United States
              Environmental Protection
              Agency
              Office of Health and
              Environmental Assessment
              Washington DC 20460
EPA-600/8-84-006A
April 1984
External Review Draft
              Research and Development
r/EPA
Health Assessment   Review
Document for           Draft
1 ,2-Dichloroethane
ir-j.l_  I      l-k-  I- 1   -.1  »
(Ethylene Dichlonde)

Part 1 of 2
                                             Cite or Quote)
                           NOTICE
              This document is a preliminary draft. It has not been formally
              released by EPA and should not at this stage be construed to
              represent Agency policy. It is being circulated for comment on its
              technical accuracy and policy implications.

-------
/n M  +                                   EPA-600/8-84-006A
(Do Not                                                A  .. -00/l
Cite or Quote)                                          April 1984
                                           External Review Draft
        Health Assessment Document
           for  1,2,- Dichloroethane
               (Ethylene Dichloride)

                       Part 1 of 2
                            NOTICE
 This document is a preliminary draft. It has not been formally released by the U.S. Environmental
 Protection Agency and should not at this stage be construed to represent Agency policy. It
 is being circulated for comment on its technical accuracy and policy implications.
                                        U.S. Environmental Protection Agency
                                        Region V, Library
                                        230 South Dearborn Street
                                        Chicago, Illinois 60604
               U.S. ENVIRONMENTAL PROTECTION AGENCY
                  Office of Research and Development
               Office of Health and Environmental Assessment
               Environmental Criteria and Assessment Office
               Research Triangle Park, North Carolina 27711

-------
                                         DISCLAIMER




           This  report is an  external draft for  review purposes only  and does  not




      constitute  Agency Policy.  Mention  of trade names or commercial  products  does




      not  constitute endorsement  or  recommendation for  use.
U,S. Environmental Protection Agency

-------
                                    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  are placed in perspective  with observed




environmental levels.
                                      111

-------
     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.  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

      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  (*).
                                       IV

-------
             Roy Albert, M.D. (Chairman)
             Elizabeth L. Anderson, Ph.D.
             Larry D. Anderson,  Ph.D.
             Steven Bayard, Ph.D.
             David L. Bayliss, M.S.
             Chao W. Chen, Ph.D.*
             Margaret M.L. Chu,  Ph.D.
             Herman J. Gibb, M.S., M.P.H.
             Bernard H. Haberman, D.V.M., M.S.
             Charalingayya B. Hiremath, Ph.D.*
             Robert McGaughy, Ph.D.
             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.


             John R. Fowle III,  Ph.D.*
             Ernest R. Jackson,  M.S.
             Casey Jason, M.D.,  Medical Officer
             David Jacobson-Kram, Ph.D.
             K.S. Lavappa, Ph.D.
             Sheila L. Rosenthal, Ph.D.
             Carol N. Sakai, Ph.D.*
             Carmella Tellone, B.S.
             Vicki L. Vaughan-Dellarco, Ph.D.
             Peter E. Voytek, Ph.D. (Director)

     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 Batriek   »  • ././;, ^ t->   >.      .-.:•-•'  •••  '
             Office of Air, Noise and Radiation
             Off ice,.of Air^Quajli&yj P.ljanni^ana-Statfdairds1-
             Research  Triangle Park, NC

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

             Jerry F.  Stara, D.V.M.       ;  '   " "<
             Office of Health and Environmental Assessment
             Environmental Criteria and A@se^sment:' Off i'ce
             Cincinnati, OH
Consultants, reviewers and contribating .authors:

             Joseph-F..Borzellecai Ph»-Du -
             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               IV.  .•"-'

             I.W.F. Davidson, Ph.D.
             Bowman Gray S@ho01*of, Medicine
             Wake Forest University
             Winston Salem, NC             -"  . "
                                       vi

-------
Larry Fishbein, Ph.D.
National Center for lexicological 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

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

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

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

Jean C. Parker, Ph.D.
Office of Waste Programs Enforcement
U.S. Environmental Protection Agency
Washington, DC
                         vn

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

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

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

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

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

Robert Tardiff, Ph.D.
1423 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

Jim Withey, Ph.D.
Department of National Health and Welfare
Tunney's Pasture
Ottawa, Ontario
CANADA K1A 01Z
                         Vlll

-------
                               TABLE OF CONTENTS


                                                                           Page

LIST OF TABLES [[[      xi

LIST OF FIGURES [[[     xvi

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

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
     4 . 1   SAMPLING [[[    4-1
     4 . 2   ANALYSIS [[[    4-1

          4.2.1   Ethylene Dichloride in Air ............................    4-1
          4.2.2   Ethylene Dichloride in Water ..........................    4-2
          4.2.3   Ethylene Dichloride in Solid Samples ..................    4-3
          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

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	   9-129

     9 .4  MUTAGENICITY	   9-131
     9.5  CARCINOGENICITY	   9-166

        9.5.1   Animal  Studies	   9-166
        9.5.2   Epidemiologic  Studies	   9-205
        9.5.3   Quantitative Estimates	   9-206
        9.5.4   Summary	   9-233
        9.5.5   Conclusions	   9-234

10.   REFERENCES  	    10~1

 Appendix  A
 Appendix  B
 Appendix  C

-------
                                 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

5-4     Comsumption of Ethylene Dichloride in 1979 and 1974	     5-7

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

5-6     Estimated Environmental Releases of Ethylene
          Dichloride in 1979	    5-11

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
          System	    7-15

7-4     Reported Occurrence of Ethylene Dichloride in Surface
          Water Systems	    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	    7-21

7-8     Total Estimated Population (in Thousands) Exposed to
          Ethylene Dichloride in Drinking Water at the Indicated
          Concentration Ranges	    7-22

7-9     Estimated Drinking Water Intake of Ethylene Dichloride
          by Adults and Infants	    7-23

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

9-2     Partition Coefficients for Ethylene Dichloride	     9-4
                                      XI

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

Table                                                                      Page

9-3     Fate of 14C-EDC in Rats 48 Hours After Oral (150 mg/kg)
          or Inhalation (150 ppra,  6-hr) Exposure	    9-6

9-4     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 Orll with Doses of EDC in Oil
          and in Water as Vehicle and After Intravenous
          Administration	    9-9

9-6     EDC Tissue Levels After 50 ppra Inhalatory Exposure	   9-14

9-7     EDC Tissue Levels After 250 ppm Inhalatory 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
          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  (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
          2-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
                                       xii

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

Table

9-16    Total Macromolecular Binding and DNA Binding in Selected
          Tissue of Rats After Exposure to   C-EDC by Oral or
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
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 mg/rag [U- C]EDB 	
Effects Associated with Acute Lethal Oral Doses of
Ethylene Dichloride in Humans 	 ,
Effects of Acute Oral Ingestion of 1 ,2-Dichloroethane
(Survey Results ) 	 ,
Effect of Ethylene Dichloride Exposure on Eye
Sensitivity to Light 	 ,
Morbidity and Lost Workdays of Aircraft Industry Gluers
Exposed to Ethylene Dichloride 	 ,
Concentrations of Ethylene Dichloride in Oil Refinery
Mineral Oil Purification Process Air 	 	 	 ,
Effects Observed in Polish Oil Refinery Workers 	
Effects of Acute Exposure to Ethylene Dichloride 	
Effect of Ethylene Dichloride on the Cornea 	
Effect of Subchronic Exposure to Ethylene Dichloride 	
Summary of Mutagenicity Testing of EDC: Gene Mutations
in Bacteria 	
Summary of Mutagenicity Testing of EDC: Higher Plants 	
Summary of Mutagenicity Testing of EDC: Gene Mutation
Tests in Insects 	
Summary of Mutagenicity Testing of EDC: Mammalian Cells
in Culture 	 	
Summary of Mutagenicity Testing of EDC: Chromosomal
Aberrations Tests 	 	 	
Summary of Mutagenicity Testing of EDC: PolA Assay 	
Summary of Mutagenicity Testing of EDC: DNA Binding
y-D
-------
                                LIST OF TABLES

Table

9-34    Design Summary for 1,2-Dichloroethane (EDC) Gavage
          Experiment in Osborne-Mendel Rats	   9-168

9-35    Terminal Survival of Osborne-Mendel Rats Treated With
          1,2-Dichloroethane (EDC)	   9-172

9-36    Squamous Cell Carcinomas of the Forestomach in Osborne-
          Mendel Rats Treated With 1,2-Dichloroethane (EDC)	   9-173

9-37    Hemangiosarcomas in Osborne-Mendel Rats Treated With
          1,2-Dichloroethane (EDC)	   9-174

9-38    Adenocarcinomas of the Mammary Gland in Female Osborne-
          Mendel Rats Treated With 1,2-Dichloroethane (EDC)	   9-175

9-39    Design Summary for 1,2-Dichloroethane (EDC) Gavage
          Experiment in B6C3F1 Mice	   9-177

9-40    Terminal Survival of B6C3F1 Mice Treated With
          1,2-Dichloroethane (EDC)	   9-178

9-41    Hepatocellular Carcinomas in B6C3F1 Mice Treated With
          1,2-Dichloroethane (EDC)	   9-181

9-42    Alveolar/Bronchiolar Adenomas in B6C3F1 Mice Treated
          With 1,2-Dichloroethane (EDC)	   9-182

9-43    Squamous Cell Carcinomas of the Forestomach in B6C3F1 Mice
          Treated With 1,2-Dichloroethane  (EDC)	   9-183

9-44    Adenocarcinomas of the Mammary Gland in Female B6C3F1 Mice
          Treated With 1,2-Dichloroethane  (EDC)	   9-184

9-45    Endometrial Polyp or Endometrial Stromal Sarcomas  in Female
          B6C3F1 Mice Treated With 1,2-Dichloroethane (EDC)	   9-184

9-46    Characterization of 1,2-Dichloroethane  (EDC) Inhalation
          Experiment in Sprague-Dawley Rats	   9-185

9-47    Design Summary for 1,2-Dichloroethane (EDC) Experiment
          in Sprague-Dawley Rats	   9-186

9-48    Survival of Sprague-Dawley Rats Exposed to EDC at  52 and
          104 Weeks	   9-188

9-49    Tumor Incidence in Sprague-Dawley  Rats Exposed to  EDC	   9-189

9-50    Mammary Tumors in Sprague-Dawley Rats Exposed to EDC	   9-193
                                      xiv

-------
                                 LIST  OF  TABLES

Table                                                                      Page

9-51    Design Summary for 1,2-Dichloroethane  (EDC) Experiment

9-52
9-53
9-51

9-55

9-56

9-57

9-58
9-59
in Swiss Mice 	
Survival of Swiss Mice Exposed to EDC at 52 and 78 Weeks 	
Tumor Incidence in Swiss Mice Exposed to EDC 	
Pulmonary Tumor Response in Strain A/st Mice
Injected with EDC 	
Mouse Skin Bioassay of 1 ,2-Dichloroethane and
Chloroacetaldehyde 	 	 	
Incidence Rates of Hemangiosarcomas in the Circulatory
Systems of Male Osborne-Mendel Rats 	
Incidence Rates of Hepatocellular Carcinomas in Male
B6C3F1 Mice 	
Upper-Bound Estimate of Risk At 1 Mg/kg/day 	
Relative Carcinogenic Potencies Among 53 Chemicals
9-196
9-197
9-198

9-202

9-204

9-220

9-220
9-223

          Evaluated by the Carcinogen Assessment Group as Suspect
          Human Carcinogens '  '3	      9-230
                                      xv

-------
                                LIST OF FIGURES


Figure                                                                     Page

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

9-4     Levels of EDC in rats after inhalatory exposure to 50 ppm
          (top) and 250 ppm (bottom).  Levels were measured at
          termination of 5-hour exposure period	   9-27

9-5     Microsomal oxidative metabolism of 1,2-dihaloethanes	   9-39

9-6     Further metabolism of 2-chloroacetaldehyde and 1-chloroso-
          2-chloroethane from microsomal oxidation	   9-41

9-7     Cytosolic metabolism of ethylene dichloride	   9-44

9-8     Growth curves for male and female Osborne-Mendel rats
          administered 1,2-dichloroethane (EDC) by gavage	  9-170

9-9     Survival comparisons for male and female Osborne-Mendel rats
          administered 1,2-dichloroethane (EDC) by gavage	  9-171

9-10    Growth curves for male and female B6C3F1 mice administered
          1,2-dichloroethane (EDC) by gavage	  9-179

9-11    Survival comparisons for male and female B6C3F1 mice
          administered 1,2-Dichloroethane (EDC) by gavage	  9-180

9-12    Point and upper-bound estimates of four dose-response models
          over low-dose region on basis of hemangiosarcomas in rats;
          dose with surface correction	  9-224

9-13    Point and upper-bound estimates of four dose-response models
          over low-dose region on the basis of liver carcinomas in
          mice; dose with surface correction	  9-225

9-14    Histogram representing the frequency distribution of the
          potency indices of 53 suspect carcinogens evaluated by
          the Carcinogen Assessment Group	  9-229
                                      xvi

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



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




pleasant odor.   EDC  has a  molecular  weight of  98.96  amu,  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/Jl and a  log octanol/water partition  coefficient of 1.48.




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




or halogen  specific  (raicrocoulometric or electrolytic  conductivity)  detector.




Alternatively, a gas chroraatograph/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,  4.523  and  3.455 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  are 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  49^4 metric  tons were




released from dispersive uses  (Seufert et al.,  1980).




     Based on  available kinetic data  and average  tropospheric hydroxyl free




radical concentration, the half-life of  EDC, the most  likely  removal  mechanism




for EDC from the atmosphere,  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 reac-




tion 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




(Elfers,  1979).  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
                                      1-2

-------
variations  in exposure  levels.   Using  Federal  Reporting  Data System  data,
Letkiewicz et al.  (1982) have  projected  that  EDC  levels  in all groundwater and
surface water systems in the United States fall below 10 pg/&, and that most are
<1.0  pg/2,.   No data  are available  indicating the presence of  EDC  in finished
foods.
      Pharmacokinetic studies with animals indicate that EDC is rapidly 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.  Tissue distribution  of EDC  is  consistent with its
lipophilic  nature, and  the chemical  crosses  the blood/brain and  placental
barriers and distributes into  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, as well  as nonreactive  glutathione  conjugates.
Elimination of unmetabolized EDC occurs  almost  exclusively via the  lungs,  is
rapid and  is consistent  with   a two-compartment  system and  Michaelis-Menten
kinetics.  Significant bioaccumulation 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).
     Limited data, primarily from the foreign literature, suggest that typical
symptoms and  signs  of   acute  toxicity  may develop  in  humans  with  repeated
occupational exposure to higher concentrations  (>60 ppm)  of  EDC  vapor.   Subtle
                                      1-3

-------
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  HOO-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.  Additional  studies  are needed,  however, particularly with humans,



to conclusively establish that EDC is not a teratogen and does not cause adverse




reproductive effects.



      Positive  responses in  different  test  systems representing  a  wide  range of




organisms  indicate that EDC is  a  weak direct-acting  mutagen capable of causing




gene  mutations.   Several  of  its  putative  metabolites,  thought  to be formed in




rats and mice, are judged  to be  more  potent mutagens than EDC.  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 did




not  produce a  statistically  significant  increase  in  the  incidence  of  lung




adenomas in strain A mice when administered intraperitoneally.   However,  benign




lung tumors (but not skin tumors) were induced in mice when applied to the skin.




No  statistically significant  increases in  tumors  occurred  in  rats  or  mice




following  lifetime inhalation  exposure.    No  case  reports  or  epidemiologic




studies concerning EDC were avilable in the published literature for analysis.




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




carcinogenicity includes: 1)  positive  findings in one oral rat study and one oral




mouse study, and supportive evidence in  two other mouse studies;  and 2)  findings




that EDC is weakly mutagenic, and that certain metabolites show evidence of even




greater mutagenic potency.   By using  the International Agency for  Research on




Cancer (IARC) classification  scheme, the level of animal evidence, combined with




the nonexistence of human data,  would constitute sufficient animal evidence that




EDC  is  a probable human  carcinogen,  the  overall IARC ranking being Group 2B.




Using the animal data  and a  linear multistage extrapolation model,  the  upper-
bound
estimate of potency for EDC is q   = 7 x  10   mg/kg/day calculated on the
basis of hemangiosarcomas in rats.  The upper-bound estimate of the cancer risk




from ingesting water contaminated with  1  pg/il  of  EDC is 2 x 10" .   The potency




index for EDC lies in the fourth quartile among 53 suspect carcinogens evaluated




by the Carcinogen Assessment  Group.   These potency  and  risk values  are upper-




bound estimates;  that is to say  that  the true values, while not  identifiable,




would not likely exceed the upper-bound, and may be lower.
                                      1-5

-------
                                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  risk assess-




ments.  When appropriate,  the authors of the document have attempted to identify




gaps in current knowledge that limit risk evaluation capabilities.
                                      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).
3.4. 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/m£
     Viscosity:             1 .4451 m Pa S (cP)
     Flashpoint:
     Closed cup             1 7°C
     Open cup               21°C
     Explosive limits  in
     air at 25°C:           6.2-1 5.6$ by volume
                                      3-1

-------
Autoignition tempera-
ture in air:           H
Vapor pressure:         ^C     kPa      Torr
                       10      5.3      40
                       20      8.5      64
                       30     13.3     100

Solubility in water:    0.0891  M (Valvani et  al.,  1981)
                       8,820 mg/£
Log Octanol/Water
Partition Coefficient:  1.48  (Valvani  et al.,  1981)
Henry's Law Constant
     O    _•(                    _ll
atm nr rool  :          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




     Taking 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, 1983), 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 U.S. EPA (!982a) and solids sampling by U.S. EPA (I982b).




4.2. ANALYSIS




4.2.1.  Ethylene Dichloride 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.   An Ascarite trap may be inserted




before the chromatography column to remove  water.   Suitable columns include 10%




SP-1000 on Supelcoport (100/120 mesh, 15'  x  1/8"  stainless  steel)  and 0.2% CW-
                                      4-1

-------
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 rnB,/minute  on the  former column and 40 m£/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 adhered to.




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




gram Tenax to be 27 & (flow rate 5-600 mil/minute; ethylene dichloride cone. <250




mg/m ; 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 Water.   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 4C 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




180°C  and  backflushed with helium  (20-60 mS,/minute,  4 minutes)  to desorb the




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




chromatography  column  packed  with  "\%  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 ug/£ with a




relative standard deviation of <1 0$  must be used.  This method is similar to EPA



method 601,624  for use with wastewater (U.S.  EPA, 19820).




4.2.3.   Ethylene Bichloride in  Solid Samples.   Ethylene  dichloride  in  solid




samples may be  analyzed by  EPA  method 8010 "Halogenated Volatile Organics" (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 4 minutes with a gas flow of 20 to  60 mil/minute.   Gas chromotographic




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




4.2.4.   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 mi with prepurged, distilled water.  The mixture is placed in a 1 00 ml 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, 24 hrs) and conditioned (270°C,  30  raft/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 4.2.2.




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




analyzed by using an apparatus identical to the one described in Section 4.2.4,




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

-------
J4.2.6.   Ethylene  Bichloride in  Tissue.   Ethylene dichloride  in tissue can be




analyzed by using an apparatus identical to the one described in Section 4.2.4,




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



bath.  The purge time is reduced to 30 minutes.
                                      4-4

-------
                         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 dehydro-

halogenation 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 oxychlorina-

tion 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 1974, =58$ of the total EDC production capacity was based upon

direct chlorination  and -^2% was based upon  oxychlorination (Pervier  et  al,

197^).  Current capacities are judged to be roughly the same.

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

liquid-phase chlorination of ethylene as follows (Archer, 1979):
                                         FeCl
                CH9  =  CH,, +  C19  (5% air) - ^ - >  C1CH5CH5C1
                   *                                      d
Most liquid-phase processes  use  ferric chloride as the  catalyst.   The chlori-

nation is  carried out  at  J40-50°C with  5% air  added  to  prevent  substitution

chlorination of the product.

     The oxychlorination  process  is  usually incorporated into an  integrated

vinyl chloride plant in which hydrogen chloride (which is recovered from cracking
                                      5-1

-------
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 + Op	—> 2C1CH_CH,,C1 + HO
                                       270°C        d  d      d


5.2. ETHYLENE BICHLORIDE PRODUCERS

     The producers of EDC in  the U.S.  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 facili-

ties 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

(197-4) and the U.S.  International Trade Commission 0974) 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 (1974) 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

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
                                      5-2

-------
                                   TABLE 5-1
                  Major Manufacturers of Ethylene Dichloridec
Manufacture
Plant Sites
     Annual Capacity
(Millions of Metric Tons)
Atlantic Richfield (ARCO)
Borden, Inc.
Dow Chemical USA
E.I. duPont (Conoco)
Ethyl Corp.
Formosa Plastics Corp.
Georgia-Pacific Corp.
B.F. Goodrich Co.
PPG Industries
Shell Chem. Co.
Union Carbide Corp.
Vulcan Materials Co.

Port Arthur, TX
Geisrnar, LA
Freeport, TX
Oyster Creek, TX
Plaquemine, LA
Lake Charles, LA
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

0.200
0.230
0.725
0.500
0.840
0.525
0.320
0.110
0.240
0.385
0.735
0.110
0.720
0.450
0.360
1 .230
0.620
0.540
0.070
0.070
0.160
Total 9.140
Note:  Capacities are flexible depending on finishing capacities for
       vinyl chloride and chlorinated solvents.
 SRI, 1983; CMR, 1983
 Operated under a toll agreement with Diamond Shamrock Corp.
'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
                                                               O
               U.S.  Production and Sales of Ethylene Dichloride
Year
1982
1981
1980
1979
1978
1977
1976
1975
1974
1973
1972
1971
1970
1965
1960
1950
Millions
Production
3.^55
4.523
5.037
5.349
4.989
4.987
3.647
3-617
4.156
4.214
3.541
3.428
3-383
1 .113
0.575
0.138
of Metric Tons
Sales
0.640
0.401
0.510
0.633
0.469
0.692
0.61 7
0.345
0.596
0.613
0.656
0.595
0.596
0.140
0.198
0.021
aUSITC, 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

-------
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 4? per year




through 198? (CMR, 1983).




5.4. ETHYLENE DICHLORIDE USES




     A major portion (84$) 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




ethylenearnines 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  tetraethyllead  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 tetraalkyllead  compounds  is always in admixture  with EDC  and/or



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




1/2 mol of ethylene dibromide per  mol  of tetraethyllead  (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 demand for




lead in gasoline  will decrease at a rate of 11 %  per year (average) through 1986.
                                      5-5

-------
                                  TABLE  5-3
                          Ethylene Bichloride Uses'
Use
Vinyl chloride monomer
Chlorinated solvents
Vinylidene chloride
Exports
Lead scavenger
Amines
Miscellaneous

1983
81
H
2
9
—
—
lb
Percent of Total
1980 1977
81 80
7 10
2
5
3
—
2b ?c

1974
78
8
—
—
3
2
9C
 CMR, 197M,  1977,  1980,  1983
b.
 includes lead scavenger
c.
 includes exports
                                5-6

-------
                                   TABLE 5-4
              Consumption of Ethylene Bichloride in 1979 and 19743
Consumption (Thousands of Metric Tons)

Vinyl chloride monomer
1 ,1 , 1 -Trichloroethane
Trichloroethylene
Perchloroethylene
Vinylidene chloride
Ethyleneamines
Lead scavenger
Exports
Preparation of polysulfides
Paints, coatings and adhesives
Extraction solvents
Cleaning of fabric and polvinyl
chloride equipment
Grain fumigation
Other uses
Total
1979 %
4420 85.0
—
89.4 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
5199.44 99.8
1974
3894.2
153-3
132.8
124.7
97.0
131 .5
97.0
167.3
—
—
—
—
—
6.8
4804.4
%
81 .0
3.2
2.8
2.6
2.0
2.7
2.0
3.5
—
—
—
—
—
0.1
99.9
aSeufert et al.,  1980;  Blackford,  1974
                                  5-7

-------
                                                     TABLE 5-5
                              Users of Ethylene Bichloride for Intermediate Purposes
                                                                                                      Vinylidene
                         1 ,1 ,1-Trichloroethane  Trichloroethylene  Perchloroethylene  Ethyleneamines  Chloride
ui
do
Dow Chem. USA

     Freeport, TX
     Plaquemine, LA

PPG Industries

     Lake Charles, LA

Union Carbide Co.

     Taft, LA
     Texas City, TX

Diamond Shamrock Corp.

     Deer Park, TX

E.I. duPont

     Corpus Christi, TX
                                  X
                                  X
X
X
X
X
                                                                                                          X
                                                                                                          X
       SRI, 1983; Blackford, 1974
      X = produces this chemical from ethylene dichloride

-------
     Exports  of EDC  have  been  increasing  in recent  years (see  Table 5-6).




Exports may grow by as much as 8% per year through the mid-8Os (CMR, 1983).  The




Japanese are expected  to  become major  importers as they close down significant




amounts of mercury  cell chlor-alkali capacity because of environmental regula-




tions.   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  in grain;  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 predominantly enter




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




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




in end-point product applications.




     A number of estimates have been generated  which  predict the  amount of EDC




that is 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
                                      5-9

-------
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 1.5/6,  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,  I979a)  indicated  that total




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




storage  are  44,000 metric  tons or  -O.Q%  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.




     For 1974,  Patterson et  al.  (1975)  estimated  total  domestic  emissions at




74,000 metric tons.   Total  domestic  emissions  for 1973 were reported at  54,000




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




below.
                                      5-10

-------
                                   TABLE 5-6
        Estimated  Environmental  Releases  of Ethylene Bichloride in 1979

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 , 1 54 61
65 191

73
138
86
1 04
5
136
180

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

	
	
	
	
5
	
	

	
46
	
50
	
	
101
aSeufert et al.,  1980
                                 5-11

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




cially at  an  integrated manufacturing facility  which 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, oxy-chlorination




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 oxy-chlorination  reactor  vent streams,  light-ends distilla-




tion 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; U.S. EPA,  I979b).   Releases from these manufacturing sites are




mostly to  the atmosphere and arise  largely  from vent  gas  streams (U.S.  EPA,




I979b).   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  which  are  generated  from  EDC manufacture  (vent gas




scrubbers, water produced during oxy-chlorination, 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




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.
                                      5-12

-------
     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  EDO were released  to the  environment  in  1979  from  lead scavenging




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




automobiles,  filling and evaporation  from gasoline  storage tanks, and combustion




of  the gasoline.   Approximately  '% 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.




      5.5.2.5.  OTHER DISPERSIVE USES —  In ether  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.  FATE AND TRANSPORT IN THE ENVIRONMENT

     The fate  and  transport 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 4), 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  [01 n] that then  reacts with water vapor to produce


•OH radicals.  The tropospheric half-life (t. .„)  of a compound is related to the

•OH radical concentration according to the expression:


          t      0-693
           1/2 ~ Kt'OH]


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
                                      6-1

-------
ranging from 0.7 to 7 mm Hg  and  at  a temperature of 23°C was 22+5 (standard



deviation of average)  cm  molecule"   sec" .



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



of Op  and  N? 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~^  cm  molecule"  sec"  with an upper limit value of



29 x 10~   cm  molecule"  sec" .  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 0? 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.



Therefore,  the measured rate constant value  of 22.0  + 5 cm  molecule"  sec"  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"   as 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

                               _1 ii   o         _i    _i
table that a value of 22.0 x 10    cnr molecule   sec   is more consistent with


                                                                            -1 4
the measured K values for other chlorinated ethanes than a value of 6.5 x 10


  3   -,   -, -1     -1
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
   .  Rate Constant
10   cm  molecule"  sec~
    Reference
CH_CHC12
CH2C1CH2C1
CH2C1CHC12
       39, 44



         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., 1981
                                    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 U
                                                    1 .75
                                                         -1 4
EDC reaction with *OH at 265°K can be estimated  as 	  cm  molecule"  sec"
or 12.6 cm  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
                     f               T
from 0.2 to 0.9 x  10  molecules cm"3 annually  (Logan et al., 1981; Crutzen and
Fishman, 1977;  Neely and  Plonka,  1978).   Singh  et al.  (1983) have  recently
estimated a mean hydroxyl radical  concentration of 0.4-0.6 x 10  molecules cm
over the troposphere of continental United States.  Therefore, a value of 0.5 x
1 0   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+2x10
molecules cm.   Assuming  equal periods of daylight and  darkness,  the average
concentration for a  24-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
                                          _11|    o          _•)     _i
value  at  ambient  temperature  (22  x   10     cnr  molecule    sec  )  and  an
                                      6-4

-------
 •OH concentration of 1 x 1 0  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 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 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 Harst




 (1978).   These investigators  found  that the chlorine-sensitized photooxidation




 of  10 ppm EDC  with H  ppm  Cl  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),  CO   (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  towards  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 1 0  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 0_.   The rate of reaction of *OH with halocarbons is one mechanism


which 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  EDC from  the  atmosphere.    Of the  chemical




processes, the most significant reaction that may account for the partial removal




of EDC from the atmosphere  is its reaction with *OH radicals.   The half-life for




the  process  has  been estimated  to  be  =36  to 127  days.   Therefore, it  is




anticipated that EDC will persist for a relatively  long time  in  the atmosphere




and may participate in intramedia transport from its source of emissions.




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




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




slowly under these conditions (Price et al., 197**;  Tabak  et al.,  1981; Ludzack




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




Under  anoxic  conditions,  Bouwer  and  McCarty  (1983)  observed  very  little




degradation of EDC with anaerobic  inoculum.   In a river die-away test, Mudder et




al. (1982) reported no degradation of EDC at concentrations of -1  ppm and above.




It  can be  concluded  from  these discussions  that biodegradation  will be  an




insignificant fate process for EDC in ambient aquatic media.






                                      6-7

-------
     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 1.6  cm,  a




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




under stirring (Chiou et al.,  1980).   Under 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 4  hours.




     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.




     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
                                      6-8

-------
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 Williamette silt loam containing 1 .6$ organic matter,
                  oc
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  NJ  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
6.4. SUMMARY
     Based on available  kinetic data and  average tropospheric  hydroxyl  free
radical concentration, the half-life  of  EDC,  the most likely removal mechanism
for EDC from  the atmosphere, has been estimated to range from 36 to 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.
                                      6-9

-------
     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.   Sizeable quantities of EDC are  judged  to be  released  to  the




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




 turing  site.  This  judgement  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.  Recent measurements




 made  at sites  in  three geographical  areas  central to  EDC production  and user




 facilities indicated EDC concentrations as high as 18H 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 Diehloride
Location
ALABAMA
Birmingham

Birmingham
ARIZONA
Phoenix
Grand Canyon
CALIFORNIA
Dominguez

Los Angeles
Oakland
Riverside
Upland

Upland
COLORADO
Denver
ILLINOIS
Chicago
KENTUCKY
Calvert City

Calvert City
LOUISIANA
Baton Rouge
Baton Rouge
Geismar
Iberville Parish
Lake Charles
Type of Site Date

April, 1977

April, 1977

urban
rural Nov-Dec 1977

May, 1976

urban
urban Aug-Sept 1977
August, 1980


Aug-Sept 1977

June, 1980

April, 1981

12 cites near Aug-Sept 1978
chem. plant
Aug-Sept 1978

March, 1977
March, 1977
Industrial Feb, Mar 1977
Jan, Feb 1977
June-Oct 1978
Analytical Number of
Method Samples

2

7 (1 ND)

GC/ECD
7 (7 ND)

1

GC/ECD
GC/ECD
GC/ECD 1 02
8

16 (11 ND)

GC/ECD 9 5

GC/ECD NR

156

GC/MS 88

25 (9 ND)
GC/MS 18 (2 ND)
10 (1 ND)
11
98
Concentration (ppt, v/v)a
Max.

98.6

99

1151 .1
—

—

1351.8
813.0
580
212.9

210

180

2820

17819.2

17800

630
2556.5
2551.5
1161.2
61000
Min.

50.7

0

38.8
	

	

173.0
38.3
89
61.8

0

100

22

0

<120

0
19.3
21.7
2.2
150
Avg.

71.7

35

216.3
	

3662.3

519.2
82.5
360
110.3

27

210

195

1631.6

5000

270
129.1
760.1
320.9
27000
Reference

Pellizzari and
Bunch, 1979
Pellizzari, I979a

Singh et al., 1980
Pellizzari, I979a

Pellizzari and
Bunch, 1979
Singh et al., 1980
Singh et al,, 1980
Singh et al., 1980
Pellizzari and
Bunch , 1 9 79
Pellizzari, I979a

Singh et al., 1980

Singh et al., 1982

Suta, 1979

Elfers, 1979

Pellizzari et al., 1979
Pellizzari, 1978a
Pellizzari, I978a
Pellizzari, I978a
Pellizzari, !979b

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

Lake Charles
Lake Charles

New Orleans
New Orleans

Plaquemlne

MISSOURI
St. Louis
NEW JERSEY
Batsto
Bound Brook

Bridgeport

Brldgewater
Burlington

Camden
Carlstadt
Clifton
Deepwater

East Brunswick

Edison

Edison

Type of Site Date

June, 1978

Sept-Oct 1978
12 sites near Sept, Oct 1978
chem. plant
October, 1978
near chem. October, 1978
plant
Jan, Feb 1977


May-June 1980

rural Feb-Dec 1979
off highway March, 1976

September, 1977

July-Aug 1978
September, 1977

urban Apr-Oct 1979
September, 1979
March, 1976
near chem. June, 1977
plant
marina July, 1976

Mar-July 1976

near waste Mar-July 1976
disposal area
Analytical Number of
Method Samples

6

GC/MS 1 1 0
144

GC/MS 91
120

11


GC/ECD 90

GC/FID-ECD-MS 42 (40 ND)
1

2

22 (18 ND)
1

GC/FID-EDC-MS 23 (21 ND)
GC/MS 16 (7 ND)
1
6 (3 traces)

2

33 (20 ND)

16 (1 trace)

Concentration (ppt, v/v)a
Max.

307.3

184000
191 545.6

41 700
40222.6

921.4


260

	
___

	

380


—
998.0

13.1

85.8

3400

14091 .5

Min.

5.2

<120
0

<120
0

2.2


65

	
— _ _

__-

0
	

	
33.4

5.9

37.1

8.6

53.6

Avg.

86.5

27500
18121.2

2900
1780.0

337.2


120

Trace
(2 samples)
Trace

Trace

170
Trace

Trace
(2 samples)
537.2
15950
7.4

61.6

1600

2712

Reference

Pellizzari and
Bunch , 1 9 79
Elfers, 1979
Suta, 1979

Elfers, 1979
Suta, 1979

Pellizzari and
Bunch, 1979

Singh et al. ,

Bozzelli et al
Pellizzari and
Bunch , 1 9 79
Pellizzari and
Bunch , 1 9 79
Bozzelli et al
Pellizzari and
Bunch , 1 9 79
Bozzelli et al













1980

., 1980




., 1979


., 1980
Pellizzari, I978b
Pellizzari and
Bunch, 1979
Pellizzari and
Bunch , 1 9 79
Pellizzari and
Bunch, 1979
Pellizzari and
Bunch, 1979
Pellizzari and
Bunch, 1979










-------
TABLE 7-1 (oont.)
Location
NEW JERSEY (cont.)
Elizabeth

Elizabeth

Fords

Hoboken

Linden

Linden
Middlesex
-~3 Newark
_J_ Newark

Newark



Passalo

Paterson

Rahway
Rutherford

Rutherford
Sayreville

Somerset
South Amboy
NEW YORK
Niagara Falls

Staten Island

Staten Island
Type of Site Date
Sept 1978-Dec 1979

near Indus- Jan-Dec 1979
trial plant
March, 1976

March, 1976

industrial June, Nov 1977

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

urban Mar 1976-Dec 1979



March, 1976

March, 1976

September, 1978
May 1978-Dec 1979

suburban May, 1979
July, 1976

July, 1978
industrial Jan-Dec 1979

residential February, 1978
(Love Canal)
urban November , 1 9 76

Mar-Apr 1981
Analytical Number of
Method Samples
GC/FID-ECD-MS 71 (53 ND)

GC/FID-ECD-MS 54

.



16

GC/MS 1 1
18
GC/FID-ECD-MS 37


160








GC/FID-ECD-MS 196

46


29
48

9



GC/ECD

(7 trace, 46 ND)

1

2 (2 ND)

(1 trace)

(1 ND)
(13 ND)
(9 trace, 28 ND)
1

(112 ND)



1

1

16
(141 ND)

(3 trace, 42 ND)
1

(17 ND)
(4 trace, 44 ND)

(2 trace, 7 ND)

3

NR
Concentration (ppt, v/v)a
Max.
2200

	

	

0

48.2

4.9
290
	
	

5800



	

	

775.5
3200

	
	

1800
	

	

50.7

4312
Min.
0

	

	

0

2.0

4.9
28
	
	

0



	

	

165.4
0

	
	

0
	

	

46.0

55
Avg.
220

2202.7

Trace

0

14.3

4.9
110
Trace
Trace

450



Trace

Trace

525.8
370

593.3
9372.8

490
Trace

Trace

48.2

256
Reference
Bozzelli et al., 1980;
Pellizzari, 1979a
Bozzelli et al., 1980

Pellizzari and
Bunch, 1979
Pellizzari, I977a;
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., 1980;
Pellizzari, 1977
Pellizzari and
Bunch , 1 9 79
Pellizzari and
Bunch , 1 9 79
Pellizzari, I978b,d
Bozzelli et al. ,
1979, 1980
Bozzelli et al., 1980
Pellizzari and
Bunch, 1979
Bozzelli et al., 1979
Bozzelli et al., 1980

Pellizzari, I978c,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
Free port

Houston

Houston
Houston

La Porte

Pasadena
UTAH
Magna
Analytical
Type of Site Date Method

June, 1980

July-Sept 1977
July-Sept 1977
July, 1977

August, 1977

August, 1977

August, 1977

urban April, 1981 GC/ECD

June-Oct 1977
March, 1980
industrial August, 1977

Apr-May 1978
industrial August, 1977

July 1976-May 1980 GC/ECD

urban NR
streets, July 1976, June,
parks, rural July 1978
highway August, 1976

July, 1976

Oct-Nov 1977
Number of
Samples

6

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

2

2

H

NR

3 (3 ND)
11
6 (3 trace)

22 (19 ND)
2

99 (5 ND)

30
10 (1 trace)

1

1

9 (9 ND)
Concentration (ppt, v/v)a
Max.

110

0
0
0

	

	

238.6

237

0
670
16390.6

29
1112.5

3000

16390.6
109.8

	

39

0
Min.

110

0
0
0

	

	

H.3

66

0
670
1002.5

0
815.8

50

73.1
30.1

	

39

0
Avg.

110

0
0
0

63.8

18.2

139.2

121

0
670
6353.5

1.3
961.2

1300

893.0
63.0

192.3

39

0
Reference

Wallace, 1981

Pellizzari, I978e
Pellizzari, I978e
Pellizzari, I978e

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

Pellizzari et all, 1979
Wallace, 1981
Pellizzari and
Bunch, 1979
Pellizzari, I979a
Pellizzari and
Bunch , 1 9 79
Pellizzari 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  (cont.)
Location
VIRGINIA
Front Royal

WEST VIRGINIA
Charleston

Charleston
Institute
Institute

Nitro
I
cr> Nitro
St. Albans

St. Albans
S. Charleston

S. Charleston

W. Belle
W. Belle

WASHINGTON
Pullman

Pullman
Analytical
Type of Site Date Method

Oct, Nov 1977


Sept, Nov 1977

Sept-Nov 1977 GC/MS
November, 1977 GC/MS
November, 1977

Oct, Nov 1977

Sept-Oct 1977 GC/MS
October, 1977

Sept-Nov 1977 GC/MS
Mar, Sept-Nov 1977

Mar-Nov 1977 GC/MS

Sept-Nov 1977 GC/MS
Sept-Nov 1977


rural Dec 1971-Feb 1975 GC/MS

rural November, 1975 GC/ECD
Number of
Samples

16


3

1) (1) ND)
2 (2 ND)
3

1)

6 (6 ND)
1

1) (D ND)
6

16 (1 5 ND)

6 (6 ND)
1)


NR

1
Concentration (ppt, v/v)a
Max.

86.0


	

0
0
	

63.8

0
—

0
63.8

37

0
61.8


—

— —
Min.

37.3


—

0
0
	

37.3

0
	

0
37.3

0

0
37.3


<5

10
Avg.

1)8.2


57.1

0
0
37.3

46.7

0
1)8.2

0
52.4

2.3

0
1)7.0


	

	
Reference

Pellizzari and
Bunch, 1979

Pellizzari and
Bunch, 1979
Pellizzari, I978e
Pellizzari, I978e
Pellizzari and
Bunch, 1979
Pellizzari and
Bunch, 1979
Pellizzari, I978e
Pellizzari and
Bunch, 1979
Pellizzari, I978e
Pellizzari and
Bunch , 1 9 79
Pellizzari, I979a;
Pellizzari, I978e
Pellizzari, I978e
Pellizzari and
Bunch, 1979

Grimsrud and
Rassmussen, 1975
Harsoh et al. , 1979
 Assume ambient temperature of 258C and atmospheric pressure of 760 mmHg.
ND = Not detected
NR = Not reported
BCD = Electron capture detection
FID = Flame ionization detection
GC = Gas chromatography
MS = Mass spectrometry

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




41 .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  spectrometry.   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  ^ig.  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 (41 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 Bichloride 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
Bichloride Concentrations (ppt)
Maximum
1353
1450
842
7300
607
2089
2505
4312
237
2820
Minimum
173
39
38
50
45
56
63
55
66
22
aSingh 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 which 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 which  have been  found in the  ground and  surface water  of the U.S.  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




(Syraons 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 m& sealed  vials, with vola-




tilization  prevented  by the  absence of head  space.   Samples  were subsequently




shipped on ice to the EPA Water Supply Research Lab 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 EDC,  six were found to contain quantifiable levels of EDC at



0.2-6.0 mg/Q,.




     The National Organics Monitoring Survey  (NOMS)  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-m2. 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 ug/1.  No detectable levels of EDC  were found during Phase




III of the study (November 1976 to January 1977).




     In Phase I of the NOMS (March to April 1976), water  samples from 87 surface




water systems were analyzed for EDC.  Of  these  87 systems, only one was found to




contain EDC, at  2.0 ug/1.   When  these  systems  were sampled  again during  the




second phase  of the  survey,  one  was found to  be  contaminated,  at 1.8  ug/1.
                                     7-n

-------
During  the  third phase of  the  NOMS (November 1976  to  January 1977),  analyses




revealed EDC contamination in one system, at 1 .25 (ig/1.




     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 1 69  water systems in  33 states were analyzed for 51 organic chemical




contaminants (SRI, 1981).   Analyses were carried out by gas chromatography 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/5,.   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/2,.




     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,   1981 a).    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 ug/
-------
      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,  1982).   A  total  of  9^5 systems  were



 sampled,  of which  466  were chosen at random and 1*79 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  466




 randomly chosen water  systems,  seven were contaminated with EDC, at concentra-




 tions ranging from 0.29-0.57 [ig/S,. 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  ng/.l.   Of the 479 nonrandom locations sampled,




 nine  were contaminated with EDC,  at concentrations between 0.33-9.8 ng/£.  Of the




 nine  positive samples,  four were from systems serving populations in excess of




 10,000 people.  The average EDC  level  for the nonrandom systems was 2.7  \ig/H.




      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 were




 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, U7 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 ug/£).   The  range  of concentrations was
                                      7-13

-------
0.5-18 |ig/H; the mean and median concentrations of the positive values were 5.6




and 1 .7 |ig/5-,  respectively.   Both the mean and median values of all samples were




<0.5 |ig/£.  Of the 47 surface water  samples,  only one (2.1$) was found  to have EDC




present (minimum quantification limit generally  0.5-1.0  ng/?,).   The concentra-




tion of the one positive sample was 19 ug/&.  The mean and median values of all




samples were both <0.5 \ig/l>




     The  combined  EDC groundwater data  and surface water  data  from the above




Federal studies  are  summarized in Tables 7-3  and 7-4, respectively.   The RWS




study was deleted from these summarizations  because the RWS data were  recorded by




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  |ig/&, and that most are below 1.0  ng/fi,.




     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 n.g/&  (Letkiewicz et al., 1932).




7.1.3.  Soil and Sediment Levels




     No data were available  on monitoring of EDC concentrations in soils, sedi-




ments 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
                                      7-14

-------
                                                    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
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
15,631
1,909
1,331
881
1 ,065
1 ,159
1 ,101
68
16
58
Number of
positive
systems
Number of
systems
sampled
1/175
0/220
0/111
3/151
0/10
2/79
1/111
11/296
1/37
0/12
1/13

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
<1 .0 ug/2,
171
220
111
118
10
77
113
285
36
12
12




Number of systems with
measured concentration
(iig/J,) of:
<1 .0
1
0
0
1
0
1
0
6
1
0
1
1 .0-5
0
0
0
2
0
1
1
1
0
0
0
>5-10
0
0
0
0
0
0
0
1
0
0
0
>10
0
0
0
0
0
0
0
0
0
0
0
 Letkiewicz et al.,  1982
bPositive systems are those with  quantified  levels  of  ethylene  dichloride.

-------
                                                     TABLE  7-4

                      Reported  Occurrence of Ethylene Bichloride  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
5,001-10,000
10,001-50,000
50,001-75,000
75,001-100,000
>100,000
Number of
systems
in U.S.
1 ,412
2,383
1,341
1,911
514
720
912
1,306
156
85
218
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 ng/P,
4
19
13
21
6
14
10
37
20
14
97
Number of systems with
measured concentration
(UK/JO of:
<1 .0
0
0
0
1
0
0
0
1
1
0
3
1 .0-5
0
0
0
0
0
1
0
0
1
0
1
>5-10
0
0
0
0
0
0
0
0
0
0
1
,,0
0
0
0
0
0
0
0
0
0
0
0
aFour systems reported as undetected with the unusually high detection limit of 2.0 pg/?, were deleted.

bPositive systems are those with quantified levels of ethylene dichloride.
Source: Letkiewicz et al., 1932

-------
                                   TABLE 7-5

               State Data on Ethylene Dichloride in Groundwatere
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
1 2 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 Range
(ug/£) (fig/A)

21
12
1 .2
1.2, 5.3

ND
7.8

ND
ND
ND


777 30-2,100
19, 160

ND
14.5 10.1-19.1
11 .6, 18.2
ND

ND
ND
1.3 1.1-1.9
2.0-2.1
ND
ND
ND
0.1-0.9
1-10
1 0-1 00
Number
of
samples

1
1
2 (1
2

1
1

1
1
2


11 (4
15 (13

H
10 (7
2
1

U
7
15
13 (11
1
154
228
4
4
1




ND)










ND)
ND)


ND)






ND)






 Letkiewicz et al.,  1982
ND = Not detected,  N/S = Not specified,  F
= Finished, D = 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 are


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 ug/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 Interna-
                                      7-18

-------
                                   TABLE 7-6
              Estimated Respiratory Intake of Ethylene Bichloride
                             by  Adults  and  Infants


Low:
Intermediate:
High:
Exposure
Ug/nr
0.25
2.5
25
Level
(ppb)
(0.062)
(0.62)
(6.20)
Intake
Adult
0.08
0.82
8.21
(ng/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 nP of air inhaled/day (infant).

aLetkiewicz et al., 1982
                                     7-19

-------
tional, 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 U.S.  fall  below 10




\ig/i, and that most are below 1.0 ug/&  (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 which contain 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,  I98lb).   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 7-7
                 Maximum Concentration Level in the Vicinity of
                           Various Emission Sources3
        Source
Maximum Annual Average EDC
 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
 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.
                                   7-21

-------
                                  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)
to concentrations (ug
<1.0 1.0-5 >S-1
69,239
126,356
195,595
(100*)
exposed
/£) of:
0 >1 0
0.0
0.0
0.0
(0.0*)
Letkiewicz et al., 1982
                                  7-22

-------
                                   TABLE 7-9
                 Estimated Drinking Water Intake of Ethylene
                       Dichloride by Adults and Infants
                                                    Intake(ug/kg/day)
Exposure Level

1 .0
5
10
100
Adult

0.029
0.14
0.29
2.9
Infant

0.24
1 .2
2.4
24.0
Assumptions: 70 kg-man,  3.5 kg-infant,  2 I of water/day (man),
             0.85 i of water/day (infant).

aLetkiewicz et al., 1982
                                   7-23

-------
                            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.




     1,2-Dichloroethane has been reported  to be non-toxic to  many economically




important plant species when directly applied to growing plants (Cast and Early,




1956).




     Toxicity of EDC to barnacles  (Barnacle nauplii)  and  unicellular  algae has




been reported (Pearson and McConnell, 1975).  The LC5Q for Barnacle nauplii was




reported  to  be  186  mg/S,.   The  effective concentration  needed to reduce the




photosynthetic ability of unicellular algae by 50% was 340 ppm (340 mg/5,).




     A 21-hour  median  tolerance  limit  of  320  ppm  (320 mg/S,) was  reported for




brine shrimp under static test conditions (Price and Conway,  1974).




     Static  24-hour  and  96-hour LC™  concentrations of >600  and   430  mg/Jl,




respectively,  have  been  determined  for  the  freshwater  bluegill   (Lepomis




macrochirus)   (Buccafusco  et  al.,  1981).   Garrett  (1957) reported  that  the




concentration of EDC needed to produce >50% mortality in the  marine pinperch




(Lagodon rhomeboides) under static conditions was 175 mg/S-.




     An acute LC(-0 of  115 mg/J, 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 LC5Qs  in the  range of  130-230 mg/5,




were determined  for  sheepshead minnows (Cyprinodon variegatus)  (Heitmuller et




al., 1981); the  observed  no-effect  concentration was reported to  be  130 mg/l.




The  estimated  acceptable concentration  of EDC in a  32-day  early life  stage
                                      8-1

-------
toxicity test with freshwater fathead minnows (Pimephales promelas) lies in the



range of 29-99 mg/J, (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  ppm.   It is appreciably soluble in water




(0.869 g/1 00 mH) (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 quantitated  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/cra2




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

                  p
(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/mJ, 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 LD,.Q 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,


mice, 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.  (1980,


1982)  found  that    C-EDC in  corn  oil given  perorally to rats  (150 mg/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 Chloroethanes

1 ,2-Dichloroethane
1 ,1 -Dichloroethane
1 ,1 ,1 -Trichloroethane
Vapor Press.
at 25°C, torr.
80
250
125
Ostwald
Water/
Air
11.3
2.7
0.93
Solubility
Blood/
Air
19.5
4.7
3.3
Coeff., 37°C
Olive Oil/
Air
W7
187
356
Adapted from 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            30.3
             37°C,  150 ppm            14.9

  Adipose tissue/blood                 8.7**

  Lung tissue/blood                    0.45**

  Liver tissue/blood                   0.72**
                              Spreafico  et  al.,  1980
                              Reitz  et al.,  1980

                              Reitz  et al.,  1980

                              Reitz  et al.,  1980

                              Reitz  et al.,  1980
 •Conversion factors:
••Dose-dependent
At 25°C/760 torr,  1  ppm in air =4.05 mg/m3
1  mg/liter air = 247 ppm
                                   9-4

-------
 air,  urine and carcass  (Table 9-3).   Spreafico et al. (1978, 1979, 1980) found



 that  25,  50  and 1 50 mg/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-^.   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. (1978, 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 (1 50 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.U minutes  for



the highest  dose in  corn oil  (150 mg/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  Fate  of    C-EDC  in  Rats  48  Hours  After  Oral  (150  mg/kg)  or
                         Inhalation (1 50 ppm, 6-hr) Exposure
Charcoal trap
 (B)
                                Oral
                                        Inhalation
                    iunole/kg
              metabolites
nmole/kg
447 +  60
 9.4 + 0.4
metabolites
Body burden 1 539 + 391
(total radio-
activity)
512
9.4
+ 135
+ 0.4
Total metabolites
(A-B)
Urine
CO trap
Total carcass
(48 hr after
exposure)
Feces
Cage wash
(1092)

926 + 348
83.1 + 11 .9
46.9 + 14.7


26.3 + 1*».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
3.3^ ± 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 umole equiva-
lents of EDC, based on the specific activity of   C-EDC (3.2 mCi/mM).

From Reitz et al., 1980.
                                     9-6

-------
  0.1
            1h    2h    3h            Sh                 8h
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 ( * ).
     Source: Spreafico et at.  (1978.  1979.  1980).
                            9-7

-------
       Table 9-4  Peak Blood and Tissue Levels After Single Oral Dosage
                  of EDC  in Male Rats
Dose
mg/kg
25
50
150
Blood
13.29
31 .94
66.78
Adipose tissue
Hg/ml or \ig/g
110.67
148.92
259.88
Lung
2.92
7.20
8.31
Liver
30.02
55.00
92.10
Adapted from Spreafico et al.,  1980
                                   9-8

-------
       Table 9-5  The Absorption Rate Constants (k ) and Area Under Curve
                 (AUC)  for Rats After Single Oral ^ith  Doses  of  EDC  in  Oil
                 and in Water as Vehicle and After  Intravenous Administration
Dose
(mg/kg)
Oral 25

50
ka
. -1
mm
0.299
0.209
0.185
0.181

(water)
(oil)
(oil, male)
(oil, female)
AUC (blood)
ug x min x ml"
466
466
1700
1685
                       150
0.109 (oil)
7297
Intravenous (water)       1

                         5

                        25
                           9

                          54

                         595
From Spreafico,  1978,  1979,  1980
                                   9-9

-------
(water) vs. k  ,  0.209 (oil); 25 mg/kg].   There appears to be no gender difference
            a,



for absorption.




     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 ug x rain x m£~  after a 25 mg/kg intravenous dose and was 466 ug x rain x




mil"  after  25 mg/kg oral  dose.  These  results  indicate  that  absorption of the




oral dose  was very extensive,  averaging  TQ% 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  (400 g)  following intragastric




intubation of equivalent doses  (100 mg/kg) in -4 m£ of water or of corn oil.  The




post-absorptive peak  blood  concentration  averaged  5  times  higher  for  water




vehicle  than  corn  oil  (84.6  vs   15.9   ug/m&);  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  3.0; water:corn oil.   These




results are in  general agreement with  those  of Spreafico et  al.  (1978,  1979,




1980) (Tables  9-4 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
                                     9-10

-------
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




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;  (U) 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.




     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 pf blood concentration of EDC, observed  by  Reitz et al. (1980,






                                     9-11

-------
     12
                          200       300       400

                               TIME (minutes)
600
    10.0
O
O
O
O
O
IU
CO
S
c
o
O
cc
O
                         40                80

                             TIME (minutes)
  Figure 9-2   Top; Blood |evell of EDC observed during and follow-
      ing a 6-hour inhalation exposure to 150 ppm EDC.  Data from
      four male Osborne-Mendel rats were fitted to a two-compart-
      ment open model as described by Gehring et al.35 The compu-
      ter plot is shown. The arrow indicates exposure termination.
      Bottom: Semi-logarithmic plot of EDC blood levels vs. time
      after exposure termination.  Source:  Reitzetal. (19RO.  1932).
                            9-12

-------
 1982)  during a  6-hour inhalation  exposure  to  150  ppm  EDC.    Blood and  body




 equilibrium  with the blood concentration maintained a plateau level  of  9  ng/m?,




 (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  during   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.M   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 and




mature breast  milk.    Urusova  (1953)  found EDC concentrated  in  breast  milk




 (5.M-6.1J 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 Inhalatory Exposure
Time of
Exposure
30 minutes
1 hour
2 hours
4 hours
6 hours
EDC ug/g or ug/ral + S.E.
Blood Liver Lung Adipose Tissue
0.48 + 0.05 0.32 + 0.02 0.14 + 0.02 2.91 + 0.22
0.92 + 0.09 0.67 ± 0.04 0.27 ± 0.03 7.49 + 0.60
1.34 ± 0.09 0.84 + 0.09 0.34 + 0.03 10.31 ± 0.94
1.34 + 0.11 1.14 + 0.17 0.42 + 0.05 11.08+0.77
1.37+0.11 1.02 + 0.10 0.38+0.02 10. 19 +1.00
From Spreafico et al.,  1980
                                  9-14

-------
          Table 9-7  EDC Tissue Levels After 250 ppm Inhalatory Exposure
Time of
Exposure
30 minutes
1 hour
2 hours
3 hours
6 hours
EDC ug/g or ug/ml + S.E.
Blood Liver Lung
6.33 ± 1.04 3-82 + 0.78 2.19 + 0.21
11.65 + 1.12 7.34+0.74 6.40+0.20
23.64 + 0.91 16. 39 ±1.18 14.07+0.47
29.36 + 1.01 20.83 ± 2.21 14.47 + 1.12
31.29 + 1.19 22.49 + L12 14.14 + 0.90

Adipose Tissue
26.75 + 3.12
82.64 + 2.19
151 .53 + 12.17
252.18 + 14.62
273-32 + 12.46
From Spreafico et  al.,  1980
                                 9-15

-------
fell to 4 ppm.  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 1 50  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  parallel  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 1 50 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 EDC in Male Rats 2 Hours
                   After Single  Oral  Doses  in  Corn Oil
Tissue
(|ig/ml, or
lig/g ± SE) 25
Blood 0.46 + 0.04
Liver 0.32 + 0.01
Kidney
Brain
Spleen
Lung <0.05
Adipose 7.80 + 0.72
Dose, mg/kg

50
5.89 + 0.41
3.81 + 0.45
3.43 + 0.26
2.96 + 0.36
1 .79 + 0.35
1 .44 + 0.10
55.78 + 6.88


150
26.60 + 1 .23
21 .62 + 3.99



1 .62 + 0.25
178.50 + 24.66
         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
From Spreafico et al.,  1978,  1979,  1980.
                                  9-17

-------
     Spreafico and his  colleagues  (1978, 1979, 1980) also investigated blood and




tissue concentrations in rats during 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 inhalatory 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 pprn



inhalation provides blood,  liver and  adipose  tissue levels  slightly  greater than




a 150 rag/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 EDC



concentrations in exhaled air of 14.5 ppm.  The breath concentration declined to



=3 ppro after 18  hours.  These values lead to an approximation of 9 hours for the



half-time of pulmonary elimination of EDC in man.  Similar observations have been



made for animals (monkey, dog, cat, rabbit,  rat and guinea pig) in early investi-



gations of the anesthetic properties and toxicities of EDC (Heppel et al., 19^5;



Kistler  and  Luckhardt, 1929;  Lehman and  Schmidt-Kehl,  1936).    In  controlled



studies  in  mice, Yllner  (1971 a) found  that up  to  ^5%  of an  intraperitoneal



injected dose of EDC (1 70 mg/kg)  was recoverable unchanged in exhaled air (Table



9-9).  The percentage of EDC recovered unchanged in exhaled air increased expo-



nentially 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,


                                      1 4
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 .8/6 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



dosage to experimental  animals have failed  to detect unchanged EDC (Spreafico  et
                                     9-19

-------
     Table 9-9  Percent Distribution of Radioactivity Excreted (48-hr)  by Mice
                         Receiving 1,2-Dichloroethane-  C
0.05
                                           Dose g/kg
                                           0.10      O.U
                                                               0.1?
  C02 (exhaled air)               13

Dichloroethane (exhaled air)      11

Urinary metabolites               73
                                             8         4         5

                                            21        46        45

                                            70        48        50
Adapted from Yllner (1971 a)
                                    9-20

-------
al.f 1980; Yllner, 19 71 a).  Mutagenic metabolites  of EDC have been reported in




rat and mouse bile (Rannug and Beije, 1979; Rannug, 1980), 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  mS, 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:  k ,  0.24 min" ; V., 43 ml; k.. ? and k?1 ,  0.02 and 0.04




rain" , 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, distri-




bution, metabolism  and  elimination, or that  three  compartment  kinetics  were




followed at all dose levels but with lower doses;  the third exponential compo-




nent, representing the third compartment, was obscured by the limit of analytical




sensitivity.  Average kinetic values for the three compartment model  were:  k ,
                                                                             6



0.094 min"  ; V   67.9 m£;  k12,  k21 ,  k^, k^ ;  0.06,  0.08,  0.01  and 0.01  min" ,
                                     9-21

-------
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 1 5 mg/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 ($).   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 (T1  .  ) 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




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 1 50 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

-------
  1   1
     0.01
                         0.5 h
                                                              V« h
Figure 9-3  Blood levels of EDC in rats after i.v. administration. ( •) 25 mg/kg i.v.;
   (O) 5 mg/kg i.v.; (A) 1 mg/kg i.v. Source:  Spreaficoet al.(1976. 1979.  1980).
                                   9-23

-------
   Table 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.
Parameter
GO, ug x ml"
K , min
V • ~1
K.. _ , mm
v • -1
K21 , mm
AUC, ug x min x
ml
T 1 /2 3 min
Dose , mg/kg
1 5 25
1 . 50 8 . 00 38 . 1 2
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
Clearance 21.98 17.46 7.98
ml x m~

Vol. distr., ml 231 239 1 62
K12

^




                     21


              el  (inhalation)

                  or
             V (Km + C)  (oral)
From 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,  T1 ,p,  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, T1 <2 and volumes of distribution, V,,



for 1 5 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. included  in  their  comprehensive  studies  on the pharmaco-




kinetics 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 estabish 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-^.




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 steady-state inhalation conditions are
                                     9-25

-------
      Table 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.
Parameter
Oil Vehicle
Ka, min"
Kel' min"
AUC, ug x min x ml"
T 1 /2 3 min
Clearance, ml x min"
Vol. Distr., ml.
Aqueous Vehicle
_1
K min
. -1
K , , mm
AUC, ug x min x ml
T 1/2 3 min
Clearance, ml x min"
Vol. Distr. , ml.
Dose, mg/kg
25 50 1 50
0.209 0.185 0.109
0.029 0.017 0.010
446 1 700 729 7
24.62 44.07 56.70
10.64 5.58 3-90
367 328 390
0.299
0.046
446
14.12
10.20
221
From Spreafico et al.,  1979,  1980.
                                 9-26

-------
       1000 er
        100 9-
1 1 1 1 1 1
li
                   0.5h
        1.5h
         2h
       1000
        0.1
                                                               Ij—•
                                                               E
                   0.5h
1h
1.5h
2h
2.5h
3h
Figure  9-4  Levels of EDC in rats after inhalatory exposure to 50 ppm
     (top) and 250 ppm (bottom).  Levels were measured at termination of
     5-hour exposure period. Adipose tissue ( • ). blood ( o), liver ( •).
     lung (A). Source: Spreaf ico et al.  (1978, 1979,  1980).
                             9-27

-------
essentially those applicable to a  single  compartment open model.   The kinetic




parameters are given  in  Table  9-12.   The  body  burden or "dose" of EDC  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 pprn  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 (ig/mi) 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 monophasic  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
                                      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                               Exposure Concentration,  ppm
                                           50                   350

CQ, jig x ml"1                              1 .42                30.92

Kel, rain"                                 0.0561                0.0313

T 1/2, min                               12.69                22.13

AUC, ug x min x ml"                      26                  1023

From Spreafico et al.,  1979,  1980
                                    9-29

-------
      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 2-Compartment Model.
             Parameter

             CQ, ,ig x ml~1                           9.0

             Kel, min~1                              0.092

             v     .  -1                              0.019
             K-p, rain
             K21 , min~1                              0.025

             AUC, ug x min x ml"                 3018
                                                   35

             Vol. Distr, ml.                      51 3
From Reitz et al.,  1980
                                  9-30

-------
 from  a two-compartment open  model.   These  results  may be  compared  with data




 obtained  by  Spreafico  et al.  for  inhalation  exposure  (Table  9-12).




      Reitz et  al.  (1982) determined  also 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




 pharmacokinetic  parameters  calculated from the model.




      Several differences  exist between  the  observed  data  and calculated para-




 meters 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 (1.4 at 50;




 8.3  at 150;  and 30.9  ug/mSl at  250 ppm), clearly demonstrate saturation  of an




 elimination  process, presumably metabolism.  Reitz et al. found peak blood levels




 following oral dosing  (30-^4 ug/mP. at 150 mg/kg) to be  similar to those  found by




 Spreafico et al. after a 250 ppm inhalation exposure.  Reitz et al. found also a




 biphasic elimination after  both oral and  inhalation exposure with the  second or




 8 phase  essentially  equal  (T1/2   =  28  rain), and  similar  to the  monophasic




 elimination calculated  by  Spreafico et al. for the 250 ppm exposure  (T.. /? =  22



min).  The relatively slow  alpha elimination  phase (T1/2, 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
                                     9-31

-------
may lie in  the  different kinetic models, species differences  and  inability of




linear kinetics to accurately describe the kinetics  of EDC body elimination.




     9.1.2.4   BIOACCUMULATION — There  is  no definitive experimental evidence




in the  literature  concerning bioaccumulation in man after  chronic  or repeated




daily exposure to EDC.  While EDC has a moderately high fat tissue/blood parti-




tion 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




Retiz 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.  These investigators observed also




that their pharmacokinetic data in rats predict  essentially complete elimination




of EDC  from the body in  24  hours  following acute oral  or  5-6 hour inhalation




exposure.   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 1 50-200  m?;  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
                                      9-32

-------
EDC derives primarily from studies performed within the last 10 years.   Metabo-



lites which have been identified either in vivo in  mice and rats or in liver and



kidney tissue crude enzyme systems are listed in Table 9-14.  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


                                       1 U
estimated by balance studies utilizing   C-EDC.



     Yllner  (1971 a)  administered intraperitoneally  doses of 50-170  mg/kg  of


1 4
  C-EDC to mice in metabolism units with CO- and  solvent traps.  Yllner recovered



97-100$ of radioactivity  in exhaled air and urine within 2*) hours.   The results



of his balance studies are summarized in Table 9-9-  For the low dose of 50 mg/kg,



Yllner  found that  86$ 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 dose being



metabolized.  For the largest dose,  170 mg/kg,  55$ was metabolized to C0? (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 CO- and urinary
                                     9-33

-------
  Table 9-1 4  Identified Metabolites of Ethylene Dichloride and Ethylene Bromide.
                                 System
                          Reference
Inorganic halide


co2


2-Chloroethanol



2-Chloroacetic acid

Bromoacetaldehyde


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
microsoraes

mouse, rat
urine

rat liver
cytosol

rat liver
cytosol,
rat urine

rat urine

blood


mouse urine

rat liver
cytosol,
tissue
Heppel and Porterfield, 1948
Nachtomi, 1970

Yllner, 19 Tib
Reitz et al., 1980

Yllner, 19Tlb
Kokarovtseva and
Kiseleva, 1978

Yllner, l9Tlb

Hill et al., 1978
Yllner, l9Tib
Spreafico et al., 1979

Livesey and Anders, 1979
Nachtomi et al., 1966

Edwards et al., 1970

Edwards et al. , 1970




Yllner, l9Tlb

Nachtomi,  1970
S,S'-ethylene-bis-glutathione    rat  liver
                    Nachtomi,  1970

-------
metabolites occurred at a dose level of between 100-140 mg/kg and 50-100 mg/kg,



respectively.



     Reitz et al.  (1980,  1982) have found that EDC is also extensively metabo-



lized in the rat.  These  investigators conducted balance studies with   C-EDC in



Osborne-Mendel rats after oral (150 mg/kg) and inhalation (150 ppm for 6 hours)



exposure.  Their results are summarized in Table 9-3.   In the oral balance study,


             1 4
150 mg/kg of   C-EDC (or, 1520 itmoles/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


                                                           1 4
unchanged in exhaled air; 5% was metabolized completely to   C0_ and 60$ of the


                         1 4
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


                                             1 4
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



limoles/kg)   or one-third  of  the  oral  dose.   Of  this dose,  2$ was  excreted



unchanged in exhaled air,  7$  was  metabolized completely to   C02,  and  84$ was


                1 4
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.    demonstrate  extensive  biotransformation  in  the rat



(70-91$) and also  suggest a dose dependency with greater metabolism  for  lower



doses,  as Yllner (1971 a)  observed in the mouse.
                                     9-35

-------
     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,



5-10$ is metabolized completely  to CO- 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 (19 71 a)  identified the  metabolites appearing in the



urine of  mice  after dosing with   C-EDC  (1 70 mg/kg)  and found six   C-metabo-
lites; the  three principal metabolites  were:   S-carboxymethylcysteine



thiodiacetic  acid  (33%) and  chloroacetic acid  (1 5%)  (Table 9-15).   However,


                                                          1 4
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 in



vivo and in vitro studies  listed  in Table  9-1 ^ and also suggest the nature  of the



reactive intermediates  involved  in the covalent binding to cellular macromole-



cules (protein and DNA) which has been demonstrated in both  in vivo and  in vitro



studies (Section 9.1.3-5).



     A.   Microsomal  Reactions;    Yllner  (1971 a) 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
                                      9-36

-------
   Table 9-1 5  Percent Distribution of Radioactivity Excreted (M-hr)  as Urinary
              Metabolites by Mice Receiving 1,2-Dichloroethane-  C or
                             2-Chloroacetic Acid-  C.
Metabolite
Chloroacetic acid
2-chloroethanol
S-carboxymethylcysteine
Conjugated S-carboxymethyl-
cysteine
Thiodiacetic acid
S , S-ethylene-bis-cysteine
Glycolic acid
Oxalic acid
After
dichloroethane
(0.17 g/kg)
16
0.3
45
3
33
0.9
—
—
After
chloroacetate
(0.10 g/kg)
13
—
39
3
37
—
4
0.2
Adapted from Yllner (1971a,b)
                                  9-37

-------
metabolite) via  2-chloroacetaldehyde.   Kokarovtseva  and Kiseleva  (1978)  have




also identified chloroethanol in the blood of rats (T  /_ ca. 9 hours), and in rat




liver tissue within 1  hour and for 24-48 hours after oral administration of EDC




(750 mg/kg).  Van Dyke and Wineman (1971)  and subsequently Salmon et al. (1978,




1981) found  that rat  microsomal  fractions,  in the presence of 0-  and NADPH,




dechlorinated a series of haloalkanes  including EDC.   The reaction for EDC had a




Vm   of  0.24  nmol/min/mg protein with a K   of 0.14 mM  (Salmon  et  al., 1981).
 ID 3.X                                      ID



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  cyto-




chrome P450 with K values of 10.3, 30  and =15, respectively.  Hill et al.  (1978)
                  3



reported that 1,2-dibromoethane was activated  to  an  irreversibly-bound  species




by microsomal mixed-function oxidases, and also provided evidence that 2-bromo-




acetyaldehyde 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 0- 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 dehydro-




genase.  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
                                     9-38

-------
NADPH
                                 H
                                   -HX
                             Hi   0  Alco. D
                          ,x.c     __
      BINDING
      PROTEIN. DNA 2.ha,oacat.
                      aldehyde fr Aid. D
                            H.
              I
       H,

2-hatoethanol
                        2-hatoacetic acid
         5'i'   P450.0,  . F   V'"1  *  "1
       -C-C-CI NADPH    X-C-C-CI-O
         BINDING
         PROTEIN. DNA
                           •1 -haloso 2-chloroethane
      BINDING
      PROTEIN. DNA'
                                                      /OH
                                              2-haloethanol
                       2-haloacetaldehyde

                             f Ald.D.
                            H»   o
                         CI-C—C*-OH

                         2 haloacetic acid

Figure  9-5  Microsomal oxidative metabolism of 1.2-dihaloethanes.
    Source: Adapted from Guengerich et al. (1980),  Anders and
    Livesey(1980).
                          9-39

-------
enzyme,  substrate  and  cofactor  concentrations.    Williams  (1959)  also  had




suggested that chloracetic acid appeared iri vivo via chloroacetaldehyde.




     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 EDC.  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




macromolecules,  or  hydrolyze  (with  release  of  hypochlorite ion)  to  form  2-




chloroethanol.




     As  a  consequence  of  microsomal  P-lJSO-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 "detoxifica-




tion"  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
                                      9-40

-------
  2-cMoroacetaldehyde  2-chloroacetate
           -Cl'
                       GSH
                             -CI"
            -t?-H
    GS
S-formylmethylglutathlone
     S-carboxymethyl glutathione
                   glutathionase
                <  r (liver, kidney)
      HOOC-CH-CH,-S-CH,-COOH

        S-carboxymethyl cyt teine
              t^CH,-
                -COOH

           'CH.-COOH

       Thiodlglycolic ecid
                                                  f   V't *   "1
                                                  [ci-c-c-ci-oj
                                           1 -chloroso-2-chloroethane
                                                   iSH     I  -H
                                                   H, H,  f
      GSH
        Hi . •«  T
    CI-C-C-SG
S-(2 chloroethyl) gtutathlone
                                                           HOH
                                                           -cr
                                              GS^JBINDING  PROTEIN.
                                                  N            DNA
                                            episutfonium ion
                                      GSH
                                      GS-CH,-CH,-SG
                                      S.S'-ethylene-bis-
                                                                 HOH
                                        glutathione
                                                  GS-CH.-CH.-OH

                                                  S42 hydroxyethyl)
                                                      glutathione
     GSH. glutathione; Aid. D. aldehyde dehydrogenase; GT. GSH tranferase;
     DH. dehydrogenase
Figure  9-6 .  Further metabolism of 2-chloroacetaldehyde and l-chloroso-2-chloro-
     ethane from microsomal oxidation. Source: Adapted from Guengerich et at. (1980)
     and Anders and Livesey < 1980).
                                   9-lM

-------
that chloroethanol was  converted by alcohol dehydrogenase to chloroacetaldehyde,




which then conjugated with GSH to give S-formylmethylglutathione, and thence by




an NAD requiring dehydrogenation to S-carboxymethylglutathione.  Johnson (I966b)



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-carboxymethylgluta-




thione 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 (1971 a) 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 adminis-




tering 2-chloroacetate  to mice (Table 9-15),  Yllner  0971b) proposed  that  2-




chloroacetic acid could also conjugate with GSH with chloride  excision forming S-




carboxyraethylcysteine and thereby enter the pathway (Figure 9-6).  Spreafico et




al. (1979) found that after rats were given oral doses of 50 and 1 50 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 which  they proposed  was formed  by microsomal




P450  oxidative  emtabolism  of  EDC,  would  be extremely  reactive and could  be




expected to  either rearrange  to 2-chloroacetaldehyde, hydrolyze  to  2-chloro-




ethanol,  or  react  directly with microsoraal  protein (Fig.  9-5).   In addition,




these workers postulated that the chloroso compound  may  react with GSH to form S-




(2-chloroethyl)glutathione,   a  half  sulfur  mustard, which could  react via  an




episulfonium ion intermediate  with macromolecules  (Fig.  9-6).  However,  in  an
                                     9-42

-------
Ames test with S. typhimurium TA1 535,  EDC  activated with rat liver microsomes,




NADPH, 0  and added  GSH,  did  not enhance mutagenic activity.  In contrast, the



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 mutagenicity.




     B.   Cytosolic reactions;  Heppel and  Porterfield  (19^8) 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.  Nachtomi 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 (Fig.  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 (I97la,b)  found small




amounts of S,Sf-ethylene-bis-cysteine in the  urine of mice injected  with EDC




(Table 9-15).   Nachtomi 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.   Nachtomi et al.  (1970) identified the  same two




compounds plus the sulfoxides  of the former in urine of EDC- and dibromoethane-

-------
    V-Y-
 CI-C-C-CI
        GT
        GSH
                -Cl
S-t2-chloro«thyl)-GSH
          HOH
          -cr
                         Ethylene
   GS\J
Episulfonium ion
                  HOH
                     *»GS-CH,-CH,OH
       I
        GSH
S42 hydroxyethyDGSH


            glutathionasa
                                                GS-CH,-CH,OH

                                                      Oxide
  GS-CH.-CH.-SG

  S.S'-ethylene bit-GSH
                               NH,
                        HOOC-CH-CH.-S-CH.-C^OH

                           S-<2 hydroxyethyl) cystalna
                                     Acetyl CoA Transferase
                        N-acetyl S-(2-hydroxyethyl) cysteine and oxide
Figure 9-7  . Cytosolic metabolism of ethylene dichtoride. Source:
     Adapted from Anders and Livesey (1930).

-------
treated  rats.    Nachtomi  et  al.  (1966)  have  also identified  N-acetyl-S-(2-




hydroxyethyDcysteine in the urine of  EDC-  and  1 ,2-dibromoethane-treated rats.




Jones and Edwards (1968) and Edwards et  al. (1970) confirmed the work of Nachtomi



with dibromoethane,  and moreover, isolated the sulfoxides of both S-(2-hydroxy-




ethyDcysteine 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  (Fig.  9-7)  has  been




suggested by Rannug  et al.  (1978,  1979, 1980) to be  responsible  for the mutagenic




action of EDC in S.  typhimurium TA1 535.  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-chloroethyDcysteine  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.



     C.   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-dibroraoethane by  a nonenzymatic  reaction  with



GSH.  The microsomal  P-H50  inhibitor, SKF 525-A,  had no effect on EDC metabolism,



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 (Fig.  9-7).  An analog



of this intermediate, S-(2-chloroethyl)  cysteine,  was nonenzymatically converted



to ethylene in the presence of glutathione and other thiols.


                                                        1 U
     D.   Oxidation of EDC to CO ;   After oral dosing of   C-EDC, Yllner (1971 a)



in the mouse and Reitz et al. (1982) in the rat found that 5-10? of the dose was



metabolized completely to CO  and water.  Yllner 0971 b) 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



COp,  Yllner  proposed that 2-chloroacetate,  arising from EDC  metabolism  (Fig.



9-5) is enzymatically hydrolyzed to glycolate by dehydrohalogenation, a portion



of which is further oxidized to oxalic acid.



     E.   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-chloroethyl)-



cysteine,  thiodiglycolic acid,  S-(2-hydroxyethyl)cysteine  and  its  mercapturic



are involved as end products.  Guengerich et al.  (1980) considered the possibi-



lity that reductive dechlorination of EDC would yield chloride ion  plus a chloro-



ethyl  radical,  which  could either  react with macromolecular targets, or lose  a



hydrogen  atom  to form vinyl chloride.  Alternatively, some other dehydrohalo-



genation  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 reduc-
                                      9-16

-------
tive  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 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 condi-




tions  produced only  30-U05t  inhibition  of  binding for EDC.  These investigators




concluded that the metabolism of EDC probably does not involve vinyl chloride as




an intermediate.




     9.1.3.4   METABOLISM, TOXICITY AND MODIFIERS — Dichloroethane metabolites,




chloroacetaldehyde,  chloroethanol (oral LD,_0  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 LD^ for rats,  770 mg/kg)  (Heppel  et  al.,  19^5,  19^6;  Woodward et




al., 19^1; 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  (Fig.  9-6).  However,




Yllner  (I97la,b)  found  that chloroacetic acid  also reacted extensively  with




sulfhydryl compounds in vivo.




     Heppel  et  al. (19^5, 19**6,  19^7) 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






                                     9-47

-------
acids, cysteine and methionine,  also protected young rats from inhalation expo-




sure.  This protective effect of sulfhydryl compounds is clearly related to the




marked depletion of glutathione levels which occurs in the livers of rats given



EDC, chloroethanol or chloroacetaldehyde (Johnson, 1965, 1966, 196?) and to the




enzyme pathways of metabolism of EDC  (Figs. 9-5  to 9-7).   Johnson (1965, 196?)




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 chloro-




acetaldehyde (Fig. 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 097*1, 1979). Like EDC,  vinylidene  chloride  is detoxified by




glutathione-dependent pathways.  Jaeger et al. 097*0 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  ct-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
                                      9-48

-------
was lowest at  6-10  pm  and  highest at 4-12 am; these  periods  correspond to the




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 cyto-




chrome  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 ) studied  also 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
                                     9.49

-------
fraction) added in vitro was accelerated  (up to 2.5-fold) by doses of ethanol up




to 4 mg/kg without causing any increase in P-450  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, rautagenesis 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 pharraacokinetic 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  (Fig.  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  (Figs.  9-6 and 9-7).   Reitz et al.
                                      9-50

-------
(1980, 1982) determined  total covalent binding and binding to DNA in rats exposed


   1 4
to   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 carcino-



genicity 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.   Nuclei  from  EDC-treated  mice



isolated 4 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  (0.05%  by



weight; an inhibitor of aldehyde dehydrogenase) which blocks the further oxida-



tion of bromoacetaldehyde  formed  in the metabolism  of  EDB (Fig.  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
                                 (1 50 mg/kg)              (150 ppm 6 hr)
Total Binding (n=4)

Liver
Kidney
Spleen3
Lung
Forestomacha
Stomach

DNA Binding (nr3)

Experiment 1

Livera
Spleen
Kidney
Stomach

Experiment 2
175
183
 65
106
160
 90
24
25
21
34
19
 2
268
263
130
14?
 71
156
45
48
22
16
19
29
21.3 ± 7.4
 5.8 + 0.7
17.4 + 2.3
   14.9
                   8.2 ± 3.3
                   1 .8 + 0.3
                   5.2 ± 3.7
                      2.8
Liver
Spleen3
Kidney
Stomach
13.9 + 2.1
2.5 + 0.3
14.5 + 6.2
6.7
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 4 hr after oral dosing or immediately following a 6-hr
inhalation exposure.


 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).
bReitz et al.,  1982.
                                  9-52

-------
 increase  of covalent binding.  Plotnick et al.  (1980)  showed  that  dietary  disul-




 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 microsomal and  cyto-




 solic metabolism  of EDC has  been demonstrated  in  vitro by several  investigative




 groups.   Van Duuren and his colleagues (Banerjee et al., 1978, I979a,b,  1980)




 observed  microsomal-activated covalent binding of both EDC and EDB to native DNA




 from salmon sperm, and to liver and lung microsomal protein  from rats and  mice.




 Binding to DNA did not occur in the absence of  microsoraes or in the  presence of




 denatured microsoraes.   Cytosolic  metabolism  produced  insignificant metabolic




 activation and binding.  The microsomal-activated binding was enhanced by  pheno-




 barbital  and  3-methylcholanthrene  pretreatment, whereas  the addition of gluta-




 thione 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   halo-




 ethylene  oxide, all of which are electrophilic  in nature.  Banerjee, Van Duuren




 and Kline 0979b) demonstrated that both 2-bromoacetaldehyde and 2-bromoethanol




 bind covalently to protein and DNA without metabolic activation.  Guengerich et




 al. (1980) have found that    C-EDC  is metabolized by both microsomes and  cyto-




 solic systems to metabolites  that covalently bind to  protein and calf  thymus DNA.




 Cytosolic metabolism  depended upon  the presence  of  glutathione  and  involved




glutathione transferases (Fig. 9-7),  although glutathione inhibited raicrosomal-




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 9-1 7  Effect of Dietary Disulfiram Upon the   V Content of Liver
  Nuclei  Isolated 24 or 48 Hours After Administration of a Single Oral Dose  of
                              1 5 mg/mg [U- •  C]EDB
Time Interval                Control                 Disulfiram


   24 hours                 68 7 + 82*                1 773 + 31 4

   48 hours                 460 + 42                 1534 + 197


•Results are expressed as dpm/pellet (mean + S.E.M.) of duplicate determinations
 on 6 animals per group at 24-hr and 5 animals per group at 48  hr.

From Plotnick et al., 1980.
                                  9-54

-------
9.1.4.    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 min.   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  min at high




doses.  The parameters of  total body elimination are compatible with a 2-compart-




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.




     Biotransformation of EDC  has been  shown to occur by  multiple pathways, in




both 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
                                     9-55

-------
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 mS,  (=1 U3-3571  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

-------
                                                                   TABLE 9-18


                                 Effects  Associated with Acute Lethal Oral Doses of Ethylene Bichloride in Humans
       Patient8
       Sex/Age
    Amount
   Ingested
     Symptoms and Signs
    Outcome and Findings
   Reference
  males/20 to 29 years
3 males/19 to 27 years
       a
Male/NS
Hale/63 years
1 50 to 200 mi
(=188 to 250.6 g)
70, 80 and 100 mil
(=«7.7, 100.2 and
125.3 g)
=62 mi (=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 U 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, 19«5
35 hours.  Punctuate
heraorrhaging in the epi-
cardium, pleura, and mucous
membranes of the stomach and
duodenum; varying degrees of
liver damage with focal
hemorrhaging in one case; yellow-
white fibrinous bundles of blood
in the heart cavities and lesser
circulatory vessels; hemolytlc
Jaundice of the endocardium,
aortal intima, and dura mater;
evidence of decomposition of
circulating erythrocytes
Deaths after 5 to 8 hours
Hyperemia and hemorrhagic
lesions (see text);
evidence of overt bleeding
into the visceral organs

Death after 6 hours
Hyperemia and hemorrhagic
lesions (see text)

Death after 22 hours
attributed to circulatory
failure.  Extensive
hemorrhagic colitis; nephrosis
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.
Kaira, 1966C
                                                                                                                      Noetzel,
Hueper and Smith,
1935

-------
                                                                   TABLE 9-18
Patient3
Sex/Age
Male/1 6 years
Amount
Ingested
50 ml
(=62.7 g)
Symptoms and Signs Outcome and Findings
Vomiting; epigastric pain; Death after 91 hours
muscle spasms, hiccups, rapid
Reference
Roubal, I947b
Male/80 years
Male/18 years
50 mfc
(=<62.7 g)
50 ml
(=62.7 g)
Male/57 years
DO mJ,
(~ 50.1  g)
pulse and lack of eyelid
response to light on the 4th
day

Elevated serum enzymes:  LDH;
SCOT; SGPT; alkaline phosphatase
glutamic dehydrogenase; RNAase.

Onset of symptoms after 1 hour:
somnolence and cyanosis.
Diarrhea after 4 hours.
Impaired blood coagulation
after 5.5 hours:  increased
prothrombin time; decrease in
clotting factors II and V;
thrombocytopenia; no increase
in fibrinolysis

Somnolence; vomiting; sinus
tachycardia; ventricular
extrasystoles; regained
consciousness after 14 hours;
dypsnea; loss of blood
pressure; cardiac arrest.
Impaired blood coagulation
observed after 21 hours;
prolonged bleeding from
venipunctures; reduction In
activity of clotting factors
II, V, VII and VIII; complete
defibrination; thrombo-
cytopenia; increased thrombin
time
Death within a few hours
Death after 17 hours
attributed to circulatory
shock.  Intravascular
thrombosis were not found.
Secchl et al., 1968'
Schonborn et al.,
I970b
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
subepicardial, subendocardial
and myocardial tissues.  The
coagulation disorder with
thrombocytopenia attributed
to disseminated intravascular
coagulopathy and hyper-
fibrinolysis
Martin et al., 1969

-------
                                                                   TABLE  9-18  (cont.)
Patient3
Sex/Age
Male/30 years
Amount
Ingested
1)0 mi
( = 50.1 g)
Symptoms and Signs
Slight cough; reddened
conjunct! vae; shock; weak,
Outcome and Findings
Death after 28 hours
Hyperemia and hemorrhagio
Reference
Garrison and
Leadingham, I954b
Male/50 years
Male/55 years
 (asthmatic)
Male/NS
Male/NS
=<30 ml
(=37.6 g)
20 mi
(=25.1  g)
                             =20 mi
                             (=25.1  g)
                             =20 ml
                             (=25.1  g)
rapid pulse.  Regained
consciousness after
3 hours; hyperactlvity
alternated with
semicomatose 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 1 0 hours
Diffuse hemorrhaglc
gastritis; bilateral
pulmonary congestion;
acute necrotizlng
bronohiolltis and
bronchitis; acute toxic
nephrosis; diffuse hepatic
necrosis; hyperemia of the
brain with scattered peri-
vascular hemorrhages in
the pons

Death after 21 hours
                                                         Death after 1 3  hours
                                                         Death within 12 hours
Lochhead and Close,
1951
Roubal, 1947°
                                Flowtow, 1952L
                                                                                         Flowtow, 1952

-------
                                                                TABLE  9-18   (cont.)
Patient*
Sex/Age
Male/1 U years
Amount
Ingested
15 mJi,
(= 18.8 g)
Symptoms and Signs
Onset of symptoms after 2 hours:
Progressive appearance of
Outcome and Findings
Death after 6 days
Extensive mid-zonal liver
Reference
Yodaiken and
Babcock, 1973
Male/32 years
8 mi
(=10 g)
Male/32 years
glass
severe headache; staggering;
lethargy; periodic vomiting;
decreased blood pressure;
oliguria; dypsnea; somnolence;
increased dypsnea; and oliguria,
hemorrhaglc nasogastric aspirate;
ecchymoses; sinus tachycardia;
cardiac arrest, pulmonary
edema; refractory hypotension.
Hypoglycemia on day 2 and
hypercalcemia on day 4.
Progressive decrease in blood
coagulation ability:  increased
prothrombin time; all clotting
factors except VIII were
markedly decreased on day U.

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
                                                                                      necrosis; renal  tubular
                                                                                      necrosis; focal  adrenal
                                                                                      degeneration and necrosis
Death after 56 hours
Bogoyavlenski et al.,
19686
Death on the 3rd day
Hyperemia and hemorrhagie
lesions (see text)
Agranovich,

-------
                                                                           TABLE 9-18   (cont.)
               Patient"
              Sex/Age
 Amount
Ingested
   Symptoms and Signs
    Outcome and Findings
                                                                     Reference
         Male/27 years
half glass
          Male/13  years
          (alcoholic)
4 drinks diluted
with orange Juice0
I
cr>
no
          Male/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; dypsnea; nausea;
cyanosis; depressed
respiratory rate; moist
rales; muffled heart sounds;
rapid pulse; extrasystoles;
anuria

Unconsciousness
Confusion; deep sleepiness;
unconsciousness; vomiting
with blood
Death after 19 hours
Hyperemia and hemorrhagic
lesions (see text); evidence
of overt bleeding into the
visceral organs
Bogoyavlenski et al.,
1968
                        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;
                        subconjunctival  hemorrhage;
                        oliguria;  paranephric hemorrhage.
                        5th day  pulmonary edema and
                        unconsciousness
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
                                                                  Morozov, 1958

-------
                                                                             TABLE 9-18 (cont.)
               Patient"
              Sex/Age
 Amount
Ingested
   Symptoms and Signs
    Outcome and Findings
                                                                                                                                   Reference
          Male/63 years
1  or 2 sips
 I
o>
OJ
          Male/1  1/2 years
          Male/79  years
          Male/2 years
          Male/23 years
1  sip
1  sip
1  sip
1 sip
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.  Hyperemia and
hemorrhagic lesions (see text)
Death the next day
Hyperemia and hemorrhagic
lesions (see text)

Death after 10 hours
attributed to heart and
circulatory failure.
Hyperemic 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 hemorrhagio
lesions (see text); evidence
of overt bleeding into the
visceral organs
Freundt et al., 1963°
                                                                                                                                Keyzer,
                                                                                                                                Weiss, 1957
Heinfried, 1953
Reinfried, 1953
           NS =  Not  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 U.S. EPA (1979) (Akimov et al., 1976,  1978;  Shchepotin and Bondarenko,  1978;




Bonitenko et al., 1974,  1977; Luzhnikov et al.,  1974; 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 consis-




tent 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,  1974, 1977; Luzhnikov et al.,  1974, 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
                                      9-64

-------
                                                                           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)
U1
         Oral
         Inhalation
211
 37
NR
NR x 20 to 30 minutes
                          110
Mild to severe poisoning characterized by
pronounced DCE odor on breath (91$), dry skin
(75$), hypotension (7W, extreme pupil dilation
(72$), mucosal cyanosis (67$), tachycardia (62%),
respiratory difficulty (59$). muscular
hypotonia (46$), decrease in tendon reflexes (46$),
loss of consciousness (12$).  Neurological syn-
dromes noted in 118 subjects:  comatose C*2$),
atactlc (12$), asthenic with autonomic vascular
Insufficiency (27$),  extrapyramidal (23$),
convulsive (7.W, psychotic (5$).  Lethality
not reported.

Aggregate findings for 218 total patients reported.
Neurological disorders (incl. unconsciousness,
respiratory depression) (100$), cardiovascular
insufficiency (incl., arrhythmias, hypotension,
reduced cardiac output, decreased peripheral
resistance) (60$), liver dysfunction (incl.,
enlargement, hyperbilirubinemia, increased
serum albumin and asparagine transaminase (35$).
Nephropathology (ollguria, proteinuria, 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 bromosulfalein, plasma bilirubin,
plasma glutamine - aparagine transaminase).
Clinical symptoms of poisoning were observed
at blood concentrations of 0.5 mg $, and coma
developed at 5 to 7 mg $.
                                                                                                       Akimov et al.,
                                                                                                       1976, 1978
Schepotln and
Bondarenko, 1978
                                                                                                                                  Luzhnlkov et al.,
                                                                                                                                  1976, 1978

-------
                                                               TABLE 9-19 (cont.)
   Route
              No. of Subjects
                              Dose
                                                                    Principal Findings
                                                                                                                             Reference
NSa
   160C
                                  NA
Oral
Inhalation
21
 3
10 to 100 mi (=12.5 to 125 g)
NR
Oral
               32
 Hemodynamic  shock due  to myocardial  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 decompensatory  (pronounced
 and  progressive hypotension, decreased cardiac
 output,  normal or slightly decreased peripheral
 resistance,  decreased myocardial  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 endothelial  cell edema,
 capillary lumen stenosis, edema of the
 myocardial interstices with leukocyte
 accumulation and microfocal hemorrhages,
 decreased glycogen, degenerative changes, and
 decreased mitochondrial enzyme activities.

 1 7 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 1 7 had hepatomegaly, icteric skin,  solera,
 and increased billrubin levels.  The other 10
 patients were comatose and had more pronounced
 symptoms of cardiac insufficiency.

Gastroenteritis and proteinuria.  Elevated
 leukocyte count and elevated alanine/aspartate
aminotransferase activities correlated with
severity of poisoning.   Coma was associated with
blood concentrations  of 15 to 30 mg J,  and
consciousness returned  at levels below 8  to
10 mg %.   Adipose tissue  and blood levels of
68  and  1.2 mg >,  respectively,  at autopsy.
                                                                                                                          Luzhnikov et al.,
                                                                                                                          1976, 1978
Andriukin, 1979
                                                                                                                          Bonitenko et al.,
                                                                                                                          1971,  1977

-------
                                                              TABLE  9-19  (cont.)
Route
Oralc
No. of Subjects Dose
7 children HR
(1 to 6 years)
Principal Findings
Clinical syndrome similar to that in adults:
severe and persistent vomiting (within 1 hour),
Reference
Hinkel, 1965
                                                                   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 hematologlc  changes.
                                                                   Typical pathological anatomical  findings.
                                                                   LD2Q reportedly ranged from 0.3  to  0.9 g/kg.
 Route is probably oral, but not specifically stated  in the EPA  O979)  summary of  this  study.

 The EPA summary of this study stated  that  "at least" 160  patients were studied.

cExposure to a. "nerve balsam" medicine that was  75t 1,2-dichlorethane  (other components not  stated  in EPA  summary).

-------
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, 19^3; Bloch, 1946; 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,  1948;  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 to poorly




characterized mixtures of EDC and other  solvents, or to 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  (Doraenici,  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 EDO




exerted a  direct  toxic effect  on the cardiopulmonary system or precipitated a




shock-like  state  which  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,  19^6;




Rosenbaum, 19^7; 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-
           i
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/m  ).   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/m ).

     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 Light*
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 Threshold3
(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
+0.1 5
no effect
-0.45
-0.25
-0.7
-0.75
-0.6
Source:  Borisova,  1957

aEye 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 was  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-minute 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, 197*0 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/nr), 6 subjects

                                    •3
perceived EDC  at 4.5 ppm  (17.5 mg/m ),  and  1 subject  detected the compound at


3 ppm (12.2 mg/nr).


     When diluted  in water,the  odor recognition threshold of EDC was determined


to be 29 ppm (v/v  water)  (Araoore 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 1$ 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  which 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 indus-




trial  exposures  (McNally and Fostvedt,  19^1;  Siegel,  19^7; Rosenbaum,  1939;




Watrous,  19^7;  Rejsek and  Rejskova,  19^7;  Delplace  et al., 1962; Suveev and




Babichenko,  1969).  In one study (Suveev and Babichenko, 1969), examination of  12




symptomatic workers that  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  (McNally and Fostvedt,   19JH),




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  0947)  discussed  experiences with cases  of EDC intoxication  in




Russian industries from 193^ to 19^5.  He  observed over  a  period of 10 years that
                                     9-71

-------
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 09^3) 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 we^e




chronically exposed  to  EDC for 7  and  9  months  during the manufacture of hexa-




chlorophene (Guerdjikoff,  1955).   EDC  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 *J times a day, the exposure concentration of EDC was about 120  ppm.  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 walking




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 44  to  H6% of  the total




exposure occurred during the gluing  operations,  when the TWA concentrations were




=28 ppm during application  and =16 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 4 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 ppra.
                                      9-76

-------
     Workers (total number not stated) who were engaged in the production of soft




tanks during the years  1951  to 1955  experienced  increased morbidity and  lost




workdays when compared with workers  in the entire factory (Table 9-21).  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.   Further




examination of 83 of the gluers revealed diseases of the liver and bile ducts (19




of 83), neurotic conditions  (13  of 83),  autonomic dystonia  (11  of 83), asthenic




conditions  (5 of 83), and goiter and hyperthyroidism (10  of 83).




     Visual-motor reactions were studied at the beginning and end of 1 4 workdays




in 1 7 of the gluers and 10 control machinists  (Kozik, 1957).   It was stated that a




device was 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  modified complex reaction test, errors were committed
                                     9-77

-------
                                                                  TABLE 9-21


                            Morbidity and Lost Workdays of Aircraft Industry Gluers Exposed to Ethylene Dichloride3

                                                               (Rates/100  Workers)
Tear

1 9 51 Cases
"•o Days
1
oi
1952 Cases
Days
1953 Cases
Days
1951 Cases
Days
1 9 55 Cases
Days
Total
Morbidity
Plant
120.2
995.8


121.0
960.9
135.6
1010.8
150.7
1175.9
127.6
978.1
Shop
159.8
1115.5


137.6
996.0
163.9
1236.5
19L8
1563.2
176.6
1162.1
Acute Gastro-
intestinal
Disorders
Plant
5.1
19.3


1.2
15.1
11.1
15.6
5.3
19.3
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
Radioulitis
Plant
5.2
99.9


5.0
HI. 8
7.5
67.3
7.9
73.8
5.9
51.1
Shop
13
127


9
91
16
116
16
182
10
90
.0
.0


.7
.5
.5
.0
.7
.8
.3
.2
Other
Diseases
Plant
31.1
351.2


31.0
335.2
35.3
338.3
10.8
386.1
37.9
315.7
Diseases of
the Muscles,
Liver and Gall Acute Chronic Tendons,
Bladder Diseases Gastritis" Gastritis" and Ganglia"
Shop
13
511


10
378
53
521
63
596
63
610
.2
.8


.8
.7
.5
.0
.8 21
.2 251.5
.3 21
.5 290
_ «• •
_


_
— _ -
.
— — -
13 6 18
61.5 15 170
838
27 11 50
"Source:  Kozik, 1957
 The incidences are reportedly elevated,  but reference  values  were  not  given.

-------
both before and after work by 1 5 of 1 7 gluers; U of 1 0 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 EDC ranged  from  =10 to 200 ppm (0.04 to




0.8 mg/£),  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/£), 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)  that  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 examina-




tions 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 1 0  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
64
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-DCEa                             Effect                                 Incidence


            62,  64 and  200 ppm             Dryness of the mouth; unpleasant sweet aftertaste;           6/10
                                             dizziness; lassitude; sleepiness; nausea;
                                             vomiting; poor appetite                                         .
                                           Burning sensation of the eyes that disappeared with          10/10
                                             adaptation to the atmosphere                                   .
                                           Epigastrium pain                                             3/10
                                           Insignificant tenderness of the epigastrium                  7/1 0
\o                                          Livers tender to palpitation with minimal enlargement        4/10
AD                                          Normal arterial blood pressure                               1 0/1 Q
                                           Relaxed pulse (60 to 65/minutes)                             6/10
                                           Intensified reflexes and autoraic neuroses                    3/10

            10  to  37 ppm                   Complaints similar to those associated with                  1/6
                                             exposure to 62, 64 and 200 ppm

            10  to  200 ppm                   Sweet aftertaste; dizziness; nausea;  vomiting; lack          6/42
                                             of appetite                                                     .
                                           Livers tender to palpitation with insignificant              3/42
                                             enlargement                                                     .
                                           Epigastrium pain                                             2/42
                                           Emaciation  (2 to 10 kg  below expected weight)                16/16
                                           Unremarkable opthalmologic examination                       1 6/1 6
                                           Unremarkable examination of upper respiratory  tract,         16/1 6
                                             lungs,  and heart
                                           Augmented reflexes and  vegetative neurosis                    3/1 6

-------
                                                       TABLE 9-23  (cont.)
          Concentration of 1,2-DCEs
                      Effect
Incidence
          10 to 200 ppra (cont.)
\)
Icteric skin coloration
Elevated urobilinogen levels

X-ray observable chronic catarrh of the stomach with
  atrophy of the mucous membrane
Periodic spasm of the pylorus
Moderate hyperchromic anemia
  (3,430,000 erythrocytes/cu ram and 60% hemoglogin)
Slight reticulocytosis (0.1  to 0.3%)
Diminished osmotic fragility of erythrocytes in NaCl
Slight leukocytes (11,200/cu mm)
Low platelet count (40,000 to 55,000 cu mm)
Decreased number of neutrophils (40 to 50%)
Slightly increased number of neutrophils (70 to T&%)
  with decreased number of lymphocytes (1 5 to 25%)
"Abnormal" distribution of white blood cells
Increased number of erythrocytes,  increased percentage
  of polyraorphonuclear neutrophils, mild stimulation
  of erythropoiesis with a less significant increase
  of leukopoiesis in the bone marrow
Elevated serum bilirubin (2.3 mg %)
1/16
"majority"
  of 16
6/16

3/16
1/1 3e

4/1 3e
6/1 3e
1/1 3e
2/1 3e
2/1 3e
6/1 3e

U/136
5/1 3e
                                                                                                       1/16

-------
                                                       TABLE 9-23 (cont.)
          Concentration of 1,2-DCEc
                      Effect
Incidence
CO
          10 to 200 ppm (cont.)
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 test
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
1/16
16/16
6/16
8/16
3/16
4/16
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)

           42 workers initially examined;  effects on  these workers prompted further examinations  of 16  workers  from
           one shift (see below).
          Q
           Heraatological examinations were performed  on  13 workers.

          f3 of the 6 with catarrh

-------
levels,  positive  Takata-Ara  liver function  teats,  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  09^7)  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  mg/S,)  in  air for  6 months  to




5 years.   "Many"  of  these workers  exhibited  non-specific functional  nervous




system    disorders  (e.g., red  dermographism,   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. 095*0  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 EDO to wash their skin.  A single atmospheric sample,  comprising 10




subsamples  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 better estimate




potential  exposure  concentrations.    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  9-24 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  hexachloroethane (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 60% 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 489 and 413



rag/kg, respectively.   In these  studies,  EDC was apparently administered in water




solution, and all mice died over a  48-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 >5QO 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

Effect of Acute Exposure to Ethylene Dichloride
Route
Oral
Oral
Oral
Oral
Oral
Inhalation
Inhalation
Species
rats
rats
mice
mice
(CD-1 , male)
mice
(CD-1 .female)
rats
rats
(female)
Number
80
6 /group
10
10
6
NS
NS
6 male or female
Sherman Strain
(100-150 weight)
1-6/group
Dose
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
0.189 g/kg (in water)
0.113 g/kg
1 000 ppm for 1 h
12,000 ppm for 0.1 h
0.2 h
Effect
LD50
LD50
10/10 dead
6/1 0 dead
no deaths
LD50
LD50
2/6, 3/6 or 1/6 dead in
11 day observation period.
No adverse effects
Adverse effect
Reference
McCollister et al.,
Smyth et al., 1969
Heppel et al., 1915
Munson et al., 1982
Munaon et al . , 1 98 2
Carpenter et al., 19
Spencer et al., 1951

1956




19

   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 ppm for 7.0 h
No adverse effect
Adverse effect
No adverse effect
Adverse effect
No adverse effect
Adverse effect
No adverse effect

-------
                                                                        -TABLE 9-24 (cont.)
 i
CO
CO
Route Species Number
Inhalation rats 20
22
HO
HO
51
1)4
32
414
22
10
30
10
11
21
10
32
31
32
30
20
33
20
20
Inhalation rats 20
15
15
12
13
mice 22
20
23
guinea pigs 1 1
12
rabbits 16
cats 3
hogs 2
raccoons 2
Dose
3000 ppm for 6 h
3000 ppm for 5 h
3000 ppm for 1 h
3000 ppm for 3 h
3000 ppm for 2 h
3000 ppm for 1 .6 h
3000 ppm for 1 .0 h
3000 ppm for 0.7 h
3000 ppm for 0.5 h
1 500 ppm for 8 h
1 500 ppm for 7 h
1 500 ppm for 6 h
1 500 ppm for 1 h
1 500 ppm for 3 h
1 500 ppm for 2 h
1 000 ppm for 8 h
1 000 ppm for 7 h
1 000 ppm for 6 h
800 ppm for 7 h
600 ppm for 8 h
600 ppm for 7 h
600 ppm for 5 h
300 ppm for 7 h
3000 ppm for 7 h
3000 ppm for 3.5 h
3000 ppra for 1 . 5 h
1 500 ppra for 7 h
1 500 ppm for 1 h
3000 ppm for 7 h
1 500 ppm for 7 h
1 500 ppm for 2 h
3000 ppm for 7 h
1 500 ppra for 7 h
3000 ppm for 7 h
3000 ppm for 7 h
3000 ppm for 7 h
3000 ppra for 7 h
Effect Reference
20/20 dead Spencer et al., 1951
22/22 dead
38 /UO dead
214/1)0 dead
6/51 dead
1/11 dead
1 /32 dead
1/414 dead
0/22 dead
7/10 dead
214/30 dead
7/10 dead
2/141 dead
1/2U dead
0/10 dead
20/32 dead
1 7/31 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
14/20 dead
0/13 dead
22/22 dead
20/20 dead
1/23 dead
114/11 dead
6/12 dead
12/16 dead
0/3 dead
2/2 dead
0/2 dead

-------
                                                                       TABLE 9-21 (Cont.)
           Houte
 Species
vo
A,
        Subcutaneous
dogs

oats

rabbits
        Subcutaneous
        Subcutaneous
                          rats
                          mice
                                          Number
        Intraperitioneal  guinea pig    I/group
                           (male)
6

NS

NS
                          albino rats   NS
10/group
1/group at
the lowest dose

10/group
                                                                   Dose
                               1 50, 300 and 600 mg/kg
                               1 dose
0.94 or 1.26 g/kg body weight

0.91 or 1.26 g/kg body weight

0.91 or 1 .26 g/kg body weight

0.91 or 1.26 g/kg body weight
0.3% 0.5, 0.75, 1.0,
 1.25 g/kg (in olive oil)
0.25, 0.38, 0.75 g/kg
 (in olive oil)
                                                                                                   Effect
                                                                                                                                 Reference
No effect on liver histology
 21 hours after injection.
No effect on serum ornithine
 carbamyl transferase (OCT)
 activity 21 hours later at
 1 50 or- 300 mg/kg.
Increased OCT activity at
 GOO mg/kg; death of 1 animal.

Death within 21 hours at
 higher dose; histology:  mild
 perilobular fatty degeneration
 in liver, swelling of tubular
 cells and mild hemorrhage In
 the kidney, lung edema his-
 tological changes more marked
 in dogs.
In dogs only: corneal opacity
 evident by 10 hours; necrosis
 of corneal endothellum; corneas
 became clear by fifth day.

0.75/kg:  50$ mortality;
0.38 g/kkg:  no deaths
                                                                                   Divincenzo and Krasavage,
                                                                                   1971
                                                                                                                           Kuewabara et al., 1968
0.38 g/kg:
0.25 g/kg:
8 Of mortality
no deaths
                                                                                                 Heppel et al., 1915
                                                                                                 Heppel et al., 1915
        h = hours

-------
followed by decreased excitement.  Progressive incoordination and salivation was




evident.  In 2*4 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 EDC.   The corneas of




the eyes in all five animals were opaque within 2*1 hours.




     9.2.2.1.2.     Dermal Exposure — Skin  absorption  LD    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  EDC 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




were  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. 0945) reported that a single  exposure to 3000 ppm EDC (12.4




mg/2,) 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.




     Heppel 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 (1 500  ppm) for a similar




duration (7 hours) was lethal to 6 of 1 2  guinea pigs and to less than one-fifth of




the rats (U 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^).




     Spencer  et  al.   (1951) subjected groups of 4 to  6  female rats  to varying




concentrations of EDC (200,  300,  1000, 3000 or 12,000 ppra) 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/3.)  for 7 hours to 12,000 ppm (48.6 mg/fc)  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 10-5^ rats after inhalation




exposure to EDC at concentrations ranging from 300 to  20,000  ppm 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  prothrombin




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 parenchyraa-




tous degeneration of the  epithelium to  complete necrosis accompanied by inter-
                                      9-92

-------
stitial edema, congestion and hemorrhage.   Changes ranging from slight conges-




tion 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 ppra.  Carpenter et al. 0949) reported




an inhalation LC,_0 in rats to be 1000 ppm in air for a 4-hour exposure.




     9.2.2.1.3-1     Central Nervous  System  and  Cardiovascular System.  Inhala-




tion of 3000 ppm EDC  (12.4  mg/P.)  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 ppra 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  0.4 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 0.1 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 1 0 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 0.5% 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

-------
                                                                  TABLE 9-25

                                                  Effect of Ethylene Bichloride on the Cornea
Route Species
Inhalation rabbits
guinea pigs
mice
rats
hogs
raccoons
vO
vo cats
VJl
Number of
Animals Dose
1 6 3000 ppn for 7 h
1 exposure
14
19
20
2
2
2
Effect Reference
No effect on eyes Heppel et al., 1944




Inhalation        red fox            1             3000 ppm  for  7 h
                                                  1 exposure
Inhalation        red fox       1  (same animal     1500 and  1000  ppm for  7 h
                                as tested above)   1  exposure at  each
                                                  concentration
                  raccoon            2

Inhalation        dog                6            1500 ppm  for 7 h,
                                                  1  exposure
Loss of appetite; barely
 detectable turbidity in eyes
 within 6 h; eyes were opaque
 2 d later; death 6 d after
 exposure.

No adverse effect on eyes
No effect on cornea of 1;
 faint turbidity in eye of 1;
 Intense clouding of both
 corneas In 4; histology;
 corneal edema, degeneration
 of corneal epithelium,
 infiltration of polymorpho-
 nuclear leucocytes into sub-
 stantia propria.
Heppel et al., 1944
Heppel et al., 1944
Heppel et al., 1944

-------
                                                               TABLE 9-25 (Cont.)
Route
Inhalation
Species
dog
Number of
Animals
10
Dose
1 000 ppm for 7 h ,
Effect
Symmetric turbidity In the
Reference
Heppel et al., 19 14
Inhalation
Inhalation
guinea pigs

rats

rabbits

dog
NS

NS

NS

 3
                                                  1  exposure
                                                  1 500 ppm for 7 h,
                                                  repeated exposure
                                                  1 500  pptn  for  7 h,  repeated
                                                  exposure
 corneas of 8 j up to 3 weeks
 required for partial
 regression.

Death in most animals
 after 4 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.
Heppel et al., 1944
Heppel et al. 1944
Inhalation
                  dog
                  10
Inhalation
                  oats              NS

                  monkeys

                  chickens

                  rodents (species,  NS)
              1000 ppm for 7 h,  5 d/wk,
              repeated exposure,
              length NS
                                1000  ppm  for  7 h,  5 d/wk,
                                repeated  exposure,  length  NS
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., 1944
                                                                                Heppel et al.,  1944

-------
                                                              TABLE  9-25  (Cont.)
Route
Subcutaneous






Inhalation
Speples
dogs
(puppies and
adults)

cats
rabbits
albino rats
dogs
Number of
Animals
6



NS
NS
NS
6
Dose
0.91 g/kg



0.91* g/kg
0.91 g/kg
0.91 g/kg
UOO ppm for 7 h, 5 d/wk,
Effect
Corneal opacity evidence within
10 hours; necrosis of corneal
endothelium; clearing by
fifth day.
No effect on cornea
No effect on cornea
No effect on cornea
Mild clouding during first
Reference
Kuwabara et al . ,



Kuwabara et al . ,
Kuwabara et al . ,
Kuwabara et al . ,
Heppel et al., 1?

1968



1968
1968
1968
W
                                                 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.
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 149 mg/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; 12%  (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 14-day study at levels of 4.9 and 49 mg/kg, which




represent 0.01  and 0.1  times the single dose  LD5Q 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



14-day exposure indicated that doses >49 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




11-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 1 4-day study were




not stated.   EDC did not alter the weights of selected  organs  (liver, spleen,




lungs, thymus, 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 14-day study, although the number of leukocytes was normal after 90 days of




exposure.   Other hematologic  parameters (hematocrit and hemoglobin evaluated




after 14 or 90 days, erythrocytes and platelets evaluated after 90 days),  coagu-




lation values  (fibrinogen  and prothrombin time evaluated after  14  days),  and




clinical chemistry  parameters  (lactic dehydrogenase, serum  glutamic-pyruvate




transaminase and blood urea nitrogen evaluated after  14 days)  were unaltered by




EDC exposure.  The hematological/coagulation/clinical chemistry parameters were



assessed in  10-12  and 16 mice/dose  in the 14-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 14 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,  spleen, lungs,  thymus  or kidneys) uptake of   Cr 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 400 ppm (1.62 mg/S,) and 100 ppm



(0.105 mg/£) for 7 hours daily,  5 days/week for 6 months (Spencer et al.,  1951).



In addition, 15 rats of both  sexes and 8 guinea pigs of both sexes were similarly



exposed to 200 ppm EDC (0.81  mg/fc) 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/Jl)



(see Table 9-26).  The  three rabbits  that were  exposed to MOO 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.  O945) 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

                                                       Effect  of Subchronlc Exposure to Ethylene Dichloride
              Route
                          Species
                                 Number
                                                       Dose
                                                                                            Effect
                                                                                                                            Reference
         Inhalation
                         rats
                              26               1000  ppm  (3.0  mg/fc)  7 h/d  x  5 d/wk   20  dead  within  15 exposures;     Heppel  et  al.,  1916
                              (average weight:                                         progressive  weakness  culmin-
                               198  g)                                                  ating in  inability to stand.
                                                                                     Histology  of  4  rats:
                                                                                      degeneration and proliferation
                                                                                      changes in renal tubular
                                                                                      epithelium;  chronic splenitis
                                                                                      in 20 rats;  pulmonary congestion
                                                                                      in 2  rats.
o
ro
Inhalation      rabbits        6               1000 ppm  (3.9 mg/Jl)   7 h/d x  5 d/wk   5 dead within  43 exposures
                              (average weight:                                         survivor  exposed  64  days
                               2480 g)
                                                                                                                             Heppel et al., 1946
        Inhalation
        Inhalation
                guinea pig


                guinea pig


                guinea pig
                        dogs
15                1000  ppm (3.9  mg/Jl)  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/Jl)  7 h/d x 5 d/wk   10 dead  within 2  exposures      Heppel et al.,  1946
(average weight:
 558  g)
16                1000  ppm (3.9  mg/Jl)  7 h/d x 5 d/wk   16 dead  within 4  exposures      Heppel et al.,  1946
(average weight:                                         lacrimation  and  marked
 900  g)                                                 inactivity during  exposure;
                                                        congestion of lung,  liver,
                                                        heart,  kidney, adrenal gland
                                                        and  spleens.
                              6 (7000-8300 g)   1000 ppm (3.9 mg/Jl 7 h/d x 5 d/wk    1 dead after 30 exposures
                                                                                    1 dead after 43 exposures
                                                                                       Heppel  et al., 1946

-------
                                                                      TABLE  9-26  (Cont.)
o
U)
Route
Inhalation
Inhalation
Species
cats
monkeys
Number
6 (2660 g
average weight)
2 (4880 g
average weight)
Dose
1000 ppm (3.9 mg/fc) 7 h/d x 5 d/wk
1000 ppm (3.9 mg/Jl) 7 h/d x 5 d/wk
Effect
2 dead after 43 exposures
congestion and fatty meta-
morphosis in liver of all.
1 dead after 2 exposures
1 dead after 43 exposures;
Reference
Heppel et al., 1946
Heppel et al., 1946
        Inhalation
                        cats
                               500 ppm 6 h/d x 5 d/wk for 6 wka
                                                                                             fatty degeneration of liver
                                                                                             and slight fatty changes in
                                                                                             kidney; focal myocarditis in
                                                                                             1 monkey.
                                     No deaths;  dilated  hearts;      Hoffmann et al., 1971
                                      blood  urea nitrogen  increased
                                      to 114 mg/100 mfc.
        Inhalation
rabbits
500 ppm 6 h/d x 5 d/wk for 6 wks
3/4 dead after 10-17 exposures;  Hoffman et al.,  19TI
 dilated hearts.
        Inhalation
guinea pigs   10
500 ppm 6 h/d x 5 d/wk for 6 wks
9/10 dead after 4-14 exposures;  Hoffman et al.,  1971
 apathy and weight loss, fatty
 degeneration and necrosis of
 myocardium, liver, kidney and
 adrenals.
        Inhalation
                        rats
                                      10
                                500 ppm 6 h/d x 5 d/wk for 6 wks
                                     All dead after 1-5 exposures;
                                      hyperemia of lungs;  fatty
                                      degeneration and  necrosis of
                                      myocardium,  liver,  kidney,
                                      and adrenals.
                                Hoffman et al.,  19 71

-------
                                                                    TABLE 9-26  (Cont.)
           Route
 Species
                                       Number
                                                             Dose
                                                                                                  Effect
                                                                                              Reference
      Inhalation
                      rats
      Inhalation
guinea pigs
 I

o
      Inhalation
      Inhalation
rabbits
                      dogs
              15 male
              1  female
              (average weight:
               167 g)
                                    23 controls
18 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
                  100 ppm (1.54 rng/d)  for
                   7 h/d x 5 d/wk for
                   60 exposures
400 ppm (1.54 mg/Jl) for
 7 h/d x 5 d/wk
400 ppm (1.54 mg/Jl) for
 7 h/d x 5 d/wk
                                400 ppm (1 .54  mg/Jl)  for
                                 7 h/d  x 5 d/wk for
                                 167-1 77 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 hlstologlcal
 change In 6 survivors of
 exposure.
3 deaths.

All dead after 97 exposures.
No hematologlcal changes.
                                     No deaths.

                                     No deaths; 6 adults: no
                                      effect on mean arterial
                                      pressure, bromsulfaleln
                                      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
                       Species
                 Number
                                       Dose
                                                                            Effect
                                                                                                                                 Reference
      Inhalation
      Inhalation
                      rats
                      rats
                                   15 male,
                                   1 5 female
                                   20 male,
                                   20 female
                                100 ppm  (1.62  mg/i)  7 h/d,
                                 5 d/wk
                                400 ppm  (1.62  mg/J,)  7 h/d,
                                 5 d/wk
 I
o
      Inhalation
guinea pigs   8  male,
              8  female
      Inhalation       guinea  pigs   2 male
                                                     100 ppm (1.62 mg/S.) 7 h/d,
                                                      5 d/ wk
                               400 ppm  (1.62 mg/d)  7 h/d,
                                 5 d/wk
Females died within 1 0
 exposures.
Males died within HO
 exposures.

60% 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
 prothrombin clotting time.

Males died within 10
 exposures.
Females died within 24
 exposures.

Sacrificed after 1, 3, 4, and
 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 testes; increased
 BUN and increased blood non-
 protein nitrogen levels; no
 change in serum phosphatase
 or plasma prothrombin clotting
 time.
Spencer et al., 1951
Spencer et al.,  19 51
                                                                                                                          Spencer et al., 19 51

-------
                                                              TABLE  9-26  (Cont.)
     Route
                 Species
Number
                      Dose
       Effect
       Reference
Inhalation      rabbits
                              2 male,
                              1 female
               100 ppm (1 .62 mg/Jl)  7 h/d,
                5 d/wk for 165 exposures
No effect on general
 appearance, behavior;
 mortality, body weight,
 histology of liver, kidney,
 lungs, heart, spleen or testes;
 organ weights, blood non-
 protein nitrogen.  BUN,
 serum phosphatase, plasma
 prothrombin clotting time.
Spencer et al., 1951
_^    Inhalation       monkeys
o
                              2 males
               400 ppm (1.62  mg/Jl)  7 h/d,
                5 d/wk
One sacrificed in moribund
 condition after 8 exposures:
 enlarged liver with in-
 creased neutral fat and
 esterified 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 hematological
 change.
Spencer et al.,  1951
Inhalation      rats
                              12 male
                               Osborne-Mendel
                              (average weight:
                               72 g)
                              12 controls
               200  ppm  (0.73  mg/Jl)  7 h/d  x  5 d/wk  8  dead  after  6 exposures
                                                    No deaths.
                                Heppel et al., 1946

-------
                                                                   TABLE 9-26  (Cont.)
          Route
Species
                                      Number
                                                           Dose
                                                                                                Effect
                                                                                                                                 Reference
     Inhalation
                     rats
 I
o
             1  male Wlatar
             11  female Wistar
             (average weight:
              219 g)
200 ppm (0.73 mg/O 7 h/d x 5 d/wk
                                   11 controls
7 dead; 1  after 73 exposures,
 6 after 11 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
Inhalation


Inhalation
                     mice
                     guinea pigs
                                   20
             12 male,
             2 female
             (average weight:
              376 g)
                                   18 controls
                               200 ppm (0.73 mg/Jl) 7 h/d x 5 d/wk   18  dead after 7 exposures.       Heppel et al.,  1916
200 ppm  (0.73 mg/Jl)  7 h/d x  5 d/wk
14 dead after 88 exposures;
 1 dead after 115 exposures.
Histology of 9 guinea pigs
 killed after 121 exposures:
 pulmonary congestion in 1,
 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.,  1916

-------
                                                                    TABLE 9-26  (Cont.)
           Route
 Species
                                       Number
                                                             Dose
                                                                                                  Effect
                                                                                                            Reference
      Inhalation
rabbits
                                    (average weight:
                                     2720)
                                      controls
                  200  ppm (0.73 mg/J,)  7 h/d x 5 d/wk
                                     No deaths.
                                     No histological changes
                                      found; no effect on red
                                      and white blood cell
                                      count, hemoglobin, and
                                      differential counts
                                      compared with controls.
                                     No deaths.
                                                                                                     Heppel et al., 1946
      Inhalation
monkeys
'-X)
 i
2 males
(average weight:
 8710 g)
                                    no controls
200 ppm (0.73 mg/fc) 7 h/d x 5 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
15 male,
1 5 female
      Inhalation
guinea pigs   8 male,
              8 female
200 ppm (0.81  mg/JO 7h/d,
 5 d/wk for 1 51  exposures
                  200 ppm (0.81  mg/fc)  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.,  19 51
                                     Final body weight
                                      significantly different
                                      from controls In males, and
                                      growth less than controls,
                                      but not sigifleant in females.
                                      Significant increase in male
                                      liver weights only; slight
                                      parenchymatous hepatic
                                      degeneration in half of
                                      guinea pigs of both sexes;
                                      no effect on hematology; slight
                                      increase in trial lipid,
                                      phospholipid, neutral fat,
                                      free and esterifled cholesterol.
                                Spencer et al., 1951

-------
                                                              TABLE 9-26  (Cont.)
     Route
 Species
                                 Number
                                                      Dose
                                                                                           Effect
                                                                                                            Reference
Inhalation
rats
15 male,          100 ppm (0.405 mg/Jl)  7 h/d,
1 5 female          5 d/wk
                   males:  151  exposures
                   females:  142 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
                                      prothrombin clotting time,
                                      total levels llpid, phospho-
                                      llpids, neutral fat in liver,
                                      free and esterified
                                      cholesterol in liver.
                                Spencer et al.,  19 51
Inhalation
guinea pigs
8 male,
8 female
Inhalation
rabbits
Inhalation
monkeys
2 male,
1 female
2 male
100 ppm (0.405 mg/1) 7 h/d,
 5 d/wk
 males:  121 exposures
 females:  162 exposures
100 ppm (0.405 mg/fc) 7 h/d,
 5 d/wk for 178 exposures
100 ppm (0.405 mg/J,) 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 prothrombin clotting
 time, gross and microscopic
 histology, total liver lipid
 phospholiplds, neutral fat
 in liver, free and esterified
 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.,  19 51
Spencer et al., 1951

-------
                                                                     TAPLE 9-26 (Cont.)
            Route
 Species
                                       Number
                                                             Dose
                                                                                                  Effect
                                                                                                            Reference
       Inhalation
                       rats
^     Inhalation
guinea pigs
       Inhalation
                       mice
              16 female
              23 male
                  100  ppm  (0.42 mg/i)  7 h/d  x  5 d/wk
                   for 4 months
10 male
 6 female
                                     30  control
              19 Juveniles
100 ppm (0.12 mg/J,) 7 h/d x 5 d/wk
 for 4 months
                  100  ppm  (0.42 mg/Jt)  7 h/d  x  5 d/wk
                   for 19  exposures
No deaths, no effect on rate
 of growth in 1 5 males
 compared with 1 5 male
 controls.
15 of 1 6 females became
 pregnant; rat pups were
 unaffected by exposure.
No histological effects in
 liver,  heart, lungs, kidney,
 adrenal glands and spleen of
 10 rats examined.

2 dead (disease of neck with
 enlarged caseous glands)
No histological effects in
 liver,  heart, lungs, kidney,
 adrenal glands and spleen of
 10 animals examined.
3 dead (disease of neck with
 enlarged caseous 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  17 wks
                                     No clinical symptoms
                                     No effect on serum levels of
                                      creatinlne, 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.  0946)  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 177 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 1 6 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 4 cats,  4 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  1/H  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 1




hours,  or  to  3000  ppm  for  2  hours/day,  5  days/week for 90  days  (Lioia,




I959a,b,c,d).   The only  significant changes observed  after the  acute 4-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/£) 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 1 0 to 1 5 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 1 0




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 measure-
                                     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 1 50 ppm do 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


1 50 ppm EDC group.  Although changes were not apparent  at  6 months, at 18 months


there was a significant decrease observed only at 5 and 1 0 ppra.


     No  treatment  related effects  were  found  on  levels  of y-glutamyl trans-


peptidase   (y-GT),  serum  glutamic-oxalacetic  transaminase   (SCOT),   serum


glumatic-pyruvic transaminase (SGPT),  serum  alkaline phosphatase,  bilirubin or


cholesterol (Spreafico et  al., 1931).   A slight, but not significant, increase in


creatine phosphokinase (CPK)  level was seen at 18 months in males exposed to 50


and  1 50  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
                                     9-114

-------
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.  SGOT  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 1 50 ppra.   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 1 U 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^jjases  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 ppm 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 endpoints.  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 subthreshold, threshold and above threshold odor perception concen-




trates.  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 9-24, 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  ppm (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 ppm 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 ppm was attributable to organ injury rather than




depression of the CNS.



     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 ppm



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 ppro  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  (1959)  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  (1947)  examined  100 workers who were




exposed to <25 ppm  for 6 months  to  5 years and found disturbances that included




"heightened  lability"  of the autonoraic  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,  19^7;  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 EDC for 7 hours/day, 5 days/week for U-6 months have shown  no




treatment-related adverse  effects  on survival,  growth,  hematology,  clinical




chemistry, organ weights or histology (Heppel et al.,  19^6; 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., 19^6).  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  ppm 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., 1U-
                                     9-119

-------
months-old) rats that were similarly exposed to 50 or 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.  (19*16)  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. (1951 )




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.  (1951).   An




unequivocal frank-effect  level (PEL) of 400 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., 1946; Spencer et al., 1951).  Spencer et al. (1951) reported that exposure




to 400 ppm EDC had no effect on rabbits (as reported  at 200 ppm), but  Heppel et




al.   (1946)  observed some mortality at  this  level.   Exposure to  500 ppra 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 m£  have been  reported  to be  lethal (see Table




9-18).  This is equivalent to  =140-3570 rag/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.f 1969) and mice (Heppel et al., 19*15) have been reported in the range of



700 mg/kg.  Higher oral doses  (up  to 2.5 g/kg) 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  of  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  ^7 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 mg/kg/day,



but not in  females  exposed to 1U9 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,  EDC)  on




the  reproductive ability  of  rats  and  effects  on their  offspring  (Murray et




al.,  1980).    The 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 initally 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 then were successively bred  to produce




two  litters,  the F. and F.._.   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  day/week) except  for day 21 of gestation




to  day 4 postpartum to allow for delivery and rearing of the young.   A  second




mating,  for  the production  of  the  F   generation took  place after the FIA




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  accomodations  (males were  caged away  from  females)  and  not to any




greater   suceptibility  of  the  males   for   developing  infection   after  EDC






                                     9-122

-------
exposure.   All 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 was  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,  I1*  or  21,  were 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
                                  9-123

-------
not affected.   There were  sporadic,  statistically significant  differences  in




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  incidence  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,  Riddle  and Borzelleca  (1981)  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 males  and 30  female  mice




(parental genration F/0) were mated to produce the first set offspring (F/1A).




After the F/1A  were weaned,  the  F/0 adults were remated  to produce the second




set of offspring (F/1B).   A parental  stock was chosen from the F/1B litter (30




females,  10 males)  to produce  a  second generation  of  offspring (F/2A).    After




weaning  the F/1B  adults were remated  to  produce  offspring (F/2B) for  use  in




teratology and  dominant lethal screening tests.




     In  this  study, the  EDC (Aldrich  Chemical Co.  99% pure) was dissolved in a




1/t  solution  of Emulphor  EL-620  (GAP Corp.,  Linden,  NJ) and  then   further




diluted  with  deionized water to  concentrations of 0.03,  0.09,  and 0.29  mg/ml




or a nominal  dose  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/ml p-dioxane  dissolved in  1.0J Emulphor




solution).  Fresh  drinking solutions were prepared twice weekly and placed in




amber  glass bottles  with  cork  stoppers  and  stainless steel  drinking tubes.
                                  9-124

-------
The authors  reported no decreases  in fluid consumptions in  either  the EDC or



the solvent control groups.



     Lane  (1981)   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  P/1A,  F/1B or F/2A  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  was no  difference  in the  litter  sizes  at birth,  pup body



weights, survival  of  pups  at days 4 and 21 of birth.  There  was  a decrease in



the survival indices  for  the F/2A generation  as compared  to values  for  the



F/1A and  F/1B 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 as well as decreased were observed in



the ratios of dead  to live. fetuses.   In  the  teratology  screening,  continous



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/1C  skeletal  specimens were



lost and, therefore, these effects could not be evaluated.



     From  this study,  (Lane et al.,  1981)   it  is  not possible  to conclusively



determine  whether  EDC has  the  potential to  cause adverse reproductive  or



teratogenic  effects  because the  doses  were  not   high  enough  to produce  any



adverse  effects,   including maternally  toxic  effects.    However,  under  the



conditions of the experiment,  it  appears that  EDC  given  in drinking  water
                                 9-125

-------
(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  by  inhalation to either  100 or 300  ppm EDC (Lot  Nos.  TA0201851L and




TA022851L,  analyzed  as >99.95&  pure by  the Dow  Chemical  Laboratories).   The




rats were  exposed 7 hrs/day on days 6  through  15 of gestation.   The rabbits




were exposed 7  hrs/day on days 6  through  18 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, threee rats




delivered  early  on  day 21;  however there were  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  or




resorptions of 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 weights 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, 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.
                                 9-1 26

-------
     In this  teratology study (Schlachter  et  al.,  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,  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 that 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  but 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  occurances



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  estrus  cycle,  changes   in  the  duration  of



various  stages  of  the  estrus  cycle,  increases  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



concentration in  the  placenta,  amniotic fluid  and  fetal tissue.   However, it



is difficult  to  evaluate or  seriously  consider these  reports since  they are



presented    with    insufficient    detail   for   critical   scientific   review.
                                  9-127

-------
Inadequacies  in  these  reports  include:   lack   of  original  data,   various



techniques and  tests mentioned but  results not  presented,  no  information  on



the source of purity of the chemical, statistical  analysis  mentioned,  but the



type of statistical tests not stated.



     The  above  comments  apply  similarily  to  the  study by  another  Russian



scientist, Urusova (1980),  who  reported that the  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  inadequencies   in  reporting.    These



inadequencies  include:     lack  of  details  concerning  the  numbers  of  women



involved  in the study,  no  data  on  women's age or physical status, no reporting



on the  concentrations and  duration of  EDC exposure after dermal exposure,  no



information on  the  repeatability  of this  finding  and  no information on number




of samples analyzed.



     Because of the  concern for EDC contamination  in  mother's milk,  Sykes and



Klein  (1957)  conducted a  study to  determine  whether oral  administrations  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 cows 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 concentration of EDC



than  the  controls,  but because  of the  small  sample  size,   and the  lack  of
                                   9-128

-------
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  injested  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.  The  available  scientific  information  on the potential  of



ethylene  dichloride  (EDC) to adversely affect  reproductive or developmental



processes   includes   a   teratoglogy  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.,  1981)  indicate that  EDC has



little  potential  for  producing  adverse  reproductive   affects or  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
                                  9~129

-------
since  only  a  few  animals  were  used  and  the  biological  effects  of  EDC-



contaminated milk were not investigated.



     In  conclusion  the  available studies  indicate  that the  EDC  has  little



ability  to adversely  affect  the  reproductive or  developmental  processes  in



laboratory animals  except at maternally  toxic levels.   However,  it should be



noted that these  studies have inadequacies which weaken the  strength  of this



conclusion.    The  multigenerational  study  by  Lane  et  al.,  1981, does  not



include  a  range of doses which 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  system,   this cannot  be



conclusively  established, especially  for humans, without  additional studies.



The  chemical similarity  of  EDC to  ethylene  dibromide  (EDB)  and  1,2-dibromo-



chloropropane  (DBCP)   might  suggest  that   additional  testing   related  to



testicular toxicity  would be appropriate.
                                   9-130

-------
9.1.  MUTAGENICITY



     Ethylene dichloride  (EDC)  has  been tested for mutagenic  activity  in bac-




teria, plants,  Drosophila, mammalian cells in vitro, and 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 and 1979,  Fabricant and Chalmers 1980,  Rannug 1980, or Simmon 1980).




GENE MUTATION STUDIES




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  without




metabolic  activation  and  stronger  positive responses  with  exogenous  hepatic




metabolic activation,  indicating that EDC is weakly mutagenic 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, and 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, (Brem et al.  197M) an analysis of duplicate experiments  carried out on at




least three different occasions revealed a twofold increase in revertant counts




for strains  TA1 530  and TA1 535  (mean  values of 50 and 5^ revertants on treated




plates versus 23 and 26 in control plates for TA1  530 and TA1 535, respectively).




No difference in revertant counts was noted  for strain TA1 538.  This response is




consistent with that expected for an alkylating agent.  The authors stated that




plate incorporation  tests could not  be performed because of the volatility of the




test agent.  Positive and negative control tests were conducted  for these experi-
                                     9-131

-------
                            TABLE 9-27



Summary of Mutagenioity  Testing  of EDC:  Gene Mutations in Bacteria
Test Activation Chemical
Reference System Strains System Information
Brem et al. Salmonella TA1 535 None 10 nmoles on filter
1971 (spot test) TA1535 disk
TA1538
^ Source: Not given
^ Purity: Not given
Results
Weak positive

Comments
1 . Could not perform
plate incorporation
tests because of
volatility.

Salmonella revertants
TA1 530
EDC 50
Water 23
Chloramphenicol 20
TA1 535 TA1 538
5U 19
26 19
31 m




-------
                                                                       TABLE 9-27 (cont.)
vo
 I
CO
CO
Test
Reference System
Principe et Salmonella/
at . 1981 mammalian
mlcrosome
assay (spot
test)




S. coelicolor
forward mutation
assay to Strr



Strains Dose uSVPlate
-S9
0 100
TA1 535 39 46
TA1 537 17 8
TA1 538 19 12
TA98 82 83
TA100 188 188

•P<0.01
Dose lilt/plate
0
2
10
20
100

0
33
10
21
77
171


Survival
100
100
100
100
100
+S9
100
103»
8
27
84
169


Comments
1 . Positive controls indicated system
working properly.
2. Positive results In Salmonella in
test with TA1 535.



spot

3. No precautions taken to prevent excessive
evaporation of EDC and insure adequate
exposure.
4. Toxicity results indicate exposure
minimal.


was

J Strr/plate
2.5
0.2
0.7
0.7
1 .0
+ 0.6
+ 0.2
+ 0.4
+ 0.4
+ 0.8





8-AG/Plate
A. nldulans
forward mutation
to 8-AGr
0
250
500
100
100
42
2.5
2.0
1.0
-f 0.9
+ 1 .1
+ 0.7




-------
                                                                   TABLE 9-27  (oont.)
Reference
McCann et al.
1975










Test
System
Salmonella/
microsome assay
(plate test)









Activation Chemical
Strains System Information
TA100 PCB-induced Concentration Tested:
rat liver S9 1 .3 x 10 (ig/plate
mix (13 junoles)


Source: Aldrieh
Chemical 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 rever-
tants/nmol)
in TA100
Comments
1 . Non-mutagenic or
extremely weak
mutagen in this study
Reproducible dose-
response curves not
obtained .

2. Metabolic activation
did not Increase
positive response.

3. Chloroethanol and
CO
-tr
Rannug 1976
Salmonella/
microsome assay
(plate test)
TA1535    Liver fractions
          from Sprague-
          Dawley or R
          strain Histar
          rats Induced
          with phenobar-
          bital with and
          without NADPH-
          generating system
          and with and
          without gluta-
          thione S-trans-
          ferases A, B
          and C.
Concentration tested:
Up to 60 iimol/plate

Source:  BDH Chemicals, Ltd.

Purity:  Not given but
         reported to be
         checked by glass
         capillary column
         chromatography
         using a flame
         ionization detector
Marginally
positive
without activation
(twofold increases);
positive response
with activation
(tenfold increases).
Spontaneous back-
ground 8-11
revertants/plate.
chloroacetaldehyde
(two putative
Intermediates  in  the
metabolism of  EDC in
mammals) tested
positive (I.e., 0.06
and 746 revertants/
(imol, respectively).

EDC activated  by  the
liver cytosol
fraction, Mixed-
function oxygenases
not involved.

NADPH-independent GSH
S-transferase  dependent
activation.

Strain differences noted
in ability to  metabolize
EDC.

Thought that mugateniclty
of EDC after activation
caused by formation of
highly reactive half
aulfur mustard,
S-(2-chloroethyl)-L-
cysteine.

-------
                                                                   TABLE 9-27 (cont.)
Reference
Test
System
Activation
Strains System
Chemical
Information
Results Comments
Rannug and Ramel
1977
Salmonella/
microsome assay
(plate test)
                                          TA1 535
Rannug et al.
1978
Salmonella/
mlcrosome assay
(plate test)
TA1 535
                                 S9  mix  from
                                 livers  of  un-
                                 induced male
                                 R strain
                                 Wistar  rats
                                 plus  NADPH
                                 generating
                                 system
Rat liver
microsome
system with
and without
NADP
                            Concentration tested:
                            Up to M5 vimol /plate

                            Source:   BDH Chemicals,  Ltd.

                            Purity:   Not given
                                                                     Concentration  tested:
                                                                     i»5  ninol/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.
                                                 Positive response
                                                 (two fold increase
                                                 without activation;
                                                 nearly tenfold
                                                 increase with
                                                 activation. )
                                                 Negative controls
                                                 yielded roughly
                                                 1 5 revertants/
                                                 plates.
                                                                                  Marginal  positive
                                                                                  response  without
                                                                                  activation (twofold
                                                                                  increase  21.8  +
                                                                                  3.06  at 15 nmol
                                                                                  v.  13.0 + 1.76
                                                                                  control);
                                                                                  positive  response
                                                                                  with  activation
                                                                                  (10-fold  increase)
                                                                                  Independent of
                                                                                  presence  of NADP.
Compared mutagenicity
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
response was
observed.

Activation of EDC
tar dependent on
NADPH.  EDC activa-
tion independent
of NADPH.

EDC major component
of EDC tar.
Mutagenlcicity of
tar increased with
metabolic activation
only with addition
of NADP.

Mutagenicity of EDC
tar not solely due
to EDC.

-------
                                                                         TABLE 9-27 (cont.)
Test
Reference System
Rannug and Beije Salmonella/
1979 isolated
perfused
rat liver



Salmonella/
(plate test)
Activation Chemical
Strains System Information
TA1 530 Isolated Concentration tested:
TA1535 perfused liver 0.1 mH (1.3 mMoles)
from male R for up to !* hours.
strain Wistar
rats.


TA1 535 Bile from male 80 mg/kg EDC i.p.;
CBA nice. removal of liver and
Results Comments
Positive. Highest 1 . Positive
response 1 5-60 responses consis-
minutes after tent with conju-
addition of EDC gation of EDC
CJ5-60 revertants with glutathlone.
compared to 7-1 0
in controls).
Positive. Greater
than twofold

-------
                                                                         TABLE 9-27  (oont.)
vo
 I
Reference
Nestman et al.
1980









Test
System
Salmonella/
mlcrosome assay
(plate test and
and desiccator
exposure) .






Strains
TA1 535
TA100
TA1 537
TA1538
TA98






Activation
System
PCB-induced
rat liver S9
mix.








Chemical
Information
Concentration tested:
Up to 9 mg/plate (91
(imoles) In desiccators.
10 mg/plate in plate tests.

Source: Chem Service
Purity: Not given
Solvent: DMSO



Results
Negative in
standard test.
Positive in
desiccator testing
in strain TA1 535.






Comments
1 . Stated that
maximum yield with
TA100 is 20
revertants above
background (i.e.,
negative). For
TA1 535 a doubling
of mutant colonies
observed. No
other data pre-
sented.
      Stolzenberg and
      Mine,  1980
Salmonella/
microsome
assay (plate test)
TA100     PCB-induced
          rat liver 39
          mix (2 mg
          protein/0.5 mfc).
Concentration tested:
Up to 10 (imoles/plate
Negative
                                                                           Source:
                                                                                    Aldrioh
                                                                                    Chemical Co.
                                                                           Purity:  99< pure
2.  Cannot adequately
    evaluate results.

1 .  All compounds
    tested in trip-
    licate with and
    without S9 mix.

2.  Experimental
    values minus
    background
    revertants.
Revertants
umoles/plate -S9 +S9
10-1
1
10

0
0
15
Toxic
0
0
No growth


-------
                                                                        TABLE 9-27 (cent.)
Reference
Barber et al.
1981
Test
System
Salmonella/
microsome assay
(vapor exposure)
Strains
TA1 535
TA100
TA1538
TA98
Activation
System
PCB-induced
rat liver S9
mix.
Chemical
Information
Concentration tested:
Up to 231 .8 (imole/plate
as determined by GLC
analysis of distilled
water samples.
Results
Negative in
standard plate
test. Positive
in desiccator
testing in strains
TA1535 and TA100.
Comments
1 . Bacteria exposed in
gas tight exposure
chambers .
2. Plastic plates
found to absorb
VO
I
                                                                          Source:  Eastman Kodak Co.
Purity:  99.98J
dibromomethane
in parallel
experiment.  Thus,
glass plates used
for all other
testing.

Weak positive
result.  0.002
revertants/nmole
TA1 535 with or
without activation.
0.001  revertants/
naiol TA1 00 with or
without activation.

Revertants selected
from each experiment
and tested to ensure
that they were
actually his .

-------
merits and indicated the systems were working properly.  Principe et al.  0981)




conducted a spot test using strains TA1 535, TA1 537, TA1 538,  TA98,  and TA100.  A




positive response was  observed  for TA1 535 when a  triangular  shaped paper disc




soaked with 1 00 \il EDC was placed on the agar in the  presence  of 39  from Aroclor




1254-induced rat liver (i.e., 103 revertants on the treated plate vs. 33 rever-




tants for the  negative control). " Negative responses were obtained with the other




strains.   A negative response was  also  obtained   in  plate  incorporation tests




conducted with TA1 00  and  TA1 535 at doses up to 100 iiSVplate.  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 1 00 and




500 ufc/plate in plate incorporation and spot tests.  There was a 100? survival in




the test conducted with S. 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 TA1 525,  TA100, and TA98 to




EDC (Aldrich,  stated to be highest  purity available) concentrations as high as 13




mg/plate (131  umoles/plate).   A weak positive 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  125*0  did not




increase the weak positive response.  Chloroacetic  acid, which is a  known mamma-




lian metabolite of EDC, and the putative intermediates  (i.e., chloroethanol and




chloroacetaldehyde see sections 9.1.3.3A),  were also tested for their mutagenic




potential.   Chloroacetic acid was  negative, but  chloroethanol yielded a weak




positive and  chloroacetaldehyde a  strong  positive result (0.06  and  7^6 rever
                                     9-139

-------
tants/nmol, respectively,  in  TA100).  The authors speculated that the weak rauta-




genic response of EDC may  be  due to relatively inefficient conversion of chloro-




ethanol to chloroacetaldehyde in the in vitro system.




     Two reports  (Rannug  1976;  and Rannug  and  Ramel 1977) compared  the muta-




genicity 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 EDC  tar because  ethylene dichloride 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 TA1 535  was dosed up to  45 ^moles/plate in the presence




or absence  of  an exogenous  metabolic  activation  system  (from livers  of  male




strain R Wistar rats) with and without NADP.  Weak positive (twofold)  increases




in revertant  frequencies  over background (13.0  + 1.76)  were observed  in  the




plates  receiving  the  highest  dose (24.8  + 3-06)  in tests  conducted  without




metabolic activation.  However,  with activation,  a strong positive response was




observed.  The revertant  count was elevated tenfold  over the  spontaneous level




(132.8  + 8.35  v.  15.5 +  1.21).   The  response  to EDC  was independent  of  the




presence of NADP;  in constrast,  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 weak positive response if tested alone, while  in  fact  a strong response




was observed for the EDC tar.




     Following these observations,  Rannug et al. (1978) tested Salmonella strain




TA1 535 with EDC (BDH Chemicals Ltd.; although the purity  was  not  reported,  the




material was checked by glass capillary chromatography using a flame ionization
                                     9-1 MO

-------
detector) at doses up to 60 (imoles/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) hypothesize that the




mutagenicity of EDC after metabolic activation is 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  ^moles/plate,  12.8  ±1.5  and 176.8  +11.1



revertants were  observed  per plate  after   treatment  with EDC  and  S(2-chloro-




ethyl )-L-cysteine, respectively.   At  5.0 umoles/plate the  observed  numbers of




revertants per plate were  13.0 + 1.1  and 195*1 (only  one  plate tested), respec-




tively.   Thus,   S-(2-chloroethyl)-L-cysteine gives   a  strong  direct  mutagenic




effect at low doses where  no effect can be  seen  for  EDC.




     In intact mammals most GSH conjugates are normally excreted in bile.  Rannug




and Bieje (1979)  reasoned that if EDC were metabolically activated by conjugation
                                     9-141

-------
with glutathione to form a half sulfur mustard, mutagenic  products  in the bile




would  be  produced in  EDC-treated mammals  or  perfused livers.   To  test this




hypothesis, an EDC concentration of 0.1  m£ (1.3 mmoles) were perfused through R




strain Wistar  rat livers for up  to 4 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 U




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




TA1 535 for  plate  incorporation  tests.  Positive  responses were obtained.  The




greatest  response (45-60 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  TA1 535 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 Hine 1980,  and Barber  et al. 1981).  The




maximum  doses  employed  in  the first  three studies  were   36  iimoles/plate,  91




limoles/plate, and 10 umoles/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  incorpora-
                                     9-142

-------
tion 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




K1 2/343/113 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 and 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  TA1 535 and TA100 to doses from 3  to 9 mg/plate  (30  to 91




umoles/plate) in desiccators.   It was reported that this treatment yielded posi-




tive results, at least for TA1 535 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 be adequately evaluated.




     Barber et  al.  (1981)  exposed  Salmonella tester strains TA1 535,  TA98, and




TA100 to four  levels  of EDC vapors  (Eastman  Kodak Co.,  99.98$ pure)  in  a 3.4



liter airtight  exposure chamber.   These exposures resulted  in  estimated  plate




concentrations  ranging  from 31 .8  to  231 .8 umoles/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




TA1 535 and TA100.   The mutagenicity of EDC in these two strains was 0.002 and




0.001  revertants/nmole,  respectively.    No  difference in  revertant  counts  or




potency of EDC was noted in comparisons  of tests done with and without metabolic
                                     9-143

-------
activation.  The positive results were  obtained by Nestmann et al.  (1980)  and




Barber  et al.  0981 )  only when  tests were  conducted  in airtight  exposure




chambers.  This suggests that standard  mutagenicity  testing of EDC (bp 83-34°)




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 weak direct-acting 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.




Eucaryotic Test Systems




     In addition to causing gene mutations in bacteria, EDC also has been shown




to cause  gene mutations in eukaryotes.




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  b?rley seeds




(variety  Bonus) with 30.3 mmoles 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  LD _.   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




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$
                                     9-144

-------
                                                                       TABLE 9-26
                                                 Summary of Mutagenicity Testing of EDC:  Higher Plants
Reference
Ehrenberg et al.
197H
Test
System
Segregating
chlorophyll
(gene) mutations
in barley.
Chemical
Information
Concentration tested:
200 seeds treated
with 30.3 mnoles/24
hours (LD Q) in a
closed vessel.
Results Comments
Positive response. 1. About 600 spike progeny were tested.
(6-8$ mutants from
treated progeny vs.
0.06$ mutants from
control progeny).
FCiricheck, 197"»
Visible mutations
in peas, 8 varieties
Concentration tested:
EDC (concentration not
reported) or water
(negative controls)
vapors for 1 hours.

Source: Not reported

Purity: Not reported
Source:  Merck Co.

Purity:  Not given

Reported postive.
5.12-28.13< of
seeds reported
to be mutated.
1.  English translation of Russian article.

2.  Germination of the treated seed varied
    with the variety tested from 1 5% to 5B<
    compared to 100$ germination of the
    control seeds.

3.  Control mutation frequency not given.

4.  Putative mutations not characterized.
                                                                                                5.   Not possible to adequately evaluate
                                                                                                    results.

-------
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.




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, Shakarnis




1970,  and  King et al. 1979).   The  fourth study demonstrated  the induction of




somatic cell mutations by EDC  (Nylander et al. 1979).



     Shakarnis  (1969,  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 w   B




males.  In  fertility  of  treated females was reduced 47$ by the  4 h treatment and




91$  by the 8 h treatment. The  F- progeny  were scored  for  lethality (measured as




the  absence of M5  or  In wa B 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 h treatment)




and  5.9$  (8  h 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 4 or 6  hours.  Immediately after  treatment




 they were  mated to Muller  5  males and  the F2  progeny scored  for  sex-linked




 recessive lethals. The  4 h treatment did not significantly reduce fertility but
                                      9-146

-------
                              TABLE 9-29
Summary of Mutagenicity Testing of EDC:   Gene Mutation Teats  in  Insects
Test Chemical
Reference System Information Results
Shakarnis, 1969 Drosophila Concentration tested: Postive response 1.
aelanogaster virgin 3-day-old
sex-linked Canton S females 2.
recessive lethal exposed to 0.07<
test. EDC gas for 1 or 8
hours at 2U-25°C.
Source: Not given 3.
Purity: Not given
Duration of
Treatment
(h) with Number of
0.071 EDC Chromosomes Scored
Control 191 0
M 2061
8 U750
Comments
Muller-5 or In wa B males.
Fertility of treated females
reduced significantly (U7< after
1 hour treatment and 91 1 after
8 hour treatment).
Canton S strain reported to be
sensitive.
Lethal Mutations
No. %
15 0.30
67 3.22
261 5.91
p<0.05

-------
                                                                   TABLE 9-29 (oont.)
Reference
Test
System
   Chemical
   Information
                                                                        Results
                                                                                                  Comments
Shakarnis, 1970
Drosophilia
melanogaster
sex-linked
recessive lethal
test.
Concentration tested:
virgin 3-day-old
radiostable D-32
strain females
exposed to 0.07< EDC
gas for 4 or 6 hours
at 24-25°C.

Source:  Not given

Purity:  Not given
Positive response     1.

                      2.
Muller-5 males.

Experiment conducted twice for
4 hour exposure and three times
for 6 hour exposure.

4 hour treatment did not
significantly reduce fertility
but 6 hour treatment reduced it
by 50%.
Duration of
Treatment
(h) with
0.07* EDC
Control
4
6
Number of
Chromosomes Scored
1904
2205
2362
Lethal
No.
1
46
78
Mutations
*
0.05
2.00
3.30
(P<0.05)

-------
                                                                                        TABLE 9-29 (cont.)
Reference
                                           Test
                                           System
   Chemical
   Information
                                                                        Results
                                                                                                  Comments
King et al.
1979
                                           Drosophila
                                           melanogaster
                                           sex-linked
                                           recessive lethal
                                           test.
vO
 I
Concentration tested:
50 mM solution of EDC
in 5% sucrose fed to
1- to 2-day-old Berlin K
males for 3 days (Near
Source:  Merck Co.
         Darmstadt, FRG

Purity:  Not given but
         stated to have
         correct melting
         point and
         elemental
         analysis.
                                                                     Positive response
Base females.

Lethal frequency increased for all
broods.  Greatest effect in brood
II, which corresponds to the
spermatid stage of spermatogenesis.
Cone.
(mM)
0
50



Brood
I-III
I
II
III
I-III
Days after
Treatment
0-9
0-3
1-6
7-9
0-9
Number of
Lethal Mutations
Chromosomes Scored No. %
22,018
1,185
1,179
156
2,520
17
6
11
2
19
0.21
0.51
3.18
1.28
1.91
P


<0.01

<0.01

-------
                                                                                       TABLE 9-29  (cont.)
Reference
Nylander et al.
1978
Test
System
Droaophila
melanogaster
somatic cell
mutations so
- - !§Tel9
stocks.
Chemical
Information Results
Concentration tested: Positive response
0.1* and 0.5* EDC in
food at 25°C and 75*
humidity during larval
development.
Source: Fisher
Comments
1 . Mutations at z locus scored in
flies of stable and unstable
genotype. Both have same phenotype
Instability thought to be caused
by transposable genetic element.
2. Survival of treated flies reduced
                                                                        Scientific Co.
                                                               Purity:   Not given
01
o
significantly in both genetically
stable (34* reduction for 0.1* and
77* reduction for 0.5* dose) and
genetically unstable stocks (86$
reduction for 0.5% dose).
Treatment
Control
Stable
Unstable
0.01*
Stable
Unstable
0.05*
Stable
Unstable
Number
Males
Scored
I»»»H1
5363
6260
2689
610
201
Number
with
Sectors
2
U
263
271
K4
50

0
0
H
9
7
2H
*
.Ot5
.075
.20
.28
.21
.88
t Value
Within
-
18
214
11
13
Differences
Genotypes
--
.7*:
.31
X
.12
t Value
Between
0.
9.
5.
Differences
Genotypes
60
16§
66'

-------
the 6 h  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




h and 6 h treatments, respectively.




     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




(sic) and  elemental analysis reported)  in  5% sucrose  for  3  days.    This  dose




approximated the LE>     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 F2 generation were scored




for  lethality.   The lethal  frequency was found  to be elevated for  all three




broods  (0.51$,  3.W!>%,  and  1.28*,  respectively, for broods  I,  II,  and  III)




compared to the negative controls (0.21*).  The highest lethal frequency (3.^*)




was  found  in  brood  II  (P<0.01),  which corresponds  primarily  to the spermatid




stage of spermatogenesis.




     Nylander 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 F1



progeny for the induction  of somatic cell sex-linked mutations  in  the eye.  A




genetically  unstable  stock  (sc   z   w+)  and   a   genetically   stable  stock




(z 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 sc z w  is




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
                                     9-151

-------
unstable stock at  0.5% EDC).  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 EDC is capable of causing




both somatic cell  and  heritable  germinal mutations in a multicellular eukaryote.




Mammalian Cells In Culture —




     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-F^ -BH^  cells were exposed in suspen-




sion culture for a period of 5 hours to EDC  concentrations ranging up to 3mM (30%




survival) in tests with exogenous rat liver S9 mix for metabolic activation and




concentrations  ranging  up to  50mM (50%  survival)  in tests conducted without




metabolic activation.   Weak positive responses were observed both in tests with




(5 mutants/10   cells/mM) and without  (1 mutant/10   cells/mM)  metabolic activa-




tion (P<0.01).  EDC was detected as a direct-acting mutagen 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.




     The consistency of positive results  obtained in higher plants, Drosophila,




and  cultured  mammalian cells  demonstrates that EDC  causes gene  mutations  in




higher eukaryotes.
                                     9-1 52

-------
                            TABLE 9-30
Summary of Mutageniclty Testing of EDC:   Mammalian Cells in Culture
Reference
Tan and Hsie,
1981






Test
System
Chinese hamster
ovary cell HGPRT
gene mutation
assay.




Chemical
Information
Concentration tested:
Up to 3 nM in tests
with rat liver S9 mix
and up to 50 mM in
tests without
metabolic activation.
LD is 1 mM with
activation and 6 mM
without activation.
Results
Positive response ,
60 v. 3 mutants/106
olonable cells for
50 and 0 mM EDC
without activation.
26 v. 3 mutants/106
clonable cells for
1 . 5 and 0 raM EDC
with activation.
Comments
1 . Mutagenic activity of EDC without and
with metabolic activation calculated
to be 1 and 5 mutants/1 0 cells/
milHraole, respectively.

2. S9 mix increases mutagenicity by
about fourfold and cytotoxicity
5- to 25-fold.

                                                3.
NADP required in S9 mix for
metabolic activation.

-------
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 197*0.  Two  were  Drosophila melanogaster X chromosome  nondis-




junction tests (Shakarnis  1969  and 1970),  and two were micronucleus  tests (King




et al. 1979 and Jenssen and Ramel  1980).




     In the report by Kristoffersson (1974) EDC  (source, purity,  and concentra-




tion not given) was reported  to cause C-mitoses in Allium root  tip cells  but not




to cause  chromosome breaks in Allium  root tip  cells, human  lymphocytes  or to




induce prophage  from  E.  coli K 39  (X).   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.




     Data  consistent  with  this  possibility  are  found in studies by Shakarnis




(1969 and 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 F1   progeny, indicative of  meiotic



nondisjunction, was seen after a 4 h treatment (0.03?  exceptional females) and 8




h 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,  in this  study 38,437  fewer  progeny  of the  4  h




treatment  group were  scored  for  exceptional progeny  than  in  the first study.




Furthermore, the longest treatment time was  6 h  (i.e.,  2 h shorter  than in the




first study), and 8,086 fewer progeny were  scored  at this  time  point than for the




longest time point in the  first  study.   It  may be  that the sample sizes were too
                                     9-154

-------
                                                                                           TABLE 9-31




                                                             Summary of Mutageniclty Testing of EDC:   Chromosomal Aberrations Teats
Ul
Reference
Shakarnis, 1969
Test
System
Drosophila
aelanogaater
X chromosome
nondisjunctlon
Chemical
Information
Concentration tested:
virgin Canton S females
exposed to 0.7$ EDC
in 1 . 5 I. desiccator
for 4 and 8 hours at
2l»-25°C.
Source: Not given
Results Comments
Statistically
significantly
(P<0.05) increases
in exceptional
female progeny
for 4 hour exposure
and male and female
progeny for 8 hour
exposure .
                                                              Purity:  Not given
0.07* EDC
Duration of
Treatment
(h)
Control
4
8
Normal Progeny
No. Females
5,848
24,125
8,437
No. Males
5,727
21,297
7,773
Exceptional
Females
No.
0
8
15
*
0
0.03*
0.18*
Progeny
Males
No. *
0 0
3 0.01
7 0.09*
                    •P<0.05

-------
                                                                                        TABLE 9-31  (cont.)
Reference
Test
System
Chemical
Information Results
Comments
                     Shakarnis,  1970
Drosophila
melanogaster
X chromosome
nondisjunction
vo
 I
Concentration tested:
3-day-old females
from radiostable D-32
strain exposed to 0.07H
EDC in a 1.5 H
desiccator for M or 6
hours at 24-25'C.

Source:  Not given

Purity:  Not given
Incidence of nondis-  1
junction greater in
treated group than
in control. Increase
not statistically
significant.          2,
Not as many individuals scored
as in 1969 study.  Sample size
may not have been sufficiently
large.

Longest exposure time 2 hours
shorter than in previous
experiment; may have been too
short.
                                                                                0.07< EDC
                                                                                Duration of
                                                                                Treatment
                                                                                   (h)
                                                         Normal  Progeny
                                                    No.  Females    No.  Males
                                                          Exceptional   Progeny
                                                          Females       Males
                                                          No.   %       No.   )
                                                                                 Control

                                                                                    1)

                                                                                    6
                                                      2172

                                                      35B4
                                               2205

                                               3H01

                                               U090
                                      0

                                      0.03

                                      0.10
                     1     0.03

                     1     0.03

                     3     0.07

-------
                                                                   TABLE 9-31  (cont.)
Reference
King et al.
1979
Test
System
Micronucleua test:
NMRI mice
Chemical
Information
Concentration tested:
2 i.p. injections of
Results
Negative
Comments
1 . 1 ,000 polychromatic erythrocytes
analyzed/animal .
                                          i» mmoles/kg (UOO mg/lcg)
                                          given 24 hours apart.
                                          1 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 micronuclei not
                                                                        given.
Jenssen and Ranel,
1980
Mioronucleua test:
CBA mice
Concentration tested:
single i.p. injection
100 mg/kg.
                                          Source:
                              BDH Chemicals,
                              Ltd.
                                          Purity:  Not given
Negative.
(0.15 ± 0.1U) poly-
eromatlc erythocytes
with micronuclei in
controls v. 0.1 7 +
0.10 in treated
animals).

-------
small or the duration of exposure too  short  in the second study to be  able to




detect a statistically significant effect.




     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 mmoles/kg (400 mg/kg)




EDC  (Merck  Co.,  purity  not  specified  but  melting  point  (sic)  and  chemical




analysis reported  to be correct).   The  authors  state this corresponds  to an




"approximate lethal  dose" (the LD1 Q for intraperitoneal injections of EDC in mice




is 250 mg/kg).   The  injections were given  2H h apart;  the animals were killed 6 h




after  the second  injection and  bone  marrow  smears made.   One thousand  poly-




chromatic erythrocytes (PCEs) were analyzed  per  animal.   Frequencies  of micro-




nuclei 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  (DBH




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




micronuclei.  The frequencies of PCEs  with micronuclei were  0.15 + C.11 in the




controls and 0.17 + 0.10 in treated animals.




     The positive  response in the  X-chromosome test  in  Drosophila (Shakarnis




1969)  suggests  EDC  is capable of causing meiotic  non-disjunction  resulting in




numerical chromosomal  abnormalities.   The negative  responses obtained  in  the




micronucleus 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  in vivo cytogenetic tests.   Such testing  is required
                                     9-158

-------
before a judgment can be made on the ability of EDC to cause chromosomal aberra-




tions.



OTHER EVIDENCE OF DNA DAMAGE




     Three other  tests  have been  conducted on the  genotoxicity of EDC.  These




tests do not measure mutagenic  events  per s_e in that they do not demonstrate the




induction  of heritable  (i.e.,  somatic or  germinal)  genetic  alterations,  but




positive results in these test systems show that DNA  has  been damaged.  Such test




systems provide supporting evidence useful for assessing genetic risk.




Bacterial Test Systems




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. 097*0 soaked  sterile  filter disks  with  10 uS, (80 umoles) 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 repsonses were said to be reproducible.  The ratio between




the zones  of  inhibition   (polA+/polA~) 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.




Eukaryotic Test Systems




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
                                     9-159

-------
                                                                            TABLE 9-32
                                                       Summary of Mutagenioity Testing of EDC:  PolA ASSAY
     Reference
 Test
System
                                            Strains
Activation
  System
 Chemical
Information
                                                                                                     Results
                                                                                                                              Comments
     Brem et al.       polA differential     E.  coll
     1971             cell killing          polA~~
                      assay                 polA*
                                        None
                 Concentration tested:
                 10 »l on filter disk
I

o
                                                                                Questionable     1.   All assays carried  out  in
                                                                                positive              duplicate on at  least
                                                                                                     three different  occasions.

                                                                                                 2.   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 Inhib.

EDC
(MS
Chloramphenlcol
A+
mm
8
15
26
A"
mm
9
51
28
A*/A"
1.26
1.11
1.00

-------
 Prodi  (1981 )  collected  blood  samples from healthy humans,  separated the lympho-




 cytes,  and cultured 5  x  10   of them in  0.2  mfc medium for 4 h  at 37°C  in  the




 presence  or  absence  of  EDC  (Carlo  Erban,  Milan,  Italy  or Merch-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  [ H]  deoxythymidylic acid




 (TdR)  after 1 h of culture (2661  + 57 dpm in untreated cells compared to 228 7 + 60




 dpm  in cells treated with  5 \iH/mH [0.06 umole/mfc] EDC).   Subsequently 2.5,  5,  and




 10 uH/mS, (0.03, 0.06 and 0.1  umole/m&) 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 4 h later. At 10 nfc/mJ, 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 71 5 +  24 and  61 2 + 26 dpm,  respectively.




 No  positive controls  were run  to  ensure that  the system  was working  properly




 although testing of chloromethyl methyl ether  (CMME)  with activation resulted in




 a doubling of dpms over  the corresponding  negative control values (1320 + 57 at 5




 nl/mS, CMME  versus 612+26 untreated).  The authors  calculated an  effective  DNA




 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
                                     9-161

-------
(673 + 45 at  5 ui/ml and 630 + 34 at 2.5 \iH/m!L  compared to control value of *vi 2 ±




26)  the  increases were not  statistically significant.   The  positive  finding




reported in this work is therefore judged to be inconclusive.




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).  Two gram aliquots of TA1 535 were incubated with 7.06




Hinol [  C] EDC/mH (sp.  act. =3.2 mCi/mmol) and varying amounts of cytosol.  DNA




alyklation values at cytosol  concentrations of 2.2, 7.8,  27, and 71$ were 8.65,




27, 107,  and  137 dpm/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  m  + 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 [14C]  EDC (sp. act. = 0.32




mCi/mmol) was administered to groups of  three animals by  gavage (150  mg/kg)  or




inhalation (150 ppm, 6 h). The animals were subsequently sacrificed and DNA was




extracted from the  liver,  spleen,  kidney, and  stomach  for  measurement  of DNA




alkylation.  Overall,  there  was three to  five  times more  DNA alkylation after




gavage than after inhalation.   The  values  ranged from  5.8  + 0.7 to 23.1  + 7.4




alkylation/10  nucleotides for gavage  versus 1.8  + 0.3  to 8.2 +  3.3  alkyla-




tion/10  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
                                     9-162

-------
                         TABLE 9-33
Summary of Mutagenioity Testing of EDC:   DNA  Binding Studies
Activation
Reference Teat System Strains System
Reitz et al. DNA alkylation TA1 535 Phenobarbital
1982 and mutageneala induced rat % Cytoaol
in Salmonella liver cytosol 2.2
vo 7.8
L 27
CT> 71
t-o
DNA alkylation Osborne- NA
in rats Mendel rat Route of
Exposure
Gavage
1 50 mg/kg


Inhalation
1 50 ppm , 6 h


Chemical Information

Revertants
4.6 + 0.82
23.5 + 3.0
80.2 + 9.6
111 + 2.6



Tissue
Liver
Spleen
Kidney
Stomach
Liver
Spleen
Kidney
Stomach
Alkylation x 1 0~6
Hue leot ides
4
12.5
49
64

Alkylation x 1 0
Nucleotldes (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 (imol EDC/ml.

2. Significant correlation
between degree of alkylation
and Increased reversion
frequency (r = 0.9976).
3. Rats sacrificed 4 h post
gavage or immediately after
inhalation.

4. Sp. act. 3.2 mCi/nmol for
bacteria; 0.32 mCi/mmol
for rats.





-------
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.




SUMMARY AND CONCLUSIONS




     Ethylene  dichloride  (EDC)  has  been shown  to cause  gene  mutations  in




bacteria,  plants,  Drosophila  and cultured Chinese  hamster ovary (CHO)  cells.




Weak  positive  responses were  observed  in the  bacterial and CHO tests  in  the




absence  of  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, a positive  response  in  another test system is needed to permit a




judgment on the generality of this  effect.  With respect to its  ability to cause




structural  chromosomal  aberrations,  sufficient testing has not been 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 in




vitro  and  in vivo mammalian cytogenetic  assays.   A sister  chromatid exchange




assay  would  also be  useful  in  assessing the  ability of EDC to cause chromosome




damage.
                                     9-16U

-------
     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 weak direct-acting



mutagen.  Several of its putative metabolites, thought to  be formed in mice and



rats, are Judged to be more potent mutagens  (e.g.,  S-[2-chloroethyl]-L-cysteine



or chloroacetaldehyde) than EDC with the potential to cause adverse  effects in



humans.
                                     9-165

-------
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 estimating




its public health impact, including a potency evaluation in relation to other




carcinogens.  The evaluation of carcinogenicity depends heavily on animal




bioassays and epidemiologic evidence.  However, other factors, including




mutagenicity, metabolism (particularly in relation to interaction with DNA),




and pharmacokinetic 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.  This section




presents an evaluation of the animal bioassays, 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 investigated  in a number of




studies in which EDC was administered to rats and mice via various routes of




administration.  Five 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.
                                        9-166

-------
 9.5.1.1  National Cancer  Institute  (1978) Rat Study—Hazleton Laboratories




 America  Inc., Vienna, Virginia,  under the sponsorship of the NCI, conducted  a




 bioassay of  1,2-dichloroethane  (EDC) using Osborne-Mendel  rats.  The NCI  pub-




 lished 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  revealed




 a purity of  98%  to 99% EDC with  4 to 10 minor peaks (Ward  1980).  However, in




 the  NCI  bioassay 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%  1,2-




 dichloroethane,  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
                                        9-167

-------
         TABLE 9-34 .  DESIGN SUMMARY FOR 1,2-DlCHLOROETHANE (EDC) GAVAGE
                      EXPERIMENT IN OSBORNE-MENDEL RATS
                                   (NCI 1978)
Group
 Initial
numbe r of
 animals
1,2-dichloro-
   ethane
   dosage3
Observation period
Treated  Untreated
(weeks)   (weeks)
Time-weighted
average dosage
over a 78-week
   period*5
Males
Untreated control
   20
                            106
Vehicle-control 20
Low-dose 50




High-dosed 50




0
50
75
50
50C
0
100
150
100
100C
0
78
7
10
18
34
— — —
7
10
18
34
— ~~~~
32
	
	
- —
9
32
	
	
	
9
23
0
47
	
	
	
	
95
	
	
	
— — —
Females

Untreated control     20

Vehicle-control       20
                  0
                  78
             106

              32
Low-dose
   50
      50
      75
      50
      50C
       0
    7
   10
   18
   34
       47
                                                            9
                                                           32
High-dosed
   50
     100
     150
     100
     100C
       0
    7
   10
   18
   34
       95
                                                            9
                                                           15
aDosage, given in rag/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.
dAll animals in this group died before the bioassay was terminated.
                                  9-168

-------
found to be inappropriate.  Early signs of toxicity necessitated several changes




in the dosages (Table 9-34).  Animal weight and food consumption per cage were




obtained weekly for the first 10 weeks and monthly thereafter.  Animals were




checked daily for mortality.  Weight depression was observed in both groups




exposed to EDC.  By 50 weeks, the weight depression averaged 12% in high-dose




rats (Figure 9-8  )•  Mortality was early and severe in dosed animals, especially




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 u for slides.  Hematoxylin and eosin stain were used rou-




tinely, and other stains were employed when necessary.  Diagnoses of tumors
                                      9-169

-------
     750
     600-
   CD
   h_
   o
  E 450-
  O
  ULJ

  £ 300 H
  o
  CO
  z
  ^ 150^
            MALE RATS
               Untreated Control
               Vehicle-Control
               Low-Dose
               High-Dose
                                                                            •750
                                                                           -600
                              -450
                              -300
                                            -150
i    '—i—i—i—i—i—i—i—i—r
 0      15      30      45       60      75
                       TIME ONTEST(Weeks)
                                                                      1
                                                         90
                                 105
                                                                             0
                             120
     750
     600-
   co
   k.
   CD
  I 450 -^
  LJJ
  O
  m
     300 -|
     150H
            FEMALE RATS
             T
                15
                                                                             750
                                    T
                                         I
T
                 T"
45       60      75
TIME ON TEST (Weeks)
                                                            Untreated Control
                                             	Vehicle-Control
                                             	Low-Dose
                                             	High-Dose
                                             	1	1	1	r—
                                                                           -600
                              -450
                                                                           -300
                              -150
            90
                                                        105
120
Figure 9-8 .   Growth  curves  for male and female Osborne-Mendel rats
              administered  1,2-dichloroethane (EDC) by gavage.
              (NCI  1978)
                                   9-170

-------
I
M
•^1
          C
          i-(
          (t
           I
          VO
        CD V)
        0. C
        3 H-
        H- <
        cn 0)
        rr M
        (0
        >-< O
        ro o
        O. B
          •a
        t- 0>
        »  l-l
        K) H-
         I cn
        a o

        n' cn
        O O
        O
        TO
        3
        n to
          3
        ^^ Q.
        PI
        O Ml
        O ID
        TO  O
        (U  01
        <:  cr
        o>  o
        TO  n
        ro  3
        •  m
        ^ m
        Z 3
        ° s-
        I— I IB
         00 ft
         >— tn
              PROBABILITY OF SURVIVAL
                                                                                                PROBABILITY OF SURVIVAL
m
O
                       O


                       VI
c
c


-

cn "


CO
O



cn ~

O)
O"




-
cn ~

CD
O~



o-
cn


NJ
O
(
i
> O O O O -
> to '*. b) oo c
I.I .1.1.

1 i ! f
r
I r- < C r-1
? c? 1 2 f
w S2 o S r r"
• § o r ,
s» o 1 J
° 5 1
•^ r~ I
- ' i
1
j 1
J ^ j
	 j _j^* I
— r1 i !
,J [ 	 1
X '"
,_j i
1 1 ,-.
r y r-J
.-1 _.j
'•'^ H
J cr-^ — 1
!JJ" H1"?
j ..,-J r H
r--1"1"' 1
r -1 J r
r' " •
_r" !
i r

c
1
, 1 1 . 1 1 1 1 1
DO O O O -
5k) *» O5 CO C
A
5

























n
^
^
>
n
D
>
H
n


.A
D
m
O

                                                          (D
                                                          CD
                                                          ?r
                                                          (/>
p p p p
o K>> '4* b>


-

«-
-

CO
0 ~
-
^
cn ~


-

a> _
°



cn ~

CO
O -


—
cn
0 ~
1 i 1 i 1 i
1 ! 1
1 ! 1
i ! 1
X r- < c
f 1 1 3 }
' rn 0 « r
OO rr Ol P
_ 0 * ™ '
S » o * 1
(DO t
3 0 1
II
^ J
1
1
—
,J
rr .--
_T 1
_X" i
ij r
-J ._!
—r' ' ' '"
, 	 r | ' ^
,- 1 r--'"J
r1 ,.JJ .J"J

1 .r!J" 1
1 ,---' 1
J r-1" j
r" r';
i j

i i . i i i
D p O O
O k) 4^ O)
0
CO
1


J —
J










1


	 l~




i j
i
1 1
i i
i
f~ f
— 1
	 1
1
J
l"
r


1 — '






0
00


*»-^











-»
O

r
J




i"
r





























r-
m
H
b

-------
and other lesions were coded according to the Systematized Nomenclature of

Pathology (SNOP) of the College of American Pathologists (1965).
               TABLE 9-35 . TERMINAL SURVIVAL OF OSBORNE-MENDEL RATS
                     TREATED WITH 1,2-DICHLOROETHANE (EDC)
                            (adapted from NCI 1978)
Group
Untreated control
Vehicle-control
Low-dose
High-dose
Weeks in
study
106
110
110
101
Males
Females
Animals alive
at end of study
4/203
4/20
1/50
0/50b
(20%)
(20%)
(2%)
(0%)
Weeks in
study
106
110
101
93
Animals
at end of
13/203
8/20
1/50
0/50*>
alive
study
(65%)
(40%)
(2%)
(0%)
aFive rats were sacrificed at 75 weeks.
t>All animals in this group died before the bioassay was terminated.
     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.
                                 9-172

-------
 characterized microscopically by acanthosis and hyperkeratosis  in  the  super-

 ficial area.  The  basal  epithelial  layer contained  papillary cords and nests  of

 anaplastic squamous 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, metastasized to  adjacent

 tissues.
    TABLE 9-36 .  SQUAMOUS CELL CARCINOMAS OF THE FORESTOMACH IN OSBORNE-MENDEL
                   RATS TREATED WITH 1,2-DICHLOROETHANE (EDC)
                            (adapted from NCI 1978)
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
3P values calculated using the Fisher Exact Test.  Treated versus pooled vehicle-
 control.
^A squamous cell carcinoma of the forestomach metastasized in one male in this
  group.
NS = not significant when P values are greater than 0.05.


     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 intestine, subcutaneous tissue,  and abdominal cavity.  The Cochran-Armitage

Trend Test indicated a significant (P  = 0.021) positive association between

dosage and the incidence of hemangiosarcomas in male rats as compared to the
                                   9-173

-------
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 = 0.0*41).
       Table 9-37.  HEMANGIOSARCOMAS IN OSBORNE-MENDEL RATS TREATED WITH
                           1 ,2-DICHLOROETHANE  (EDO)
                            (adapted from NCI 1978)
Group
Untreated control
Matched vehicle-
Males
0/20 (0%)
0/20 (0%)
P value3 Females
0/20 (0*)
0/20 (0$)
P value3

 control

Pooled vehicle-
 control

Low-dose

High-dose0
1/60 (2%)
9/50 (18?)

7/50 (14*)'
             0/59 (0%)
0.003

0.016
4/50 (8$)    0.041

4/50 (8$)    0.041
 P values calculated using the Fisher Exact Test (one-tailed).  Treated
 versus pooled vehicle-control.
 Only 48 animals were examined for hemangiosarcomas of the large intestine.
°0nly 49 animals were examined for hemangiosarcomas of the spleen and
 adrenals and 48 for hemangiosarcomas of the pancreas.
NS = Not significant.

     In addition to stomach carcinomas and hemangiosarcomas, EDC-treated

female rats showed significant increases in the incidence of mammary adenocarcinoraas

(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

(P<0.001 ) positive association between 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)
                                      9-174

-------
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)
                            (adapted from NCI 1978)
                                Adenocarcinoma of
Group                           the mammary gland         P value3


Untreated control                  2/20 (10?)

Matched vehicle-control            0/20 (O/O

Pooled vehicle-control             1/59 (2$)

Low-dose                           1/50 (2?)                NS

High-dose                         18/50 (36$)              0.0008


 P values calculated using the Fisher Exact Test (one-tailed).  Treated
 versus pooled vehicle-control.
NS = Not significant.


     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 squamous cell

carcinomas of the forestomach, hemangiosarcomas of the circulatory system, and fibromas

of the subcutaneous tissue occurred in male rats.  There was also a statistically signifies

increase in the incidence of adenocarcinomas of mammary gland and hemangiosarcomas

of the circulatory system in female rats.

9.5.1.2.  National Cancer Institute (1978) Mouse Study — Hazleton Laboratories 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
                                       9-175

-------
by the NCI (1978) and were also reported by Weisburger (1977),  the International




Agency for Research on 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 thereafter.




Animals were checked daily for mortality and signs of toxic effects.  No dose-




related mean body weight 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 1 5.  The estimated probabilities of




survival for male and female mice in the control and EDC-dosed groups are shown




in Figure 9-31-  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 a 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
                                     9-176

-------
         TABLE 9-39 •   DESIGN SUMMARY FOR 1,2-DICHLOROETHANE (EDC) GAVAGE
                           EXPERIMENT IN B6C3F1 MICE
                            (adapted from NCI 1978)
Group
 Initial   1,2-dichloro-  Observation period
number of     ethane      Treated  Untreated
 animals      dosage3     (weeks)   (weeks)
Time-weighted
average dosage'3
Males
Untreated control
Vehicle- control
Low-dose


High-dose

Females
Untreated control
Vehicle -control
Low-dose



High-dose


20
20
50


50

20
20
50



50


aDosage, given in tng/kg body
days per week.
''Time-weighted average dosage

0
75
100
0
150
200
0

0
125
200
150
0
250
400
300
0

78
8
70
____
8
70
___

78
8
3
67
— • — — •
8
3
67

weight, was administered by
= (dosage x weeks received)
weeks
receiving chemical
90
12
	
	
12
— __
13
91
32
	
	
	
13
___
	
13
gavage

0
97
	
— __
195
~" — *~

0
149
	
	
_^ —
299
	
— —
five consecutive
                                   9-177

-------
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)
                            (adapted from NCI 1978)
Group
Untreated control
Vehicle -control
Low-dose
High-dose
Weeks in
study
90
90
90
91
Males
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%)
     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

paraplast, and sectioned at 5 u 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
                                      9-178

-------
        50
              	 Untreated Control
              	Vehicle-Control
              	Low-Dose
              	High-Dose
                                                             MALE MICE
              T
             T
              15
                         T~
                          30
                           45       60      75
                          TIME ON TEST (Weeks)
                                                                               50
                                                                             -40
                                                                             -30
                                                                             -20
                                                                         -10
                                  90      105     120
       50
I  40-
(0
O
»-
O  30^
UJ

0  20 H
o
CD
10-
                 Untreated Control
                 Vehicle-Control
                 Low-Dose
                 High-Dose
                                                           FEMALE MICE
                   I
                  15
                                                                              50
                                                                             -40
                                                                            -30
                                                                            -20
                                                                        -10
30      45       60      75
        TIM EON TEST (Weeks)
                                                      90
                                                           105     120
Figure 9-1O   Growth curves  for male and female B6C3F1 mice administered
              1,2-dichloroethane  (EDC) by gavage.  (NCI 1978)
                                   9-179

-------
00
O
            Tl
           TO
           C
            CD


            ^O
         .  C
          to 1
          o n
          i-l O
          o B
          m -o
          rf 05
          =r i
          Ql H.
          3 cn
          fD O
            3
          r— N 01
          m
          o ••*>
          o o
          Oq (D
          0)

          •^ 2
          0) 3
          TO ^*
          (D
          . Hi
            n>
          n f
          I—I
             03

          VD n

          OO T)
          •*~* H-

             s
             H-
             o
             m

             TO
             H-

             fD
             3
                                   PROBABILITY OF SURVIVAL
             PROBABILITY OF SURVIVAL
o o o o o r*
b K> '*• o> bo o

o —

01 ~~

•
to _
O


— |v
m
O
2
ffi 8-
—1

^^
^^ -^
n
* ^J


(O
o -

O -
tn

_,
Si
0
, l.l.ii
1 !
1 !
X r- < C rr
— .. O fl> T
fQ ^ .L. ^ 1
3" < — . *^ 1
» J. O CO i

1 1 |i 1
3 0
3 o r1
0 3 '
-I r
,-' .
1:
i
I !
JJ .-?

J 	 ^ r--* '

, «J , f
r — "~ 1 i"1
-r1-1"1" j f
f"" rj^f





















1 X""fl
i _F "" _L'

Tl
m
m

O
m
D O O O O -*
b io '-^ b> oo o
m

O
2

m
CD
0)

-------
 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~H  HEPATOCELLULAR CARCINOMAS IN B6C3F1 MICE TREATED
                          WITH 1,2-DICHLOROETHANE (EDC)
                            (adapted from NCI 1978)
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
3P values calculated using the Fisher Exact Test (one-tailed).  Treated versus
 pooled vehicle-control.
NS = not significant.


     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)
                                     9-181

-------
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.  ALVEOIAR/BRONCHIOLAR ADENOMAS IN B6C3F1  MICE TREATED
                         WITH 1,2-DICHLOROETHANE (EDC)
                            (adapted from NCI 1978)
Group
Untreated controls
Matched vehicle-
controls
Pooled vehicle-
controls
Low-dose
High-dose^
Males P value3
0/20
0/19
0/59
1/47
15/48
(0%)
(0%)
(3%)
(2%) NS
(31%) 0.0025
Females
1/19
1/20
2/60
7/50
15/48
(5%)
(5%)
(3%)
(14%)
(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.
     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.
                                    9-182

-------
            TABLE  9-43 .  SQUAMOUS CELL CARCINOMAS OF THE FORESTOMACH
              IN B6C3F1 MICE TREATED WITH 1,2-DICHLOROETHANE  (EDC)
                            (adapted from NCI 1978)
Group
Untreated control
Matche i vehicle-
contr )l
Pooled vehicle-
contr )1
Low-do ;e
High-d >se
Males P value3
0/20 (0%)
1/19 (5%)
1/59 (2%)
1/46 (2%) NS
2/50 (4%) NS
Females P value3
0/20 (0%)
1/20 (5%)
1/60 (2%)
2/50 (4%) NS
5/48 (10%) NS
aP values calculated using the Fisher Exact Test (one-tailed).  Treated versus
 vehic Le-control.
NS = not significant.
     The incidence of adenocarcinomas of the mammary gland in female mice

treate< with EDC is presented in Table 9-44 .  The Cochran-Armitage Trend

Test indicated a significant (P = 0.007) positive association between dosage

and th< incidence of adenocarcinomas of the mammary gland when compared to the

pooled controls.  The Fisher Exact Test confirmed these results with a

significant (P _<_ 0.003) comparison of both high-dose (7/48) and low-dose

(9/50) groups to the pooled vehicle-control groups (0/60).

     Er.dometrial 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 and combined incidence.  The Fisher Exact Test

showed a statistically significant (P = 0.014) incidence in the high-dose

(5/47) group.
                                         9-183

-------
       TABLE  9-44.  ADENOCARC1NOMAS OF THE MAMMARY GLAND IN FEMALE B6CJF1
                   MICE TREATED WITH ],2-DICHLOROETHANE (EDC)
                            (adapted from NCI 1978)
Group
  Adenocarcinoma of
  the mammary gland
P value3
Pooled vehicle-control

Matched vehicle-control

Low-dose

High-dose
      0/60 (0%)

      0/20 (0%)

      9/50 (18%)

      7/48 (15%)
0.0005

0.0026
aP values calculated using the Fisher Exact Test (one-tailed).  Treated versus
 pooled vehicle-control.
        TABLE 9-45 .   ENDOMETRIAL POLYP OR ENDOMETRIAL STROMAL SARCOMAS IN
            FEMALE B6C3F1 MICE TREATED WITH 1,2-DICHLOROETHANE (EDC)
                            (adapted from NCI 1978)
Group
    Endometrial polyp or
endometrial stromal sarcomas
P value3
Pooled vehicle-control

Matched vehicle—control

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.
     In summary, the NCI study in B6C3F1  mice demonstrated a statistically

significant increase in incidences of hepatocellular carcinomas and alveolar/

bronchiolar adenomas in male mice and a statistically significant increase in

incidences of alveolar/bronchiolar adenomas,  mammary carcinomas,  and endometrial

tumors in female mice.
                                      9-184

-------
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 ppm for 7 hours per exposure.



9.5.1.A  Maltoni et al. (1980) Rat Study—A more recent inhalation study con-

ducted 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 .
      TABLE 9-46 .  CHARACTERIZATION OF 1,2-DICHLOROETHANE (EDC) INHALATION
                       EXPERIMENT IN SPRAGUE-DAWLEY RATS
                             (Maltoni et al. 1980)
Component
Purity
1,2-Dichloroethane

1,1-Dichloroethane

Carbon tetrachloride

Trichloroethylene

Perchloroethylene

Benzene
99.82%

 0.02%

 0.02%

 0.02%

 0.03%

 0.09%
                                        9-185

-------
            TABLE 9-47 .  DESIGN SUMMARY FOR 1,2-DICHLORUETHANE (EDC)
                       EXPERIMENT IN SPRAGUE-DAWLEY RATS3
                             (Maltoni et al. 1980)
Group
I

II

III

IV

V

VI

Concentration Sex
250-150 ppmc M
F
50 ppm M
F
10 ppm M
F
5 ppm M
F
Controls in chambers M
F
Controls M
F
Animals**
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.
     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

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-
                                      9-186

-------
 after.  All detectable gross pathologic  changes were recorded.  A  complete




 autopsy was performed on each animal, with histopathologic examinations conducted




 on  the following:  brain, Zymbal glands, retrobulbar glands, interscapular




 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 mentality varied with the different groups, but there




 appears to be no direct relationship between mortality and exosure to EDC




 (Table 9-48 ).  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 ex-




 posed 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 sur-




 vival rate was 83.7%.




     The results of histopathologic 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 ex-




posed to EDC at 250-150 ppm,  50 ppm,  and 5 ppm,  when compared to controls in the
                                      9-187

-------
                 TABLE 9-48 .   SURVIVAL  OF  SPRAGUE-DAWLEY  RATS  EXPOSED  TO  EDC  AT  52  AND 104  WEEKS3
                                               (Maltoni  et al.  1980)
Dose
Group (ppm)
I 250-15Qb

£ II 50
00
III 10

IV 5

V Controls
in chambers
VI Controls

Sex
M
F
M
F
M
F
M
F
M
F
M
F
Initial
numbe rs
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
Numbe r
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.
''After a few weeks the dose was reduced  to  150 ppm because  of high  toxicity at  the  250 ppm  level.

-------
TAHLK  9-49  TUMOR INClliKNCK HI M'RACUF-IIAWI KY RATS  KKPOSKI)  TO K.DC
                        (M.iltnnl ct  al.  IIHII)
An lm,i Is wi tli
M.imtn.ir v Lumoi s
Animals3

Croup <>incp"t rnt i on Sex
I -J50- 1 "7(1 ppjnr M
F
M ,-inrl F
I t 50 ppm M
P
1 M and F
5§ '11 10 ppm M
F
M and F
IV 5 ppm H
F
M and F
V Controls In M
chambers F
M and F
VI Controls M
F
M and F
Total

Number
at start
90
90
180
90
90
180
90
90
180
90
90
180
90
90
180
90
90
180
1080

Correct ed
numbe r
89
90
179
90
90
180
89
90
179
90
90
180
90
90
180
90
90
180
1070

Total
numbe r
1 1
52
63
10
58
68
5
43
48
11
65
76
8
38
46
5
52
57


Percent1'
12. 1
57.7
35.2
1 I.I
64.4
37.8
5.6
47.8
26.8
12.2
72.2
42.2
8.9
Iti.l
25.5
5.5
57.8
31.7

Average
1 at encv
lime
(wkK
92.9
78.6
81 .1
HH.S
78.5
80. o
60.8
79.4
77.5
110.2
83.2
87.1
85.5
83. 1
83.6
92.0
85.5
86.1


Total
number
0
2
2
0
1
1
0
0
0
0
2
2
1
0
1
1
0
1

/.vmbal

Pe rcent b
„_
2.2
I.I
	
1 .1
0.6
—
—
-__
2.2
1.1
1.1
	
0.6
I.I
	
0.6

gland carcinoms
A ve r a Re
latency
t i me
(uk)d
_-_
78.0
78.0
	
86.0
86.0
—
	
.__
47.D
47.0
78.0
	
78.0
74.0
	
74.0


Tot a 1
numbe r
0
0
0
1
2
1
4
0
4
2
6
8
1
3
4
0
3
3

r urior s
LCI

Percent"
...
	
—
1. 1
2.2
1.7
4.6
2.2
2.2
6.7
4.4
1.1
1. )
2.2
	
3.3
1.7


ikemi as
A ve r a p,c
la tency
t Ime
(wk)''
...
	
— -
HI! .0
7-1.5
76.3
74. o
74.0
133.5
83.5
90.0
17.0
64.3
59.5
	
79.1
79.1


Nephroblns tomap
Average
la t encv
Total time
number Percent" (wV)^
D 	 	
II 	 	
„
I,
O 	 	
II 	 	
II 	 	
(1 	 	
II 	 	
1 I.I 8'..n
(1 	 	
1 0.7 84.0
I,
D 	 	
0
0 	 	
(1 — —
,1
1
                                                                                                                           (coitt t miptt  on the  f o I I nwt IK;

-------
TABLE
     9-49.
             (Continued)
Animals with tumors
Angiosar comas



Group Concentration Sex
I 250-150 ppmf M
F
M and
v£> I I 50 ppm M
1 F
H1 ,
vo M a nd
O
HI 10 ppm M
F
M and
IV 5 ppm M
F
M and
V Controls In M
chambers F
M and
VI Controls M
F
M and
Liver
Average
1 a tency
Total time
number Percent*1 (wk)d
0 — 	
0 	
F 0
l\ 	 	 	 	
0 — - 	
F 0 	

0 ' 	 	
0 	 	
F 0
i\ 	 __ 	
0 	 	
F 0 	
0 	 	
0 	 	
F 0
„
0 	 	
F 0
Other Sites
Average
latency
Total time
number Percent*1 (wk)d
0 	 	
0
0 	 	
0 	 	
0 — 	
0 	 	

0 	 	
0 	 	
0 — — — — — —
0
0 	 	
0 	 	
0 	 	
0 	 	
0 	 	
0 	 	
0 	 	
0 	 	
Angiomas and f ibroangl omas
Liver
Ave rage
latency
Total time
number Percent*1 (wk)^
0 	 	
0 	 	
0 	 	
o — —
0 	 	
0 	 	

0 	 	
o — —
0 	 	
0 	 	
0 	 	
o — —
o — —
o — —
0 	
o — —
0 	 	
o — —



Total
numbe r
0
0
0
0
0
0

0
0
0
1
1
i
0
0
0
1
0
1
Other sites
Average
latency
t imp
Percent*1 (wk )d
	 	
	 	

—
	 	
	 	

	 	
	
	
1.1 125. f»
1.1 1)1. 0
l.l J2K.O
_-_
	 	
	
1.1 130.0
	 	
0.7 1)0.0

-------
UHl.h 9-49.   (cout Inucd)
Animals with tumors



Group Con < f*n t rat ion Sex
I 25H-I50 ppm1" M
F
M and F
I I 5li ppm M
F
M and F
111 10 ppm M
F
M and F
IV 5 ppm M
F
H and F
V Controls in M
chambers F
M and F
VI Controls M
F
M and F
Hc'patomas
Average
latency
Total t line
number Percent*" (wk)<*
0
n — —
0 	 	
(i — —
0 	 	
0 	 	
0 — —
0 	 	
0 	 	
rj — —
0 — 	
0 	 	
„
0 	 	
0 -— 	
0 	 	
0 	 	
n — —
Korestomach epithelial


Total
number
II
II
II
1
0
1
0
1
1
1
0
1
2
1
3
2
1
3


Percent0
	
	
	
1 . 1
	
0.6
	
1.1
0.6
1.1
	
0.6
1. 1
1.1
1.7
2.2
1.1
1.7
Average
latency
t Imc
	
	
	
60.0
	
60.0
	
78.0
78.0
101.0
	
101.0
78.5
6).o
73.3
110.0
102. U
107.3
Skin carcinomas
Aver.-iRe
la tencv
Tot a 1 1 1 me
number Percent0 (wk)^
0 	 	
O 	 	
0 	 	
1 1.1 56.0
() 	 	
1 0.6 56.0
I 1.1 94.0
(1 	 	
1 0.6 94.0
0 	 	
0 	 	
I) — —
0
0 	 	
II
II 	 	
0 	
I) — —



Total
number Percent0
0 	
(1 	
0
(i 	
0 	
II 	
0 	
0 	
o —
1 1.1
0 	
1 (1.6
0 	
1 1.1
1 II. n
0 	
0 	
0 	

A ver.i^e
litpnrv
1 Imc

	
—

	
	

	
—
io.o
	
30.0
—
78. 0
7H.O

	
	 i

-------
TAB1F. 9-49-   Out Inued)
Animals with tumors




tiroup
I


I 1


II 1


IV


V


VI


Exposure
Results




Concent rat Ion
250-150 ppme

M
50 ppm

H
10 ppm

H
5 ppm

M
Controls in
chninbe rs
H
Controls

M
by inhalation to F.DC
after 148 wk (end of
^The percentages refer to the
^ ra'!e
""Af t PT a





Sex
M
F
and F
M
F
and F
M
F
and F
M
F
and F
M
F
and F
M
F
and F

Nen rob las to

Total
number Percent0
0 	
0 	
II 	
0 	
0 	
0 	
0
0 	
0 	
II —
0 	
0 	
[1 	
II 	
II —
0 	
0 	
u —
in air at 250-150, 50 ppm,
expert me n t ).
correct ed

few werks the dose was reduced
numbe rs .
airy p
to 1 5O ppn, be c i u
I nccpha I 1 c tumors
mas Other
Average
la t encv
time Total
(wk )(' number
	 0
	 0
	 0
	 1
	 1
2
	 0
2
	 T
	 0
	 0
	 0
— 1
	 1
2
	 0
3
	 i
10 ppn, and 5 ppm, / hr/


se of the high toxicltv

sites


Peri en t ^
	
	

I.I
I.I
I.I
l.l
	
2.2
	
	
	
. . i
1 .1
1 . 1
	
3.3
1.7
day, 5 day





others

Total
numbe r
9
1 1
20
14
b
20
4
4
8
Ib
9
25
8
4
12
7
8
n
s wk, for 78 wk.


Ben l^n
numbe r
7
5
12
14
3
17
2
3
5
15
5
20
7
2
9
7
5
12



Ma 1 1 pnant
uumbe r
2
ft
8
0
3
3
2
1
3
1
4
5
1
2
5
0
)
3




Number
15
54
f.9
20
5b
76
1 )
43
5b
30
h5
95
17
38
55
14
5b
70



Total -f
Number nf
dl f f'.rent
t urn-i r=: •' t iimr. r-
IVrrent ho.irinq l"ln,il'
1 h . 9 1 . _>
h 1 1 . O 2 . ' '
)8.5 1.?
22.2 1 . »-
b2.2 1.7
42.2 1 . '
14. b 1 .1
47.R 1...
H . 3 1.5
33.3 1.4
72.2 I.1-
52.8 1 . "
18.9 1 . 1
'i>.: i . u
30. h 1."
1 5 . b I.I
b2.: i .
<«.9 1.1,



at the 250 ppm level.

-------
                                        TABLE
                                               9-50
MAMMARY TUMORS IN SPRAGUE-DAWLEY RATS
    EXPOSED TO EDC
(Maltoni et al. 1980)
vO
OJ
Animals
Group Concentration Sex
I 250-150 ppmd M
F
M and F
II 50 ppm M
F
M and F
III 10 ppm M
F
M and F
IV 5 ppm M
F
M and F
V Controls in M
chambe rs F
M and F
VI Controls M
F
M and F
Total
Number
at start
90
90
180
90
90
180
90
90
180
90
90
180
90
90
180
90
90
180
1080
Corrected
number3
89
90
179
90
90
180
89
90
179
90
90
180
90
90
180
90
90
180
1078

Total
numbe r
11
52
63
10
58
68
5
43
48
11
65
76
8
38
46'
5
52
57

Mammary
Percent15
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

tumors
Average
latency
time
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
tumors/tumor-
bearing animals
1
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
. — i
                                                                                   (continued on the following page)

-------
                                                                TABLE  9-50  (l-onttnuod)
Mammary tumorse
HlsLolotjlcal evaluation
Mammary tumors

Group Concentration Sex
I 250-150 ppm 4.1 7 i . 0
2 3.7 M.I)


^Fhc percent n^cs ref e r to  the  corrected mmbe rs.
cAverage  age  at the onset  of  first mamma ry  tumor per aninwl,  dctpctfd at the periodic control  or  at  autopsv.
^After a  Few  weeks the dose was reduced to  150 ppm, because  of  the hiRh toxirlty  at  the   tunv> r <; nf MIP  same or different  tvpes (flbro.ldr nnrn;i ^ ,  r a rri nnm \ ^ ,  >>a rr oms ,  c.i rri nos ;i rcoTiai ) nnv
 be present  in the same animal.
* The pe rcentaj^e^ rt'f •=> r t n  tor al numbe r of 
-------
 chamber,  but  not  significant when compared  to  controls  outside  of  the  chamber.




 Although  the  author  stated  that  the  onset of fibromas and  fibroadenomas was




 age-correlated, the  incidences of these  tumors  observed  in the  various 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.







 9.5.1.5   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 ppm, and 5 ppm, respectively, 7 hours per day, 5 days per week, for 78




 weeks.  After  several days of 250 ppm exposure, the mice 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 until




 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%.
                                  9-195

-------
            TABLE  9-51.   DESIGN SUMMARY FOR 1,2-DICHLOROETHANE (EDC)
                           EXPERIMENT IN SWISS  MICEa
                             (Maltoni et al. 1980)
Animals"
Group Concentration Sex
I 250-150 ppmc M
F
II 50 ppm M
F
III 10 ppm M
F
IV 5 ppm M
F
V Control M
F
Numbe r
90
90
90
90
90
90
90
90
115
134
aExposed 7 hours/day, 5 days/week, for 78 weeks.
bSwiss mice, 11 weeks old at start.
cAfter a few weeks the dose was reduced to 150 ppm,  because of high
 toxicity at the 250 ppm level.
     The results of histopathologic analysis of various tumors are shown in

Table 9-53 .  These results do not indicate a statiscally 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 dose levels that appear

to have been less than maximally tolerated doses.
                                   9-196

-------
                       TABLE  9-52  .  SURVIVAL OF SWISS MICE EXPOSED TO EDC AT 52 and 78 WEEKS3
                                             (Maltoni et al. 1980)
Survivors at

Dose
Group (ppm) Sex
I 250-150 ppmb M
F
II 50 M
F
III 10 M
F
IV 5 M
F
V Control M
F

Initial
numbers
90
90
90
90
90
90
90
90
115
134
52
of
Number
56
75
68
83
74
86
54
84
91
127
weeks
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
of
Number
26
44
30
49
34
50
26
68
42
76
weeks
age
Percent
28.9
48.9
33.3
54.4
37.8
55.6
28.9
75.6
36.6
56.8
Survivors
52 weeks
start of
after
from
study
Number Percent
39
58
46
73
59
72
42
84
72
112
43.3
64.4
51.1
81.1
65.6
80.0
46.7
93.3
62.6
83.6
      were 11 weeks old at the  start of the experiment.
^After a few weeks the dose was reduced to 150 ppm, because of high toxicity at the 250 ppm level.

-------
| |UUJ )
                                                                                                                       646
4V4 4'x 17 b'Vt (M X
t ' 14 (Ml 51 7/15 O'l 1
W54 ','5 4 5'Bi 4'1 •?
iv< re n x'4z 7't i
X ' (> 4 / ' 4 4 0 ' i B 5 ' V '/
ll'hl 6V 7 U'V1? V'l 1
r'\t 7'4 1 1 0'iZ I'l t
4'64 ^'5 5 5'1 5 1 V Z
5''/5 ft 4 r^B 4'» i?
•J-S4 /•«, 1)1 U'',/ 6V <,
7'6(, 4-t; i; ^'t// fV 7
h'64 6'4 4 f'W Vf {
0'64 i"t ? Z'iH H'l I
li'O ri 1 ^'tW 9'C (
ll'i/ Z' 1 1 	 	 0
Ijl1)") 31U->TJ"H 4rf>|UIIU p(1") j3U.I.O-lu,| JjqUJPIU
r»ui|) lt3"J. .MU |i-'l"l
A-'U J 1 1' [ A JU<1 j I/ |
dJt'JHA\' t)HtJ.)AV
•.t )uja^l)R'| bt?UUUK1|>l' A 1 L'UIHU | l)t|
SJOUI1) LHlrt <'IU(UV
3 dnoj;)













QO

i








                                                     (OHM  'I1'  )•' l»"l|iw)
                                        O.L (HM)dX.H  H:)1H  S.SlttS  Nl   IIN'*II.JW1
                                                                                           t"C— A

-------
fABLF.  9-53  (continued)


Animals with tumors



Anglosarcomas




Croup Concentration Sex
1 2r)0-lSU ppm1" H
F
M and F
v-° II 50 ppm H
.1 F
VO M and F
III 10 ppm M
F
M and F
IV 5 ppm M
F
M and F
V Controls In M
rhnmbers F
H and F
VI Controls M
F
H and F
Nephroblastomas
Ave rage
latency
Total time
number Percent0 (wk)"1
II 	 	
0 	 	
0 	 	
0 	 	
0 	 	
0 	 	
0 — 	
o
(I — —
(1 — —
0 — 	
1) — —
0 — 	
0 	 	
0 	 	
0 	 	
0 	 	
0 	 	
Kidney adenocarclnomas
Average
latency
Total time
number Percent0 (wk)^
0 	 	
0 	 	
0 	 	
(1 — —
(1 — —
,)
o
0 	 	
0 — 	
0 	
1 1.1 «.2.U
1 0.6 62.0
0
n — -_.
0 	 	
I)
(1 — —
I) 	 	
Liver
Ave r.ige
latency
Total time
number Percent0 (uk)1'
0
0 	 	
0 	 	
(1 — —
1) — 	
0 	 	
0 	 	
u — —
„
0 	
0 	 	
0 	 	
0
U 	 	
(1 — —
0 	 	
{1 	 	
1) — —



Total
number
1
- 0
1
0
CP
0
0
I)
(1
o
0
0
0
u
0
1
0
1
Other sites
Aver, --Re
latencv
t imp
Pert/put c (wk )
1.2 -io.ii
	 	
O.h 4(1. u
	 	
	 	
	 	
	 	
	 	
—
125. o
131.0
	 UH.ll
	 	
	 	
	 	
(1.9 '^.0
	 	
0.4 M.O
                                                                                          (continued on  the  fnllnuliiK

-------
TABI.K  9-53  (r/HitlniM-il)








vO
to
0
O

















Group Concentration Sex
1 2V.-I5" ppm" M
F
M and F

1 1 VI ppm H
F
M and F
lit HI ppm M
F
M and F
IV 5 ppm M
F
M and K
V Omtrolq In M
chamber* F
M a fid F

Angfoma'
Liver
Average
latent y
Tot a 1 1 1 me
mimher Percent c (wk )^
II 	 	
n 	 	
n — —

ti — —
1 1.1 81.0
1 <>.ft fll.ll
n — —
n — —
o — —
i) — —
0 — —
it — —
II 11.9 Ii9.ll
II 	 	
I) O.'i h4.ll

; and f Ihronngiomas
Other iltei
Aver.tge
1 a I enc v
Tot a I 1 1 me
number Percent1" (wk)<*
1, 	 	
II 	 	
II 	 	

1 I.I 7h.ll
1 I.I 83.11
2 I.I 79. "i
II --- 9t.(l
1 I.I 91.11
1 II. h
H 	 	
(1 	 	
11 	 	
1 n.9 76.11
11 	 	
1 11.4 7h.H


Hepiit omai
Ave rage
latency
Total time
number Percentr (wk)1*
11 	 	
O 	 	
(I 	 	

()
M 	 	 	
o
0 	 	
(1 — —
0 	 	
M.
1) — —
0 	 	
4 !.•> R2.ll
O 	 	
4 l.h H2.ll


Knre* t ornach epithelial
tumors
Avenue
1 .1 lenc v
Total ( Ine
number Pnrrentr (wV I'1
(I 	 	
II 	 	
() 	

II 	 	
II 	 	
(1 	
II 	
|l 	 	
(1 	 	
II 	 	
1) 	 	
II 	 	
II 	 	
II 	 	
II 	 	
                                                                                                              (r.mt i.i.u-.l on  t lie  rnllowlnr  p

-------
TMMK   9-53
Anlmils wtth Lunvirs




Uronp (>>ncent rat Ion Sex
I J5H-I5II ppm* H
F
'1 an.l
M ''ii ppm M
vO f
1 M and
rJ
3 HI 1" Prra M
F
M ami
l
IV 'i ppn M
F
M and
V Controls In M
rlnmbprs F
H and
Exposure by Inhalation to KUC In
Results niter 119 weeks (end of
"Animals alive after 24 wkf when
"Average time from the start of
eAfter a few weeks the dose was
Skin


Total
number
1)
(1
F ('
I)
1
F 1

(1
0
F (I

II
1
F 1
1
1
F 2
air at 2511-
experlment )
t lie first t
the t'xperlme
reduced to 1
epithelial tumors Subcutaneous saicotnas
Averapp Average
lat en< v latent v
t l"ip Total t Imp
Percent0 (wk )"* number I'erci-iii'" (wk)"1
l,
	 — o 	 —
	 	 o 	 	
1,
I.I IIK.II o — —
o.n lui.n (i — —

	 	 1 I.I 77. II
1 I.I 25.11
2 I.I SI. il

	 — 0 	 	
1.1 7'>.tl I)
O.b 79.11 II 	 	
'1.9 lOt.tl 1 !).<> IDh.ll
11.7 77.O II 	 	
I'.K 91.11 U (1.4 IDh.d
1511 ppm, 5(1 ppm, 10 ppm, ami ^ ppm, 7 hr/tl.iv, 5 davs/wk,
nmor (a lenkemli) was observed.
nt to netettlon .it the period)' (ontrol or at .nitopsy.
V pfm, he'-.iiisr of hlRh toxi> Itv .'t the .|r»o ppm level.



Total
nninbe r
II
2
2
1
It
s

1
2
1

1
6
5
1
4

lor 7H


Others


Be 11 it'll
number
H
2
2
II
(
1

1
1
2

;l
)
1
1
2

t-k.





Malignant
nnmhe r
II
(1
(1
1
1
2

(1
1
1

1
1
2
y
2
2







Nnmbe r
2
U
1 1
9
15
2
-------
9.5.1.6  Theiss et al. (1977) Mouse Study—Theiss et al.  (1977) conducted a

pulmonary tumor bioassay with EDC and other organic contaminants of drinking

water in the United States.  The compounds were injected  intraperitoneally 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

examined 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 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 non-significant 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 EDC
                             (Theiss et al. 1977)
Dose/injection Number of
(mg/kg) injections
Oa 24
20 24
40 24
100 24
Fraction
surviving
46/50
14/20
16/20
20/20
(92%)
(70%)
(80%)
(100%)
Number of
tumors/mouse
0.39 _+ 0.06
0.21 +_ 0.06
0.44 +_ 0.11
0.75 + 0.17
aTricaprylin vehicle
                                     9-202

-------
9.5.1.7  Van Duuren et al. (1979) Mouse Study—Van Duuren et al. (1979) con-




ducted a bioassay of EDC and of a suspected metabolite, chloracetaldehyde, as




initiators, promoters, and complete carcinogens using two-stage skin tests.




Female non-inbred ICR/Ha Swiss mice were used, with treatment beginning at 6-8




weeks of age.  The experimental group consisted of 30 animals, exclusive of the




no-treatment groups or those receiving repeated application of phorbol myristate




acetate (PMA).  The results of this study are given in Table 9-55 .  The com-




pounds, in 0.1 ml 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 recorded




monthly.  Animals in poor health or with large tumor masses were killed.  Ani-




mals were completely autopsied at the termination of the experiment or at death.




Tissue sections were fixed in 10% formalin, processed, blocked in paraffin, and




stained with hematoxylin and eosin for pathologic diagnosis.  The results indi-




cate that neither EDC nor chloroacetaldehyde induced a statistically 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 statis-




tically significant increase in the incidence of benign lung papillomas.




     In 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 DMA 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.
                                       9-203

-------
                               TABLE 9-55,   MOUSE SKIN BIOASSAY OF 1 ,2-OICIILOROKTHANE  ANU  CHLOROACETAI.DEHYDE
                                                        (adapted from Van Duuren el  al.  1979)
Initial ton-promot iona
Dose Days lo
mg/appllcat Ion/ first
Compound mouse tumor
1,2-IHchloroethane 12b.O 337


Chloroac.'taldehvde 1.0 152
1
O PMA controls
*• (120 mice) 0.0025 141
(90 mice ) 0.0050 449
Acetone (0.1 ml) 	 	
No treatment
(100 mice)

Repeated application15
Mice with Dose Days to Mice with No. of mice
paplllomas0/ mg/appll ca t ion/ first papillomasC/ with distant
total paplllomas mouse tumor total paplllomas tumors"
J/3 12h.O 	 0 2(i lung
P<0.0005
3 stomach
42.0 	 0 17 lung
1 stomach
3/J (1 ) 1.0 	 0 14 luii);
1 stomach


9/10 (1)
6/7 (2)
	 0.1 ml 	 0 1 1 lung
2 s tomach
	 	 	 0 30 lung
5 stomach
aAll applications were to  the  dorsal  skin  by  mlcroplpette.   1 ,2-dl chlorocthane was administered once  only  in 0.2
 ml acetone, followed 14 days  later  by  5 ug of  PMA  In  O.2 ml  acetone three times weekly.  Chloroaceta1dehyde
 was administered once only  in  O.I ml  acetone,  followed  14  days  later  by 2.5 up, of PMA in 0.1  ml  acetone  three
 times weekly.
" I , 2-d 1 rh 1 o roe t hane was administered  In 0.2 ml  arrlone.  Ch loroace t al dehyde was administered lliroe  t imns  weekly
 in O.I ml  acetone.    (
cNnmbe r of  mice with srjnamous  cell carcinomas arc given  in  a  parentheses.
dA! I lung tumors are benign  paplllomas; stomach tumors are  paplllomas  of the forestomach and squamou--- cell  ca I—
 clnomas of the forestomach.   P values  are given only  where  significant, i.e., P < (1.05.

-------
     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).  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 con-




ducted 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 con-




cluded that unless unusual and unexpected synergisms occurred, the presence of




other animals in the test rooms would not have affected the results.




     Reitz et al. (1982) proposed that basic metabolism rate differences,




specifically the saturation of the detoxification/excretion mechanism, occurred




between the single gavage dose (and presumably other routes) and the longer-




term inhalation dose.  Their calculations suggest that EDC is metabolized by




non-linear kinetics, and that the inhalation study did not produce peak blood




levels sufficiently high to overcome the detoxification mechanism and produce




a detectable incidence of tumors.




     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.  Thus, the




results indicate the EDC is carcinogenic in rats and mice, and is a weak




mutagen.







9.5.2  Epidemiologic Studies




     No epidemiologic studies have been published regarding the carcino-




genicity of EDC.
                                      9-205

-------
9.5.3  Quantitative Estimates




     This quantitative section deals with estimation of the unit risk for EUC




as a potential carcinogen in air and water, and with the potency of EDC rela-




tive to other carcinogens that have been evaluated by the CAG.  The unit risk




for an air or water pollutant is defined as the lifetime risk of cancer to




humans from daily exposure to a concentration of 1 ug/nH of the pollutant in




air by inhalation, or to a concentration of 1 ug/L in water by ingestion.




     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 evaluating environmental hazards.  For practi-




cal reasons the correspondingly low levels of risk cannot be measured directly




either by animal experiments or by epidemiologic studies.  Low-dose extrapola-




tion 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 irre-




versible damage to DNA.  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 muta-




enesis is associated with a linear (at low doses) non-threshold dose-response




relationship.  Indeed, there is substantial evidence from mutagenicity studies




with both ionizing radiation and a wide variety of chemicals that this type  of




dose-response model is the appropriate one to use.  This is particularly true at




the lower end of the dose-response curve; at higher doses, there can be an




upward curvature, probably reflecting the effects of multistage processes on




the mutagenic response.  The linear non-threshold dose-response relationship
                                        9-206

-------
is also consistent with  the relatively few epidemiologic studies of  cancer




responses to specific agents that contain enough information to make the eval-




uation 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 evidence also exists from animal




experiments (e.g., the initiation stage of the two-stage carcinogenesis model




in rat liver and mouse skin).  Linearity is also supported when the mode of




action of the carcinogen in question is similar to that of the background can-




cer production in the exposed population.




     Because its scientific basis, although limited, is the best of any of the




current mathematical extrapolation models, the non-threshold model, which is




linear at low doses, has been adopted as the primary basis for risk extrapo-




lation to low levels of the dose-response relationship.  Any risk estimates




made with such a model should be regarded as conservative,  representing the




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.




     The unit risk estimate based on animal bioassays is only an approximate




indication of the absolute risk in populations exposed to known carcinogen




concentrations.  This is true for several reasons.  First,  there are important




species differences in uptake,  metabolism,  and organ distribution of carcino-




gens, as well as species differences in target site susceptibility, immuno-




logical responses, hormone function, dietary factors, and disease.   Second,




the concept  of equivalent doses for  humans  as compared to animals,  based on




a ratio of weight to surface area,  is virtually without experimental verifi-




cation  as  regards carcinogenic  response.   Finally,  human populations are




variable with respect to genetic constitution and diet, living environment,




activity  patterns, and other cultural factors.
                                        9-207

-------
     The unit risk estimate can give a rough indication of the relative




carcinogenic potency of a given agent, as compared with that of 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.




     The quantitative aspect of carcinogen risk assessment is addressed here




because of its possible value in the regulatory decision-making process, e.g.,




in setting regulatory priorities, evaluating the adequacy of technology-based




controls, etc.  However, the imprecision of presently available technology




for estimating cancer risks to humans at low levels of exposure should be




recognized.  At best, the linear extrapolation model used here provides a




rough but plausible estimate of the upper limit of risk—that is, with this




model it is not likely that the true risk would be much more than the estimated




risk, but it could be considerably lower.  The risk estimates presented in




subsequent sections should not be regarded, therefore, as accurate representa-




tions of the true cancer risks even when the exposures involved are accurately




defined.  The estimates presented may, however, be factored into regulatory




decisions to the extent that the concept of upper-risk limits is found to




be useful.






9.5.3.1  Procedures for the Determination of Unit Risk—






9.5.3.1.1  Low-dose extrapolation model.  The mathematical formulation chosen to




describe the linear non-threshold dose-response relationship at low doses is the




linearized multistage model.  This model employs enough arbitrary constants to




be able to fit almost any monotonically increasing dose-response data, and it




incorporates a procedure for estimating the largest possible linear slope (in
                                      9-208

-------
 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 H


 where
                           qi  >_ 0,  i =  0, 1,  2,  ....  k


 Equivalently,

                   Pt(d)  = 1  - exp [-(qj^d  +  q2d2 + ... +  q^d1*-)]


 where

                               P  (d)  =  P(d) - P(0)
                               t         1  - P(0)


 is the extra risk over background  rate  at  dose d.

      The  point estimate of the coefficients q^, i =  0, 1, 2,  ...,  k,  and

 consequently,  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, GLOBAL79, developed  by

 Crump and Watson (1979).  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 qj.  When-

ever qj > 0, at low doses  the extra risk Pt(d) has approximately the form

Pt(d) = q* 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.
                                      9-209

-------
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 q* for the linear




coefficient, the resulting maximum value of the log-likelihood Lj satisfies the




equation







                         2 (L0 - LX) = 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 alway linear.  This is conceptually




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.)




     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 experiment,




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
                            NiPi
                                      9-210

-------
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 i   dose group estimated by fitting the multistage


model to the data, and h is the number of remaining groups.  The fit is determined

                              P
to be unaccepatable 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 coefficents.


9.5.3.1.2  Selection of data.  For some chemicals, several studies in different


animal species, strains, and sexes, each run at several doses and different


routes of exposure, are available.  A choice must be made as to which of the


data sets from several studies to use in the model.  It may also be appropriate


to correct for metabolism differences between species and for absorption factors


via different routes of administration.  The procedures used in evaluating


these data are consistent with the approach of making a maximum-likely risk


estimate.  They are as follows:


     1.  The tumor incidence data are separated according to organ sites or


tumor types.  The set of data (i.e., dose and tumor incidence) used in the


model is the set where the incidence is statistically significantly higher


than the control for at least one test dose level and/or where the tumor incidence


rate shows a statistically significant trend with respect to dose level.  The

data set that gives the highest estimate of the lifetime carcinogenic risk,


q.j*, is selected in most cases.  However, efforts are*made to exclude data


sets that produce spuriously high risk estimates because of a small number


of animals.  That is, if two sets of data show a similar dose-response relationship,


and one has a very small sample size, the set of data having the larger sample


size is selected for calculating the carcinogenic potency.
                                      9-211

-------
     2.  If there are two or more data sets of comparable size that are


identical with respect to species, strain, sex, and tumor sites, the geometric


mean of q*, estimated from each of these data sets, is used for risk assessment,


The geometric mean of numbers A^, A£, •••, Am is defined as




                            \" i  X Ao X ••* X A )




     3.  If two or more significant tumor sites are observed in the same study,


and if the data are available, the number of animals with at least one of the


specific tumor sites under consideration is used as incidence data in the model.




9.5.3.1.3  Calculation of human equivalent dosages.  Following the suggestion of


Mantel and Schneiderman (1975),  it is assumed that mg/surface area/day is an


equivalent dose between species.  Since, to a close approximation, the surface


area is proportional to the two-thirds power of the weight, as would be the case


for a perfect sphere, the exposure in mg/day per two-thirds power of the weight


is also considered to be equivalent exposure.  In an animal experiment, this


equivalent dose is computed in the following manner.


Let


     Le = duration of experiment


     le = duration of exposure


     m = average dose per day in mg during administration of the agent (i.e.,

         during le), and


     W = average weight of the experimental animal


The lifetime exposure is then
                         d -
                             L  x W2/3
                              e
                                     9-212

-------
      9.5.3.1.3.1   Oral.   Often exposures are not given in units of mg/day,  and




 it becomes  necessary to  convert the given exposures into mg/day.   Similarly,  in




 drinking water studies,  exposure is expressed as ppm in the water.  For example,




 in most feeding studies  exposure is given in terms  of ppm in the  diet.   In




 these cases,  the  exposure in mg/day is






                                 m = ppm x F  x r






 where ppm is  parts  per million of the  carcinogenic  agent  in the diet  or water,




 F  is  the weight of  the food  or water consumed per day in  kg,  and  r is the




 absorption  fraction.  In  the absence of  any  data to the  contrary,  r is  assumed




 to be equal to one.   For  a uniform diet,  the  weight of  the  food consumed is




 proportional  to the  calories required, which  in  turn  is proportional  to the




 surface  area,  or  two-thirds  power of the  weight.  Water demands are also




 assumed  to  be  proportional to  the surface  area,  so  that
                                m a ppm x




or
                                     W2/3
                                            ppm
As a result, ppm in the diet or water is often assumed to be an equivalent




exposure between species.  However, this is not justified for the present




study, since the ratio of calories to food weight is very different in the




diet of man as compared to laboratory animals, primarily due to differences




in the moisture content of the foods eaten.  For the same reason, the amount




of drinking water required by each species also differs.  It is therefore




necessary to use an empirically derived factor, f = F/W, which is the




fraction of an organism's body weight that is consumed per day as food,




expressed as follows:
                                   9-213

-------
                                            Fraction of body
                                           weight consumed as


                    Species         W       ffood      ^water
Man
Rats
Mice
70
0.35
0.03
0.028
0.05
0.13
0.029
0.078
0.17
Thus, when the exposure is given as a certain dietary or water concentration in


ppm, the exposure in mg/W^/3 ±s



                             * F = ppm x f x W = p   x f x w1/3
                 rW2/3    w2/3         w2/3



When exposure is given in terms of mg/kg/day = m/Wr = s, the conversion is


simply


                                  m   = s x W1/3
                                rW2/3





     9.5.3.1.3.2  Inhalation.  When exposure is via inhalation, the calcula-


tion of dose can be considered for two cases where 1) the carcinogenic agent is


either a completely water-soluble gas or an aerosol and is absorbed propor-


tionally to the amount of air breathed in, and 2) where the carcinogen is a


poorly water— soluble gas which reaches an equilibrium between the air breathed


and the body compartments.  After equilibrium is reached, the rate of absorption


of these agents is expected to be proportional to the metabolic rate, which in


turn is proportional to the rate of oxygen consumption, which in turn is a


function of surface area.



     9.5.3.1.3.2.1  Case 1 — Agents that are in the form of particulate matter or


virtually completely absorbed gases, such as sulfur dioxide, can reasonably be
                                         9-214

-------
 expected to be absorbed proportionally to the breathing rate.  In this case the

 exposure in mg/day may be expressed as

                                m = I x v x r


 where 1 = inhalation rate per day in m3, v = mg/m3 of the agent in air, and

 r = the absorption fraction.

      The inhalation rates, I, for various species can be calculated from the

 observations of the Federation of American Societies for Experimental Biology

 (FASEB 1974) that 25 g mice breathe 34.5 liters/day and 113 g rats breathe 105

 liters/day.  For mice and rats of other weights,  W (in kilograms), the surface

 area proportionality can be used to find breathing rates in m3/day as follows:


                    For mice, I = 0.0345 (W/0.025)2/3 m3/day

                    For rats, 1 = 0.105 (W/0.113)2/3 m3/day


 For humans, the value of 30 m3/day* is adopted as a standard breathing rate

 (International Commission on Radiological Protection 1977).  The equivalent

 exposure in mg/W2/3 for these agents can be derived from the air intake data

 in a way analogous to the food intake data.  The  empirical factors for the air

 intake per kg per day,  i = I/W, based upon the previously stated relationships,

 are tabulated as follows:


                     Species            W           i = I/W
Man
Rats
Mice
70
0.35
0.03
0.29
0.64
1.3
*From "Recommendation of the International Commission on Radiological Protection,
 page 9.   The average breathing rate is 10'  cm3 per 8-hour workday and 2 x 10'  cm
 in 24 hours.
                                         9-215

-------
Therefore, for particulates or completely absorbed gases, the equivalent




exposure in mg/W^/-' is
                        d =   m  = Ivr  = iWvr =

                            w2/3   w2/3   w2/3






     In the absence of experimental information or a sound theoretical argument




to the contrary, the fraction absorbed, r, is assumed to be the same for all




species.






     9.5.3.1.3.2.2  Case 2 — The dose in rag/day of partially soluble vapors  is


                                                                      2 / 3
proportional to the 02 consumption, which in turn is proportional to W '  and




is also proportional to the solubility of the gas in body fluids, which can be




expressed as an absorption coefficient, r, for the gas.  Therefore, expressing



                              2/3
the 0? consumption as Oo = k W '  , where k is a constant independent of species,




it follows that




                            m = k W^/3 x v x r




or




                            d -   m   - kvr

                                w2/3






As with Case 1, in the absence of experimental information or a sound theoretical




argument to the contrary, the absorption fraction, r, is assumed to be the  same




for all species.  Therefore, for  these substances a certain concentration in




ppm or ug/m3 in experimental animals is equivalent to the same concentration




in humans.  This is supported by  the observation that the minimum alveolar




concentration necessary to produce a given "stage" of anesthesia is similar




in man and animals (Dripps et al.  1977).  When the animals are exposed via




the oral route and human exposure  is via inhalation or vice versa,  the
                                        9-216

-------
 assumption  is made, unless  there  is pharmacokinetic evidence  to  the  contrary,


 that  absorption is equal  by either exposure route.



 9.5.3.1.4   Calculation of the unit risk from animal studies.  The risk associated


 with  d mg/kg2/3/day ±s obtained from GLOBAL79 and, for most cases of interest  to


 risk  assessment, can be adequately approximated by P(d) = 1 - exp (-q*d).  A


 "unit risk" in units X is simply  the risk corresponding to an exposure of X =  1.


 This  value  is estimated simply by finding the number of mg/kg2/3/day that cor-


 responds to one unit of X,  and substituting this value into the  above relation-


 ship.  Thus, for example, if X is in units of ug/m3 in the air,  then for case


 1, d  = 0.29 x 701/3 x 10~3  mg/kg2/3/day, and for case 2, d =  1, when ug/m3 is


 the unit used to compute parameters in animal experiments.


      If exposures are given in terms of ppm in air, the following calculation


 may be used:


                     1 ppm  = 1.2 x molecular weight (gas) mg/m3
                                   molecular weight (air)



 Note  that an equivalent method of calculating unit risk would be to use mg/kg


 for the animal exposures, and then to increase the jt*1 polynomial coefficient


 by an amount


                        (Wh/Wa)J/3  j = 1,  2,  .... k



 and to use mg/kg equivalents for the unit risk values.



     9.5.3.1.4.1  Adjustment for less than  the natural  lifetime of an experiment.


 If the duration of experiment Le is less than the  natural lifespan of the test


animal L,  the  slope q*,  or more generally the  exponent  g(d),  is increased by

                           o
multiplying a  factor (L/L£)  .   We  assume that  if  the  average  dose d  is  continued,


 the age-specific rate  of  cancer will continue  to  increase as  a constant  function
                                         9-217

-------
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 demon-


strated 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 q*, or more generally the exponent g(d), would also


increase by at least the third power of age.  As a result, if the slope q*  [or


g(d)] is calculated at age Le, it is expected that if the experiment had been


continued for the full lifespan L at the given average exposure, the slope q*

                                                      o
[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)]



9.5.3.2  Unit Risk Estimates—


9.5.3.2.1  Data available for potency calculation.  The carcinogenic potency of


EDC can only be estimated from animal data, since no human epidemiologic studies


are available at  the present  time.  EDC was shown to be carcinogenic  in both


rats and mice in  a gavage study  by the National Cancer Institute (NCI  1978).   The


only available inhalation study  (Maltoni  et al. 1980) did not  indicate any  signi-


ficant  carcinogenic effects  in either rats or  mice.  The  reason  for  this non-


responsiveness by inhalation is  not known.  In  their evaluation  of  inhalation


and gavage studies on  EDC, Hooper et al.  (1980) concluded that the  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;
                                        9-218

-------
     (2)  The route of exposure does make a difference concerning the carcino-
          genic action of EDC; or

     (3)  An artifact has been introduced by the intercurrent mortality.

    Two approaches have been used herein to estimate the cancer risk from

inhalation of EDC:  direct estimation based on the results of the EDC gavage

study, and indirect estimation based on the results of ethylene dibromide (EDB)

inhalation and gavage studies.

     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 (see

         Table 9_56.

     2.  Time-to-death data (see Table B-l,  Appendix B).

     For mice, hepatocellular carcinomas in male mice (Table 9-57 ) were used.

Again, this tumor site was selected because it is the most sensitive in mice.


9.5.3.2.2  Choice of low-dose extrapolation models.  In addition to the multi-

stage model currently used by the CAG for low-dose extrapolation,  three more

models, the probit, the Weibull, and the one-hit, are also employed for pur-

poses of comparison.  These models cover almost the entire spectrum of risk

estimates that could be generated from existing mathematical extrapolation
                                     9-219

-------
       TABLE  9-5$  INCIDENCE RATES OF HEMANGIOSARCOMAS IN THE CIRCULATORY
                       SYSTEMS OF MALE OSBORNE-MENDEL RATS
                                   (NCI 1978)
      Human (animal)
     dose (mg/kg/day)a                          Incidence rates
0
4.85 (47)
9.80 (95)
0/40
9/48
7/27
aHuman equivalent dose is calculated by d x (5/7) x (78/104) x (D.5/70)1/^
 = 0.103 x d, where d is the experimental dose in mg/kg/day, administered
 5 days per week for 78 weeks.  The lifespan for rats is assumed to be 104
 weeks.  The body weights are assumed to be 0.5 kg for rats and 70 kg for
 humans.  This dose conversion assumes that the doses in mg per surface area
 are equivalent between species.  For comparison, the risk is also calculated
 under the assumption that doses in mg/kg/day (i.e., without surface cor-
 rection) are equivalent between species.
             TABLE 9.57. INCIDENCE RATES OF HEPATOCELLULAR CARCINOMAS
                               IN MALE B6C3F1 MICE
                                   (NCI 1978)
      Human (animal)
     dose (mg/kg/day)a                          Incidence rates
          0                                          1/19
          4.76 (97)                                  6/47
          9.57 (195)                                 12/48

aHuman equivalent dose is calculated by d x (5/7)  x (78/90)  x (0.035/70)WJ
 = 4.91 x 10~2 x d, where d is the animal dose in  mg/kg/day,  administered
 5 days per week for 78 weeks.  The lifespan for mice is assumed to be 90
 weeks.  The body weights are 0.035 kg for male mice and 70  kg for humans.
 For comparison, the risk is also calculated under the assumption that doses
 in mg/kg/day (i.e., without surface correction) are equivalent between
 species.
                                9-220

-------
models.  Generally statistical in character, these models are not derived




from biological arguments, except for the multistage model, which has  been




used to support the somatic mutation hypothesis of carcinogenesis (Armitage




and Doll 1954, Wittemore  1978, Whittemore and Keller 1978).  The main




difference among these models is the rate at which the response function




P(d) approaches zero or P(0) as dose d decreases.  For instance, the probit




model would usually predict a smaller risk at low doses that 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 multi-




stage 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 para-




meters in the multistage model are zero.  This, of course, is not reasonable




without knowing, a priori, what the carcinogenic process for the agent is.




Although the multistage model appears to be the most reasonable or at  least




the most general model to use, the point estimate generated from this model




is of limited value because it does not help to determine the shape of the




dose-response curve beyond experimental exposure levels.  Furthermore, point




estimates at low doses extrapolated beyond the experimental doses could be




extremely unstable and could differ drastically,  depending on the amount of




the lowest  experimental dose.  Since upper-bound estimates from the multi-




stage model at low doses are relatively more stable than point estimates,




it is suggested that  the upper-bound estimate for the risk (or the lower-




bound estimates for the dose) be  used in evaluating the carcinogenic potency




of a suspect carcinogen.  The upper-bound estimate can  be taken as a




plausible estimate if  the true dose-response curve is actually linear at




low doses.   The upper-bound estimate means that the risks are not likely to




be higher,  but could be lower if  the compound has a concave  upward dose-
                                 9-221

-------
response curve or a threshold at low doses.   Another reason one can, at




best, obtain an upper-bound estimate of the  risk when animal data are used




is that the estimated risk is a probability  conditional to the assumption




that an animal carcinogen is also a human carcinogen.  Therefore, in




reality, the actual risk could range from a  value near zero to an upper-




bound estimate.






9.5.3.2.3  Calculation of the carcinogenic potency of EDC.  Using the incidence




data in Tables 9-56 and 9-57  and tne corresponding human equivalent doses, with




and without body surface correction, the maximum likelihood estimates of the




parameters in each of four extrapolation models are calculated and  presented in




Table A-l of Appendix A.  These models can be used to calculate point estimates




of risk at given doses or at doses for given levels of risk.  The upper-bound




estimates of the risk at 1 mg/kg/day, calculated by various models  using




different data sets, are presented in Table 9-58'  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 Weibull models are extremely unstable and predict  a wide




range  of risk, depending on the  data base used.  Figures  9~12and 9-13compare




the  maximum likelihood estimates of the four models  over  the  relatively low




dose range.




     For the  reasons discussed  previously, the  CAG  recommends  that  the




estimate q* =  6.9 x  10~2 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




factor.  This  value  will  also  be used  to  estimate  the  risk  of  EDC by




inhalation.
                                          9-222

-------
                                       TABLE  9-58.   UPPER-BOUND ESTIMATE OF RISK AT  1 MG/KG/DAY
*£>

NJ
NJ
OJ
Data base
Hemangiosarcomas ,
dose with surface
correction
Hemangiosarcomas ,
dose without
surface correction
Hepatocellular carcinomas,
dose with surface
correction
(with
5.4
(6.
7.0
(1.
3.8
Multistage
time factor) Probit Weibull One-hit
x 10~2 0.29 0.28 5.4 x 10~2
9 x 10~2)
\
x 10~3 0.17 0.19 7.0 x ID'3
3 x ID"2)
x ID'2 2.4 x 10-2 4.3 x 10-2 3>8 x 10-2
        Hepatocellular  carcinomas,
           dose without  surface
           correction
3.0 x 10~3
1.1  x 10~5         1.9 x 10~3       3.0 x  10~3

-------
NJ
                 O



                 PI
   o  o
   fi  H'
    I  3
   D. it
   O
C  cn  D>
H  (D  3
iti    Q.
CD  >-<
o  n>
n>  OP
                                                                                                            Carcinogenic  Response
          en
       c
      •a

n o  rt>
o 3  n

i-i o  a*
n> 3  Q
o
             n>
                3
      
      cn
   P> rt
   cn H-
•^^ H- B
2 cn &
O    rt
M o rt>
   MI cn

vo 3- o
          oo
             s  o
             cm  c
             H- M
             o
             cn  Q.
             0)  O
             i-t  cn
             o  n>
             O   I
                g^
                (I
             cn  cn
             3  3
                cn
             i-i  n
             so
             "  S
             cn  O
   a. (-•
   O  cn
   cn
   a>  o

      m
      M
                           (D
                                     O
                                     CT
             §   ^   C  2
             •o   p   -a   r~
              »   m   5   m
              CT
              O
              C

              a

              A
              (0

              i
              0)
                       CT
                       O
                       C
                       3
                       a.

                       (i>
                       in
                       ^

                       3
                       Q>

                       (C
                                               Q)
                                               (Q
                                               (I)
                                               cp
                                               rr
                                           a
                                            i
                                 a
                                  i
                                  I
                                  l
                                 a
                                  i
                                  i
                                  i
                                 D
                                 C
                                 •o
                                 •a
                                      (—
                                      m
    0)
    x
-   I
0)   5r
                                               O
                                               o
                                               Q.

                                               0)
                                               cn


                                               §
                                               cu
                                                                                                                  O
                                                                                                                            01
                                                                                                                                                                             N)
                                                                                                                                                                             (D
                                                                                                                                                                             cn
                                                                                                                                                                             o

-------
     0.0529
             0.0
             0.6
     Dose in mg/kg/day
1.2
             Multistage
             Probit
             Weibull
             One-hit
— — ^—• — •— — —   M.L.E. (maximum likelihood estimate)
 	A	A	A  Upper-bound estimate
                                                    M.L.E.
                   •+  Upper-bound estimate
                   • -  M.L.E.
                                          •O
                   D   Upper-bound estimate
                       M LE.
                   O   Upper-bound estimate
FIGURE  9_ig Point  and upper-bound estimates of four dose-response models
            over  low-dose region on  the  basis of liver carcinomas in mice;
            dose with surface correction.   (NCI 1978)
                                  9-225

-------
9.5.3.2.4  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 ug/L (1 ppb) of EDC is







      d = 1 ug/L x 2 L/day x 10~3 mg/ug x 1/70 kg = 2.9 x 10~5 mg/kg/day







Therefore, the risk associated with 1 ug/L of EDC in drinking water is







                    P = 6.9 x 10~2 x 2.9 x 10~5 = 2 x I0~b







9.5.3.2.5  Risk associated with 1 ug/m3 of EDC in air.  Two approaches are




used herein to estimate the carcinogenic potency of EDC by inhalation:




(1) direct estimation based on the EDC gavage study, and (2) an indirect




estimation based on EDB studies.




     9.5.3.2.5.1  Direct estimate based on EDC gavage study.  It was estimated in




the previous section that the carcinogenic potency of EDC by gavage is q* = 6.9 x




10~2 mg/kg/day.  If the relative absorption rate of EDC by inhalation and gavage




in animals is known, it is possible to estimate carcinogenic potency by inhalation




if the potency calculated from the gavage study is adjusted proportionately.




As an approximation, it can be assumed that the effective dose  by inhalation  is




only one-third of that by gavage.  This assumption is motivated by  the observation




(Reitz et al. 1982) that DNA alkylation after gavage was 2 to 5 times higher




than after inhalation.  To calculate potency in terms of ug/m3, it  is assumed




that the daily intake of air for a 70-kg person is 20 m3.  Thus, for 1 ug/m3  of




EDC in air, the  corresponding dose in mg/kg/day is







           (1/3) x  (1 ug/m3) x (20 rag/day) x (1/70 kg) x (10~3  mg/ug)




                            = 9.52 x 10~5 mg/kg/day
                                        9-226

-------
On the basis of the above calculations, the carcinogenic potency of EDC by


inhalation is estimated as follows:



             q* = (6.90 x ID"2) x (9.52 x 1CT5) = 6.6 x 1CT6 ug/m3.




     9.5.3.2.5.2  Indirect estimate based on EDB studies.  The carcinogenic poten-


cy of EDC by inhalation can be estimated by utilizing the following assumption:



                             P(EDC, I) = P(EDC, G)
                             P(EDB, I)   P(EDB, G)



where P(EDC, I) and P(EDC, G) indicate the potencies of EDC by inhalation


and by gavage, respectively, and P(EDB, I) and P(EDB, G) indicate the potency


of EDC by inhalation and by gavage, respectively.


     The potencies for EDC and EDB by gavage are estimated from the hemangio-


sarcomas in male rats in the NCI studies on EDC (NCI 1978) and EDB (NCI 1979).


On the basis of gavage studies, it is estimated that EDB is approximately 10


times more potent than EDC.  The estimation and the data used for calculating


the EDB potency by gavage are presented in Appendix C.


     The potency of EDB by inhalation is calculated as P(EDB, I) = 6.77 x 10~5


ug/m3, on the basis of nasal cavity tumors in male rats (NCI 1979).  The cal-


culation and data are presented in Appendix C.  On this basis, the potency of


EDC by inhalation is calculated as follows:



                        P(EDC.I) = P(EDC, G> x P(EDB, I)
                                   P(EDB, G)



                                 = _1 x 6.77 x 10~5
                                   10



                                 = 6.8 x 10-6 Ug/m3
                                   9-227

-------
Practically no difference exists between this estimate and that calculated




directly from the EDC gavage study data.







9.5.3.3  Comparison of Potency with Other Compounds—One of the uses of




quantitative potency estimates is to compare the relative potencies of carcino-




gens.  Figure 9-14is a histogram representing the frequency distribution of




potency indices of 53 suspect carcinogens evaluated by the CAG.  The data




summarized by the histogram are presented in Table 9-59 •  The potency index is




derived from q*, the 95% upper bound of the linear component in the multistage




model, and is expressed in term of (mMol/kg/day)~~ .  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 7 x 10™.  This figure




is derived by multiplying the slope q* = 6.9 x 10~2 mg/kg/day and the




molecular weight of EDC, 98.9.  This places the potency index for EDC in the




fourth quartile of the 53 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 linear extrapolation from




the observational range.  These indices may not be appropriate for the compari-




son of potencies if linearity does not exist at the low-dose range, or if
                                     9-228

-------
zu
18
16
14
& 12
uu
LU
oc
"" 8
6
4
2
0
*
4th

3rd 2nd 1 st
QUARTILE | QUARTILE QUARTILE QUARTILE ~

—


—

~
—
—
ill


II
•x'-i-i'i'i'
ivivivi
**•*******•*'
vX'Xv
XvXv'
•XvXv




2 0
I




X;";Xvi
XvXv'
2
0




>1 4xiQ+2 2xio+3
XvX'X'
•X'XvX
"vX'X*''
XvX'X'
:•:$$:$:
8$ffK:
l-i-ix'Sliii
x-x-jxij
•:•:•:•:•:•:•:•
—
—
—
~"
—
_

$ 6 $
^vivX-iv
n^u g^ -
468
                           LOG OF POTENCY INDEX
Figure 9-16  Histogram representing the frequency distribution  of  the
            potency indices of  53 suspect carcinogens  evaluated by the
            Carcinogen Assessment Group.
                               9-229

-------
TABLE  9-59.  RELATIVE CARCINOGENIC POTENCIES AMONG 53 CHEMICALS EVALUATED BY
    THE CARCINOGEN ASSESSMENT GROUP AS SUSPECT HUMAN CARCINOGENS1'2'3
Slope Molecular
Compound (mg/kg/day)"1 weight
Acrylonitr ile
Af la toxin B,
Aldrin
Allyl chloride
Arsenic
B[a]P
Benzene
Benzidene
Beryllium
Cadmium
Carbon tetrachloride
Chlordane
Chlorinated ethanes
1 , 2-dichloroethane
hexachloroe thane
1,1,2, 2-tetrachloroethane
1,1, 1-trichloroethane
1,1, 2-trichloroethane
Chloroform
Chromium
DDT
Dichlorobenzidine
1 , 1-dichloroethylene
Dieldrin
0.24(W)
2924
11.4
1.19xlO-2
15(H)
11.5
5.2xlCT2(W)
234 (W)
1.40
6.65(W)
1. SOxlO'1
1.61
6.9xlO-2
1.42x10-2
0.20
1.6xlO-3
5.73x10-2
7x10-2
41(W)
8.42
1.69
1.47xlQ-1(I)
30.4
53.1
312.3
369.4
76.5
149.8
252.3
78
184.2
9
112.4
153.8
409.8
98.9
236.7
167.9
133.4
133.4
119.4
100
354.5
253.1
97
380.9
Potency
index
1x10+1
9xlO+5
4xlO+3
9x10-1
2xlO+3
3xlO+3
4x10°
4xlO+A
1x10+1
7x10+2
2x10+1
7x10+2
7x1 00
3x10°
3x10+1
2x10-1
8x10°
8x1 00
4xlO+3
3xlO+3
4xlO+2
1x10+1
lxlO+4
Order of
magnitude
(logio
index)
+ 1
+6
+4
0
+3
-t-3
+1
+5
+1
+3
+1
+3
+1
0
+ 1
-1
+1
+ 1
+4
+3
+3
+1
+4
                                                (continued  on the  following page)
                                  9-230

-------
TABLE 9-59 (cont.)
Compound
Dinitrotoluene
Diphenylhydrazine
Epichlorohydrin
Bis(2-chloroethyl)ether
Bis (chloromethyl) ether
Ethylene dibromide (EDB)
Ethylene Oxide
Heptachlor
Hexachlorobenzene
Hexachlorobutadiene
Hexachlorocyclohexane
technical grade
alpha isomer
beta isomer
gamma isomer
Methylene chloride
Nickel
Nitrosamines
Dimethylnitrosamine
Diethylnitrosamine
Dibutylnitrosamine
N-nitrosopyrrolidine
N-nitroso-N-ethylurea
N-nitroso-N-methylurea
N-nitroso-diphenylamine
PCBs
Slope
(mg/kg/day)"
0.31
0.77
9.9xlO"3
1.14
9300(1)
8.51
0.63(1)
3.37
1 .67
7.75xiO~2
4.75
11 .12
1.84
1.33
6.3X10"11
1 J5(W)
25. 9 (not by q.,«)
43.5(not by q^)
5.43
2.13
32.9
302.6
4.92x10~3
..3*
Molecular
weight
182
180
92.5
143
115
187.9
44.0
373.3
284.4
261
290.9
290.9
290.9
290.9
84.9
58.7
74.1
102.1
158.2
100.2
117.1
103.1
198
324
Potency
index
6x1 0+1
1 xi 0+2
9x1 O"1
2x1 0+2
1 x1 0+6
2x1 0+3
3x1 O"1"1
1x1 0+3
5x1 O*2
2x1 0+
3x1 o*3
5x1 0+J;
4x1 O"1"2
5x1 0~2
7x1 0+
2x1 0+3
4x1 O"1"3
9x1 O^2
2x1 0*^
4x1 0+^
3x1 0*
1x10
1 x1 O"1"3
Order of
magnitude
(log
index;
+2
+2
0
+2
+6
+3
+1
+3
+3
+1
+3
+3
+3
+3
-1
+2
+3
+4
+3
+2
+4
+4
0
+3
  9-231

-------
                        TABLE 9-59 .   (continued)
Order of
Compound
Phenols
2,4 ,6-trichlorophenol
Tetrachlorodioxin
Te trachloroethylene
Toxaphene
Trichloroethylene
Vinyl chloride
Remarks:
1 . Animal slopes are
Slope
(mg/kg/day) L
1.99xlO-2
4.25xl05
3.5xlO-2
1.13
1.9xlO"2
1.75x10-2(1)

95% upper-limit sl<
magnitude
Molecular Potency (logio
weight index index)
197.4 4x10°
322 lxlO+8
165.8 6x10°
414 5x10+2
131.4 2.5x10°
62.5 1x10°

Dpes based on the linearized
+ 1
+8
+ 1
+3
0
0

mult istage
    model.  They are calculated based on animal oral studies, except for those
    indicated by 1 (animal inhalation), W (human occupational exposure), and H
    (human drinking water exposure).  Human slopes are point estimates based on
    the linear non-threshold model.

2.  The potency index is a rounded-off slope in (mMol/kg/day)~^ and is calcu-
    lated by multiplying the slopes in (mg/kg/day)-1 by the molecular weight
    of the compound.

3.  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.
                                9-232

-------
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.^  Summary



9-5.U.1  Qualitative—Although several 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,




hemangiosarcomas of the circulatory system, and fibromas of subcutaneous




tissue in male rats.  There was also a statistically significant increased




incidence of adenocarcinomas of the mammary gland and hemangiosarcomas of the




circulatory system in female rats.




     In the NCI (1978) studies in mice, 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 postive 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 1 51




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 organ sites as compared  to control animals.




     The study by Theiss et al.  (1977),  a pulmonary bioassay in which EDC was




administered intraperitoneally to strain A mice,  produced a statistically




insignificant increase in the incidence of lung tumors in treated  animals.
                                     9-233

-------
     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 tumorigen.   This




study did not show a statistically significant increase in skin carcinomas;




a significantly 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 were available




in the published literature.






9.5.4.2  Quantitative—Data from studies using both rats and mice were used




to estimate the carcinogenic potency of EDC.   For rats, data on hemangiosar-




comas in the circulatory system were used.  For mice, data on hepatocellular




carcinomas were used.  The carcinogenic potencies 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 is q* = 7 x 10~2 mg/kg/day,




calculated on the basis of hemangiosarcomas using the time-to-death data.




The upper-bound estimate of the cancer risk from 1 ug/nH of EDC in air is




7 x 10~6.  The upper-bound estimate of the risk from 1 ug/L of EDC in




drinking water is 2 x 10~6.  The cancer risk of EDC by inhalation is calcu-




lated by two methods:  1) a direct estimation based on the EDC gavage study,




assuming that the absorption rate by inhalation is one-third of that by the




oral route; and 2) an indirect estimation from the EDB inhalation study.   The




potencies calculated from both approaches are practically identical.




     The potency index for EDC, defined as q* x molecular weight, lies in  the




fourth quartile among the 53 suspect carcinogens evaluated by the Carcinogen




Assessment Group.






9.5.5  Conclusions




     There is evidence that EDC is a potential human carcinogen.  This conclusion
                                  9-234

-------
is based on:   1) positive findings in one oral rat study and one oral mouse




study and supportive evidence in two other mouse studies;  2) a positive mutage-




nicity response in multiple tests; and 3) demonstrated evidence of the presence




of reactive metabolites and covalent binding to DNA.




     The estimated carcinogenic potency of EDC places it in the 4th quartile of




the 53 suspect carcinogens evaluated by the CAG.




     Applying the International Agency for Research on Cancer (1ARC) classifica-




tion scheme,  this level of evidence in animals would  be considered sufficient




for concluding that EDC is a potential human carcinogen with a rank of 2B.
                                 9-235

-------
               United States
               Environmental Protection
               Agency
              Office of Health and
              Environmental Assessment
              Washington DC 20460
EPA-600/8-84-006A
April 1984
External Review Draft
               Research and Development
x>EPA
Health Assessment    Review
Document for            Draft
1 ,2-Dichloroethane
._ .   .       I-N-  i i   -i»
(Ethylene Dichloride)

Part  2 of 2
                                              Cite or Quote)
                            NOTICE
              This document is a preliminary draft. It has not been formally
              released by EPA and should not at this stage be construed to
              represent Agency policy. It is being circulated for comment on its
              technical accuracy and policy implications.

-------
(DO Not                                   EPA-600/8-?4:°°6*
Cite or Quote)                                c      , D APnl ^98*
                                            External Review Draft
        Health Assessment Document
           for  1,2,- Dichloroethane
               (Ethylene Dichloride)
                       Part 2 of 2
                            NOTICE
 This document is a preliminary draft. It has not been formally released by the U.S. Environmental
 Protection Agency and should not at this stage be construed to represent Agency policy. It
 is being circulated for comment on its technical accuracy and policy implications.
                                       U.S. Environmental Protection Agency
                                       Region V, Library
                                       230 South DearL'C-Ti C'.rest
                                       Chicago, Illinois  60604
               U.S. ENVIRONMENTAL PROTECTION AGENCY
                  Office of Research and Development
               Office of Health and Environmental Assessment
               Environmental Criteria and Assessment Office
               Research Triangle Park, North Carolina 27711

-------
                                         DISCLAIMER




           This  report is an  external draft for  review purposes only  and does  not




      constitute  Agency Policy.  Mention  of trade names or  commercial  products  does




      not constitute endorsement  or  recommendation for  use.
U.S. Environmental Protection Agency
                                              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  are placed  in perspective  with observed




environmental levels.
                                      111

-------
     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.  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-U080

             D. Anthony Gray, Ph.D.
             Life and Environmental Sciences Division
             Syracuse Research Corporation
             Syracuse, New York  13210-H080

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

     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 (»).
                                       iv

-------
             Roy Albert, M.D. (Chairman)
             Elizabeth L. Anderson, Ph.D.
             Larry D. Anderson, Ph.D.
             Steven Bayard, Ph.D.
             David L. Bayliss, M.S.
             Chao W. Chen, Ph.D.*
             Margaret M.L. Chu, Ph.D.
             Herman J. Gibb, M.S., M.P.H.
             Bernard H. Haberman, D.V.M., M.S.
             Charalingayya B. Hiremath, Ph.D.*
             Robert McGaughy, Ph.D.
             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.


             John R. Fowle III, Ph.D.*
             Ernest R. Jackson, M.S.
             Casey Jason, M.D., Medical Officer
             David Jacobson-Kram, Ph.D.
             K.S. Lavappa, Ph.D.
             Sheila L. Rosenthal, Ph.D.
             Carol N. Sakai, Ph.D.*
             Carmella Tellone, B.S.
             Vicki L. Vaughan-Dellarco, Ph.D.
             Peter E. Voytek, Ph.D. (Director)

     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
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
                                      VI

-------
Larry Fishbein, Ph.D.
National Center for lexicological 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

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

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

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

Jean C. Parker, Ph.D.
Office of Waste Programs Enforcement
U.S. Environmental Protection Agency
Washington, DC
                         vn

-------
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

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

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

Robert Tardiff, Ph.D.
1423 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

Jim Withey, Ph.D.
Department of National Health and Welfare
Tunney's Pasture
Ottawa, Ontario
CANADA K1A 01Z
                         viii

-------
                                TABLE OF CONTENTS


                                                                            Page

. LIST  OF  TABLES	       xi

 LIST  OF  FIGURES	      xvi

 1.    SUMMARY  AND  CONCLUSIONS	     1-1

 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
      3.1   STRUCTURE	     3-1
      3.5   PHYSICAL PROPERTIES OF  PURE ETHYLENE  DICHLORIDE	     3-1

 1 .    SAMPLING AND ANALYSIS  OF ETHYLENE DICHLORIDE	     1-1

      1 . 1   SAMPLING	     1-1
      4.2   ANALYSIS	     1-1

           1.2.1    Ethylene  Dichloride in Air	     1-1
           1.2.2    Ethylene  Dichloride in Water	     1-2
           1.2.3    Ethylene  Dichloride in Solid  Samples	     1-3
           1.2.1    Ethylene  Dichloride in Blood	     1-3
           1.2.5    Ethylene  Dichloride in Urine	     1-3
           1.2.6    Ethylene  Dichloride in Tissue	     1-1

 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
      5.1   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	   5-11

 6.    FATE  AND TRANSPORT IN THE ENVIRONMENT	     6-1

      6.1   ATMOSPHERE	     6-1
      6.2   AQUATIC MEDIA	     6-7
      6.3   SOIL	     6-8
      6.1   SUMMARY	     6-9
                                       IX

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

                                                                           Page

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	   9-85
        9.2.3    Summary of Acute, Subchronic  and Chronic  Toxicity	   9-115

     9.3  REPRODUCTIVE  AND TERATOGENIC EFFECTS	   9-122

        9.3.1    Summary	   9-129

     9.4  MUTAGENICITY	   9-131
     9.5  CARCINOGENICITY	   9-166

        9.5.1    Animal  Studies	   9-166
        9.5.2    Epidemiologic Studies	   9-205
        9.5.3    Quantitative Estimates	   9-206
        9.5.4    Summary	   9-233
        9.5.5    Conclusions	   9-234

10.   REFERENCES  	    10-1

 Appendix  A
 Appendix  B
 Appendix  C

-------
                                 LIST OF TABLES


Table                                                                      Page

5-1     Major Manufacturers of Ethylene Dichloride	     5-3

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

5-3     Ethylene Dichloride Uses	     5-6

5-4     Comsumption of Ethylene Dichloride in 1979 and 1974	     5-7

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

5-6     Estimated Environmental Releases of Ethylene
          Dichloride in 1979	    5-11

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
          System	    7-15

7-4     Reported Occurrence of Ethylene Dichloride in Surface
          Water Systems	    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	    7-21

7-8     Total Estimated Population (in Thousands) Exposed to
          Ethylene Dichloride in Drinking Water at the Indicated
          Concentration Ranges	    7-22

7-9     Estimated Drinking Water Intake of Ethylene Dichloride
          by Adults and Infants	    7-23

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

9-2     Partition Coefficients for Ethylene Dichloride	     9-4
                                      xi

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

Table                                                                      Page

9-3     Fate of li4C-EDC in Rats 18 Hours After Oral (150 mg/kg)
          or Inhalation (150 ppm,  6~hr) Exposure ........................    9-6

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 Orll with Doses of EDC in Oil
          and in Water as Vehicle and After Intravenous
          Administration ............................. . ..................    9-9

9-6     EDC Tissue Levels After 50 ppm Inhalatory Exposure ........... ...   9-11

9-7     EDC Tissue Levels After 250 ppm Inhalatory 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 (18-hr) by
          Mice Receiving 1 ,2-Dichloroethane-   C ......................     9-29

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 (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
          2-Compartment Model ......................................... . .   9-30

9-H    Identified Metabolites of Ethylene Dichloride and
          Ethylene Bromide ..............................................   9-31

9-15    Percent Distribution of Radioactivity Excreted (18-hr) as
          Urinary Metabolites by Mice Receiving  1 ,2-Dichloroethane-  C

-------
                            LIST OF TABLES  (oont.)

Table                                                                      Page

9-16    Total Macromolecular Binding and DMA Binding in Selected
          Tissue of Rats After Exposure to   C-EDC by Oral or
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
iii
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 mg/mg [U- C]EDB 	
Effects Associated with Acute Lethal Oral Doses of
Ethylene Dichloride in Humans 	
Effects of Acute Oral Ingestion of 1 ,2-Dichloroethane
(Survey Results) 	
Effect of Ethylene Dichloride Exposure on Eye
Sensitivity to Light 	
Morbidity and Lost Workdays of Aircraft Industry Gluers
Exposed to Ethylene Dichloride 	
Concentrations of Ethylene Dichloride in Oil Refinery
Mineral Oil Purification Process Air 	
Effects Observed in Polish Oil Refinery Workers 	
Effects of Acute Exposure to Ethylene Dichloride 	
Effect of Ethylene Dichloride on the Cornea 	
Effect of Subchronic Exposure to Ethylene Dichloride 	 ,
Summary of Mutagenicity Testing of EDC: Gene Mutations
in Bacteria 	
Summary of Mutagenicity Testing of EDC: Higher Plants 	 ,
Summary of Mutagenicity Testing of EDC: Gene Mutation

Summary of Mutagenicity Testing of EDC: Mammalian Cells
in Culture 	
Summary of Mutagenicity Testing of EDC: Chromosomal
Aberrations Tests 	 ,
Summary of Mutagenicity Testing of EDC: PolA Assay 	 ,
Summary of Mutagenicity Testing of EDC: DNA Binding


9-54

9-58

9-65

9-72

9-78

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

9-132
9-145

9-147

9-153

9-155
9-160

          Studies.	   9-163
                                      xiii

-------
                                LIST OF TABLES

Table

9-3*4    Design Summary for 1,2-Dichloroethane (EDC) Gavage
          Experiment in Osborne-Mendel Rats	   9-168

9-35    Terminal Survival of Osborne-Mendel Rats Treated With
          1,2-Dichloroethane (EDC)	   9-172

9-36    Squamous Cell Carcinomas of the Forestomach in Osborne-
          Mendel Rats Treated With 1,2-Dichloroethane (EDC)	   9-173

9-37    Hemangiosarcomas in Osborne-Mendel Rats Treated With
          1,2-Dichloroethane (EDC)	   9-171

9-38    Adenocarcinomas of the Mammary Gland in Female Osborne-
          Mendel Rats Treated With 1,2-Dichloroethane (EDC)	   9-175

9-39    Design Summary for 1,2-Dichloroethane (EDC) Gavage
          Experiment in B6C3F1 Mice	   9-177

9-10    Terminal Survival of B6C3F1 Mice Treated With
          1,2-Dichloroethane (EDC)	   9-178

9-11    Hepatocellular Carcinomas in B6C3F1 Mice Treated With
          1,2-Dichloroethane (EDC)	   9-181

9-12    Alveolar/Bronchiolar Adenomas in B6C3F1 Mice Treated
          With 1,2-Dichloroethane (EDC)	   9-182

9-13    Squamous Cell Carcinomas of the Forestomach in B6C3F1 Mice
          Treated With 1 ,2-Dichloroethane  (EDC)	   9-183

9-11    Adenocarcinomas of the Mammary Gland in Female B6C3F1 Mice
          Treated With 1 ,2-Dichloroethane  (EDC)	   9-181

9-15    Endometrial Polyp or Endometrial Stromal Sarcomas  in Female
          B6C3F1 Mice Treated With 1,2-Dichloroethane (EDC)	   9-181

9-16    Characterization of 1,2-Dichloroethane  (EDC) Inhalation
          Experiment in Sprague-Dawley Rats.	   9-185

9-17    Design Summary for  1,2-Dichloroethane (EDC) Experiment
          in Sprague-Dawley Rats	   9-186

9-18    Survival of Sprague-Dawley Rats Exposed to EDC at  52 and
           101 Weeks	   9-188

9-19    Tumor Incidence  in Sprague-Dawley  Rats  Exposed to  EDC	   9-189

9-50    Mammary Tumors in Sprague-Dawley Rats Exposed to EDC	   9-193
                                       xiv

-------
                                LIST  OF  TABLES

Table

9-51    Design Summary for 1,2-Dichloroethane (EDC) Experiment
          in Swiss Mice	    9-196

9-52    Survival of Swiss Mice Exposed to EDC at 52 and 78 Weeks	    9-197

9-53    Tumor Incidence in Swiss Mice Exposed to EDC	    9-198

9-54    Pulmonary Tumor Response in Strain A/st Mice
          Injected with EDC	    9-202

9-55    Mouse Skin Bioassay of 1,2-Dichloroethane and
          Chloroacetaldehyde	    9-204

9-56    Incidence Rates of Hemangiosarcomas in the Circulatory
          Systems of Male Osborne-Mendel Rats	    9-220

9-57    Incidence Rates of Hepatocellular Carcinomas in Male
          B6C3F1 Mice	    9-220

9-58    Upper-Bound Estimate of Risk At 1 Mg/kg/day	    9-223

9-59    Relative Carcinogenic Potencies Among 53 Chemicals
          Evaluated by the Carcinogen Assessment Group as Suspect
          Human Carcinogens ' '3	      9-230
                                       xv

-------
                                LIST OF FIGURES


Figure                                                                     Page

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

9-4     Levels of EDC in rats after inhalatory exposure to 50 ppm
          (top) and 250 ppm (bottom).  Levels were measured at
          termination of 5-hour exposure period .........................   9-27

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

9-6     Further metabolism of 2-chloroacetaldehyde and 1-chloroso-
          2-chloroethane from microsomal oxidation ......................   9-4 1
9-7     Cytosolic metabolism of ethylene dichloride .....................   9-44

9-8     Growth curves for male and female Osborne-Mendel rats
          administered 1 ,2-dichloroethane (EDC) by gavage ...............  9-170

9-9     Survival comparisons for male and female Osborne-Mendel rats
          administered 1 ,2-dichloroethane (EDC) by gavage ...............  9-171

9-10    Growth curves for male and female B6C3F1 mice administered
          1,2-dichloroethane (EDC) by gavage ............................  9-179

9-11    Survival comparisons for male and female B6C3F1 mice
          administered 1 ,2-Dichloroethane (EDC) by gavage ...............  9-180

9-12    Point and upper-bound estimates of four dose-response models
          over low-dose region on basis of hemangiosarcomas in rats;
          dose with surface correction ..................................  9-224

9-13    Point and upper-bound estimates of four dose-response models
          over low-dose region on the basis of liver carcinomas in
          mice; dose with surface correction ............................  9-225

9-T4    Histogram representing the frequency distribution of the
          potency indices of 53 suspect carcinogens evaluated by
          the Carcinogen Assessment Group ...............................  9-229
                                      xvi

-------
                                10.   REFERENCES








Agranovich, B.Y.A.   19*18.   Clinical Treatment and  Pathology of  Toxicologic-




chemical Injuries of the  Liver  in the Case  of Industrial Poisoning.   Moscow,




USSR, Academy of Medical Science,   p. 132-U3.   (Summarized in NIOSH,  1976).








Akimo, G.A., V.M. Buchko and I.P.  lolesnichenko.   1976.   Changes  in the nervous




system  in  acute dichloroethane  poisoning.   Voenno-meditsinskiy Zhurnal.    5:




35-37.








Akimo, G.A., V.M. Buchko and I.P.  Kolesnichenko.   1978.   Neurological disorgers




in acute dichloroethane poisoning.  Zh.  Nevropatol.  Piskhiatr.  78(5):  687-692.








Altshuller, A.P.  1979.   Lifetimes  of organic  molecules  in the troposphere and




lower stratosphere.   Advances in Environmental Science and Technology.  10:  181-




219.








Altshuller, A.P. and  P.L.  Hanst.  1975.  Report on  the Problem of Halogenated Air




Pollutants and  Stratospheric Ozone.   EPA-600/9-75-008,  Office of  Research and




Development, U.S. Environmental  Protection Agency, Washington, DC








Ambrose, A.  1950.   Toxicological studies of compounds investigated  for use  as




inhibitors  of biological processes.   II.  Toxicity  of  ethylene chlorohydrin.




Arch. Inc. Hyg. Occup.  Med.   2:  591-597.
                                     10-1

-------
Amoore, J.E.  and D. Venstrom.  1966.  Sensory analysis of odor qualities in term




of the stereochemical theory.   J.  Food. Science.   31: 118.








Anders, M.W.  and J.C.  Livesey.   1980.   Metabolism of 1,2-dihaloethanes,  in




"Banbury Report No.  5, Ethylene Bichloride:  A Potential Health Risk?", B. Ames,




P. Infante and R. Reitz, eds., Cold Spring Harbor Laboratory, p.  331-3^3.








Andriukin, A.A.   1979.   Toxic effects of dichloroethane  on  the  cardiovascular




system.  Klin.  Med.   57:  U3-47 (Rus.)   (Summarized in Chem. Abstr.   79: 2088,




197).








Anonymous.  19**6.  Ethylene Bichloride, in Annual Report of the Chief Inspector




of Factories for the Year 19^5.  London, His Majesty's Stationery Office,  p. 77.




(Summarized in NIOSH, 1976).








Appleby, A.  1976. Atmospheric Freons  and Halogenated Compounds.  EPA-600/3-76-




108, Office of Research and Development,  U.S. Environmental Protection Agency.








Archer,  W.L.    1979.    1,2-Dichloroethane.   In;   Kirk-Othmer Encyclopedia of




Chemical Technology,  Third  Edition.,  Grayson,  M.  and  Eckroth, D.,  eds.   John




Wiley and Sons, Inc., NYJl  Vol. 5, p.  72*1-727.








Archer,  W.L.   1979.    Other Chloroethanes.   In:   Kirk-Othmer Encyclopedia of




Chemical Technology,  Third Edition,  Grayson, M. and Eckroth, D., eds.   John Wiley




and Sons, Inc., NY. Vol. 5, p. 722-7*»2.
                                      10-2

-------
Armitage,  P. and R. Doll.  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  Bichloride.   Unpublished report  prepared  for the  U.S.  Environmental




Protection Agency under contract number EPA-68-01-3899  and Auerbach Associates,




Inc., number AAI-2431 -1OU-TN-1 ,  Philadelphia,  PA, 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.  (Ger.) (Summarized




in NIOSH,  1976).








Banerjee,  S.  and B.L.  Van Duuren.  1978.   Interaction of activated carcinogenic




intermediates of ethylene dihalides with protein and DNA in mice and rat tissue




in vitro.   Proc.  Amer. Assoc.  Cancer Res.   19: 67.








Banerjee,  S.  and B.L. Van  Duuren.   I979a.   Binding  of carcinogenic halogenate




hydrocarbons to cellular raacromolecules.   J. Natl. Cancer Inst.  63: 707.








Banerjee,  S.,  B.L. Van Duuren and S.A. Kline.   1979b.  Interaction of potential




metabolites of the carcinogen ethylene dibromide with protein and DNA in vitro.




Biochera. Biophys. Res. Comm.  90: 1214-1220.








Banerjee,   S.,  B.L.  Van Duuren  and  F.I.  Oruambo.   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. and H.S.  Telford.   1966.  Effect of sublethal doses  of  fumigants  on




stored-grain insects.  Technical Bulletin  50.  Washington  Agricultural  Experi-




ment Station, Washington State University.








Barber, E.D., W.J. Donish and K.R.  Mueller.  1981.  A procedure for the  quanti-




tative  measurement   of  the   mutagenicity  of  volatile  liquids  in  the  Ames




Salmonella/microsome assay.  Mutat. Res.   90:  31-48








Barrows, M.E., S.R.  Petrocelli, K.J. Macek and J.J.  Carrell.  1980.  Bioconcen-




tration  and  elimination  of  selected  water  pollutants   by  bluegill  sunfish




(Lepomis macrochirus).   In:  Dynamics,  Exposure,  and Hazard Assessment of Toxic




Chemicals.  R. Haque, ed.  Ann Arbor Science,  Ann Arbor,  MS.  p. 379-392.








Barsoum, G.S. and K.  Saad.  1934.  Relative toxicity of certain chlorine  deriva-




tives of the aliphatic series.  Quart.  J. Pharra.  Pharmacol.  7: 205-214.








Bellamy, R. and W. Schwartz.   1975.  Engineering and Cost Study of Air Pollution




Control  for the  Petrochemical  Industry:   Vinyl Chloride  Manufacture   by  the




Balanced Process. Volume 8.  EPA-450/3-73-006h.  Houdry Division of Air Products




and Chemicals,  Inc.  Report  prepared for U.S.  Environmental Protection  Agency,




Research Triangle Park, NC.








Benoit, D.A.,  F.A.  Puglisi and D.L. Olson.  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): 189-97.
                                      10-4

-------
 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(6): 977-984.








 Berck, B.  1965.   Sorption  of ethylene dibromide, ethylene  dichloride, and carbon




 tetrachloride by cereal products.  J. Agric. Food Chem.  13: 248-254.








 Blackford, J.L.   1974.   Ethylene  Dichloride.  In:  Chemical Economics Handbook.




 Menlo Park, CA:  Stanford Research Institute, p. 651 .5031 a-651 .5032z.








 Bloch,  W.    1946.    Two poisonings  from dichloroethane  used  for  purposes  of




 inebriation.  Schweiz Med. Wochenschr.   76:  1078-1079.   (Ger.)   (Summarized  in



 NIOSH, 1976).








 Bogoyavlenski, V.F., S.K.H. Salikhova and E.V. Karpova.  1968.  Clinical aspects




 and  therapy  for  ethylene dichloride   poisoning.  Sov.  Med.    31:  107-109.




 (Summarized in NIOSH, 1976).








 Bolt, H.M.  1978.  Pharmacokinetics of vinyl chloride.  Gen.  Pharmacol.   9: 91.








 Bonitenko, Y.Y.   1974.   Aspartate-amino-transf erase isoezymes in acute dichloro-




 ethane poisoning.  Gigiena Truda i Professional'nye Zabolevaniga.   7:  46-47.








 Bonitenko, Y.Y.,  A.A. Bruk, Ye. A. Belouson  and M.F.  Lebeden.   1977.   Veomno-




Meditsinskiy Zhurnal.  11:  75-77.
                                     10-5

-------
Borisova, M.K.   1957.  Experimental data for determination of the  maximum  allow-




able concentration of  dichloroethane in the atmosphere.  Gig.  Sanit.  22:  13-19.




(Rus.) (Summarized from English Translation).








Bouwer, E.J. and P.L.  McCarty.  1983.  Transformations of 1- and 2-carbon haloge-




nated  aliphatic  organic  compounds  under  methanogenic  conditions.     Appl.




Environ. Microbiol.  45: 1286-1294.








Bozzelli, J.W.  and B.B.  Kebbekus.   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.,  B.B.  Kebbekus  and A.  Greenburg.   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 Protec-




tion.








Brass, K.   1949.   Concerning a  lethal  dichloroethane poisoning.   Deutsch.  Med.




Wochenschr.  74: 553-554.  (Ger.)  (Summarized in NIOSH, 1976).








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.








Bray, H., W.  Thorpe and D. Vallance. 1952.  The liberation of chloride ions from




organic chlorocompounds by tissue extracts.  Biochem. J.  51 : 193-201.
                                     10-6

-------
Brem,  H.,  A.B.  Stein and  H.S.  Rosenkranz.   1974.   The mutagenicity  and DNA-




modifying effects of haloalkanes.  Cancer Res.  34: 2576-2579.








Brodzinsky, R. and H.B. Singh.  1982.  Volatile Organic Chemicals in the Atmos-




phere:  Assessment of Available Data.  Prepared for the U.S. EPA under contract




No. 68-02-3452 by Stanford Research Institute International, Menlo Park, CA.








Brown, R.H. and Purnell, C.J.   1979.   Collection and Analysis of Trace Organic




Vapor Pollutants in Ambient Atmospheres.  The performance of a Tenax GC adsorbent




Tube.  J. Chromat.  1 78 : 79-90.








Bryzhin, F.F.  1945.  Pathomorphological changes of internal organs in connection




with poisoning by ethylene  dichloride through the digestive  tract.   Farmakol.




Toksikol.  8(5):  43-49. (Rus.)  (Summarized in NIOSH,  1976).








Brzozowski, J.,  J. Czajka, T. Dutkiewicz,  I.  Kesy and  J.  Wojcik.  1954.   Work




hygiene and the health condition of workers  occupied  in combating the Leptino-




tarsa decemlineata with HCH and dichloroethane.  Med.  Pracy.   5:  89-98.  (Pol.)



(Summarized in NIOSH, 1976).








Buccafusco, R.J.,  S.J. Ellis and G.A. LeBlanc.  1981.  Acute toxicity of priority




pollutants to bluegill (Lepomis macrochirus).  Bull. Environ. Contam.  Toxicol.



26(4):  446-52.
                                     10-7

-------
Burns, L.A., D.M. Cline and R.R.  Lassiter.   1982.  Exposure Analysis  Modeling




System  (EXAMS):   User Manual  and System Documentation.   U.S.  EPA Report  No.




EPA-600/3-82-023, Environmental  Research Laboratory,  Office of  Research  and




Development, U.S. Environmental Protection Agency, Athens,  GA.   443 p.








Butler, R., I.J.  Solomon and A. Snelson.  1978.  Rate constants  for the  reaction




of OH with halocarbons in  the presence of 0_ + N?.  J. Air Pollut. Control Assoc.




28(11): 1131-1133.








Byers, D.H.  1943.  Chlorinated solvents in common wartime use.   Ind. Med.  12:




440.








Catalytic.  1979.  Draft Summary Report of BAT-308 Responses.








Callahan, M.A., M.W. Slimak, N.W. Gabel,  I.P.  May,  C.F.  Folwer,  J.R. Freed, et




al.   1979.   Water-Related Environmental Fate of 129 Priority Pollutants.  Vol.




II.    U.S.  EPA  Report No.  EPA-440/4-79-029b,  Office  of  Water   Planning  and




Standards,  Office of  Water and Waste Management, U.S.  Environmental Protection




Agency, Washington, DC.








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 dichloro-




ethane.   Folia.  Med.  Cracov.   1: 169-192.   (Pol.)   (Summarized  from English




Translaton).
                                      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




Bichloride.   Chemical  Marketing Reporter, June  10,  1974.








CMR  (Chemical   Marketing  Reporter).    1977.    Chemical  Profile  -  Ethylene




Bichloride.   Chemical  Marketing Reporter, May 16, 1977.








CMR  (Chemical   Marketing  Reporter).    1980.    Chemical  Profile  -  Ethylene




Bichloride.   Chemical  Marketing Reporter, May 5,  1980.








CMR  (Chemical  Marketing  Reporter).   1982.    Chemical  Profile  -  Lead Alkyls.




Chemical Marketing Reporter,  February  1,  1982.
                                     10-9

-------
CMR  (Chemical  Marketing  Reporter).    1983.    Chemical  Profile  -  Ethylene




Dichloride.  Chemical Marketing Reporter,  June 13,  1983.








Chiou, C.T.,  L.J. Peters and V.H. Freed.  1979.  A physical concept of soil-water




equilibria for nonionic organic compounds.  Science.   206:  831-832.








Chiou, C.T.,  V.H. Freed, L.J. Peters and  R.L.  Kohnert.   1980.   Evaporation  of




solutes from water.   Environ. International.  231-236.








Coleman,  W.E., R.D.  Lingg,  R.G.  Melton  and F.C. Koppler.   1976.   The occurrence




of volatile organics in five drinking water supplies using gas  chromatography-




mass spectrometry.   Identif. Amal.  Org.  Pollut. Water.  (Chem.   Congs.   North




Amer.  Cont.) 1st 1975, Ann Arbor Sci.,  Ann Arbor,  MI.   p.  305-327.








The  Condensed  Chemical Dictionary.   1977.   Nine edition.   G.G. Hawley,  ed.,




Van Nostrand Reinhold Company,  NY.








Conkle, J.P.,  B.J.  Camp and B.E. Welch.   1975.  Trace composition of  human



respiratory gas.  Arch. Environ. Health.   30(6):  290-295.








Cooper, C.V.,  L.D. White and R.E. Kupel.  1971.  Qualitative detection limits for




specific compounds utilizing gas chromatographic fractions, activated charcoal,




and a mass spectrometer.








Cox, C.B.   1972.  Regression model and  life tables.   J.  Roy.  Stat. Soc.  B.  3*<:




187-220.
                                     10-10

-------
Crump, K.S.  and W.W. Waston.  1979.  GLOBAL79.  A Fortran program to extrapolate




dichotomous animal carcinogenicity data to low dose.  Natl. Inst. Environ. Health




Science Contract  No.  1-ES-2123-







Crump, K.S., H.A. Guess and L.L. Deal.  1977.  Confidence intervals and test of




hypotheses  concerning  dose-response  relations  inferred  from animal  carcino-




genicity data.  Biometric.  33:  437-451 •








Crutzen,  P.J.  and J.  Fishman.    1977.   Average  concentrations  of  *OH  in  the




troposphere and the budgets of CH^, CO, H2, and CH-CCl^.  Geophy.  Res. Lett.   4:




321-324.








Cupitt, L.T.   1980.  Fate  of Toxic  and Hazardous Materials in the Air Environ-




ment.   U.S.  EPA  Report No. EPA-600/S3-80-084, Environmental  Sciences  Research




Laboratory, U.S.  Environmental  Protection Agency, Research Triangle Park,  NC.




Available through NTIS Order No. PB30-221948, Springfield, VA.








Daffer, P., K. Crump and M. Masterman.   1980.   Asymptotic theory for analyzing




dose-response survival data with application to low-dose extrapolation problem.




Mathematical Biosciences.   50:  207-230.








Danz, M.  and  H.  Urban.   1979.   Carcinogens, promoters,  and  suspicious drugs:




Their identical  short-term effect in a  promoting activity  test (PAT).   Exp.




Pathol.  17: 181-18U.
                                     10-11

-------
Deinzer,  M, F. Schaumburg and E.  Klein.   1978.  Environmental Health  Sciences




Center.  Task Force Review of Halogenated Organics  in Drinking Water.   Environ.




Health Persp.  24: 209-239.








Delplace, Y. A.  Cavigneaux and G.  Cabasson.  1962.   Occupational disorders due to




methylene chloride and dichloroethane.   Arch.  Mai. Prof.  23: 816-817.   (Fr.)




(Summarized in NIOSH, 1976).








Billing,  W.L., N.B.  Tefertiller and G.J.  Kallos.   1975.   Evaporation rates and




reactivities of methylene chloride, chloroform, 1,1,1-trichloroethane,  trichlo-




roethylene,  tetrachloroethylene,   and  other  chlorinated compounds  in  dilute




aqueous solutions.  Environ. Sci.  Technol.  9(9):  833-838.








Doll,  R.   1971.   Weibull distribution  of cancer:    Implications for  models of




carcinogenesis.  J. Roy. Stat. Soc.  A13: 133-166.








Domenici, F.  1955.  Granosan intoxication.   Rass.  Clin-Sci.   31: 70-73.  (Ita.)




(Summarized in NIOSH, 1976).








Dowty, B.J., D.R.  Carlisle and J.L. Laseter.  1975.  New Orleans drinking water




sources tested by  gas chromatography-mass spectrometry.  Environ. Sci. Technol.




9:  762-765.








Dripps, R.D., J.E. Eckenhoff and L.D. Vandam.  1977.  Introduction to anesthesia:




The principles  of  safe  practice.    5th  edition.    W.B.  Saunders  Company,




Philadelphia, PA.  p. 121-123.
                                     10-12

-------
Drury, J.S. and A.S. Mammons.  1979.   Investigations  of  selected  environmental




pollutants:  1,2-dichloroethane.  Oak  Ridge National  Laboratory/EIS-143.   U.S.




Environmental Protection Agency 560/2-78-006.








Edwards, K., H. Jackson and A.R.  Jones.  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., S.  Osterman-Golkar,  D. Singh  and  U.  Lundquist.  1974.   On the




reaction kinetics and  mutagenic activity of methylating and -halogenating gaso-




line additives.  Radiation Botany.  15: 185-194.








Eimutis, E.G. and R.P. Quill.   1977.  Source Assessment:  Non-criteria Pollutant




Emissions.   EPA-600/2-77-107e.  Monsanto  Research  Corp.,  Dayton, OH.   Report




prepared for the U.S.  Environmental Protection  Agency,  Research Triangle Park,




NC.








Elfers,  L.  1979.  "Monitoring of Ambient Levels of Ethylene Dichloride (EDO in




the Vicinity of EDC Production and User Facilities," prepared for U.S.  Environ-




mental Protection Agency by PEDCO Environmental, Inc., Cincinnati, OH.  NTIS PB-




298303,  EPA 600/4-79-029.








Ellis, C.R. and P.O. Morrison. 1967.   Small chamber tests of ethylene dibromide




and ethylene dichloride on adult grain-infesting cleoptera.  Canad.  J. Zoology.



45:
                                     10-13

-------
Ehrenberg, L., S. Osterman-Golkar,  D.  Singh and U. Lindquist.   1974.   On  the




reaction kinetics and mutagenic activity of methylating and B-halogenethylating



gasoline additives.   Had. Botany.  15:  185-194.








Ethylene Dichloride.  In;  Annual Report  of  the Chief Inspector of Factories for




the Year 1945.  London,  His Majesty's Stationary Office, p. 77.   1946.








Ewing, B.B., E.S.K.  Chian, J.C.  Cook,  C.A.  Evans,  P.K.  Hopke and E.G.  Perkins.




1977.   Monitoring  to Detect  Previously  Unrecognized  Pollutants  in  Surface




Waters.   EPA-560/6-77-015.   U.S. Environmental  Protection Agency.   Office  of




Toxic Substances,  p. 63-64,  73.








Fabricant,  J.D.  and J.G.  Chalmers,  Jr.   1980.    Evidence of mutagenicity  of




ethylene dichloride  and  structurally related compounds.  In:  Banbury Report  5.




Ethylene  Dichloride:   A  potential health  risk?   B.N. Ames,  P.  Infante  and




R. Reitz, eds.   Cold  Spring Harbor Laboratory  Press,  Cold Spring Harbor,  NY.




p. 309-329.








Federal Register.  197L   36(288): 22542.








Federation of American Societies for Experimental Biology  (FASEB).   1974.   2nd




edition.  Vol. III.  Philip L. Altman and Dorothy S.  Dittman, Eds.  Bethesda, MD.




Library of Congress  No.  72-87738.








Finnegan, R.J. and K.E.  Stewart.  1962.  Control  of the pine root collar weevil,




Hylobius radicus.  J. Econ. Entomol.  55(4): 483-486.
                                     10-14

-------
Filser, J.G. and H.M.  Bolt.  1979.  Pharmacokinetics of halogenated ethylenes in




rats.  Arch. Toxicol.  12: 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.   Science of  the  Total




Environment.  11: 223-257.








Flowtow, E.  1952.  Poisoning  due to chlorinated hydrocarbon compounds, particu-




larly  1,2-dichloroethane.   Chem.  Tech. (Berlin)   6:  253-254.   (Summarized  in




NIOSH, 1976).








Freundt, K.J., H. Eberhardt and U.M. Walz.  1963.  Lethal peroral poisoning with




1,2-dichloroethane  and  2,2-dichlorodiethylether.   Int.  Arch.  Gewerbepathol.




Gewerbehyg.  20: 1*1-18.   (Ger.)  (Summarized in NIOSH,  1976).








Fujii, T.   1977.   Direct  aqueous injection  chromatograph-mass spectrometry for



analysis of organohalides in water  at  concentrations below the part per billion




level.  J. Chromatogr.  139: 297-302.








Garrett, J.T.  1957.   Toxicity investigations on aquatic and marine life.  Public




Works.  88(12): 95-96.








Garrett, J.T.   1957.   Toxicity considerations in pollution  control.   Indust.




Wastes.  2: 17-19.
                                     10-15

-------
Garrison, S.C. and R.S. Leadinghara.   1954.  A fatal case of ethylene dichloride




poisoning in  an occupational therapy  department  of a neuropsychiatric hospital.




Amer. J. Phys. Med.  33: 230-237.








Gast, R. and J. Early.  1956.   Phytotoxicity  of  solvents and emulsifiers used in




insecticide formulations.  Agricul.  Chem.  ?: 42-45, 136-139.








Gehring, P.J.,  P.G.  Watanabe and J.D.  Young.  1977-   The relevance  of  sode-




dependent  pharmacokinetics  in  the  assessment   of  carcinogenic   hazard  of




chemicals.  Cold Spring Harbor Conf.  Cell Proliferation  4: 187.








Gehring, P.J., P.G. Watanabe and G.E. Blau.   1976.  Pharmacokinetic studies in




the evaluation of the toxicological  and  environmental hazards of chemicals.  In:




Advances in Modern Toxicology,  M.A.  Mehlman, R.E. Shapiro  and H.   Blumenthal,




eds., vol. 1, part 1, Washington, DC:  Hemisphere Publishing Co.








Gehring, P.J., P.G. Wantabe and  C.N. Park.   1978.   Resolution  of dose response




toxicity  data for chemicals  requiring  metabolic  activation:   Example - vinyl




chloride.  Toxicol. Appl. Pharmacol.  44: 581.








Gikalov, G.S.,  K.A.  Chemanaev and M.U.  Janda.  1969.   Clinic  and treatment of




dichloroethane  poisoning.   Voen-med. Zh.   4:  78-79-   (Rus.)    (Summarized in




NIOSH, 1976).








Gold, L.  1980.  Pages 209-226  in B. Ames, P. Infante and R. Reitz, Eds.  Banbury




Report No. 5.  Ethylene dichloride:  A potential  health  risk? Cold Spring Harbor




Laboratory.
                                     10-16

-------
Grimsrud, E.  and R. Rasmussen.  1975.  Survey and analysis of halocarbons in the




atmosphere  by  gas   chromatography—mass  spectrometry.  Atmos.  Environ.    9:




1014-1017.








Guarino, A. and N. Lioia.  1958.  Clinical  and histopathological findings on a




fatal poisoning with  Granosan.  Folia. Med. (Napoli).  41: 676-690.  (Summarized




in NIOSH, 1976).








Guengerich,   P.P.,   W.M.  Crawford,   J.Y.   Domoredzki,   T.L.   McDonald   and




P.G. Watanabe.  1980.  In vitro 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.  (Fr.)   (Summarized in NIOSH,  1976).








Hadengue, A.  and R. Martin.   1953.   A case of fatal poisoning by dichloroethane.




Soc.  Med. Leg.  33:  247-249.  (Fr.)  (Summarized in NIOSH, 1976).








Handbook of Environmental Data on Organic Chemicals.  1977.  K. Verschueren, ed.,




Van Nostrand  and Co., NY.








Hanst, P.L.  1978.  Part  II.  Halogenated pollutants: Noxious trace gases in the




air.  Chemistry.  51(2):  6-12.
                                     10-1 7

-------
Hardie, D.W.F.  1964.  Chlorocarbons and chlorohydrocarbons.   In:   Kirk-Othmer




The  Encyclopedia  of Chemical  Technology,  2nd  ed.,  volume  5,  Interscience




Publishers, NY.  p. 152-154.








Hawley, G.G.   1981.   The Condensed Chemical  Dictionary.   Tenth  edition.  Van




Nostrand Reinhold Co., NY. p. 431.








Hayes, F.D., R.D. Short and J.E. Gibson.  1973.  Differential toxicity of mono-




chloroacetate, monofluoroacetate and monoiodoacetate in rats.  Toxicol.   Appl.




Pharmacol.  26: 93-102.








Heitmuller, P.T., T.A. Hollister and P.R. Parrish.  1981.   Acute  toxicity of 54




industrial  chemicals to  sheepshead minnows  (Cyprinodon  variegatus).    Bull.




Environ. Contam. Toxicol.  27(5): 596-604.









Hellman, T.M. and F.H. Small.  1973-  Characterization of odors from the petro-




chemical industry.  Chem. Eng. Progr.  69:  75.








Hellman, T.M. and F.H. Small,  n.d.  Characterization off the odor properties of




101 petrochemicals using sensory methods.  J. Air Pollution Control Assoc.  24:



979-982.








Heppel, L.A., P.A. Neeal,  T.L. Perrin, K.M. Endicott and V.T. Porterfield.  1945.




Toxicology of 1,2-dichloroethane.   III.   Its acute toxicity and the  effect of




protective agents.  J. Exp. Pharmacol.  Ther.  83: 53-63.
                                     10-18

-------
Heppel, L.A., P.A. Neal,  T.L.  Perrin,  K.M. Endicott and V.T. Porterfield.  19*16.




Toxicology of  1 ,2-dichloroethane.   V.  Effects  of  daily inhalations.  J. Ind.




Hyg. Toxicol.  28: 113-120.








Heppel,  L.A.,   V.T.  Porterfield  and N.G.  Sharpliss.   19*17.   Toxicology  of




1,2-dichloroethane.  IV.   Its detoxification  by L-cystine,  D.L.-methionine and




certain  other   sulfur-containing  compounds.   J. Pharraacol.  Exp.  Ther.    9"1 :




385-391.









Heppel, L.A. and  V.T. Porterfield.   19*18 •   Enzymatic dehalogenation of certain




brominated and chlorinated compounds.  J. Biol. Chem.  1?6: 763-769.








Heukelekian, H. and M.C. Rand.  1955.  Biochemical oxygen demand of pure organic




compounds.  J.  Water Pollut.  Control Assoc.   29: 10*10-1053.








Hill, D.T.,  T.-W.  Shih,  T.P.  Johnston and  R.F.  Struck.   1978.   Macromolecular




binding and  metabolism  of the carcinogen 1,2-dibromoethane.   Cancer Research.




28: 2*138.








Hinkel, O.K.  1965. Oral dichloroethane poisoning in children.  Deutsche Gesund-




heitswesen.  20: 1327-1331.








Hobbs, F.D.  and J.A.  Key.  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.
                                     10-19

-------
Holman, H.T., H. Birnstiel and P. Jobst.  1971.  On  the  inhalation  toxicity of




1,1- and 1,2-dichloroethane.  Arch.  Toxicology.  27:  248.








Hooper, K.,  L. Gold and B. Ames.  1980.  Pages 65-82  in B. Ames, P. Infante and R.




Reitz, Eds.   Banbury Report No.  5.   Ethylene dichloride:  A  potential  health



risk?  Cold Spring Harbor Laboratory.








Howard, C.J. and  K.M.  Evenson.   1976.  Rate  constants for  the  reactions  of OH




with  ethane  and some  halogen  substituted ethanes at  296°K.    J.  Chem.  Phys.




64(11 ): 4303-4306.









Howe, R.B. and  K.S.  Crump.   1982.   A  computer program to extrapolate  quantral




animal animal toxicity data to low doses.   OSHA Contract  41  USC252C3.








Hueper, W.C. and C. Smith.  1935.  Fatal ethylene  dichloride  poisoning.  Amer. J.




Med. Sci.   189:  778-784.








Hulst, J.P.L.,  A.J.  Steenhauer  and  D.L.  Keede.   1946.    Lethal poisoning  with




dichloroethane.   Ned. Tijdschr Geneeskd.  90: 406-407.   (Out.)  (Summarized in



NIOSH, 1976).








lenistea,  C.  and M.D.  Mezincesco.    1943.    Fatal poisoning  by  ingestion  of




ethylene dichloride.  Bull. Acad. Med. Roum.  8: 614-617.   (Fr.)  (Summarized in



NIOSH, 1976).








International Agency  for  Research  on  Cancer.  1979.  Monograph on the evaluation




of carcinogenic  risk  of chemicals  to  humans.   20:  429-448.
                                     10-20

-------
 International Commission on Radiological Protection  (ICRP).  1977.  Recommenda-




 tion of the ICRP, Pub. No.  26,  adopted  Jan.  17, 1977.  Oxford, England:  Pergamon



 Press.








 Irish,  D.   n.d.   Halogenated hydrocarbons, J.  Aliphatics.   In;   Industrial




 Hygiene  and  Toxicology.  Vol.  2,  F.A.  Patty,  ed.    NY:    Interscience,  p.



 1 28 0-1 28 4.









 Irish, D.D.  1963.  Aliphatic Halogenated Hydrocarbons.   In;  Industrial Hygiene




 and Toxicology.   Second revised  edition,  D.W.  Fassett and D.D.  Irish,  eds.,




 Interscience Publishers.








 Ivanetick, K.M., S. Lucas,  J.A. March,  M.R. Ziman, I.D. Katz and J.J. Bradshaw.




 1978.  Organic  compounds.   Their  interaction  with and  degradation  of hepatic




 microsomal drug-metabolizing enzymes in vitro.  Drug Met. Dispos.  6(3): 218-225.








 Jaeger, R.J., R.B. Conolly and S.D. Murphy.  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.








 Jaeger, R.J.  1979.  Time-related variation of non-protein sulhydryl concentra-




 tions in rat tissue and human  blood.   Int.  Arch. Occup. Environ.  Health.   42:



 141-148.








Jakobson,  I.,  J.E.  Wahlberg, B. Holmberg and G. Johansson.  1982.  Uptake via the




blood  and  elimination  of 10 organic solvents following epicutaneous exposure of




anesthetized guinea pigs.   Toxicol. Appl. Pharmacol.   63: 181-187.
                                     10-21

-------
Jenssen, D. and C.  Ramel.   1980.   The micronucleas  test  as  part  of a  short-term
mutagenicity test program  for the prediction of carcinogenicity evaluated  by 1 43
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.  19663.  Metabolism of  iodomethane in  the rat.  Biochem. J.  98:
38-43.


Johnson, M.K.   I966b.   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.  and K.  Edwards.    1968.   The  comparative  metabolism  of ethylene
dimethanesulphonate and ethylene dibromide.   Experientia.  24:  1100-1101.


Jordi,  A.  1944.   Industrial  poisonings  due to symmetrical 1,2-dichloroethane.
Z. Unfallmed. Berufskr.  37:  131-136.  (Ger.)  (Summarized  in NIOSH,  1976).


Kaira,  P.M.  1966.   Alimentary oral dichloroethane  poisoning.   Klin.  Med.  Mosk.
44: 143-146.   (Rus.)   (Summarized in NIOSH,  1976).


Kaplan, E. and P. Meier.   1958.   Nonparametric  estimation from incomplete  obser-
vations.   J. American Statistical Assoc.   52: 457-481 .
                                     10-22

-------
Kellam,  R.G.  and  M.G.  Dusetzina.    Human  exposure to  ethylene  dichloride:




Potential for regulation via EPA's proposed  airborne carcinogen policy.   In:



Banbury Report No.  5  Ethylene dichloride:  A potential health  risk?   B.  Ames,



P. Infante and R. Reitz,  eds.  Cold Springs Harbor Laboratory,  p. 265-271*.








Kenaga, E.E.  1980.  Predicted bioconcentration factors and soil sorption coeffi-



cients of pesticides and other chemicals.   Ecotoxicol. Environ. Safety.  4:  26-




38.








Keyzer,  J.L.    19UU.    Lethal  poisoning  by  dichloroethane.    Ned.  Tijdschr.




Geneeskd.  88: 6H1 .  (Out.)  (Summarized in NIOSH, 1976).








King, M.T., H. Beikirch,  K. Echkhardt,  E.  Gock and D. Wild.  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: 33-43.








Kirk-Othmer.   1964.  Encyclopedia of Chemical Technology.








Kiriohek, lu.  F.  1974.  Effects of 1,2-dichloroethane.   Usp.  Khira. Mutageneze



Sel.  232-235.








Kistler,  G.H.  and A.B.  Luckhardt.   1929.   Die 13  wichtigsten chlorkohlenw-



asserstaffe der fattreihe vom standpunkt der gewerbehydiene.  Anesth. Analg.  8:




65-78.
                                     10-23

-------
Kokarovtseva, M.G. and N.J.  Kiseleva.   1978.  Chloroethanol, one of  the  toxic




metabolites of 1 ,2-dichloroethane.   Farmakol. J.  Toksikol.   41:  118-120.








Kovar, J.  and D. Krewski.   1981 .   RISKS 1 :    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.   (Rus.)   (Summarized




from English Translation).








Kristoffersson,   U.     1974.    Genetic  effects  of  some  gasoline   additives.




Hereditas.  78:  319.








Krohne,  H.E. and  D.L.  Lingren.   1958.    Susceptibility  of  Life  Stages  of




Sitophilus oryza to various furaigants.  J. Econ.  Entomol.  51(2): 157-158.








Lane,  R.W.,  B.L.  Riddle  and J.F. Borzelleca.  1982.   Effects of 1,2-dichloro-




ethane and 1,1,1-trichloroethane in drinking water on reproduction and develop-




ment in mice.  Toxicol. Appl. Pharmacol.  63: 409-421.








Larkin, R.L., J.V. Crable, L.R. Catlett and M.J.  Seymour.  1977.  Collaborative




testing  of  a gas  chromatographic charocoal  tube  method  for seven  organic




solvents.  Amer.  Inc. Hyg. Assoc.  38: 543-553.








Lehmann,  K.B. and  L.  Schmidt-Kehl.  1936.   The  pharmacology of some ethylene-




halogen compounds.  Arch. J. Hyg.  116: 131-143.
                                     10-24

-------
Letkiewicz, F.,  P.  Johnston, J.  Colman,  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., L.E. Vincent and H.E.  Krohne.   19 54.   Relative effectiveness  of




10 fumigants to adults of 8 species  of stored-product insects.  J. Econ.  Entomol.




17(5):  923-926.








Lindgren, D.L. and L.E.  Vincent.  1959.   J.  Econ.  Entomol.   52:  1091.








Lindgren, D.L.,  W.B.  Sinclair and L.E. Vincent.   1968.   Residues  in raw and




processes  foods   resulting  from  post-harvest  insecticidal  treatments.   In;




Residue Reviews,  volume  21,  F.A. Gunther,  ed.,  Springer-Verlag,  NY.  p.   1-122.








Livesey, J.C.  and M.W. Anders.  1979.  In vitro metabolism of 1 ,2-dihaloethane  to




ethylene.  Drug.  Metab.  Dispos.   7: 199-203.








Livesey, J.C., M.W. Anders, P.W. Langvardt, C.L.  Putzig and H.R.  Reitz.   1982.




Sterochemistry  of  the   glutathione-dependent   biotransformation of   vicinal-




dihaloalkanes to  alkanes.  Drug  Metab.  Dispos.   10:  201-204.








Lochhead, H.B. and H.P.  Close.  1951 .   Ethylene dichloride  plastic  cement — A




case of fatal poisoning.  JAMA.   146:  1323.








Logan,  J.A., M.J. Prather, S.C.  Wofsy and M.B. McElroy.   1981.  Tropospheric




Chemistry:   A global perspective.   J.  Geophys.  Res.  86:  7210-7254.
                                     10-25

-------
Lovelock, J.E.  1971*.  The electron capture detector:   Theory and  practice.   J.




Chromat.  99: 3-12.








Lowenheim, F. and M. Moran.  1975.   Ethylene Bichloride.  In:  Faith, Keyes,  and




Clark's  Industrial Chemicals, 4th  ed.,  John  Wiley and  Sons,  Inc.,  NY.    p.




392-396.








Ludzack, F.J. and M.B.  Ettinger.  1960.  Chemical Structures resistant to aerobic




biochemical stabilization.  J. Water Pollut. Control Fed.   32: 1173-1200.








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, L.I. and  A.S.  Savina.   1976.  Myocardiopathies  in acute poisonings.




Therapeuticheskii Arkhiv.  H8(9): 8U-88.








Luzhnikov, E.A., A.A. Andryukin,  A.S. Savina., et al.  197^-   To title provided.




Therapeuticheskiy Arkhiv.  U6(2): 131-135.








Luzhnikov, E.A., L.I. Petrova, A.S.  Savina,  L.G. Kostomareva and A.V. Zakharova.




1978.   Lesion  of the heart  in  exotoxic  shock.    Kardiologiia.    18: 83-68.




(Summarized in Chem. Abstr.).








Lyman,  W.J.,  W.F.  Reehe and D.H.  Rosenblatt.   1982.   Handbook of  Chemical




Property  Estimation Methods.    Environmental  Behavior of  Organic  Compounds.




McGraw-Hill Book Co., New York,  NY.  960 p.
                                     10-26

-------
Mabey, W.B.,  J.H. Smith,  R.T. Podoll, et al.  1981.   Aquatic  Fate  Process  Data




for Organic Priority Pollutants.  U.S. EPA,  Monitoring  and Data Support Division,




Washington, DC.  EPA-U40/U-81-014.








Maltoni, C., L. Valgimigli and C.  Scarnato.   1980.   In;   Banbury Report No.  5.




Ethylene dichloride: A potential health risk?  B. Ames, P. Infante and R. Reitz,




eds.  Cold Spring Harbor Laboratory,  p. 3-33•








Mantel, N. and M.A. Schneiderman.  1975.  Estimating "safe" levels, a hazardous




undertaking.   Cancer Res.  35: 1379-1386.








Martin, G., K.  Knorpp, K. Huth, F. Heinrich and C. Mittermayer.  1969.  Clinical




features,  pathogenesis  and  management  of  dichlorethane  poisoning.   Ger.  Med.




Mon.  14: 62-67.  (Summarized in NIOSH, 1976).








Martin, G., K. Knorpp,  K. Huth,  F.  Henrich and D. Mittermayer.  1969.  Clinical




features, pathogenesis and management  of dichloroethane  poisoning.   Ger.  Mad.




Mon.  11: 62-67.








McCann, J., V. Simmon, D. Streitwiesser  and B.N.  Ames.   1975.  Mutagenicity of




chloroacetaldehyde, a possible metabolic  product of 1 ,2-dichloroethane (ethylene




dichloride),  chloroethanol (ethylene chlorohydrin), vinyl  chloride,  and cyclo-




phosphamide.   Proc. Natl. Acad.  Sci.  USA.  72(8): 3190-3193.








McCollister,  D.D.,  R.L. Hollingsworth,  F. Oyen and V.K. Rowe. 1956.  Comparative




inhalation toxicity of fumigant  mixtures.  AMA Arch. Ind.  Health.  13: 1-7.
                                     10-27

-------
McConnell, G., D.M. Ferguson and C.R.  Pearson.   1975.   Chlorinated hydrocarbons




and the environment.  Endeavor.  34(121):  13-18.








McCormack, W.B., R. Moore and C.A. Sandy.   1981.   Lead Compounds  (organolead).




In:  Kirk-Othmer Encyclopedia of Chemical Technology, Third edition, Grayson,  M.




and Eckroth, D., eds. John Wiley and Sons,  Inc., NY. Vol.  14,  p.  186-189.








McKenna, M.J.,  J.A.  Zempel, E.O. Madrid,  W.H.  Braun and P.J. Gehring.   1978.




Metabolism and pharmacokinetic profile of vinylidene chloride  in  rats following




oral administration.  Toxicol. Appl. Pharmacol.   45: 821-835.








McNally, W.D. and  G. Fostvedt.  1941.   Ethylene Bichloride - Poisoning.   Ind.




Med. 10: 373-374.








McPherson,  R.W.,  C.M.  Starks and G.J.  Fryar.   1979.   Vinyl  chloride monomer.




What you should know.  Hydrocarbon Processing.  3: 75-88








Melcher, E.G.,  R.R. Langner and R.O. Kagel.  1978.  Criteria for  the evaluation




of methods for the collection of organic pollutants  in air using solid sorbents.




Am. Inc. Hyg. Assoc.  39(5): 349-361.








Menschick,  H.   1957.   Acute poisoning from inhalation of symmetrical dichloro-




ethane.   Arch.  Gewerbepathol.  Gewerbehyg.   15:  241-252.  (Ger.)  (Summarized  in




NIOSH, 1976).
                                     10-28

-------
Metcalf, R.L.  1981.  Insect Control Technology.  In; Kirk-Othmer Encyclopedia of


Chemical Technology, Third edition, Grayson, M. and Eckroth, D., eds. John Wiley


and Sons, Inc.,  NY. Vol. 13, p.
Mill,  T.,  W.R. Mabey,  D.C.  Bomberger, T.-W.  Chon et  al.   1982.   Laboratory


Protocols for Evaluating the Fate of Organic Chemicals  in Air  and  Water.   U.S.


EPA  Report No.  EPA-600/3-8 2-022, Environmental  Research  Laboratory,  Office of


Research and Development, U.S. Environmental Protection Agency, Athens,  GA.






Miller, V.,  R.J.  Dobbs  and S.I. Jacobs.  1970.   Ethylene chlorohydrin  intoxi-


cation with fatality.  Arch.  Dis. Child.  45: 589-590.






Morgan, A.,  A. Black and  D.R.  Belcher.  1972.   Studies  on the absorption of

                                                                   38
halogenated  hydrocarbons  and  their  excretion  in breath  using    Cl  tracer


techniques.  Ann.  Occup. Hyg.  15:  273-282.






Morozov, G.N.   1958.    On acute dichloroethane  poisoning.   Pharm.  Toxicol.


(USSR).  21:  80-83.  (Summarized in  NIOSH,  1976).






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.,


Kansas City,  MO.  Sept.  1982.  p. 52-53.






Munson, A.E.,  V.M.  Sanders,   K.A. Douglas,  L.E.  Sain,  B.M. Kaufmann and  K.L.


White, Jr.   1982.   In vivo assessment  of immunotoxicity.  EHP,  Environ.  Health


Perspect.   H3:  11-52.
                                     10-29

-------
Murray, J.S., K.S. Rao, J.T. Young  et  al.   1980.   Ethylene  dichloride:  single




generation inhalation reproduction study in  rats.  Unpublished study, Toxicology




Research Laboratory, Dow Chemical U.S.A. Midland, MI.








Nachtomi,  E.   1970.   The metabolism  of ethylene  dibromide in the rat.   The




enzymatic reaction with glutathione iri vivo, and in vitro.  Biochem. Pharmacol.




19: 2853-2860.








Nachtomi, E., E. Alumot and A. Bondi.  1966.  The metabolism of ethylene dibro-




mide in the rat.   I.  Identification  of detoxification products in urine.  Isreal




J. Chem.  4: 239-246.








Nakajima, T. and  A.  Sato.   1979.   Enhanced activity of liver drug-metabolizing




enzymes  for  aromatic and  chlorinated hydrocarbons  following food deprivation.




Toxcol. Appl Pharmacol  50(3): 5^9-556.








NAS  (National Academy  of Science).   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.
                                     10-30

-------
NCI  (National  Cancer Institute).   1978.   Bioassay of  1,2-dichloroethane  for




possible carcinogenicity.   NCI carcinogenesis technical report  series  no.  55.



Department of  Health,  Education,  and  Welfare Publication  No.  (NIH)  78-1361.




Washington, DC:  Government Printing Office.








NCI  (National  Cancer Institute).   1979.   Bioassay  of ethylene dibromide  for




possible carcinogenicity.








Neely, W.B. and J.H. Plonka.  1978.  Estimation of time-average hydroxyl radical




concentration in the troposphere.  Environ. Sci.  Technol.  12: 317-321.








Nestmann, E.R., E.  Lee,  T.  Matula, G.R. Douglas and J.  Mueller.   1980.   Muta-




genicity of consituents indentified in pulp and  paper  mill  effluents using the




Salmonella/mammalian-microsome assay.   Mutat.  Res.   79: 203-212.








NIOSH (National Institute for  Occupational  Safety and Health).  1974.  Manual of




analytical Methods,  Health,  Education,  and  Welfare  Publication  No.  (NIOSH)




75-1 21 .








NIOSH Technical Information.  1975.  Collaborative Testing of Activated Charcoal




Sampling Tubes for Seven  Organic  Solvents.  Health, Education and Welfare Publi-




cation 75-184.








NIOSH.  (National  Institute  for Occupational Safety and  Health).  1976.  Criteria




for a Recommended  Standard.  Occupational Exposure to Ethylene Dichloride.  U.S.




Department of  Health,  Education,  and  Welfare.   Superintendent  of  Doc.,  U.S.




Government Printing Office,  Washington, DC.  1 57 p.
                                     10-31

-------
Noetzel, 0.   1944.   Lethal poisoning  with  ethylene chloride.   Chem.  Z.   58:




146-117.  (Ger.)  (Summarized  in NIOSH, 1976).








Noweir, M.H., E.A. Pfitzer and T.F.  Hatch.   1972.   Decomposition  of chlorinated




hydrocarbons; A review.   J. Am. Ind. Hyg.  Assoc.   33(7):
Nylander, P.O.,  H.  Olofsson and  B.  Rasmuson.   1979.   The  use of  Drosophila




melanogaster as a test for indirect mutagens.   Mutat.  Res.   64:  122-123.








Ohta, T., M. Morita and I. Mizoguchi.   1976.   Local distribution of chlorinated




hydrocarbons in the ambient air in Tokyo.   Atmos. Environ.   10:  557-560.








Okuno, T., M. Tsiji, Y. Shintani and H. Watanabe.  1974.   Gas chromatography of




chlorinated hydrocarbons in urban air.   Hyogo-ken Kogai Kenkyusho Kenyo Hokoku.




6: 106.  (CA (1977), 87: 72564f).








Ollivier, H., P. Grillo-Abade,  G. Helvadjian  and J. Quicke.   1954.   Report  of a




fatal  case  of  intoxication by  dichloroethane.   Soc.  Med.  Leg.   34: 261-264.



(Fr.)  (Summarized in NIOSH, 1976).








Otterson, E.J.  and C.U.  Guy.    1964.   A method of atmospheric solvent vapor




sampling on activated charcoal  in connection with gas chromography.   Trans.  26th




Annual Meeting, Amer. Conf.  of  Governmental Industrial Hygienists, Philadelphia.




p. 37-43.
                                     10-32

-------
Page, G.W.  1981 .   Comparison of groundwater and surgaoe water for patterns and




levels of contamination by toxic substances.  Environ. Sci. Technol.  15:  1*175-




1181.








Page, B.D.  and B.P.C.  Kennedy.   1975.   Determination of  methylene  chloride,




ethylene dichloride, and trichloroethylene as solvent residues in spice oleore-




sins, using  vacuum  distillation and electron  capture gas  chromatography.   J.




Assoc. Off. Anal.  Chem.  58(5): 1062-1068.








Paparopoli, G. and  V.  Cali.   1956.  Collective  intoxications with chlorinated




hydrocarbons  in dock workers.   Folia.  Med.   (Napoli)   39:  819-831.    (Ita.)




(Summarized in NIOSH, 1976).








Patterson, R., M.  Bornstein, R. Hall and E. Garshick.   1975.   Assessment  of




Ethylene Dichloride  as a Potential Air  Pollution Problem, vol.  III.    Report




number GCA-TR-75-32-G(3), GCA Corp.,  Bedford, MA.  25 p.








Pavlova, I.V., P.A.  Rosenberg,  N.K. Byalko and N.A. Gel'fon. 1965.  Biochemical




changes in  the blood  during acute intoxications  by some  chlorinated  hydroc-



arbons.   Prof. Zabol. V.  Khim. Prom.  p.  217-224.  (Rus.)  (Summarized  in NIOSH,




1976).








Pearson, C.R.  and G. McConnell.  1975.  Chlorinated C1  and  C~ hydrocarbons in the




environment.   Proc.  R.  Soc. Lond. B.  189:  305-332.marine
                                     10-33

-------
PEDCO Environmental  Inc.   1979.    Monitoring of  ambient  levels  of  ethylene




dichloride (EDC) in  the  vicinity  of EDC production and uses  facilities.   EPA,




Contract No.  68-02-2722.








Pellizzari, E.D.  1971.  Electron capture  detection  in gas chromatography.  J.




Chromat.  98: 323-361.








Pellizzari, E.D.  I977a.   "The Measure of Carcinogenic Vapors in Ambient Atmos-




pheres,"  NTIS  PB-269582,  EPA-600/7-77-055,  prepared for  U.S.  Environmental




Protection Agency by Research Triangle Institute, Research Triangle Park,  NC.








Pellizzari, E.D.   I977b.  "Analysis of  Organic  Air  Pollutants by  Gas Chroma-




tograph and Mass Spectroscopy," Research Triangle  Institute,  Research Triangle




Park, NC, EPA 600/2-77-100.








Pellizzari,  E.D.    I978a.    Preliminary   assessment  of  halogenated  organic




compounds in man and environmental media.  Monthly technical progress report No.




2  (January 1-30, 1978).  Prepared for the U.S. Environmental Protection Agency,




Research Triangle Park,  NC.  Contract No. 68-01-4731.








Pellizzari,  E.D.    I978b.    Preliminary   assessment  of  halogenated  organic




compounds in man and environmental media.  Monthly technical progress report No.




5  (April  1-April  30,  1978).  Prepared  for the U.S.  Environmental Protection




Agency, Research Triangle Park, NC.   Contract No. 68-01-4731.
                                     10-34

-------
Pelizzari,  E.D.   I978c.  Preliminary assessment of halogenated organic compounds




in  man  and  environmental  media.   Monthly  technical  progress  report  No.  10




(September 1-30,  1978).   Prepared for  the U.S.  Environmental  Protection  Agency,



Research Triangle Park,  NC.   Contract  No. 68-01-4731.








Pellizzari,  E.D.   I978d.    Preliminary  assessment  of  halogenated  organic




compounds in man  and environmental media.  Monthly technical progress report No.




11 (October 1-30, 1978).  Prepared for the U.S.  Environmental  Protection  Agency,




Research Triangle Park,  NC.   Contract  No. 68-01-4731.








Pellizzari,  E.D.   1978e.     "Quantification  of  Chlorinated  Hydrocarbons  in




Previously  Collected   Air   Samples,"   Research  Triangle  Institute, Research




Triangle Park, NC, EPA 450/3-78-112, NTIS PB 289804.








Pellizzari,  E.D.    1978.     Quantification  of  chlorinated  hydrocarbons  in




previously  collected  air  samples.    U.S.  Environmental  Protection  Agency,




Research Triangle Park,  NC.   EPA-450/3-78-112.








Pellizzari, E.D.   1978.  Measurement  of carcinogenic vapors  in  ambient atmos-




pheres.   EPA-600/7-78-062.








Pellizzari, E.D.   I979a.  Information  on the Characteristics  of Ambient  Organic




Vapors in Areas of High  Chemical Production.   Prepared  for U.S. EPA by Research




Triangle Institute,  Research Triangle  Park,  NC.
                                     10-35

-------
Pellizzari, E.D.   I979b.   "Organic  Screening in Lake  Charles,  LA,  Using  Gas




Chromatography Mass Spectrometry  Computer  Techiques,"  EPA Contract 68-02-2714,




Research Triangle Institute, Research Triangle Park, NC. (August).








Pellizzari, E.D.  and  J.E.  Bunch.    1979.    Ambient  air  carcinogenic  vapors:




Improved sampling  and analytical  techniques.   Prepared  by Research  Triangle




Institute  for  Environmental Sciences  Research Laboratory, U.S.  Environmental




Protection Agency,  Research Triangle Park,  NC.  Contract No. 68-02-2764.








Pellizzari, E.D.,  M.D.  Erickson  and  R.A.  Zweidinger.   1979.    Formulation  of




preliminary assessment of halogenated  organic compounds in man and environmental




media.  U.S. Environmental Protection Agency,  Research Triangle Park,  NC.   EPA-



560/13-79-006.








Pellizzari, E.D., M.D. Erickson  and  R.A. Zweidinger.  1979.   Formulation  of a




preliminary assessment of halogenated  organic compounds in man and environmental




media.   U.S.  Environmental Protection Agency,  Office  of Toxic  Substances,




Washington, DC.  U.S.  EPA 560/13-79-006. NTIS  PB  80-112170.  Contract  No.  68-



01 4731 •








Perocco, P. and G.  Prodi. 1981.   DNA damage by haloalkanes in human lymphocytes




cultured in vitro.   Cancer Letters.  13: 213-218.








Pervier, R., R. Barley,  D. Field,  B. Friedman, R. Morris and W.  Schwartz.  1974.




Survey Reports on Atmospheric  Emissions From  the Petrochemical  Industry,  Vol.




II.  Environmental Protection Agency, Contract No.  68-02-0255.   Available  from




the National Technical Information Service,  Springfield, VA., PB-244958, p.  135.
                                     10-36

-------
Piet,  G.J.,   P.   Slingerland,  F.E.   de  Grund,   M.P.M.   v.d.  Heuvel   and




B.C.J. Zoeteman.   1978.   Determination  of very  volatile halogenated  organic



compounds in water by means  of direct head-space analysis.  Anal. Lett.  All(5) '•



U37-UH8.








Plotnick, H.B., W.W.  Weigel,  D.E.  Richards,  K.L.  Cheever  and  C.  Kommirreni.



1980.  Dietary disulfiram  enhancement of the toxicity of ethylene dibromie.   In:



Banbury Report No. 5, Ethylene dichloride:  A potential  health risk?   B.  Ames,



P. Infante and R.  Reitz,  eds., Cold Spring Harbor Laboratory,  p.  279-286.








Price, K., G.  Waggy and R. Conway.  197^.  Brine shrimp bioassay and seawater BOD



(biochemical oxygen demand)  of petrochemicals.  J. Water Pollution Control Fed.




16(1): 63-77.








Rannug, U.  1976.   Mutagenic studies of EDC tars as a model for tests of complex



mixtures  of chemicals in  the environment.   Nordforsk Meljoevardssekr.   Publ.  (2



Org. Miljoegifter  Vaten,  Nord. Symp. Vattenforsk,  12th)  561-565.








Rannug, U.   1980.   Genotoxic effects  of 1 ,2-dibromoethane and  1 ,2-dichloro-



ethane.  Mutat. Res.   76:  269-295.








Rannug, U.  1980.   The use of different metabolizing systems in the elucidation



of the mutagenic  effects  of ethylene dichloride   in  Salmonella.   In;   Banbury



Report No. 5,  Ethylene dichloride:  A potential health risk?  B. Ames,  P.  Infante



and R. Reitz,  eds., Cold  Spring Laboratory, p. 83-92.
                                     10-37

-------
Rannug, U.  and  C. Ramel.   1977.   Mutagenicity of  waste  products from  vinyl




chloride  industries.    Journal  of  Toxicology  and  Environmental  Health.    2:




1019-1029.








Rannug, U. and B. Beiji.   1979.   The mutagenic effect of 1,2-dichloroethane on




Salmonella  typhimurium.   II. Activation  by  the  isolated  perfused rat  liver.




Chemico-Biological Interactions.  24: 265-285.








Rannug,  U.,  A.  Sundvall  and  C.   Ramel.    1978.    The mutagenic  effect  of




1,2-dichloroethane on Salmonella typhimurium I.  Activation through conjugation




with glutathione in vitro.  Chemico-Biological Interactions.  20: 1-16.








Rao, K.S.,  J.S.  Murray, M.M. Deacon,  J.A. John, L.L.  Calhoun  and J.T.  Young.




1980.   Teratogenicity and  reproductive studies  in  animals  inhaling  ethylene




dichloride.  Bandury Report.  5: 149-166.








Reichert, D. and D. Henschler.  1978.  Uptake and hepatotoxicity of 1,1-dichloro-




ethylene  by  the  isolated  blood-perfused rat  liver.   Int. Arch.  Occup. Environ.




Health.   41 : 169-178.








Reinfried, H.  1958.   On lethal poisonings due to ingestion of 1 ,2-dichloroethane




containing  rubbing compounds.   Deutsch. Gesundheitswes.  13:  778-779.  (Ger.)




 (Summarized  in NIOSH, 1976).
                                     10-38

-------
Reitz,  R.H.,  T.R.  Fox,  J.Y.  Domoradzki,  J.F.  Quast,   P.   Langvardt   and




P.G. Watanabe.   1980.    In;    Banbury Report  No.  5,  Ethylene  dichloride:    A




potential health risk?   B. Ames, P.  Infante  and R.  Reitz,  eds.,  Cold  Spring




Harbor Laboratory,  p.  ^35-^^8.








Reitz, R.H.,  T.R. Fox, J.C. Ramsey, J.F. Quast,  P.W. Langvardt and P.G. Watanabe.




1982.  Pharmacokinetics and micromolecular interactions of ethylene  dichloride




in rats after inhalation or gavage.   Toxicol.  Appl.  Pharmacol.   62:  190-204.








Rejsek, K.  and M. Rejskova.  19**7.  Intoxication with symmetrical  dichloroethane.




Cas. Lek. Cesk.   86: 207-209.   (Cze.)  (Susmmarized  in NIOSH,  1976).








Rohmann, E.,  D. Zinn and J. Kulz.  1976.  Electroencephalographic  observations in




childhood poisonings and their  therapeutic  consequences.    Kinderaeztl.  Prax.




37: 209-216.   (Ger.) (Summarized  in  NIOSH,  1976).








Rosenbaum,  N.  1939.  Use  of dichloroethane  in  industry from the standpoint of




occupational   hygiene,  in  [Dichloroethane].   Moskow,   Chap.  IV,  p. 109-113-




(Rus.)  (Summarized in  NIOSH,  1976).








Rosenbaum,  N.D.   19^7.  Ethylene dichloride as an industrial poison.  Gig. Sanit.




12(2): 17-21,  (Rus.)   (Summarized from English Translation).








Roubal, J.  19^7.   Two fatal cases of intoxication with symmetric dichloroethane




ingestion.  Cas. Lek. Cesk. 86: 203-206.  (Cze.)  (Summarized  from NIOSH, 1976).
                                     10-39

-------
Salmon, A.G., W.C. Mackrodt,  R.B.  Jones, S.K.  Basu and J.  Ashby.   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.  450): Abst. 253, 277.








Salvini, M. and  B. Mazzucchelli.  1955.  Intoxication from fumes of carbon tetra-




chloride and dichloroethane used as fumigant parasiticides.   Minerva. Med.  49:




2295-2304.  (Ita.)  (Summarized from NIOSH,  1976).








Sato,  A.  and  T.  Nakajima.   1979.   A  structure-activity relationship  of some




chlorinated hydrocarbons.  Arch. Environ. Health.   34(2): 69-75.








Sato, A., T. Nakajima and Y. Koyama.  1980.  Effects of chronic ethanol  consump-




tion on hepatic  metabolism of  aromatic and chlorinated hydrocarbons in rats.  Br.




J. Ind. Med.  37: 382-386.








Sato,  A.,  T.  Nakajima and Y. Koyama.  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,  A.A. Crawford, J.A. John et al.  1979.  The effects of inhaled




ethylene  dichloride  on  embryonal  and  fetal development  in rats and  rabbits.




Unpublished study, Toxicology Research Laboratory,  Dow Chemical U.S.A.  Midland,




MI.
                                     10-40

-------
Schoborn, H.,  W. Prellwitz and P. Baum.  1970.  Consumption coagulation pathology




of  1 ,2-dichloroethane  poisoning.    Klin.  Wochenschr.    48:822-824.    (Ger.)




(Summarized from NIOSH,  1976).








Schwartz, W.A., F.G. Higgins, Jr.,  J.A. Lee, R. Newirth and J.W. Pervier.  1974.




Engineering and  Cost Study  of  Air Pollution  Control  for  the  Petrochemical




Industry.    vol.  3.    Ethylene  Bichloride  Manufacture  by  Oxychlorination.




EPA-450/3-73-006c.  Houdry  Division, Air Products and Chemicals, Inc.   Report




prepared for U.S.  Environmental Protection Agency, Research Triangle Park,  NC.








Secchi, G.C.,  G.  Chiappino,  A.  Lotto  and N.  Zurlo.   1968.   Actual  chemical




composition of the "commercial trieline"  and their hepatotoxic effect -  Clinical




and enzymological  studies.   Med. Lav.   59:  486-497.    (Ita.)    (Summarized  in




NIOSH, 1976).








Seufert, F.B., P.  Brown, J.A.  Oatway, M. Bornstein, W. Ostrowski and R. Home.




1980.   1,2-Dichloroethane:  Technical control options analysis.   Prepared by CGA




Corp., Bedford, MA, for  U.S. Environmental  Protection  Agency.   Control  No.   68-




01-5960 as cited in Letkiewicz et al.,  1982.








Shackelford, W.M.  and L.H.  Keith.  1976.  Frequency of Organic Compounds Identi-




fied in Water.  EPA-600/4-76-062.   Environmental  Research Laboratory, Office of




Research and Development,  U.S. Environmental Protection Agency,  Athens, GA.  p.




134.
                                     10-41

-------
Shakarnis, V.F.  19&9.  Induction of  X-chromosome  nondisjunction  and  recessive




sex-linked recessive lethal mutations in  females of  Drosophila  tnelanogaster  by




1,2-dichloroethane.  Sov.  Genetics.   5:  1666-1671.








Shakarnis, V.F.   1970.   Effect of  1 ,2-dichloroethane.   Vesnik  Leningradsku-




nivsiteta Seviya Biologii.  25(1):  153-156.








Sharael, R.E.,  R. Williams,  J.K. O'Neill,  R. Eller,  R. Green, K.D.  Hallock and




R.P. Tschirch.   1975.    Preliminary  Economic  Impact Assessment  of  Possible




Regulatory Action  to  Control  Atmospheric  Emissions  of Selected  Halocarbons.




EPA-450/3-75-073.  Arthur D. Little,  Inc.,  Cambridge, MS,  Report  prepared for




U.S. Environmental Protection Agency, Research Triangle Park,  NC.








Shchepotin, B.M. and  Ya. D. Bondarenko.  1978.  Clinical syndromes and pathogenic




treatment principles of dichloroethane intoxications.  Vrach. Delo.  7: 13^-139-








Siegel, I.M.  19*47.  Ethylene dichloride.   JAMA. (Lett).   133:  577.








Simmon, V.F.   1980.   Review of nonbacterial tests  of the genotoxic activity of




ethylene dichloride.  In;   Banbury Report 5.  Ethylene dichloride:  A potential




health  risk?    B.N.  Ames, P.  Infante  and  R. Reitz,  eds.   Cold  Spring Harbor




Laboratory Press.  Cold Spring Harbor, NY.  p. 97-103.








Singh, H.B.  1977.   Atmospheric halocarbons.  1977.  Evidence of  favor of reduced




average hydroxyl radical  concentrations in the troposphere.   Geophy. Res. Lett.




1(3): 101-104.
                                     10-1*2

-------
Singh, H.B.  1978.  Letter to the editor.  Atmos. Environ.  12: 1809.




Singh, H.B., L. Salas, D. Lillian,  R.R.  Arnts  and A. Appleby.  1977.  Generation
                                    •
of accurate halocarbon primary standards with permeation  tubes.   Environ.  Sci.


Technol.  11(5): 511-513.




Singh, H.B., L.J.  Salas,  H. Shigeishi, A.J. Smith, E. Schibner and L.A. Cavanagh.


1979.   Atmospheric  distributions,  sources and  sinks  of  selected  halocarbons,


hydrocarbons, SFg and N02.  Grant No. 8038020. EPA-600/3-79-107.




Singh, H.B., L.J.  Salas,  A. Smith and H. Shigeishi.   1980.   Atmospheric measure-


ments  of  selected toxic  organic  chemicals -  interim  report  1979.   Grant  No.


805990. EPA-600/3-80-072.




Singh, H.B., L.J.  Salas,  A.  Smith and P. Shigeshi.   1980.   Atmospheric measure-


ments  of  selected toxic  organic chemicals:  Halogenated  alkanes,  chlorinated


aromatics, aromatic hydrocarbons  and  secondary organics.  Prepared by SRI Inter-


national  for  Environmental   Sciences  Research  Laboratory,  U.S.  Environmental


Protection  Agency, Research  Triangle Park, NC.   Interim  report.   SRI Project


777*4.




Singh, H.B., L.J. Salas,  A.J.  Smith and H. Shigeishi.  1981.  Measurements of


some  potentially  hazardous  organic chemicals in urban environments.   Atmos.


Environ. 15(U): 601-612.
                                     10-U3

-------
Singh, H.B.,  L.J. Salas and R.E.  Stiles.  1982.  Distribution of Selected Gaseous




Organic Mutagens and Suspect  Carcinogens in Ambient Air.  Environ. Sci.  Technol.




16(12): 872.








Singh, H.B.,  L.T. Salas and  R.E. Stiles.  1983.   Selected man-made halogenated




chemicals in the air and oceanic environment.   J.  Geophys. Res.   88: 3675-3683.








Singh, H.B.,  L.J.  Salas,  R.  Stiles and H.  Shigeishi.   1983.  Measurements  of




Hazardous Organic Chemicals  in  the Ambient Atmosphere.   Grant No.  CA  805990.




U.S.  Environmental   Protection  Agency,  Office  of Research  and  Development,




Research Triangle Park, NC.  EPA-600/3-83-002,  Avail.  NTIS, PB-83 156935.








Sipes, I.G.  and A.J. Gandolfi.  1980.   In vitro  comparative  bioactivation  of




aliphatic halogenated hydrocarbons.  Dev.  Toxicol. Environ. Sci.   8: 501-506.








Sjoberg, B.  1952.   Thermal  decomposition of chlorinated  hydrocarbons.  Svensk




Kern.  Tidskr.  64: 63.








Smirnova, N.A. and H.P. Granik.  1970.  On  the remote effects of acute occupa-




tional poisoning with some carbohydrates and their derivatives.  Gig. Tr.  Prof.




Zabol.  U(5): 50-51.  (Rus.)  (Summarized in NIOSH,  1976).








Smyth, H.F.,  Jr.  1956.  In:  Handbook of Toxicology,  Vol.  I., W.S.  Spector, ed.








Snapp, O.I.  1958.  Trunk sprays for control of the peach tree borer.  J.  Econ.




Entomol.  51(4): 557-558.
                                     10-44

-------
Snelson, A., R.  Butler  and F. Jarke.   1978.   Study  of removal  processes  for




halogenated air pollutants.  EPA-600/3-78-058,  Office  of Research and  Develop-




ment, U.S. Environmental Protection Agency,  Washington, DC.








Sopikov, N.F. and A.K.  Gorshunova.   1979-   Investigation of  the uptake, distri-




bution, and excretion of ethylene dichloride in rats.  Gig. Tr.  Prof.  Zabol.   4:




36-10.








Spence, J.W. and P.L. Hanst.   1978.  Oxidation of chlorinated ethanes.   J.  Air.




Pollut. Control Assoc.   28(3):  250-255.








Spencer, B.C., V.K.  Rowe,  E.M.  Adams,  D.D. McCollister and  D.D.  Irish.   1951.




Vapor toxicity of  ethylene dichloride  determined by experiments  on  laboratory




animals.  Industrial Hygiene  and Occupational Medicine.  4:  482.








Spreafico, F.,  E. Zuccato, F.  Murcurei et al.  1978.  Metabolism of 1 ,2-dichloro-




ethane in  experimental  animals,  Report  Nos.  1 and  2 to Chemical Manufacturers




Association, New York,  NY.








Spreafico, F.,  E. Zuccato, F. Murcurei et al.  1979.  Distribution and metabolism




of 1 ,2-dichloroethane (EDC)  in  experimental  animals.    Report  Nos.  3  and 4  to




Chemical Manufacturers  Association, New York,  NY.
                                     10-45

-------
Spreafico, F., E.  Zuccato,  F. Murcurci  et al.   Pharmacokinetics of  ethylene




dichloride in  rats treated by  different routes  and  its long-term  inhalatory




toxicity.  In:  Banbury Report No. 5, Ethylene dichloride:   A  potential  health



risk?  B. Ames, P.  Infante and R.  Reitz, eds., Cold Spring Harbor Laboratory,  p.




107-133.








SRI  International.   1979.  Population  exposure  to EDC.   Menlo Park, CA: SRI




International.  U.S. EPA  Contract No.  68-02-2835, as cited  in  memorandum  from




David R. Patrick to Lester Grant,  U.S. EPA, Research Triangle Park, NC, December




14, 1982.








SRI International.   1983.  1983 Directory of Chemical Producers: United States  of




America.  SRI International, Menlo Park, CA. p. 584,  771, 949,  965,  583.








SRI  (Stanford Research  Institute).    1981.   National  Screening Program for




Organics  in Drinking Water.   Boland,  PA:  SRI International.  EPA  Contract No.




68-01-4666.








Stoelting, R.K. and E.I. Eger.  II. Percutaneous loss of nitrous oxide,  cyclo-




propane, ether, and haloethane in man.  Anesthesiology.  30: 278-283.








Stolzenberg, S.J.  and C.H.  Hine.   1980.   Mutagenicity of 2- and 3-carbon halo-




genated  compounds  in the Salmonella/mammalian-microsome test.   Environmental




Mutagenesis.  2: 59-66.








Storer,  R.D.,  T.A. Bank and R.D.  Conolly.   1982.  In vivo genotoxic effect  of




1,2-dichloroethane in male B6C3F1 mice.  Toxicologist.  2: 129.
                                     10-46

-------
Storey, C.L., L.D. Krik and G.C.  Mustakas.   Fate of EDC-CCl^  (75:25)  residues




during milling  and oil  extraction of  soybeans.   J.  Econ.  Entomol.    65(4):




11 26-1129.








Stover, E.L.  and  D.F. Kincannon.   1983.   Biological treatability  of  specific




organic  compounds  found in  chemical indusry  wastewater.    J. Water.  Pollut.




Control. Fed.  55:  97-109.








Stuhlert,  H.   19^9-   Fatal  poisoning from  ethylene  chloride.   Dtsch.  Med.




Wochenschr.  7*»:  15M2-15U3.  (Ger.)  (Summarized in NIOSH,  1976).








Suta,  B.E.   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, NC, under Contract




No.  68-02-2835,  Task 17.   Final Report.








Suveev, I.M. and M.E.  Babichenko.  1969.  Clinical picture and treatment of acute




poisoning  with dichloroethane  vapors.   Gig.  Tr. Prof.  Zabol.   13O):  50-51.




(Rus.)  (Summarized in NIOSH,  1976).








Sykes, J.F.  and  A.F.  Klein.   1957.  Chloro-organic residues  in milk  of  cows




orally administered ethylene  dichloride.    J.  Assoc. Off.  Agric.  Chem.   MO:




203-209.








Sykes, J.F.  and  A.K.  Klein.   1957.   Participating Chemists.   Chloro-organic




residues in milk of cows orally administered ethylene dichloride.   Assoc.  Off.




Agric. Chem.  40(1):  206-209.
                                     10-U7

-------
Sykes, J.F. and A.K.  Klein.   1957.   Fumigant  residue-in-milk study.   Chloro-




organic residues in milk of cows orally administered ethylene dichloride.  Assoc.




Off. Agric. Chem.   40(1):  203-206.








Symons,  J.M.,  T.A.  Bellar,   J.K.  Carswel,  et al.  1975.   National  organics




reconnaissance survey  for halogenated  organics.    J.  Am.  Water Works  Assoc.




67(11): 634-647.








Tan, E.L. and A.W. Hsie.   1981.  Mutagenicity and cytotoxicity of haloethanes as




studied in the CHO/HGPRT system. Mutat.  Res.   90:  183-191.








Tabak, H.H., S.A. Quave,  C.I.  Mashni and E.F. Earth.  1981.   Biodegradability




studies with organic priority pollutant compounds.  J. Water Pollut.  Control Fed.




53: 1503-1518.








Theiss, J., G.  Stoner, M.  Schimkin  and E.I Weisburger.  1977.  Test  for carcino-




genicity of organic contaminants of  United States drinking water  by  pulmonary




tumor response in  Strain A mice. Cancer Res.   37:  2717-2720.








Troisi, F.M. and D.  Cavallazzi.  1961.  Fatal  poisoning from  the inhalation of



dichloroethane fumes.   Med.  Lav.   52: 612-618.  (Ita.)   (Summarized  in NIOSH,




1976).








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(4):  27-236.
                                     10-48

-------
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-^, 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. EPA.  n.d.  A Study of Industrial Data on Candidate Chemicals for Testing.




EPA-560/5-77-006.  SRI International, Menlo Park, CA.








U.S. EPA.  n.d.  Determination of Population Density Around Ethylene Dichloride




Production Facilities.  EPA Contract Numer 68-02-2836.








U.S. EPA.  1977.  National Organics Monitoring Survey.  Office of Water Supply,




as cited in Letkiewicz et al., 1982.








U.S. EPA.   1979.   Health Assessment Document for  1,2-Dichloroethane (Ethylene




Dichloride).  Draft  report prepared  by  the Environmental Criteria and Assessment




Office, Office of Health  and Environmental Assessment, Office of  Research and




Development, U.S. Environmental  Protection Agency,  Research Triangle Park,  NC.








U.S. EPA.  1979.  Monitoring  of ambient levels  of  ethylene dichloride (EDO in




the vicinity of EDC  production and user facilities.   EPA-600/4-79-029.  Environ-




ment Monitoring and  Support Laboratory, Research Triangle Park,  NC.  131  p.
                                     10-49

-------
U.S. EPA.  1979.  Monitoring of Ambient Levels of EDC Near  Production  and  User




Facilities.  EPA-600/4-79-029.  Environmental  Protection  Agency,  Environmental




Monitoring and Support Laboratory,  Research Triangle Park, NC.   131  p.








U.S. EPA.  I979a.  Population Exposure to EDC.  Draft report.  SRI International,




Menlo Park, CA.  Contract Number 68-02-2835.








U.S. EPA.   I979b.  Investigations  of Selected Environmental Pollutants:  1,2-




Dichloroethane.  EPA 560/2-78-006.








U.S. EPA.  I98la.   Community Water  Supply Survey.   Office  of Drinking Water,  as




cited in Letkiewicz et al., 1982.








U.S.  EPA.    I98lb.   Draft  criteria document  for  1 ,2-dichloroethane.    U.S.




Environmental Protection Agency, Office of  Drinking Water,  Washington, DC.,  as




cited in Letkiewicz et al., 1982.








U.S. EPA.  1981.  The  determination  of halogenated chemical indicators of indus-




trial contamination in water by the  purge-and-trap  method,  method  502.1.   U.S.




Environmental  Protection  Agency, Environmental Monitoring  and  Support Labora-




tory, Cincinnati,  OH.   EPA 600/4-81-059.








U.S. EPA.   I982a.  Handbook for sampling and  sample preservation  of water and




wastewater.  Environmental Monitoring and Support Laboratory, U.S. Environmental




Protection Agency, Cincinnati,  OH.  EPA 600 A-82-029.
                                     10-50

-------
U.S. EPA.  1982b.   Test methods for evaluating  solid  waste,  physical/chemical



methods.   Office of  Solid  Waste and  Emergency Responses, U.S.  Environmental



Protection Agency, Washington, DC.   SW-846.








U.S. EPA.  19820.  Methods for Organic  Chemical Analysis of Municipal and Indus-



trial Wastewater.   J.E. Longbottom and  J.J.  Lichtenburg, eds.   Environmental



Monitoring  and  Support  Laboratory,   U.S.  Environmental  Protection  Agency,




Cincinnati, OH.  EPA-600/4-82-057.








U.S. EPA.  1982.  Groundwater Supply  Survey, as cited in Letkiewicz  et al., 1982.








USITC (International Trade Commission).  1975-1983. Synthetic Organic Chemicals.



U.S. Production and Sales, U.S. International Trade Commission.








USITC (International  Trade  Commission).    1977-   Synthetic Organic  Chemicals.



USITC Publication Number 20.








USTC  (Tariff  Commission).    1952-197*1.    Synthetic  Organic  Chemicals.    U.S.



Production and Sales.   United States Tariff Commission.








Valvani, S.C., S.H. Yalkowsky and T.J. Roseman.   1981.   Solubility and parti-



tioning.   IV.  Aqueous solubility and  octanol-water  partition coefficients  of



liquid nonelectrolytes.  J.  Pharm. Sci.  70(5):  502-507.








Van Duuren, B., B. Goldschmidt, G. Loewengart et al.   1979.  Carcinogenicity of



halogenated olefinic and aliphatic hydrocarbons  in mice.   J.  Natl. Cancer Inst.



63:  1133-11139.
                                     10-51

-------
Van Dyke,  R. and C. Wineman. 1971-  Enzymatic dechlorination.  Dechlorination of




chloroethane and propanes in vitro.  Biochem.  Pharmacol.  20:  463-470.








Vesar, Inc.  1975.   Identification  of Organic Compounds in Effluents from Indus-




trial Sources.  Final Report to EPA Office of Toxic Substances, Washington,  DC,




EPA-560/3-75-002.








Vincent,  L.E.  and D.L. Lindgren.   1965.   Influence  of fumigation and  age  on




carbon  dioxide  production  of some stored-product  insects.   J.  Econ.  Entomol.




58(4): 660-664.








Von  Oettinger,  W.F.   1964.   The Halogenated  Hydrocarbons of  Industrial  and




Toxicological Importance.  Elsevier,  Amsterdam, London, New York.








Vozovaya,  M.   1971.   Changes  in the esterous cycle  of  white  rats chronically




exposed to the  combined  action  of gasoline and dichlorethane vapors.   Akush.




Genekol.  (Kiev).  47(12): 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.  1975.  The effect of low concentrations of benzene, dichloroethane,




alone  and their  combination  on the reproductive  function animals and  on  the




development of progeny.  Gig. Tr. Prof. Zabol.  7: 20-23
                                     10-52

-------
Vozovaya, M.  1976.  Effect of low concentrations of benzene and dichloroethane



separately and combined on the reproductive  function  animals.   Gig.  Sanit.   6:



100-102.








Vozovaya, M.   1977.   The  effect of  dichloroethane on  the sexual cycle  and



embryogenesis of  experimental animals 1977.  Akusk. Ginekol.  (Moscow).  2: 57-59.








Wadhi, S.R.  and T. Scares.  196U.   Comparative toxicity of some fumigants to the



Red Scale,  Aonidrella  aurantii (Maskell)  (Hemiptera:   coccoidea).  Indian  J.



Entomol.  27: 85-88.








Wallace, L.   1981 .  "Measurements of Volatile Organic Compounds in Breathing Zone



Air, Drinking Water and Exhaled Breath  (Preliminary Draft).  Cited in Brodzinsky



and Singh, 1982.








Ward, J.  1980.  Pages 35-5M in B. Ames, P. Infante  and  R.  Reitz,  Eds.   Banbury



Report No. 5.  Ethylene dichloride: A potential health risk.  Cold Spring Harbor



Laboratory.








Watrous, R.M.  19U7. Health hazards of the pharmaceutical industry. Br. J. Inc.



Med.  1J: 111-125.








Weast, R.C.   1980,  Handbook of Chemistry and Physics, Sixty-First edition.  CRC



Press Inc.,  Boca  Raton, FL.   p.  C-309.








Weisburger,   E.    1977.   Carcinogenicity  studies on halogenated  hydrocarbons.



Environ.  Health  Perspect.  21:  7-16.
                                     10-53

-------
Weiss, F.  1957.  Lethal oral  poisoning  from dichloroethane.   Arch.  Gewerbe-



pathol.  Gewerbahyg.  15:  253-26U.   (Ger.)   (Summarized  in  NIOSH,  1976).








Wendel, H.   19^.   Lethal  poisoning from dichloroethane  (ethylene  chloride).



Pharmazie.  3: 398-400.  (Ger.)  (Summarized in NIOSH, 1976).








Whitney, W.  1961.   Fumigation hazards as related to the  physical,  chemical,  and



biological properties of fumigants.   Pest.  Control.  29(7): 16-21.








Whittemore, A. 1978.  Quantitative theories  of  carcinogenesis.  In:  Advances in



cancer research.  Vol. 27.   Academic Press,  New York,  NY.   p.  55-58.








Whittemore, A. and  B.  Keller.   1978.   Quantitative theory of  carcinogenesis.



Society for Industrial and Applied Mathematics  Review.   20: 1-30.








Williams, R.T.  1959.  Detoxication  mechanisms.  London,  p. 55.








Wilson, J.T., C.G.  Enfield,  W.J. Dunlap, R.L. Cosby, D.A. Foster and L.B.  Baskin.



1981.  Transport and  fate of selected  organic  pollutants  in a sandy soil.   J.



Environ. Qual.  10(4): 501-506.








Wirtschafter,  Z.T.  and  E.D.   Schwartz.    1939.   Acute  ethylene  dichloride



poisoning.  J. Ind. Hyg. Toxicol.   21:  126-131.

-------
Withey, J.R. and B.T. Collins.  1980.  Chlorinated aliphatic hydrocarbons used in



teh 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., B.T.  Collins  and  P.G.  Collins.   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.








Woodward, G., S. Lange,  K. Nelson and H. Calvery.  1981.  The acute oral toxicity



of acetic, chloroacetic, dichloroacetic and trichloroacetic acids.  J. Ind. Hyg.



Toxicol.  23: 78-82.








Yasoda, S.K. and E.  Dan  Loughran.  1977.  Air sampling methods  for S-tetrachloro-



ethane and other related chlorinated hydrocarbons.   J.  Chromat.  137-283-292.








Yllner,  S.   1971 a.   Metabolism of  chloroacetate-1-  C in  the  mouse.   Acta.



Pharmacol. Toxicol.   30: 69-80/
                                                      1 U
Yllner, S.  I971b.   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).
                                     10-55

-------
                                   APPENDIX A

                COMPARISON AMONG DIFFERENT EXTRAPOLATION MODELS


     Four models used for low-dose extrapolation, assuming the independent

background, are:


Multistage:                 P(d) = 1 - exp [~(q±d + ... + qkdk)]


where q^ are non-negative parameters;
Probit:                               A + Bln(d)
                            P(d) =    /  F(x) dx
where f(.) is the standard normal probability density function;


Weibull:                    P(d) = 1 - exp [-bdk]


where b and k are non-negative parameters; and


One-hit:                    P(d)  = 1 - exp [~bd]


where b is a non-negative parameter.

     The maximum likelihood estimates (MLE) of the parameters in the multi-

stage and one-hit models are calculated by means of the program GLOBAL82,

which was developed by Howe and Crump (1982).   The MLE estimates of the

parameters in the probit and Weibull models are calculated by means of the

program RISK81,  which was developed by Kovar and Krewski (1981).

     Table A-l presents the MLE of parameters  in each of the four models

that are applicable to a data set.
                                     A-l

-------
                    TABLE A-l.  MAXIMUM LIKELIHOOD ESTIMATES OF THE PARAMETERS  FOR  EACH  OF  THE FOUR
                                        EXTRAPOLATION MODELS BASED ON DIFFERENT DATA  BASES
Data base
Hemangiosarcomas in male
rats, dose with surface
correction
N) Hemangiosarcomas in male
rats, dose without
surface correction
He pa to cellular carcinomas
in male mice, dose with
surface correction
Heptocellular carcinomas in
male mice, dose without
Multistage
ql = 3.65 x
q2 = 0

qi = 7.03 x
q2 = 0

q^ = 1.03 x
(\2 ~ 1 «^7 x

q: = 8.18 x
q? = 9.25 x
Pro bit
io-2


10~"3


io-2
10~3

io-4
10~6
A =
B =

A =
B =

A =
B =

A =
B =
-1.43
0.34

-1.99
0.34

-2.75
0.86

-4.92
0.86
Weibull
b
k

b
k

b
k

b
k
= 9.08 x 10 2
= 0.52

= 3.84 x 10~2
= 0.52

= 8.07 x 10~~3
= 1.49

= 1.84 x 10~4
= 1.49
One-hit
b = 3.65 x 10~2


b = 7.03 x 10~3


b = 2.32 x 10"2


b = 3.04 x 10~3

surface corrections

-------
                                   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 l-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* = 6.9 x ID"2 mg/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 lifespan for control animals is also




less than 90 weeks (approximately 70% of the control animals died before 90




weeks).
                                       B-l

-------
            TABLE B-l.   TIME-TO-DEATH IN WEEKS  FROM  HEMANGIOSARCOMAS
                 IN MALE OSBORNE-MENDEL RATS  FED  EDC BY  GAVAGE
Control (vehicle and untreated):

     28,  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,  10.6,  106,  106,  106,  106,  108,  110,  110,  110,  110.

Low-dose  group:

     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:

     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,  7A(H),  74,  74,  76(H),
     76,  78(H),  83(H),  84,  89,  101.
aH indicates death from hemangiosarcoma.
                                 B-2

-------
                                   APPENDIX C




CALCULATION OF EDB CARCINOGENIC POTENCIES BY GAVAGE AND INHALATION ROUTES







     The data used for calculating the potency of EDB by gavage are presented in




Table C-l.  The data from the high-dose group are not used because the dose




pattern from the high-dose group was drastically modified during the experi-




ment, resulting in a time-weighted dose almost identical to that of the low-




dose group.  Using the life-table approach (Kaplan and Meier 1958), the




probability of cancer at week 49 (the end of the study) is 0.37 with the 95%




one-sided upper-confidence limit at 0.46.  Thus, the upper-bound estimate of




the slope in the one-hit model, P = 1 - exp (-bd), is:







               b = t-ln (l-P)]/d




                 = [-In (1-0.46J/5.01




                 = 0.12 mg/kg/day.







The lifetime cancer potency, adjusting for the less-than-lifetime study, is






              B - 0.12 x (90/49)3 = 0.74 mg/kg/day







where 90 weeks is the lifespan that is also used in the EDC calculation when




time-to-event data are used.




     Therefore, EDB is approximately 10 times more potent than EDC (i.e.,




0.74/0.069).
                                        C-l

-------
     The data used for calculating the potency of EDB by inhalation are




presented in Table C-2.  The carcinogenic potency of EDB by inhalation is




calculated on the basis of nasal cavity tumors in male rats from the NCI




inhalation study on EDB (NCI 1979), using the linearized multistage model.




The potency of EDB by inhalation is







                              6.77 x 10-5/(ug/m3).
                                        C-2

-------
            TABLE C-l.  TIME-TO-DEATH IN WEEKS FROM HEMANGIOSARCOMAS
                 IN MALE OSBORNE-MENDEL RATS FED EDB BY GAVAGE
                                   (NCI 1978)
Low-dose group (38 mg/kg/day)a

     31(H)b,  32,  34(H),  34(H), 35,  36,  37,  38(H),  39,  39,  40,  41,
     42, 43,  43,  43,  43,  43,  43,  44,  44,  44,  46, 46, 47,  47, 48(H),
     48, 48,  48,  48,  49(H),  49(H),  49(H), 49(H), 49(H),  49(H),  49,
     49, 49,  49,  49,  49,  49,  49,  49,  49,  49,  49, 49.
aThe animals were fed EDB 5 days per week for 47  weeks  (out  of  49  weeks  observa-
 tion period).  The human equivalent dose is  (38  mg/kg/day)  x (5/7)  x (47/49)  x
 (0.5/70)1/3 = 5.01 mg/kg/day.
bH indicates that the animals died from hemangiosarcomas.   It is assumed that
 all animals observed with tumors at week 49  died from  the  tumors.  This
 assumption seems reasonable, since the time  from exposure  to tumor  death was
 very short.
              TABLE C-2.   INCIDENCE OF  NASAL  CAVITY  TUMORS  IN MALE
                FISCHER 344 RATS  ADMINISTERED EDB  BY INHALATION
                                   (NCI 1979)
Human equivalent
  dose (ug/m^)a
                                Animal  dose
                                  (ppm)
Response
0
1.39 x
5.57 x
                                      0
                                     10
                                     40
 0/50 (0%)
 39/50 (78%)
 41/50 (82%)
aHuman equivalent  dose  = d  x (5/7)  x  (6/24)  =  0.178  x  d.
 to the unit of ug/m3  by using  1  ppm  =  7.83  x  10^  ug/nH.
                                                          The  dose  is  converted
                                 C-3

-------