer Res. 40: 2170-2173-
                                      10-3

-------
Bang,  Y.H.;  Telford,  H.S.  (1966)  Effect  of sublethal  doses of  fumigants  on



stored-grain  insects. Washington Agricultural  Experiment  Station,  Washington



State University; Technical Bulletin  50.








Barber, E.D.; Donish, W.J.; Mueller, K.R.  (1981) A procedure for the quantitative




measurement   of   the   mutagenicity   of   volatile   liquids   in   the   Ames



Salmonella/microsome  assay. Mutat.  Res. 90: 31-48








Barrows,  M.E.;  Petrocelli,  S.R.; Nacek, K.J.;  Carrell,  J.J.  (1980)  Bioconcen-



tration  and  elimination  of  selected water  pollutants  by  bluegill  sunfish



(Lepomia  macrochirus).  In:   Haque,  R.,  ed.  Dynamics,   exposure,  and  hazard



assessment of toxic chemicals.  Ann  Arbor, MS: Ann Arbor Science, pp. 379-392.








Barsoum, G.S.; Saad, K. (1934) Relative toxicity of  certain  chlorine derivatives



of the aliphatic series. Quart.  J.  Pharm. Pharmacol. 7: 205-214.








Bellamy,  R.;  Schwartz, W. (1975) Engineering  and cost study  of air  pollution



control  for  the petrochemical  industry:  vinyl chloride  manufacture by  the



balanced  process, v.  8.  Marcus Hook,  PA: Air Products and  Chemicals,  Inc.  EPA



report no. EPA-450/3-73-006H. Available from: NTIS, Springfield, VA; PB 252 247.








Benoit,  D.A.;  Puglisi,  P.A.;  Olson,  D.L.  (1982)  A fathead  minnow  Pimphales



promelas  early life  stage toxicity  test method evaluation and exposure to four



organic chemicals. Environ. Pollut. Ser. A. 28(3):  189-197.
                                      10-4

-------
Berg, E.L.  (1982)  Handbook  for sampling  and  sample preservation  of  water and



wastewater. Cincinnati, OH:  U.S. Environmental Protection Agency, Environmental



Monitoring and Support  Laboratory.  EPA report no.  EPA  600/4-82-029-  Available



from: NTIS, Springfield, VA; PB83-1245D3.








Berck, B. (1965)  Sorption of ethylene dibromide, ethylene dichloride, and carbon



tetrachloride by cereal products. J. Agric. Food Chem. 13: 2*48-254.








Berck, B. (1974)  Fumigant residues of carbon tetrachloride, ethylene dichloride,



and ethylene dibromide  in wheat,  flour, bran, middlings,  and bread.  J. Agric.



Food Chem. 22:  977-984.








Blackford,  J.L.  (1974) Ethylene Dichloride.  In:   Chemical  economics  handbook.



Menlo Park, CA: Stanford Research Institute,  pp.  651.5031a-651.5032z.








Bloch,  W.  (1946)  Two  poisonings   from dichloroethane  used  for purposes  of



inebriation. Schweiz Med. Wochenschr.  76: 1078-1079.







Bogoyavlenski,  V.F.;  Salikhova,  S*K.H.; Karpova,   E.V.  (1968) Clinical aspects



and therapy for ethylene dichloride poisoning. Sov. Med. 31:  107-109.








Bolt, H.M. (1978) Pharmacokinetics  of vinyl chloride. Gen. Pharmacol.  9: 91.








Bonitenko,   Y.Y.   (1974)   Aspartate-amino-transferase   isoenzymes   in  acute



dichloroethane  poisoning. Gig. Tr.  Prof. Zabol. 7: 46-47.
                                      10-5

-------
Bonitenko,  Y.Y.;  Bruk,   A.A.;  Belouson,  Ye.  A.;  Lebede,  M.F.   (1977)  Acute



dichloroethane  poisoning and intensive treatment procedure. Voen.-Med.  Zh. 11:



75-77.







Borisova, M.K.  (1957) Experimental  data  for  determination of the maximum allow-



able concentration of dichloroethane  in  the  atmosphere. Gig. Sanit. 22:  13-19.







Bouwer,   E.J.;   McCarty,  P.L.  (1983)  Transformations  of  1-  and  2-carbon



halogenated  aliphatic organic  compounds under  methanogenic  conditions.  Appl.



Environ.  Microbiol.  15:  1286-1294.







Bozzelli,  J.W.; Kebbekus,  B.B. (1979)  "Analysis of  Selected  Volatile  Organic



Substances in Ambient Air," prepared  for New Jersey Department of Environmental



Protection,  New Jersey Institute  of Technology,  NJ. NTIS PB 80-144694.







Bozzelli, J.W.; Kebbekus,  B.B.; Greenburg, A.  (1980)  Analysis of selected toxic



and carcinogenic  substances in ambient  air. Prepared for  Office of Cancer and



Toxic Substances Research,  New Jersey Department of Environmental Protection.







Brass, K. (1919) Concerning a  lethal  dichloroethane poisoning. Med. Wochenschr.



74: 553-551*.







Brass, H.J.  (1981)  An overview of  organics  data: community water supply survey



and  rural water  survey.  Technical  Support Division,  Drinking  Water  Quality



Assessment Branch, U.S.  Environmental Protection Agency.
                                       10-6

-------
Bray, H.;  Thorpe, W.; Vallance, D.  (1952)  The liberation of chloride ions from



organic chlorocompounds by tissue extracts. Biochem. J. 51: 193-201.








Brem,  H.;  Stein,  A.B.;  Rosenkranz,   H.S.  {197*0  The  mutagenicity and  DNA-




modifying effects of haloalkanes. Cancer Res. 34: 2576-2579.








Brodzinsky, R.;  Singh,  H.B.  (1982)  Volatile organic  chemicals  in the atmosphere:



assessment of available data.  Research  Triangle Park,  NC:  U.S.  Environmental



Protection  Agency,  Environmental  Sciences   Research  Lab.   EPA  report  no.



EPA-600/3-83-027A. Available from:  NTIS,  Springfield, VA; PB83-195503.








Brown, R.H.;  Purnell,  C.J.  (1979) Collection and  analysis of trace organic vapor



pollutants in ambient atmospheres.  The performance of a Tenax  GC adsorbent tube.



J. Chromatogr. 178: 79-90.








Bryzhin, F.F. (1945) Pathomorphological changes of internal organs in connection



with  poisoning  by ethylene dichloride through the  digestive tract.  Fannakol.




Toksikol.  8:  43-49.








Brzozowski,  J.;  Czajka,  J.; Dutkiewicz,  T.;  Kesy,   I.;  Wojcik,  J.  (195*0 Work



hygiene  and  the  health  condition  of  workers  occupied   in  combating  the



Leptinotarsa decemlineata with HCH and dichloroethane. Med. Pr. 5: 89-98.








Buccafusco, R.J.; Ellis, S.J.;  LeBlanc,  G.A.  (1981) Acute toxicity of priority




pollutants to bluegill (Lepomis macrochirus). Bull. Environ. Contain. Toxicol.



26(4): 416-1)52.
                                      10-7

-------
Burns,  L.A.;  Cline,  D.M. ;  Lassiter,  R.R.  (1982)  Exposure  analysis  modeling




system   (EXAMS):  user  manual   and   system   documentation.  Athens,  GA:  U.S.



Environmental Protection  Agency, Environmental Research Laboratory,  EPA report



no. EPA-600/3-82-023. Available  from:  NTIS,  Springfield, VA; PB82-25B096.








Butler,  R. ; Solomon, I.J.; Snelson, A. (1978) Rate constants  for the reaction of




OH with halocarbons in the presence  of Op + Np. J. Air Pollut. Control Assoc.




28(11):  1131-1133.








Byers,  D.H.  (1943) Chlorinated  solvents in  common wartime  use.  Ind.  Med. 12:
Catalytic.  (1979)  Draft Summary Report of BAT-308  Responses.







Callahan, M.A.; Slimak,  M.W. ; Gabel, N.W.; May, I. P.; Fowler, C.F. (1979) Water-



related environmental  fate of 129 priority pollutants, v. 2. Washington, DC: U.S.



Environmental  Protection Agency,  Office of Water  Planning  and  Standards;  EPA



report   no.   EPA-440/4-79-029b.   Available   from:  NTIS,   Springfield,   VA;



PB80-204381.







Calvert,  J.G.  (1976)  Hydrocarbon involvement  in photochemical  smog  formation in



Los  Angeles atmosphere. Environ. Sci.  Technol. 10: 256-262.







Cetnarowicz,   J.   (1959)  Experimental  and  clinical   studies  on  effects  of



dichloroethane.  Folia. Med.  Cracov. 1: 169-192.
                                       10-8

-------
Chemical and Engineering News. (1979) Key Chemicals -  Vinyl  Choride.  C and EN,




June 24, 1979, p. 18.







Chemical and Engineering News. (1982) Key chemicals - Vinyl Chloride.  C and EN,




June 21, 1982, p. 18.








Chemical and Engineering News. (1983) Key Polymers -  Vinyl Chloride.  C and EN,




June 20, 1983, p. 17.








Chemical Economics Handbook. (1975) SRI International.








Chemical Economics Newsletter. (1978) SRI International.








CMR   (Chemical   Marketing   Reporter).  (1974)  Chemical   Profile  -   Ethylene



Dichloride. Chemical Marketing Reporter, June 10,   1974.








CMR   (Chemical   Marketing   Reporter).  (1977)  Chemical   Profile  -   Ethylene



Dichloride. Chemical Marketing Reporter, May 16, 1977.








CMR   (Chemical   Marketing   Reporter).  (1980)  Chemical   Profile  -   Ethylene



Dichloride. Chemical Marketing Reporter, May 5, 1980.








CMR  (Chemical  Marketing  Reporter).  (1982)  Chemical   Profile  -  Lead Alkyls.



Chemical Marketing Reporter, February 1, 1982.








CMR   (Chemical   Marketing   Reporter).  (1983)  Chemical   Profile  -   Ethylene



Dichloride. Chemical Marketing Reporter, June 13,   1983.
                                      10-9

-------
Chiou, C.T.; Peters,  L.J.;  Freed,  V.H.  (1979)  A physical concept of soil-water



equilibria  for nonionic organic compounds. Science 206: 831-832.







Chiou,  C.T.;  Freed,  V.H.;  Peters, L.J.;  Kohnert,  R.L.  (1980)  Evaporation  of



solutes from water. Environ.  Int.  231-236.







Coleman, W.E.; Lingg, R.D.; Melton, R.G.; Koppler,  F.C.  (1976) The occurrence of



volatile organics  in  five drinking water supplies using gas chromatography-mass



spectrometry.  Zdentif. Anal.  Org. Pollut.  Water.  (Chem.  Congs.  North  Amer.



Cont.)  1st  1975, Ann  Arbor  Science, Ann  Arbor,  MI. pp. 305-327.







The Condensed Chemical Dictionary. (1977) 9th ed. G.G. Hawley, ed., Van Nostrand



Reinhold Company,  NY.







Conkle,  J.P.;  Camp,  B.J.;  Welch,   B.E. (1975)  Trace  composition of  human



respiratory gas. Arch.  Environ.  Health.  30:  290-295.







Cooper,  C.V.;  White,  L.D.; Kupel, R.E.  (1971)  Qualitative detection limits for



specific compounds utilizing gas chromatographic fractions, activated charcoal,



and a mass  spectrometer.  J. Amer. Ind.  Hyg.  Assoc.  32:  383-386.







Cox,  C.B.   (1972)  Regression  model and life tables. J. Roy.  Stat.  Soc.  B. 31:



187-220.







Crump, K.S.; Guess.  H.A.;  Deal,  L.L.  (1977)  Confidence intervals and test of



hypotheses   concerning   dose-response   relations   inferred    from   animal



carcinogenicity data. Biometrics. 33:
                                      10-10

-------
Crutzen,  P.J.;  Fishman,   J.   (1977)  Average   concentrations  of  *OH  in  the



troposphere and the budgets of CHj.,  CO,  H_, and CH^CCl,. Geophys. Res. Lett. 4:




321-324.







Cupitt, L.T. (1980) Fate of toxic and hazardous materials in the air environment.




Research Triangle Park, NC: U.S. Environmental Protection Agency, Environmental



Sciences Research  Laboratory;  EPA report  no. EPA-600/3-80-084.  Available from:



NTIS, Springfield, VA; PB80-221948.








Daffer, P.; Crump, K.; Masterman, M. (1980) Asymptotic theory  for  analyzing dose-




response  survival data with  application to  low-dose extrapolation problem.



Math. Biosci. 50: 207-230.








Danz, M.;  Urban,  H.  (1979) Carcinogens, promoters,  and suspicious drugs: Their



identical short-term effect in a promoting activity  test (PAT). Exp. Pathol. 17:



181-184.








Davidson,  I.W.F.  (1984)   The  biological  basis for  extrapolation  of  dose-



carcinogenic  response  relationship  of   laboratory  animals.  A review  paper



prepared for the Carcinogen Assessment Group.








Dedrick, R.L.  (1973) Animal scale-up. J.  Pharmacokinet. Biopharm. 1:  1435-461.








Dedrick, R.L.;  Bischoff,  K.B.  (1980)  Species similarities in pharmacokinetics.




Fed.  Proc. 39: 54-59.
                                     10-11

-------
Deinzer,  M.;  Schaumburg,  F.;  Klein,  E.  (1978)  Environmental  health sciences



center. Task force review of halogenated organics  in drinking water. EHP Environ.




Health Perspect. 24:  209-239.







Delplace, Y.;  Cavigneaux,  A.;  Cabasson, G.  (1962) Occupational  disorders due to




methylene chloride  and dichloroethane.  Arch.  Mai.  Prof.  Hyg.  Toxicol.  Ind.




23: 816-817.







Dilling,  W.L.;  Tefertiller,  N.B;  Kallos,  G.J.   (1975)  Evaporation  rates and



reactivities   of   methylene   chloride,    chloroform,   1,1,1-trichloroethane,



trichloroethylene,   tetrachloroethylene,   and  other  chlorinated  compounds  in



dilute  aqueous solutions.  Environ.  Sci. Technol.  9:  833-838.








Doll,  R.  (1971) Weibull  distribution of  cancer: Implications  for  models of




carcinogenesis.  J.  Roy. Stat.  Soc.  A 13:  133-166.








Domenici, F.  (1955) Granoaan intoxication. Bass.  Clin-Sci. 31:  70-73-








Dowty,  B.J.;  Carlisle, D.R.;  Laseter, J.L.  (1975)  New  Orleans  drinking  water



sources tested  by  gas chromatography-tnass spectrometry.  Environ. Sci.  Technol.




9: 762-765.







Dripps, R.D.; Eckenhoff,  J.E.;  Vandam, L.D. (1977)  Introduction to anesthesia:



The principles of safe practice. 5th ed. Philadelphia,  PA: W.B. Saunders Company,




 pp. 121-123-
                                       10-12

-------
Drury,  J.S.;  Haramons,  A.S.  (1979)  Investigations  of  selected  environmental



pollutants:  1, 2-dichloroethane.  Washington,  DC: U.S. Environmental  Protection



Agency, Office of Toxic Substances; EPA  report  no.  EPA-560/2-78-006.  Available



from: NTIS, Springfield, VA;  PB-295-865.








Edwards, K.; Jackson,  H.;  Jones,  A.R.  (1970) Studies with alkylating esters. II.




A chemical interpretation  through metabolic studies of the antifertility effects



of ethylene dimethanesulphonate and ethylene dibromide.  Biochem. Pharmacol. 19:




1783.








Ehrenberg,  L.;  Osterman-Golkar,  S.;  Singh,   D.;  Lundquist,  U.  (197U)  On the



reaction kinetics and mutagenic  activity of methylating and halogenating gaso-




line additives. Radia. Hot. 15: 185-19^.








Eimutis,  E.G.;  Quill,  R.P.  (1977) Source  assessment:   non-criteria  pollutant



emissions.  Research Triangle  Park, NC:  U.S.  Environmental  Protection Agency,



Industrial Environmental Research Laboratory.  EPA report no. EPA-600/2-77-107e.



Available  from: NTIS,  Springfield,  VA; PB270 550.








Elfers, L.  (1979) Monitoring of  ambient  levels  of ethylene dichloride (EDC) in



the vicinity of EDC production and user  facilities. Research Triangle Park, NC:



U.S.  Environmental Protection  Agency,  Environmental  Monitoring and  Support




Laboratory. EPA report no. EPA-600/iJ-79-029. Available  from: NTIS, Springfield,




VA; PB-298  303-
                                     10-13

-------
Ellis, C.R.; Morrison, P.O. (1967) Small chamber tests of ethylene dibromide and



ethylene  dichloride  on adult  grain-infesting  coleoptera.  Can.  J.  Zool.  45:



U35-118.







Ehrenberg,  L.;  Osterman-Golkar,  S.;  Singh,  D.;  Lindquist,  U.  (197*0  On  the



reaction kinetics  and rnutagenic activity of methylating and B-halogenethylating



gasoline additives.  Radia.  Hot. 15:  185-191.







Ethylene Dichloride. In:  Annual report of  the Chief  Inspector of Factories for



the year  1915.  London, United  Kingdom, His Majesty's Stationary Office; p. 77.



1916.







Ewing,  B.B.;  Chian,  E.S.K.;  Cook,   J.C.; Evans,  C.A.;  Hopke,   P.K.  (1977)



Monitoring  to  detect  previously  unrecognized  pollutants  in surface waters.



Washington,   DC:   U.S.  Environmental  Protection  Agency,   Office  of  Toxic



Substances;  pp. 63,  61,  73.  EPA  report  no.  EPA-560/6-77-015.  Available from:



NTIS, Springfield,  VA; PB-273 319/1.







Fabricant,  J.D.; Chalmers,  J.G., Jr. (1980) Evidence of mutagenicity of ethylene



dichloride  and  structurally related compounds.  In: Ames,  B.N.; Infante, P. and



Reitz, R.,  eds. Ethylene dichloride: a potential  health risk? Cold Spring Harbor,



NY: Cold Spring Harbor Laboratory; pp. 309-329.  (Banbury  report no. 5).







Federal Register.  (1971)  36(288):  22512.







Finnegan,  R.J.; Stewart,  K.E.  (1962) Control of the pine root collar weevil,



Hylobius radicus.  J. Econ. Entomol.  55:  183-186.
                                      10-11

-------
Filser, J.G.;  Bolt,  H.M.  (1979) Pharmacokinetics  of halogenated  ethylenes in



rats. Arch. Toxicol. 42:  123.








Fishbein,  L. (1976) Industrial mutagens  and  potential  mutagens.  I. Halogenated




aliphatic derivatives. Mutat.  Res.  32: 267-308.








Fishbein,  L. (1979) Potential  halogenated industrial carcinogenic and mutagenic



chemicals.  III.  Alkane halides, alkanols  and ethers.  Sci.  Tot.  Environ.  11:




223-257.







Flowtow, E. (1952) Poisoning due to chlorinated hydrocarbon compounds, particu-



larly 1,2-dichloroethane.  Chem.  Tech. (Leipzig) 6:  253-251.








Freireich,  E.J.;  Gehan, E.A.; Rail,  D.P.;  Schmidt,  L.H.;  Skipper,  H.E.  (1966)



Quantitative comparison of toxicity of anticancer agents  in mouse, rat, hamster,



dog, monkey, and man. Cancer Chemother.  Rep.  50:  219-244.








Freundt, K.J.; Eberhardt,  H.; Walz,  U.M. (1963) Lethal  peroral  poisoning with



1,2-dichloroethane  and  2,2-dichlorodiethylether.   Int.  Arch.  Gewerbepathol.




Gewerbehyg. 20: 41-48.








Fujii,  T.  (1977)  Direct aqueous injection chromatograph-mass  spectroznetry for



analysis of organohalides  in water  at concentrations  below the parts per billion




level.  J.  Chroraatogr. 139: 297-302.








Garrett, J.T.  (1957) Toxicity  investigations on aquatic and marine life. Public




Works 88:  95-96.
                                     10-15

-------
Garrett, J.T. (1957) Toxicity considerations in pollution control. Ind. Wastes.




2: 17-19.







Garrison,  S.C.;  Leadingham, R.S.  (1951) A  fatal  case of  ethylene  dichloride



poisoning  in an  occupatonal  therapy department of a neuropsychiatric hospital.




Am. J. Phys. Med. 33: 230-237.







Gast,  R.;  Early, J.  (1956)  Phytotoxicity of  solvents  and  emulsifiers used in



insecticide formulations. Agric. Chem.  11: H2-H5,  136-139.







Gehring,   P.J.;   Watanabe,  P.G.;   Blau,  G.E.   (1980)  Risk  assessment  of



environmental carcinogens utilizing pharmacokinetic  parameters.  Ann.  NY Acad.



Sci.  329:  137-152.







Gehring, P.J.; Watanabe,  P.G.;  Blau, G.E.  (1976) Pharmacokinetic  studies in the



evaluation of the  toxicological and  environmental  hazards of  chemicals.  In:



Mehlman,   M.A.;  Shapiro,   R.E.;  Blumenthal,   H.,   eds.,   Advances  in  Modern



Toxicology. Washington,  DC:  Hemisphere Publishing  Co. v.  1,  part  1.







Gehring,   P.J.;  Watanabe,  P.G.;  Young,  J.D.  (1977)  The  relevance  of dose-



dependent   pharmacokinetics  in  the  assessment  of  carcinogenic  hazard  of



chemicals. Cold  Spring Harbor  Conference, Cell Proliferation U:  187.







Gehring,  P.J.;  Watanabe,  P.G.; Park,   C.N.  (1978)  Resolution  of dose response



toxicity  data  for  chemicals   requiring  metabolic  activation:  Example  -  vinyl



chloride.  Toxicol.  Appl. Pharmacol. 44: 581.
                                      10-16

-------
Cikalov,  G.S.;  Chemanaev, K.A.;  Janda, M.U.  (1969) Clinic  and  treatment  of



dichloroethane poisoning. Voen-Med. Zh. 1:  78-79.








Gillette, J.R. (1976)  Application of pharmacokinetic principles in extrapolation




of animal data to humans. Clin. Toxicol. 9: 709-722.








Gocke, E.; Wild, D.;  Eckhardt,  K.;  King, M. (1983) Mutagenicity studies with the




mouse spot test. Mutat. Res. 117: 201-212.








Gold, L. (1980) Human Exposures to  Ethylene Dichloride. In: Ames, B.N.j Infante,




P.;  Reitz,  R.,  eds.  Ethylene dichloride:  a  potential  health  risk? Cold Spring



Harbor, NY: Cold Spring Harbor Laboratory;  pp. 209-226. (Banbury report no. 5).








Grimsrud,  E.;  Rasmussen, R. (1975) Survey and analysis  of halocarbons in the



atmosphere  by   gas   chromatography-mass  spectrometry.   Atmos.   Environ.  9:




1014-1017.







Guarino,  A.; Lioia, N. (1958) Clinical and  histopathological findings on a  fatal




poisoning with Granosan.  Folia. Med.  (Naples). 41: 676-690.








Guengerich, P.P.;  Crawford,  W.M.; Domradizki,  J.Y.;  McDonald, T.L.; Watanabe,



P.G.  (1980)  In  yitro  activation of  1,2-dichloroethane by microsomal and




cytosolic enzymes. Toxicol.  Appl.  Pharmacol. 55:  303-317.








Guerdjikoff,  C.  (1955) Acute and chronic occupational intoxication by symmetric



dichloroethane.  Doctoral thesis No.  2325.  Geneva,  Switzerland,  University of



Geneva,  Faculty  of Medicine. 107 p.
                                      10-17

-------
Hadengue, A.;  Martin,  R.  (1953)  A  case of fatal  poisoning  by dichloroethane.



Soc. Med. Leg. 33: 247-249-







Handbook of Environmental Data on Organic Chemicals.  (1977) K. Verschueren, ed.,



New York, NY:  Van Nostrand and  Co.







Hanst, P.L.  (1978)  Part II.  Halogenated pollutants:  Noxious trace gases in the



air. Chemistry 51: 6-12.







Hardie,  D.W.F.  (1964)  Chlorocarbons  and  chlorohydrocarbons.  In:  Kirk-Othmer



encyclopedia  of  chemical  technology,  2nd ed., v. 5,  New York,  NY: Interscience



Publishers; pp.  152-154.







Hawley,  G.G.  (1981) The Condensed  Chemical Dictionary.  10th ed. New York, NY:



Van Nostrand  Reinhold Co.; p.  431.







Hayes,  F.D.;  Short, R.D.;  Gibson,  J.E.  (1973)  Differential toxicity of mono-



chloroacetate,  monofluoroacetate and  monoiodoacetate in rats.  Toxicol.  Appl.



Pharmacol.  26: 93-102.







Heitmuller,  P.T.; Hollister,  T.A.;  Parrish,  P.R. (1981) Acute  toxicity of 54



industrial  chemicals   to  sheepshead  minnows  (Cyprinodon   variegatus).  Bull.



Environ.  Contam. Toxicol.  27(5): 596-604.







Hellman,  T.M.; Small, F.H. (1974) Characterization of the odor  properties  of 101



petrochemicals  using   sensory  methods.  J.  Air  Pollution   Control  Assoc.  24:




979-982.
                                      10-18

-------
Hellnan, T.M.;  Small,  F.H.  (1973)  Characterization of odors  from  the  petro-



chemical industry. Chera.  Eng. Prog.  69: 75.








Heppel, L.A.;  Neal, P.A.; Pen-in, T.L.; Endicott,  K.M.; Porterfield, V.T.  (19^5)



Toxicology of 1,2-dichloroethane.  III.  Its  acute toxicity  and  the  effect  of




protective agents. J. Exp. Pharmacol. Ther.  83: 53-63-








Heppel, L.A.;  Neal,  P.A.;  Perrin, T.L.; Endicott, K.M.; Porterfield, V.T. (19t6)



Toxicology of 1,2-dichloroethane. V. Effects of daily inhalations. J. Ind. Hyg.




Toxicol. 28:  113-120.








Heppel,  L.A.;   Porterfield,  V.T.;  Sharpliss,  N.G.  (1947)   Toxicology  of



1,2-dichloroethane.  IV.  Its  detoxification  by  L-cystine,   D,L-methionine and



certain  other  sulfur-containing  compounds.  J.  Pharmacol.  Exp.  Ther.  91:




385-394.







Heppel,  L.A.;  Porterfield,  V.T.  (1948) Enzymatic  dehalogenation  of certain



brominated and  chlorinated compounds. J. Biol. Chem. 176:  763-769.








Heukelekian,  H.;  Rand, M.C.  (1955)  Biochemical  oxygen demand of pure organic




compounds. J. Water Pollut.  Control  Aasoc. 29: 1040-1053.








Hill,  D.T.;   Shin,  T.-W.; Johnston,  T.P.;  Struck,   R.F.  (1978)  Macromolecular



binding and   metabolism of the carcinogen  1,2-dibromoethane.  Cancer  Res. 28:




2438.
                                      10-19

-------
Hinkel,  G.K.   (1965)   Oral  dichloroethane   poisoning   in  children.   Dtsch.



Gesundheitswes. 20: 1327-1331.







Hobbs, F.D.; Key, J.A.  (1978) Emissions Control Options for the Synthetic Organic



Chemicals Manufacturing Industry.  Draft  report.  EPA  contract number 68-02-2572.



Hydroscience,  Inc.,  Knoxville, TN.  Report prepared for  Office  of Air Quality



Planning and Standards, U.S.  Environmental Protection Agency, Research Triangle



Park, NC.







Holman, H.T.; Birnstiel, H.;  Jobst, P. (1971) On the inhalation toxicity of 1,1-



and  1,2-dichloroethane. Arch. Toxicol. 27: 218.







Hooper, K.;  Gold, L.; Ames,  B.  (1980)  The carcinogenicity potency of ethylene



dichloride  in  two animal  bioassays: a  comparison  of   inhalation  and  gavage



studies.  In:  Ames, B.N.;  Infante, P.;  Reitz,  R.,  eds.  Ethylene dichloride: a



potential health risk? Cold Spring Harbor,  NY: Cold Spring Harbor  Laboratory; pp.



65-82.  (Banbury report no.  5).







Howard, C.J.;  Evenson, K.M.  (1976) Rate constants for the  reactions of OH with



ethane  and  some  halogen  substituted  ethanes  at  296°K.  J.  Chem.  Phys.  64:



4303-4306.







Howe,  R.B.;  Crump, K.S.  (1982) A computer  program to extrapolate  quantral animal



animal  toxicity data to low doses. OSHA Contract 41  USC252C3.
                                      10-20

-------
Howe,  R.B.  (1983) GLOBAL83:  an experimental  program developed  for  the U.S.



Environmental Protection Agency as an update to GLOBAL82:  a computer program to



extrapolate quantal animal toxicity  data  to low doses (May  1982).  Ruston, LA:



K.S. Crump and Co., Inc. Unpublished.








Hueper, W.C.;  Smith,  C. (1935)  Fatal  ethylene dichloride poisoning.  Amer. J.




Med. Sci. 189: 778-784.







Hulst,  J.P.L.;  Steenhauer,  A.J.;  Keede,  D.L.  (1946)  Lethal poisoning with




dichloroethane. Ned. Tijdschr Geneeskd. 90: 406-407.








lenistea, C.;  Mezincesco, M.D.  (1943)  Fatal poisoning by ingestion of ethylene




dichloride. Bull. Acad. Med. Roum. 8: 614-617.








International  Agency for Research on Cancer. (1979) Monograph on the evaluation



of  carcinogenic  risk of chemicals to humans. 20: 429-448.








Irish,  D.D.  (1963) Aliphatic  Halogenated Hydrocarbons. In:  Fassett,  D.W. and



Irish,  D.D., eds.  Industrial Hygiene and Toxicology:  2nd revised ed.,  New York,



NY:  Interscience  Publishers; pp. 1280-1284.








Ivanetick, K.M.; Lucas, S.;  March,  J.A.; Ziman, M.R.;  Katz, I.D.; Bradshaw,  J.J.



(1978)  Organic  compounds.  Their interaction  with and  degradation of  hepatic



microsomal drug-metabolizing enzymes i.n vitro. Drug Met. Dispos. 6:  218-225.








Jaeger, R.J.; Conolly,  R.B.;  Murphy,  S.D.  (1974) Effect of 18-hour fast and



glutathione  depletion  on  1,1-dichloroethyl-induced hepatotoxicity  and  lethality




in  rats.  Exp.  Mol.  Pathol. 20:  187-198.
                                      10-21

-------
Jaeger, R.J.  (1979) Time-related variation of non-protein aulhydryl  concentra-



tions in rat  tissue and human  blood. Int.  Arch. Occup. Environ. Health 42:  141-



118.







Jakobson, I.; Wahlberg, J.E.;  Holmberg, B.; Johansson, G. (1982) Uptake via the



blood and elimination  of  10 organic  solvents  following epicutaneous exposure of



anesthetized  guinea pigs.  Toxicol. Appl.  Pharmacol.  63: 181-1&7.







Jenssen,  D.;  Ramel, C.  (1980) The micronucleas  test  as  part  of  a  short-term



mutagenicity  test program  for  the prediction of carcinogenicity evaluated by 143



agents  tested. Mutat.  Res. 75: 191-202.







Johnson,  M.K. (1965)  The  influence  of  some aliphatic compounds  on rat liver



glutathione levels. Biochem. Pharmacol.  14:  1383-1385.







Johnson,  M.K. (1966a)  Metabolism of  iodomethane in the  rat.  Biochem.  J. 98:




38-43.







Johnson,  M.K.  (1966b) Studies on  glutathione S-alkytransferase of  the  rat.



Biochem.  J. 98:  44-56.







Johnson,  M.K. (1967) Metabolism of chloroethanol in the rat. Biochem.  Pharmacol.




 16:  185-199.







Jones,   A.R.;  Edwards,  K.   (1968)   The  comparative  metabolism of ethylene



dimethanesulphonate and ethylene dibromide. Experientia 24: 1100-1101.
                                      10-22

-------
Jordi, A.  (1911)  Industrial  poisonings due to  symmetrical  1 ,2-dichloroethane.



Z. Unfallmed. Berufskr. 37: 131-136-







Kaira,  F.M.  (1966)  Alimentary  oral  dichloroethane  poisoning.  Klin.  Med.




(Moscow). 11:
Kaplan,  E. ;   Meier,   P.  (1958)  Nonparametric   estimation  from  incomplete




observations. J. Am. Stat. Assoc. 52: 457-181.








Kellara,  R.G.;  Dusetzina, M.G.  (I960) Human  exposure to  ethylene dichloride:



Potential  for  regulation via  EPA's  proposed  airborne carcinogen  policy.  In:



Ames, B.N. ; Infante, P.;  Reitz,  R., eds. Ethylene  dichloride: a potential health



risk?  Cold Spring  Harbor,   NY:  Cold  Spring  Harbor  Laboratory;  pp. 265-271.



(Banbury report no. 5).








Kenaga,  E.E.  (1980)  Predicted  bioconcentration  factors  and  soil  sorption



coefficients of pesticides and other chemicals. Ecotoxicol. Environ. Safety. 1:




26-38.








Keyzer,  J.L.   (1911)  Lethal   poisoning   by  dichloroethane.   Ned.  Tijdschr.



Geneeskd.  88: 611.








King, M.T. ; Beikirch, H.  ; Echkhardt, K.; Gock, E. ; Wild, D.  (1979) Mutagenicity



studies with x-ray contrast media, analgesics,  antipyretics,  antirheumatics , and



some  other pharmaceutical drugs in  bacterial,  Drosophila  and  mammalian test



systems. Mutat. Res. 66:  33-13-
                                     10-23

-------
Kirk-Othmer. (1964) Encyclopedia  of Chemical Technology.







Kirichek, lu. F.  (1974) Effects of 1,2-dichloroethane. Usp. Khim. Mutagen.  Sel.



232-235.







Kistler, G.H.; Luckhardt, A.B. (1929)  Die 13 wichtigsten Chlorkohlenwasserstaffe



der Fattreihe vom Standpunkt  der  Gewerbehydiene. Anesth. Analg. 8: 65-78.







Kokarovtseva,  M.G.;  Kiseleva, N.J.   (1978)  Chloroethanol,  one  of the  toxic



metabolites of  1,2-dichloroethane. Farmakol. Toksikol. 41: 118-120.







Kovar,  J.;  Krewski,  D.   (1981)  RISK81:  A  computer  program  for  low-dose



extrapolation  of quantal  response  toxicity  data.  Department  of Health  and



Welfare, Canada.







Kozik, I. (1957) Problems of occupational hygiene in the use of dichloroethane in



the aviation industry. Gig. Tr. Prof.  Zabol.  1:  31-38.







Krasovski, G.N.  (1976) Extrapolation  of  experimental  data from  animal  to  man.



Environ.  Health  Perspect.  13:  51-58.







Kristoffersson,  U.  (1974) Genetic effects of  some  gasoline additives. Hereditas



78: 319.







Krohne, H.E.; Lingren, D.L. (1958)  Susceptibility of life stages of Sitophilus



oryza  to various fumigants.  J.  Econ.  Entomol.  51(2):  157-158.
                                      10-24

-------
Lane, R.W.; Riddle; B.L.;  Borzelleca,  J.F. (1982) Effects of 1,2-dichloroethane



and  1,1,1-trichloroethane in drinking water  on  reproduction and development in



mice. Toxicol. Appl. Pharmacol. 63: 409-421.








Larkin,  R.L.;  Crable,  J.V.;  Catlett,  L.R.;  Seymour,  M.J.  (1977)  Collaborative




testing  of .a gas  chromatographic charocoal  tube  method  for seven  organic




solvents. Amer. Ind. Hyg.  Assoc. 38: 543-553-








LeBlanc, G.A.  (1980) Acute toxicity of priority pollutants to water flea (Daphnia




magna).  Bull.  Environ. Contam.  Toxicol.  24: 684-691.








Lehmann, K.B.; Schmidt-Kehl, L.  (1936) The  pharmacology of some ethylene-halogen




compounds. Arch. J. Hyg.  116:  131-143.








Letkiewicz, P.;  Johnston, P.;  Colman,   J.;  et  al.  (1982) Occurrence  of 1,2-



Dichloroethane in Drinking Water, Food and Air. EPA Contract No. 68-01-6185, Task



11.








Lindgren, D.L.; Vincent,  L.E.;  Krohne, H.E.  (1954) Relative effectiveness of 10




fumigants to  adults of 8  species of stored-product  insects.  J.  Econ. Entomol.



47(5): 923-926.








Lindgren, D.L.; Vincent,  L.E.  (1959) J.  Econ. Entomol. 52:  1091.








Lindgren,  D.L.;  Sinclair;  W.B.;  Vincent,  L.E.  (1968)  Residues  in  raw  and



processed  foods  resulting  from  post-harvest  insecticidal  treatments.  In:



Gunther, F.A., ed. Springer-Verlag, NY:  pp.  1-122. Residue reviews, v. 21.
                                     10-25

-------
Livesey,  J.C.;  Anders,  M.W. (1979) Jn  vitro metabolism of 1,2-dihaloethane to



ethylene. Drug Metab. Dispos.  7:  199-203.







Livesey,  J.C.;  Anders,  M.W.;  Langvardt, P.W.; Putzig, C.L.; Reitz,  H.R. (1982)



Sterochemistry  of   the  glutathione-dependent   biotransformation  of  vicinal-



dihaloalkanes to altcanes.  Drug Metab. Dispos.  10: 201-204.







Lochhead, H.B.; Close, H.P. (1951)  Ethylene  dichloride  plastic cement — a case



of fatal  poisoning.  J. Am.  Med. Assoc.  146:  1323.







Logan,  J.A.; Prather,  M.J.;  Wofsy,  S.C.;  McElroy, M.B.  (1981)  Tropospheric



chemistry: a global  perspective.  J. Geophys.  Res. 86: 7210-7254.







Longbottom, J.E.; Lichtenberg, J.J. (1982) Methods for organic chemical analysis



of  municipal and  industrial  wastewater. Cincinnati,  OH:   U.S.  Environmental



Protection Agency,  Environmental Monitoring and Support Laboratory;  EPA report



no. EPA-600/4-82-057. Available from: NTIS,  Springfield, VA; PB83-201798.







Lovelock, J.E.  (1971) The  electron capture detector:  theory and  practice.  J.



Chromat.  99: 3-12.







Lowenheim,  F.;  Moran,  M.  (1975)  Ethylene  dichloride.  In: Faith,  Keyes,  and



Clark's industrial chemicals,  4th ed.  New York,  NY: John Wiley and Sons, Inc.;



pp. 392-396.







Ludzack,  F.J.;  Ettinger, M.B.  (1960)  Chemical structures resistant  to aerobic



biochemical stabilization.  J.  Water Pollut.  Control Fed.  32: 1173-1200.
                                      10-26

-------
Lutz, W.K.  (1979)  In vivo covalent  binding of organic  chemicals to DNA  as a



quantitative indicator  in  the  process of chemical  carcinogenesis.  Mutat.  Res.



65: 289-356.







Luzhnikov,  E.A.;  Andryukin,  A.A.;  Savina.,   A.S.;  et  al.   (1971)   [No  title




provided]. Ter. Arkh. 46(2):  131-135.







Luzhnikov, E.A.; Petrova,  L.I.;  Savina,  A.S.j Kostomareva, L.G.; Zakharova, A.V.



(1978) Lesion of the heart in exotoxic shock.  Kardiologiya.  18: 83-88.








Luzhnikov, L.I.; Savina, A.S. (1976) Myocardiopathies in acute poisonings. Ter.



Arkh. 18: 81-88.







Lyman, W.J.; Reehe, W.F.;  Rosenblatt, D.H. (1982) Handbook of chemical property



estimation methods. Environmental  behavior  of  organic  compounds.  New York,  NY:



McGraw-Hill Book Co.







Mabey, W.B.; Smith, J.H.;  Podoll, R.T.; et al. (1981) Aquatic fate process data



for organic priority pollutants. Washington,  DC:  U.S.  Environmental Protection



Agency, Monitoring and Data Support Division;  EPA report no. EPA-140/1-81-011.








Maltoni,  C.;  Valgimigli,  L.;  Scarnato,  C.  (1980)  Long-term  carcinogenic



bioassays on ethylene  dichloride administered by inhalation  by  rats and mice.



In: Ames, B.N.;  Infante,  P.; Ritz,  R.,  eds.  Ethylene  dichloride:  a potential



health  risk? Cold  Spring  Harbor, NY: Cold  Spring Harbor Laboratory; pp.  3-33-



(Banbury report no. 5).
                                     10-27

-------
Martin, G.; Knorpp, K.; Huth,  K.;  Heinrich, F.; Mittermayer, C. (1969) Clinical



features,  pathogenesis and  management  of  dichlorethane poisoning.  Ger.  Med.



Mon. m: 62-67.







McCann,  J.;  Simmon, V.;  Streitwiesser,  D.; Ames,  B.N.  (1975)  Mutagenicity of



chloroacetaldehyde, a. possible metabolic product of 1,2-diehloroethane (ethylene



dichloride),  chloroethanol (ethylene chlorohydrin), vinyl chloride, and cyclo-



phosphamide.  Proc.  Natl.  Acad.  Sci.  U.S.A.  72:  3190-3193*







McCollister,  D.D.;  Hollingsworth, R.L; Oyen,  F.; Howe, V.K. (1956) Comparative



inhalation toxicity of fumigant mixtures. AHA Arch.  Ind. Health 13: 1-7.







McConnell, G.; Ferguson, D.M.; Pearson, C.R. (1975) Chlorinated hydrocarbons and



the environment.  Endeavour 3U:  13-18.







McCormack, W.B.;  Moore, R.; Sandy, C.A.  (1981)  Lead compounds (organolead). In:



Grayson,  M.   and  Eckroth,   D.,   eds.   Kirk-Othmer  encyclopedia  of  chemical



technology, 3rd  ed. New York,  NY:  John Wiley and  Sons, Inc. v. 1U, pp. 186-189.







McKenna,  M.J.; Zempel, J.A.;  Madrid,  E.O.; Braun,  W.H.;  Gehring,  P.J.  (1978)



Metabolism and pharmacokinetic profile of vinylidene chloride in rats following



oral administration. Toxicol.  Appl.  Pharmacol.  45:  821-835.







McNally,  W.D.; Fostvedt,   G.  (19^1)  Ethylene  dichloride  - poisoning.  Ind. Med.



10: 373-37H.
                                      10-28

-------
McPherson, R.W.; Starks, C.M.; Fryar, G.J.  (1979)  Vinyl chloride monomer. What



you should know. Hydrocarbon Process. 3: 75-88







Melcher, R.G.; Langner, R.R.; Kagel, R.O. (1978) Criteria for the evaluation of



methods for the  collection  of organic pollutants  in  air using solid sorbents.



Am. Ind. Hyg.  Assoc. 39: 349-361.







Menschick, H.  (1957)  Acute  poisoning from  inhalation of symmetrical dichloro-



ethane. Arch.  Oewerbepathol. Gewerbehyg. 15: 241-252.







Metcalf, R.L.  (1981) Insect  control technology.  In: Grayson, M. and Eckroth, D.,



eds. Kirk-Othmer  encyclopedia  of chemical  technology,  3rd ed.,  New York, NY:



John Wiley and Sons, Inc., v. 13, p. 467.







Hill,  T.; Mabey,  W.R.;  Bomberger,  D.C.;  Chou,  T.-W.;  Hendry, D.G.   (1982)



Laboratory protocols  for evaluating the  fate of organic  chemicals  in air and



water. U.S. Environmental Protection Agency, Athens,  GA: Environmental Research



Laboratory,  U.S.  EPA  report  no.  BPA-600/3-82-022,  Available  from:   NTIS,



Springfield, VA; PB83-150888.







Miller, V.; Dobbs, R.J.; Jacobs, S.I. (1970) Ethylene chlorohydrin intoxication



with fatality. Arch. Dis. Child. U5: 589-590.
Morgan,  A.;  Black,  A.;  Belcher,  D.R.  (1972)  Studies on  the  absorption of



halogenated  hydrocarbons  and  their  exci



techniques. Ann. Occup. Hyg. 15: 273-282.
                                                                   38
halogenated  hydrocarbons  and  their  excretion  in  breath  using    Cl  tracer
                                     10-29

-------
Morozov,  G.N.  (1958)  On  acute  dichloroethane  poisoning.  Pnarmacol.  Toxicol.



Engl. Transl.  21:  80-83.







Mudder,   T.I.   (1982)  Development  of  empirical  structure  biodegradability



relationships  and  biodegradability testing protocol  for  volatile  and slightly



soluble priority pollutants. Presented at Div. Environ.  Chem.,  Amer. Chem. Soc.;



Sept., Kansas  City, MO;  1982.  pp.  52-53.








Munson, A.E.;  Sanders,  V.M.; Douglas,  K.A.; Sain, L.E.; Kaufmann,  B.M.;  White,



K.L.,  Jr.  (1982)  In vivo assessment  of  immunotoxicity.  EHP  Environ.  Health



Perspect. 43:  41-52.








Murray, J.S.;  Rao, K.S.;  Young,  J.T.;  et al.  (1980) Ethylene dichloride:  single



generation  inhalation reproduction study in rats [unpublished study].  Midland,



MI: Toxicology Research  Laboratory, Dow Chemical U.S.A.








Nachtomi, E.;  Alumot, E.;  Bondi,  A. (1966)  The metabolism of ethylene dibromide



in the rat. I.  Identification of detoxification products in urine. Isr. J. Chem.



4: 239-246.








Nachtomi,  E.  (1970)  The metabolism  of ethylene  dibromide  in  the  rat.  The



enzymatic reaction with glutathione .in vivo  and in vitro.  Biochem.  Pharmacol.



19: 2853-2860.








Nakajima,  T.;  Sato,  A.  (1979)   Enhanced  activity of  liver drug-metabolizing



enzymes for aromatic  and  chlorinated  hydrocarbons  following food  deprivation.



Toxicol. Appl.  Pharmacol.  50:  549-556.
                                      10-30

-------
National  Academy  of  Sciences.  (1978)  Non-fluorinated  haloraethanes  in  the



environment.  Panel on  low molecular  weight-halogenated  hydrocarbons on  the



coordinating committee for  scientific and technical assessments of environmental



pollutants.








National  Science Foundation  Panel  on  Manufactured Organic  Chemicals in  the



Environment. (1975) SRI  International Data.








National  Cancer  Institute.  (1978)  Bioassay of  1,2-dichloroethane  for possible



carcinogenicity.  Washington, DC: Department  of Health, Education,  and Welfare



publication no.  (NIH) 78-1361.  (NCI carcinogenesis  technical  report series no.




55).







National  Cancer  Institute.  (1979)  Bioassay of  ethylene dibromide  for possible



carcinogenici ty.








Neely,  W.B.;  Plonka,  J.H.  (1978)  Estimation of  time-average  hydroxyl radical



concentration in the troposphere. Environ.  Sci. Technol. 12: 317-321.








Nestraann,  E.R.;   Lee,  E.; Matula,  T.;   Douglas;  G.R.;  Mueller,  J.  (1980)



Mutagenicity of consituents identified in pulp and paper mill effluents  using the



Salmonella/mammalian-microsome assay. Mutat.  Res. 79:  203-212.








National  Institute  for  Occupational   Safety  and  Health.   (1974)  Manual  of



analytical  methods.   Health,  Education, and  Welfare  Publication  No. (NIOSH)



75-121.
                                     10-31

-------
National  Institute  for  Occupational  Safety  and  Health.  (1975)  Technican



information.  Collaborative testing  of activated  charcoal  sampling  tubes  for



seven organic solvents. Health, Education and Welfare publication 75-184.








National  Institute  for Occupational  Safety and Health.  (1976)  Criteria for a




recommended standard. Occupational exposure to  ethylene dichloride. Washington,



DC: U.S. Department of Health,  Education, and Welfare.








Noetzel,  0.   (1914)  Lethal poisoning with  ethylene  chloride.  Chem.  Ztg.  68:




146-147.








Noweir,  M.H.; Pfitzer, E.A.;  Hatch, T.F.  (1972)  Decomposition of chlorinated




hydrocarbons: a review. J. Am.  Ind.  Hyg. Assoc> 33: 454-460.








Nylander,  P.O.;  Olofsson,  H.;  Rasmuson,   B.  (1979)  The  use  of Drosophila



melanogaster  as a test for indirect  mutagens. Mutat. Res. 64: 122-123*








Ohta,  T.;  Morita, M.; Mizoguchi, I.  (1976)  Local  distribution of chlorinated



hydrocarbons  in the ambient air in Tokyo. Atmos. Environ. 10: 557-560.








Okuno,  T.;  Tsiji, M.;  Shintani,  Y.;  Watanabe,  H.  (1974) Gas chromatography of



chlorinated  hydrocarbons  in  urban   air.   Hyogo-ken  Kogai   Kenkyusho Kenykyuo




Hokoku.  6: 106. (CA  (1977), 87: 72564f).








Ollivier,  H.; Grillo-Abade,  P.;  Helvadjian,  G.;  Quicke,  J. (1954) Report of  a



fatal  case of intoxication by  dichloroethane. Soc. Med.  Leg. 34:  261-264.
                                      10-32

-------
Otterson, E.J.; Guy, C.U. (1964) A method of atmospheric solvent vapor sampling



on activated charcoal in connection with gas chromatography. Trans. 26th Annual



Meeting,   American   Conference   of   Governmental   Industrial   Hygienists;



Philadelphia; pp. 37-43.








Page, B.D.; Kennedy, B.P.C.  (1975) Determination of methylene chloride, ethylene




dichloride, and trichloroethylene  as  solvent residues in spice oleoresins, using



vacuum  distillation and electron  capture gas  chromatography.  J.  Assoc.  Off.



Anal. Chem. 58: 1062-1068.








Page, G.W. (1981) Comparison of groundwater and surface water  for patterns and



levels of  contamination  by  toxic  substances.  Environ. Sci. Technol.  15:  1475-



1481.








Paparopoli,  G.;  Cali,   V.  (1956)  Collective  intoxications  with  chlorinated



hydrocarbons in dock workers.  Folia.  Med. (Naples) 39:  819-831.








Parker,  J.C.;  Davidson,  I.W.F.   (1984)  Extrapolation  of  the  carcinogenic



uncertainty  in risk assessment.  Risk management  and  decision  making p.  38.



Presented at the Society of Risk Analysis.








Patterson,  R.;  Bernstein,   M.;  Hall,  R.; Garshick,  E.  (1975)  Assessment  of



ethylene dichloride as a potential air pollution  problem,  v.  III.  Bedford, MA:




GCA Corp. Report no. GCA-TR-75-32-G(3);  25 p.
                                     10-33

-------
Pavlova, I.V.; Rosenberg,  P.A.;  Byalko,  N.K.;  Gel Ton, N.A. (1965)  Biochemical



changes   in  the   blood   during  acute   intoxications   by  some  chlorinated




hydrocarbons.   In:   Letavet,   A.A.,  ed.   Professional'nye   zabolevaniia   v



tchimicheskoi  promyshlennosti  [Occupational diseases in the chemical industry];




pp. 217-224.








Pearson, C.R.;  McConnell,  G. (1975) Chlorinated  C   and  C~ hydrocarbons in the




environment.  Proc.  R. Soc. London B  189: 305-332.








PEDCo  Environmental,  Inc.  (1979)  Monitoring  of  ambient  levels  of  ethylene



dichloride  (EDO) in the vicinity of EDO production and  user  facilities. Research



Triangle   Park,   NC:  U.S.   Environmental  Protection  Agency,  Environmental



Monitoring  and  Support Lab.; EPA report no.  EPA/600-4-79-029.  Available from:




NTIS, Springfield,  VA; PB-298303.








Pellizzari,  E.D. (197t) Electron capture  detection in  gas chromatography.  J.




Chromatogr.  98:  323-361.








Pellizzari,  E.D. (1977a)  The measure  of carcinogenic  vapors in ambient atmos-



pheres. Research Triangle  Park,  NC:  Research Triangle  Institute; EPA report no.



EPA 600/7-77-055. Available  from: NTIS, Springfield, VA; PB-269582.








Pellizzari,  E.D. (1977b) Analysis of organic air pollutants by gas chromatograph



and mass spectroscopy. Research  Triangle Park,  NC:  Research Triangle Institute;



EPA report  no. EPA  600/2-77-100.
                                      10-34

-------
Pellizzari, E.D.  (1978)  Measurement of carcinogenic  vapors in  ambient atmos-



pheres.  Research Triangle  Park,  NC:  U.S.   Environmental  Protection  Agency,



Environmental Sciences Research Lab.; EPA report no. EPA-600/7-78-062. Available



from: NTIS, Springfield,  VA; PB-283023-







Pellizzari, E.D. (1978a)  Preliminary assessment of  halogenated organic compounds



in man and environmental  media. Monthly technical progress report no. 2  (January



1-30,  1978).  Contract  no.  68-01-1(731.   Research  Triangle  Park,  NC:  U.S.



Environmental Protection Agency.







Pellizzari, E.D. (1978b)  Preliminary assessment of  halogenated organic compounds



in man and environmental  media. Monthly technical progress report  no.  5 (April  1-



30,   1978).   Contract  no.   68-01-1731.  Research Triangle   Park,  NC:  U.S.



Environmental Protection Agency.







Pellizzari, E.D. (1978c)  Preliminary assessment of  halogenated organic compounds



in  man  and  environmental  media.  Monthly   technical  progress  report   no.   10



(September  1978). Contract  No.  68-01-4731.  Research  Triangle  Park,  NC:  U.S.



Environmental Protection Agency.







Pellizzari, E.D.  (1978d)  Preliminary assessment of  halogenated organic compounds



in man and environmental  media. Monthly technical progress report  No.  11  (October



1978). Contract No.  68-01-1731.  Research  Triangle  Park, NC: U.S. Environmental



Protection Agency.
                                      10-35

-------
Pellizzari,  E.D.   (1978e)  Quantification   of   chlorinated   hydrocarbons  in



previously collected air samples. Research Triangle Park,  NC: U.S. Environmental



Protection Agency, Office  of Air  Quality  Planning  and Standards; EPA report no.



EPA-H50/3-78-112. Available from: NTIS, Springfield, VA; PB289804.








Pellizzari,  E.D.  (1979a)  Information on the Characteristics of ambient organic




vapors in areas of high chemical production. Research Triangle Park, NC: Research




Triangle Institute.








Pellizzari,  E.D.  (1979b)  Organic  screening  in  Lake  Charles,  LA,   using gas



chromatography  mass  spectrometry computer techniques,  EPA  contract  no.  68-02-



271**, Research  Triangle Park,  NC: Research Triangle Institute.








Pellizzari,  E.D.;  Bunch,  J.E.  (1979) Ambient air carcinogenic  vapors: improved



sampling   and   analytical   techniques.   Research  Triangle   Park,   NC:   U.S.



Environmental  Protection  Agency,  Environmental  Sciences  Research   Lab.;  EPA



report no. EPA-600/2-79-081. Available  from:  NTIS, Springfield, VA; PB-297932.








Pellizzari,  E.D.; Erickson,  M.D.;   Zweidinger,  R.A.  (1979)  Formulation  of  a



preliminary  assessment  of  halogenated organic compounds in man  and environmental



media. Washington,  DC: U.S.  Environmental Protection  Agency,   Office of Toxic



Substances,  EPA report  no. 560/13-79-006. Available from:  NTIS,  Springfield, VA;




PB 80-112170.








Perocco,  P.; Prodi, G. (1981) DNA  damage  by haloalkanes in human lymphocytes




cultured in  vitro. Cancer  Lett. 13:  213-218.
                                      10-36

-------
Pervier, J.W.; Barley, R.C.;  Field,  D.E.;  Friedman, B.M.; Morris,  R.B.  (1971)



Survey reports on atmospheric emissions from the petrochemical industry,  v. II.



Marcus  Hook,  PA:   Air   Products   and  Chemicals,   Inc.;   EPA   report   no.



EPA/450-73-005b.  Available from:  NTIS,  Springfield,  VA; PB-244958; p. 135.








Piet, G.J.;  Slingerland,  P.; de  Grund, F.E.;  v.d. Heuvel,  M.P.M. ;  Zoeteman,



B.C.J.  (1978)  Determination  of  very volatile halogenated organic compounds in



water by means of direct head-space analysis. Anal.  Lett. 11:
Plotnick,  H.B.;  Weigel,  W.W. ;  Richards,  D.E. ;   Cheever  K.L. ;  Kommirreni,  C.



(1980) Dietary disulfiram enhancement of the toxicity of ethylene dibromie. In:



Ames, B.N.; Infante, P.; Reitz,  R. ,  eda. Ethylene  dichloride: a potential health



risk?  Cold Spring  Harbor,  NY:  Cold  Spring  Harbor Laboratory;  pp. 279-286.



(Banbury report no. 5).








Price, K.; Waggy, G.;  Conway, R.  (1974)  Brine shrimp bioassay and seawater BOD



(biochemical oxygen demand)  of petrochemicals.  J.  Water Pollut. Control Fed. 46:




63-77.








Rannug, U. (1976) Mutagenic studies of EDC tars as a model for tests of complex



mixtures of chemicals  in  the environment.  In:  Organiska Miljoegifter i Vatten,



12th Nordiska Symposiet om Vattenforskning. Publ. Nordforsk Miljoevardssekr. :2.








Rannug, U. ; Ramel, C.  (1977) Mutagenicity of waste products from vinyl chloride



industries. J. Toxicol. Environ. Health 2: 1019-1029.
                                     10-37

-------
Rannug,   U.;   Sundvall,  A.;   Rarael,   C.  (1978)  The   mutagenic   effect   of



1,2-dichloroethane on Salmonella  typhimurium. I. Activation through conjugation




with glutathione jLn vitro. Chera.  Biol.  Interact. 20:  1-16.







Rannug,  U.;  Beiji,  B.   (1979)  The mutagenic  effect  of  1,2-dichloroethane  on




Salmonella typhimurium.  II. Activation by the isolated perfused rat liver. Chen.




Biol. Interact. 24: 265-285.








Rannug,    U.    (1980a)   Genotoxic    effects   of    1,2-dibromoethane    and




1,2-dichloroethane.  Mutat. Res.  76:  269-295.








Rannug,  U. (1980b) The  use of different metabolizing  systems in the elucidation



of the mutagenic  effects of ethylene dichloride in Salmonella.  In:  Ames,  B.N.;



Infante,  P.; Reitz, R.,  eds. Ethylene dichloride: a potential health risk? Cold



Spring Harbor, NY: Cold Spring  Laboratory;  pp.  83-92. (Banbury report no. 5K








Rao,  K.S.; Murray, J.S.;  Deacon, M.M.;  John,  J.A.;  Calhoun  L.L.;  Young,  J.T.



(1980)  Teratogenicity  and  reproductive  studies  in  animals  inhaling  ethylene



dichloride.  In: Ames, B.N.; Infante,  P.;  Reitz, R., eds.  Ethylene dichloride: a



potential health risk? Cold Spring Harbor, NY: Cold Spring Harbor Laboratory, pp.




149-166.  (Banbury  report no. 5).








Reichert,    D.;    Henschler,    D.   (1978)    Uptake   and  hepatotoxicity   of



1,1-dichloroethylene  by the  isolated blood-perfused  rat  liver.  Int.  Arch.




Occup. Environ. Health  41:  169-178.
                                      10-38

-------
Reinfried, H.  (1958) On  lethal poisonings due to ingestion of 1,2-dichloroethane



containing rubbing compounds. Dtsch. Gesundheitswes. 13=  778-779.








Reitz, R.H.; Fox,  T.R.;  Domoradzki,  J.Y.;  Quast,  J.F.;  Langvardt,  P.; Watanabe,



P.O.  (1980)  Pharmacokinetic  and   macromolecular  interactions   of  ethylene



dichloride:  Comparison  of  oral  and  inhalation  exposures.  In:  Ames,  B.N.;



Infante, P.; Reitz, R.,  eds. Ethylene dichloride: a potential health risk? Cold



Spring Harbor, NY: Cold Spring Harbor  Laboratory,  pp.  135-148.  (Banbury report




no. 5).








Reitz, R.H.;  Fox,  T.R.;  Ramsey,  J.C.;  Quast, J.F.;  Langvardt,  P.W.; Watanabe,



P.O.  (1982)  Pharmacokinetics  and  micromolecular  interactions  of  ethylene



dichloride in rats after inhalation or gavage. Toxicol.  Appl.  Pharmaeol. 62: 190-



204.








Rejsek, K.;  Rejskova,  M.  (1947) Intoxication with symmetrical  dichloroethane.




Cas. Lek. Cesk.  86: 207-209.








Rohmann,  E.;  Zinn, D.;  Kulz,  J.  (1976) Electroencephalographic  observations in



childhood  poisonings  and their  therapeutic consequences.  Kinderaerztl.  Prax.




37: 209-216.








Rosenbaum,  N. (1939)  Use  of dichloroethane  in industry  from the standpoint of



occupational hygiene, in [dichloroethane]. Moscow, Chap. IV, pp. 109-113*








Rosenbaum,  N.D.  (1947) Ethylene dichloride as an industrial poison. Gig. Sanit.



12(2):  17-21.
                                     10-39

-------
Roubal, J. (1947) Two fatal cases of intoxication with symmetric dichloroethane




ingestion. Cas. Lek. Cesk. 86: 203-206.








Salmon,  A.G.;  Mackrodt,  W.C.;  Jones,  R.B.;  Basu,  S.K.;  Ashby,  J.  (1978)  An




investigation  of  the dichlorination of a series  of haloalkanes  by  rat  liver




microsomes and its possible role in the expression of toxic phenomenon. Toxicol.




Appl. Pharmacol. 45: Abst. 253, 277.








Salvini,  M.; Mazzucchelli,  B. (1958)  Intoxication  from  fumes  of  carbon tetra-



chloride  and dichloroethane used as  fumigant parasiticides. Minerva  Med.  49:



2295-2304.








Sato,  A.;  Nakajima,  T.  (1979)  A  structure-activity  relationship  of  some



chlorinated hydrocarbons. Arch. Environ. Health.  34:  69-75.








Sato, A.; Nakajima, T.; Koyama, Y. (1980) Effects of chronic ethanol consumption



on hepatic metabolism  of aromatic and  chlorinated hydrocarbons in rats.  Br.  J.




Ind. Med. 37: 382-386.








Sato, A.; Nakajima, T.;  fCoyama, Y.  (1981) Dose-related effects of a single dose



of  ethanol  on  the  metabolism in  rat   liver  of  some aromatic  and  chlorinated



hydrocarbons. Toxicol.  Appl.  Pharmacol.  60: 8-15.








Schlachter, M.M.;  Crawford,  A.A.;  John, J.A.;  et  al.  (1979)  The  effects  of



inhaled  ethylene  dichloride  on  embryonal  and  fetal  development in  rats  and




rabbits,  [unpublished  study].  Midland, MI:  Toxicology Research Laboratory,  Dow



Chemical  U.S.A.
                                      10-40

-------
Schoborn, H.; Prellwitz, W.; Baura,  P.  (1970)  Consumption coagulation pathology



of 1,2-dichloroethane poisoning. Klin. Wochenschr. 18: 822-824.








Schwartz, W.A.;  Higgins,  F.G.,  Jr.;  Lee,  J.A.; Newirth, R. ; Pervier, J.W. (1974)



Engineering  and cost  study of  air  pollution  control  for the  petrochemical



industry, v.  3.  Ethylene  dichloride  manufacture by oxychlorination. Marcus Hook,



PA:  Air  Products  and  Chemicals,   Inc.;  EPA  report  no.  EPA-450/3-73-006c.



Available from:  NTIS, Springfield, VA; PB-240492.








Secchi,  G.C.;   Chiappino,  G.;   Lotto. A.;  Zurlo,  N.  (1968)  Actual  chemical




composition of the "commercial  trieline"  and their hepatotoxic effect - Clinical



and enzymological studies.  Med. Lav. 59:  486-497.








Seufert, F.B.; Brown, P.; Oatway,  J.A.; Bernstein, M.; Ostrowski,  W.; Home, R.



(1980) 1,2-Dichloroethane:  technical control options analysis.  Bedford,  MA: CGA



Corp., EPA contract no. 68-01-5960.








Shackelford,  W.M.; Keith, L.H.  (1976) Frequency of organic compounds identified



in water.  Athens,  GA:  U.S.   Environmental  Protection  Agency;  Environmental



Research Laboratory,  EPA report  no.  EPA-600/4-76-062.  Available  from:  NTIS,



Springfield,  VA; PB-265470.  p.  134.








Shakarnis,  V.F.  (1969)  Induction  of X-chromosome nondisjunction  and recessive




sex-linked recessive lethal mutations  in females  of  Drosophila melanogaster by



1,2-dichloroethane. Sov.  Genet. Engl. Transl.  5:  1666-1671.
                                     10-41

-------
Shakarnis,  V.F.  (1970)  Effect of  1,2-dichloroethane.  Vesnik  Leningradskuniv-




siteta Seviya Biologii  25:  153-156.








Shamel,  R.E.;  O'Neill,  J.K.;  Williams, R.  (1975)  Preliminary  economic  impact




assessment  of  possible  regulatory  action  to control  atmospheric  emissions  of




selected  halocarbons.  Cambridge,  MS:  Arthur D.  Little,  Inc.,  EPA  report  no.



EPA-450/3-75-073.  Available from:  NTIS,  Springfield, VA; PB-247115.








Shchepotin,  B.M.;  Bondarenko, Ya.  D.  (1978) Clinical syndromes and pathogenic



treatment principles  of dichloroethane  intoxications.  Vrach. Delo 7: 134-139.








Siegel,  I.M. (1947) Ethylene dichloride [letter]. J. Am. Med. Assoc. 133: 577.








Simmon,  V.F.  (1980)  Review  of nonbacterial  tests of the genotoxic activity of



ethylene dichloride.  In:  Ames,  B.N.;  Infante,  P.;  Reitz, R.,  eds.  Ethylene



dichloride: a potential health risk? Cold  Spring  Harbor, NY: Cold Spring Harbor




Laboratory; pp.  97-103. (Banbury report no.  5).








Singh,  H.B.  (1977) Atmospheric halocarbons. 1977. Evidence of favor of reduced



average  hydroxyl radical concentrations in  the troposphere. Geophy. Res. Lett.




H:  101-101.








Singh, H.B.; Salas, L.J.; Lillian, D.; Arnts, R.R.; Appleby, A.  (1977) Generation



of  accurate halocarbon  primary  standards  with  permeation tubes.  Environ. Sci.




Technol.  11: 511-513-








Singh,  H.B.  (1978) [Letter to the editor]. Atmos. Environ.  12:  1809.
                                      10-42

-------
Singh,  H.B.;  Salas,  L.J.;  Shigeishi,  H.;  Smith,  A.J.;  Scribner,   E.  (1979)



Atmospheric  distributions,   sources  and   sinks  of   selected   halocarbons,



hydrocarbons,   SFfi  and  N0?.  Research  Triangle  Park,  NC:  U.S.  Environmental



Protection Agency,  Environmental  Sciences Research  Lab.;  EPA report no.  EPA-




600/3-79-107.  Available from:  NTIS,  Springfield, VA;  PB80-1M2706.








Singh,  H.B.;  Salas,  L.J.;  Smith,   A.;  Shigeishi,   H.   (1980)  Atmospheric



measurements of selected  toxic  organic chemicals. Research Triangle  Park,  NC:



U.S. Environmental Protection Agency, Environmental Sciences Research Lab.; EPA



report no. EPA-600/3-80-072. Available from: NTIS, Springfield,  VA; PB80-198989.








Singh, H.B.; Salas, L.J.;  Smith, A.J.; Shigeishi,  H. (1981) Measurements of some



potentially hazardous organic chemicals in urban environments.  Atmos. Environ.



15: 601-612.








Singh, H.B.; Salas, L.J.; Stiles, R.E.  (1982)  Distribution of selected gaseous



organic mutagens and suspect carcinogens in ambient air. Environ.  Sci. Technol.



16: 872.








Singh,  H.B.;  Salas,  L.T.; Stiles,  R.E.  (1983a)  Selected  man-made  halogenated



chemicals in the air and oceanic environment. J. Geophys. Res. 88: 3675-3683.








Singh,  H.B.;  Salas,  L.J.; Stiles,  R.; Shigeishi,  H.  (19&3b)  Measurements of



hazardous organic chemicals in the  ambient atmosphere.  Research Triangle Park,



NC: U.S.  Environmental Protection Agency,  Environmental Sciences Research Lab.;




EPA  report  no.  EPA-600/3-83-002,   Available  from:  NTIS,  Springfield,  VA;




PB-83156935.
                                     10-43

-------
Sipes,  I.G.;  Gandolfi,  A.J.  (1980)  jn  vitro  comparative  bioactivation  of



aliphatic halogenated hydrocarbons.  Dev. Toxicol. Environ. Sci. 8: 501-506.








Sjoberg, B. (1952) Thermal decomposition of chlorinated hydrocarbons. Sven. Kern.




Tidskr. 64: 63.








Smirnova, N.A.; Granik,  H.P.  (1970)  On the remote effects of acute occupational



poisoning with  some  carbohydrates and their derivatives. Gig.  Tr. Prof. Zabol.



14: 50-51.








Smyth, H.F., Jr. (1956) In: Spector, W.S.,  ed.  Handbook of toxicology, v. 1.  PA:



W.B.  Saunders  Company.








Snapp, O.I.  (1958)  Trunk sprays  for control  of the peach tree borer.  J. Econ.



Entomol. 51(1): 557-558.








Snelson,  A.; Butler,   R.;  Jarke,  F.  (1978)  Study  of removal  processes  for



halogenated  air  pollutants.  Research  Triangle Park,  NC:   U.S.  Environmental



Protection  Agency,  Environmental   Sciences   Research  Lab.;  EPA  report  no.



EPA-600/3-78-058. Available from: NTIS, Springfield, VA; PB-284066.








Sopikov,   N.F.;  Gorshunova,   A.K.  (1979)   Investigation   of   the   uptake,



distribution, and excretion  of  ethylene  dichloride  in  rats.  Gig. Tr.  Prof.



Zabol. 4: 36-40.








Spence, J.W.; Hanst,  P.L. (1978) Oxidation of chlorinated ethanes.  J. Air Pollut.



Control Assoc. 28: 250-255.
                                      10-44

-------
Spencer, H.C.; Rowe, V.K.;  Adams,  E.M.;  McCollister, D.D.;  Irish,  D.D.  (195O




Vapor toxicity of ethylene  dichloride  determined by experiments  on  laboratory




animals. Ind. Hyg.  Occup. Med. H:  t82.








Spreafico,  P.;  Zuccato,  E.;  Marcuri,  F.;  et   al.   (1978)   Metabolism  of




1,2-dichloroethane  in experimental animals,  report  nos.  1 and 2  Hew  York,  NY:




Chemical Manufacturers Association.








Spreafico,  F. ;  Zuccato,  E.;  Marcuri,  F. ;   et  al.  (1979)  Distribution  and




metabolism of 1,2-dichloroethane  (EDO  in experimental animals.  New  York,  NY:




Chemical Manufacturers Association, report nos. 3 and M.








Spreafico, F.; Zuccato,  E.; Marcuri, F.;  Sironi, M.;  Paglialunga, S.; Madonna,



M.; Mussini, E. (I960)  Pharmacokineties of ethylene dichloride in  rats treated by



different routes and its long-term inhalatory toxicity. In: Ames, B.N.; Infante,




P.;  Reitz,  R.,  eds.  Ethylene dichloride: a  potential  health risk? Cold Spring




Harbor, NY: Cold Spring Harbor Laboratory; pp. 107-133- (Banbury report no. 5).








SRI  International.  (1979) Assessment of human exposures to ethylene dichloride.




Prepared by SRI International for Office of  Air Quality Planning and Standards,



U.S.  Environmental  Protection Agency,  Research Triangle  Park, KC, EPA contract




no.  68-02-2835, Task 17.  Final Report.








SRI  International.  (1983) 1983 Directory  of chemical producers: United States of




America. Menlo Park, CA: SRI  International;  pp. 584, 771, 9^9, 965, 583.
                                      10-15

-------
Stanford  Research  Institute.  (1981) National screening program for organics in




drinking  water.  EPA contract  no.  68-01-4666.  Boland,  PA: SRI International.








Stoelting,  R.K.; Eger,  E.I., (1969)  II.  Percutaneous loss of  nitrous oxide,




cyclopropane, ether, and haloethane in  man.  Anesthesiology 30: 278-283.








Stolzenberg, S.J.; Hine, C.H. (1980) Mutagenicity of 2- and 3-carbon halogenated




compounds in  the  Salmonella/ma mmalian-microsome test.  Environ.  Mutagen.  2:




59-66.








Storer, R.D.; Bank, T.A.; Conolly,  R.D.  (1982)  In vivo genotoxic effect of 1,2-




dichloroethane  in male  B6C3F1 mice.  Toxicologist  2:  129.








Storey, C.L.; Krik, L.D.; Mustakas, C.C.  (1972)  Fate of EDC-CCl^ (75:25) residues




during milling  and oil  extraction of soybeans.  J. Econ. Entomol. 65: 1126-1129-








Stover, C.L.; Kincannon, D.F. (1983) Biological treatability of specific organic




compounds found  in chemical industry wastewater. J.  Water. Pollut. Control. Fed.




55: 97-109.








Stucki, G.; Brunner, W.; Staub,  D.;  Leisinger, T.  (1981) Microbial Degradation of




Chlorinated C1  and C2  Hydrocarbons. In: Leisinger,  T.; Cook,  A.M.;  Hutter, R.;



Nuesch,   R.,   eds.   Microbial  degradation  of   xenobiotics  and  recalcitrant




compounds. London, United Kingdom:  Academic  Press; pp.  131-137.








Stucki,   G.;   Krebner,  U.;   Leisinger,   T.    (1983)  Bacterial   growth  on




1,2-dichloroethane. Experimentia  39:  1271-1273.
                                      10-46

-------
Stuhlert,  H.   (1919)  Fatal  poisoning  from  ethylene  chloride.  Dtsch.  Med.




Wochenschr. 71: 1512-1513.







Suveev, I.M.;  Babichenko, M.E.  (1969)  Clinical picture and  treatment  of acute




poisoning with dichloroethane vapors.  Gig. Tr. Prof. Zabol. 13: 50-51.








Sykes, J.F.; Klein, A.K.  (1957) Chloro-organic  residues  in milk of cows orally



administered ethylene dichloride.  J.  Assoc. Off. Agric. Chem. 10: 203-209.








Sykes, J.F.; Klein, A.K. (1957)  Participating chemists. Chloro-organic residues



in  milk  of cows  orally administered  ethylene  dichloride. Assoc.  Off.  Agric.




Chera. 10: 206-209.








Sykes, J.F.; Klein, A.K.  (1957) Fumigant  residue-in-milk study. Chloro-organic



residues in milk  of cows orally administered ethylene dichloride.  Assoc. Off.



Agric. Chem. 10: 203-206.








Symons,  J.M.;  Bellar,  T.A.;  Carswel,  J.K.;  et al.  (1975)  National  organics



reconnaissance survey for halogenated  organics. J. Am. Water Works Assoc. 67:




631-617.








Tan,  E.L.;  Hsie,  A.W.  (1981) Mutagenicity and cytotoxicity  of haloethanes as



studied in the CHO/HCPRT system. Mutat. Res. 90: 183-191.








Tabak,  H.H.;  Quave,  S.A.;  Mashni, C.I.;  Barth,   E.F.  (1981) Biodegradability



studies with organic priority pollutant  compounds.  J. Water Pollut. Control Fed.




53:  1503-1518.
                                     10-17

-------
Theiss,  J.;  Stoner,  G.;  Schimkin,  M.;   Weisburger,   E.L.   (5977)  Test  for



carcinogenicity  of  organic  contaminants  of United  States drinking water  by



pulmonary tumor response  in strain  A  mice.  Cancer Res. 37: 2717-2720.








Troisi,  P.M.;  Cavallazzi,  D.   (1961)  Fatal  poisoning  from the  inhalation  of




dichloroethane fumes. Med. Lav.  52:  612-618.








Tsuruta, H.  (1975)  Percutaneous absorption of organic solvents. 1. Comparative



study of the  in  vivo percutaneous  absorption of chlorinated  solvents  in mice.




Ind. Health. 13: 27-236.








Tsuruta, H. (1977)  Percutaneous  absorption  of organic solvents. 2. A method for



measuring the penetration rate of chlorinated solvents through  excised rat skin.




Ind. Health. 15: 3-4, 131-139.








Urusova, T.P. (1953) The  possible presence  of dichloroethane in human milk with



exposure in industrial conditions.  Gig.  Sanit.  18:  36-37.








U.S. Environmental  Protection Agency, (n.d.) Determination of population density



around ethylene dichloride production  facilities. EPA contract no. 68-02-2836.








U.S.  Environmental Protection  Agency.  (1977)  National  organics  monitoring



survey. Office of Water Supply,  as  cited in Letltiewi.cz et  al.,  1982.








U.S.  Environmental  Protection  Agency.  (1979)  Health assessment  document  for



1,2-dichloroethane  (ethylene dichloride).  Draft  report.  Research Triangle Park,



NC: Environmental Criteria and  Assessment  Office.
                                      10-43

-------
U.S. Environmental Protection Agency.  (1979a) Population exposure to EDC [draft



report]. Contract no. 68-02-2835.  Menlo Park, CA: SRI International.








U.S. Environmental Protection  Agency.  (1981) The determination  of halogenated



chemical indicators  of industrial contamination  in  water  by  the purge-and-trap



method, method  502.1. Cincinnati,  OK: U.S.  Environmental Protection  Agency,



Environmental Monitoring and  Support  Laboratory. EPA report no.  EPA 600/4-81-




059.








U.S. Environmental  Protection Agency.  (198la)  Community water  supply  survey.



Washington, DC:  U.S.  Environmental Protection Agency, Office of Drinking Water.








U.S. Environmental Protection  Agency.  (198lb) Draft  criteria document for 1,2-



dichloroethane.  Washington,  DC: U.S. Environmental Protection Agency, Office of



Drinking Water.








U.S. Environmental Protection Agency.  (1982a) Groundwater supply survey. (Cited



in Letkiewicz et al., 1982.)








U.S. Environmental Protection Agency.  (1982b) Test methods for  evaluating solid



waste:  physical/chemical  methods.  Washington, DC: U.S. Environmental Protection



Agency, Office of Solid  Waste and Emergency Responses, EPA  report no.  SW-846.



Available from:  NTIS, Springfield, VA; PB82-172156.








U.S. Environmental Protection  Agency. (1984, November  23)  Proposed guidelines



for carcinogen risk assessment. Federal Register 19: 46294-46301.
                                     10-49

-------
U.S. International  Trade Commission.  (1975-1983)  Synthetic organic chemicals.




U.S.  Production and Sales, U.S.  International Trade  Commission.








U.S. International  Trade Commission.  (1977) Synthetic organic chemicals. USITC




publication no. 20.








U.S.   Tariff  Commission.   (1952-1974)  Synthetic   organic   chemicals.  U.S.




Production and Sales. United States Tariff  Commission.








Valvani,   S.C.;   Yalkowsky,   S.H.;    Roseman,   T.J.    (1981)   Solubility   and




partitioning. IV. Aqueous solubility  and octanol-water partition coefficients of




liquid nonelectrolytes.  J. Pharra.  Sci.  70:  502-507.








Van  Duuren,  B.;  Goldschmidt,   B.;  Loewengart,   G.;  Smith,  A.;  Mechionne,  S.;




Seldman,  I.;  Roth,  D.   (1979)  Carcinogenicity of  halogenated olefinic  and




aliphatic hydrocarbons in mice.  J. Natl.  Cancer  Inst.   63:  11«33-11»39.








Van  Dyke,  R.; Wineman,  C.  (1971) Enzymatic  dechlorination.  Dechlorination of




chloroethane and propanes in vitro. Biochem.  Pharmacol. 20: 463-^70.








Vesar, Inc.  (1975)  Identification of  organic  compounds  in effluents from indus-




trial sources. Springfield,  VA:  U.S.  Environmental  Protection Agency,  General




Technologies  Division,  EPA report no. EPA-560/3-75-002.  Available  from: NTIS,




Springfield, VA PB-2U161M.
                                      10-50

-------
Vincent, L.E.; Lindgren, D.L. (1965) Influence of  fumigation  and  age on carbon



dioxide  production  of  some  stored-product  insects.  J.   Econ.  Entornol.  58:




660-664.







Von'  Oettinger,  W.F.  (1961)  The halogenated  hydrocarbons  of industrial  and




toxicological importance.  Amsterdam, The Netherlands: Elsevier.








Vozovaya,  M.  (1971) Changes  in  the esterous  cycle of white  rats  chronically



exposed  to the combined  action  of gasoline  and dichlorethane vapors.  Akush.




Ginekol. (Moscow) 17: 65-66.








Vozovaya,  M.  (1975)  The effect of  low  concentrations of benzene dichloroethane



alone and  their combination on the  reproductive  function  of animals and on the




development of progeny.  Gig. Tr.  Prof.  Zabol. 7: 20-23-








Vozovaya,  M.  (1976)  Effect  of low  concentrations of benzene and dichloroethane



separately and combined on the reproductive function of animals. Gig. Sanit. 6:




100-102.








Vozovaya,  M.  (1977) The  effect of  dichloroethane  on  the  sexual  cycle  and




embryogenesis of experimental animals 1977. Akush. Ginekol. (Moscow) 2: 57-59.








Wadhi,  S.R.; Scares, T.  (1961)  Comparative toxicity of some fumigants to the red



scale,  Aonidrella aurantii  (Maskell) (Hemiptera: coccoidea). Indian  J. Entomol.



27: 85-88.
                                      10-51

-------
Wagner, J.C. (1975) Fundamentals of clinical pharmacokinetics.  1st ed. Hamilton,




IL:  Drug Intelligence  Publications,  Inc.








Wallace, L.  (1981) Measurements  of volatile organic  compounds  in breathing zone




air, drinking water and exhaled breath (preliminary draft).  (Cited in Brodzinskjy




and Singh,  1982.)








Ward, J. (1980) The carcinogenicity of ethylene dichloride in Osborn-Mendel rats



and  B6C3F1  mice.  In:  Ames,  B.N.;  Infante,  P.;  Reitz,  R.,  eds.  Ethylene



dichloride:  a potential health risk?  Cold  Spring  Harbor,  NY: Cold Spring Harbor




Laboratory;  pp. 35-54.  (Banbury  report  no.  5).








Watrous, R.M. (1947)  Health hazards of  the pharmaceutical industry. Br. J. Ind.




Med. 4:  111-125.








Weast,  R.C.  (1980) Handbook of Chemistry and Physics, 61st ed. Boca Raton, PL:




CRC Press Inc.; p. C-309.








Weisburger,  E.  (1977) Carcinogenicity studies  on halogenated  hydrocarbons. EHP




Environ. Health Perspect.  21: 7-16.








Weiss,  F. (1957)  Lethal  oral  poisoning from dichloroethane.  Arch. Gewerbepathol.




Gewerbehyg.  15: 253-264.








Wendel,  H.  (1948)  Lethal  poisoning  from  dichloroethane (ethylene chloride).




Pharmazie  3: 398-400.
                                      10-52

-------
Whitney, W. (1961)  Fumigation hazards as related to the physical, chemical, and



biological properties of fumigants. Pest Control 29: 16-21.








Whittemore, A. (1978) Quantitative theories of  carcinogenesis.  In:  Advances in




cancer research,  v. 27.  New York, NY: Academic Press; pp.  55-58.








Whittemore, A.; Keller,  B.  (1978)  Quantitative theory of  carcinogenesis.  SI AM



Rev. 20: 1-30.








Williams, R.T. (1959) Detoxication mechanisms. London, United Kingdom, p. 55.








Wilson, J.T.;  Enfield,  C.G.;  Dunlap, W.J.; Cosby,  R.L.; Poster, D.A.; Baskin,



L.B. (1981) Transport and  fate of  selected organic pollutants in a sandy soil.



J. Environ. Qual.  10: 501-506.








Wirtschafter,  Z.T.;  Schwartz, E.D.  (1939)  Acute ethylene  dichloride poisoning.




J. Ind. Hyg. Toxicol. 21: 126-131.








Withey, J.R.;  Collins,  B.T.  (1980)  Chlorinated aliphatic  hydrocarbons used in



the food industry:  the comparative pharmacokinetics of methylene chloride,  1,2-



dichloroethane, chloroform and  trichloroethylene after i.v.  administration in



the rat. J. Environ. Pathol.  Toxicol. 3: 313-332.








Withey,  J.R.;  Collins,   B.T.;  Collins,  P.G.  (1982)  Effect  of  vehicle  on  the



pharmacokinetics and  uptake  of  four halogenated hydrocarbons  from  the gastro-



intestinal  tract  of the  rat.  Pre-print paper  submitted  to  J.  Appl. Toxicol.



December, 1982.
                                     10-53

-------
Woodward, G.; Lange, S. ; Nelson, K.; Calvery, H. (1981) The acute oral toxicity



of acetic, chloroacetic, dichloroacetic and trichloroacetic acids. J. Ind.  Hyg.



Toxicol. 23: 78-82.






Yasoda,   S.K.;   Dan   Loughran,   E.   (1977)    Air   sampling   methods   for



S-tetrachloroethane  and other related chlorinated  hydrocarbons.  J.  Chromatogr.



137-283-292.






Yllner,  S.  (1971a)  Metabolism  of  chloroacetate-1-1 C  in  the mouse.  Acta



Pharmacol.  Toxicol.  30: 69-80.
                                                        It
Yllner,  S.  (1971b)  Metabolism of  1,2-dichloroethane- C  in the  mouse.  Acta



Pharmacol.  Toxicol.  30:  257-265.
 Yllner,  S.  (n.d.) Chlorinated aliphatic hydrocarbons used in the  foods industry.



 The  comparative  pharmacokinetics  of methylene  chloride,   1,2-dichloroethane,



 chloroform, and  trichloroethylene  after  i.v.  administration  in  the  rat.  J.



 Environ. Pathol.  Toxicol.  3: in press.






 Yodaiken,  R.E.;   Babcock,  J.R.  (1973)   1,2-Dichloroethane  poisoning.   Arch.



 Environ. Health.  26: 281-281.
                                      10-54

-------