United States
               Environmental Protection
               Agency
               Office of Health and
               Environmental Assessment
               Washington DC 20460
EPA-600/8-82-005B
December 1983
External Review Draft
               Research and Development
vvEPA
Health Assessment
Document for
Tetrachloroethylene
(Perch loroethylene)
  Review
  Draft
  (Do Not
  Cite or Quote)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.

-------
                                      EPA-600/8-82-005B
                                      Jan. 1984
                                      External Review Draft
Health  Assessment  Document
                    for
        Tetrachloroethylene
        (Perchloroethylene)
                    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
            Office of Research and Development
        Office of Health and Environmental Assessment
         Environmental Criteria and Assessment Office
            Research Triangle Park, NC 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.
003PE2/A                                                              12/7/83

-------
                             Docket No. BCAP-HA-78-3
                      DRAFT HEALTH ASSESSMENT DOCUMENT FOR
                    TETRAOfljOROETHYLENE (PERCHLOROETHYLENE)
                                (ORD-FRL-2548-2)

AGENCY:  Environmental Protection Agency

ACTION:  Revision Clarifying the Carcinogenicity Conclusions of the Draft Health
         Assessment Document for Tetrachloroethylene (Perchloroethylene), and
         Reopening of Public Comment Period.

SUMMARY:  The External Review Draft of the EPA document, Draft Health
Assessment Document for Tetrachloroethylene (Perchloroethylene),
EPA-600/8-82-005B dated December 1983 was announced on December 23, 1983
in the Federal Register as being available for public review and comment from
January 5, 1984 through March 5, 1984.  On February 3, errata for pages 1-4 and
9-45 were distributed to all parties who had requested a copy of the document
from EPA and inserted in all undistributed copies.

     Because of a need to further clarify the conclusion regarding the
carcinogenicity findings, the Agency is issuing a revised statement of these
conclusions..  This statement replaces the last four lines on page 1-3 and all
of page 1-4, and also replaces the Conclusions section 9.4.3 on page 9-45.  The
revised statement is as follows:

     Tetrachloroethylene has been demonstrated to induce malignant tumors of
the liver in both male and female mice of the B6C3F1 strain.  This constitutes
a signal that tetrachloroethylene might be a carcinogen for humans.
The technical adequacy and the strong nature of the positive response in
the 1977 NCI tetrachlorethylene bioassay study makes it likely that a repeat
bioassay would also be positive.  In fact, the recent National Toxicology
Program (NTP) study, currently under audit, showed similar positive results.
This bioassay study, if validated, would strengthen the evidence for carcinogen-
icity of tetrachloroethylene.

     It should be recognized that there is a substantial body of opinion in
the scientific community to the effect that the mouse liver overreacts to
chlorinated organic compounds in contrast to that of the rat, and that the
induction of liver cancer in the mouse represents only a promoting action for
spontaneous liver tumors which normally occur with substantial incidence.
Furthermore, this promoting action might be related to liver damage associated
only with high level exposures to chlorinated agents such as tetrachlorethylene.
However, the evidence is inconclusive either for this restrictive position on
the mouse liver carcinogenic response to chlorinated organics or to the position
that the mouse liver is as good as any other mammalian indicator of carcinogenicity
for these compounds.

-------
     According to a literal interpretation of the criteria of the International
Agency  for Research on Cancer  (IARC), the animal data supporting the carcinogenicity
of tetrachloroethylene might be classified as limited.  Also, since existing
human epidemiologic data for tetrachloroethylene are inconclusive, its overall
IARC ranking might be classsified as Group 3, meaning, according to IARC language,
that perchloroethylene cannot be classified as to its human carcinogenicity.

     It should be recognized that Group 3 covers a broad range of evidence:
from inadequate to almost sufficient animal data.  Because of the stength of the
mouse liver cancer response, tetrachloroethylene is at the upper end of this
range.  Hence, the classification of the carcinogenicity of tetrachloroethylene
under the IARC criteria for animal evidence could be limited or almost sufficient
depending on the nature of the bioassay evidence as it exists today and on the
differing current scientific views about the induction of liver tumors in mice
by chlorinated organic compounds.  Therefore, the overall IARC ranking of
tetrachloroethylene is Group 3 but close to Group 2B, i.e. the more conservative
scientific view would regard tetrachloroethylene as being close to a probable
human carcinogen, but there is considerable scientific sentiment for regarding
tetrachloroethylene as an agent that cannot be classified as to its carcinogen-
icity for humans.

     In consideration of the above action, the public comment period will be
reopened for 30 days beginning March 23, 1984 and ending April 23, 1984.  In
addition to this notice, copies of this revision will be forwarded to those who
have already received copies of the draft health assessment document and errata
from the ORD Publications Office,. Center for Environmental Research Information
(CERI), in Cincinnati.  During the 30-day public comment period, requesters may
obtain copies of these materials, as follows:

     Single copies will be available from ORD Publications - CERI-FRN, U.S.
Environmental Protection Agency, 26 West St. Clair Street, Cincinnati, Ohio
45268. Tel. (513) 684-7562.

     These documents also will be available for public inspection and copying
at the EPA library at Waterside Mall, 401 M Street, S.W., Washington, D.C. 20460.

     Comments on the revision should be submitted in writing by close of business
on April 23,  1984 to:  Project Officer for Tetrachloroethylene (Perchloroethylene),
Environmental Criteria and Asssessment Office (MD-52), U.S. Environmental
Protection Agency, Research Triangle Park, North Carolina 27711.

FOR FURTHER INFORMATION CONTACT:  Ms. Diane Chappell, 919/541-3637.
March 15, 1984                                       /s/
    Date                                      Bernard D. Goldstein
                                            Assistant Administrator
                                         for Research and Development

-------
 "twoi*5
              UNITED STATES ENVIRONMENTAL PROTECTION AGENCY

                            WASHINGTON, O.C. 20460
                              February 3, 1984
FROM:
TO:
                                                                    OFFICE OF
                                                             RESEARCH AND DEVELOPMENT
SUBJECT:  ERRATA
El IzabethTTfArj^erson
Director, Office of Health  and
  Environmental  Assessment  (RD-689)

Recipients of the Second External  Review Draft of the Health
Assessment Document for Tetrachloroethylene  (Perchloroethylene),
December 1983, EPA-600/8-82-005B
     Because of errors on two pages  1n  the Tetrachloroethylene document (pages
1-4 and 9-45), we are providing you  with  replacement pages for Insertion Into
the document.  The shaded areas on the  replacement pages Indicate the material
that has been revised.

     We apologize for any Inconvenience these  changes may have caused the
recipients.
Attachments-2

-------
              UNITED STATES ENVIRONMENTAL PROTECTION AGENCY

                            WASHINGTON, D.C. 20460
FPCM:
                                                                    OFFICE OF
                                                              RESEARCH AND DEVELOPMENT
SUBJECT:  Dra£t^fjear±*h Assessment Document for Tetrachloroethylene
                           rson
          Director, Office of Health and
             Environmental Assessment
TO:
Addressees
     The attached second external review draft of the Health Assessment
Document for Tetrachloroethylene is provided for your information.

     The draft document was made available for public review and comment on
January 5, 1984 (December 23, 1983, 48 FR 56847) and the Agency is accepting
public comments until March 5, 1984.  If you would like to cement on this
document, please address your conments to:

     Project Officer for Tetrachloroethylene
     Environmental Criteria and Assessment Office (MD-52)
     U.S. Environmental Protection Agency
     Research Triangle Park, North Caroline  27711

Conments should be in writing and should be submitted by close-of-business
March 5, 1984.

     A limited supply of copies will be available from the ORE publications
office in Cincinnati (FTS 684-7562).

     After receipt of all comments, the EPA Science Advisory Board will review
the subject dco.:rsent in ~i public meeting.  This meeting will be announced in a
subsequent Federal Register notice.

Attachment

-------
                                    PREFACE

     The Office of Health and Environmental Assessment has prepared this health
assessment to serve as a "source document" for EPA use.   Originally the health
assessment was developed for use by the Office of Air Quality Planning and
Standards to support decision-making regarding possible regulation of PCE
as a hazardous air pollutant.  However, at the request of the Agency's Work
Group on Solvents the assessment scope was 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 dose-response
relationships are discussed, where appropriate, so that the nature of the
adverse health responses is placed in perspective with observed environmental
levels.
003PE2/A                                                               12/1/83

-------
                               CONTENTS


                                                                        Page

1.   EXECUTIVE SUMMARY	    1-1

2.   INTRODUCTION	    2-1

3.   GENERAL BACKGROUND INFORMATION	    3-1
     3.1  PHYSICAL AND CHEMICAL PROPERTIES	    3-1
     3.2  PRODUCTION	    3-1
     3. 3  USE	    3-3
     3.4  EMISSIONS	    3-4
     3.5  ENVIRONMENTAL FATE AND TRANSPORT	    3-5
          3.5.1  Ambient Air	    3-5
          3.5.2  Water	    3-9
     3.6  LEVELS OF EXPOSURE	    3-10
          3.6.1  Mixing Ratios	    3-10
     3. 7  ANALYTICAL METHOD	    3-19
          3.7.1  Ambient Air	    3-19
          3.7.2  Water	    3-23
          3.7.3  Biological Media	    3-25
          3.7.4  Calibration	    3-25
          3.7.5  Storage and Stability of PCE	    3-25
     3.8  REFERENCES	    3-27

4.   ECOSYSTEM CONSIDERATIONS	    4-1
     4.1  EFFECTS ON AQUATIC ORGANISMS AND PLANTS	    4-1
          4.1.1  Effects on Freshwater Species	    4-1
          4.1.2  Effects, on Aquatic Plants	    4-2
          4.1.3  Effects on Saltwater Species	    4-2
     4. 2  BIOCONCENTRATION AND BIOACCUMULATION	    4-3
          4.2.1  Levels of PCE in Tissues of Aquatic Species	    4-4
     4.3  BEHAVIOR IN WATER AND SOIL	    4-13
     4.4  SUMMARY	    4-15
     4. 5  REFERENCES	    4-16

5.   COMPOUND DISTRIBUTION AND RELATED PHARMACOKINETICS	    5-1
     5.1  HUMAN AND ANIMAL STUDIES	    5-1
          5.1.1  Absorption	    5-1
          5.1.2  Distribution	    5-4
          5.1.3  Metabolism	    5-6
          5.1.4  Excretion and Elimination	    5-13
          5.1.5  Estimates of Biological Half-life	    5-24
          5.1.6  Interaction of PCE with Other Compounds	    5-24
          5.1. 7  Summary	    5-25
     5.2  REFERENCES	    5-28
                                      IV
003PE2/A                                                              12/1/83

-------
                             CONTENTS (continued)
6.    TOXIC EFFECTS	    6-1
     6.1  HUMANS	    6-1
          6.1.1  Effects on the Liver	    6-1
          6.1.2  Effects on Kidneys	    6-5
          6.1.3  Effects on Other Organs/Tissues	    6-5
          6.1.4  Behavioral and Neurological Effects	    6-6
     6.2  ANIMALS	    6-10
          6.2.1  Effects on the Nervous System	    6-10
          6.2.2  Effects on the Liver and Kidney	    6-19
          6.2.3  Effects on on the Heart	    6-24
          6.2.4  Effects on the Skin and Eye	    6-24
     6.3  ADVERSE EFFECTS OF SECONDARY POLLUTANTS	    6-24
     6.4  SUMMARY OF ADVERSE HEALTH EFFECTS AND ASSOCIATED LOWEST
            OBSERVABLE EFFECT CONCENTRATIONS	    6-24
           6.4.1  Inhalation Exposure	    6-24
           6.4.2  Oral Exposure	    6-26
           6.4.3  Dermal Exposure	    6-26
     6.5   REFERENCES	    6-27

7.    TERATOGENICITY, EMBRYOTOXICITY, AND REPRODUCTIVE EFFECTS	    7-1
     7.1  ANIMAL STUDIES	    7-2
          7.1.1  Mice	    7-2
          7.1.2  Rats	    7-3
          7.1. 3  Rabbits	    7-6
     7.2  SUMMARY	    7-7
     7. 3  REFERENCES	    7-8

8.    MUTAGEN IC ITY	8-1
     8.1  GENE MUTATION TESTS	    8-1
          8.1.1  Bacteria	    8-1
          8.1.2  Drosophila	    8-12
     8.2  CHROMOSOME ABERRATION TESTS	    8-13
          8.2.1  Whole-Mammal Bone Marrow Cells	    8-13
          8.2.2  Human Peripheral Lymphocytes	    8-14
          8.2.3  Drosophila	    8-15
     8. 3  OTHER TESTS INDICATIVE OF DNA DAMAGE	    8-15
          8. 3.1  DNA Repair	    8-15
          8.3.2  Mitotic Recombination	    8-18
     8.4  DNA BINDING STUDIES	    8-21
     8.5  STUDIES INDICATIVE OF MUTAGENICITY IN GERM CELLS	    8-21
     8. 6  MUTAGENICITY OF METABOLITES	    8-22
     8. 7  SUMMARY AND CONCLUSIONS	    8-24
     8. 8  REFERENCES	    8-26
003PE2/A
12/1/83

-------
                             CONTENTS (continued)
9.   CARCINOGENICITY	    9-1
     9.1  ANIMAL STUDIES	    9-1
          9.1.1  National  Cancer Institute Bioassay (1977a)	    9-1
          9.1.2  Dow Chemical Company Inhalation Study (Rampy et al.,
                  1978)	    9-10
          9.1.3  Intraperitoneal Administration Study (Theiss et al.
                  1977)	    9-12
          9.1.4  Skin Painting Study (Van Duuren et al.,  1979)	    9-13
     9.2  EPIDEMIOLOGIC STUDIES	    9-15
          9.2.1  Kaplan (1980)	    9-15
          9.2.2  Blair et al. (1979)	    9-18
          9.2.3  Katz and Jowett (1981)	    9-18
          9.2.4  Lin and Kessler (1981)	    9-20
          9.2.5  Asal (personal communication, 1983)	    9-20
     9.3  QUANTITATIVE ESTIMATION	    9-21
          9.3.1  Procedures for the Determination of Unit Risk	    9-23
          9.3.2  Unit Risk Estimates	    9-31
          9.3.3  Comparison of Potency with Other Compounds	    9-39
     9.4  SUMMARY AND CONCLUSIONS	    9-39
          9.4.1  Qualitative	    9-39
          9.4.2  Quantitative	    9-44
          9.4. 3  Conclusions	    9-45
     9. 5  REFERENCES	    9-46
          APPENDIX A.  Comparison Among Different Extrapolation
                       Models	    A-l
          APPENDIX B.  Time-To-Event Data and Calculations of
                       Potency	    B-l
                                       VI
003PE2/A                                                              12/1/83

-------
                                LIST OF TABLES

Table                                                                 Page

3-1  Major U. S.  producers of PCE	     3-3
3-2  Ambient air mixing ratios of PCE measured at sites around
      the world	     3-11

4-1  Levels of PCE in tissues of marine organisms,  birds,  and
      mamma Is	     4-5
4-2  Accumulation of PCE by dabs	     4-9
4-3  Concentration of PCE and trichloroethylene in mollusks and
      fish near the Isle of Man	     4-12

5-1  Estimated uptake of six individuals exposed to tetrachloro-
      ethylene while at rest and after rest/exercise	     5-3
5-2  Alcohol and diazepam effects upon tetrachloroethylene (PCE)
      in blood and breath concentrations, 5.5-hour exposures	     5-26

6-1  Summary of the effects of tetrachloroethylene on animals	     6-11
6-2  Toxic dose data	     6-17

8-1  Summary of mutagenicity testing of PCE	     8-2
8-2  Results of bacterial tests of different purities and
      sources of PCE	     8-3

9-1  Incidence of hepatocellular carcinomas in B6C3F1 mice fed
      PCE	     9-3
9-2  Cumulative survival of Sprague-Dawley rats exposed to PCE
      for 12 months	     9-12
9-3  Pulmonary tumor response to PCE	     9-13
9-4  Incidence rate of hepatocellular carcinomas in female
      B6C3F1 mice	     9-32
9-5  Upper-bound (point) estimates of risk at various dose levels
      based on data of hepatocellular carcinomas in female mice
      (NCI)	     9-36
9-6  Expired PCE unchanged as percentage of recovered radio-
      activity	     9-37
9-7  Relative carcinogenic potencies among 53 chemicals evaluated
      by the Carcinogen Assessment Group as suspect human
      carcinogens	     9-41

A-l  Maximum likelihood estimate of  the parameters for each of
      the four extrapolation models  based on  hepatocellular
      carcinomas in female mice	     A-2

B-l  Time-to-event data and calculations of potency	     B-l
                                     VI 1
 003PE2/A                                                               12/1/83

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

3-1  Locations of U.S.  PCE production facilities producing more
       than 100 mi 11 i on pounds	     3-4

4-1  Accumulation and loss of PCE by dabs	    4-10
4-2  Relation between flesh and liver concentrations of PCE
       in dabs	    4-11

5-1  Predicted post-exposure alveolar air concentrations of PCE
       at various times against duration of exposure	     5-5
5-2  Mean and range of breath concentrations of PCE after exposure
       of individuals to single or repeated exposures	    5-15
5-3  Mean and range of breath concentrations of five individuals
       during post-exposure after five separate exposures to 96,
       109, 104, 98 and 99 ppm	:	    5-16
5-4  PCE in blood and exhaled air following exposure to PCE for
       4 hours	    5-18
5-5  Trichloroacetic acid in blood following exposure to PCE
       for 4 hours	    5-21
5-6  Urinary excretion of trichloroacetic acid following exposure
       to PCE for 4 hours	    5-22

8-1  Dose response curves of Perchlor 200 and Perchlor 230 using
       Salmonella tester strains TA100 and TA1535	     8-8
8-2  Induction of mitotic recombination by PCE in Saccharomyces
       cerevisiae D7	    8-19

9-1  Growth curves for male and female mice in the PCE chronic
       study (NCI)	    9-5
9-2  Survival comparisons of male and female mice in the PCE
     chronic study (NCI)	    9-6
9-3  Growth curves for male and female rats in the PCE chronic
       study (NCI)	    9-7
9-4  Survival comparisons of male and female rats in the PCE
       chronic study (NCI)	    9-8
9-5  Histogram representing the frequency distribution of the
       potency indices of 53 suspect carcinogens evaluated
       by the Carcinogen Assessment Group	   9-40
                                     VI 1 1
003PE2/A                                                              12/1/83

-------
                             AUTHORS AND REVIEWERS

The principal authors of this document are:

Chao W.  Chen, Carcinogen Assessment Group, U.S.  Environmental Protection
     Agency, Washington, D.C.

I.W.F.  Davidson, Department of Physiology and Pharmacology, Bowman Gray
     School of Medicine, Winston-Salem, N.C.

Vicki Vaughan-Oellarco, Reproductive Effects Assessment Group, U.S.  Environ-
     mental Protection Agency, Washington, D.C.

Herman Gibb, Carcinogen Assessment Group, U.S. Environmental Protection Agency,
     Washington, D.C.

Mark Greenberg, Environmental Criteria and Assessment Office, U.S. Environmental
     Protection Agency, Research Triangle Park,  N.C.

Charalingayya B. Hiremath,  Carcinogen  Assessment  Group,  U.S.  Environmental
     Protection Agency, Washington, D.C.

Jean C.  Parker, Office of Solid Waste, U.S.  Environmental Protection Agency,
     Washington, D.C.
                                       IX
 003PE2/A                                                              12/1/83

-------
The following individuals were asked to review an early draft of this document
and submit comments:

Dr. Joseph Borzelleca
Dept.  of Pharmacology
The Medical College of Virginia
Virginia Commonwealth University
Richmond, VA  23298

Dr. Benjamin Van Duuren
Institute of Environmental Medicine
New York University Medical Center
New York, NY  10016

Dr. Herbert Cornish
Dept.  of Environmental and Industrial Health
University of Michigan
Ypsilanti, MI  48197

Dr. I. W. F.  Davidson
Dept.  of Physiology/Pharmacology
The Bowman Gray School of Medicine
Winston-Salem, NC  27103

Dr. Lawrence Fishbein
National Center for Toxicological Research
Jefferson, AR  72079

Dr. John G. Keller
P. 0.  Box 10763
Research Triangle Park, NC  27709

Dr. John L. Laseter
Director, Environmental Affairs, Inc.
New Orleans,  LA  70122

Al1 Members of the
Interagency Regulatory Liaison Group
Subcommittee on Organic Solvents
003PE2/A                                                              12/1/83

-------
Participating Members of The Carcinogen Assessment Group
Roy E. 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.
Bernard H. Haberman, D.V.M., M.S.
Charalingayya B. Hiremath, Ph.D.
Robert E. McGaughy, Ph.D.
Dharm W. Singh, D.V.M., Ph.D.
Todd W. Thorslund, Sc.D.

Participating Members of the Reproductive Effects Assessment Group

Peter E. Voytek, Ph.D.  (Director)
John R. Fowle III, Ph.D.
Carol Sakai, Ph.D.
Ernest Jackson, M.D.
K.S. Lavappa, Ph.D.
Sheila Rosenthal, Ph.D.
Vicki Vaughan-Dellarco, Ph.D.
Members of the Agency Work Group on Solvents
Elizabeth L. Anderson
Charles H. Ris
Jean Parker
Mark Greenberg
Cynthia Sonich
Steve Lutkenhoff
James A. Stewart
Paul Price
William Lappenbush
Hugh Spitzer
David R. Patrick
Lois Jacob
Arnold Edelman
Josephine Brecher
Mike Ruggiero
Jan Jablonski
Charles Delos
Richard Johnson
Priscilla Holtzclaw
Office of Health and Environmental Assessment
Office of Health and Environmental Assessment
Office of Health and Environmental Assessment
Office of Health and Environmental Assessment
Office of Health and Environmental Assessment
Office of Health and Environmental Assessment
Office of Toxic Substances
Office of Toxic Substances
Office of Drinking Water
Consumer Product Safety Commission
Office of Air Quality Planning and Standards
Office of General Enforcement
Office of Toxic Integration
Office of Water Regulations and Standards
Office of Water Regulations and Standards
Office of Solid Waste
Office Water Regulations and Standards
Office of Pesticide Programs
Office of Emergency and Remedial Response
 003PE2/A
                                       XI
                                    12/7/83

-------
The following individuals attended a review workshop to discuss draft EPA
documents on organic compounds which included an early draft of this
document:

Dr. Mildred Christian
Argus Laboratories
Perkasie, PA  18944

Dr. Rudolf Jaeger
Institute of Environmental Medicine
New York, NY  10016

Dr. Benjamin Van Duuren
Institute of Environmental Medicine
New York University Medical Center
New York, NY  10016

Dr. Herbert Cornish
School of Public Health
University of Michigan
Ann Arbor, MI  48197

Dr. I. W. F. Davidson
Dept.  of Physiology/Pharmacology
The Bowman Gray School of Medicine
Winston-Sal em, NC'  27103

Dr. John Egle
Dept.  of Pharmacology
Virginia Commonwealth University
Richmond, VA  23298

Dr. John G.  Keller
P. 0.  Box 10763
Research Triangle Park, NC  27709

Dr. Norman Trieff
Dept.  of Preventive Medicine
University of Texas Medical Branch
Galveston, TX  77550

Dr. Thomas Haley
National Center for Toxicology Research
Jefferson, AK  72079

Dr. James Withey
Food Directorate
Bureau of Food Chemistry
Ottawa, Canada
                                       xi i
003PE2/A                                           .                   12/1/83

-------
                             1.   EXECUTIVE SUMMARY

     Tetrachloroethylene (PCE) is a moderately volatile chlorinated hydrocarbon
which has important commercial applications in the dry cleaning of fabrics and
in the degreasing of fabricated metal parts.   It is estimated that approximately
300,000 metric tons were produced in the United States in 1980.   Approximately
90 percent of  production is estimated to  be released eventually to the atmos-
phere.  Because  PCE  is  relatively insoluble in water  (150  mg/1)  and has a
vapor pressure of  0.19  pascals at 20°C,  PCE  in natural waters would be con-
veyed to  the  atmosphere rapidly,  through evaporation.   There are no known or
expected natural  sources of emissions.
     PCE has been detected in the ambient (natural environment) air of a variety
of urban  and  nonurban  areas  of the  United  States  and other regions of the
world.  Levels can range from trace amounts in rural areas to as much as 10 parts
per billion (ppb)  or 0.068 mg/m3 in some  large  urban centers.   The global
average background  level is estimated at  about 25  parts per  trillion (ppt) or
0.175 x 10-3 mg/m3.  It is detected less frequently in water, because it is not
appreciably soluble; but  PCE  has been monitored in some surface and drinking
waters, generally  at levels of  1  ppb or  less.  In  certain instances  involving
contamination of groundwater,  much higher levels have been reported.   Although
there is  very  limited  information on the behavior of PCE in soil, PCE can be
expected to leach through soils of low (< 0.1 percent) organic carbon content.
The amount of PCE adsorbed to soils is dependent on the partition coefficient,
the organic carbon  content, and the  concentration  of  PCE  in  the liquid phase.
     In the troposphere,  a region of the  atmosphere  extending  to between 8
and 16  kilometers  above the earth's  surface,  PCE  undergoes  photochemical  deg-
radation  to the  extent  that   its  estimated  lifetime  is  appreciably less than
1 year.   Little  PCE is  expected  to  be conveyed to the  stratosphere.  Recent
studies have  shown  that,  in   real atmospheres, neither  atomic  chlorine-  nor
hydroxy radical-induced photooxidation  of PCE generates substantial concen-
trations  of ozone  or other oxidants; thus,  PCE is  not  believed  to  be a signi-
ficant factor in production of photochemically-induced pollution often experi-
enced near  large urban  centers.  Because of the reduced solar flux in winter
and seasonal variations in hydroxy radical concentration, PCE levels in ambient
air are expected to  be  higher in winter than in summer.  On a daily basis, PCE
levels fluctuate considerably.
003PE4/F                             1-1                              11/22/83

-------
     Inhalation is the principal  route by which PCE enters the body.   Ingestion
of drinking water contaminated by PCE is a secondary source of exposure.   During
inhalation, PCE  is absorbed by the blood and distributed throughout the body.
Controlled studies with human volunteers (at 100 ppm) have shown that pulmonary
steady-state conditions are reached  between 3  and 4  hours.  Physical activity
increases uptake.  At  levels  approaching those found or expected in ambient
air, the time  required to reach steady-state  conditions may be considerably
longer.   Once  body equilibrium  or  steady-state has  been reached, no further
uptake is possible.   There  is evidence that PCE will  partition selectively
into  lipid-rich  tissues with chronic  or  long-term  exposure  to even  low
ambient air concentrations  until  steady-state is attained.  Because  of  its
lipophilic nature, PCE  is expected to cross membrane  barriers  in  the body.
Most  inhaled  PCE is  excreted via  the lungs in unchanged  form.  Both con-
trolled and  occupational  exposures  of  humans indicate that  the  principal
urinary excretion  product of PCE metabolism is trichloroacetic  acid.   Con-
trolled studies  with  humans have demonstrated that  PCE metabolism  (urinary
trichloroacetic acid)  represents 2  percent or less  of the amount of PCE ab-
sorbed.   Studies in rats and mice suggest that metabolism  of PCE is  a saturable
process.   In humans,  saturation  would not be  expected until exposure levels
approximate 100 ppm (678 mg/m3).
     Toxicity testing in experimental animals,  coupled with limited  human data
derived principally from overexposure situations,  suggests that long-term expo-
sure of humans to environmental  levels of PCE is not likely to present a  serious
health concern.
     Decrements in task performance and coordination are the first gross  signs
of central  nervous system (CNS)  depression and behavioral  alterations observed
in controlled human studies in which individuals were exposed to about 100 ppm
(678 mg/m3) for  up to 7 hours.   More sensitive tests, however, would have to
be performed to  determine if PCE affects  the  nervous  system  at even lower
concentrations.
     Evidence  in  rodent  species  suggests that PCE has the potential to cause
liver damage with acute or prolonged exposure at levels that,  in humans,  would
cause only slight  CNS depression.   However, there are  insufficient data to
estimate the lowest  levels  of PCE that  are  associated with adverse effects
upon the liver in humans.
     The lowest  observed-adverse-effect  level  (LOAEL), based on CNS dysfunc-
tion, is about 100 ppm (678 mg/m3).  However, the LOAEL may not be sufficiently

003PE4/F                             1-2                              11/22/83

-------
protective of human  health  when  one considers that intermittent or prolonged
exposure of animals  to  PCE  has been observed  to  result in liver and kidney
damage at  levels  exceeding  200 ppm (1,356 mg/m3).   It  should be noted that
liver damage  in humans  is  generally associated  with  short-term exposures
greatly in excess of  100 ppm (678 mg/m3).  The LOAEL is defined  as the lowest
exposure level  in a  study  or group of  studies which produces statistically
significant increases in frequency  or severity of  adverse  effects  between  the
exposed population and its  appropriate control.
     The mammalian animal tests performed to date do not indicate any signifi-
cant teratogenic potential  of PCE in the species  tested.  On this basis,  there
is no evidence  to suggest  that the conceptus  is  uniquely  susceptible  to the
effects of PCE.   The anatomical  effects observed reflect delayed development
and can be considered reversible.   It is  important to note,  however, that  the
reversible nature of an embryonic/fetal  effect in one species might, in another
species,  be  manifested  in  a more  serious  and irreversible manner.  At the
current time, the teratogenic  potential of PCE for humans must be considered
unknown.
     Tetrachloroethylene has  been  evaluated  for  its ability to cause gene
mutation,  chromosomal  aberrations, unscheduled  DMA synthesis,  and  mitotic
recombination.  These tests were  conducted  using  bacteria,  Drosophila, yeast,
cultured mammalian cells,  whole  mammal  systems,   and cytogenetic analyses  of
exposed  humans.   Certain technical  and commercial samples of  PCE  elicited
increased  responses  in  the  Ames  bacterial  test,  a yeast mitotic recombination
assay, a host-mediated assay using  Salmonella, and DNA  repair tests.  Exogenous
metabolic  activation was not required for detection of  these  increased effects.
In general,  the responses  were weak and observed at high concentrations that
were cytotoxic; dose-dependent relationships were  not established.  The positive
findings may be the  result of mutagenic contaminants and/or added  stabilizers.
There have been several other tests of  commercial and technical  samples of PCE
which have been reported as negative.   The epoxide of PCE, which is thought to
be the  active biological intermediate,  was found  to be positive in bacterial,
studies.
     Tetrachloroethylene has been demonstrated to  induce malignant  tumors of
the liver  in  both male and female mice  of the B6C3F1 strain.  This constitutes
limited  evidence  that PCE  may  be carcinogenic in humans.   It should be recog-
nized  that a substantial part of  the scientific community believes that  the
 003PE4/F                              1-3                               11/22/83

-------
mouse liver overreacts  to  chlorinated organic compounds,  in contrast to that
of the  rat, and  that the induction of  liver  cancer in the mouse represents
only a promoting action for spontaneous liver tumors which normally occur with
substantial incidence.
     According to  the  criteria  of the International Agency  for  Research on
Cancer  (IARC), the data  supporting the carcinogenicity of  PCE must be classi-
fied as  "limited."  Because  existing  human epidemiological  data  for  PCE is
inconclusive,  its overall IARC ranking should be Group 3,  corresponding to the
conservative  scientific  view that PCE  is  probably carcinogenic in humans.
003PE4/F                             1-4                              11/22/83

-------
 Replacement Page 1-4 for EPA-600/8-82-005B, "Health Assessment  Document
                      for Tetrachloroethylene (Perchloroethylene)
 mouse liver overreacts to chlorinated organic compounds, in contrast to that
 of the rat, and that the Induction of liver  cancer in the mouse represents
 only a promoting action for spontaneous  liver tumors which normally occur with
 substantial Incidence.
      According to the criteria of the International  Agency for Research on
 Cancer (IARC), the data supporting the carcinogenlcity of PCE must be classi-
 fied as "limited."  Because existing human epidemiological data for PCE is
                                                           ::^^^^^x•^:^•^xw.^x.^x•^x««v:::^::
 inconclusive, its overall IARC ranking should be  Group 3,;!meaning that there :•$•:
*&<<<<<<'X<<
-------
                               2.   INTRODUCTION

     Tetrachloroethylene (PCE)  is  one member  of a  family of unsaturated
chlorinated aliphatic compounds.  Other  common names/acronyms are perchloro-
ethylene, Perk,  PER,  and  PERC.   Its synonyms  include  carbon dichloride,
tetrachloroethene, and 1,1,2,2-tetrachloroethylene.
     Tetrachloroethylene, though a water and solid waste contaminant, is pri-
marily of interest  in  ambient air exposure situations.  It is released into
ambient  air  as a result of  evaporative  losses during production, storage,
and/or use.    It  is  not known to be  generated  from  natural sources.   It has
negligible photochemical  reactivity  in  the  troposphere and  is  removed by
scavenging mechanisms, principally via hydroxyl radicals.
     The scientific data base  is limited with  reference to the effects  of  PCE
on humans.   Effects on  humans have generally  been  ascertained  from  studies
involving individuals  occupationally or accidentally  exposed.   During  such
exposures, the concentrations  associated with  adverse effects on  human  health
were either  unknown or  far in excess of concentrations found or  expected  in
ambient air.   Controlled PCE exposure studies have been directed toward eluci-
dating the effects on the central nervous system, effects  on clinical chemistries,
and pharmacokinetic parameters of PCE exposure.
     Since epidemiological studies have not been able to assess adequately the
overall  impact of PCE on human health, it has been necessary to rely greatly
on animal studies to derive indications of potential harmful effects.  Although
animal data  cannot  always be extrapolated to humans, indications of probable
or likely effects among animal species increase confidence that similar effects
may occur in  humans.
     This document is intended to provide an evaluation of the scientific  data
base concerning  PCE.   It is believed that the literature  has been comprehen-
sively reviewed and critically evaluated through September 1983.  The publica-
tions  cited  in this document  represent  a  majority  of the  known  scientific
references to PCE.   Reports which had little  or no bearing upon the issues
discussed were not cited.
 003PE1/B                              2-1                             11/22/83

-------
                      3.    GENERAL BACKGROUND INFORMATION

3.1  PHYSICAL AND CHEMICAL PROPERTIES
     Tetrachloroethylene,  also called PCE (1,1,2,2-tetrachloroethylene or per-
chloroethylene),  is a colorless, heavy liquid with a chloroform-like odor. It
is used as a solvent for organic substances and is commercially important as  a
solvent in the dry  cleaning  of fabrics and  in the degreasing of metals.  It
has a molecular weight of 165.85 and is relatively insoluble in water (150 mg/1)
(Handbook of Chemistry and Physics, 1976; Chemical and Process Technology Ency-
clopedia, 1974).   Its CAS registry number is 127184.   In air, at standard tem-
perature and pressure,  1  part per million (ppm)  is equivalent to 6.78 mg/m3.
     Tetrachloroethylene has  negligible photochemical reactivity (Dimitriades
et al., 1983) and, in the troposphere, is decomposed via free radical mechanisms.
When in contact with water for prolonged periods, PCE slowly decomposes to yield
trichloroacetic and  hydrochloric  acids.   Upon prolonged storage in light, it
was reported to  decompose  slowly to trichloroacetyl  chloride and phosgene by
autooxidation (Hardie,  1966).   At 700°C, it decomposes, when in contact with
activated charcoal, to hexachloroethane and hexachlorobenzene (Gonikberg, 1956).
Tetrachloroethylene has a boiling point of 121.1°C at 760 mm Hg and a vapor pres-
sure of 14 torr at 20°C.  MacKay et al. (1982) have calculated a vapor pressure
of 19 torr at 25°C.
     The chemical  reactivity  of PCE  has  been discussed  by  Bonse  and  Henschler
(1976)  in  terms  of  the  electron-inductive  effect of  the chlorine atoms,  which
reduce  electron  density about the ethylene bond.   This  effect,  in  combination
with a  steric  protective  effect afforded by the chlorine atoms, provides in-
creased  stability  against  electrophilic  attack.   This  is  exemplified  in the
reaction of PCE with ozone.   Compared to ethylene and less-substituted chlorina-
tion hydrocarbons,  PCE  has a  low rate of reaction (Williamson and  Cvetanovik,
1968).

3.2  PRODUCTION
     Tetrachloroethylene may  be produced by  several processes:
     1.   Chlorination  of  trichloroethylene:

               CHC1 = CC12 +  C12 |j^ CHCI2 CC13
 003PE4/B                             3-1                               11/22/83

-------
          2CHC12CC13 + Ca(OH)2 2iLi C12C = CC12 + CaCl2 + 2H20

     2.   Dehydrochlorination of S-tetrachloroethane:
          CHC12-CHC12 + C12 	> CC12 = CC12 + 2HC1

     3.   Oxygenation of S-tetrachloroethane:
          2CHClaCHCl2 + 02 	> 2CC12 = CC12 + 2H20

     4.   Chlorination of acetylene:
                            ?nn°r
          CC12 = CC12 + C12 -^-^ CC13-CC13
          ru - ru * o rn rn  20Q-4000C 4CC12 = CC12 + 2HC1
          LH _ LH + J LLI3LLI3 catalyst

     5.   Chlorination of hydrocarbons:
          C3H8 + 8 C12 	»  CC12 = CC12 + CC14 + 8HC1
          (propane)
               2CC14 	»  CC12 = CC12 + 2C12

     6.   Oxychlorination of 1,2-dichloroethane:
          2C2H4C12 + 5C12 	> C2H2C14 + C2HC15 + 5HC1
          C2H2C14 + C2HC15 	» C2HC13 + 2HC1 + CC12 = CC12
          7HC1 + 1.75 02 	» 3.5 H20 + 3.5 C12
          2C2H4C12 + 1.5 C12 + 1.75 02 	»  C2HC13 + CC12 = CC12 + 3.5 H20

     The majority  of  PCE producted in the United  States  is  derived  from the
oxychlorination  of 1,2-dichloroethane or  via Chlorination  of  hydrocarbons
(Lowenheim and Moran, 1975).
     In 1980,  347,859  metric tons of  PCE were  produced (U.S.  International
Trade  Commission,  1980).   The major producers and  production  capacities are
shown  in Table 3-1.  Locations of U.S. production facilities are shown in Fig-
ure 3-1.
     Imports of PCE may be sizeable, although they are partially offset by ex-
ports.   Most  of  the PCE imported is  produced in Belgium,  Italy,  France, and
Canada (Chemical Marketing Reporter, 1979a).


003PE4/B                             3-2              '                11/22/83

-------
                        TABLE  3-1.   MAJOR  U.S.  PRODUCERS  OF  PCE3
     Organization                                 Yearly  capacity,  MTa
     Dow Chemical                                         320
     PPG                                                  200
     Vulcan                                               200
     Diamond Shamrock                                     165
     Ethyl  Corporation
     E.  I.  du Pont de Nemours                             160

a                      ^
 Adapted from Chemical Economics Handbook (SRI,  1982).   MT = Metric tons.
 Recently announced withdrawal  from production (Halogenated Solvents
 Industry Alliance, 1983)
C0utput for captive use only.

3.3  USE
     Tetrachloroethylene has the following uses  (Gosselin et al., 1976; Fishbein,
1977):   (1) dry cleaning solvent; (2) textile scouring solvent; (3) dried vege-
table fumigant; (4) rug and upholstery cleaner;  (5) stain, spot, lipstick, and
rust remover; (6) paint remover; (7) printing ink ingredient; (8) heat transfer
media ingredient; (9) chemical  intermediate in the production of other organic
compounds;  and (10) metal degreaser.
     Testimony provided by the International Trade Commission to the Secretary
of the  Treasury  in  March  1979  stated  that  dry cleaning consumes  approximately
75 percent  of  U.S.  PCE production  and  imports  (Chemical  Marketing  Reporter,
1979a).  About 72  percent of commercial dry cleaning plants are estimated to
use PCE (U.S. EPA, 1979).

3.4  EMISSIONS
     Emissions of  PCE arise  during  its  production,  from  its  use  as  a chemical
intermediate in  industrial processes, from storage containers, during disposal,
and  from its use as  a solvent.   Because  emissions  are almost  exclusively to
the  atmosphere,  the  information presented  in this  section focuses on  air.
Few  data are available concerning  discharges to  water.   The  section dealing

003PE4/B                             3-3                              11/22/83

-------
 100 mllliv»n  pounds
   Figure 3-1.   Locations of U.S. PCE production facilities producing more
                than 100 million pounds.

                Source:  Fuller, 1976.
003PE4/B
3-4
11/22/83

-------
with monitoring  indicates that discharges to water probably occur.  Emissions
estimates reflect a diversity of sources throughout the country.   Dry cleaning
operations are  located primarily in urban areas.  Approximately 26,000 estab-
lishments are estimated to exist,  according to Bureau of Census data (U.S.  EPA,
1979).
     In 1977, global emissions were estimated at 570,000 ± 285,000 metric tons
(Singh  et  al.,  1979).   It was also estimated  that emissions accounted  for
approximately 90  percent  of  the amount of PCE produced (300,000 metric tons)
in the United States.

3.5  ENVIRONMENTAL FATE AND TRANSPORT
     The potential  for  ambient air and water  mixing  ratios of PCE  to  pose a
hazard  to  human health  is  influenced  by many processes.   Such  factors  include
transformation  into  secondary  pollutants  of concern and degradation rates in
air and water.

3.5.1 Ambient Air
3.5.1.1  Tropospheric Reactivity-- Reaction  with  the  hydroxyl radical  (-OH) is
the principal process by which many organic compounds, including PCE, are scav-
enged from the  troposphere.  Hydroxyl  radicals are produced when 0$ is irradi-
ated, resulting in excited atomic  oxygen, which  then  reacts with  water vapor.
The tropospheric  lifetime  of a compound  is  related  to the -OH mixing ratio
according to  the  expression:
                                          _
where  k  is the rate constant of reaction.
     Singh et  al.  (1979, 1981) calculated a tropospheric residence of PCE of
about  68 days.  This calculation was based on an average 24-hour -OH abundance
of  10^ molecules  cm •*  in the  boundary  layer  of  a polluted  atmosphere.  Justi-
fication for this  -OH mixing ratio stems from the field studies of Calvert (1976)
and  from Singh and coworkers  (1979a).  Because this  -OH mixing ratio is more
typical  of  summer months, Singh et al.  (1981)  suggested that a seasonally  ad-
justed mixing  ratio would result  in a  longer  chemical  residence  time.   If a
seasonally-averaged  -OH  mixing  ratio of 4 x  105 molecules cm  3  (a level  supported


003PE4/B                            3-5                              11/22/83

-------
by the field measurements of Campbell et al. ,  1979) and a weighted global  aver-
age temperature  (265°K), an average  residence time of  PCE would be calculated
to be 292 days or 0.8 years (Singh et al.,  1979).
     Estimations of  a  residence  time of PCE of one  year or less have been
reported by  a  number of  investigators  (Altshuller, 1980; Singh et al., 1978a;
Singh, 1977; Crutzen et  al.,  1978;  Lillian et al.,  1975; Yung et al., 1975;
Pearson and McConnell,  1975).
     Dimitriades et  al.  (1983)  calculated  a very low tropospheric reactivity
for PCE based on observations that ambient  levels  of PCE are constant.   Atmos-
pheric consumption of PCE is 0.02 percent per daylight hour.
     Higher  levels of  -OH have been  reported for the southern hemisphere com-
pared to those in the northern hemisphere (Singh,  1978).  This gradient probably
is due to the fact that carbon monoxide levels are much higher in the northern
hemisphere,  thus reducing  -OH  levels (Singh,  1978).   Measurements of PCE and
other reactive halocarbons  indicate  that mixing ratios  are  higher in the nor-
thern hemisphere where the  -OH  mixing ratio is low and where most of the PCE
is released  (Singh et al.,  1978b).
     Chamber studies indicate that PCE, on  irradiation in the presence of other
atmospheric  constituents, can be  transformed  into secondary products.  This
area of  study  has  been recently reviewed and further explored by Dimitriades
et al.  (1983).   These investigators have confirmed that PCE, under smog chamber
conditions,  reacts to  form  Og and  ozone precursors by  means  of a Cl-initiated
photooxidation mechanism.  However, such photooxidation is not expected to occur
in the real  atmosphere at  a rate  high enough for  substantial 03 production.
It is the  authors'  contention  that Cl atoms are effectively scavenged by the
hydrocarbons normally  present in  the  atmosphere; thus,  PCE  was judged to con-
tribute  less to  03 production than equal concentrations  of  ethane.   Ethane is
regarded by  the authors to be a boundary species separating the reactive vola-
tile organics from the unreactive ones.
     Studies on  PCE  reactions  with Og, 0,  and  «OH have indicated that rate
constants are lower than with Cl  (Dimitriades et al., 1983).
     Gay et  al.  (1976) had determined  that  trichloroacetyl chloride was a
photooxidation product of  PCE  in smog chamber studies.   Dimitriades et  al.
(1983)  found that,  on  irradiation,  the  only  product observed was CO.
Phosgene was not detected.  When  2 ppm (13.6 mg/nr*)  PCE  and  20 ppb trichloro-
acetyl chloride  were irradiated together,  the phosgene  level reached 0.1 ppm.

003PE4/B                             3-6                              11/22/83

-------
     Phosgene production from the photochemical oxidation of PCE in the presence
of other  substances  has been reported by others  (Lillian et al., 1975; Gay  et
al., 1976).  The  extent to which phosgene may be formed in real atmospheres,
based on smog chamber results,  would also be expected to be minimal.
3.5.1.2   Tropospheric Removal  Mechanisms  for  PCE--The reaction  sequence by
which PCE may be scavenged from the troposphere is as follows (Graedel , 1978):

        C2C14 + HO- - >   HOC(C1)2C(C1)2-
        HOC(C1)2C(C1)2- + Og - >  HOC(C1)2C(C1)202-
        HOC(C1)2C(C1)202- - oxygen - »  HOC(C1)2C(C1)20-
                           abstraction
        HOC(C1)2C(C1)20- - >  HOC(C1)2- + COClj,
        HOC(C1)2- + 02 - »  COC12 + H02-
     Howard (1976) suggested that the reaction path for the atmospheric oxida
tion of  PCE  may follow the scheme below, leading to the production of oxalyl
chloride:

          C2C14 +  -OH - » C2C12OH-


                      - > CC12CC10H- + Cl-                          (3-3)


          CCl2CCljjOH- + 02 - »• 02CC12CC12OH


          02CC12CC12OH + NO - » COC1CC12OH- + N02 + Cl •


          •OH + COC1CC12OH - > COC1COC1 + H20 + Cl •
Compared  to  other ethylene compounds studies,  Howard (1976) reported that PCE
exhibits  unusually  low reactivity toward  hydroxyl  radicals.


003PE4/B                              3-7                               11/22/83

-------
     Snelson et  al.  (1978) suggested that trichloroacetyl chloride and phos-
gene would  hydrolyze to  the  corresponding  chloroacetic acids and hydrogen
chloride  via  homogeneous gas phase  hydrolysis.   The  acids then would pre-
sumably be washed out of the atmosphere.
     The environmental significance of the production of phosgene from PCE has
been discussed by  Singh  and coworkers (Singh et al.,  1975; Singh,  1976).   As
PCE emissions are  likely to be higher in urban areas, the reactivity of this
halocarbon may result in concentrations of phosgene in the low ppb range during
adverse meteorological conditions  in and around urban centers (Singh, 1976).
It should be  noted that overt adverse health damage would not be expected at
these phosgene levels.  Considering the smog chamber results of Dimitriades et
al. (1983), in which PCE was found to have negligible reactivity,  it appears
unlikely  that phosgene  would be produced at  other than trace levels.  The
results of Singh and coworkers  indicate  that  negligible tropospheric  loss  via
gas phase hydrolysis would be expected, because phosgene is very stable in the
gas phase. The two  important  sinks cited were heterogeneous decomposition  and
slow liquid phase hydrolysis.   Singh (1976) concluded that phosgene is removed
slowly from the  atmosphere.   Rainfall  appears to lower atmospheric levels of
phosgene (Singh et al.,  1977a).   On the other hand, phosgene has been reported
to hydrolyze to  C02  and  HC1 rapidly  in liquid water.  Manogue  (1958)  reported
a half-life of 0.1 second at 25°C.   Thus, it is not expected to persist in the
troposphere because of rainout and hydrolysis in aqueous aerosols.
     The observed diurnal variations in PCE levels suggest that PCE has higher
mixing ratios  in the morning and evening hours than at other times (Lillian et
al.,  1975; Singh et al.,  1977a).  Ohta et al.  (1977) reported that mixing ratios
tended to be highest on cloudy days and lowest on rainy days.
     Solar flux is a major factor in the rate at which PCE is removed from the
atmosphere.   Singh et al. (1977a) suggested that the reduced solar flux in winter
months would permit a much longer transport of PCE because of reduced reactivity.
The effect of  solar flux was calculated by Altshuller (1980) who estimated that
a 1 percent consumption  of  PCE  by  reaction with  -OH would  take 14 days during
the month of January as opposed to one day in July.
     While the studies of Gay et al.  (1976) indicate that trichloroacetyl  chlo-
ride may be formed through chlorine atom migration in an epoxide intermediate,
evaluation of  -OH and oxygen atom rate constants indicate that less than 1 per-
cent of PCE in ambient air reacts with atomic oxygen and, of the activated epox-
ides formed, only  a small percentage undergo rearrangement (Graedel,  1978).

003PE4/B               .              3-8                              11/22/83

-------
3.5.2  Water
     Jensen and Rosenberg (1975) investigated the degradability of PCE (~ 0.1
to 1.0 ppm)  in  both open and closed systems,  with sea water and fresh water.
In open aquaria, with 20 1  of sea water,  PCE levels decreased 50 percent  within
200 hours (daylight).  In a closed system,  levels decreased approximately 25 per-
cent over the same  interval  (daylight).   It was reported that PCE levels in
boiled, deionized water in a closed system  did not exhibit any significant de-
crease after 8  days.   Analysis  was by headspace collection, followed by gas
chromatography-electron capture detection  (GC-ECD)  quantification.   Accuracy
and  limits  of detection were not reported.   Variation  in detector sensitivity
was checked daily by an injection of PCE in hexane.
     Dilling et al.  (1975) reported that PCE in water slowly decomposes to form
trichloroacetic and  hydrochloric  acids.  The  evaporation rate was determined
by dissolving 1 ppm (w/w)  PCE in  200 ml of water.   Solution temperature was
approximately 25°C.   The solution was stirred magnetically in a sealed system.
Quantification  was  made  by  mass  spectroscopy.   The  evaporation rate  of  PCE,
determined  from measurements  over  a 2-week period, was characterized by  a 50
percent  decrease  in the  initial  mixing ratio in 24 to 28 minutes.   The stir-
ring speed  had  a  marked  effect on  the evaporation rate.  With  no stirring ex-
cept for 15 seconds every 5 minutes, the time required for 50 percent depletion
ranged from 72  to 90 minutes.   The evaporative half-life was 27 ± 3 minutes.
Addition of dry,  granular  bentonite clay (500  ppm)  appeared to increase the
rate of  disappearance by 33 percent at 20 minutes.   However, when the clay was
allowed  to  remain in contact with purified water for several days and then added
to the solution,  there was no change in the  rate compared  with control.  In
closed system investigations, Dilling et al. (1975) used dry, powdered dolomi-
tic  limestone,  bentonite,  and  peat moss to determine the adsorption rate for
PCE.  With  500  ppm  bentonite, there was a 22 percent absorption after 30 minutes.
There was no further absorption.   Addition of limestone resulted in a 50 percent
depletion in 20 ± 2 minutes.  Addition of silica sand had no effect on the dis-
appearance  rate.  When 500  ppm peat moss was  added,  up to  0.4  ppm PCE was ab-
sorbed after 10 minutes.  At longer times,  no further absorption was observed.
It was concluded  that evaporation probably is the major pathway by which PCE
i s lost  from water.
     In  reactivity  studies,  Dilling et  al.  (1975)  found that  sunlight had the
greatest effect on  the rate of PCE disappearance.   The PCE level  in  water was

003PE4/B                            3-9                              11/22/83

-------
6 |jM.  Over a  12-month period, in which PCE solutions were stored  in the dark
as well as in the light,  PCE levels decreased from 1 ppm (0 time),  to 0.63 ppm
(6 months), to 0.35 ppm (12 months) in samples stored in the dark.   In the light-
exposed solution, the level decreased to 0.52 ppm (6 months), and 0.24 ppm (12
months).
     Schwarzenbach et al.  (1979), in measurements of PCE levels in Lake Zurich,
reported findings compatible with those of Dilling et al.  (1975) in that evapora-
tion is the dominant elimination process from surface waters. The annual release
to the atmosphere was estimated by applying a steady-state mass balance model.
Based on  vertical  concentration  profiles  from the lake, about 240 kg PCE was
released from the central  basin annually.

3.6  LEVELS OF EXPOSURE
3.6.1.   Mixing Ratios
3.6.1.1  Ambient Aii—A wide  variety of halogenated aliphatic  hydrocarbons,
including PCE, have been detected in ambient air.  Ambient measurements of PCE
have been  conducted  in  both the United States and other  areas  of  the world.
These determinations provide  a basis for assessing the levels to which human
populations may be exposed.
     Measured ambient air concentrations differ widely and undoubtedly reflect
the influences of a variety of factors, e.g., meteorological conditions, tropo-
spheric reactivity,  diurnal variations, sampling times, and  source emissions.
     Table 3-2 provides summary information regarding background and urban con-
centrations of PCE.  It should be noted that, in general,  these values reflect
short sampling times.
     Measurements of ground-level samples by Singh et al.  (1978b),  in both the
northern  and  southern  hemispheres,  gave average background levels of 0.040 ±
0.012 ppb (2.7 x 10"4 ± 0.08 x 10"4 mg/m3) and 0.012 ± 0.003 ppb (0.081 x lo"3
±0.02 x 10 3 mg/m3), respectively.   Globally,  the average background  level  of
PCE was 0.026  ±  0.007.7 ppb (1.7  x  lo"4 ±  0.47 x 10"4 mg/m3);  the  coefficient
of variation was 27 percent.   The average urban level of PCE was  found to be
about 30 times the background  level.
     Evidence  for  considerable variability in ambient air  levels  of  PCE was
shown by Lillian et al. (1975).  The authors attributed the variability of PCE
to its tropospheric reactivity (reaction with hydroxyl radicals).
003PE4/B                             3-10                             11/22/83

-------
TABLE 3-2.   AMBIENT AIR MIXING RATIOS OF PCE MEASURED AT SITES AROUND THE WORLD
Location
Alabama3
Birmingham
Arizona
Grand Canyon
Phoenix
California
San Bernardino Mtns.
oo Badger Pass
i
i— • Point Arena
Stanford Hills
Point Reyes
Dominguez
El Cajon
La Jolla
Los Angeles
Menlo Park
Mill Valley
Mt. Cuyamaca
Date of Reported Concentration, ppra (mg/mj)
Measurement Max i urn Minimum Average
April 12-22, 1977 0.008 0 0.001 ± 0.003

Nov. 28-Oec. 5, 1977 0 0 0
Apr. 23-May 6, 1979b 3.696 (0.025) 0.129 0.9938 ± 0.7155
(0.0008)
Fall. 1972^ 0.09 (6.1 x 10"4)
May 12-16, 1976 0.03 (2 x 10~4)
1976 0.03 (2 x 10~4)
Nov. 24-30, 1975 0.04 (2.6 x 10"4)
Dec. 2-12. 1975 0.043 (2.9 x lo"4)
May 14, 1976 2.9
April 9, 1975 0.31
Apr 9, 1974-Jan 6, 1976 2.3 0 0.53 ± 0.63
Sept. 22. 1972- 2.2 0.067 1.1 ± 0.45
April 19, 1979
Nov. 24-30, 1975 0.20 ± 0.21
Jan. 1-27, 1977 0.065 ± 0.075
Mar. 15, 1975 0.22
Reference
Pellizzari. 1979

Pellizzari, 1979
Singh et al., 1981
(0.0067 ± 0.0048)
Simmonds et al. , 1974
Singh et al. , 1977a
Singh et al. , 1978a
Singh et al. , 1977b
Singh et al. , 1977b
Pellizzari. 1977
Su and Goldberg, 1976
Su and Goldberg, 1976
Simmonds et al., 1974;
Singh, 1976;
Singh et al. , 1977a;
Su and Goldberg, 1976
Singh et al. , 1977a
Singh et al. , 1979
Su and Goldberg, 1976

-------
                                                                         TABLE 3-2.   (continued)
ro
Location
California
Oakland
Palm Springs
Riverside
San Jose
Santa Monica
Upland
Colorado
Denver
Delaware
Delaware City
Louisiana
Baton Rouge, Gel soar,
and Plaquemine
Maryland
Baltimore
Michigan
Detroit
New Jersey
Bayonne
Date of Reported Concentration, ppm (mg/m3)
Measurement Max i urn Minimum Average

June 30-July 8, 1979 0.64 0.12 0.31 * 0.17
May 5-11. 1976 1.1 0.12 0.28 ± 0.084
April 25, 1977- 0.98 0.37 0.49 ± 0.13
July 12, 1980
Aug. 21-27, 1978 . 1.1 ± 0.036
April 6, 1974 2.3
Aug. 13-Sept. 23, 1977 1.1 0.01 0.19 ± 0.35
Jun 16-26, 1980 0.47 0.24 0.39 ± 0.077

July 8-10. 1974 0.51 (0.0034) <0.02 (<0.0001) 0.24 (0.0016)

Fall. 1978 0.18 (0.001) .001 (0.007 x lo"3) 0.017 (0.118 x 10"3)

July 11-12, 1974 0.29 (0.0019) <0.02 (<0.0001) 0.18 (0.0012)

Oct 27-Nov 5. 1978b 2.16 (0.004) <0.1 (<0.001) 0.35 (0.002)
March, 1973-Dec. , 1973 8.2 (0.0055) 0.30 (0.0020) 1.63 (0.0110)
Reference

Singh et al. , 1979;
Singh et al. , 1981
Singh et al. , 1978a
Singh et al. , 1979;
Singh et al. . 1979




1980

Su and Goldberg, 1976
Pellizzarl, 1979
Singh et al. , 1980

Lillian et al. . 1975






Pellizzari et al. . 1979b

Lillian et al., 1975

Evans et al . , 1979
Lillian et al. , 1975





-------
                                                                     TABLE  3-2.   (continued)
Location
  Date of
Measurement
 Reported Concentration, ppra (mg/mj)
Maximum                        Minimum
Average
Reference
New Jersey

  New Brunswick

  New Brunswick
                                                   0.5 (0.003)

                                                   0.12 (0.0081)
Seagirt
Sandy Hook
Boundbrook, Rahway,
Edison and Passaic
Batsto3
Bridgeport3
Burlington3
Camden3
Carlstadt3
Edison3
Elizabeth3
Fords3
Middlesex3
Newark3
June 18-19, 1974
July 2, 1974
Sept 18-22, 1978
Feb. 26-Oec. 29. 1979
Sept. 22, 1977
Sept. 19, 1977
April 3-Oct. 24, 1979
Sept. 28-30, 1978
March 24, 1976-
Sept. 24, 1978
Sept. 15, 1978-
Oec. 29, 1979
Mar. 26, 1976-
Sept. 27. 1978
July 23-28, 1978
Mar. 23, 1976-
Dec. 29, 1979
0.88 (0.059)
1.4 (95 x 10"«)
58 (0.394)c
0.53
0.041

30
5.5
8.7
14
5.6
0.21
32
0.10 (0.067)
0.15 (10 x 10~«)
trace
0
0

0
1.1
0.11
0
0
0
0
0.32 (0.0022

30.9 (0.210)
0.034
0.020
0.027
1.8 ± 6.2
3.5 ± 2.3
2.8 ± 3.1
2.0 ± 3.1
2.8 ± 2.7
0.068 ± 0.091
1.3 ± 3.1
                                                                   Lillian et al.,  1976

                                                                   Lillian and Singh, 1974

                                                                   Lillian et al.,  1975

                                                                   Lillian et al.,  1975


                                                                   Pellizari et al., 1979

                                                                   Bozzelli et al.,  1980

                                                                   Pellizzari and Bunch, 1979

                                                                   Pellizzari and Bunch, 1979

                                                                   Bozzelli et al.,  1980

                                                                   Pellizzari et al. , 1979

                                                                   Pellizzari et al., 1979; Pelliz-
                                                                   zari, 1978; Bunn et al., 1975

                                                                   Bozzelli et al. ,  1980;
                                                                   Pellizzari, 1979

                                                                   Pellizzari et al. , 1979;
                                                                   Pellizzari, 1977

                                                            0.090  Bozzelli and Kebbekus, 1979

                                                                   Bozzelli and Kebbekus, 1979;
                                                                   Bozzelli et al.,  1980;
                                                                   Pellizzari, 1977

-------
TABLE 3-2.   (continued)
Location
New Jersey
Rahway
Rutherford3
3
Somerset
South Amboy
New York
New York City
OJ
i Niagara Falls and
£ Buffalo
Whiteface Htn.
Ohio3
Wilmington
Texas
Houston
Aldine3
Deer Park3
El Pasoa
Freeport3
Houston
Date of
Measurement
Sept. 20-22. 1978
May 1. 19 78- Dec 29, 1979
July 18-26. 1978
Jan. 27-Dec. 29, 1979
June 27-28. 1974
Aug 18-27. 1978°
Fall 1978
Sept. 17. 1974
July 16-26, 1974

Sept 16-25, 1978b
June 22-Oct. 20, 1977
July 29-30, 1976
April 5-May 1, 1978
Aug. 9, 1976
July 27, 1976-
May 24, 1980
Reported Concentration,
Maxium
5.0
9.2
0.068
2.2
9.75 (0.0661)
10.61 (0.0721)
2.0 (0.014)
0.19 (12.8 x 10~4)


4.52 (0.030)
0.037
0.15
0.39

1.3
ppm (oig/in3)
Minimum
2.7
0
0
0
1.0 (0.006)
0.16 (0.001)
0.02 (0.122 x 10"3)
0.02 (0.13 x 10"3)


<0.1 (<0.001)
0
0.01
0.11

0
Average
3.8 ± 1.2
0.89 ± 0.14
0.036 ± 0.26
0.21 ± 0.53
4.5 (0.030)
1.00 (0.006)
1.0 (0.0068)

0.15 ± 0.015

0.11 (0.001)
0.012
0.07
0.15
0.12
0.33
Reference
Bozzelli and Kebbekus, 1979;
Bozzelli et al. . 1980
Bozzelli et al. , 1980
Bozzelli and Kebbekus, 1979
Bozzelli et al. . 1980
Lillian et al. , 1975
Evans et al. , 1979
Pellizzari et al. , 1979b
Lillian et al. , 1975
Lillian et al. , 1975

Evans et al. , 1979
Pellizzari et al. , 1979
Pellizzari et al. , 1979
Pellizzari, 1979
Pellizzari et al., 1979
Pellizzari et al. , 1979;
                                          Singh et al..  1980

-------
                                                                            TABLE 3-2.   (continued)
Location
Texas
LaPorte3
Pasadena
Utah3
oo Hagna
^ a

-------
     Some of the  highest air  levels of PCE reported have been associated with
waste disposal  sites.  Pellizzari (1978) reported levels to ranges at sites in
New Jersey that ranged from trace amounts to a maximum of 58 ppb (0.394 mg/m3)
in  a  14-minute sampling period.  PCE was  adsorbed using Tenax cartridges.
     Coefficients of variation for most of the recent studies reported by Singh
et al. have been less than 30 percent.
     Howie (1980)  reported ambient air levels of  PCE  in the  vicinity of  laun-
dries to  be  as  high  as  32 ppb (0.22 mg/m3).   In this  study of indoor PCE con-
centrations, outdoor samples provided background data.  Measurements were made
by  adsorbing PCE  onto  charcoal filters,  followed  by  desorption with carbon
disulfide and quantification by GC-ECD and GC-MS.   Outdoor samples were collec-
ted for 24 hours.   Of 124 measured samples, 56 had 24-hour levels  of less than
1 ppb.  Replicate  sample analyses  were  reported to give an overall precision
of better than 20 percent for both indoor and outdoor samples.
3.6.1.2  Water—Various studies  have  shown that PCE is found in both natural
and municipal waters.   A  review by Deinzer et al. (1978) has summarized many
of the findings.  Love and Eilers (1982), in their review,  reported that halo-
genated solvents  such  as  PCE are seldom detected  in  concentrations greater
than a few micrograms per liter in surface waters.
3.6.1.2.1  Natural waters.    Surface waters,  such  as rivers and lakes, are the
most important  sources  of drinking water in  the United States.  Attempts have
been made to show  an epidemiological link  between  the presence of  halogenated
organic compounds  in drinking water and cancer (Harris and Epstein, 1976) but
a cause-effect relationship has not been established.
     Dowty et al.  (1975a,b) detected PCE by GC-MS techniques in  untreated Miss-
issippi River water  as  well  as  in treated water.   An  approximate six-fold re-
duction  in  concentration  occurred after  sedimentation  and chlorination.
Tetrachloroethylene  in  water  from  a  commercial  deionizing charcoal filtering
unit showed  a  marked increase over the amount  found  in  finished  water from
treatment facilities or commercial  sources of bottled water.  The value of
charcoal  filtering  to   remove  organics  from water requires  further study.
     Suffet et  al. (1977) reported detection of PCE in river waters supplying
drinking  water  to Philadelphia, Pennsylvania.  The Belmont  Water Treatment
Plant, with an average capacity of 78 million gallons per day,  obtains influent
from the Schuykill River.
     In a study designed  to  detect pollutants in  surface water at different
U.S. sites, Ewing et al. (1977) identified PCE among the pollutants.  Detection
003PE4/B                             3-16                             11/22/83

-------
limits were not reported.   The highest level  (45 ug/1)  was reported for surface
water in Ashtabula,  Ohio,  located along the  south edge of Lake Erie.   In the
vicinity of  Philadelphia, 3 ug/1 was detected.  Less than 1 ug/1 was reported
along the  Ohio  River and  at the confluence  of  its tributaries.   Similarly,
1 ug/1 or  less  was  reported for all sites sampled in  the Great Lakes  and at
six  sites  along  the  Tennessee River.   An exception was Chattanooga where 13
ug/1 was reported.  In the greater Chicago area, the highest level  found was 5
ug/1.  In  all  samples  taken in California,  Oregon, and  Washington,  PCE was
either not  detected  or was  found at a concentration of 1 ug/1  or less.  Sam-
pling sites included those in the vicinity of Los Angeles Harbor,  Santa Monica
Bay, and San Francisco Bay,  at three sites along the Willamette River in Oregon,
and  two in the Puget Sound area.
     Tetrachloroethylene was  among  a  number  of halogenated organics found in
community drinking water supply wells in Nassau County, New York.   Because con-
tamination, 16 of these wells were closed by the New York State Health Depart-
ment  (Ewing  et  al.,  1977).  The maximum  detected  level of PCE  in the contami-
nated wells  was 375 ug/1.  Since  PCE  is generally not used as  a  cesspool
cleaning agent, previous industrial dumping may be the source of contamination.
     Pearson and  McConnell  (1975)  found an average PCE concentration of 0.12
ppb  in  Liverpool  Bay sea  water; the maximum concentration found was 2.6 ppb.
Sediments  from  Liverpool  Bay  were found to contain 4.8 ppb (w/w).   No direct
correlation was found between PCE concentration in sediments and in the waters
above.  Rainwater collected near an organochlorine manufacturing site was found
to contain 0.15 ppb  (w/w) PCE (Pearson and McConnell,  1975); it was not detected
in well  waters.   Upland waters of two  rivers  in  Wales were  found  to contain
approximately  0.15  ppb PCE; similar levels  of  trichloroethylene  were found
(Pearson and McConnell, 1975).
      Lbchner (1976)  found  that levels of PCE  in  Bavarian lake waters  ranged
from 0.015 to  3 ppb  (0.015 x 10~^ to 2.7 x  10~3 mg/1).   European surface
waters were  reported to have uniform PCE concentrations ranging from 0.2 x 10  3
to 0.002 mg/1.   Analyses of river, canal, and sea water, all containing effluent
from production and  user sites  in four countries, revealed  PCE concentrations
ranging  from 0.01 to 46 ppb  (0.01  to  46 ug/liter) (Correia et al. ,  1977).
3.6.1.2.2   Municipal waters.   Bellar  et  al.  (1974)  measured  the concentration
of PCE in water obtained from sewage treatment plants  in several cities.  Before
treatment,  the average PCE concentration was  6.2 ug/1.   The  treated  water

003PE4/B                             3-17                              11/22/83

-------
before chlorination  contained  3.9  pg/l  PCE.  After chlorination, the effluent
contained 4.2 (jg/1 PCE.
     Tetrachloroethylene has been  detected in  the drinking water of a number
of U.S. cities.  These include Evansville, Indiana (Keith et al., 1977); Kirk-
wood,  Missouri  (Keith  et al.,  1977); New  Orleans,  Louisiana  (Dowty et al.,
1975); Jefferson  Parish, Louisiana (Dowty et  al.,  1975b);  Cincinnati,  Ohio
(Keith et al., 1977); Miami, Florida (Keith et al.,  1977); Grand Forks,  North
Dakota (Keith  et  al.,  1977);  Lawrence,  Kansas (Keith et al.,  1977); New York
City (Keith et al., 1977); and Tucson, Arizona (Keith et al.,  1977).
     Concentrations  recorded for  the  above cities were less than 1 ug/1.   An
exception was  Jefferson  Parish,  which had a measured concentration of  5 ppb
(5 ug/1).   Keith  et  al.  (1977) did not  detect PCE  in the drinking water of
Philadelphia.   Tetrachloroethylene was found in Evansville tap water from July
1971 to December 1972.   The Ohio River Basin, a heavily industralized area, is
upstream from  Evansville and  serves as a  major source of drinking water for
that community.
     Dowty  et  al.  (1975b)  determined levels of PCE  in the drinking water for
New Orleans.   Considerable  variation in the relative concentrations  of the
various halogenated compounds was observed from day  to day.
     Contamination of drinking water by PCE was recently investigated by Wake-
ham et al.  (1980).   It was reported that elevated concentrations of PCE were
found  in drinking  water  transported in vinyl-coated asbestos-cement pipes  in
areas of the  town  of Falmouth, Massachusetts.  Tetrachloroethylene  is used  as
a solvent during the application of the vinyl coating to the pipe during manu-
facturing.   It was suggested  that  residual  solvent  leaches into the water
carried in these pipes.
     Using  a  charcoal  trap  with flame ionization detection, Wakeham and co-
workers (1980)  detected  levels ranging  from 140  to  18,000  ppb in unflushed
pipes.   In other parts of the distribution system, levels were less than 2  ppb.
The authors reported  that  vinyl-coated  asbestos-cement pipe has been used  in
parts of the  northeastern  United States over  the past decade  in response  to
concerns that  water  carried in uncoated pipes  could contain asbestos fibers.
     In municipal  waters supplying  the  cities of  Liverpool,  Chester,  and
Manchester, England, 0.38 ppm (w/w) PCE was found (Pearson and McConnell, 1975).
     Munich (Germany)  drinking water was  analyzed by  Lbchner (1976).  Samples
taken at various sampling points and times gave a range of 1.1 x 10 ? to 2.4 x

003PE4/B                             3-18                             11/22/83

-------
10 3 mg/1.   Raw sewage at Munich contained 0.088 mg/1  PCE.   On  mechanical  clari-
fication,  the 24-hour average concentration of PCE  was 0.0068 mg/1.

3.7  ANALYTICAL METHODOLOGY
     Tetrachloroethylene has been analyzed in air and in water, as well as in
biological  fluids,  by a variety of methods.   Separation of  PCE  from other  com-
pounds is usually  carried  out  by gas chromatography (GC).   Quantification is
usually made by  either electron capture detection  (ECD) or mass spectroscopy
(MS).  These analytical methods, GC/ECD or GC/MS, have a lower  limit of detec-
tion of a few ppt.

3.7.1  Ambient Air
     Because PCE levels in air are typically in the sub-ppb range, the sampling
and analysis techniques have been designed to detect trace  gas  levels.
3.7.1.1  Sampling and Sources of Error—Because of  the low  levels occurring in
ambient air, sampling  techniques  have focused on adsorption onto solids such
as charcoal  (Evans et al., 1979) or on concentration methods that increase the
amount of PCE to above detection limits (Rasmussen  et al.,  1977).   In the  upper
troposphere, PCE has been sampled by pumping air into stainless steel or glass
containers until there is a positive pressure relative to the surrounding  atmos-
phere (Singh et al., 1979).
     Evans et  al.  (1979)  sampled PCE using a method based  on adsorption onto
activated charcoal,  followed by desorption by carbon  disulfide/methanol.   The
precision of the  analytical  method,  expressed as a coefficient of  variation
for  the total  measurement system (including sample collection, handling,  and
preparation) was reported as 16 percent.  When 49 quality control samples  were
analyzed,  the  overall  percent  recovery  from  the  charcoal tubes was  70.2 ±1.7
percent.   When the total measurement system was independently checked by using
Tenax  GC, another  solid adsorbent, the paired  data  were correlated with  a
coefficient of 0.82, with the average Tenax result exceeding the average char-
coal  result  by 21  percent.   Samples were  in  the  sub-  to  low-ppb range.  Evans
et  al. (1979)  reported that PCE is stable on charcoal tubes for at least one
month  at 0°C.   The lower limit of detection for the total  measurement method
(to  include GC/ECD) was estimated at 0.68 ug/m3 (0.1 ppb).
     Pellizzari and  Bunch  (1979)  reported the use of  Tenax  GC,  a  porous poly-
mer  based on 2,6-diphenyl-p-phenylene oxide, to adsorb PCE from ambient air.
Recovery was made  by thermal desorption  and  helium  purging into  a  freezeout
003PE4/B                             3-19                             11/22/83

-------
trap.   The estimated  detection  limit,  when high resolution GC/MS is used,  is
0.38 ppt (2.5 x 10 6 mg/m?).   Accuracy of analysis was reported at ± 30 percent.
Included among the inherent analytical  errors were (1) the ability to accurate-
ly determine  the  breakthrough  volume,  (2) the percent recovery from the sam-
pling cartridge after a period of storage, and (3) the reproducibility of ther-
mal desorption  from  the  cartridge and its  introduction  into  the analytical
system.   To minimize  loss  of sample, cartridge  samplers  should be enclosed in
cartridge holders and placed in a second container that can be sealed, protec-
ted from  light,  and  stored at 0°C.  The advantages reported for Tenax include
(1) low water  retention,  (2)  high thermal  stability, and  (3) low background
levels (Pellizzari, 1974,  1975, 1977;  Pellizzari  et al., 1976).  Singh et al.
(1982) have  cautioned that Tenax suffers  from serious  artifact problems.
     Krost et  al.  (1982) reported an estimated  detection limit of 0.3 ppt for
PCE using high  resolution  GC/MS.   The detection  limit was calculated on the
basis of  the  breakthrough  volume  for a known amount of Tenax GC at 10°,  21°,
and 32°C.   Field sampling and analysis precision of the Tenax method was  found
to range  from  ± 10 to ± 40 percent relative standard deviation for different
substances when replicate field sampling cartridges were examined.
     Knoll et al. (1979) reported resolution of PCE from other chlorinated hy-
drocarbons with Carbopak C-HT,  a graphitized thermal  carbon black treated with
hydrogen at 1000°C.   Carbowax  20M,  reacted with  nitroterephthalic acid, was
reported not  to  give  good  separation.   Porapak  T, a  porous polymer  based on
ethylene glycol dimethacrylate, was reported to give good separation.
     A freezeout concentration  has  been developed by Rasmussen and coworkers
(1977) to determine  trace  levels  of PCE  in the  presence of other compounds.
The detection  limit was  reported  at 0.2 ppt  (1.36 x  10  6  mg/m?) for 500-ml
aliquots of ambient air samples measured by GC coupled with EDC.   When freeze-
out is complete, PCE remains behind, and such gases as oxygen and nitrogen are
passed through  as  the freezeout loop  is  heated.  The carrier gas sweeps the
contents onto the column.
     Singh et  al.  (1979,  1982) have employed the cryogenic  trapping of air
containing trace  levels of PCE and other compounds of interest.   During sam-
pling, traps  are maintained at  liquid  oxygen  temperature.  Traps were made of
stainless steel  packed  with  a  4-inch bed of glass beads or glass wool.   Ali-
quots are thermally desorbed and injected directly into the gas chromatograph.
Both electric  heating and  hot water desorption  techniques were  found to be
satisfactory.
003PE4/B                             3-20                             11/22/83

-------
     Makide et al.  (1980)  employed  stainless steel  canisters for sampling of
air containing PCE at levels of about 20 ppt.   Canisters were polished electro-
chemically.  Canisters were  evaluated  to 10 4 Pa at 200°C  before sampling.
The composition of the samples was reported to have  remained unchanged for over
a year.
     The monitoring method  recommended  by the National  Institute for Occupa-
tional Safety  and Health (1977) utilizes  adsorption onto charcoal followed by
desorption with carbon disulfide.   This method is recommended for the range 96
to 405 ppm (655 to 2749 mg/m3).  The coefficient of  variation for the analyti-
cal and  sampling  method  is 0.052.  A reported disadvantage is that the char-
coal may be overloaded, thus limiting the amount of  sample that can be collec-
ted.  This may be obviated by using more than one adsorber.
     Budde and Eichelberger (1979) reported  that carbon  adsorption methods
generally  have more  disadvantages than those methods using  porous  polymers.
The advantage  of  porous polymers  coupled  with thermal desorption, as  contras-
ted with  solvent  desorption,  is higher sensitivity, because the total sample
is measured and there is no background from the solvent.  However, because the
total sample is measured,  multiple samples must be collected to insure against
accident and loss of sample, and to obtain information on the precision of the
method.  Tenax GC was reported to be superior to other polymers for organics
analysis.  Samples  are  taken by pulling  air  through glass  tubes  packed with
Tenax  GC,  60/80  mesh and  supported by plugs of glass wool.   After a suitable
sampling period  (about  2  to 4  hours  in  urban areas),  tubes are  capped and
stored.  Samples  are  thermally desorbed  (250 to 270°C) for 3 minutes under  a
10-ml helium flow.
     Criteria  for evaluating methods using solid sorbents to collect organic
compounds  from air  have  been discussed  by  Melcher  et al.  (1978).   Among  the
factors to be  considered are effects of (1) size of collection tube (2) break-
through  volume,  (3)  humidity,  (4) temperature, (5)  migration, (6) desorption
efficiency, and (7) concentration.
3.7.1.2  Analysis—The sampling methods which use solid adsorbents or cryogenic
techniques have  the trap  connected to the gas chromatograph by multiple-port
gas  sampling  valves.   With solid traps,  the  collected  organics are quickly
heated  and the desorbed organics  are  passed through capillary  columns.   A
number  of  coating materials in the  capillary  columns  have  been successfully
used  for separating PCE.   These  materials  include  (1)  SF-96 on 100/120 mesh

003PE4/B                             3-21                            11/22/83

-------
Chromosorb W (Cronn et al.,1977); (2) SP-2100 on 80/100 mesh Supelcoport (Singh
et al., 1979); (3) 80/100 mesh Carbopak C-HT, Porapak T, and SP-2100/0.1 percent
Carbowax 1500 on 100/ 120 mesh Supelcoport (Knoll et al., 1979).   In the method
used  by  Evans  et al.  (1979)  in  field  studies,  a 1.8-m glass column with a
2-mm  i.d. , packed with 0.1 percent SP-1000 on Carbopack C, 80/100 mesh was used
to separate  PCE  from  other  organics  in  ambient  air  samples.  Twenty-four-hour
samples were adsorbed onto  charcoal.  After  desorption  with carbon  disulfide/
methanol, a 1.0-ul aliquot was injected into the gas chromatograph.   The separa-
tion  conditions  included  an oven temperature of 125°C (all transfer lines at
least 170°C), and the carrier gas was 5 percent methane in argon.  Quantifica-
tion  was made  by ECD (Nickel 63; ECD  temperature  of 218°C) and a  standing
current of 0.5  amperes.   To  insure  that  the cell  was not contaminated, the
sensitivity of the detector was  evaluated by comparing the standing current
with the pulse frequency curve.
     Electron capture detection is a method of choice used by a number of inves-
tigators (Singh  et al.,  1977, 1979, 1982; Rasmussen et al., 1977).   Singh et
al.  (1979) maintained the ECO at a higher temperature (325°C) than did Evans et
al.  (1979), because it was found that the ECD response increased with an increase
in temperature.   The  identity of PCE was confirmed by determining its ionization
efficiency as well as the EC thermal response.
     More recently, Singh  et al.  (1982) maintained  the  ECD  at 275°C with a
carrier flow rate  of  40 ml/min  to analyze for  PCE  and 11 other halogenated
organics in ambient air.   Identity of PCE and other  compounds was established
from  retention times  on  multiple columns, the  ECD  thermal response, and the
ECD ionization efficiency.   Lillian and Singh (1974) reported that the accuracy
associated with GC-ECD measurements of compounds having ionization efficiencies
exceeding 50 percent  is 75  percent or greater.   Using two  ECDs in series, PCE
was found to have an  ionization  efficiency of 70 percent.   In a  comparison of
GC-ECD with GC-MS, Cronn et  al.  (1976)  judged GC-ECD to be superior in  repro-
ducibility for quantitating  halocarbons.   Of four halocarbon standards  (PCE
not among them)  measured by  GC-ECD,  the coefficients of variation ranged from
1.4 to 4.3 percent, compared to  a range of 4 to 19  percent when  11  halocarbon
standards were measured by GC-MS.  A close agreement between the levels of PCE
and other halocarbons determined by GC-ECD and GC-MS on the same ambient air
samples was obtained  by Russell  and Shadoff  (1977).
     GC-ECD was used  by Pellizzari et al.  (1979) to measure PCE in ambient air
samples.   Samples were  adsorbed  onto charcoal and desorbed with a mixture of
003PE4/B                             3-22                             11/22/83

-------
methanol and carbon disulfide, and aliquots were separated on a 2.5-mm (i.d.)
Pyrex column containing  0.2  percent  Carbowax 1500  on Carbopack C.   The esti-
mated detection limit was 2.5 x 10 6  mg/m3 (0.38 ppt).
     Makide et al.  (1980) separated trace levels of PCE from other halogenated
organics on a silicone OV-101 column  (10 percent by weight coated on Chromosorb
W-HP, 80-100 mesh) of 5-mm i.d. and 3 m long.   Samples were transferred to the
column cooled at -40°C during preconcentration.   Separation was carried out by
raising column  temperature 5°C per minute up to 70°C.  Methane was added to
the carrier gas (nitrogen) to improve the signal-to-noise ratio and to stabilize
the baseline.  Quantification was made by a constant-current ECD.   The detection
limit for  PCE was  reported as  <0.05  ppt.   Precision was reported to be within
2 percent.
     To measure PCE at levels expected to occur in  occupational air, flame ion-
ization detection has been used.   The analytical method S335, suggested by the
National Institute  for Occupational  Safety and  Health  (1977) for organic  sol-
vents in air,  utilizes  adsorption onto charcoal, followed by desorption with
carbon  disulfide.   PCE is separated  by  GC.  The  method is  recommended  for the
range 96 to 405 ppm (655 to 2749 mg/m3).  The coefficient of variation for the
analytical  sampling method is 5.2 percent.  With the method, interferences are
minimal, and those that do occur can be eliminated  by altering chromatographic
conditions.

3.7.2  Water
3.7.2.1  Sampling--A  variety  of  techniques and methods are  commonly  used to
sample  trace  levels  of  PCE  and other halogenated  organics  in water samples.
Coleman et  al.  (1981) have reported  that  the Grob  closed-loop-stripping tech-
nique is an  excellent tool to  monitor organics  in  water at  the  ppt  level.   It
was  reported that  a million-fold concentration of most low  and  intermediate
molecular weight organics can be achieved.  Quantisation is performed by spik-
ing the initial water sample with a  series of internal standards, stripping at
30°C  for two hours,  and by chromatographing the CS2 extract on a wall-coated
open-tubular capillary.
3.7.2.1.1  Gas purging and trapping.   In this method, finely divided gas bubbles
are  passed  through the  sample, transferring the organic compounds to the gas
phase.  The gas is then passed through a solid adsorbent in a trap.  Compounds
are  desorbed  at elevated temperature by backflushing with a carrier gas into

003PE4/B                             3-23                             11/22/83

-------
the gas chromatograph (Budde and Eichelberger, 1979).   Since the boiling point
of PCE is 121°C, Tenax GC would be an effective absorbent.
     The purge  and  trap  procedure is widely  used,  as  is  the purging device
developed by Bellar and Lichtenberg (1974).   For most organic compounds, detec-
tion limits  as  low  as  1 ^9/1 can  be obtained  when GC/MS is  used for analysis.
Mieure (1980) reported that adding salt to the sample or increasing its tempera-
ture dramatically improves the removal of most organic compounds.
     Otson and  Williams (1982)  have described a modified purge and trap tech-
nique  for evaluation  of  volatile  organic pollutants in water.   The detection
limit reported for PCE was 0.1 ug/1 with ECD,  and 1 ug/1 with flame ionization
detection (FID).  Tenax GC was used as packing for the combined trap/chromatog-
raphic column.
3.7.2.1.2  Headspace analysis.  This  method  describes  static sampling of the
vapor phase that is in equilibrium with the aqueous sample.   The concentration
in the  headspace  is  proportional  to the  concentration  in the water (Kepner et
al., 1964).  In this procedure, trace organics in the  range of 10 to 100 ug/1
can be  sampled  (Mieure,  1980).   Mieure (1980) reported a detection limit for
PCE (analyzed by ECD) of 0.01 ug/1.  With flame ionization detection,  the limit
was 32 ug/1.  Typically,  a 1-  to 2-ml sample of the headspace is  removed and
injected into the gas  chromatograph.  Headspace extraction  coupled with mixed
column separation and  ECD analysis was reported by Castello et al.  (1982)  to
be suitable for rapid screening of drinking water supplies.
3.7.2.1.3  Liquid/liquid extraction.  Mieure  (1980) reported that recovery of
PCE from water spiked with 2.3 to 90 ug/1 ranges from 100 to 113 percent.   The
precision ranges  from  10  to 12 (RSD).   These results  were  obtained from a
round-robin study,  by  the American Society for Testing and Materials  (ASTM)
Committee D19 on  Water,  using liquid/liquid  extraction.  The extractant was
not identified.
     Budde and Eichelberger (1979) cautioned that a disadvantage to this method
is that  very volatile  compounds may be  lost  during extract concentration or
during  solvent  elution from the  gas  chromatograph.  Methylene  chloride was
recommended as the only general-purpose solvent.
     Sheldon and  Hites (1978) used methylene  chloride  in a  sampling procedure
applied  to  the  identification  of  PCE and 98 other organic compounds in river
water.    Grab samples  were collected in  amber glass  bottles and samples for
solvent  extraction  were   immediately  preserved  by acidifying to pH  2  with
hydrochloric acid and  by  adding 250 ml  of methylene chloride.  The analytical
003PE4/B                             3-24                             11/22/83

-------
techniques used were those reported by Jungclaus et al.  (1978).   Solvent extrac-
tion efficiencies were  not  determined.   While PCE was previously detected in
vapor stripping analysis  of  prior samples,  it was not detected  in the water
samples cited in their report.
3.7.2.2  Analysis—Schwarzenbach  et al.  (1979)  used ECD to measure PCE  levels
in water  samples.   Volatile  organics  were purged and adsorbed onto charcoal.
Desorption was by carbon disulfide.  Quantification was made by FID and ECD.
     Dowty et  al.  (1975a)  used Tenax GC  in trapping  purgeable organics from
water samples.  The polymer  containing the trapped organics was  placed  in the
GC injection  port  maintained at 200°C.   Final separation was made on a glass
capillary column coated  with Pluronics  121.   The effluent of the column was
split to allow for FID and ECD.

3.7.3  Biological  Media
     Ramsey and Flanagan  (1982)  have  described a gas chromatographic method
reported to be suitable for analysis of PCE and other organics present in blood.
Detection was both by flame ionization and ECD.   Approximately 200 ul of blood
or 200 mg tissue is required for analysis.

3.7.4  Calibration
     Singh et  al.  (1982)  found that primary  standards of PCE in the  low-ppb
range could  be satisfactorily calibrated using  permeation  tubes maintained
either at 30° or 70°C.  Permeation tubes were standard FEP or TFE Teflon.   All
permeation tubes were conditioned for two weeks or longer.   Errors in the per-
meation rate  were  ± 10  percent.   Singh  et al. (1982) found that long-term (2
years) stability of  primary  standards (10 ppm) of PCE in aluminum containers
was excellent.

3.7.5  Storage and Stability of PCE
     Sampling  of exhaled  breath commonly  is accomplished by  use  of Saran bags
or glass pipettes.   Temperature and storage time of the samples before analysis
are factors to be considered in obtaining accurate data.
3.7.5.1  Glass Sampling Tubes—Evaluation of  glass sampling  tubes was made by
Pasquini  (1978).   Serial  alveolar breath samples were collected in the tubes
and the concentrations of PCE were analyzed by a gas chromatograph equipped with
a flame ionization detector.   Analysis of vapor retention over 169 hours indi-
cated  that  glass  tubes can  be  acceptable containers  for breath  samples  if
003PE4/B                             3-25                             11/22/83

-------
precautions are  taken.  Moisture, temperature, and tube surface and condition
can greatly alter vapor retention.
     In tubes  filled  with  breath samples taken at room temperature and also
stored at room temperature, the mean percent loss of PCE was 64.8 ± 9.4.   Par-
titioning of PCE between the vapor and liquid states appears to be a reasonable
explanation for vapor retention loss.   It was shown for trichloroethylene that
if storage tubes were maintained at 37°C, vapor retention was greater.   It was
also greater if si 1 iconized tubes were used.
3.7.5.2  Saran, Teflon and Tedlar Containers—Saran bags as storage containers
for PCE vapors  have  been  evaluated by Desbaumes and Imhoff (1971).   Although
it was concluded that Saran can be an acceptable container, the diffusion rate
was appreciable  over  a  24-hour storage period.  Storage temperature was not
reported.
     Teflon containers were  judged  by Drasche et al.  (1972) to be more suit-
able than  Saran  even  though  losses  of  PCE due  to adherence  to Teflon surfaces
were appreciable.  Within the first 30 minutes after introduction of a mixture
(relative humidity = 45 percent) of benzene, trichloroethylene, and PCE into a
Teflon bag, vapor  concentrations of each dropped 40 to 60 percent.   However,
when the bag was heated to 100°C for 30 minutes after the mixture had been stored
for 44 hours at 25°C, concentrations rose to the initial values.
     Knoll et  al.  (1979)  reported that PCE, when stored at ambient tempera-
tures  for  10 days  or  less  in Tedlar bags, was  stable.  When the vapor mixture
is heated to 70°C, PCE is stable for no more than 5 hours.
003PE4/B                             3-26                             11/22/83

-------
3.8  REFERENCES


Altshuller, A.  P.   Lifetimes of organic molecules in the troposphere and lower
     stratosphere.   Adv. Environ.  Sci. Technol. 10:181-219, 1980.

Bellar, T. A.,  and J.  J.  Lichtenberg.  Determining volatile organics at the
     microgram per litre levels by gas chromatography.   J. Am. Water Works
     Assoc. 66:739-744, 1974.

Bellar, T. A.,  J.  J.  Lichtenberg,  and R.  C. Kroner.  The occurrence of organo-
     halides in chlorinated drinking waters.  J. Am. Water works Assoc. 66:703-
     706, 1974.

Bonse, G., and D.  Henschler.  Chemical reactivity, biotransformation, and toxi-
     city of polychlorinated aliphatic compounds. Crit. Rev. Toxicol. 4:395-409,
     1976.

Bozzelli, J. W., and B. Kebbekus.   Analysis of selected volatile organic sub-
     stances in ambient air.  New Jersey Institute of Technology.  PB80-144694,
     Newark, New Jersey, 1979.

Bozzelli, J. W., B. Kebbekus, and A. Greenberg.  Analysis of selected toxic
     and  carcinogenic substances in ambient air in New Jersey.  New Jersey
     Institute of Technology, Newark, New Jersey, 1980.

Brodzinsky, R., and H. B.  Singh.  Volatile organic chemicals in the atmosphere:
     an assessment of available data.  Final Report.  SRI International, Menlo
     Park, CA.   Prepared for the U.S. EPA, 1982.

Budde, W.  L., and J.  W. Eichelberger.  An EPA manual for organics analysis
     using gas chromatography—mass spectrometry, 1979.

Bunn, W.  W., E. R.  Deane, D. W. Klein, and R. D. Kleopfer.  Sampling and
     characterization of air for organic compounds.  Water Air Soil Pollut.
     4(3-4):367-380, 1975.

Calvert,  J. G.   Hydrocarbon  involvement in photochemical smog.  Environ. Sci.
     Technol.  10:256-262, 1976.

Campbell, M. J., J. C. Sheppard, and B. F. Aiu.  Measurement of hydroxyl
     radical concentrations  in boundary layer air by monitoring CO oxidations.
     Geophys.  Res.  Lett. 6:185-188, 1979.

Castello, G., T. C. Gerbino, and S. Kanitz.  Gas chromatographic analysis of
     halocarbons in drinking water by headspace extraction and mixed column
     separation.  J. Chromat.  247:263-272, 1982.

Chemical  Marketing Reporter, June 18, 1979b.

Chemical  Marketing Reporter, May 7, 1979.
003PE4/B                              3-27                              11/22/83

-------
Coleman, W. E., R. G. Melton, R. W. Slater, F. C. Kopfler, S. J. Voto, W. K.
     Allen, and T. A. Aurand.  Determination of organic contaminants by  the
     Grob closed-loop-stripping technique.  J. Am. Water Works Assoc.
     73(2):119-125, 1981.

Considine, D. M., ed.  Chemical and Process Technology Encyclopedia.  New York.
     NY:  McGraw Hill, 1974.

Correia, Y., G.  J. Martens, F. H. Van Mensch, and B. P. Whim.  The occurrence
     of trichloroethylene, tetrachloroethylene, and 1,1,1-trichloroethane in
     Western Europe in air and water.   Atmos. Environ.  11:1113-1116, 1977.

Cronn, D.  R., R. A. Rasmussen, and E.  Robinson.'  Phase I report:  measurement
     of tropospheric halocarbons by gas chromatography-mass spectrometry.
     Washington State University, August 1976.

Cronn, D.  R, R.  A. Rasmussen, and E.  Robinson.  Report for Phase II:  Measure-
     ment of tropospheric halocarbons by gas chromatography-mass spectrometry.
     Washington State University, October 1977.

Cronn, D.  R., R. A. Rasmussen, E. Robinson, and D. E. Harsch.  Halogenated
     compound identification and measurement in the troposphere and lower
     stratosphere.  J. Geophys. Res.  82(37):5935-5944, 1977b.

Crutzen, P. J., I. S. A. Isaksen, and J. R. McAfee.  The impact of the chloro-
     carbon industry on the ozone layer.  J. Geophys. Rev. 83:345-363, 1978.

Deinzer, M., F.  Schaumburg, and E. Klein.  Environmental Health Sciences
     Center Task Force Review on halogenated organics in drinking water.
     Environ. Health Persp.  24:209-239, 1978.

Desbaumes, E., and C. Imhoff.  Use of Saran bags for the determination of
     solvent concentration in the air of workshops.  Staub-Reinhalt Luft
     31(6);36-41, 1971.

Dilling, W. L., N. B. Tefertiller, and G. J. Kallos.  Evaporation rates  and
     reactivities of methylene chloride, chloroform, 1,1,1-trichloroethane,
     trichloroethylene,  tetrachloroethylene, and other chlorinated compounds
     in dilute aqueous solutions.  Environ. Sci.  Technol. 9(a):833-838,  1975.

Dilling, W.  Interphase transfer processes.  II.   Evaporation rates of chloro-
     methanes, ethanes,  ethylenes, propanes, propylenes from dilute aqueous
     solutions.   Comparisons with theoretical predictions.  Environ. Sci.
     Technol. 11(4):405-409, 1977.

Dimitriades, B., B. W. Gay, Jr., R. R. Arnts, and R. L. Seila.  Photochemical
     reactivity of perchloroethylene:   a new appraisal. J. Air Pollut. Control
     Assoc. 33(6):575-587, 1983.

Dowty, B.  J., D. R. Carlisle, and J.  L. Laseter.   New Orleans drinking water
     sources tested by gas chromatography - mass spectrometry.  Environ. Sci.
     Technol.  9:762-765, 1975a.
003PE4/B                             3-28                              11/22/83

-------
Dowty, B.  J. ,  D.  R.  Carlisle, J.  L.  Laseter,  and J. Storer.  Halogenated
     hydrocarbons in New Orleans drinking water and blood plasma.  Science
     187:75-77, 1975b.

Drasche, H. ,  L. Funk, and R.  Herbolsheimer.  Storing of air samples for the
     analysis of contaminants especially, of chlorinated hydrocarbons.  Staub-
     Reinhalt.  Luft 32(9):20-25, 1972.

Evans, G.  F.,  R.  E.  Baumgardner, J.  E. Bumgarner, P. L. Finkelstein, J. E.
     Knoll, and B.  E. Martin.  Measurement of perchloroethylene  in ambient
     air.   EPA-600/4-69-047.   U.S.  Environmental Protection Agency, August
     1979.

Evans, G.  F.   Measurement of perchloroethylene in ambient air.   Proceedings of
     the International  Technical Conference on Toxic Air Contaminants, October
     9-10,  1980.

Ewing, B.  B.,  E.  S.  K.  Chian, J.  C.  Cook, C. A. Evans, P. K. Hopke, and E. G.
     Perkins.   Monitoring to detect previously unrecognized pollutants in
     surface waters.  EPA-560/6-77-015.  U.S. Environmental Protection Agency
     July 1977.

Fishbein,  L.   Potential industrial  carcinogens and mutagens.  EPA-560/5-77-005,
     Office of Toxic Substances, U.S. Environmental Protection Agency.  May,
     1977.

Fuller, B.  B.   Air pollution assessment of tetrachloroethylene,  Mitre Corp.,
     February,  1976.

Gay, B. W., P.  L. Hanst, J. J. Bufalini, and R. C. Noonan.  Atmospheric oxida-
     tion of chlorinated ethylenes.    Environ. Sci. Technol. 10(1):  58-67,
     1976.

Gonikberg,  M.  G.  , V. M. Zhulin, and V. P. Butuzor.  Bull. Acad.  Sci. USSR
     Dir.  Chem. Sci. 7^9:1956 (English translation).

Gosselin et al.  Clinical toxicology  of commercial products.  4th Edition,
     1976.

Graedel, T. E.   Bell Laboratories to  H. B. Singh, Personal  Communication, SRI
     International, October,1978.

Grimsrud,  E.  P., and R. A. Rasmussen.  Survey and analysis  of halocarbons in
     the atmosphere by gas chromatography-mass spectrometry.  Atmos. Environ.
     9:1014-1017, 1975.

Halogenated Solvents Industry Alliance.  Comments submitted to the U.S. Environ-
     mental Protection Agency, August 1983.

Handbook of Chemistry and Physics, 57th Edition, CRC Publishing  Co., Cleveland,
     Ohio,  1976.

Hardie, D.  W.  F.   Chlorocarbons and Chlorohydrocarbons.  In:  The encyclopedia
     of chemical technology, 2nd Edition,  1966.


003PE4/B                              3-29                              11/22/83

-------
Harris, R.H., and S. S. Epstein.  Drinking water and cancer mortality in
     Louisiana.  Science  193:55, 1976.

Howard, C. J.  Rate constants for the gas-phase reactions of OH radicals with
     ethylene and halogenated ethylene compounds.  J. Chem. Phys. 65(11):148-154,
     1976.                                                        ~

Howie, S.  J.  Ambient perchloroethylene levels inside coin-operated laundries
     with dry-cleaning machines on the premises.  Environmental Monitoring
     Systems Laboratories, U.S. Environmental Protection Agency, December 1980.

Huybrechts, G., J. Olgregts, and K. Thomas.  Trans. Faraday Soc. 63:  1647,
     1967.

Jensen, S., and R. Rosenberg.  Degradability of some chlorinated aliphatic
     hydrocarbons in sea water and sterilized water.  Water Res. 9:659-661,
     1975.

Jungclaus, L. A., V. Lopez-Avila, and R. A. Hites.  Environ. Sci. Technol.
     12:88, 1978.

Keith, L.  H., A.  W. Garrison, F. R. Allen, H. H. Carter, T. L. Floyd, J. D.
     Pope, and A. D. Thruston, Jr.  Identification of organic compounds in
     drinking water from thirteen U.S. cities:  chap. 22.  In:  Identification
     and analysis of organic pollutants in water.  L. H. Keith, ed.  Ann Arbor
     Science, 1977.

Kepner, R. E.,  H. Maarse, and J. Strating.  Gas chromatographic head space
     techniques for the quantitative determination of volatile components in
     multicomponent aqueous solutions.  Anal. Chem. 36: 77-82, 1964.

Knoll, J.  E., M.  A. Smith, and M. Rodney Midgett.  Evaluation of emission test
     methods for halogenated hydrocarbons.  Volume 1.  EPA-600/4-79-025.  U.S.
     Environmental Protection Agency, March 1979.

Krost, K.  J., E.  D. Pellizzari, S. G. Walburn, and S. A. Hubbard.  Collection
     and analysis of hazardous organic emissions.  Anal. Chem. 54:810-817,
     1982.

Lillian, D.  , and H. B. Singh.  Absolute determination of atmospheric halo-
     carbons by gas phase coulometry.  Anal. Chem. 46(8):1060-1063, 1974.

Lillian, D., H. B. Singh, A. Appleby, L. Lobban, R. Arnts, R. Gumpert,  R.
     Hague, J.  Toomey, J. Kazazis, M. Antell, D. Hansen, and B. Scott.
     Atmospheric fates of halogenated compounds.  Environ. Sci. Technol.
     9(12):1042-1048, 1975.

Lillian, D., H. B. Singh, and A. Appleby.  Gas chromatographic analysis of
     ambient halogenated compounds.  J. Air Pollut. Control Assoc.  26(2):
     141-143, 1976.

Lochner, F.   Perchloroethylene:  taking stock.  Umwelt 6:434-436, 1976.
     (English translation).
003PE4/B                             3-30                              11/22/83

-------
Love, 0.  T.,  and R.  G.  Eilers.   Treatment of drinking water containing trich-
     loroethylene and related industrial solvents.  J. Am. Water Works Assoc.
     74:413-425, 1982.

Lovelock, J.  E.   Atmospheric halocarbons and stratospheric ozone.  Nature
     252:292-294, 1974.

Lowenheim, F.  A., and M.  K. Moran.  Perchloroethylene.  In:  Faith, Keyes, and
     Clark's Industrial  Chemicals, Fourth edition, 1975.  pp. 604-611.

MacKay, D.,  A.  Bobra, D.  W. Chan, and W. Y.  Shiu.  Vapor pressure correlations
     for low-volatility environmental chemicals.  Environ. Sci. Technol.
     16:645-649, 1982.

Makide, Y.,  Y.  Kanai, and T. Tominaga.  Background atmospheric concentrations
     of halogenated hydrocarbons  in Japan.  Bull. Chem. Soc. Japan. 53:2681-2682,
     1980.

Mathias, E., E.  Sanhueza,  I. C. Hisatsune, and J. Heicklen.  The chlorine  atom
     sensitized oxidation and ozonolysis of C?C1...  Can. J. Chem. 52:3852-3862,
     1974.                                   *  ^

McConnell, G.,  D. M. Ferguson, and C. R. Pearson.  Chlorinated hydrocarbons
     and the environment.   Endeavor  34(121):13-18, 1975.

Melcher, R.  G. ,  R. R. Langner, and R. 0. Kage.l.   Criteria for the evaluation
     of methods for the collection of organic pollutants in air using solid
     sorbents.   Am.  Ind.  Hyg. Assoc. J. 39:349-361, 1978.

Mieure, J. P.   Determining  volatile organics in water.  Environ. Sci. Technol.
     14(8):930-935, 1980.

Monogue, W.  H.   Diss. Abstr. 18:984, 1958.

Murray, A. J.,  and J. P.  Riley.   The determination of  chlorinated aliphatic
     hydrocarbons in air,  natural waters, marine organisms, and sediments.
     Anal. Chim. Acta  65:261-270, 1973.

National  Institute for Occupational Safety and Health.  Manual of analytical
     methods, 2nd Edition,  Part II.  NIOSH Monitoring  Methods, Vol. 3,  April
     1977.

Ohta, T., M. Morita, and I. Mizoguchi.  Local distribution of chlorinated
     hydrocarbons in the ambient  air  in Tokyo.  Atmos.  Environ.  10:557-560,
     1976.

Ohta, T., M. Morita, I.  Mizoguchi, and T. Tada.   Washout effect and diurnal
     variation for chlorinated hydrocarbons  in ambient air.  Atmos. Environ.
     11:985-987, 1977.

Otson, R., and D. T. Williams.  Headspace chromatographic  determination of
     water pollutants.   Anal. Chem.   54:942-946,  1982.

Pasquini, D. A.   Evaluation of glass  sampling tubes for industrial breath
     analysis.   Am.  Ind.  Hyg. Assoc.  J.   39(l):55-62,  1978.

003PE4/B                              3-31                             11/22/83

-------
Pearson, C. R.,  and G. McConnell.  Chlorinated Cx and C2 hydrocarbons in the
     marine environment.  Proc. Soc. London B 189:305-332, 1975.

Pellizzari, E. 0.   Development of method for carcinogenic vapor analysis in
     ambient atmospheres.  EPA-650/2-74-121, July 1974.

Pellizzari, E. D.   Development of analytical techniques for measuring ambient
     atmospheric carcinogenic vapors.  EPA-600/2-75-075, November 1975.

Pellizzari, E. D.   The measurement of carcinogenic vapors in ambient atmos-
     pheres.  EPA-600/7-77-055, June 1977.

Pellizzari, E. D.   Measurement of carcinogenic vapors in ambient atmospheres.
     EPA-600/7-78-062, April 1978.

Pellizzari, E. D.   Information on the characteristics of ambient organic
     vapors in areas -of high chemical production.  Research Triangle Institute,
     Research Triangle Park, NC, 1979.   Available from U.S.  Environmental
     Protection Agency.

Pellizzari, E. D. ,  and J. E. Bunch.   Ambient air carcinogenic vapors:  improved
     sampling and analytical techniques and field studies.  EPA-600/2-79-081,
     May 1979a.   U.S. Environmental  Protection Agency.

Pellizzari, E. D.,  J. E. Bunch, R. E. Berkley, and J. McRae.  Collection and
     analysis of trace organic vapor pollutants in ambient atmospheres.   The
     performance of a Tenax GC cartridge sampler for hazardous vapors.   Anal.
     Lett.  9:45-63, 1976.

Pellizzari, E. D.,  M. D. Erickson, and R. A. Zweidinger.  Formulation of a
     preliminary assessment of halogenated organic compounds in man and
     environmental  media.  EPA 560/13-79-006.  U.S.  Environmental Protection
     Agency, July 1979b.

Ramsey, J.  D.  and R.  J. Flanagan.  Detection and identification of volatile
     organic compounds in blood by headspace gas chromatography as an aid to
     the diagnosis  of solvent abuse.  J. Chromat.  	:423-444, 1982.

Rasmussen,  R.  A.,  D.  E. Harsch, R. H. Sweany, J. P.  Krasnec, and D. R.  Cronn.
     Determination  of atmospheric halocarbons by a temperature-programmed gas
     chromatographic freezeout concentration method.  J. Air Pollut. Control
     Assoc. 27(6):579-581, 1977.

Russell, J. W.,  and L. A. Shadoff.  The sampling and determination of halo-
     carbons in ambient air using concentration on porous polymer.  J.  Chromat.
     134:375-384,  1977.

Schwarzenbach, R.  P., E. Molnar-Kubica, W. Giger, and S. G.  Wakeham.  Distribu-
     tion,  residence time, and fluxes of tetrachloroethylene and 1,4-dichlorobenzene
     in Lake Zurich, Switzerland.  Environ. Sci. Techno!. 13(11):1367-1373,
     1979.

Sheldon, L. S.,  and R. A. Hites.  Organic compounds in the Delaware River.
     Environ.  Sci.  Technol. 12(10):1188-1194, 1978.


003PE4/B                             3-32                             11/22/83

-------
Simmonds, P.  G.,  S.  L.  Kerrin, J. E. Lovelock, and F. H. Shair.  Distribution
     of atmospheric halocarbons in the air over the Los Angeles basin.  Atmos.
     Environ.   8:209-216, 1974.

Singh, H. B.   Phosgene in the ambient air.  Nature 264:(5585):428-429, 1976.

Singh, H. B.   Atmospheric halocarbons:  evidence in favor of  reduced average
     hydroxyl  radical concentration in the troposphere.  Geophys. Res. Lett.
     4(3):101, 1977.

Singh, H. B.   Personal communication, 1978.

Singh, H. B.,  D.  Lillian, and A. Appleby.  Anal. Chem.  47:860-864, 1975a.

Singh, H. B.,  D.  Lillian, A. Appleby, and L. Lobban.  Atmospheric formation of
     carbon tetrachloride from tetrachloroethylene.  Environ. Lett. 10(3):253-256,
     1975b.

Singh, H. B.,  L.  J.  Salas, and L. A. Cavanagh.  Distribution, sources, and
     sinks of atmospheric halogenated compounds.  J. Air Pollut. Control
     Assoc. 27(4):332-336, 1977b.

Singh, H. B.,  L.  Salas, D. Lillian, R. R. Arnts, and A. Appleby.  Generation
     of accurate halocarbon primary standards with permeation tubes.   Environ.
     Sci. Technol.  11:511-513, 1977c.

Singh, H. B.,  L.  Salas, H. Shigeishi, and A. Crawford.  Urban-nonurban relation-
     ships of halocarbons, SF6, N.^0, and other atmospheric trace constituents.
     Atmos. Environ. 11:819-828, 1977a.

Singh, H. B. ,  L.  J.  Salas, H. Shigeishi, and E. Scribner.  Global distribution
     of selected halocarbons, SF6, and N20.  Phase II  interim report.  SRI
     International, Menlo Park, California, May 1978b.

Singh, H. B.,  L.  J.  Salas, H. Shigeishi, and A. H. Smith.  Fate of halogenated
     compounds in the atmosphere.  Interim Report 1977, EPA-600/3-78-017,
     1978b.

Singh, H. B.,  L.  J.  Salas, H. Shigeishi, A. J. Smith,  E. Scribner, and L.  A.
     Cavanagh.  Atmospheric distributions, sources, and sinks of selected
     halocarbons, hydrocarbons, SF6 and N.^O.  EPA-600/3-79-107.  U.S.  Environ-
     mental Protection Agency.  November 1979.

Singh, H. B.,  L.  J.  Salas, R. Stiles, and H. Shigeishi.  Atmospheric measurements
     of selected hazardous organic chemicals.  Second year interim report.  SRI
     International, Menlo Park, CA.  1980.

Singh, H. B.,  L.  J.  Salas, A. J. Smith, and H. Shigeishi.  Measurements  of
     some potentially hazardous organic chemicals in urban environments.
     Atmos. Environ. 15:601-612, 1981.

Singh, H. B.,  L.  J. Salas, R. Stiles, and H. Shigeishi.  Measurements  of
     hazardous organic chemicals in the ambient atmosphere,  final  report.   Pre-
     pared for the U.S. EPA by SRI  International, Menlo Park, CA,  August  1982.


003PE4/B                             3-33                              11/22/83

-------
Snelson, A., R.  Butler, and F. Jarke.  Study of removal processes for halogenated
     air pollutants.  EPA-600/3-78-058.  Environmental Sciences Research
     Laboratory, U.S. Environmental Protection Agency, Research Triangle Park,
     N. C., 1978.

SRI International Chemical Economics Handbook, C-2 Chlorinated Solvents.
     (February, 1982)  p. 632, 3001 H.

Su, C.  W., and E.D.  Goldberg.  Environmental concentrations and fluxes of some
     hydrocarbons.   In: Marine Pollutant Transfer.  H. L. Windom, ed., 1976.
     pp 353-374.

Suffet, I. H., L. Brenner, and J. V. Radziul.  GC/MS  identification of trace
     organic compounds in Philadelphia waters, chap.  23.  In: Identification
     and analysis of organic pollutants in water, L.  H. Keith, ed.  Ann
     Arbor Science, 1977.

U.S. Environmental   Protection Agency.  Draft environmental impact statement:
     perchloroethylene - background  information for proposed standards.
     EPA-450/3-79-029a.  August 1979.

U.S. International   Trade Commission.   Synthetic organic chemicals, United
     States Production and Sales, 1980.

Wakeham, S.  G., A.   C. Davis, R. T. Witt, B. W. Tripp, and N. M. Frew.  Tetra-
     chloroethylene contamination of drinking water by vinyl-coated asbestos-
     cement pipe.   Bull. Environ. Contam. Toxicol. 25:639-645, 1980.

Williamson, 0.  G.,   and R. J. Cvetanovic.  Rates of reaction of ozone with
     chlorinated and conjugated olefins.  JACS 90:4248, 1968.

Yung, Y. L., M. B.   McElroy, and S. C.  Wofsy.  Atmospheric halocarbons:  a
     discussion with emphasis on chloroform.  Geophys. Res. Lett.   2(9):397-399,
     1975.
003PE4/B                              3-34                              11/22/83

-------
                       4.   ECOSYSTEM CONSIDERATIONS

4.1  EFFECTS ON AQUATIC ORGANISMS AND PLANTS
     Tetrachloroethylene (PCE) has been tested for acute toxicity in a limited
number of aquatic species.   The information presented in this chapter presents
observed levels  reported to result  in adverse effects under  laboratory condi-
tions.   It is recognized that such parameters of toxicity are not easily extra-
polated to environmental situations.   Test populations themselves may not be
representative of the entire species, in which susceptibility of various life-
stages to the test substance may vary considerably.  Guidelines for the utili-
zation of these  data in the development of  criteria  levels  for  PCE  in water
are discussed elsewhere (U.S.  EPA, 1979).
     The toxicity of PCE  to fish and other aquatic organisms has been gauged
principally by  flow-through  and static testing methods (Committee on Methods
for Toxicity  Tests  with Aquatic Organisms,  1975).   The flow-through method
exposes the organism(s) continuously to a  constant concentration of  PCE while
oxygen is continuously  replenished  and waste products are removed.  A static
test, on  the  other  hand, exposes  the organism(s)  to  the  added  initial concen-
tration only.   Both types  of  tests  are  commonly  used  as  initial  indicators  of
the potential of substances to cause adverse effects.

4.1.1  Effects on Freshwater Species
     Alexander  et  al.  (1978)  used  both flow-through (measured)  and  static
(unmeasured)  methods to investigate the acute  toxicity of four chlorinated
solvents,  including PCE,   to  adult   fathead  minnows  (Pimephales promelas).
Studies were  conducted in  accordance  with  test methods described  by  the
Committee on Methods  for Toxicity Tests with Aquatic Organisms (1975).
     The static  and  flow-through results for the 96-hour experiments  indicated
that PCE  was  the most toxic  of the  solvents  tested.   The lethal  concentration
(96-hour  LC50)  necessary  to kill 50 percent  of  the  fathead minnows  in  the
flow-through  test was 18.4 mg/1 (18.4 ppm); the 95 percent confidence limits
were 14.8  to  21.3 mg/1  (14.8  to 21.3 ppm).   In  comparison,  the static experi-
ments gave  a  96-hour LC50 of  21.4 mg/1 (21.4 ppm); the  95 percent confidence
limits were  16.5 to 26.4 mg/1  (16.5 to 26.4  ppm).  Fish affected during expo-
sure were  transferred to  static freshwater  aquaria  at  the end  of exposure.
Only  those  fish severely  affected  by  high concentrations did  not recover.
003PE1/D                           4-1                           11/22/83

-------
     When  the  minnows were exposed  to  sublethal  levels for short  exposure
intervals, only  reversible  effects  were observed.  Endpoints  evaluated were
loss of  equilibrium,  melanization,  narcosis,  and  swollen,  hemorrhaging gills.
The effective  flow-through  concentration (EC50) of PCE  that produced one or
more of these reversible effects was 14.4 mg/1 (14.4 ppm).
     The 96-hour LC50) in a static test with the bluegill (Lepomis macrochirus),
was reported as 12.9 mg/1 (12.9 ppm) (U.S. EPA, 1978,  1980).  The most sensitive
species tested is the rainbow trout  (Salmo gairdneri).  The LC5o determined by
a  flow-through measured  procedure was 5.28 mg/1 (5.28 ppm) (U.S. EPA, 1980).
With embryo-larval  test  procedures,  a chronic  value of  0.840 mg/1 (0.840 ppm)
was obtained by the U.S.  EPA (1980)  for the fathead minnow.
     With  the  freshwater invertebrate  Daphnia magna,  a 48-hour EC5(j value of
17.7 mg/1  (17.7  ppm) was obtained  (U.S.  EPA,  1980).   The  midge Tanytarsus
dissimi1 is was more resistant, with  a 48-hour  LC5o value of 30.84 mg/1 (30-84
ppm) determined under static, measured conditions.

4.1.2  Effects on Aquatic Plants
     As cited  in  the U.S.  EPA Ambient  Water  Quality  Criteria  Document (U.S.
EPA, 1980),  no adverse effects on chlorophyll  a or cell  numbers  of  the fresh-
water  alga Selenastrum capricornutum were  observed at exposure concentrations
as high as 816 mg/1 (816 ppm).
     For the  saltwater  species  Skeletonema costatum  a  96-hour EC50  of about
500 mg/1 (500 ppm) was determined for effects on chlorophyll a and cell  number.
This alga  species is more resistant  than the alga Phaeodactylum tricornutum for
which  the  EC50 value was determined to  be 10.5 mg/1  (10 ppm) (Pearson and
McConnell, 1975).

4.1.3  Effects on Saltwater Species
     Pearson and McConnell  (1975) investigated the acute  toxicity  of PCE on
the dab (Limanda 1imanda), barnacle  larvae (Barnacle nauplii), and on unicell-
ular algae (Phaeodactylum  tricornutum).   The LC50 was 5 mg/1  (5 ppm) for the
dab.  The  48-hour LC5o for barnacle  larvae was 3.5 mg/1  (3.5 ppm).
     Toxicity to the unicellular  alga was assessed by measuring alterations in
the uptake of  carbon from atmospheric  carbon  dioxide during photosynthesis.
                                                            14
Uptake of  carbon dioxide was measured by the use of sodium-  C-carbonate.   The
EC50 was 10.5 mg/1  (10.5 ppm).
003PE1/D                           4-2                           11/22/83

-------
     Data collected by  the  U.S.  Environmental  Protection Agency (1980) indi-
cate that, for  mysid  shrimp (Mysidopsis bahia),  the LC5u was 10.2 mg/1 (10.2
ppm) in a 96-hour static,  unmeasured procedure.   Chronic testing over the life
cycle of the mysid shrimp  resulted in a chronic value of 0.450 mg/1 (0.45 ppm)
(U.S. EPA, 1980).  The chronic value is 0.044 times the 96-hour LC5u.

4.2  BIOCONCENTRATION AND  BIOACCUMULATION
     An  indicator of  the potential  for  a substance  to  result  in cumulative or
chronic toxic effects in aquatic species is the bioconcentration factor (BCF).
Bioconcentration refers to  the  increased concentration of a substance within
an organism  (e.g.,  fish)  relative to the  ambient  water concentration under
steady-state conditions.  As defined by Veith et al. (1979), the bioconcentration
factor is a constant of proportionality between the concentration of the chemical
in fish and in the water.   This can be more clearly expressed as

                    Cf   Id
                    C~ = K~ = KRCF   at steady state.  (4-1)
                     w    ?

Bioaccumulation, a term often erroneously used in place of bioconcentration, can
be defined as  that  process which includes bioconcentration and any uptake of
toxic substances through consumption of one organism by another.  The BCF alone,
however, may not be the most useful measure of the overall fate of a substance
in water  or  of its  potential for producing toxic effects, for all chemicals.
      In  the absence  of direct  measurement,  a measure  commonly  used to assess
the degree to which a compound may be bioconcentrated is the octanol-water par-
tition coefficient.   Estimates for the octanol/water partition coefficient range
from 339 to 871  (Neely et al., 1974; U.S. EPA, 1980; Chiou et al., 1977).  The
partition coefficient has  been shown to be directly related to bioconcentration
potential in fish  (Neely  et al., 1974).  In guidelines recently set forth by
the American Society for Testing and Materials (ASTM), a log partition coeffi-
cient exceeding  a value of three was considered an  indication of a high proba-
bility of measurable bioaccumulation in aquatic species (ASTM, 1978).   Compounds
that exhibit a large  log coefficient generally are those  with low water solu-
bility and high  solubility in organic solvents.  Although a compound may demon-
strate a  high BCF or  log partition coefficient, other environmental factors that
act  to reduce this potential often exist.  The compound may be  rapidly hydrolyzed


003PE1/D                           4-3                            11/22/83

-------
or degraded by other mechanisms.  Measurable uptake by the organism may be pre-
cluded if the tissue depuration rate for the substance is great.
     With regard  to  PCE,  the BCF was calculated to be 34 and 49 in two fish
species (U.S. EPA, 1980;  Neely  et al.,  1974).  Neely et  al. (1974) found that
the BCF for PCE and other chemicals was linearly related to the respective parti'
tion coefficients.   For  PCE,  the log partition coefficient was 2.88,  and the
BCF, determined  in trout  (rainbow)  muscle, was 39.6 ±  5.5.   The trout were
exposed to  two  undefined  levels of PCE for an undefined period of time.   The
extent to which the levels approached the acute LC5U level for this species or
whether a steady-state was achieved was not reported.   Preliminary data in the
U.S. Environmental Protection  Agency  study (1980)  with  bluegill indicated a
BCF of 49.   The  log  partition coefficient was 2.53.   The depuration rate was
rapid, with a half-life of less than one day.
     Although these studies suggest that PCE does have bioconcentration poten-
tial, the extent  to  which this potential  can  be manifested in the form of
adverse effects  can  be gauged  only  from the results of toxicological studies.

4.2.1  Levels of PCE in Tissues of Aquatic Species
     Pearson and McConnell (1975) suggested that chronic and sublethal  effects
of PCE may  result from exposure to low concentrations  of PCE, if the halo-
carbon can  be bioaccumulated.   As a first step in  addressing the question of
bioaccumulation, these investigators determined levels of PCE  in a variety of
invertebrate and vertebrate species (Tables 4-1 and 4-2).
     Among marine  invertebrates,  wet  tissue concentrations of PCE were found
to range  from 0.001  to 0.009 ppm.  The highest concentration (0.008 to 0.009
ppm) found  was  in the crab  (Cancer pagurus).   Higher  levels  were found in
marine algae (0.013  to  0.022 ppm).   In tissues of  fish,  a range of 0.0003 to
0.041 ppm was found.   Concentrations  in the  livers of three species of fish
were found to greatly exceed those found in the flesh.   Tissue levels  from all
species are shown in Table 4-1.  Concentrations reported for fish-eating birds
and marine  mammals were  for selected tissues  such  as  fish liver, sea bird
eggs, and seal  blubber.   If the reported tissue concentrations for birds and
mammals are  converted  to  a whole-body  weight  basis, concentrations are much
lower and closer to concentrations measured  in  seawater,  indicating  little
or no bioconcentration and biomagnification (U.S.  EPA, 1981).
003PE1/D                           4-4                           11/22/83

-------
                           TABLE 4-1.   LEVELS  OF  PCE  IN  TISSUES  OF  MARINE  ORGANISMS,  BIRDS, AND MAMMALS
en
Species
Invertebrates
Plankton
Plankton
Ragworm (Nereis diversicolor)
Mussel (Mytilus edulis)


Cockle (Cerastoderma edule)
Oyster (Ostrea edulis)
Whelk (Buccinum undatum)
Slipper limpet (Crepidula
fornicata)
Crab (Cancer pagurus)
Shorecrab (Carcinus maenus)
Hermit crab (Eupagurus
bernhardus)
Source
Liverpool Bay
Torbay
Mersey Estuary
Liverpool Bay
Firth of Forth
Thames Estuary
Liverpool Bay
Thames Estuary
Thames Estuary
Thames Estuary
Tees Bay
Liverpoool Bay
Firth of Forth
Firth of Forth
Firth of Forth
Thames Estuary
Trichlorg-
ethylene
Tissue (ppm x 103)
0.05 - 0.4
0.9
Not detected
4 - 11.9
9
8
6 - 11
2
Not detected
9
2.6
10 - 12
15
12
15
5
PCE
(ppm x 103)
0.05 - 0.5
2.3
2.9
1.3 - 6.4
9
1
2-3
0.5
1
2
2.3
8-9
7
6
15
2
            Shrimp  (Crangon crangon)
Firth of Forth
16

-------
TABLE 4-1.   (continued)
Species
Starfish (Asterias rubens)
Sunstar (Solaster sp. )
Sea Urchin (Echinus esculentus)
Marine Algae
Enteromorpha compressa
Ulva lactuca
Fucus vesiculosus
Fucus serratus
Fucus spiral is
Fish
Ray (Raja clavata)
Plaice (Pleuronectes platessa)
Flounder (Platyethys f lesus)
Dab (Limanda limanda)

Source
Thames Estuary
Thames Estuary
Thames Estuary
Mersey Estuary
Mersey Estuary
Mersey Estuary
Mersey Estuary
Mersey Estuary
Liverpool Bay
Liverpool Bay
Liverpool Bay
Liverpool Bay
Trichloro-
ethylene
Tissue (ppm x 103)
5
2
1
19 - 20
23
17 - 18
22
16
flesh 0.8 - 5
liver 5-56
flesh 0.8 - 8
liver 16 - 20
flesh 3
liver 2
flesh 3-5
liver 12 - 21
PCE
(ppm x 103)
1
2
1
14 - 14.5
22
13 - 20
15
13
0.3 - 8
14 - 41
4-8
11 - 28
2
1
1.5 - 11
15 - 30

-------
                                          TABLE 4-1.  (continued)
Species
Mackerel (Scomber scombrus)

Dab (Limanda limanda)

Plaice (Pleuronectes platessa)
Sole (Solea solea)

Red gurnard (Aspitrigla
cuculus)
Scad (Trachurus trachurus)
Pout (Trisopterus luscus)
Spurdog (Squalus acanthi as)
Mackerel (Scomber scombrus)
Clupea sprattus
Cod (Gadus morrhua)


Sea and Freshwater Birds
Gannet (Sula bassana)

Source
Liverpool Bay

Redcar, Yorks
Thames Estuary
Thames Estuary
Thames Estuary

Thames Estuary

Thames Estuary
Thames Estuary
Thames Estuary
Torbay, Devon
Torbay, Devon
Torbay, Devon



Irish Sea

Tissue
flesh
1 iver
flesh
flesh
flesh
flesh
guts
flesh
guts
flesh
flesh
flesh
flesh
flesh
flesh
Air
bladder

1 iver
eggs
Trichloro-
ethylene
(ppm x 103)
5
8
4.6
2
3
2
11
11
6
2
2
3
2.1
3.4
0.8
<0.1


4.5 - 6
9-17
PCE
(ppm x 103)
1
not
5.1
3
3
4
1
1
2
4
2
1
Not
1.6
<0.
3.6


1.5
4.5

detected










detected

1



- 3.2
- 26
Shag (Phalacrocerax aristotelis)   Irish Sea
eggs
          2.4
1.4

-------
                                                      TABLE  4-1.   (continued)
00
Species
Razorbill (Alca torda)
Kittiwake (Rissa tridactyla)
Swan (Cygnus olor)

Moorhen (Gallinula chloropus)


Mallard (Anas platyrynchos)
Mammals
Grey Seal (Halichoerus grypus

Common Shrew (Sorex araneus)

Source
Irish Sea
North Sea
Frodsham Marsh

Merseyside

Merseyside
Fame Island
Frodsham Marsh
Tissue
eggs
eggs
1 iver
kidney
liver
muscle
eggs
eggs
blubber
1 iver
-
Trichloro-
ethylene
(ppm x 103)
28 - 29
33
2.1
14
6
2.5
6.2 - 7.8
9.8 - 16
2.5 - 7.2
3 - 6.2
2.6 - 7.8
PCE
(ppm x 103)
32-39
25
1.9
6.4
3.1
0.7
1.3 - 2.5
1.9 - 4.5
0.6 - 19
0 - 3.2
1
            Levels for trichloroethylene  included  for comparative  purposes.



           Source:  Pearson and McConnell, 1975.

-------
                     TABLE  4-2.   ACCUMULATION  OF  PCE BY  DABS
Period of
Tissue Exposure (days)
flesh 3-35
liver 3-35
flesh 3 - 35
liver 3-35
flesh 10
liver 10
Mean Exposure
Concentration (ppm)
0.3
0.3
0.03
0.03
0.2
0.2
Mean
Concentration in
Tissue (ppm x 100)
2,800a (13)
113,000 (14)
60 (9)
7,400b (9)
1,300 (7)
69,000 (7)
Accumu-
lation
Factor
x 9
x 400
x 5
x 200
x 6
x 350
 Numbers in parentheses are number of specimens  analyzed.
aOne fish had a flesh concentration of 29.7 ppm  and was  omitted from calculations.
 One fish had flesh concentration of 50.3 ppm and was  omitted from calculations.
Source:   Pearson and McConnell,  1975.

     The average concentration of PCE in seawater taken  from Liverpool  Bay,  an
area where  many species  of  organisms were collected, was  0.00012  ppm.   A
comparison of this value with those presented in Table 4-1 indicates an uptake
of as much as 75-fold.   It was the authors' contention that, based on their  ob-
servations, there is little indication that bioaccumulation occurs in the food
chain.
     As shown  in  Table 4-2,  dabs (Limanda 1 imanda) exposed to 0.3 ppm for 3
to  35  days  were found to  have  a  BCF (liver) for  PCE of  400.   It was not
reported whether  this  period  of exposure approximated a steady-state for PCE.
After dabs  were  returned  to clean seawater,  the  level of PCE dropped to 1/100
of  the  original  level  in 4 days and  to  1/1000 of  the initial  level after 11
days (Figure 4-1).  The ratio between liver and  flesh concentrations is approx-
imately 100  to  1.   The relationship between flesh  and  liver concentrations
in the dab is shown in Figure 4-2.
     Dickson and  Riley (1976) detected PCE  in three  species of mollusks and
in  five  species  of  fish  collected near  Port  Erin,  Isle of Man.  Levels of PCE
in  various  tissues  are shown  in Table 4-3.   Relative  to the  PCE concentration
in  seawater, there  was only a slight enrichment in the  tissues (< 25 times).
Tetrachloroethylene  had  one  of  the lowest  mean bioconcentration factors.
003PE1/D                           4-9                           11/22/83

-------
                100
              -fie
              Ul

              X.
              01  -
              O  1
                0.1
                                    d\     B
                   B^  -
O LIVER ACCUMULATION
0 LIVER LOSS
                   0          16         32
                        EXPOSURE TIME, diyt
Figure 4-1.  Accumulation and  loss of PCE by dabs.

            Source:  Alexander  et al., 1978.
003PE1/D
            4-10
11/22/83

-------
              100
           Q.
           a


           I
           in
           z
           ui
           UJ

           O
           oc
           O
oc
I-
III
t-
               10
               0.1
              0.01
                                I


                        EXPOSURE LEVELS, ppm

                               O  0.3

                               Q  0.2

                               A  0.02
                           AA
                                 I
                                10              100


                         TETRACHLOROETHYLENE IN LIVER, ppm
                                                    1000
   Figure 4-2.   Relation between flesh and  liver concentration of PCE  in  dabs.



                 Source:   Alexander et al.,  1978.
003PE1/D
                          4-11
11/22/83

-------
    TABLE 4-3.   CONCENTRATION OF PCE AND TRICHLOROETHYLENE IN MOLLUSKS AND
                           FISH NEAR THE ISLE OF MAN
Species
Eel (Conger conger)
brain
gin
gut
1 iver
muscle
Cod (Gadus morhua)
brain
gill
heart
1 iver
muscle
PCE
(mg x

6
2
3
43
1

3
3
8
2
Trichloroethylene
106/g dry weight tissue)

62
29
29
43
70

56
21
11
66
8
     skeletal tissue
     stomach

Coalfish (Pollachius birens)

     alimentary canal
     brain
     gill
     heart
     1 iver
     muscle

Dogfish (Scyl1iorhinus canicula)

     brain
     gill
     gut
     heart
     liver
     muscle
     spleen

Bib (Trisopterus luscus)

     brain
     gut
     1 iver
     muscle
     skeletal tissue
         6
         2
        12
        13
         4
         0.3
                              306
                               71
 70
  8
 40
176
 41
274
479
 41
307
143
187
185
003PE1/D
4-12
11/22/83

-------
                            TABLE  4-3.   (continued)
                                         PCE
Species
                                                         Trichloroethylene
                                           (mg x
                                                       dry weight tissue)
Baccinum undatum
     digestive gland
     muscle
Modi ol us modi ol us
                                           33
                                           39
digestive tissue
mantle
muscle
Pecten maximus
gill
mantle
muscle
ovary
testis
-
63
16

88
40
24
-
176
56
250
33

detected
-
-
-

Source:   Dickson and Riley, 1976.

4.3  BEHAVIOR IN WATER AND SOIL
     The potential of any substance for bioconcentration is influenced by many
factors, including the rate at which it volatizes and its reactivity.
     In the  laboratory study by Oil ling et al. (1975), the measured half-life
of PCE ranged from 24 to 28 minutes in water.   Factors affecting the evaporation
of PCE  were  surface  wind speed,  agitation of  the  water,  and water and  air
temperatures.   Reactivity  of  PCE in water was measured by exposing  sealed
quartz  tubes  containing  1  ppm  PCE to sunlight  for  one year.  At 6 months, the
level of PCE had declined to 0.52 ppm,  and at 1 year, to 0.25 ppm.   Oil ling et
al.  (1975)  reported that  the  presence of 3 percent NaCl  (as  in  seawater)
caused  about  a  10 percent decrease in the evaporation  after 40 percent had
already evaporated.  The  addition  of  500 ppm  clay appeared  to increase the
rate of disappearance  to 85 percent soluble depletion  at  20 minutes.  These
experiments were conducted to simulate the evaporation of PCE under conditions
more nearly  like  those found in the environment.    Evaporation  of PCE  from the
hydrosphere is a rapid process.
003PE1/D
                                   4-13
                                                                 11/22/83

-------
     The field  studies  of  Zoeteman et al. (1980)  suggest  that PCE is more
persistent in natural water environments  than is indicated by  laboratory mea-
surements.   In a field study of the persistence  of a variety of organic chemi-
cals  in  different  aquatic  environments in the  Netherlands,  Zoeteman  et al.
(1980) estimated  the persistence of  PCE  in  river  water from  3 to  30 days
(half-life).   In lakes and groundwaters,  the half-life was  estimated at 10-fold
higher.   Estimates  were  derived from monitored values  of  samples  collected
between two sites along the Rhine River,  into which no discharges  were expected.
PCE was analyzed by GC-MS.
     Estimates  of  the persistence  of PCE in rivers, lakes, and  ponds,  by
calculation according to Smith  et al. (1980) are  in  general  agreement with
the field  results  of Zoeteman and coworkers (1980).  The half-life of PCE is
obtained from the expression:

                                  t1/2  =  0.693.                         (4-2)
                                             kv

                  where kc is the volatilization rate constant.

     Using the  data  provided  by Smith et al. (1980),  the  tl,2 (days) is as
follows:   ponds, 9 to 20; lakes, < 1 to 20;  rivers, < 1 to  20.
     Bouwer et  al.  (1981)  found that PCE and other halogenated organics have
the potential  to leach rapidly through soil.   When secondary treated municipal
wastewater containing from  1  to 10 ng/1  PCE was applied to soil columns,  at
rates typical of  high-rate  land application systems and under conditions  in
which volatilization was prevented, PCE was  detected in the effluent.   Leaching
of PCE through  soil  was suggested by  Zoeteman  et  al.  (1980) as a  probable
factor in the contamination of groundwater supplies in the  Netherlands.
     The potential  for halogenated organics, including  PCE,  to  contaminate
groundwater supplies via leaching from surface waters was examined by Scwarzen-
bach and Westall  (1981).   In batch and column experiments  with various types
of sorbents and organics designed to simulate field conditions, these investi-
gators found  that  the partition coefficient for a particular compound can be
estimated  from  its  octanol/water partition  coefficient and from the fraction
of organic carbon  in the sorbent.  A  high  degree  of correlation  was  found
between the partition coefficient and organic carbon content when  the fraction
003PE1/D                           4-14                          11/22/83

-------
of organic carbon  was  greater than 0.1 percent.  A partition coefficient of
0.56 ± 0.09 was found  for PCE, using natural aquifer material (organic carbon
= 0.15 percent) from a field  site  in Switzerland.  It was concluded that, for
concentrations typically encountered in  natural  waters,  sorption of PCE and
other organics of comparable 1ipophilicity by aquifer materials  is reversible.
The expression

                                   S =  kpC,                           (4-3)
                  where S = concentration in solid phase
                         kp = partition coefficient
                         C = concentration in liquid phase

was found satisfactory to describe sorption equilibrium.

4.4  SUMMARY
     The available data  for PCE  indicate that acute and chronic  toxicity to
freshwater aquatic life  can occur  at concentrations around 5280 and 840 ug/1,
respectively.   For saltwater aquatic life, the acute and  chronic toxicity values
are 10,200 and 450 ug/1,  respectively.
     Tetrachloroethylene does  not  appear  to biomagnify or concentrate as it
moves up the food chain.   The available data suggest that the bioconcentration
potential of  PCE  is  low, and  it  appears to  be eliminated rapidly  from aquatic
organisms.
     Contamination of  groundwater  supplies  by PCE  leaching through soil could
be a  concern,  particularly  in situations  in which  soils of low organic carbon
content are involved.
003PE1/D                           4-15                          11/22/83

-------
   4.5  REFERENCES

   Alexander, H.  C.,  W.  M. McCarty, and E. A. Bartlett.  Toxicity of perchloro-
        ethylene, trichloroethylene, 1,1,1-trichloroethane, and methylene chloride
        to fathead minnows.   Bull.  Environ. Contain. Toxicol. 20:344-352, 1978.

   American Society for Testing and Materials.  Estimating the hazard of chemical
        substances to aquatic life. J.  Gavins, K.  L. Dickson, and A. W. Maki,
        eds. , STP 657.   Committee D-19 on Water, 1978.

   Bouwer, E.  J., P.  L.  McCarty, and J. C. Lance.   Trace organic behavior in  soil
        columns during rapid infiltration of secondary wastewater.  Water Res.
        15(1):151-160,  1981.

   Chiou, C.  T.,  V.  H.  Freed, D. W. Schmedding, and R. L. Kohnert.  Partition
        coefficient and bioaccumulation of selected organic chemicals.  Environ.
        Sci.  Technol.   11:475-478,  1977.

   Committee on Methods for Toxicity Tests with Aquatic Organisms:  methods  for
        acute toxicity tests with fish, macroinvertebrates, and amphibians.    Ecol.
        Res.  Series,  EPA 600/3-75-009,  1975.

   Dickson, A.  G., and J.  P. Riley.  The distribution of short-chain halogenated
        aliphatic hydrocarbons in some marine organisms.   Marine Pollut. Bull.
        7(9):167-169, 1976.

   Dilling, W.  L., N.  B. Tefertiller, and G.  J. Kallos.  Evaporation rates and
        reactivities  of methylene chloride, chloroform, 1,1,1-trichloroethane,
        trichloroethylene, tetrachloroethylene, and other chlorinated compounds
        in dilute aqueous solutions.  Environ. Sci. Technol. 9(a):833-838, 1975.

   Neely, W.  B.,  D.  R.  Branson, and G.  E.  Blau.  Partition coefficient to
        measure bioconcentration potential of organic chemicals in  fish.
        Environ.  Sci.  Technol. 8:1113,  1974.

   Pearson, C.  R. , and G.  McConnell.  Chlorinated Cx and C^ hydrocarbons in
        the marine environment.  Proc.  Roy. Soc. London B 189:305332, 1975.

   Schwarzenbach, R.  P.  and J. Westall.  Transport of nonpolar organic compounds
        from surface  water to groundwater.  Laboratory sorption studies.  Environ.
        Sci.  Technol.  15:1360-1367, 1981.

   Smith, J.  H.,  D.  C.  Bomberger, Jr.,  and D. L. Haynes.   Prediction of the
        volatilization rates of high-volatility chemicals from natural water
        bodies.   Environ.  Sci. Technol. 14(11):1332-1337, 1980.

   U.S.  Environmental Protection Agency.   In-depth studies on health and environ-
        mental  impacts of selected water pollutants.  Contract No.  68-01-4646,
        Duluth, MN, 1978.

   U.S.  Environmental Protection Agency.   Tetrachloroethylene:  water quality
        criteria.  Federal Register 44(52):15966-15969, 1979.
003PE1/D                           4-16                          11/22/83

-------
   U.S.  Environmental  Protection Agency.   Tetrachloroethylene:   ambient water
        quality criteria.   Office of Water Regulations and Standards, EPA
        440/5-80-073,  October 1980.

   U.S.  Environmental  Protection Agency.   Environmental  risk assessment of tetra-
        chloroethylene,  draft report.   Office of Toxic Substances.   14 September,
        1981.

   Veith,  G.  D.,  D.  L.  DeFoe, and B.  V.  Bergstedt.   Measuring and estimating the
        bioconcentration factor of chemicals in fish.   J.  Fish.  Res.  Board Canada
        36:1040-0145,  1979.

   Zoeteman,  B.  C.  J.,  K.  Harmsen, J.  B.  H.  J.  Linders,  C. F. H.  Morra, and
        W.  Slooff.   Persistent organic pollutants in river water and ground
        water of the Netherlands.   Chemosphere 9:231-249,  1980.
003PE1/D                           4-17                          11/22/83

-------
            5.   COMPOUND DISTRIBUTION AND RELATED  PHARMACOKINETICS

5.1  HUMAN AND ANIMAL STUDIES
5.1.1  Absorption
5.1.1.1  Pulmonary--Inhalation is the  principal  route by which tetrachloro-
ethylene (PCE) enters the  body.   During inhalation, PCE is  absorbed  by the
blood  via  alveolar air.   The principal  approaches  used  in calculating the
amount absorbed are  those  involving  serial  breath analysis, i.e., measuring
the amount exhaled.
     The magnitude of PCE uptake  into the body (dose,  burden) depends  primari-
ly on  several parameters:  inspired  air  concentration, pulmonary ventilation,
duration of exposure, and  the rates  of diffusion into and  solubility in blood
and the various tissues.   The concentration of PCE in  alveolar air,  in equili-
brium with pulmonary  venous  blood,  approaches a minimum difference with the
concentration in the inspiratory  air until  a steady-state condition  is reached.
After  tissue and  total body  equilibrium  is  reached during  exposure, uptake  is
balanced by  elimination through  the lungs and by  other  routes,  including
metabolism.  The difference between alveolar and inspiratory air concentrations,
together with the  ventilation  rate  (about 6 1/min at  rest), provides  a means
of calculating uptake during exposure:

                           Q = (Cins - Calv> * ' T                    <5-D

where Q is the quantity absorbed; C is the air concentration in milligrams per
liter; V  is  the  alveolar ventilation rate in liters per minute; and T is the
duration of exposure  in  minutes.   The percent retention is  defined as  (C.
C i )/C.   x 100,  and percent retention  x quantity  inspired  (V  • T  • C.   )  is
equal to uptake.
     In serial determinations of PCE in alveolar breath and in blood, during
and following exposure (1, 3, 5.5, and 7.5 hr/.day) of  males and females to 25,
50, 100, and 150  ppm  (170, 339,  678, and 1017 mg/m3), Hake  et al. (1976) con-
cluded that the  compound was rapidly absorbed  and  rapidly excreted via the
lungs.  The amount absorbed  at a given vapor  concentration  was  reported to  be
related to the respiratory minute volume.  The minute  volume is defined as the
product of the  tidal volume and  the  respiratory  frequency over a 1-minute
period.

003PE1/E                             5-1                              11/22/83

-------
     In a  study  by  Monster  et  al.  (1979),  six male volunteers were exposed  in
a chamber  for  4  hours to 72 ± 2 ppm PCE  (488 ± 13 mg/nr1) and to 144 ± 7 ppm
PCE (977 ± 47 mg/m3) while at rest.  Absorption of PCE by the lungs was reported
to decrease with continuation  of exposure  (p <0.05); approximately 25 percent
less PCE was  absorbed in the  fourth hour  as compared with the first hour of
exposure.   The effects of a workload (bicycle ergometer) were determined in a
separate exposure of the volunteers to 142 ±  6 ppm PCE (963 ±  41 mg/ma);
individuals exercised for  two  30-minute periods  during  the  4-hour exposure
period.  A 2-week interval  occurred between each exposure mode.   During each
mode,  the  individuals inhaled  vapors  through a gas mask and exhalations were
                  ®
made into a Tedlar  bag.
     The total uptake, shown  in  Table 5-1, was influenced more by lean body
mass than  by  respiratory minute  volume or adipose tissue.   Uptake is defined
as the amount of  PCE absorbed per minute.   The inter-individual  coefficient  of
variation of body burden predicted by measurements of PCE in exhaled air or  in
blood  was  about  25  percent.  The individual uptake at 144 ppm PCE  (977 mg/m3)
was 2.1 times  higher  than that at  72 ppm  (488 mg/m3) when individuals were at
rest.  During  exercise,  total  uptake increased  about 40  percent; exercise had
no effect  on the half-life  of  elimination  or on the rate constant  of elimina-
tion.  Minute volume was three-fold higher than that observed when the indivi-
duals  were at  rest.   An  alveolar  retention of  approximately 60  percent was
calculated at  the end of the 4-hour exposure period.  Alveolar retention is
defined as the percentage difference between the amount of PCE inhaled and the
amount exhaled.
     Monster (1979)  suggested  that the decrease in absorption (declining lung
clearance) as  exposure continued  reflected the small  degree to which PCE was
metabolized,  in  contrast to trichloroethylene,  which has a more constant rate
of absorption  but is  metabolized to a much greater degree.   There  would be no
reason to  expect that chronic  exposure to environmental levels of PCE would
not follow the  patterns  of absorption and lung clearance observed by Monster
and coworkers.   In  the  linear  pharmacokinetic  range, these  parameters would
remain proportional  to  the  exposure  concentration down  the dose-response
curve.
5.1.1.2  Percutaneous--Under  most  circumstances  of  use, absorption of PCE
through the skin is of minor consideration.  Stewart and Dodd (1964) observed
absorption in each of five individuals after one thumb of each was immersed in

003PE1/E                             5-2                              11/22/83

-------
          TABLE 5-1.   ESTIMATED UPTAKE OF  SIX INDIVIDUALS  EXPOSED TO
           TETRACHLOROETHYLENE WHILE AT REST AND AFTER REST/EXERCISE

              Uptake,  according to TCE exposure
              	concentration	
                                142 ppm              Lean  body  Minute volume
           72 ppm    144 ppm   (rest and  Body mass,,    mass,       at rest,
Subject  (at rest)  (at rest)  exercise)      kg         kg         1/min
A
B
C
D
E
F
370
490
530
500
390
450
670
940
1000
1210
880
970
1060
1500
1400
1510
1320
1120
70
82
82
86
67
77
62
71
71
74
61
61
7.6
11.6
10.0
11.3
12.3
8.8
Source:   Monster, 1979.

a beaker of PCE, which was located in a ventilated hood.   By measuring concen-
trations in alveolar breath samples,  the investigators concluded that there is
little  likelihood that toxic amounts of PCE will be absorbed through the skin
during normal use or exposure to the  compound.
     Riihimaki and  Pfaffli  (1978)  concluded  that PCE concentrations found in
the work environment were  not  likely to result  in a significant amount of  PCE
being absorbed  through the skin.   In their experiment, three individuals who
wore full facepiece respirators to prevent pulmonary absorption and were dressed
in thin  cotton  pajamas and socks were  exposed to 600  ppm PCE (4069  mg/m3)  for
3.5 hours.   During each midhour, for  a period of 10 minutes, each person exer-
cised on a  bicycle  ergometer.   The periodic exercise  was employed  to simulate
work conditions.  Tidal and alveolar air  (mixed) were collected in  polyester-
lined polyethylene  bags.   Concentrations  of  PCE in blood and  in exhaled air
were determined  up  to 20  and  50  hours,  respectively.  The amount  absorbed
(assuming 98  percent  is  exhaled)  was  calculated at  7 ppm PCE (47.6 mg/m3).
     Jakobsen et  al.  (1982) found that PCE was absorbed through the skin  of
anesthetized guinea pigs after epicutaneous exposure.   A maximum blood concen-
tration  was  observed  after 1 hour, followed by  a  rapid decrease during the
003PE1/E
5-3
11/22/83

-------
rest of the exposure time.  PCE was applied to a clipped area of the skin and
sealed under a glass ring.
     Kronevi et al. (1981) found that PCE, when applied to the skin of guinea
pigs, caused  considerable local  skin changes within  15  minutes  of topical
exposure.    Jakobson  et al.  (1982) suggested  that  these  skin changes might
reflect a defense reaction against penetrating agents.

5.1.2  Distribution
     Tetrachloroethylene  is believed  to partition in body tissues  having  a
high  lipid  content (Stewart,  1969).   This site probably  accounts  for  the
prolonged retention of  PCE.   Such  retention in adipose tissue is suggested by
the blood/air and  fat/blood partition coefficients of 16 and 90, respectively
(Monster, 1979). However,  the time needed to saturate adipose tissue  to 50
percent of its equilibrium concentration is high,  about 25 hours.
     In an abstract, ,Hake et al.  (1976) reported that "the solvent was  rapidly
absorbed and excreted  via  the lungs with a small  portion accumulating  in the
body which  was  slowly  excreted."   Male  and female volunteers were exposed for
1.3, 5.5, and 7.5  hr/day to 25,  50, 100, and 150 ppm PCE (170,  339, 678, and
1017 mg/m3).
     Because of selective partitioning of PCE in adipose tissue,  a long  period
(about 2 weeks)  is necessary  to eliminate PCE from the body completely  upon
cessation of exposure  (Monster et  al.,  1979;  Fernandez et  al., 1976).  Uptake
of  PCE by adipose  tissue will take place during repeated  exposures until an
equilibrium is reached; further exposure will  not result in further accumulation
(Schumann et al., 1980).
     Guberan and  Fernandez (1974)  used a mathematical  model  to  calculate
uptake and  distribution of PCE in the  body,  and  they predicted  that fatty
tissues would  show the  slowest  rate  of PCE depletion because of  the  high
solubility  of PCE  in  fatty tissues.  Serial  breath concentration decay  data
obtained from 25  volunteers  exposed to between 50  and  150 ppm PCE (339 and
1017 mg/m3) for  up to  8 hours were used in developing the  model.   As shown in
Figure 5-1,  theoretical curves of concentration in alveolar air (C , )  divided
by  the concentration  in inspired air (C.  ) versus exposure time for various
post-exposure times can be used to estimate unknown concentrations to which an
individual may be exposed.
     Savolainen et  al.  (1977) found that exposure of two  strains of rats to
200 ppm PCE (13,560 mg/m3) for 6 hr/day for 4 days resulted in partitioning of
003PE1/E                             5-4                              11/22/83

-------
      a
      I/I

     o"
      >
      •3
     o
                           345678

                          DURATION OF EXPOSURE, hours
Figure 5-1.
003PE1/E
Predicted post-exposure alveolar  air  concentrations  of  tetrachloro-
ethylene at various times  against duration  of  exposure.
Source:   Guberan and Fernandez,  1974.


                        5-5
11/22/83

-------
PCE in perirenal fat, brain, and liver.   Statistical analyses were not performed.
When behavioral patterns were observed subsequent to the last exposure, ambula-
tion  frequency  of exposed  rats  increased (p <0.05)  compared  to controls.
Rearing frequency, defecation and  urination frequency, and preening frequency
were not affected by exposure.

5.1.3  Metabolism (Animal Studies)
     The hepatotoxic,  carcinogenic,  and  mutagenic potentials of  a number of
chlorinated  ethylene  compounds  (U.S. Department of Health,  Education, and
Welfare, 1977a,b,c; Viola et al.,  1971;  Maltoni  and Lefemine, 1974;  Creech and
Johnson, 1974; Waxweiler et al., 1976) have generated considerable interest in
the metabolic pathways of these compounds.   Certain relatively inert chemicals
may be activated by biotransformation to carcinogenic intermediate metabolites
that  could  have the  potential  to  induce  a carcinogenic  lesion.   Thus, the
relationship of the metabolism of the various chlorinated ethylenes,  including
PCE, to their toxicity is an important consideration.
     The cytochrome P-450 dependent mixed-function oxidases of mammalian liver
microsomes have been  demonstrated  to  oxidize the carbon-carbon double  bond in
olefins to an epoxide ring (Liebman and Ortiz,  1968; Watabe and Maynert, 1968;
Liebman and  Ortiz, 1970; Maynert et al., 1970).   The  stability of the  epoxide
ring  varies,  depending  on  the  configurations of the oxirane compound.   Thus,
this  activated  intermediate metabolite may interact covalently with a  variety
of  cellular  macromolecules.   If the  chemical  interacts  with nucleic  acids
and/or  proteins  that are essential to  cellular  function,  the reaction may
result  in alteration  of cellular  metabolism, resulting in cellular necrosis,
or  in carcinogenic or mutagenic lesions (Jerina and Daly, 1974).
     The formation of an epoxide  intermediate for  a  chloroethylene compound
was originally  postulated by Powell  in 1945.  Later,  Yllner  (1961) and Daniel
(1963)  speculated  that  PCE might be oxidized to  an  epoxide as an  intermediate
metabolite during  its biotransformation.   Recent interest in this hypothesis
has resulted from findings  that vinyl chloride  is  carcinogenic   in man and
animals (Viola  et  al.,  1971;  Creech and Johnson, 1974; Maltoni and Lefemine,
1974;  Lee and Harry,  1974)  and the observation  that  this  carcinogenicity is
probably caused by formation of an epoxide intermediate,  chloroethylene oxide.
This mechanism was proposed by van Duuren  in 1975.  Tetrachloroethylene epoxide,
as  well  as   other  chloroethylene  epoxides, have  been synthesized i_n  vitro

003PE1/E                             5-6                              11/22/83

-------
(Kline et al.,  1978;  Kline  and van Duuren,  1977;  and Derkosch,  1976).   These
epoxides are unstable (have  short half-lives) and  are highly reactive.
                                              14
     Yllner (1961) studied  the  metabolism of   C-labeled PCE in mice exposed
for 2 hours by  inhalation to doses of 1.3 mg/g.  Seventy percent of the ab-
sorbed radioactivity was expired in air,  20 percent was excreted in the urine,
and less than 0.5 percent was eliminated  in the feces.   Of the total amount of
radioactivity excreted in the urine, 52 percent was identified as trichloroace-
tic acid, 11 percent  as oxalic  acid, and  a trace was dichloroacetic acid.  No
radiolabeled monochloroacetic  acid, formic  acid,  or  trichloroethanol  was
detected.  Of the  remaining radioactivity in the urine,  18 percent was not
extractable with ether, even after hydrolysis.
                        14
     Daniel  (1963) fed    C-labeled PCE to rats and  found that  excretion was
largely of unchanged compound through the lungs (half-time of expiration was 8
hours).   Only 2  percent of  the radioactivity was excreted  in the urine, and
equimolar proportions  of  trichloroacetic  acid and inorganic  chloride were  the
only metabolites detected.
     Other  investigators  (Ogata et al.,  1971;  Ikeda  and  Ohtsuji, 1972;  Ikeda,
1977; Moslen et  al. ,  1977;  Dmitrieva,  1967; Tada and Nakaaki, 1970; Boillat,
1970; Barchet et al., 1972) have reported that trichloroacetic acid (TCA)  is a
urinary metabolite of PCE in experimental animals and humans.
     The most likely  pathway for  such  product  formation  would be via epoxida-
tion of the double bond.  The resulting chloro-oxirane compound is known to be
unstable and rearranges spontaneously.   However,  the  stability  of  symmetric
oxiranes such as the one  formed from PCE  is greater than that of the asymmetric
oxiranes such as those formed  from vinyl  chloride,  vinylidene  chloride, and
trichloroethylene.  Henschler and  his colleagues (Bonse et al., 1975; Greim et
al. , 1975; Henschler et al., 1975; Bonse  and Henschler, 1976) have studied the
chemical reactivity,  metabolism,  and mutagenicity of the chlorinated ethylene
series,  including  vinyl chloride,  trichloroethylene,  and tetrachloroethylene.
These investigators have  reported  a correlation between biological activity and
chemical structure:   those  chlorinated ethylenes that  are symmetrical,  such as
cis-  and trans-l,2-dichloroethylene and  PCE, are  relatively stable and not
mutagenic.  In contrast,  the asymmetrical ethylenes, vinyl chloride, vinylidene
chloride, and trichloroethylene,  are unstable and mutagenic.  Although these
investigators recognized  that  oxiranes (epoxides)  may be formed by  all  six of
the  chlorinated  ethylenes,  they concluded that the  asymmetrical oxiranes  are

003PE1/E                             5-7                              11/22/83

-------
far less stable than the symmetrical ones, and therefore are more highly elec-
trophilic  and  may react  directly  with  nucleophilic  constituents  of  cells more
readily, thereby  exerting  mutagenic or carcinogenic effects.  The results of
the mutagenic  tests conducted  by these  investigators  correlate with this
structure-activity relationship.
     Evidence  for the involvement of  the microsomal  mixed-function oxidase
system  in  the  metabolism of  PCE was  shown by  Moslen et  al.  (1977).   Rats that
were pretreated with phenobarbital or Arochlor-1254 (polychlorinated biphenyls),
which  are  inducers of the hepatic  mixed-function oxidase system,  showed  a
significant  increase  in total  trichlorinated urinary  metabolites  and TCA
excretion  following a  single oral administration of 0.75 ml/kg PCE.  Hepato-
toxicity of  PCE was  enhanced by  Arochlor-1254 pretreatment, as  evidenced  by
doubling of  serum glutamic-oxaloacetic  transaminase (SCOT)  levels,  and by  the
appearance of  focal areas  of vacuolar  degeneration  and  necrosis  of  the liver.
However, Cornish  et al.  (1973)  did  not  observe a  potentiation  of  PCE  toxicity
following  intraperitoneal injection of 0.3 to 2.0 ml/kg PCE to rats pretreated
with phenobarbital.   (The  LD50  for the mouse is 2.9 ml/kg [Klaasen and Plaa,
1966]).  However,  elevation   of SCOT was  noted at  all  dose  levels  in this
study.
     Costa and Ivanetich's  recent  j_n  vitro  studies  (1980)  concerning the
extent  of  metabolism  of  PCE   provide further  evidence of  the central  role  of
hepatic microsomal cytochrome P-450 in the metabolism  of PCE.   Two distinct
forms of P-450, one  induced  j_n vivo in  rats  by phenobarbital  and the other
induced by pregnenolone-16«-carbonitrile,  were  shown  to be  active  in the  i_n
vitro binding  and metabolism of  PCE.  P-448,  induced by B-napthoflavone, was
not active in the j_n vitro system.
     Spectral binding of PCE  to P-450 in microsomes was measured by the differ-
ence in absorbance between 386 nm and  418 nm.   Metabolism was estimated j_n
vitro by the maximal  rate  of production (V   ) of TCA per minute  per  nmole of
	                                      max
P-450.   The spectral binding  constant (K  ) associated with P-450 from uninduced
rats was 0.4 mM.   Phenobarbital-induced P-450 had no effect on K , but P-450
from pregnenolone~16«-carbonitrile-induced rats increased K .
     V     was  significantly   increased  (P <0.01) increased  above controls
      max
(uninduced rats)  when  the  microsomal preparation was derived  from  either  of
the inducers.  Phenobarbital   induction resulted in a lowering of the Michaelis-
Menton  (K  )  constant, indicating an  increased  affinity for the  associated

003PE1/E                             5-8                              11/22/83

-------
enzymes.   The K  was increased when the microsomes were derived from pregneno-
lone-16<*-carbonitrile induction.  The  P-448  inducer,  p-napthoflavone,  had no
effect on any of these parameters.
     The rate  of  carbon  monoxide-inhibitable NADPH oxidation by PCE was ele-
vated  significantly  (P<  0.01)  above  noninduced controls by phenobarbital-
induced  microsomes.   Induction  by  pregnenolone-16«-carbonitrile was without
effect in  this  system.   Metyrapone,  a specific inhibitor of P-450,  reduced
spectral binding,  NADPH  oxidation, and  metabolism by  PCE  80, 65, and 75
percent, respectively.   SKF-525A reduced  spectral binding and metabolism 19
and 33  percent,  respectively.   The microsomal preparation used in the system
was derived  from  phenobarbital-treated rats.   The major metabolite in the j_n
vitro  system was  observed  to be unbound TCA.  Trichloroethanol was not found
in  appreciable  amounts.   Levels of P-450, as  well  as cytochrome b5,  heme,
NADPH-cytochrome reductase, and glucose-6-phosphatase, were not reported to be
altered, whether the inducer was phenobarbital or pregnenolone-16<*-carbonitrile.
     Kaemmerer et  al.  (1982)  reported that feeding rats  25  ppm PCE in feed
resulted in  a  statistically significant induction of cytochrome P-450 in rat
liver  homogenates 14 days after administration, compared to untreated controls.
When 500 ppm was  used,  the increase occurred after 7 days.   The authors also
reported that  blood  coagulation was impaired in rats.  The authors concluded
that PCE interferes  with basic  physiologic functions,  particularly cell meta-
bolism.
     Takano  and  Miyazaki  (1982) reported that  the  PCE inhibited glutamate,
succinate,  and malate oxidation in the presence of rat liver mitochondria.  The
lesser inhibition of succinate oxidation suggested to the authors that PCE may
act as an  uncoupler  in the electron transport process between  NADH and coenzyme Q.
Ogata  and  Hasegawa (1981)  have reported similar  findings  for PCE and other
halocarbons  in succinate oxidation by rat liver mitochondria.
     The extent of metabolism of PCE and its binding to hepatic macromolecules
in  mice  (B6C3F1) and rats (Sprague-Dawley) was investigated by Schumann et al.
(1980).  Animals  were  exposed for  6  hours via inhalation to  10 and 600 ppm
14C-labeled  PCE  (67.8  and  4068 mg/m^).  Purity was determined to  be greater
than 98  percent.   Following inhalation of  10 ppm  (67.8 mg/m3)  14C-labeled PCE
by  mice, urinary  metabolites  were  observed to account for 62.5 percent of the
total  radioactivity  recovered, though  unchanged PCE in expired air accounted  for
only 12.0  ±1.3  percent.   The reverse  situation was observed after a  single

003PE1/E                             5-9                               11/22/83

-------
oral dose of 500 mg  14C-labeled  PCE/kg body weight: 82.6 percent of the total
recovered radioactivity was in the form of unchanged PCE in expired air in the
mouse.  This shift  in  the mouse in the major route  of elimination demonstrates
a saturation of oxidative metabolism.  Metabolites accounted for 10.3 percent.
When compared  to the  extent of metabolism  in  rats  (Pegg et  al., 1979), it was
calculated that the mouse  metabolized 8.5  and 1.6  times more PCE than  rats at
10 ppm (67.8 mg/m3) and 500 mg/kg, respectively.
     Following exposure of  rats  and mice  to  10  and  600 ppm 14C-labeled PCE
(67.8 and 4068 mg/m3),  no  radioactivity was observed to be bound to purified
hepatic DMA.   The  detection limit was 14.5 alkylations/106  nucleotides at the
600 ppm (4068 mg/m3) level.  Lack of binding to DMA also was observed following
a single oral  dose of 500 mg/kg  14C-labeled  PCE.  However, at times of peak
binding to hepatic macromolecules, 4.7-,  5.8-, and 4.4-fold more radioactivity
was irreversibly bound  in  the mouse than  in the rat following exposure to 10
or 600 ppm  (67.8  or 4068 mg/m3), or 500 mg/kg,  respectively.  The extent of
hepatic macromolecular bindings was determined subsequent to homogenization of
the liver.  The  authors noted that  the  60-fold increase  in exposure  level
resulted in a 10-fold increase in the extent of binding in both rats and mice.
This result suggests  that  metabolism of PCE is a saturable process.   Binding
was determined at intervals up to 72 hours post-exposure.   The lack of binding
to DNA was  suggested  by the authors as  evidence indicating that PCE  lacks
genotoxic potential.  An absence of binding to DNA does not preclude such an
event that occurs below the detection limit of alkylation.   Effects of treatment
on body weight and liver are reported in Chapter 6.   A briefer discussion of
the results of Schumann and coworkers was presented by Watanabe et al.  (1980).
     A low rate of metabolism (2.8  mmole/kg) of PCE was reported by Bolt et
al.  (1982) during  exposure of  newborn female Wistar rats for 10 weeks  at 2000
ppm (13,560 mg/m3).   The  reported metabolic rate  was  7  umole/hr/kg,  a rate
30-fold less than that reported for trichloroethylene.
     In the oral and  inhalation  exposure of male Sprague-Dawley rats,  Pegg et
al.  (1979) found  that by either route,  PCE was eliminated in unchanged form,
predominantly  in  expired air.   In  contradiction to  evidence pertaining to
humans and other species, Pegg and coworkers found oxalic acid to be the major
urinary metabolite.  TCA was not detected.
     The protocol  was  similar to that used by  Schumann et  al. (1980)  in ex-
posures of mice  and rats.   Rats were exposed via  inhalation (6 hours) to 10

003PE1/E                             5-10                             11/22/83

-------
and 600 ppm 14C-labeled PCE (67.8 and 4068 mg/nv*).  In the gavage experiment,
1 or 500 mg 14Olabeled PCE/kg body weight was administered as a single dose.
     After oral administration (500 mg/kg) or inhalation (600 ppm;  4068 mg/m^),
89 percent of  the  radioactivity  was recovered in expired air as PCE;  1 to 2
percent remained in the carcass.   The difference  was accounted for by 14carbon
dioxide (C02)  and  urinary  and fecal metabolites.   Pulmonary elimination was
observed to be monophasic with a half-life of about 7 hours;  it was independent
of dose or route of administration.   Radioactivity remaining  in the carcass 72
hours  after  exposure by either  route was primarily  distributed  in liver,
kidney, and fat tissue.   After oral administration of 1 mg/kg, only 72 percent
of the  radioactivity  was  recovered in expired air as PCE.   Recovery of 14CO;j
and nonvolatile metabolites  increased.   The  authors suggested that, in rats,
metabolism of PCE is a saturable process.
     In blood, PCE levels peaked 1 hour after oral administration.   Disappear-
ance followed apparent first-order kinetics for up to 36 hours after exposure,
with a  half-life  of  6 hours and  an  elimination  constant of  0.12/hr.  After
termination of  exposure to 600 ppm  (4068 mg/m5),  PCE  levels  in blood  declined
similarly with  a  half-life of 7  hours and an  elimination constant  of  0.10/hr.
     Whole liver  radioactivity  and  that associated irreversibly with tissue
macromolecules was assayed 0, 6, 24, and 72 hours after termination of exposure
to 10  or  600  ppm  14C-labeled  PCE  (67.8  or 4068 mg/m3)  and 72  hours  after oral
administration.  Whole-liver  radioactivity in livers from inhalation-exposed
animals decreased  exponentially  during  the  72-hour collection period as  a
biphasic process.  The  ratio of bound  to  metabolized  radioactivity 72 hours
after  oral or inhalation exposure was not significantly  different  between  the
high and the  low dose.
     Nonprotein liver  sulfhydryl  levels were not found  to be  depleted, thus
suggesting to  Pegg  et al.  that glutathione was  not involved in metabolism.
The low rate of turnover of nonextractable radioactivity in liver macromolecules
also suggested  that  accumulation of bound PCE metabolites could  potentially
occur  upon repeated  exposure.   Such "accumulation" (or preferably, extent of
binding)  would be expected to plateau  as the rate of uptake,  elimination,
metabolism, and protein turnover  reached  equilibrium.
     Vainio et  al.  (1976)  looked at the  effects of PCE on liver metabolizing
enzymes jjri vivo in the rat.  Oral administration of 2.6 mmol/kg PCE was associ-
ated with a statistically significant lowering of concentrations of 3,4-benzpy-
rene hydroxylation and p-nitroanisole-o-methylation.
003PE1/E                             5-11                              11/22/83

-------
     Plevova et  al.  (1975)  showed that 6 hours  of  exposure to 177 ppm PCE
(12,000 mg/m3)  20  hours prior to administration of 44 mg/kg  intraperitoneal.
pentobarbital sodium would  lengthen  pentobarbital sleeping  time by  30  percent
in Wistar rats.   This effect was possibly mediated through hepatic drug metabo-
lizing enzyme activity.   Also,  changes in spontaneous motor activity induced
by  intraperitoneal  injection of  pentobarbital,  diazepam, amphetamine, and
partly by chlorpromazine  were enhanced by previous inhalation of PCE.  This
response probably resulted from an effect on metabolism rates.
     Although TCA  has  been  observed by several investigators to be a urinary
metabolite of PCE,  the  excretion  of  total trichloro compounds, as measured by
the nonspecific  Fujiwara  colorimetric  reaction after  oxidation, exceeded  that
of  TCA.   In  some cases,  this excretion  was  assumed to be  trichloroethanol
(Muenzer and Heder, 1972; Ikeda et al., 1972).   In other studies,  that portion
that was not  TCA could not be demonstrated  to be trichloroethanol  (Hake  and
Stewart,  1977).    In one report, ethylene  glycol was claimed to be a prominent
metabolite in the rat (Moslen et al., 1977).
     Leibman and Ortiz  (1975, 1977) proposed a scheme (see below)  for possible
pathways of PCE metabolism.   The formation of PCE epoxide by the hepatic mixed-
function oxidase system may be followed by hydration of the epoxide to PCE glycol
Because of the  symmetric  arrangement of  the  epoxide and glycol intermediates,
rearrangement of both  would yield trichloroacetyl  chloride, which hydrolyzes
rapidly to TCA and, to a much lesser extent,  TCE in the urine.  Possibly,  this
response could be caused by dehydrohalogenation to oxalic acid, which has  been
reported by Pegg et al. (1979) and Yllner (1961).

               C1C = CC12
              C10C	CC19—>• C1..C-COC1 -*• Cl-jC-COOH
                c. \^   j   L-      3            3
                    0
                    I
              C19C 	CC19-* CKC-COC1—^ C1,C-COOH
                *\     \   *
                 OH    OH

     Incubation of  PCE  and rat liver supernatant with a nicotinamide adenine
dinucleotide phosphate (NADPH), reduced generating system confirmed the produc-
tion of  TCA.   Nicotinamide  adenine dinucleotide,  reduced  (NADH),  did not
003PE1/E                             5-12                             11/22/83

-------
promote the  formation  of  TCA.   Expoxide hydrase inhibition, produced by the
addition of  cyclohexane to  the  incubation mixture, did  not  have  any effect
upon TCA formation.  Leibman and Ortiz  (1977) concluded  that, if the epoxide-
diol pathway  is  operative,  tetrachloroethylene  oxide is not a substrate for
hydration  by epoxide  hydrase,  or that  the  epoxide  and  glycol  rearrange to
trichloroacetyl chloride at similar rates.

5.1.4  Excretion and Elimination
     PCE is  removed  from  the body in two principal ways:  elimination as an
unchanged  compound in  exhaled air and through elimination of its metabolites.
During exhalation, the concentration of PCE  in expired air is a function of a
number of  factors:   (1) duration of exposure (until steady-state  is reached)
and the concentration  in inhaled air, (2)  rate of respiration,  (3) time elapsed
following  exposure, and (4) total  body lipid and other  tissue repositories.
The principal  approach used to  measure  the  amount and kinetics of elimination
via the lungs is serial breath analysis of alveolar air.
     From  controlled exposure  studies,  Stewart  and coworkers (Stewart, 1969;
Hake et  al.,  1976;  Stewart et al., 1970;  Stewart et al., 1961a,b; Stewart et
al., 1977;  Stewart  et  al.,  1974) concluded that PCE is   rapidly excreted from
the lungs  and is principally excreted in  unchanged  form.  These findings have
recently been  confirmed in  the controlled exposure studies of Monster et al.
(1979), who  used serial breath analysis and  found  that  between  80 and 100
percent of PCE is eliminated unchanged  via the lungs.
     The principal urinary  metabolite of PCE is TCA, although trichloroethanol
(TCE) has  been reported by  the nonspecific Fujiwara reaction to be a secondary
metabolite.   Other minor  metabolites reported include oxalic acid, dichloro-
acetic acid, and ethylene glycol.
5.1.4.1  Controlled  Studies—The  concentration  in  alveolar  air in the most
immediate  post-exposure period  (up to  2 hours)  is  a reflection  of the PCE
concentration  to which the  individual   was most  recently exposed  (Stewart et
al., 1970;  Stewart  1961b).   The breath decay curves shown in Figure 5-2 were
obtained from five  males  experimentally exposed to an average PCE concentra-
tion of  101 ppm (685  mg/m3) for  7  hd/day on 5  consecutive days.   The  curves
show that  a high percentage of absorbed  PCE was excreted during the 2-week
period  following exposure.   A single 7-hour  exposure of 15  volunteers  to an
average PCE  concentration of 101 ppm (685 mg/m3) resulted in a similar alveolar

003PE1/E                             5-13                             11/22/83

-------
desaturation curve  (Stewart  et  al.,  1970).   The range of concentrations from
which the  average  was  obtained  was 62 to 137  ppm (420 to 929 mg/m3).  The
increase of initial alveolar air concentration after repeated exposure (Figures
5-2 and  5-3) was  suggested by the authors to be a result of the accumulation
of PCE  in  the  body tissues.   However, as shown by Schumann et al.  (1980),  an
equilibrium is rapidly reached where the body burden of PCE would not increase
to a  value greater  than that  of the previous day  (Figure 5-2).  The "hump" in
the curve  (Figure  5-3)  is unexplained but was  not considered by Stewart and
co-workers to be artifactual.   Breath samples, collected in glass pipettes  and
Saran bags, were analyzed by infrared spectroscopy.
     After  an  exposure of six volunteers  to  approximately  100 and 200 ppm PCE
(678  and  1356  mg/m3),  analyses  of the breath decay curves indicated that (1)
exposures  of similar duration yielded decay curves with similar elimination rate
constants,  (2)  the  average concentration in the expired air was proportional
to the  vapor concentration for exposures of  similar  duration,  and (3) the
length of  time that PCE  could be measured in expired air was proportional  to
both  the  vapor concentration  and the duration  of exposure,  i.e., it  is pro-
portional  to  the acquired  body  burden, which  would  plateau upon  repeated
exposure (Stewart et al., 1961a).
     The concentration of PCE in the blood of those individuals exposed to  194
ppm (1316  mg/m3)  for  83  and 187 minutes approached a plateau near the end  of
the third  hour of  exposure.   After exposure,  PCE was  rapidly cleared  from the
blood and was undetectable after 30 minutes.
     Monster et  al. (1979),  in agreement  with  the general  findings of Stewart
and co-workers (Stewart,  1969; Hake et al.,  1976; Stewart et al., 1970;  Stewart
et al.,  1961a,b; Stewart et al.,  1977; Stewart et al., 1974), found that 80 to
100 percent  of the  PCE absorbed was excreted  unchanged; metabolism to urinary
TCA accounted for less than 2 percent.  Physical exercise resulted in increased
uptake and an  increase of PCE in  blood  levels;  similar  observations were made
by Stewart et al. (1974).
     In  the  study  by  Monster et al.  (1979),  discussed  previously in  Section
5.1.1.1, the concentration of PCE in exhaled air decreased when exposed indivi-
duals exercised.  Recovery in exhaled air was 78 percent (exercise) as compared
to 92 percent  when the individuals were  at  rest.  Exercise  had  no effect on
the half-life  of  PCE  elimination or on  the  rate  constant of elimination.
Minute volume and lung clearance were three-fold higher than the values obtained

003PE1/E                             5-14                             11/22/83

-------
      UJ
      Z
      UJ
         £ 100
      UJ
      O
      CE
      O

      I
      O
      <
      cc

      UJ
      I-
        O
I
UJ
O

O
O
10
     iT~\   I    I    FT
   7 hr VAPOR EXPOSURES
                                      I   I    I   \    I    I   I    I
                1   I    I    I   I    I   I   I    I   I
                    1   2  3   4   5   6   7  8   9  10  11  12   13   14

                                     TIME, days
Figure 5-2.
003PE1/E
 Mean and range of breath concentrations  of  tetrachloroethylene
 after exposure of individuals  to  single  or  repeated exposures.
 Source:  Stewart et al.,  1970.

                         5-15
                                                             11/22/83

-------
        30 ir-l  I  I  I  I  I  I  I  I  I  I  I   I  I  II
                                      I
                                      I
                                           I  I  I  I  I   I  I  I-
                                   MEAN AND RANGE AFTER .
                                   101 ppm 7 hr/day for 5 days,
                                   5 SUBJECTS
                                   MEAN AND RANGE AFTER
                                   101 ppm 7 hr, 15 SUBJECTS
                                                      .AJ-TT
cc
»-
LLJ
        0.1
              I  I  I  I  I  I   I  I  I  I  I  I   I  I  I  I  I  I   i  I  I  I.|  I
             15 30   60   90  120  150 180 210  240 270  300  330 360
                             POST EXPOSURE, hours
Figure 5-3.   Mean  and  range of breath concentrations of  five  individuals during
             post-exposure after five separate exposures to 96,  109, 104, 98,
             and 99  ppm  tetrachloroethylene.

             Source:   et al., 1970.
003PE1/E
                               5-16
11/22/83

-------
when the individuals were at rest.   While exercise decreased the amount of PCE
in exhaled air, uptake increased about 40 percent (Figure 5-1).
     The concentrations  of  PCE  in  blood and in exhaled air during the post-
exposure period are  shown  in Figure 5-4.   Contrary to the finding of Stewart
et al.  (1961a)  that  blood  concentrations of PCE were undetectable 30 minutes
after exposure, Monster  et  al.  (1979) found that the decrease of PCE in the
blood paralleled the decay in expired air.   The slopes of the curves  in Figure
5-3 suggest that the half-lives of PCE in exhaled air and blood for three body
compartments are for (1) tissues with high blood flow, (2) lean tissue,  and (3)
adipose tissue.  The concentration of TCA (from metabolism) in the blood increased
for 20 hours post-exposure before declining.  Monster (1979) suggested that the
increase in TCA concentration is smaller and that the maximum is reached earlier
in contrast to exposures to trichloroethylene or methyl  chloroform.   The biolo-
gical half-life of TCA  in blood  is  the  same  for all  solvents  (about  80 to  100
hours).   Upon repeated exposure to PCE, TCA would increase (Monster et al., 1979).
However, TCA would  be expected to eventually plateau and not increase beyond
steady-state.
     The time  course of the PCE concentrations in  blood  and in expired air
indicates that a long period (greater than 275 hours) is necessary to complete-
ly eliminate PCE from the body (Fernandez et al., 1976;  Monster et al., 1979).
Fernandez et al. (1976)  found that 2 weeks were necessary to completely eliminate
PCE  from the  body  after exposure to  100 ppm (678 mg/m^)  for  8  hours.  These
findings agree with  those of Monster et al.  (1979).   In these chamber studies,
24 volunteers  were  exposed  for  1 to 8 hours  to  vapor containing  100,  150,  and
200  ppm  PCE (678,  1017, and 1356 mg/m3).   When the  exposure  time  increased,
the  PCE concentration in alveolar air would  plateau at steady-state.
     Monster (1979)  determined  that the values  for  partition  coefficients  and
lung clearance  measurements between blood  and  vapor  after exposure of four to
six  individuals to 70 to 140 ppm PCE  (475 and 950 mg/m3) for 4 hours  indicated
that (1) alveolar air concentration of PCE in the first few hours after exposure
will be  proportional to exposure concentration and  to  the concentration  in
blood and  other rapidly exchangeable tissues,  and  (2) during  that  later  phase
of elimination,  the  alveolar air concentration will  be  proportional to the
concentration  in adipose tissue.  As  previously stated, the partition coeffici-
ent  (37°C)  for  PCE between  venous blood and  alveolar air of 16 and the partition
coefficient between  fat and blood  of  90  suggests  that  adipose  tissue  is  a

003PE1/E                              5-17                             11/22/83

-------
         §8
         <  rJ
         I  ffl
         X  Z
            UJ
1000

 100

  10

   1

  0.1

0.01
                   = A
I   I  I   I  I   I   I  I   I  I   I   I  I
 O  72 ppm PCE AT REST
 A 144 ppm PCE AT REST
 D 142 ppm PCE AT REST AND WORKLOAD
                      EXHALED AIR
                   -I
                                 50           100
                                POST EXPOSURE, houn
                                 150
Figure 5-4.  Tetrachloroethylene in blood and exhaled air  following  exposure
             to tetrachloroethylene for 4 hours.  Each  point  represents  the
             geometric mean ± standard deviation of six individuals.
             Source:  Monster et al. , 1979.
003PE1/E
           5-18
                                                        11/22/83

-------
primary storage site.  Monster estimated that 25 hours would be necessary for
PCE to saturate adipose tissue to 50 percent of its equilibrium concentration
with plasma.
     Metabolic considerations  investigated  in this  study are discussed in
Sections 5.1.3 and 5.1.4.
     Evidence that blood levels of PCE may be useful  in determining individual
uptake was obtained in a single exposure to 70 and 140 ppm (475 and 950 mg/m3)
for 4  hours  (Monster and  Houtkooper, 1979).   Concentrations of PCE in blood,
urine, and  exhaled air were  determined  at  2 and 20  hours  after  exposure.
While a high degree of correlation was obtained between exhaled air concentra-
tion and  blood concentration,  linear and multiple linear regression analysis
showed an inter-person coefficient of variation of 20 to  25 percent for blood
measurements at 2 and 20 hours and in exhaled air at 2 hours.
     Verberk and  Scheffers  (1980) suggest that measurements of PCE  in  expired
air are useful in monitoring  exposure in the general  population as well as  in
populations of occupationally exposed people.   PCE was measured in the expired
air of residents  living near 12 dry-cleaning shops.   Residents were classified
into two  groups,  corresponding to the distance they  lived from the  shops.   In
all cases, the residents  living nearest the shop, the house next  to the shop,
had higher levels of PCE  in expired  air  than  residents who lived one house
farther away from the shop.
     In the study by Stewart and Dodd (1964), elimination of PCE via the lungs
following percutaneous absorption was shown to be minor.  Each of five indi-
viduals immersed  one thumb in a beaker of PCE located in  a ventilated hood.  At
intervals of 10 minutes, the concentration of PCE in exhaled air was measured.
Before and periodically during each skin exposure,  samples of breathing zone
air were  analyzed to preclude solvent  vapor  contamination.   The mean peak
breath concentration  after a 40-minute immersion was 0.31 ppm (2.1 mg/m3);  2
hours  after  exposure,  the  mean breath concentration  was  0.23  ppm  (1.6  mg/m3).
Five  hours after  exposure,  PCE was  still  detectable  (0.16 to  0.26 ppm; 1.1  to
1.8 mg/m3).
     The  elimination of PCE  through the skin was•found by Bolanowska and
Golacka (1972) to be approximately  0.02  percent  per  hour of the dose  inhaled.
     As previously mentioned, in both controlled and occupational exposures of
humans to PCE, the principal urinary excretion product is TCA.  Trichloroethanol
has been  reported by a  nonspecific  assay  as  a  metabolite.  TCE was  indirectly
measured  by chromate oxidation of urine to TCA.
003PE1/E                             5-19                              11/22/83

-------
     Hake and Stewart (1977) found only traces of TCA in 24-hour urine specimens
from individuals exposed  to 150 ppm PCE (1017 mg/m3) and below.  No TCE was
detected.
     Ogata et al.  (1971) found TCA in the urine of four individuals exposed to
87 ppm PCE  (590 mg/m3)  for  3  hours at  a  level of  1.8 percent of the total PCE
dose.   TCE  could not  be detected, but the  urine  contained  1 percent of an
unidentified chlorine-containing  compound.   Urine was  collected for 67 hours
into the post-exposure period.
     In the previously discussed studies of Monster and co-workers (Monster et
al., 1979;  Monster  and  Houtkooper,  1979),  urinary TCA was found to represent
less than 1 percent of  the absorbed dose of PCE.   In blood, TCA continued to
increase until 20  hours post-exposure.  From about 60  hours after exposure,
the concentration decreased exponentially.   A base level of 0.6 mg of TCA per
day was found  in  the urine of  subjects  prior to  exposure.  Results of blood
and urine concentrations are shown in Figures 5-5 and 5-6.  The ratio of TCA  .
                                        3                                  urine
to TCA. ,   .  was three-fold  higher in the period 0 to 22 hours after the start
       blood
of  the exposure.   The  relatively high concentration  in urine possibly was
caused by an unknown  compound measured by the non-specific  Fujiwara reaction;
TCA in blood was measured by gas chromatography.   The unknown compound was not
PCE or TCE.
     Exposure combined  with exercise  resulted in 20 percent higher levels of
excreted TCA, while uptake  of PCE increased 40 percent.   The TCA concentration
at  20  hours after  exposure was 1.6  times  the concentration at  the  end of
exposure.    Inferences drawn from these results regarding metabolism of PCE are
discussed in  Section  5.1.3.   Monster and Houtkooper (1979)  concluded that TCA
is not a reliable indicator of  exposure to PCE.
5.1.4.2  Occupational Studies — In a study  involving  six  dry-cleaning-plant
workers exposed  to PCE,  an increase  in urinary  TCA was  observed over the
50-hour sampling period (National Institute of Occupational  Safety and Health,
1974).   A  control  group not  exposed to  significant  quantities  of PCE also
evidenced a  similar increase.    The average length of exposure for these indivi-
duals  was 17 months.   The  worker evidencing  the  highest  level  of TCA  in the
urine  had been  exposed  to  an 8-hour time-weighted average  of 168 to 171 ppm
PCE (1139 to 1160 mg/m3).
     TCA and TCE  (specific detection  method not  discussed) were  found in the
urine  of 40 workers exposed to PCE concentrations ranging from 58 to 134 ppm

003PE1/E                             5-20                             11/22/83

-------
               1 -
         I   0.6 H
          I
             0.4 —
         §   0.2 H
         ffi
              0.1 —
O 72 ppm PCE AT REST
4144 ppm PCE AT REST

0142 ppm PCE AT REST AND WORKLOAD
                        I  I  I  I  I   I  I  I   I  I  I  I   I  I  I  |
                              50         100         150

                          TIME AFTER EXPOSURE, houn
Figure 5-5.  Trichloroacetic acid in blood following exposure  to  tetrachloro-
             ethylene for 4 hours.  Each point represents  the  geometric mean
             ± the standard deviation of six subjects.

             Source:   Monster et al., 1979).
003PE1/E
              5-21
11/22/83

-------

0

1*
< 8
H 6
UJ
Z 4
cc
3 2
ft
1 1 1 1 1
i i

i i
72ppmPCE WppmPCE 142ppmPCE
AT REST AT REST AT REST AND
T WORKLOAD
— —
^^
"" rri
-1 [*,*,
*






¥1
T





^ —
Tji,
                     0 22 46 70     0  22  46 70     0  22 46 70

                            TIME AFTER START EXPOSURE, hours
Figure 5-6.  Urinary excretion of trichloroacetic  acid  following  exposure to
             tetrachloroethylene for 4  hours.   Each  point  represents  the  geo-
             metric mean ± the standard deviation  of six subjects.

             Source:  Monster et al., 1979.
003PE1/E
5-22
11/22/83

-------
(393 to 909  mg/m3)  (Medek and Kovarik, 1973).  The  maximum levels observed
were 41 mg  TCA  and  116 mg TCE per  liter  of urine.   Seventy-two percent of
these workers reported  subjective  complaints.   No relationship was proposed
between specific  complaints  and  specific exposures  or  TCA and TCE levels.
Exposed groups were not compared  with blind controls.
     Muenzer and  Heder  (1972)  reported that urinary TCA was found in 124 of
200 dry-cleaning-plant  employees.   Seventy-one  individuals had more than 10
mg/1.   Liver  function  tests  for  exposed and unexposed  (control) groups were
comparable.   The  general  room air in the work places contained between 200  to
300 ppm PCE  (1357 to 2035 mg/m3).  An  association between  workroom air con-
centrations and TCA levels was not made.
     Ikeda et al. (1972)  reported evidence  that TCA  and TCE concentrations  in
the urine increased in proportion to environmental concentrations of PCE up to
50 ppm  (339  mg/m3).   In this  study, urine samples were  collected from 34 male
industrial workers  who  had been  exposed to PCE vapors for 8 h/day for 6 days
per week.   Concentrations  of  PCE in the work place ranged from 10 to 400 ppm
(68 to  2713  mg/m3).   The  plateau observed in the urinary excretion curve for
TCA suggested to  the investigators  that the capacity of humans to metabolize
PCE is  limited.   The maximum level  of TCA observed was approximately 50 mg/1
of urine.   For  TCE, the maximum concentration reported was approximately 25
mg/1.   TCE  was  measured indirectly by oxidation of urine with chromic oxide.
     In another  study,  Ikeda  and Ohtsuji  (1972) reported a wide variation  in
the TCA  and TCE  levels in urine  from  occupational ly exposed workers.  One
group of  four workers had been exposed to  a concentration  range of 20 to 70
ppm (136  to  475  mg/m3), while 66 workers  in another  group  had  been exposed  to
between 200  and  400 ppm (1357 and  2713 mg/m3).   The urine from the smaller
group contained  between 4 and 35 mg TCA  and  4 to 20 mg of TCE per liter of
urine.   In  the  larger group,  TCA levels were  32  to 97 mg/1 and TCE  levels
ranged from 21 to 100 mg/1.
     In their most  recent study, Ohtsuji  et al.  (1983) obtained additional
evidence  that confirms  reports by others  that  PCE is metabolized to a limited
extent  (approximately  2,percent)  in humans.  Personal monitoring of exposure,
using carbon  felt dosimeters,  was  carried  out  in  two  groups of workers (36
males and 25  females).  Comparison  of urinary total   trichlorocompounds (expres-
sed as  TCA)  with occupational  levels of PCE suggested that metabolic  capacity
becomes  saturated at  about  100  ppm (678  mg/m3)  in  air.  Metabolite  levels
increased in  a quasi-linear fashion up  to 100 ppm PCE (678  mg/m3)
003PE1/E                             5-23                             11/22/83

-------
     High levels of TCA (greater than 60 mg/1 of urine) also were reported by
Weiss (1969) and by Haag (1958) in studies of individuals  exposed occupational-
iy-
5.1.4.3   Population Studies--Pre1imi nary  results  reported  by  Ziglio (1981)
suggest  that  plasma TCA  can be a reliable indicator of the extent of chronic
exposure  to PCE.   Ziglio  chose 24 individuals in Milan, a city in which both
trichloroethylene and  PCE were present  in tap water at  concentrations as high
as 150 to 200 ug/1  (Giovanardi, 1979).   Subjects were divided into two groups:
exposed  (those who drank water with halocarbon levels > 50 ug/1) and nonexposed
(those who drank water with levels < 1 ug/1).   When exposure increased,  plasma
TCA  levels  increased.  However, the TCA levels were suggested by Ziglio to be
more indicative  of  the extent of exposure to trichloroethylene, because this
halocarbon is metabolized to a much greater extent than is PCE.

5.1.5  Estimates of Biological Half-life
     Monster  et  al. (1979)  determined from the concentration curves  of  PCE in
blood and exhaled air after exposure (Figure 5-4) that PCE was eliminated from
the body at three different rate constants with corresponding half-lives of 12
to 16,  30 to  40, and  55 hours, respectively; this result  suggested that there
are  three major  body  compartments  for PCE.   The half-life, derived  from the
data of  Stewart  et  al. (1970), was  calculated to be 65  hours.   The long half-
life does not pertain to  the major portion  of  the  exposure but to  a small
percentage of  the body burden  (Hake et  al., 1976).  TCA in  blood was  reported
by Monster  et al.   (1979)  to  have  a predominant half-life  (60  hours after
exposure) of 75 to 80 hours.
     Ikeda (1977) and Ikeda and  Imamura (1973) reported  that  the mean bio-
logical  half-life for PCE  urinary  metabolites is 144  hours.  A possible sex
difference,  indicated  from  exposures  of nine males and four females, has not
been confirmed.  The  estimated biological  half-life of PCE stored in adipose
tissue is 71.5 hours  (Guberan and Fernandez,  1974).

5.1.6  Interaction of PCE with Other Compounds
     Stewart et al.  (1976) conducted a study designed to determine the effects
of alcohol  and diazepam (Valium  ) on 12 individuals exposed to  25 and 100  ppm
PCE  (170 and 678 mg/m3) for 5.5 hours.   Administration of alcohol to individuals
during exposure  to  25 ppm  (170 mg/m3) significantly  increased blood  levels of

003PE1/E                             5-24                             11/22/83

-------
PCE (p < 0.01), but there was no effect during exposure at 100 ppm (678 mg/m^).
Diazepam and alcohol  each  raised breath levels of PCE during exposure at 25
ppm (170 mg/m3) but not at 100 ppm (678 mg/m^).   Results are shown in Table 5-2.
Neither diazepam nor alcohol changed the effects of PCE as measured by behavioral
and neurological tests.

5.1.7  Summary
     Inhalation  is  the predominant route  of  exposure  for humans, although
contamination  of potable  water  supplies indicates that oral ingestion repre-
sents an additional  route  of exposure.  Percutaneous  absorption  is  of minor
consideration.   In all species tested, PCE is  rapidly absorbed and distributed
to major body  compartments.   Because  of its high octanol/water and fat/blood
partition  coefficients,  PCE can  be expected  to selectively partition into
lipid-rich tissues until an equilibrium is reached between uptake and elimina-
tion, after  which  additional  accumulation  would not occur.  While  most of the
PCE  is  rapidly excreted,  up to 2 weeks or more may be required to completely
eliminate  PCE  from  the human body (Fernandez et  al. ,  1976).   The estimated
biological  half-life  of  PCE in adipose tissue  is  about 70  hours.  PCE would
also  be  expected to partition  into lungs,  liver,  kidney, spleen, and lean
muscle until equilibrium  is reached.   The rate of tissue loading  in humans,
with a  given inspired air concentration, is increased with .physical  activity
and with exposure duration.
     PCE is postulated to be metabolized via oxidation by the cytochrome P-450
mixed-function oxidase system.  An epoxide intermediate,  formed during metabo-
lism,  is believed  to  represent  a  species  capable of binding covalently to
tissue macromolecules.  Jji vitro experiments  (Costa and Ivanetich, 1980) have
shown that a cytochrome P-450 activation system from rats binds and metabolizes
PCE; TCA was identified as the principal reaction product.  The _i_n vivo studies
by Pegg  et al. (1979) in  rats  and by Schumann et al.  (1980)  in mice have
demonstrated  irreversible  binding of  PCE  and/or  its  metabolites to  hepatic
macromolecules;  binding  to DNA, however, was not observed.   Differences in
metabolism of  PCE in animals and humans, who have  shown very little metabolism
of PCE,  seem to exist.   In view of these  metabolic differences between humans
and  rodents, little binding of PCE metabolites to  human hepatic macromolecules
would be expected.
     Pegg  et al.  (1979) and Schumann  et  al.  (1980)  have suggested  that the
metabolism of  PCE  is a saturable, dose-dependent  process and that the extent
003PE1/E                             5-25                             11/22/83

-------
                  TABLE 5-2.  ALCOHOL AND DIA2EPAM EFFECTS UPON TETRACHLOROETHYLENE (PCE) BLOOD  AND  BREATH  CONCENTRATIONS (5.5-HOUR EXPOSURES)
cr
 i
re
CT:
PCE in blood, ppm
2 hours into exposure
PCE in
chamber,
ppm
25
100
PCE
alone
1.65
(35)
8.25
(63)
PCE and
alcohol
2.92C
(15)
7.95
(29)
PCE andb
diazepam
1.76
(23)
8.47
(41)
PCE
alone
11.03
(35)
33.2
(68)
PCE in breath, ppm
2 hours into exposure 30
PCE and
alcohol3
12.35d
(15)
32.3
(28)
PCE andb
diazepam
11.72
(23)
35.5
(44)
PCE
alone
6.40
(35)
17.62
(64)
minutes post-exposure
PCE and
alcohol
7.40C
(14)
13.83C
(29)
PCE and
diazepam
6.96d
(22)
17.35
(42)
           Alcohol blood levels of 30 to 100 rug percent.
           Diazepam blood levels of 7 to 30 mg percent.
          cSignificantly different from PCE alone at p <0.1.
           Significantly different from PCE alone at p <0.05.
          Numbers in parentheses indicate the number of determinations.
          Source:  Stewart et al., 1977.

-------
of reactive intermediate formation plateaus with increasing dose.   This situa-
tion would explain why  exposure  to mice at 500  mg  PCE/kg body weight would
result in a carcinogenic response (see Chapter 9) in the National  Cancer Institute
bioassay but not an increase at a level  of 1000 mg/kg.
     Although TCA appears to be a predominant  urinary metabolite in humans and
rodents, metabolism  in  humans  (in the form of  urinary  TCA)  accounts for 2
percent or less of the absorbed dose.   The estimated mean biological  half-life
for all  PCE  urinary  metabolites  in humans  is  144  hours.   Minor metabolites
reported include  TCE, oxalic acid, and ethylene  glycol.   However,  Pegg et al.
(1979) observed  oxalic  acid to be the principal urinary metabolite  in rats.
TCA was  not  detected upon incubation of a  rat  hepatic microsomal  activation
system with  PCE.   Costa  and  Ivanetich (1980)  observed TCA  to be the  principal
reaction product.
     Elimination  of  PCE  in  expired air differs  somewhat  between humans, mice,
and rats.  Monster and  coworkers (1979) have  shown that humans may exhale as
much  as  98 percent  of  PCE  in unchanged  form.   Exercise  increases  uptake
(decreases pulmonary  elimination)  of  PCE in the body.   Repeated exposures to
levels  of  72  ppm (488  mg/m^)  or greater  during sedentary conditions were
observed to result in more PCE being eliminated unchanged in expired air; this
situation  is  consistent  with saturation of metabolism.   At found or expected
ambient air levels of exposure, the fate of PCE would not be altered regardless
of exposure concentration in the linear pharmacokinetic  range.
     Elimination  as  the  unchanged compound in expired  air also is the pre-
dominant process  in  rats (Pegg et al., 1979),  upon inhalation or oral exposure to
PCE.   However,  in mice,  urinary metabolites  represent  the chief  elimination
process  at low exposures but not at  high  exposures  (Schumann  et  al.,  1980).
Upon  high  oral dose exposure of mice, pulmonary elimination of unchanged PCE
predominates  (Schumann et al., 1980).
     The dose level  is  a factor  of  particular  importance  when one considers
the potential  effects of PCE in  humans.   There  appears  to  be no evidence  that
a  different  metabolite  or a distinctively  different pharmacokinetic pattern
exists  at  high,  as compared to  low,  exposure levels.   Monster et al.  (1979)
has shown  that a  two-fold increase in exposure level resulted  in a doubling of
uptake;  a  shift   in the  nature of  the urinary metabolites was not observed.
 003PE1/E                             5-27                             11/22/83

-------
5.2  REFERENCES


Barchet et al.   Chemical investigations for environmental protection. Deutsch
     Lebensm - Rundsch. 68:69, 1972.  Cited in:  Chem. Abstr. 77:32869,  1972.

Boillat, M. A.   The value of the determination of trichloroacetic acid  in  the
     urine for the detection of trichloroethylene and perchloroethylene  poison-
     ing.  An inquiry  in several works.  Praventivmedizin --  Revue de Med.
     Prev. 15(6):447-456, 1970.

Bolanowska, W., and J. Golacka.  Absorption and elimination of tetrachloro-
     ethylene in humans under experimental conditions.  Medycyna Pracy  23(2):
     109-119, 1972.  (English translation)

Bolt, H. M., R.  J.  Laib, and J. G.  Filser.  Reactive metabolites and carcino-
     genicity of halogenated ethylenes.  Biochem. Pharmacol.  3J.:(l):l-4, 1982.

Bonse, G., and D. Henschler.  Chemical reactivity, biotransformation, and
     toxicity of polychlorinated aliphatic compounds.  CRC Crit. Rev. Toxicol.
     4:  395-409, 1976.

Bonse, G., Th.  Urban,  D. Reichert,  and D. Henschler.  Chemical reactivity,
     metabolic oxirane, etc.  Biochem. Pharmacol. 24(19):1829-1834, 1975.

Cornish, H. H., B.  P.  Ling, and M.  C. Barth.  Phenobarbital and organic  solvent
     toxicity.   Am. Ind. Hyg. Assoc. J. 34:487-492, 1973.

Costa, A. K., and K. M. Ivanetich.  Tetrachloroethylene metabolism by the
     hepatic microsomal cytochrome  P-450 system.  Biochem. Pharmacol.
     29:2863-2869,  1980.

Creech,  J. L.,  Jr., and M. N. Johnson.  Angiosarcoma of the liver in the
     manufacture of polyvinyl chloride.  J. Occup. Med. 16:150, 1974.

Daniel,  J.  The metabolism of 36Cl-labeled trichloroethylene  and tetrachloro-
     ethylene in the rat.  Biochem. Pharmacol. 12:795-802, 1963.

Derkosch.  Cited in:   CRC Crit. Rev. Toxicol. 4:395-409, 1976.

Dmitrieva, N.  V.  Contribution to the metabolism of tetrachloroethylene. Gig.
     Tr. Prof.  Zabol.  11:54-56, 1967.  (English translation)

Fernandez, J.,  E. Guberan, and J. Caperos.  Experimental human exposures to
     tetrachloroethylene vapor and  elimination in breath after inhalation.
     Am. Ind.  Hyg.  Assoc. J. 37:143-150, 1976.

Giovanardi, A.   Riv. Ital. Ig. 39:323, 1979.

Greim, H., G.  Bonse, Z. Radwan, D.  Reichert, and D. Henschler.  Mutagenicity
     ui vitro and potential carcinogenicity of chlorinated ethylenes as  a
     function of metabolic oxirane  formation.  Biochem. Pharmacol. 24(21):
     2013-2017, 1975.
003PE1/E                             5-28                              11/22/83

-------
Guberan, E.,  and J.  Fernandez.   Control of industrial exposure to tetrachloro-
     ethylene by measuring alveolar concentration:  theoretical approach using
     a mathematical  model.  Brit.  J. Ind.  Med. 31:159, 1974.

Haag, T. P.   Concerning the question of the determination and decomposition of
     perchloroethylene.  Arch.  Toxikol. 17:204-205, 1958.

Hake, C. L.,  and R.  D. Stewart.  Human exposure to tetrachloroethylene:
     inhalation and skin contact.   Environ. Health Persp. 21:231-238,  1977.

Hake, C. L. ,  R. D.  Stewart, A.  Wu, and S.  A. Graff.  Experimental human expo-
     sures to perchloroethylene.  I.  Absorption and excretion.  Toxicol.
     Appl. Pharmacol.  37:175, 1976 (abstr).

Henschler, D., G. Bonse, and H. Greim.  Carcinogenic potential of chlorinated
     ethylenes -- tentative molecular rules.  Proc. Third WHO-IARC Meeting,
     Lyon, November 3, 1975.

Ikeda, M.  Metabolism of trichloroethylene and tetrachloroethylene in  human
     subjects.  Environ. Health Persp. 21:239-245, 1977.

Ikeda, M. , and H. Ohtsuji.  A comparative study on the excretion of  Fujiwara
     reaction positive substances in the urine of  humans and  rodents given
     trichloro or tetrachloro derivatives of ethane or ethylene.  Brit. J.
     Ind. Med. 29:94-104,  1972.

Ikeda, M., and T. Imamura.  Biological half-life  of trichloroethylene  and
     tetrachloroethylene  in human subjects.   Int.  Arch.  Arbeitsmed.  3^1:209-224,
     1973.

Ikeda, M., H. Ohtsuji, T.  Imamura, and Y. Komoike.  Urinary excretion  of  total
     trichloro-compounds,  trichloroethanol, and trichloroecetic  acid as a
     measure of exposure  to trichloroethylene and  tetrachloroethylene.  Brit.
     J.  Ind.  Med. 29:329-333,  1972.

Jakobson, I., J. E. Wahlberg,  B.  Holmberg,  and G.  Johansson.   Uptake via  the
     blood and elimination of  10  organic solvents  following epicutaneous  expo-
     sure of anesthetized  guinea  pigs.  Toxicol.  Appl.  Pharmacol. 63:181-187,
     1982.

Jerina,  D. M., and J.  W.  Daly.  Arene  oxides:  A  new  aspect of drug  meta-
     bolism.   Science  185:573,  1974.

Kaemmerer, K., J. Fink,  and M.  Kietzmann.   Studies on  the  pharmacodynamics of
     perchloroethylene.   Der praktische Tierarzt  2:171-182, 1982.

Klaassen, C. D., and  G.  L.  Plaa.  The  relative effects  of  various chlorinated
     hydrocarbons on  liver and  kidney  function in mice.  Toxicol. Appl.
     Pharmacol.  9:129-131,  1966.

Kline,  S. A.,  and B.  L.  Van Duuren.   Reactions of epoxy-l,l,2-trichloroethane
     with nucleophiles.   J. Heterocyclic Chem. 14:455-458,  1977.

Kline,  S. A.,  J. J. Solomon, and  B.  L. Van  Duuren.   Synthesis and  reactions of
     chloroalkene epoxides.  J. Org.  Chem.  43:3596-3600, 1978.

003PE1/E                             5-29                              11/22/83

-------
Kronevi, T. , J. F. Wahlberg, and B. Holmberg.  Skin pathology following epicu-
     taneous exposure to seven organic solvents.  Int. J. Tiss. React. III(l):
     21-30, 1981.

Lee, F. I., and D. S. Harry.  Angiosarcoma of the liver  in a vinyl chloride
     worker.   Lancet 1:1316, 1974.

Liebman, K. C., and E.  Ortiz.  Styrene epoxide - an intermediate  in microsomal
     oxidation of styrene to its glycol.  Pharmacologist 10:203,  1968.

Liebman, K. C., and E.  Ortiz.  Epoxide intermediates  in microsomal oxidation
     of olefins to glycols.   J. Pharmacol. Explt. Therap. 173:242, 1970.

Leibman, K. C., and E.  Ortiz.  Microsomal metabolism  of chlorinated ethylenes.
     Paper presented at the Sixth  International Congress on Pharmacology.
     1975.

Leibman, K. C., and E.  Ortiz.  Metabolism of halogenated ethylenes. Environ.
     Health Persp. 21:91-97, 1977.

Maltoni, C., and G.  Lefemine.  Carcinogenicity bioassays of vinyl chloride. I.
     Environ.  Res. 7:387, 1974.

Maynert, E. W., R. L. Foreman, and T. Watabe.  Epoxides as obligatory inter-
     mediates  in the metabolism of olefins to glycols.  J. Biol.  Chem. 245:
     5254,  1970.

Medek, V.,  and J. Kovarik.   The effect of perchloroethylene on the health of
     workers.   Pracovni Lekarstvi  25(8):339-341, 1973.

Monster, A. C.  Difference in uptake, elimination and metabolism  in exposure
     to trichloroethylene,  1,1,1-trichloroethane and  tetrachloroethylene.
     Int.  Arch. Occup.  Environ. Health 42:311-317, 1979.

Monster, A. C., and J.  M. Houtkooper.  Estimation of  individual uptake of
     trichloroethylene, 1,1,1-trichloroethane and tetrachloroethylene from
     biological parameters.   Int.  Arch. Occup. Environ. Health 42:319-323,
     1979.

Monster, A. C., G. Boersma,  and H. Steenweg.   Kinetics of tetrachloroethylene
     in volunteers;  influence of exposure concentration and work  load.  Int.
     Arch.  Occup. Environ.  Health  42:303-309, 1979.

Moslen, M.  T., E. S.  Reynolds, and S. Szabo.   Enhancement of the  metabolism
     and hepatotoxicity of trichloroethylene and perchloroethylene.  Biochem.
     Pharmacol. 26:369-375,  1977.

Muenzer, M. , and K.  Heder.   Results of an industrial  hygiene survey and medical
     examinations of drycleaning firms.  Zbl. Arbeitsmed. 2^:133-138, 1972.

National Institute of Occupational Safety and Health.   Health Hazard Evaluation
     Report No. 73-86-114,  1974.

Ogata, M.,  and T. Hasegawa.   Effects of chlorinated aliphatic hydrocarbons on
     mitochondrial oxidative phosphorylation in the rat with reference to the
     effects of chlorinated aromatic hydrocarbons.  Ind. Health 19:71-75, 1981.

003PE1/E                             5-30                             11/22/83

-------
Ogata, M. ,  Y. Takatsuka, and K. Tomokuni.  Excretion of organic chlorine
     compounds in the urine of persons exposed to vapours of trichloroethylene
     and tetrachloroethylane.   Brit. J.  Ind. Med. 28:386-391, 1971.

Ohtsuji, T., K. Sato, A. Koizumi, M. Kumai, and M. Ikeda.  Limited capacity
     of humans to metabolize tetrachloroethylene.  Int. Arch. Occup. Environ.
     Health 51:381-390, 1983.

Pegg, D. G., J. A. Zemple, W.  H. Braun,  and P. G. Watanabe.  Disposition  of
     (14C) tetrachloroethylene following oral and inhalation exposure  in  rats.
     Toxicol. Appl. Pharmacol. 51:465-474, 1979.

Plevova, J. , E. Frantik, and D. Chladkova.  Problem of interaction between
     drugs and some air pollutants.  Proc. Eur. Soc. Toxicol. 16:303-306,
     1975.

Riihimaki, V., and P. Pfaffli.  Percutaneous absorption of solvent vapors  in
     man.  Scand. J. Work Environ.  Health  4:73-85, 1978.

Savolainen, H., P. Pfaffli, M. Tengen, and H. Vainio.  Biochemical and behavioral
     effects of inhalation exposure to tetrachloroethylene and dichloromethane.
     J. Neuropath. Exp. Neurology 36(6):941-949,  1977.

Schumann,  A. M.,  J. F. Quast, and P. G.  Watanabe.  The pharmacokinetics and
     macromolecular interaction of  perchloroethylene in mice and  rats  as  related
     to oncogenicity.  Toxicol. Appl.  Pharmacol.  55:207-219, 1980.

Stewart, R. D.  Acute tetrachloroethylene  intoxication.  J. Am. Med. Assoc.
     208(8):1490-1492, 1969.

Stewart, R. D., and H. C. Dodd.  Absorption of carbon  tetrachloride, tri-
     chloroethylene, tetrachloroethylene,  methylene chloride, and 1,1,1-trichloro-
     ethane through the human  skin.  J.  Ind. Hyg. 25:439-446, 1964.

Stewart, R. D., H. H. Gay, D.  S. Erley,  C.  L. Hake, and A. W. Schaffer.  Human
     exposure  to  tetrachloroethylene vapor.  Arch. Environ.  Health  2:40-46,  1961a.

Stewart, R. 0., E. D. Baretta, H. C. Dodd, and T. R. Torkelson.   Experimental
     human exposure to tetrachloroethylene.  Arch. Environ.  Health  20:224-229,
     1970.

Stewart, R. D., C. L. Hake, H. V. Forster, A. J.  Lebrun, J.  E. Peterson,  and
     A. Wu.  Tetrachloroethylene—development of  a biologic  standard for the
     industrial worker by breath analysis.  Report No. NIOSH NCOW-ENVM-PCE-74-6,
     1974.   172 pp.

Stewart, R.  D., C. L. Hake, A. Wu,  J.  Kalbfleisch, P.  E. Newton,  S.  K.  Marlow,
     and M.  V. Salama.  Effects of  perchloroethylene/drug  interactions on
     behavior  and neurological  function.   Final  Report.  Cincinnati, OH,  National
     Institute for Occupational Safety and Health, April 1977.

Stewart, R.  D.,  D. S.  Erley,  A. W.  Schaffer,  and  H.  H. Gay.  Accidental vapor
     exposure  to  anesthetic concentrations of  a  solvent  containing tetrachloro-
     ethylene.   Ind. Med. Surg. 30:327-330,  1961b.


003PE1/E                             5-31                              11/22/83

-------
Tada, 0., and K. Nakaaki.  Urinary excretion of alkali-pyridine reactants  in
     humans exposed to perchloroethylene vapor.  Rodo Kagaku 45:588, 1969.
     Cited in Chem. Abstr. 72:47126, 1970.

Takano, T. , and Y. Miyazaki.  Effect of chlorinated ethanes and ethylenes  on
     electron transport  in rat liver mitochondria.  J. Toxicol. Sci. 7:143-
     149, 1982.

U.S. Department of Health, Education and Welfare.  Public Health Service,
     National Institute  of Health, National Cancer Institute.  Bioassay of
     tetrachloroethylene for possible carcinogenicity.  DHEW Pub. No. (NIH)
     77-813, 1977a.

U.S. Department of Health, Education and Welfare, Public Health Service,
     National Institute  of Health, National Cancer Institute.  Bioassay of
     trichloroethylene for possible carcinogenicity.  DHEW Publication No.
     (NIH) 76-802, 1977b.

U.S. Department of Health, Education and Welfare, Public Health Service
     (NIH), NIEHS.  Vinyl chloride related compounds.  Environ. Health Persp.
     21:333, 1977c.

Vainio, H. M., G. Parkki, and J. Marniemi.  Effects of aliphatic chlorohydro-
     carbons on drug metabolizing enzymes in rat liver jji vitro.  Xenobiotica
     6(10):599-604, 1976.

Verberk, M.  M.  and T.  M. L. Scheffers.  Tetrachloroethylene in exhaled air of
     residents near dry-cleaning shops.  Environ. Res. 2J.:432-437, 1980.

Viola, P. L., A. Brigotti, and A. Caputo.  Oncogenic response of rat skin,
     lungs and bones to  vinyl chloride.  Cancer Res. 3_1:516, 1971.

Watabe, T., and E. W.  Maynert.  Role of epoxides in the metabolism of olefins.
     Pharmacologist 10:203, 1968.

Watanabe, P. G.  , R. H. Reitz, A. M. Shumann, M. J. McKenna, J. F. Quast, and
     P. J. Gehring.  Implications of the mechanisms of tumorigenicity for
     risk assessment.   Dev. Toxicol. Environ.  Sci.  6:69-89, 1980.

Waxweiler, R. J., et al.  Neoplastic risk among workers exposed to vinyl chloride.
     Ann. N. Y.  Acad.  Sci. 271:39, 1976.

Weiss, G.  A course of observations of trichloroacetic acid excretion in
     occupationally caused perchloroethylene poisoning.  Zentr. Arbeitsmed.
     Arbeitschutz. 19:143-146, 1969.  (German)

Yllner, S.  Urinary metabolites of C14-tetrachloroethylene in mice.  Nature
     191:820-821, 1961.

Ziglio, G.  Human exposure to environmental trichloroethylene and
     tetrachloroethylene:  preliminary data on population groups in
     Milan, Italy.  Bull. Environ. Contam. Toxicol. 26:131-136, 1981.
003PE1/E                             5-32                              11/22/83

-------
                               6.   TOXIC EFFECTS

6.1  HUMANS
     The known effects of tetrachloroethylene (PCE) on humans have been estab-
lished primarily from  individuals  accidentally  or occupationally exposed to
high (in some cases,  unknown) concentrations of  PCE.   Exposure to high concen-
trations of  PCE causes a variety of toxicological  effects in  humans.  Effects
upon the central nervous system (CMS)  are generally the most noticeable follow-
ing acute  or excessive  occupational  exposures.   Effects upon  the  liver and
kidney usually are observed after an elapsed period of exposure to high concen-
trations.

6.1.1  Effects on the Liver
6.1.1.1  Acute Exposure Situations—Transient, mild hepatitis was diagnosed by
Stewart (1969)  in a worker occupationally exposed  to  high,  anesthetic concen-
trations of  PCE in a  tank car  for  less  than  30  minutes.  Infrared analysis  of
the patient's  exhaled breath 1.5 hours  after exposure  showed  105 ppm  PCE (712
mg/m3).  Urinary urobilinogen  levels  were elevated on  the  ninth day of the
post-exposure  period.   The  serum  glutamic-oxaloacetic  transaminase  (SCOT)
level showed a slight increase on the third and fourth days.  Stewart concluded
this patient had  experienced marked depression of the  CMS  followed  by  tran-
sient, minimal liver injury.   The increase in urinary urobilinogen was suggest-
ed as one  indicator of hepatic injury caused by PCE.   Elevations in the levels
of urobilinogen and  other  indicators  of  liver  damage  have  been reported in
other  case studies  involving acute exposures (Stewart et al., 1961a; Saland,
1967).
     Stewart  et al.   (1961a)  reported a case in which  a male construction
worker  became  semicomatose after being  overexposed to  fumes from a  petroleum-
based  solvent,  during a  3.5-hour period.  The solvent  was reported  to contain
50 percent Stoddard  Solvent  and 50 percent  PCE.   A  neurological  examination
conducted  1 hour after  collapse indicated  no abnormality  of  function;  the
liver  was  not palpable.   During a 6-week period  subsequent to the  incident,
neither clinical  jaundice  nor  neurological  symptoms  were observed.   Simulated
exposure conditions  suggested  that the  estimated  average concentration  of  PCE
in  the work  environment,  during exposure,   was 393  ppm (2666 mg/m3).  The
average exposure for the first 3 hours was estimated to  be  275 ppm (1864 mg/m3).

003PE5/A                             6-1                              11/22/83

-------
The exposure for  the  remaining 30 minutes with  the  air hose turned off was
1100 ppm  (7458  mg/m3).   Clinical  tests revealed evidence  of  impaired  liver
function, beginning 9 days  following exposure; urinary  urobilinogen and total
serum bilirubin were  elevated.   On the fourteenth post-exposure day,  urinary
urobilinogen was in the normal range, but a slight elevation of alkaline phos-
phatase was observed.   Serum  glutamine-pyruvic transaminase (SGPT) and SGOT
values were in  the  normal range throughout the post-exposure period, with the
exception that  on  the eighteenth day, SGPT was  slightly elevated.   On the
sixteenth post-exposure day,  the  PCE concentration in expired air was sharply
elevated.   However, the overall decay curve slope was not altered appreciably.
The investigators suggested that such an acute exposure may represent a contin-
uing insult to the liver in view of the observations  that PCE had an exceedingly
long exponential  decay in  expired air,  indicating  slow  release  from  body
tissues.   On the twenty-first day, no  detectable PCE was  found  on the exposed
worker's breath.
     Elevated SGOT  values   and  one enlarged liver were  reported  by Saland
(1967).   Nine firefighters  were exposed  to an unknown  concentration of PCE
upon responding to a complaint about fumes.   All  the  firefighters were  exposed
without masks to  unknown  levels above the odor  threshold  for 3 minutes.  No
irritation of the  eyes, mucous  membranes, or respiratory  tract  occurred.  For
some inexplicable reason, all  the firefighters were admitted to a hospital  for
study 12  days after the  incident.  All clinical  tests indicated no  abnormali-
ties; however,  eight  of nine  individuals had elevated SGOT levels.   In seven
individuals, SGOT  values  had  returned to normal  by  22  days after  testing.
Hepatomegaly and splenomegaly were found in one individual 12 days post-exposure.
After 63 days post-exposure,  the liver was not palpable.
     An enlarged  liver  and  obstructive jaundice  were diagnosed  by Bagnell and
Ellenberger (1977)  in  a 6-week-old,  breast-fed infant.   While this situation
is not uncommon  in infants,  the  infant  had  been indirectly exposed to PCE.
The child's father  worked  as  a leather  and  suede  cleaner in a dry-cleaning
establishment where PCE vapors  were  present.   During regular lunchtime  visits
to the exposure  site,  the  mother had been exposed to the same vapors.   These
visits lasted between 30 and  60 minutes.   The concentration of PCE in the work
place was unknown,  although it  was believed to be  excessively high  because  of
reported episodes  of  dizziness.   In  the infant, bilirubin,  SGOT,  and  serum
alkaline  phosphatase  were   elevated; other  blood and urinary parameters of

003PE5/A                             6-2                              11/22/83

-------
liver function were normal.  Normal liver function was found in both parents.
Analysis of  the  mother's blood 2  hours  after one of her  lunchtime  visits
indicated a  PCE  level  of 0.3 mg per  100  ml.   One hour after a  visit,  her
breast milk contained 1.0 mg PCE per 100  ml.   After 24 hours,  the concentration
of PCE  in the breast milk decreased to 0.3 mg per 100 ml.   Chlorinated hydro-
carbons were not  found  in the mother's urine.   One week after breast feeding
was discontinued, serum bilirubin and serum alkaline phosphatase levels  in the
infant  returned  to  a  normal  range.   The  findings  suggest  that  the neonatal
liver may be sensitive to toxicological effects of PCE, although other causal
factors in this instance cannot be ruled  out.
     Levine et al. (1981) reported a diffuse and marked fatty  metamorphosis  of
the  liver of a  53-year-old male dry cleaner who succumbed to  an overexposure
of PCE  over  an  unspecified period of  time.  During autopsy, the PCE concen-
tration in the  liver  was 240 mg/kg.   PCE was also detected in the blood (4.5
mg/1), brain (69 mg/kg), kidney (71 mg/kg), and liver (30  mg/kg).   The authors
noted that the liver was  extremely fatty  and that an  ethanol concentration of
0.20 percent (w/v) was found in the blood.
     Hake and  Stewart (1977)  reported mild liver injury,  as  indicated by
elevated serum enzymes,  in a 60-year-old male  dry cleaning operator who was
overcome by  PCE  vapors.   The investigators estimated that the individual  had
been  lying in a  pool of  solvent for about 12 hours.   Although kidney and skin
damage  also  were  evident,  the  individual  was released after 21  days of  hospi-
tal ization and appeared fully recovered.
6.1.1.2   Chronic Exposure Situations--Hepatotoxic  effects  associated with
persons working  with unknown  concentrations of  PCE over extended periods have
been reported by a number of investigators (Coler and Rossmiller, 1953;  Franke
and  Eggeling,  1969; Hughes, 1954; Trense and  Zimmerman,  1969;  Meckler and
Phelps, 1966;  Larsen  et al.,  1977; Moeschlin, 1965; Dumortier et al., 1964).
Liver function parameters  that may be observed to be altered as a result of
excessive  PCE  exposure  include  sulfobromophthalein  retention time, thymol
turbidity,  serum bilirubin,  serum protein  patterns, cephalin-cholesterol
flocculation,  serum alkaline  phosphatase,  SGOT,  and  serum lactic acid de-
hydrogenase  (LDH).   However,  these parameters may be observed as a result of
many different causes that are completely dissociated with PCE.
     Effects observed after exposure to PCE at high or unknown levels included
cirrhosis of the liver  (Coler and  Rossmiller,  1953),  toxic  hepatitis  (Hughes,

003PE5/A                             6-3                              11/22/83

-------
1954; Meckler and  Phelps,  1966),  liver cell necrosis (Trense and Zimmerman,
1969; Meckler  and Phelps,  1966),  and enlarged liver (Meckler  and  Phelps,
1966).  In some cases, liver dysfunction parameters returned to normal  follow-
ing cessation of exposure (Hughes,  1954).   In one  case,  the liver was enlarged
6 months after cessation of exposure (Meckler and  Phelps,  1966).
     Liver dysfunction was evidenced in a 24-year-old male who was admitted  to
the hospital with  abdominal  pains  and blood-tinged vomitus  (Larsen  et al.,
1977).  Three days before  hospitalization,  this individual was exposed to PCE
vapors emanating from recently dry-cleaned clothes.  In  this study,  SCOT values
increased four  to  five  times above normal 1 day after initial symptoms.   Two
days  later,  SCOT  returned  to a normal  range.  The subject was found to have
renal insufficiency  and  mild proteinuria.   Despite diureses,  serum creatinine
and blood area  increased constantly.   Serum bilirubin was normal throughout
hospitalization.  Variations in the  levels of SCOT and other liver function
parameters during  the post-exposure  period indicate  that repeated testing
during this interval  is required for complete diagnosis.
     Larsen et  al. (1977)  also described an incident in which a  female dry-
cleaning worker was hospitalized in a comatose state with a grand mal  seizure.
Three days  prior to  hospitalization,  she had worn clothing  that had  been
recently dry-cleaned.  Upon  her admission,  SGOT and serum  LDH levels were re-
ported to be strongly elevated; levels gradually decreased during hospitalization.
Blood and protein  were  found in the urine.  Elevated serum creatinine levels
decreased after peritoneal dialysis.   A cause-effect relationship is unproven.
     Chmielewski et al.  (1976) found that the activities of alanine and aspara-
gine aminotransferase were significantly  elevated  (t „ __ = 2.032) in  a group
of 16 of 25 workers compared to non-exposed controls.   This group of 16 workers
had been exposed to PCE vapors in the range of 59  to 442 ppm (400 to 3000 mg/m3)
for periods ranging from 2 months to 27 years.   Aminotransferase activity in a
group of nine workers who were exposed to levels of PCE  at or below 29 ppm (200
mg/m3) was normal.  These enzyme imbalances were indicative of liver cell  injury
by PCE.   Low urinary  excretion of 17-ketosteroids  were observed in seven of  nine
persons whose exposures were less than 29 ppm and  4 of 16 persons whose exposures
ranged from 59 to 442 ppm PCE.   Abnormal electroencephalogram (EEC) tracings were
reported in 10 persons.   Six people were diagnosed as having a pseudo-neurotic
syndrome, and in four other cases the researchers  were led to diagnose or suspect
encephalopathy.

003PE5/A                             6-4                              11/22/83

-------
6.1.2  Effects on Kidneys
     Diminished excretion of urine (5 to 10 ml  of urine per hour), uremia, and
elevated serum creatinine  was  observed in a woman exposed to PCE vapors ema-
nating from clothes that had been dry-cleaned (Larsen et al., 1977).  As  pre-
viously described, she  had been admitted to the hospital  in a comatose state
with a grand  mal  seizure.   During hospitalization, urine  excretion and serum
creatinine  returned  to normal.   Renal  biopsy  suggested toxic nephropathy.
Liver dysfunction also  was evidenced by increased SGOT and  bilirubin levels
(Section 6.1.1).   Protein and blood were detected in the urine.   A cause-effect
relationship was unproven.
     In another  situation  in which  an  individual  (male)  had worn  clothing
permeated with PCE  vapors, elevated serum creatinine and  blood  uremia  were
observed (Larsen  et al. , 1977).   This  individual was admitted to  the hospital
with abdominal pains  and  blood-tinged vomitus.   Mild proteinuria  and leuko-
cytes and  erythrocytes were  found  in the  urine.   Despite diuresis,  serum
creatinine and blood urea  increased.  Serum creatinine decreased with peritoneal
dialysis, over a  19-day treatment period.   Renal biopsy evidenced  necrosis  in
the renal tubules.
     Hake and Stewart (1977) reported renal damage, indicated by a proteinuria
that lasted 20  days  and hematuria  lasting  for  8 days,  in  a  60-year-old male
dry-cleaning worker who was overcome by PCE vapors.   Liver damage and skin
damage also were  reported.  The  individual was reported fully recovered  upon
release from the hospital  (21 days post-exposure).

6.1.3  Effects on Other Organs/Tissues
6.1.3.1 Effects on the  Pulmonary  System—A  21-year-old  male was found  uncon-
scious in a library,  presumably  from a  massive but unknown  concentration of
PCE.  After his  admission  to  the  hospital,  the  principal  clinical  feature was
consistent  with  acute  pulmonary edema (Patel et al. ,  1977).  Bubbling  rales
were heard  over  the  entire lung  field.   Recovery  was  complete  4 days  after
hospital  admission.   Liver and kidney  function tests  in  this patient  were
normal.
     Levine et al. (1981)  reported pulmonary damage in a 53-year-old male dry-
cleaner who succumbed  upon overexposure  to PCE  vapors.  Autopsy  findings  were
congestion  superimposed on mildly fibrotic and diffusely emphysematous lungs
with  apical bullous emphysema.   The PCE  concentration in lung tissue was

003PE5/A                             6-5                              11/22/83

-------
30 mg/kg.   A co-worker, also overcome by vapors, was reported to have recovered
without complications  after hospitalization.   The  PCE  concentration in the
work place air at the time of the incident was not reported.
     Alkaline phosphatase in leukocytes is a defense mechanism against bacterial
infection, and  it  plays  an active part in phagocytosis.  In an investigation
of the  effects  of  PCE on alkaline phosphatase activity in human neutrophilic
leukocytes, Friborska  (1969) found that activity was within  the normal  range.
In this study of occupational exposure, seven workers were exposed to PCE and
four had  been exposed to both PCE and  trichloroethylene.   For controls, 20
unexposed  individuals  were  used.   Trichloroethylene exposure, as opposed to
PCE exposure, raised the activity of the alkaline phosphatase above the control
level.   For those  individuals  exposed to both  compounds,  no synergistic or
additive effect was observed.
     A  slight depression  in the  total white blood cell  count of three of nine
firefighters exposed for 3 minutes to unknown concentrations of PCE was reported
by Saland (1967).   The white cell count was made 12 days after the firefighters
were exposed to PCE.
6.1.3.2   Effects on the Skin--The effects  of  PCE upon  the skin range from a
mild to moderate burning sensation  when direct  contact occurs  for 5 to 10
minutes, to a marked erythema after prolonged exposure,  and finally,  blistering
if PCE  is trapped under clothing or in shoes (Hake and Stewart, 1977).
     Stewart and  Dodd (1964) reported  that  individuals experienced a  mild
burning sensation  on  their  thumbs after immersion of the thumbs in a solution
of PCE  for 5  to 10 minutes.  After  the  thumbs  were withdrawn, burning per-
sisted  without  a decrease in  intensity for 10  minutes  before  gradually sub-
siding  after 1  hour.   A  marked erythema was present in  all cases  and subsided
between 1 and 2 hours post-exposure.
     Morgan (1969)  reported erythema and blistering over  30 percent of the
body of a worker who was anesthetized for 30 minutes after PCE overexposure in
a coin-operated laundry.
     Ling  and Lindsay  (1971)  reported severe burns when an  individual,  upon
losing  consciousness,  fell  into  a pool of  PCE on the floor.   The  burns  gradu-
ally healed within 3 weeks following exposure.

6.1.4  Behavioral  and Neurological Effects
     In nearly all  the occupational  situations involving short-term high-level
exposures to PCE,  an initial characteristic response is depression of the CNS.
003PE5/A                             6-6                              11/22/83

-------
Subchronic exposures to high levels produce characteristics of a neurasthenic
syndrome; the most  frequently  reported  subjective complaints are dizziness,
headache, nausea,  fatigue,  and  irritation  of the eyes, nose,  and  throat.
These  symptoms  are typical  of many common  ailments.   Sensitivity  may vary
greatly among individuals.
6.1.4.1   Effects of Short-term Exposures—Stewart (1969)  reported  normal
neurological findings, except  for  the  Romberg test,  in an  individual  who was
found  unconscious  at  the  bottom of a  tank car.   Analysis of the man's breath
1.5 hours after  exposure  showed approximately 105 ppm PCE (712 mg/m3).  The
Romberg test is  designed  to detect swaying motions when the  subject stands
with eyes closed.   Upon  return to work, the  individual  reported  being very
fatigued after 4  hours  of light work.   The individual's fatigue with  slight
exertion diminished over  the  2 days following his return to work.   Abnormal
results of  the  Romberg  test are suggested  to be  the earliest indications of
signs of intoxication caused by PCE.
     Rowe et al.  (1952)  reported that six  individuals exposed to an average
PCE concentration  of  106  ppm (719 mg/m3) (range  = 83 to 130 ppm; 663  to 881
mg/m3) did  not  evidence  CMS effects.   At an average  PCE concentration of 216
ppm  (1465  mg/m3), all four  individuals  exposed  for  45 minutes to  2 hours
experienced slight  eye  irritation,  which developed 20 to 30 minutes into the
exposure period.   Minimal, transient eye irritation led the authors to suggest
that the vapor concentration causing this effect  in unacclimatized individuals
lies between 100  and  200 ppm (678 and 1356 mg/m3).   Dizziness and sleepiness
also were noted.   Recovery  from  all symptoms was  complete within 1  hour  after
exposure.  An exposure to an average concentration of 280 ppm PCE (1899 mg/m3)
for up to 2 hours resulted in complaints of 1ightheadedness, burning sensation
in the eyes, congestion  of frontal sinuses, and  tightness  about  the mouth.
Transient nausea was reported by one individual.   The subjects felt that motor
coordination was  impaired and mental  effort was  required  for coordination.
Motor  coordination  was  accomplished only with mental effort when two  indivi-
duals  were  exposed to an average PCE concentration  of 600  ppm  (4070  mg/m3).
Recovery  was  complete within  1 hour after exposure.   An  average  exposure
concentration of  1060  ppm (7190 mg/m3) for 1 minute  was intolerable to three
of four  individuals.   None  experienced  functional disturbances.  Recovery  was
rapid.
     No  behavioral  or  neurological  effects were  reported by Carpenter (1937)
when four individuals were exposed to 500 ppm (3391 mg/m3) PCE for 70  minutes.
003PE5/A                             6-7                               11/22/83

-------
Short-term exposures to higher concentrations resulted in subject's reports of
mental fogginess, lassitude, inebriation, loss of inhibition, and vertigo.   At
an exposure  level  of 1500 ppm (10,174 mg/m3),  shortness  of breath, nausea,
mental sluggishness,  and difficulties in maintaining  balance  were reported
during the post-exposure  period.  Tinnitus,  ringing of the  ears, was reported
upon exposure to 2000 ppm PCE (13,565 mg/m3) for 7.5 minutes.
     Weichardt and  Lindner  (1975) recorded  subjective  responses of  headaches,
giddiness, numbness,  alcohol  intolerance,  and intolerance  of  fats  and fried
foods as  a result  of exposures to between 11 to 45 ppm PCE (75 to 305 mg/m3)
for approximately 3 hours.  Very little information about the work environment
was available.   In  addition,  the study failed  to  include  suitable  controls.
     In a case  study involving a nursing mother exposed  daily for 30 to 60
minutes to unknown  but excessive PCE concentrations,  Bagnell and Ellenberger
(1977) reported subjective complaints of dizziness.
6.1.4.2   Effects of Long-term  Exposures—In  a comprehensive 3-month chamber
study of  six males and six females,  designed to elicit interactions between
ethanol and diazepam (Valium ) and PCE, Stewart and coworkers (Stewart et al.,
1977; Hake and  Stewart,  1977)  found that exposure  to 25 and 100 ppm PCE  (170
and 678 mg/m3)  alone or  in  combination with  diazepam or alcohol had no effect
on the EEC tracings.  A battery of neurological  and behavioral  tests,  consisting
of the  following,  were administered  at  the  peak blood levels  of  exposure:
Michigan  eye-hand  coordination,  rotary  pursuit, Flanagan coordination,  and
Saccade eye,  velocity, and dual attention  tasks.   EEG's  were recorded  for
spectral  density analysis during the exposures.  The  only  decrement  in  the
behavioral tests  reported by the authors was a "non-consistent significant
detrimental effect of PCE alone (100 ppm; 678 mg/m3) in the performance of the
Flanagan  coordination  test."   This  test requires subjects  to follow a spiral
pathway with a pencil, touching the  sides  of the  pathway  as  few times  as
possible.
     Each subject was exposed 5.5 hr/day.  The exposure duration was 11 weeks.
Monday and Tuesday were  generally control days.  Thursday was  an  intermediate
exposure  day.   Wednesday  and Friday were  100-ppm-PCE exposure  days.  No  other
unusual behavioral  or neurological  findings were  noted for exposures to PCE
alone.  However,  subjective complaints were noted for the  nine subjects who
completed the  study.   One subject accounted for one-third the incidence of
headache  complaints  and two-thirds the incidence of nausea  complaints reported.

003PE5/A                              6-8                              11/22/83

-------
The same  subject  complained  of eye, nose,  and throat irritation on all  but 8
of the  55 test days.  Analysis of the times of occurrence of these subjective
symptoms  revealed that  there was  no relationship to PCE vapor exposure.   In
fact, the incidence decreased  somewhat when the 100-ppm-PCE (678 mg/m3) expo-
sure concentrations were  compared  to non-PCE exposures.  The absence of EEC
abnormalities, such as were found in an earlier study by Stewart and co-workers
(1974), suggest that  EEC  observations may not be reliable indicators of early
signs of PCE narcosis.  In their earlier study,  impairment of the Flanagan co-
ordination test was also  occasionally noted during exposures to 150 ppm for
7.5 hours.   Chmielewski et al. (1976) observed abnormal EEC tracings in 10 of
16 individuals exposed occupationally, from 2 months to 27 years, to 59 to 442
ppm PCE (400 to 3000 mg/m3).   However, 6 of 16 cases were further diagnosed as
pseudo-neurotic syndrome, and  in 4  of 16 cases the  researchers  "were led to a
diagnosis or suspicion  at least of  encephalopathy."   No  additional  details
were given.
     Stewart et al.  (1970)  conducted an experiment with 16 healthy technical
employees ranging  in  age  from 24 to  64 years.   Five of the male  subjects,
ranging in  age  from 36 to 64  years,  were repeatedly exposed to  100 ppm (678
mg/m3)  PCE  for  7  hours on 5 consecutive days.  The remainder of the subjects
were exposed for one 7-hour period.  When queried every hour during the exposure
about  subjective  response,  25 percent of  the subjects reported that they  had
developed a  mild frontal headache.   Sixty  percent  complained  of mild  eye,
nose, or  throat irritation developing within the first 2 hours of exposure and
usually subsiding  before  7  hours had elapsed.  Forty percent of the subjects
reported  that  they felt  "slightly sleepy" when  inactive in the  chamber.
The  authors  wrote, "Unfortunately,  it  was  not  possible to confine these
same subjects  in  the  exposure  chamber for a  comparable  control  period,  so  the
clinical  significance  of  the slightly sleepy sensation cannot  be  assessed."
Twenty-five  percent of the  subjects "noted  some  difficulty in  speaking,
analogous to that noted during the  early phases  of  intoxication."   The  single
untoward  objective  response  was  an abnormal  Romberg test occurring in  three
of  the  subjects  within the  first 3 hours of exposure to 100 ppm (678 mg/m3).
With greater mental  effort,  these three individuals were  able  to perform a
normal  test  when given  a  second chance.   Those subjects repeatedly exposed had
fewer  subjective complaints.   One subject, who had  low-grade chronic sinusitis,
developed a  mild  frontal  headache during the course of each exposure.   Two out

003PE5/A                              6-9                              11/22/83

-------
of five subjects consistently reported mild eye and throat irritation and light-
headedness.   Sensations of sleepiness and speech difficulty were not reported.
All other neurological test results were normal.   The group exposed repeatedly
responded more  comparably  to  subjects in two  subsequent, more  sophisticated
3-week  studies  performed by  Stewart et al.  (1974 and  1977)  with repeated
exposure  to  100 ppm  (678  mg/m?)  PCE alone and  in combination  with either
diazepam or ethanol.
     Similar subjective  complaints,  as  well  as neurological  effects, of PCE
also were reported  in other studies  (Method,  1946;  Lob,  1957;  Eberhardt and
Freundt, 1966; Gold, 1969).

6.2  ANIMALS
     Reported toxic effects associated with PCE exposure in laboratory animals
include effects  on  the CNS, cardiovascular system,  skin, liver,  kidney, and
the immune system.
     A number of previous reviews (NIOSH, 1976; Fuller, 1976;  U.S. Environmental
Protection Agency,  1980; Walter et al., 1976; Parker et al. ,  1978; FishbeiX
1976; U.S.  Environmental  Protection Agency,  1977) support the  assessment of
the toxic effects  of PCE in animals  as  presented below.  Summaries of these
toxic effects and of toxic dose data appear in Tables 6-1 and  6-2.

6.2.1  Effects on the Nervous System
     Effects of acute exposure to PCE are very much dominated  by CNS depression.
Abnormal weakness,  handling intolerance, intoxication, restlessness, irregular
respiration, muscle incoordination, unconsciousness,  and ultimately, death are
among the symptoms,  considered  to be manifestations  of  effects on the CNS,
which have been observed in animals exposed to excessive levels.
     Symptoms of acute  CNS depression have been seen in experimental  animals
(National Institute  of Occupational Safety and  Health,  1976; Fuller,  1976;
U.S. Environmental  Protection  Agency, 1980;  Walter  et  al. , 1976;  Parker et
al., 1978;  Fishbein,  1976;  U.S.  Environmental  Protection  Agency,  1977) and  in
dogs treated  with  therapeutic  (anthelmintic)  doses  of  PCE  (Miller,  1966;
Snow, 1973;  Christensen and Lynch, 1973) PCE.
     Rowe et  al. (1952)  reported  that behavioral  changes  were not  observed  in
rats, guinea  pigs,  rabbits, or monkeys  exposed  repeatedly  for 7  hr/day at
vapor concentrations of PCE up to 401 ppm (2720 mg/m3).

003PE5/A                             6-10                             11/22/83

-------
                        TABLE 6-1.   SUMMARY OF THE EFFECTS OF TETRACHLOROETHYLENE ON ANIMALS
Animal
species
    Dose
concentration
   Route of
administration
Exposure variables
     Effects
    Reference
Rabbit
  (female)
Rabbit
Mouse
Guinea
 pig
  N/A
  13 mmole/kg
  200 ppm
Mouse
Guinea
pig
Guinea
pig
200 ppm
100 ppm
200 ppra
  400  ppm
  skin
  oral
  inhalation
                                inhalation
                                inhalation
                                inhalation
  inhalation
 single application
   single instillation
   (eye)

 single dose
                                                  4 hours
                                                  single exposure
                                     4  hours/day
                                     6  days/week
                                     1-8 weeks

                                     7  hours/day
                                     5  days/week
                                     132 exposures

                                     7  hours/day
                                     5  days/week
 7 hours/day
 5 days/week
 169 exposures
primary eye and
  skin irritant
marked increase in
  serum enzymes, i.e. ,
  alkaline phosphatase,
  SCOT, and SGPT within
  24 hours

moderate fatty infiltration
  of the liver 1 day
  after exposure but not
  3 days after

fatty degeneration of
  the liver
                                               increased liver weights
                                                 in  females
increased liver weights
  with some fatty degenera-
  tion in both males and
  females - slight increase
  in lipid content and
  several small fat vacuoles
  in liver

more pronounced liver
  changes than at 200 ppm,
  slight cirrhosis was
  observed - increased liver
  weight, increase in neutral
  fat and esterified choles-
  terol in the liver, moderate
  central fatty degeneration,
  cirrhosis
Ouprat et al., 1976



Fujii et al., 1975





Kyi in et al., 1963




Kyi in et al. , 1965



Rowe et al.  , 1952



Rowe et al., 1952
Rowe et al. ,  1952

-------
                                                                      TABLE 6-1.  (continued)
               Animal
               species
    Dose
concentration
   Route of
adainistrati on
               Guinea
                pig
(Ti
 I
               Rabbit
               Rat
               Monkey

               Rabbit
              Rat


              Rat





              Rat





              Rabbit


              Rabbit
Exposure variables
     Effects
    Reference
  2500 ppm
  inhalation
  100-400 ppn



  2500 ppm



  2500 ppra


  1600 ppm





  3000-6000





  15 ppn
  2212 ppm
  Q5 ara/1)
  inhalation
  inhalation
  inhalation





  inhalation


  Inhalation
 18 7-hour
 exposures
 7 hours/day
 5 days/week
 6 months

 28 7-hour
 exposures
inhalation
Inhalation
1-13 7-hour
exposures
18 7- hour
exposures
 single exposure
 up to 8 hours
 3-4 hours/day
 7-11 months

 45 days
 4 hrs/day
 5 days/week
loss of equilibrium,
  coordination, and strength,
  increase in weights of liver
  and kidney, fatty degenera-
  tion of the liver, cloudy
  swelling of tubular epithe-
  lium of the kidney

no abnormal growth,
  organ function, or
  histopathologic findings

central nervous system
  depression without
  unconsciousness

loss of consciousness
  and death

drowsiness, stupor, increased
  salivation, extreme restless-
  ness, disturbance of equili-
  brium and coordination, biting
  and scratching reflex

increase in liver weight, in-
  crease in total lipid content
  of liver accompanied by a few
  diffusely distributed fat
  globules

depressed agglutlnin
  formation

liver damage
  indicated by elevated
  SGPT, SCOT. SGLOH:
  marked reduction of
  Schmidt index
Rowe et al., 1952
Rowe et al., 1952



Rowe et al., 1952



Rowe et al., 1952


Rowe et al., 1952





Rowe et al., 1952





Hazza, 1972


Mazza, 1972

-------
                                                        TABLE 6-1.  (continued)
Animal
species
Rat



Rat




Rat







Rat

Rat

Rabbit

Dose Route of
concentration administration Exposure variables
70 ppra inhalation 8 hours/day
5 days/week
150 exposures
(7 months)
230 ppm inhalation 8 hours/day
5 days/week
150 exposures
(7 months)

470 ppm inhalation 8 hours/day
5 days/week
150 exposures
(7 months)




2750-9000 inhalation single exposure
ppn
19000 ppm inhalation 30-60 minutes

15 ppm inhalation 3-4 hours/day
7-11 months
Effects Reference
no pathological findings Carpenter, 1937



similar, but less severe, Carpenter, 1937
pathological findings as
with 470 ppm - congestion
and light granular swelling
of kidneys
congested livers with cloudy Carpenter, 1937
swelling, no evidence of
fatty degeneration or
necrosis: evidence of kid-
ney injury - increased
secretion, cloudy swelling.
and desquamation of kidneys:
congestion of spleen
no deaths Carpenter, 1937

congested livers with granular Carpenter, 1937
swelling, some deaths
moderately increased Navrotskii et al., 1971
urinary urobi 1 inogen,
Rabbit
2211 ppm
(15 mg/1)
inhalation
45 days
  pathomorphological
  changes in the
  parenchyma of liver
  and kidneys

significant reduction
  of glomerular filtration
  rate and the renal
  plasma flow; decrease
  of highest excretory
  tubular capacity
  (kidney damage)
                                                                                                               Brancaccio et al.,  1971

-------
                                                                       TABLE 6-1.  (continued)
CTi
Animal Dose
species concentration
Mouse 2.5 ml/kg
(Swiss
male,
10 animals)

House 5.0 ml/kg
(10
Animals)

Rabbit 2211 pprn
(15 mg/1)

House N/A
House N/A
Dog
Dog
Dog
Rat 300 ppro
Route of
administration Exposure variables
intraperitoneal

intrapeHtoneal
(urine samples were collected 24 hours
inhalation 45 days

intraperitoneal
intraperitoneal
intraperitoneal
intraperitoneal
intraperitoneal
inhalation 7 hours/day
days 6-15 of
gestation
Effects
100 mg percent or more
protein found in one of
six mice - proximal con-
voluted tubules were
swollen in all animals
and necrotic in one
two of four mice had
100 mg percent or
more protein in urine
post-injection)
increased plasma and urine
levels of adrenal cortical
and adrenal medullar hor-
mones; increased excretion
of principal catecholamine
metabolite (not statistically
significant)
liver dysfunction
L050
elevated SGPT
caused phenol sulfo-
nephthalein retention
indicating kidney dysfunction
LD50
decreased maternal
weight gains,
increased fetal
Reference
Plaa and Larson, 1965



Mazza and Brancaccio,
1971

Klaassen and Plaa, 1966
Klaassen and Plaa, 1966
Klaassen and Plaa, 1967
Klaassen and Plaa, 1967
Klaassen and Plaa, 1967
Schwetz et al. ,
1975
                                                                                              reabsorptions

-------
                                                        TABLE 6-1.   (continued)
Animal
species
    Oose
concentration
   Route of
administration
Exposure variables
     Effects
                                                                                                         Reference
House
  300 ppm
  inhalation
Rat
              44.2 ppm
Mouse         15-74 ppm



Rat           15 ppm
fiat
Dog
(male
beagles)
  73  and
  147 ppm
  0.5-1.0%
  v/v
  5000 &
  10000 ppm
                    inhalation
                    inhalation
                    inhalation
                                inhalation
  inhalation
 7 hours/day
 days 6-15 of
 gestation
                    entire gestation
                    period
                    5 hours/day
                    3 months
                    4 hours/day
                    5 months
                    4  hours/day
                    4  weeks
 7  minutes  house  air
 followed by 10
 minutes of tetrachloro-
 ethylene 8 M9/k9
 epinephrine given  I.V.
 (1)  a  control dose
 after  2 minutes  of
 breathing  air (2)  chal-
 lenge  dose  after 5  min-
 utes of breathing  test
 compound
maternal liver weights
  increased relative to
  body weight; increased
  incidences of fetal
  subcutaneous edema,
  delayed ossification of
  skull bones, and split
  sternebrae

decreased levels of ONA
  and total nucleic acids
  in the liver, brain,
  ovaries, and placenta

decreased electroconductance
  of muscle and "amplitude"
  of muscular contraction

EEC changes and proto-
  plasmal swelling of
  cerebral cortical cells,
  some vacuolated cells and
  signs of karyolysis

EEC and electromyogram
  changes; decreased
  acetylcholinesterase
  activity

cardiac sensitization
  (development of serious
  arrhythmia or cardiac
  arrest) was  not induced
  at the concentrations
  tested (other similar
  compounds gave positive
  results at same concen-
  tration,
                                                                                                                 Schwetz et al.,
                                                                                                                 1975
                                                                Aninina, 1972
                                                                                                     Dmitrieva,  1968
                                                                                                     Omitrieva,  1966
                                                                                                                 Dmitrieva, 1966
Reinhardt et al. ,  1973

-------
                                                       TABLE 6-1.  (continued)



CTi
M
CTi



Animal
species
Cat
Cat
House
Mouse
Rabbit
Cat
Dog
Dose
concentration
3000 ppm
14,600 ppm
40 rag/1
5,900 ppm
4-5 ml/kg
5 ml/kg
4 ing/ kg
9000 ppm
Route of
administration Exposure variables
inhalation 4 hours
inhalation 1-2 hours
inhalation
oral
oral (in oil)
oral (in oil)
inhalation
Effects
no anesthesia
anesthesia
minimal fatal concentration
death in 2-9 hours from
central nervous system
depression
death in 17-24 hours
death within hours
narcosis, marked
Reference
Lehmann, 1911


Lehmann and Schmidt-
Kehl, 1936
Lamson et al . ,
Lamson et al . ,
Lamson et al . ,
Lamson et al . ,
Lamson et al . ,
1929
1929
1929
1929
1929
                                                                              salivation, "narrow
                                                                              margin of safety"
Dog
4-25 ml/kg
oral (in oil)
death in 5-48 hours
Lamson et al. ,  1929

-------
                                                                    TABLE 6-2.  TOXIC DOSE DATA
(Ti
Description
of exposure*
LOsb
LDsb
E050
LD&0
ED50
LOsb
E050
E050
'-Dso
ED50
LOso
LD50
LDSO
LCLo
L050
Species
mouse (male)
mouse
mouse
mouse
mouse
dog
dog
dog
mouse
mouse
mouse
mouse
mouse
mouse
rat
Route of
administration
oral
interperitoneal
interperitoneal
interperitoneal
interperitoneal
interperitoneal
interperitoneal
interperitoneal
subcutaneous
subcutaneous
oral
(undiluted)
oral
(in oil)
oral
inhalation

Dose
concentration
6100 mg/kg
2.9 ml/kg
28 mM/kg
4700 mg/kg
2.9 ml/kg
28-32 mM/kg
34 mM/kg
24 mM/kg
2.1 ml/kg
21 mM/kg
3400 rag/ kg
0.74 ml/kg
7.2 mM/kg
1.4 mf/kg
390 mM/kg
27 mM/kg
0.109 ml
0.134 ml
8850 mg/kg
23000 mg/m3

Toxic effect
endpoint
death
death
liver
dysfunction
death
liver toxicity
death
liver damage
kidney
dysfunction
death
liver toxicity
death
death
death
death
death
Time
36 hr
24 hr

24 hr
24 hr
24 hr
24 hr
10 days

unknown
unknown
unknown
2 hr

Reference
Wenzel and Gibson, 1951
Klaassen and Plaa, 1966

Gehring. 1968
Klaassen and Plaa, 1967
Klaassen and Plaa, 1967
Klaassen and Plaa, 1967
Plaa et al. , 1958
Plaa et al. , 1958
Dybing and Dybing, 1946
Dybing and Oybing, 1946
Handbook of Toxicology, 1959

Withey and Hall, 1975

-------
                                                                        TABLE 6-2.  (continued)
 I
M
CO
Description
of exposure
LCLo
LCLo
LDLo
LDLo
LOLo
LDLo
Species
rat
rat
dog
dog
cat
rabbit
Route of
administration
inhalation
inhalation
oral
intravenous
oral
oral
Dose
concentration
4000 ppm
4000 ppm
4000 mg/kg
85 rag/ kg
4000 mg/kg
5000 mg/kg
Toxic effect
endpoint
death
death
death
death
death
death
Time
4 hr
4 hr
unknown
unknown
unknown
unknown
Reference
Handbook of Toxicology, 1959

Arch. Hyg. Bakteriol.
116:131, 1936
Carpenter et al. , 1949
Clayton, 1962
Lamson et al. , 1929
                    LCL   -  the  lowest concentration of a substance, other than an LCSu, in air which has been reported to have caused death in
                    humans  or animals.

                    LDL   -  the  lowest dose of a substance, other than an LDSo, that is introduced by any route other than inhalation over any
                    given period of time and reported to have caused death in humans or animals introduced in one or more divided
                    portions.

-------
     A single 4-hour  exposure  to 2270 ppm PCE  (15400  mg/m3)  caused rats to
suffer an 80-percent  loss of both avoidance and escape responses (Goldberg et
al., 1964).  Savolainen  et  al.  (1977) demonstrated behavioral impairment in
rats exposed  to 200  ppm  (1357  mg/m3) PCE vapor for 6  hr/day for 4 days.
Marked increases in  the  frequency of ambulation in the open  field were most
significant 1 hour after exposure when these responses  were compared to control
responses.   High tissue  concentrations  of PCE were detected in fat and brain
tissue after a relatively short exposure.   A significant decrease in the ribo-
nucleic acid (RNA) content of the brain was measured as well as an increase in
nonspecific cholinesterase activity.
     Dmitrieva and coworkers  (Dmitrieva,  1966;  Dmitrieva and Kuleshov, 1971;
Dmitrieva et al.,  1968; Dmitrieva, 1973; Dmitrieva, 1968) have reported altera-
tions of EEC patterns in rats exposed to as low as  15 ppm PCE (102 mg/m3) 3 to
4 hr/day for  7  to 11 months.  Such effects have not been observed by Western
investigators at much higher levels  nor were details provided in the reports
of these Russian investigators to adequately assess the quality of the studies.

6.2.2  Effects on the Liver and Kidney
     PCE is generally regarded  as being both  hepatotoxic and  nephrotoxic when
exposure is excessive and prolonged.
     Carpenter (1937) exposed three groups of 24 albino rats each to PCE vapor
concentrations averaging  70, 230, or  470  ppm  (475,  1560, or 3188 mg/m3)  for  8
hr/day, 5  days/week,  for  up  to  7 months.   The maximum exposure for any  animal
during the  7-month period was 150 days  (1200  hours).  A group  of 18  unexposed
animals served as controls.
     The rats  exposed to 470 ppm  (3188 mg/m3)  for 150 days  followed  by a
46-day rest period developed cloudy  and congested  livers with  swelling;  there
was  no evidence of  fatty degeneration or  necrosis.  These  rats also had  in-
creased renal secretion  with cloudy  swelling and desquamation of kidneys, as
well as congested spleens with increased pigment.   The pathologic changes were
similar but less  severe  in  the rats  exposed to 230 ppm PCE (1560 mg/m3).  In
some instances,  there was congestion  and light granular swelling of the kidneys
after 21 exposure days.   After 150 days  of  exposure,  followed by a 20-day
rest, congestion was  found in the kidney and spleen.  The livers showed reduced
glycogen storage.   Microscopic  evidence of damage  to liver, kidney,  or  spleen
in rats exposed at 70 ppm (475 mg/m3) for  150 days was not observed.  In addi-
tion, microscopic examination  of heart, brain, eye, or  nerve  tissue did  not
003PE5/A                             6-19                             11/22/83

-------
reveal any damaging effects in any of the chronically exposed rats.   Functional
parameters, including  icteric  index,  Van den Bergh  test  for bilirubin,  and
blood  and  urine analysis,  were  normal after the  exposures.  -Fertility  of
female rats, as measured by a fertility index (actual number of litters/possible
number of  litters), was  increased  slightly after repeated exposures to 230 or
470 ppm PCE (1560 or 3188 mg/m3).   No deaths or signs of anorexia or diminished
activity were observed during the chronic exposures.
     Carpenter also tried  to determine the highest concentration of PCE vapor
that would  not  anesthetize rats  exposed for 8 hours.  Exposure to 31,000 ppm
(210,273 mg/m3) was  lethal  within a few minutes.   Rats exposed to 19,000 ppm
(128,871 mg/m3) died  after 30  to 60 minutes.   Animals  that were exposed to
19,000 ppm  (128,877 mg/m3)  and removed from  the inhalation  chamber just prior
to unconsciousness developed congestion  and  granular swelling of the liver.
Similar liver effects  were seen  after exposure at 9000  ppm (61,047 mg/m3).
There was  also marked  granular swelling of the  kidneys.  A  single exposure at
9000, 4500, or 2750 ppm  (61,047,  30,523, or  15,261 mg/m3) did not cause death
to any of  the  rats in this study;  however,  post-mortem examinations  of  the
rats exposed to those  concentrations revealed only  a slight increase in the
prominence of liver and kidney markings.
     Rowe et al.  (1952) exposed rabbits,  monkeys,  rats, and guinea pigs  to PCE
vapor for  7  hours,  5  days/week,  for up to 6 months.   Exposure concentrations
ranged from 100 to 2500  ppm (678  to  16,957 mg/m3).   Three of the four species
tested -- rabbits,  monkeys, and rats -- showed no  effects of repeated  exposures
to concentrations,  up  to  400 ppm (2713 mg/m3).  There were no adverse  effects
on growth,  liver weight,  or lipid content,  or  gross or microscopic anatomy
observed in any animal.  In contrast, guinea pigs  showed marked susceptibility
to PCE  in this study.   The liver weights of female  guinea pigs increased
significantly after 132  7-hour  exposures at 100 ppm (678 mg/m3).  At  200 ppm
(1356 mg/m3), there was  a slight depression  of growth in female guinea pigs
and  increased liver weights in both males and  females.   Slight  to moderate
fatty degeneration of  the  liver  also was observed.  These  effects were more
pronounced in guinea pigs  that received  169  7-hour exposures at  400 ppm (2713
mg/m3).  At this concentration,  there also were increased amounts of  neutral
fat  and esterified  cholesterol  in livers.   Gross  and microscopic examination
of the tissues  revealed  slight to moderate  fatty  degeneration  in the liver
with  slight  cirrhosis.  Rowe  et  al.  stated  that at 395  ppm (2680 mg/m3),

003PE5/A                             6-20                             11/22/83

-------
increased kidney weights  also  were observed in guinea pigs but not in other
species.   Guinea pigs  have  been  shown to be extremely sensitive to toxicity
testing.
     Klaassen and Plaa (1967) showed that intraperitoneal  injections of PCE to
mongrel male and female dogs can  produce damage to the kidney and  liver.   They
estimated the ED50  (effective  dose in 50 percent of  the animals tested) for
liver  and kidney damage  as  well  as the 24-hour LD50 value (lethal  dose in 50
percent of the animals treated).   The ED50 values were measured by sulfobromo-
phthalein (BSP),  SGPT, glucose,  protein, and  phenolsulfonephthalein  (PSP),
indicators of liver or kidney dysfunction.  The LD50 was 2.1 ml/kg (21 mmoles/
kg) and  the  ED50  for elevation of  SGPT was 0.74 ml/kg (7.2 mmoles/kg).  The
ED50  for diminution of PSP excretion was 1.4  ml/kg (14 mmoles/kg).  After
administration,  effects on the liver and kidneys were determined by microscopic
examination.  At near-lethal doses,  PCE produced moderate neutrophilic infil-
trations  in  the  sinusoids and portal areas; necrosis was not observed.   Near
the EDr0, vacuolization of centralobular hepatocytes in about half the animals
was observed.  After  a single interperitoneal  injection at 0.75 x 1-0™,  SGPT
peaked at 2 days and rapidly declined throughout the 9-day measurement period.
Kidney dysfunction  was deemed  significant when  PSP  excretion was less than  39
percent  (determined  24 hours after interperitoneal injection).  At near  PSP
excretion--EDcn doses, only mild dilation of the collecting  ducts was seen  in
some of  the kidneys.
     In  a similar  study  using Swiss-Webster mice,  Klaassen  and Plaa  (1966)
determined  that  the 24-hour LD™ was 2.9 ml/kg (28 mmoles/kg).  The ED™ for
BSP retention was  2.9 ml/kg (32 mmoles/kg).   The  ED5Q  for  elevation  of SGPT
was 2.9  ml/kg (28 mmoles/kg).  When  ethanol  (5 gm/kg) was administered  by
gavage for  3 days  prior to injection of 1.0 ml/kg PCE, neither PSP excretion
nor BSP   retention  was significantly altered.   At BSP-ED™, mice exhibited
predominantly inflammatory  changes with trace to marked  quantities of lipid
accumulation.
     Effects  upon  the livers  of  rats exposed continuously for  3 months to  0.7
                             3
and 2.7  ppm (4.5 and 19  mg/m ) were reported by Bonashevskaya  (1977).  In  the
low-exposure  group, an increase  in  the  activity  of succinate  dehydrogenase
(SDH)  was reported.   At  the high  level,  SDH  was slightly decreased  in the
central  lobular  sections while activity  in the  peripheral  zones  was  either
unchanged or increased.   The activities  of glucose-6-phosphate dehydrogenase
(G6PDH)  were similar in  pattern  to that for SDH.   RNA content  was  reported to
003PE5/A                             6-21                             11/22/83

-------
be decreased.   LDH  activity  was either unchanged or  increased.   The  enzyme
variations  indicated  the subject's adaptation to exposure.   No significant
histomorphological  changes  in the  liver  were reported  at  either exposure
level.
     A subcutaneous injection  (0.4  ml  daily,  for 8 weeks) of  PCE  to groups  of
rats that  were fed a diet containing  high protein had  less  of an adverse
effect upon the  liver than  PCE alone  (Dumitrache et al.,  1975).   PCE was
observed to  induce  liver  hypertrophy compared to the  reaction of  the controls
(p <0.001).  Hypertrophy  was  most pronounced in the  low-protein  group.   In
treating rats  fed a diet containing low protein, cholesterol (p  <0.001) and
total liver  lipids  (p <  0.001) were elevated compared to the reaction of the
controls.   Twelve rats were used in each of the four groups.
     Kyi in et al.  (1963)  noted moderate fatty degeneration of the liver with a
single 4-hour  exposure to 200  ppm PCE  (1356 mg/m3) in female  albino mice that
were sacrificed 1 day after  exposure.   Degeneration was not observed in mice
sacrificed 3 days after exposure.  The mice were exposed to PCE concentrations
of 200,  400, 800,  or 1600 ppm (1356, 2713,  5426,  or 10,852 mg/m ) for 4 hours.
Tissues  were studied  microscopically to assess the extent of  necrosis  and  fat
infiltration of the liver.   Moderate  to massive infiltration was observed in
                                                            3
mice killed 1 or  3  days after exposure at 400 ppm (2713 mg/m ) or more, but no
cell necrosis  was observed  even after 4 hours exposure  up to  1600 ppm PCE
            3
(10,852 mg/m ).   Kylin et al.  (1963)   exposed four groups  of 20 albino mice
                          3
to 200 ppm  PCE  (1356  mg/m ).   Each  group was  exposed  for 4  hr/day, 6 days  per
week, for  1, 2,  4, or 8 weeks.   Microscopic  examinations  were performed on
livers and  kidneys  of the exposed mice and controls.   Fatty degeneration was
particularly marked and  tended to be more  severe with longer  exposure  to PCE.
Chemical  determination of the  liver fat content was performed in addition to
the histologic  examination.  Correlation between the  histologically evaluated
degree of fatty degeneration and the concentration of extracted fat was +0.74.
Liver fat  content of  the exposed animals  was  between 4 and  5  mg/g of body
weight,  as  compared to 2  to  2.5  mg/g for the  control  animals.   The actual  fat
content of  the  livers did not  increase with  duration of exposure as did the
extent of  the  fatty infiltration.   No liver  cell necrosis  was  observed.   No
effect on the kidneys was reported.
     Mazza  (1972)  exposed 15 male rabbits  for 4 hr/day,  5 days  a  week, for  45
                                   3
days, to  2790  ppm PCE (18,924 mg/m ).    Mazza looked at the effect of PCE on

003PE5/A                             6-22                             11/22/83

-------
serum enzyme levels in an attempt to determine the specific location of initial
liver injury as well as the severity of the damage to the  liver.  The Schmidt
Index, which is the  sum of SCOT and the SGPT divided by the serum glutamate
dehydrogenase (GDH),  was used as an indication of hepatic disorders.   Enzymatic
determinations  were made before exposure and 15,  30,  and 45 days after exposure
to PCE.  All three of the enzymes  showed an increase in activity, but the GDH
increased the most;  GDH reduced the Schmidt  Index from 6.70 to 1.79.  Mazza
concluded that  this reduction indicates the prevalence of mitochondrial  injury
over cytoplasmic injury in the liver.
     Mazza and  Brancaccio  (1971)  exposed  10 rabbits  for 4 hr/day,  5 days per
                                               3
week, for 45 days, to 2790 ppm  PCE  (18,924 mg/m  ).  These  investigators  found
a moderate, but not  statistically significant,  increase in levels  of adrenal
cortical and medullar  hormones—plasma and urinary corticosteroids and cate-
cholamines--including increased excretion of 3-methoxy-l-hydroxymandelic- acid,
the principal catecholamine metabolite.
     In another study,  Brancaccio  et al. (1971)  exposed 12 male rabbits  for  4
                                                                   3
hr/day, 6 days  per week,  for 45 days,  to 2280 ppm PCE (15,465 mg/m ) to look
at effects  on  kidney function.   They noted a reduction in glomerular filtra-
tion and renal  plasma  flow and a  decrease  in the maximum tubular  excretion
when  measured  upon  cessation  of  the  exposure regimen.   Brancaccio  et al.
concluded that PCE causes kidney damage, primarily in the renal tubule.   These
findings agreed with earlier histological  findings of Pennarola and Brancaccio
(1968)  in  which kidney  injury,  following  exposure to  PCE,  appeared to be
primarily in the renal tubule.
     Plaa and  Larson (1965)  dosed  mice with PCE  by interperitoneal  injection.
Ten mice received  2.5  mg/kg and 10 others  received 5.0 mg/kg.   Urine samples
were  collected  from surviving  mice 24 hours  after  the injection  of PCE.
Protein was  found  in the  urine  of  one  of the  six surviving mice injected with
the  lower dose  and in two of  four survivors  of  the higher dose at  levels of
100 or more mg percent.  None of the survivors had greater than 150 mg percent
glucose  in  the urine.   The  kidneys  of the mice given  the  lower dose were
examined microscopically.   The proximal convoluted tubules were swollen in all
animals and necrotic in one.
     Fujii  (1975)  observed an  increase in serum  enzyme  activities (i.e.,
alkaline phosphatase,  SCOT,  and SGPT)  within 24 hours after a single dose of
13 mmole PCE/kg given orally to rabbits.  These changes in serum enzyme activi-
ties, indicative of  liver damage, were mild and transient.
003PE5/A                             6-23                             11/22/83

-------
6.2.3  Effects on the Heart
     The possible  cardiovascular  effects  of PCE have not been systematically
investigated.   Reinhardt  et al.   (1973)  noted that PCE  does  not  appear to
sensitize the myocardium  to epinephrine.   In this study of dogs, a response
that was considered to be indicative of cardiac sensitization was the develop-
ment of  a seriously  life-threatening arrhythmia or cardiac arrest following a
challenge dose  of epinephrine.   PCE inhalation exposure for 10 minutes at
                                                            3
concentrations  of  5000  or 10,000 ppm (33,915 or 67,830 mg/m ) did not result
in a positive response in any of the 17 dogs.
     The investigators  noted  the possibility that PCE  has the  potential for
cardiac sensitization, as do many organic solvents.   However, the PCE concentra-
tion that failed  to  elicit a positive response (10,000 ppm) would be a level
immediately hazardous to life, based on CNS effects.
     Christensen and Lynch  (1933) observed depression of the heart and respira-
tion in  five  dogs, each given a single oral dose  ranging from 4 to 5.3 ml/kg
PCE  (approximately the  LD5Q for  dogs).  Autopsy showed fatty infiltration of
both heart  and  liver tissue.   The small  intestine was  extremely shriveled  and
showed marked inflammation.
6.2.4  Effects  on the Skin  and Eye
     Duprat et  al. (1976) have shown PCE to be a primary eye and skin irritant
in rabbits.   Instillation of the chemical into the eye produced conjunctivitis
with epithelial  abrasion.   However,  healing  of  the ocular mucosa was  complete
within 2 weeks.   PCE had a severe  irritant  effect when  a single application
was made to the skin of the rabbit.
6.3  ADVERSE EFFECTS OF SECONDARY POLLUTANTS
     The  level  of phosgene derived from photodecomposition  of PCE  is not
likely to result  in  serious long-term effects.   In  occupational settings  or
under certain  conditions  in which phosgene  is  directly  formed  at high tem-
peratures from  halocarbons (cigarette  smoke, welding), sufficient warning
from the generation  of extremely  irritating  hydrogen  chloride  vapors would
prevent exposure to harmful concentrations of phosgene.
6.4  SUMMARY OF ADVERSE HEALTH EFFECTS AND ASSOCIATED  LOWEST OBSERVABLE
     EFFECT CONCENTRATIONS
6.4.1  Inhalation  Exposure
     A number  of  case reports  describe accidental or occupational exposure to
PCE.   However,  the duration and  extent  of  exposure  either were unknown or
involved excessively  high concentrations.   The few  controlled  human  studies
003PE5/A                             6-24                             11/22/83

-------
available generally provide  information  on  effects resulting from short-term
                                          3
exposures at levels near 100 ppm (678 mg/m ).   Effects associated with chronic
exposures, on the other hand, are available  from animal  experiments.
6.4.1.1  Effects Associated With Intermittent or Prolonged Exposure—The data
available from both human and animal exposures to  excessive concentrations of
PCE indicate that the CNS,  liver, and kidneys  may be adversely affected.   In a
controlled  study  involving  humans,  Stewart  and coworkers  (1977)  exposed 12
                                                     3
individuals  to 25  and 100 ppm  PCE  (170  and 678 mg/m ), 5.5  hr/day,  for 3
                                                3
months (55  exposures).  At  100  ppm  PCE (678 mg/m )  there was  a non-consistent
detrimental effect upon coordination, as  measured by the Flanagan test, but no
adverse effects  on  the remainder of a battery of neurological and behavioral
tests and  no alterations in  the EEC patterns.  There were  no  observed  effects
at 25 ppm (170 mg/m3).
     Both subchronic  and  chronic inhalation exposures of animals support the
observations in humans.  Carpenter  (1937) observed dose-related congestion and
swelling  in  the  livers and kidneys  of rats exposed for 8  hr/day, 5 days per
                                                            3
week, for  7  months,  to 230 and  470 ppm  (1559 and 3187 mg/m  ).   Rowe et al.
(1952) observed  degenerative  changes in  the livers of guinea pigs,  a species
that appears more  sensitive to PCE, following an 8-month exposure to 200 and
                           3
400 ppm (1356 and 2712 mg/m ),  for  7 hr/day, 5 days per week.
     Carpenter (1937)  reported  a no-observable-effects  level  (NOEL) at 70  ppm
             3
PCE (475 mg/m ) in. rats exposed 8 hr/day, 5  days per week, for 7 months.   His-
tologic  examination  of the  liver,  kidneys,  spleen, heart, brain, eye,  and
nerve tissue,  as  well as blood and  urine analyses, were performed.   The NOEL
is defined  as  that exposure level  at which  there are no statistically signi-
ficant  increases  in  frequency  or  severity of effects  between  the  exposed
population and the appropriate  control.
6.4.1.2   Effects Associated With Short-term Exposure—Controlled studies  in
humans have  provided  information on a progression  of  effects upon  the CNS,
ranging from lightheadedness to narcosis, and subsequently to death.   A summary
of the  estimated dose-response relationships  for  acute effects of single,
short-term exposures  of humans  is presented below:
          >4000 ppm PCE:  possibly life-threatening
           2000 ppm PCE:  light narcosis, possible liver
                          damage
           1000 ppm PCE:  lightheadedness
003PE5/A                             6-25                             11/22/83

-------
              200 ppm PCE:   possible reversible fatty changes
                            in the liver
              100 ppm PCE:   possible first signs of CNS depression,
                            giddiness, slight eye irritation
               75 ppm PCE:   very slight eye irritation
                5 ppm PCE:   odor threshold
     Stewart et al.  (1970)  reported 1ightheadedness in 25 percent of the 15
volunteers exposed  for  7  hours to an average  PCE  concentration of 101 ppm
         3
(685 mg/m ).   Two  subsequent  studies by Stewart et al.  (1974,  1977),  using
more updated technology and  testing methodology, did not confirm this  result
even when exposures  to  100  ppm PCE were combined  with  diazepam or alcohol.
Rowe et al.  (1952)  reported 1ightheadedness  in four individuals exposed for 2
                               3
hours to  216 ppm PCE  (1465 mg/m ), but  not in  six  individuals exposed 2 hours
                    3
to 106 ppm (719 mg/m ).   The observations in these studies follow the expected
pattern for  a  non-specific anesthetic  effect.  These  results  show  that  a
short-term exposure  to  PCE  in the range of about 100 to 200 ppm (678 to 1356
    3
mg/m ) will  result  in some  individuals experiencing the first gross signs of
CNS depression and  behavioral  alterations.   Test  results also  indicate that
short-term exposures  may  damage  the liver.   Kyi in et  al.  (1963)  reported
transient moderate fatty infiltration in livers of mice exposed for 4 hours to
                      3
200 ppm PCE  (1356 mg/m  ).  However,  liver damage in humans is generally asso-
                                                                         3
ciated with  short-term  exposures  greatly in excess of  100  ppm (678 mg/m ).
6.4.2  Oral Exposure
     As summarized  in Table  6-2,  the  acute oral  toxicity of  PCE  has  been
determined in  rats,  mice,  cats,  rabbits, and  dogs.  There are  no subchronic
oral exposure  studies and  only one chronic study—the NCI bioassay.   Neither
of these  studies  identifies  either a NOEL or  lowest-observed-adverse-effect
level (LOAEL).
6.4.3  Dermal Exposure
     Although PCE  can be absorbed  through unbroken skin,  absorption was esti-
mated to  be  minor  (Stewart  and Dodd, 1964).   Toxic quantities would probably
not be absorbed through this route.
003PE5/A                             6-26                             11/22/83

-------
6.5  REFERENCES FOR CHAPTER 6


Aninina, T.  Effect of aliphatic hydrocarbons and fluorinated and chlorinated
     derivatives on the content of nucleic acids in animal tissues during
     embryogenesis.  Tr. Permsk. Cas. Med. Inst.  110:69-71, 1972.

Archiv Hyg. Bakteriol. (Munchen)  116:131, 1936.

Bagnell, P. C.; H. C. Ellenberger.  Obstructive jaundice due to a chlorinated
     hydrocarbon in breast milk.  J. Can. Med. Assoc. 117: 1047-1048, 1977.

Blair, A., P. Decoutle; D. Graumen.  Causes of death among laundry and
     dry-cleaning workers.  Am. J. Public Health 69(5): 508-511,  1979.

Bonashevskaya, T.  I.  Certain Results of a morphological and functional
     investigation of the lungs in a hygienic assessment of atmospheric
     pollution.  Gig. Sanit. 2:15-20, 1977.   (Translation)

Brancaccio, A., V. Mazza; R. DiPaola.  Renal  function in experimental
     tetrachloroethylene poisoning.  Folia Med.  54:233-237, 1971.   (English
     translation)

Brown, D.  P.  Memorandum.  Division of Surveillance, Hazard Evaluations, and
     Field Studies, National Institute for Occupational Safety and Health.
     August 24, 1978.

Carpenter, C. P.   The chronic toxicity of tetrachloroethylene.  J. Ind. Hyg.
     Toxicol. 19:323-336, 1937.

Carpenter, C. P.,  H.  F. Smyth, Jr.; V. C. Pozzani.  The assay of  acute
     vapor toxicity and the grading and  interpretation of results on 96
     chemical compounds.  J. Ind. Hyg. Toxicol.  31:434,  1949.

Chmielewski, J., R. Tomaszewski;  P. Glombiowski; W. Kowalewski; S. R.
     Viwiatkowski; W. Szozekocki; A. Winnicka.  Clinical  observations of
     the occupational exposure to tetrachloroethylene.  Bull. Inst.  Marit.
     Trop. Med. Gdynia 27(2):197-295, 1976.

Christensen, B. V., and H. J. Lynch.  The effect of anthelmintics on the
     host.  I. Tetrachloroethylene  II.  Hexylresorcinol.   J. Pharmacol.  Exp.
     Thera. 48:311-316, 1933.

Clayton, J. W.  The toxicity of fluorocarbons with special reference to chemi-
     cal constitution.  J. Occup. Med.   4:262-273, 1962.

Coler, H.  R.; H.  R.  Rossmiller.   Tetrachloroethylene exposure in  a small
     industry.   Ind.  Hyg. Occup.  Med. 8:227,  1953.

Dmitrieva,  N. V.   Maximum permissible concentration of tetrachloroethylene in
     factory air.   Hyg. Sanit.  ^31:387-393, 1966.  (English translation)

Dmitrieva,  N. V.   Bioelectric activity and electric conducting properties  of
     muscles exposed  to chlorinated  hydrocarbons.  Farmakologiya  i Toksikologiya
     31(2):228-230,  1968.   (English  translation)


003PE5/A                              6-27                              11/22/83

-------
Dmitrieva, N. V.  Changes in the bioelectrical activity in the cerebral cortex
     of rats with the narcotic effect of substances with different polarization
     properties.  Experimental1 naya Khirurgiya i Anestezidogiya  6:72-75,
     1973.   (English translation)

Dmitrieva, N. V., and E. V.  Kuleshov.  Changes in the bioelectric activity and
     electric conductivity of the brain in rats chronically poisoned with
     certain chlorinated hydrocarbons.   Hyg.  Sanit.  36:23-29, 1971.  (English
     translation)

Dmitrieva, N. V.; E. V.  Kuleshov; E. K. Orjonikidze.  Changes in the
     impedance and bioelectrical activity of the cerebral cortex of rats under
     the action of anesthetic drugs.  Zhur. vysshei Nervnoi Deyatel 'nosti
     18(3):463-468, 1968.  (English translation)

Dumitrache, S., I. Gontea, and V. Stanciu.  Role of proteins in the resistance
     of the organism to tetrachloroethylene.   Revista Igiena Bact. Virus Parazit.
     Epidem. Pneum. Igiena 24(3):147-151, 1975.  (translation).

Dumortier, L.;  G. Nicolas, F. Nicolas.   A case of hepato-nephritis syndrome
     due to perchloroethylene.  Arch. Mai. Prof. 25:519-522, 1964 (English
     translation)

Duprat, P.;  L.  Delsaut; D. Gradiski.  Irritant potency of the principal
     aliphatic chloride solvents on the skin and ocular mucous membranes of
     rabbits.  Europ. J. Toxicol.  3:171-177, 1976.

Dybing, F. ; 0.  Dybing.  The toxic effect of tetrachloroethane and tetra-
     chloroethylene in oily solution.  Acta Pharmacol.  2:223-226, 1946.

Eberhardt, H. ;  K. J. Freundt.  Tetrachloroethylene poisoning.  Arch. Toxikol.
     (Berlin) 21:338-351, 1966.  (English translation)

Fishbein,  L.  Industrial mutagens and potential mutagens..  I.  Halogenated
     aliphatic derivatives.   Mutat. Res.  32:267-308, 1976.

Franke, W.;  F.  Eggeling.  Clinical and statistical studies on employees of
     chemical cleaning plants exposed to perchloroethylene.  Med. Welt 9:453-460,
     1969.   (English translation)

Friborska, A.  The phosphatases of peripheral white blood cells in workers
     exposed to trichloroethylene and perchloroethylene.  Brit.  Med. J. Ind.
     Med.  26:159-161, 1969.

Fujii, T.  The variation in the liver function of rabbits after administration
     of chlorinated hydrocarbons.  Jpn. J. Ind. Health  17:81-88, 1975.
     (English translation)

Fuller, B. B.  Air pollution "assessment of tetrachloroethylene."  Mitre
     Technical  Report - 7143.  February 1976.

Gehring, P.  Hepatotoxicity of various chlorinated hydrocarbon vapors relative
     to their narcotic and lethal properties in mice.  Toxicol.  Appl.  Pharmacol.
     13:287-298,  1968.

Gold, J. H.  Chronic perchloroethylene poisoning.  Can. Psychiatric Assoc. J.
     14:627-630,  1969.
003PE57A                             6-28                             11/22/83

-------
Goldberg, M. E.; H. E. Johnson; U. C. Pozzani; H. F. Smyth, Jr.  Effect of
     repeated inhalation of vapors of industrial solvents on animal behavior.
     I.  Evaluation of nine solvent vapors on pole-climb performance in rats.
     Am. Ind. Hyg. Assoc. J.  25:369-375, 1964.

Hake, C. L.; R. D. Stewart.  Human exposure to tetrachloroethylene:  inhala-
     tion and skin contact.  Environ. Health  Persp. 21:231-238, 1977.

Handbook of Toxicology, Volumes II-V, Philadelphia:  W. B. Saunders Co.,  1959.
     Volume V., p. 76.

Hughes, J.  P.  Hazardous exposure to a so-called safe  solvent.  J. Am. Med.
     Assoc. 156:234-237, 1954.

Klaassen, C. D.; G. L. Plaa.  Relative effects of various chlorinated
     hydrocarbons on  liver and kidney function in mice.  Toxicol.  Appl.
     Pharmacol.  9:139-151, 1966.

Klaassen, C. D.; G. L. Plaa.  Relative effects of various chlorinated
     hydrocarbons on  liver and kidney function in dogs.  Toxicol.  Appl. Pharmacol
     10:119-131, 1967.

Kylin,  B.;  I. Sumegi; S. Yllner.  Hepatotoxicity of inhaled trichloroethylene
     and tetrachloroethylene - long-term exposure.  Acta Pharmacol. Toxicol.
     (Kbh).  22:379-385, 1965.

Kylin,  B. ;  H. Reichard;  I. Sumegi; S. Yllner.  Hepatotoxicity  of  inhaled
     trichloroethylene,  tetrachloroethylene,  and chloroform—single exposure.
     Acta Pharmacol.  Toxicol.  20:16-26, 1963.

Lamson, P.  D.; B. H.  Robbins; C.  B. Ward.  The pharmacology and toxicology
     of tetrachloroethylene.  Am. J. Hyg.  9:430-444,  1929.

Larson, N.  A.; B. Nielsen; A. Ravin-Nielsen.   Perchloroethylene intoxica-
     tion—a hazard in the use of coin laundries.   Ugeskr. Laeg.  39(5):270-275,
     1977.   (English  translation)

Lehmann, K. B.   Experimental  studies on the  influence  of technically and
     hygienically  important gases and vapors  on  the organism.  Arch. Hyg.
     74:1-60,  1911.   (German)

Lehmann, K. B.; L. Schmidt-Kehl.  The 13 most important chlorinated  hydro-
     carbons of the aliphatic series from the standpoint of occupational
     hygiene.  Arch.  Hyg.  116:131-268, 1936.   (German)

Levine, B.; M.  F.  Fierro;  S. W. Goza; J. C.  Valentour.  A tetrachloro-
     ethylene  fatality.  J. Forensic Sci. 26(1):206-209,  1981.

Ling,  S.; W. A. Lindsay.   Perchloroethylene  burns.  Brit. Med. J.  3(5766):115,
     1971.

Lob, M.  Dangers  of perchloroethylene.  Arch.  Gewerbepath. Gewerbehyg. 16:45-52,
     1957.   (English  translation)
003PE5/A                              6-29                              11/22/83

-------
Mazza, V.  Enzyme changes in experimental tetrachloroethylene  intoxication.
     Folia Med.  55(9-10):373-381, 1972.  (English translation)

Mazza, V.; A. Brancaccio.  Adrenal cortical and medullar hormones  in
     experimental tetrachloroethylene poisoning.  Folia Med.   54:204-207,
     1971.  (English translation)

Meckler, L. C.; D. K. Phelps.  Liver disease secondary to tetrachloroethylene
     exposure.  J. Am.  Med.  Assoc. 197(8):144-145, 1966.

Method, H. C.  Toxicity of tetrachloroethylene.  J. Am. Med. Assoc. 131:1468,
     1946.

Miller, T. A.  Anthelmintic activity of tetrachloroethylene against various
     stages of Ancylostoma cam'urn in young dogs.  Am. J. Vet.  Res.  27(119):
     1037-1040, 1966.

Moeschlin, S.  Poisons, diagnosis and treatment.  New York, N.Y.:  Gruner  and
     Stratton, 1965.

Morgan, B.  Dangers of perchloroethylene.  Brit. Med. J. 2:513,  1969.

National Institute for Occupational Safety and Health.  Criteria for a  recom-
     mended standard...occupational exposure to tetrachloroethylene (perchloro-
     ethylene).  HEW publication no. (NIOSH) 76-185.  U.S. Department of HEW,
     PHS, CDC, NIOSH.  July, 1976.

Navrotskiy, V. K.; Kashin, I.  L. Kulinskaya; L. F. Mikhaylovskaya; L. M.
     Shuster, Z. N. Burlaka-Vouk; B. V. Zadorozhniy.  Comparative  assessment
     of the toxicity of a number of industrial poisons when inhaled in  low
     concentrations for prolonged periods.  Trudy S'ezda Gigen Vkramkoi:
     SSR 8:224-226, 1971.  (translation)

Parker, J. C.; L. J. Bahlman;  N. A. Feidel; H. P. Stein; A. W. Thomas;  B.  S.
     Woolf; E. J. Baier.  Tetrachloroethylene  (perchloroethylene).  Current
     NIOSH Intelligence Bulletin #20.  Am. Ind. Hyg. Assoc. J.   39:3, 1978.

Patel, R. ; N. Janakiraman; W.  D. Towne.   Pulmonary edema due to  tetrachloro-
     ethylene.  Environ. Health Persp. 21:247-249, 1977.

Plaa, G. L.;  R. E. Larson.  Relative nephrotoxic properties of chlorinated
     methane, ethane, and ethylene derivatives in mice.  Toxicol.  Appl.
     Pharmacol.  7:37-44, 1965.

Plaa, G. L.;  E. A. Evans; C. H. Hine.  Relative hepatotoxicity of  seven
     halogenated hydrocarbons.  J. Pharmacol.  Expt. Ther.  123:224-229, 1958.

Reinhardt, C. F. ; L. S. Mullin; M. B. Maxfield.  Epinephrine-induced cardiac
     arrhythmia potential of some common  industrial solvents.  J.  Occup. Med.
     15:953-955, 1973.

Rowe, V. K.;  D. D. McCollister; H. C. Spencer; E. M. Adams; D. D.  Irish.
     Vapor toxicity of tetrachloroethylene for laboratory animals  and human
     subjects.  Arch. Ind. Hyg. Occup. Med.  5:566-579, 1952.


003PE5/A                             6-30                            11/22/83

-------
Saland, G.   Accidental exposure to perchloroethylene.  N. Y. State J. Med.
     67:2359-2361, 1967.

Savolainen, H.;  P. Pfaffli; M. Tegen; H. Vainio.  Biochemical and behavioral
     effects of inhalation exposure to tetrachloroethylene and dichloromethane.
     J. Neuropathol.  Exp. Neurol.   36:941-949, 1977.

Schwetz, B. A.,  B. K. Leong; and P. J. Gehring.  The effect of maternally
     inhaled trichloroethylene, perchloroethylene, methyl chloroform, and
     methylene chloride on embryonal and fetal development in mice and rats.
     Toxicol.  Appl. Pharmacol.  32:84-96, 1975.

Snow, 0. H.  The effects of pyrantel pamoate and tetrachloroethylene on
     several blood enzyme levels in the greyhound.  Aust. Vet. J.  49:269-272,
     1973.

Stewart, R. D.   Acute tetrachloroethylene intoxication.  J. Am. Med. Assoc.
     208(8):1490-1492, 1969.

Stewart, R. D.;  H. C. Dodd.  Absorption of carbon tetrachloride, trichloro-
     ethylene, tetrachloroethylene, methylene chloride,  and 1,1,1-trichloroethane
     through the human skin.  Am.   Ind. Hyg.  Assoc. J. 25:439-446, 1964.

Stewart, R. D.;  D. S. Erley; A. W. Schaffer; H. H. Gay.  Accidental  vapor
     exposure to anesthetic concentrations of a solvent  containing tetrachloro-
     ethylene.   Ind.  Med. Surg.  30:327-330, 1961a.

Stewart, R. D.;  H. H. Gay; D. S. Erley; C. L. Hake; A. W. Schaffer.  Human
     exposure to tetrachloroethylene vapor.   Arch. Env.  Health. 2:40-46,
     1961b.

Stewart, R. D.;  E. D. Baretta; H.   C. Dodd; T. R. Torkelson.   Experimental
     human exposure to tetrachloroethylene.   Arch. Environ. Health 20:224-229,
     1970.

Stewart, R. D.;  C. L. Hake; H. V.   Forster; A. J.  Lebrum; J. E. Peterson;
     A. Wu.  Tetrachloroethylene:   development of a biological standard  for
     the industrial worker by breath analysis.  Report no. NIOSH-MCOW-ENVM-
     PCE-74-6.   National Institute of Occupational Safety and Health, 1974.

Stewart, R. D.; C. L. Hake; A. Wu; J. Kalbfleisch; P. E. Newton; S.  K. Marloro;
     M. V.  Salama.  Effects of perchloroethylene/drug interaction on
     behavior and  neurological function.  Final report,  National Institute  for
     Occupational  Safety and Health, April 1977.

Trense, E.; H.  Zimmerman.  Fatal inhalation poisoning with chronically-acting
     tetrachloroethylene vapors.   Zbl. Arbeitsmed. _19: 131-137,  1969.  (English
     translation)

U.S. Environmental Protection Agency.  An assessment  of  the need for limitations
     on trichloroethylene, methyl  chloroform, and perchloroethylene.  Draft
     final  report, Volumes I,  II,  III.  Office of Toxic  Substances.  EPA contract
     no. 68-01-4121,  1977.
003PE5/A                             6-31                              11/22/83

-------
U.  S. Environmental Protection Agency.  Ambient water quality criteria for
     tetrachloroethylene.  EPA 440/5-80-073, 1980.

Walter, P.; A. Craigmill; J. Villaume; S. Sweeney; G. L. Miller.  "Chlorinated
     hydrocarbon toxicity (1,1,1-trichloroethane, trichloroethylene and tetra-
     chloroethylene)" - a monograph.  Nat. Tech.  Infor. Serv.   Springfield,
     Va.  PB-257 185/9, May 1976.

Weichardt, H; J. Linder.  Health hazards caused by perchloroethylene  in
     dry-cleaning plants from the point of view of occupational medicine and
     toxicology.  Staub-Reinhalt.   Luft 35(11):416-420, 1975.   (English transla-
     tion)

Wenzel, D. G.; R. D. Gibson.  Toxicity and anthelmintic activity of
    ' n-butylidine chloride.  J. Pharm. Pharmacol.  3:169-176, 1951.

Withey, R. J.; J. W. Hall.   The joint action of perchloroethylene with
     benzene or toluene in rats.  Toxicol.  4:5-15,  1975.
003PE5/A                              6-32                              11/22/83

-------
         7.   TERATOGENICITY,  EMBRYOTOXICITY,  AND REPRODUCTIVE EFFECTS


     Because of its widespread use,  PCE has been studied for teratogenic

potential.   Teratology studies have  been performed in rats,  mice,  and rabbits,

using doses of PCE which, in some studies,  produced slight signs of maternal

toxicity.  Other studies in chicken  embryos (Elovaara et al., 1979) have

indicated that PCE disrupts embryogenesis in a dose-related  manner.   However,

since administration of PCE directly into the air space of chicken embryo is

not comparable to administration of  dose to animals with a placenta, it is not

possible to interpret this result in relationship to the potential of PCE to

cause adverse effects in animals or  humans.

     The following discussion of studies subscribes to the basic viewpoints

and definitions of the terms "teratogenic"  and "fetotoxic" as summarized and

stated by the U.S. Environmental Protection Agency (1980):


          Generally,  the  term  "teratogenic"  is  defined as  the tendency  to
     produce physical  and/or  functional  defects in offspring  i_n  utero.   The
     term "fetotoxic"  has  traditionally been used to describe a wide variety
     of  embryonic  and/or  fetal  divergences from the  normal  which cannot be
     classified as  gross  terata (birth  defects)  -- or which  are of  unknown or
     doubtful  significance.   Types  of effects which  fall  under  the  very  broad
     category  of  fetotoxic effects are  death,  reductions in fetal  weight,
     enlarged  renal  pelvis edema,  and  increased incidence  of supernumerary
     ribs.   It should be emphasized, however, that the phenomena of terata and
     fetal   toxicity  as  currently defined are  not separable  into precise  cate-
     gories.   Rather,  the spectrum of  adverse  embryonic/fetal  effects is
     continuous,  and all deviations from  the  normal  must be considered  as
     examples of developmental  toxicity.  Gross morphological terata represent
     but one  aspect  of  this  spectrum,  and while  the  significance of such
     structural changes is more  readily evaluated, such effects are not neces-
     sarily more  serious  than certain effects which  are ordinarily  classified
     as  fetotoxic--fetal death  being the most obvious example.

          In view  of the spectrum of effects at issue, the Agency suggests
     that  it might be useful  to  consider  developmental  toxicity  in terms  of
     three  basic  subcategories.  The first  subcategory would be embryo  or
     fetal   lethality.   This  is,  of course,  an  irreversible effect  and  may
     occur with or without the  occurrence of gross terata.  The second subcate-
     gory would be teratogenesis and would encompass those changes  (structural
     and/or functional) which are induced prenatally, and which are  irreversible.
     Teratogenesis includes structural defects apparent  in the fetus, functional
     deficits  which  may become  apparent only  after birth, and any other  long-
     term effects  (such as carcinogenicity) which are attributable  to i_n utero
     exposure.  The  third category  would be embryo or  fetal toxicity as com-
     prised of those  effects which  are potentially reversible.  This subcategory
     would  therefore include  such effects as weight reductions,  reduction in
     the degree of  skeletal  ossification,  and delays  in organ maturation.


003PE2/E                             7-1                               11/22/83

-------
          Two major problems with a definitional scheme of this nature must be
     pointed out, however.   The first is that the reversibility of any phenom-
     enon is extremely  difficult  to prove.   An organ such as the kidney, for
     example, may  be  delayed  in development and then  appear to "catch up."
     Unless a series  of specific  kidney function tests are  performed  on the
     neonate, however,  no  conclusion  may be drawn concerning permanent organ
     function changes.   This  same  uncertainty as  to possible  long-lasting
     aftereffects  from  developmental  deviations is  true for  all examples of
     fetotoxicity.   The  second problem  is that the  reversible nature of an
     embryonic/  fetal effect  in one species  might, under a given agent,  react
     in another species in a more serious and  irreversible manner.
7.1  ANIMAL STUDIES
7.1.1  Mice
     Schwetz et al. (1975) reported a study of Swiss-Webster mice exposed to
                                                          3
PCE via inhalation at concentrations of 300 ppm (2034 mg/m ) for 7 hours daily
on days 6 through 15 of presumed gestation.  Day 0 of gestation was designated
the day a vaginal plug was observed.  This concentration was cited as twice
the maximum allowable exclusion limit for human industrial exposure, with the
                           ®                     •*
Threshold Limit Value (TLV)  of 100 ppm (678 mg/m ).   Concurrent controls were
exposed to filtered air.
     Following maternal Caesarean-sectioning on day 18 of gestation, all
fetuses were examined for external anomalies.   One-half the fetuses were
examined for soft tissue malformations using a free-hand sectioning technique,
and the other one-half of the fetuses were cleared, stained, and examined for
skeletal malformations.  One fetus in each litter was processed and examined,
using histopathological techniques.  Seven litters were examined.  The pups in
the exposed group were significantly smaller,  as measured by decreases in
fetal body weight.  Also, slight but not statistically significant increases
in the number of runts were observed, as well  as increases in the numbers of
fetuses with subcutaneous edema, delayed ossification of the skull, delayed
ossification of the sternebrae, and splits in sternebrae.  No other remarkable
malformations were reported in fetuses.  Increases in the absolute and relative
mean maternal liver weights were reported.  No evidence of teratogenicity of PCE
was found at the concentration tested.
7.1.2  Rats
     Schwetz et al. (1975) also administered PCE to Sprague-Dawley rats by
                              3
inhalation (300 ppm; 2034 mg/m ) for 7 hours daily, on days 6 to 15 of gesta-
tion.  Control rats were exposed to filtered air.  Day 0 of gestation was

003PE2/E                             7-2                               11/22/83

-------
designated as the day when spermatozoa were observed in smears of vaginal
contents.   Rats were sacrificed on day 21 of gestation.  Caesarean sections
were performed, and fetuses were examined for external  malformations.   One-
half of the fetuses in each litter was examined for soft tissue malformations
and the remaining one-half of the fetuses were examined for skeletal malforma-
tions.   One fetus from each litter was randomly selected for serial sectioning
and histological evaluation.  Average maternal body weight gain was slightly
reduced in the rats exposed to PCE.   A slight but statistically significant
increase in resorption was reported in 9 of 17 PCE-exposed litters evaluated.
Exposure of dams to PCE produced no effect on the average number of implanta-
tions per litter, fetal sex ratios,  or fetal body measurements.  No soft
tissue or skeletal anomalies were reported in the offspring of rats exposed to
PCE.  No evidence of teratogenicity of PCE was found at the concentration
tested.
     Beliles et al. (1980) exposed Sprague-Dawley rats to PCE at 300 ppm
          3                                                           •
(2034 mg/m ) for 7 hours daily, 5 days per week.   Controls were administered
filtered air.  One-half of the rats were exposed for 3 weeks prior to mating.
All rats were exposed during gestation on either days  0 through 18 or days 6
through 18, with day 0 designated as the day when spermatozoa were observed in
smears of vaginal contents.  It should be noted that the information in the
report indicated that a Monday through Friday treatment period was used.   It
is  unclear, based on information in the report, if the rats were indeed exposed
on  the gestational days cited.  Three rats (days 6-18) died on the second  day
of  pregestational treatment.  Signs of ataxia and loss of balance were observed
in  all of the other rats of this group on the same day.  The authors thought
this response was most likely due to the high levels in the inhalation chamber
during the last 2 hours of  the day.  Measurements taken 15 minutes before  the
                                              3
end of the exposure showed  568 ppm (4061 mg/m ) but could have been higher
previously.  The maternal body weight gain in the PCE-treated  rats was not
statistically different from that in controls during the pregestational period.
Inhibition of maternal body weight gain occurred during the first week, as
well as increases  in mean absolute, but not  relative,  kidney and liver weights.
No  embryotoxic  effects were observed which were attributable to maternal
exposure  to  PCE, except for delays in skeletal ossification.   This  effect,
however,  is  thought to be a reversible effect and is not considered a malformation
as  such.  No teratogenic effects were observed.

003PE2/E                             7-3                                11/22/83

-------
     Nelson et al. (1980) evaluated the ability of PCE to elicit a behavioral
teratogenic response in Sprague-Dawley rats.   In a pilot dose-range finding
study, groups of 3 rats were exposed by inhalation to 1800 or 3600 ppm (12,204
              3
or 24,408 mg/m ) PCE.   Narcotization was observed in dams.   Therefore, a
                                   3
concentration of 900 ppm (6102 mg/m ) PCE was used in the pilot study.  Signs
of maternal toxicity such as severe reductions in average food intake and
decreased average body weight gain were observed.  Food consumption and body
weight gain were also reduced but were not statistically significant in rats
                             3
exposed to 900 ppm (6102 mg/m ) PCE on days 14 to 20 of gestation.   Dams and
pups exposed to 100 ppm (678 mg/3) PCE on days 14 to 20 of gestation did not
show any adverse effects as compared to the controls.
     In the behavioral testing study, 102 pregnant rats were exposed to PCE by
inhalation as follows:

     (1)  900 ppm, days 7-13 of gestation (N=19)
     (2)  900 ppm, days 14-20 of gestation (N=21)
     (3)  sham exposed, days 7-13 of gestation (N=13)
     (4)  sham exposed, days 14-20 of gestation (N=19)
     (5)  100 ppm, days 14-20 of gestation (N=15)
     (6)  sham exposed, days 14-20 of gestation (N=15)

     Seven behavioral tests were performed on days 4 through 46 postparturition,
using one male and one female per litter.  One group of rats, consisting of a
male/female pair from each litter, was tested for ascent on a wire mesh
screen and rotorod balancing.  One male and one female rat per litter were
tested for open-field activity, activity wheel, and avoidance conditioning.  A
third pair was tested for operant conditioning.  Catecholamines (norepinephrine
and dopamine), acetylcholine, and protein, measured in the brain tissue, were
evaluated in 10 pups (no more than 2 per litter per treatment group) at birth,
or at 21 days postparturition.  Histopathological evaluation of brains for
neuropathology was performed in an unreported number of pups.
                                                 3
     Rats from dams exposed to 900 ppm (6102 mg/m ) PCE on days 7 to 13 of
gestation, performed less well on discrete testing of ascent and rotorod
tests, but only on certain days of testing.  Offspring exposed to 900 ppm
          3
(6102 mg/m ) on days 14 to 20 of gestation performed less well on one test day
in the ascent test, but later performed better than controls in the rotorod
test, and were relatively more active than controls in the open-field tests.
Acetylcholine levels were reduced in 21-day-old rats of dams exposed to 900 ppm

003PE2/E                             7-4                               11/22/83

-------
          3
(6102 mg/m ) PCE.   Dopamine levels were reduced in rats of dams exposed on
days 7 to 13 of gestation.
                                                      3
     In the group of rats exposed to 100 ppm (678 mg/m ) PCE during days 14-20
of gestation, no significant differences were observed between the offspring
of these animals and their controls on any of the behavioral tests.  The
authors summarized that "there were generally few behavioral or neurochemical
                                                                               o
differences observed between offspring of animals exposed to 900 ppm (6102 mg/m )
PCE during either days 7-13 or 14-20 of gestation.   When significant differ-
ences did appear, they occurred more often when the group exposed during days
14-20 of gestation was compared with its control group."
     It should be noted that the field of behavioral teratology is in its
early stages of development (Buelke-Sam and Kimmel, 1979), and such behavioral
alterations cannot at this time be interpreted in terms of human effect.  It
also must be noted that these observed changes may be related to maternal
toxicity and do not represent a direct toxic effect.
     Tepe et al. (unpublished, 1982) exposed Long-Evans hooded female rats to
                               3
1000 ± 125 ppm (6780 ± 847 mg/m ) PCE vapors to ascertain if exposure before
mating and during pregnancy was more detrimental to the embryo than exposure
during pregnancy alone.  Four treatment groups (30 rats each) were utilized in
a two-by-two factorial design:  exposure to PCE for two weeks (6 hours daily,
5 days per week) before mating, through day 20 of pregnancy; PCE before mating
and filtered air during pregnancy; filtered air before and PCE during pregnancy;
and filtered air throughout.  One-half the dams in each treatment group were
sacrificed on day 21 by ether anesthesia.  Elevations in relative maternal
liver weights and reduction in fetal body weight were observed without altera-
tion in maternal weight-gains in groups exposed during pregnancy.  An excess
of skeletal  variations were seen in the group exposed before mating and during
pregnancy; and excessive soft tissue variations (e.g. kidney dysplasia) occurred
in the group exposed during pregnancy alone.  These effects are consistent
with embryotoxicity.   Elevation in ethoxycoumaren dealkylase activity (an
indicator of P45Q activity) was observed in maternal livers but not fetal
livers with  pregnancy  exposure.  Ethoxyresorufin dealkylase activity (an
indicator of ?..„ activity) was not elevated in maternal livers with PCE
exposure and was not detectable in fetal livers.  No other effects were observed
in pregnant  rats exposed for 6 h per day on days 0-20 of gestation.
     Hanson  et al.  (unpublished, 1982) conducted a  study on postnatal evaluation
                                                                          3
of offspring of the female  rats exposed to 1000 ± 125 ppm (6780 ±  847 mg/m  )  PCE
before and/or during pregnancy.  The four treatment groups are those described
003PE2/E                             7-5                                11/22/83

-------
above (Tepe et al., 1982).   The purpose of the postnatal  evaluation was to deter-
mine if the reduction in body weight or the excess skeletal and soft tissue
variants observed in term fetuses from the Tepe et al.  (1982) study persisted,
and if PCE possessed transplacental carcinogenic activity or neurobehavioral
toxicity.  Weight gain and survival of offspring up to 18 months of age, and
frequency of any gross lesions observed at 6- and 18-month autopsies, were not
influenced by prenatal exposure to PCE.  Neurobehavioral  tests of general acti-
vity in open field tests at 10 and 20 days of age, and in running wheels from 40
to 100 days of age, did not indicate treatment-related effects.   Likewise, results
from the visual discrimination test on offspring from 130 to 170 days of age
were negative.   Prenatal exposure to PCE did not exert a detrimental effect on
any of the parameters of postnatal maturation examined.
7.1.3  Rabbits
     Beliles et al. (1980) also investigated the teratogenic potential of PCE
in rabbits.  The rabbits were divided into six groups as follows:

                                                 Days of Exposure
Group          Cone (ppm)             Pregestational             Gestational
1
2
3
4
5
6
0 (control)
0 (control)
500
500
500
500
none
5 days/week;
none
5 days/week;
none
5 days/week;

3 weeks

3 weeks

3 weeks
0-21
0-21
0-21
0-21
0-21
0-21
After the pregestational exposure period was complete, rabbits from each group
were mated.   A positive identification of spermatozoa in the vaginal canal was
taken as evidence of mating and designated as day 0 of gestation.   The two
control groups (1 and 2) were exposed to filtered air and the exposure groups
                                      3
(3, 4, 5, and 6) to 500 ppm (3390 mg/m ) PCE, 7 h per day.   It is unclear if
the animals were exposed on each day, i.e., on days 0 through 21 of gestation
or only on a Monday through Friday basis.  All rabbits could not have been mated
on the same calendar day; thus, all rabbits would not have been at the same stage
of gestation during the exposure period.
     Mean body weights of rabbits during pregestational and gestational exposure
indicated no significant difference between controls and treated groups.
Reduced food consumption during the approximate period of days 10 through 22
of gestation was observed in groups 3 and 5 and may have been related to PCE

003PE2/E                             7-6                               11/22/83

-------
exposure.   During the investigation, 21 female rabbits died (9 in the control
groups and 12 in the treatment groups).   Placental  abnormalities were found at
all exposure levels.  In four litters examined in group 5, 19 anomalies were
found (p < 0.05 compared to group 1 and 2 controls.)  Rank Sum analysis did
not confirm this significance and the authors judged it to reflect a change
confined to a few litters.   Fifteen of the 19 abnormal placentas in group 5
occurred in two litters.  Histopathologic evaluation failed to reveal any
significant change in the placentas.  Of the skeletal changes noted in the
fetuses examined in each group, neither the frequency nor the character of
changes in the treated groups indicated an adverse effect on fetal growth and
development, nor any teratogenic effects.

7.2  SUMMARY
     The mammalian animal tests performed to date do not indicate any signifi-
cant teratogenic potential  of PCE.  On this basis, there is no evidence that
suggest that the conceptus is uniquely susceptible to the effects of PCE.  The
anatomical effects observed reflect delayed development and can be considered
reversible.  The minor behavioral changes observed probably reflect maternal
nutritional deprivation rather than a direct effect of PCE.  It is important
to note, however, that the reversible nature of an embryonic/fetal effect in
one species might,  in another species, be manifested in a more serious and
irreversible manner.  Thus, the teratogenic potential of PCE for humans must
be considered unknown.
 003PE2/E                             7-7                               11/22/83

-------
7.2  REFERENCES
Beliles, R. P.; Brusick, D. J.; Mecler, F. J. (1980).  Teratogenic-mutagenic
     risk of workplace contaminants:  trichloroethylene, perch!oroethylene, and
     carbon disulfide.  U.S. Department of Health Education and Welfare, Contract
     No. 210-77-0047.

Buelke-Sam, J.; Kimmel, C.  A. (1979).  Development and standardization of
     screening methods for behavioral teratology.  Teratology 20:17-30.

Elovaara, E. ;  Hemminki, K.; Vainio, E. (1979).  Effects of methylene chloride,
     trichloroethane,  trichloroethylene, tetrachloroethylene and toluene on
     the development of chick embryos.  Toxicol. 12:111-119.

Lanham, S.   Studies on placental transfer:  trichloroethylene.   Ind. Med.
     39:46-49, 1970.

Manson, J.  M.; Tepe, S. J.; Lowrey, B.; L. Hastings  (1982).  Postnatal evaluation
     of offspring exposed prenatally to perchloroethylene.  Unpublished.


Nelson, B.  K.; Taylor, B.  J.; Setzer, J. V.; Hornung, R. W. (1980).  Behavioral
     teratology of perchloroethylene in rats.  J. Environ. Pathol.  Toxicol.
     3:233-250.

Schwetz, B. A.; Leong, B.  K.; Gehring, P.  J. (1975).  The effect of maternally
     inhaled trichloroethylene, perchloroethylene, methyl chloroform, and
     methylene chloride on embryonal and fetal development in mice and rats.
     Toxicol.  Appl. Pharmacol. 32:84-96.

Tepe, S. J.; Dorfmveller,  M. K.; York, R. G.; J. M. Manson (1982).  Teratogenic
     evaluation of perchloroethylene in rats.  Unpublished.

U.S. Environmental Protection Agency.  Proposed guidelines for registering
     pesticides in the United States.  43 FR, #163,  August 22, 1978.  pp.
     37382-37388.

U. S. Environmental Protection Agency.  Proposed health effects test standards
     for Toxic Substances Control Act test rules and proposed good laboratory
     practice standards for health effects.  44 FR,  #145, July 26, 1979,  pp.
     44089-44092.

U.S. Environmental Protection Agency.  Determination not to initiate a rebut-
     table presumption against registration (RPAR) of pesticide products con-
     taining carbaryl  availability of decision document.  Fed.  Regist. 45:
     81,869-81,876, December  12, 1980.
003PE2/E                             7-8                               11/22/83

-------
                               8.1   MUTAGENICITY

     The objective of this mutagenicity evaluation is to determine whether
tetrachloroethylene has the potential  to cause mutations in germ and somatic
cells of humans.  This qualitative  assessment is  based on data derived from
several  short-term tests that measure  different types of genetic alterations:
gene mutation, chromosomal aberrations, unscheduled DMA synthesis, and mitotic
recombination (Table 8-1).  These tests were conducted using bacteria, Drosophila,
yeast, cultured mammalian cells, whole mammal systems, and cytogenetic analyses
of exposed humans.  Consideration will also be given to studies concerning the
mutagenicity of known and expected  metabolites.

8.1  GENE MUTATION TESTS
8.1.1  Bacteria
     The ability of tetrachloroethylene to cause  gene mutations in bacteria has
been studied by several investigators.  Many of these investigators used the
Ames Salmonel1 a/microsome test or modifications of that test.  Different purities
of tetrachloroethylene (stabilized* and low-stabilized materials) have been
evaluated.   (Bacterial studies are summarized in  Table 8-2.)
     Tetrachloroethylene is a volatile chemical,  and thus, the standard Ames/
Salmonella plate test, in which precautions are not taken to prevent escape of
evaporated material, is not entirely suitable for its testing.  Williams and
Shimada (1983,  sponsored by PPG Industries, Inc.), however, modified the standard
plate procedure by exposing the bacteria to the test agent in a sealed chamber.
     *Stabilization is the intentional addition of material to increase the
stability of tetrachloroethylene.  Typically, the added stabilizers are acid
and free radical scavengers (Dr. A. Philip Leber of PPG Industries, Inc.,
personal communication, September 1983).
                                    8-1

-------
       TABLE 8-1.   SUMMARY  OF MUTAGENICITY  TESTING  OF  TETRACHLOROETHYLENE
A.  Gene Mutation Tests                 Results*
    Ames/Salmonella assay                 +**,-
    Escherlchla coll K12/343/113 (  )
    Multi-purpose test
    Saccharomyces cerevlslae D7 reverse
    mutation test (lvl-1 locus)
    Drosophlla sex-linked recessive
    lethal test
    Host-mediated tests In mice: bacteria  wk
                                 yeast
B.  Chromosomal Aberration Tests
    Rat bone marrow assay
    Mouse bone marrow assay
    Peripheral lymphocytes from
    exposed humans
    Drosophlla sex chromosome loss
    assay
    Rat dominant lethal assay
                                           _t
                                                            References
                                                      Williams and Shlmada 1983,
                                                      Margard 1978, SRI  Inter-
                                                      national 1983, NTP 1983,
                                                      Bartsch et  al. 1979, Cerna
                                                      and Kypenova 1977  (abstract)
                                                      Henschler 1977, Grelm et al.
                                                      1975
                                                      Call en et al. 1980,
                                                      Bronzettl et al. 1983
                                                      Bellies et  al. 1980
                                                       Bellies  et  al.  1980,
                                                       Cerna  and Kypenova  1977
                                                       (abstract)
                                                       Bronzettl et al.  1983
                                                       Rampy  et al.  1978,
                                                       Bellies  et al.  1980
                                                       Cerna  and Kypenova
                                                       1977  (abstract)
                                                       Ikeda  et al.  1980
                                                       Bellies et al.  1980
                                                       Bellies et al.  1980
C.  Other Tests Indicative of DNA Damaging Activity
    Unscheduled DNA synthesis In WI-38     -f
    Hepatocyte primary culture/DMA repair  *,-tt
    test
    Mltotlc recombination tests In         +,-
    Saccharotnyces cerevlslae D7
                                                       Bellies et al.  1980
                                                       Williams and Shlmada
                                                       1983, Williams 1983
                                                       Callen et al.  1980,
                                                       Bronzettl et al. 1983
D.  DNA Binding Studies
    Whole mice
E.  Germ Cell Tests
    Altered sperm morphology
                                     mouse   rat
                                                       Schumann et al.  1980
                                                       Bellies et al.  1980
      * + designates positive; - negative; wk weak response.  Dose-response
 relationships were not established for the reported * results or wk results.
     **AHhough Increases several fold over background were observed, the positive
 results are considered weak because large amounts of material were needed to
 elicit the responses.  Positive results were only obtained using airtight
 chambers (except  for the study by Cerna and Kypenova 1977).
      Questionable evidence for weak or borderline activity In specific data sets.
     ftPos1t1ve results were found with vapor phase exposure and negative results
 were obtained using conventional phase exposure.
                                         8-2

-------
                      TABLE 8-2.   RESULTS  OF  BACTERIAL  TESTS OF DIFFERENT PURITIES AND SOURCES OF TETRACHLOROETHYLENE
co
Test system/strain
Ames/Salmonella
TA98, TA100, TA1535
Ames/Salmonella
TA98, TA100, TA1535
TA1537, TA1538
Purity/source
Perch! or- 200-
low-stabil ized
99.93% purity
PPG Industries,
Inc.
Perchlor 230-
stabil ized
99.80% purity
PPG Industries,
Inc.
Concentrations Metabolic
tested activation
1% v/v for TA98, Aroclor-
TA1538, and induced rat
TA1537; 0.1 S9 mix
1.0, 2.5, 5.0,
7.5, and 10%
for TA100 and
TA1535
1% v/v for TA98, Aroclor-
TA1538, and induced rat
TA1537; 0.1 S9 mix
1.0, 2.5, 5.0,
7.5, and 10%
Protocol
Gas-phase
exposure in
airtight
chamber's
Gas-phase
exposure in
airtight
chambers
Reported
result Reference
Positive
at 2.5% (>97%
toxicity) in
base-pair
substitution-
sensitive strain
(two to tenfold
increases
+/- M.A.*)
dose-response
not established
Positive
at 2.5% (>97%
toxicity)
in base pair-
substitution
Williams
and
Shimada
1983
Wi 1 1 i ams
and
Shimada
1983
                                                      for TA100 and
                                                      TA1535
                                                                    sensitive
                                                                    strains (three to
                                                                    ten-fold increase
                                                                    +/- M.A.)
                                                                    dose-response
                                                                    not established
          Ames/Salmonella
          TA100, TA1535
High purity
Perchlor
low-stabil ized,
99.98+% purity,
PPG Industries,
Inc.
0.1, 1.0, and
2.5% v/v
Aroclor-
induced rat
S9 mix
Gas-phase
exposure
in airtight
chambers
Negative
Wi11i ams
  and
Shimada
1983
              *+/- M.A. designates response similar in the presence and absence of
         metabolic activation
                                                         (continued on the following page)

-------
                                                          TABLE  8-2.   (continued)
Test system/strain
Ames/Salmonella
TA98, TA100, TA1535
TA1537, TA1538
Ames/Salmonella
TA98, TA100, TA1535
TA1537, TA1538
Purity/source
Nonstabilized
high purity
Detrex
Industries, Inc.
Stabilized
99.84% purity
Detrex
Industries, Inc.
Concentrations
tested
0.01, 0.05,
and 0.1 ml/
plate
0.01, 0.05,
and 0.1 ml/
plate
Metabolic
activation
Aroclor -
induced r-at
S9 mix
Aroclor -
induced rat
S9 mix
Protocol
Standar-d
plate test
in airtight
chambers
Standar d
plate test
in airtight
chambers
Reported
result
Negative
Refer ence
Margard
1978
Positive Margard
(twofold 1978
increases
in frameshift-
CO
sensitive
strains and
TA100) at 0.1
ml/pi ate (160
mg/plate). >90%
toxicity.  S9 mix
increased r-esponse
(10-to 17-fold
increases)
Ames/Salmonella
TA98, TA100, TA1535
TA1537
Ames/Salmonella
TA98, TA100, TA1535
TA1537
99+% purity 0.025, 0.05,
Aldrich 0.1, 0.5, 1.0
and 1.5 added
to petri plate
at bottom of
desiccator-
Technical grade 3, 10, 33, 100,
Fisher 333 ug/plate
Ar odor-induced
female and male
rat liver S9 mix
and Aroclor-- in-
duced female and
male mouse liver
S9 mix
Aroclor- induced
rat liver S9 mix
Aroclor-induced
hamster- liver
S9 mix
Gas-phase Negative
exposure in
airtight
chambers
Preincu- Negative
bation assay
10 min. 37°C
SRI
Inter-
national
1983
NTP 1983
                                                                                          (continued on the following page)

-------
                                                           TABLE 8-2.   (continued)
00
Test system/strain
Ames/Salmonella
TA100
Ames/Salmonella
tester- strains
not reported
E. coli K12/343/113 (>)
Host-mediated assay
ICR mice/Salmonella
TA1950, TA1951,
TA1952
Host-mediated assay
male and female CD
mice/Salmonella
TA98
Purity/source
99. 7% purity
Merck-Darmstadt
Not
reported
analytical grade
Merck-Darmstadt
Not
reported
91.43% purity
North Strong
Division
Chemicals
Concentrations
tested
0 to 663
mg/plate
(4xlO-3n)
Not
reported
0.9 mM
Not
reported
Inhalation at
100 ppm and
500 ppm for
5 consecutive
days
Metabolic
activation
Phenobar bital-
induced mouse
1 iver microsomes
with and without
cofactors

Phenobar bital-
induced mouse
1 iver- micro-
somes
Female mice
Male and
female mice
Protocol
Standard
plate test
Spot test
Liquid
suspension
2 hours at
37°C
Not
reported
Bacteria in-
jected intra-
per itoneally
after last
exposure and
removed 3
hours
Reported
result
Negative
Positive
Negative
Positive
(mortality
not
reported)
Positive
(twofold
increases
in revertants
from 100 ppm
in males;
and fourfold
increases
in revertants
from 500 ppm
females).
Reference
Bartsch
et al.
1979
Cerna and
Kypenova
(1977,
abstract)
Greim et
al. 1975
Cerna and
Kypenova
(1977,
abstract)
Beliles
et al.
1980

-------
In their procedure, a known volume of test chemical  was added to a glass petri
plate containing a magnetic bar to ensure continuous stirring for even dissipation
of vapors.  The chamber was initially incubated at room temperature for 20
minutes, and then exposure was continued at 37°C for 18 hours,  after which  the
bacterial test plates were removed from the chamber, covered with lids, and
incubated another 30-54 hours at 37°C.
     In the Williams and Shimada (1983) study,  three types of material  (provided
by PPG Industries, Inc.) were tested in the presence and absence of S9 mix
(derived from livers of Aroclor-induced rats):   Perchlor 200 (low-stabilized,
99.93% purity), high purity Perchlor (low-stabilized, 99.98+% purity), and
Perchlor 230 (stabilized, 99.80% purity).*  Perchlor 200 and Perchlor 230 were
evaluated in tester strains TA98, TA1537, and TA1538 at 1% (v/v) and in tester-
strains TA100 and TA1535 at 0.1, 1.0, 2.5, 5.0, 7.5, and 10% (v/v).  High-
purity Perchlor was evaluated in TA100 and TA1535 at 0.1, 1.0,  and 2.5% (v/v).
These concentrations represent the predicted concentration of the compound  in
the gas phase based upon calculations that consider the chamber volume and  absolute
temperature of the chamber, atmospheric pressure, and density of the test compound.
(For example, 0.06, 0.62, 1.55, 3.1, 4.65, and 6.2 ml of tetrachloroethylene
was added to the chamber- for a 0.1, 1.0, 2.5, 5.0, 7.5, and 5% v/v vapor- tar-get
concentration, respectively.)  Positive responses were obtained for Perchlor
200 and 230 at 2.5% v/v (or 1.55 ml per desiccator)  but not for high-purity
Perchlor at the concentrations tested.   A slightly higher response was observed
with stabilized material.  Positive results were repeated in a second experiment
and were similar in the presence or absence of S9 mix.  For example, in the
absence of S9 mix Perchlor 200 (at 2.5%) increased the number of revertant
colonies in the base-pair substitution-sensitive strains TA1535 (six to tenfold
increases) and TA100 (two to threefold increases); Perchlor 230 (at 2.5% v/v) also
caused increases in revertant numbers in TA1535 (approximately tenfold increases)
     *Perchlor 200 contained 0.012 (% by weight) of hydroquinone monomethyl
ether (HQMME) which provides minimal  stabilization;  Perchlor 230 contained  0.011%
HQMME, 0.07% cyclohexene oxide, and 0.05% B-ethoxy propionitrile; high-purity
Perchlor contained 0.01% HQMME (written communication from Dr. A. Philip Leber
of PPG Industries, Inc., 1983).

                                    8-6

-------
and TA100 (approximately threefold increases).   (See Figure 8-1  for an  illustra-
tion of these responses).   Negative results were found in frameshlft-sensitive
strains (TA98 and TA1538)  for both Perch!or 200 and 230 in the presence or
absence of S9 mix.  At the next highest concentration (5% v/v) the revertant
counts decreased to zero as toxicity became an  important factor in the  tests.
The authors also indicated the test materials were toxic (> 97% killing) at
the concentration (i.e., 2.5% v/v) that induced the mutagenic responses as
determined by a simultaneous cytotoxicity test.  Although the test materials
may be quite toxic at 2.5% v/v, the investigator's method of quantifying
cytotoxicity may not be accurate because the cell  density that was used on
each plate for the determination of toxicity is several orders of magnitude
lower than that used for determination of mutagenicity (i.e., 107-109 cells).
Nevertheless, it should be noted that because of toxicity, the mutagenic
responses observed for Perchlor 200 and 230 are within a narrow range of
concentrations.  Because of this narrow range of effective concentrations and
the concentration increments tested, a clear dose-response was not demonstrated.
     In the Williams and Shimada (1983) study,  high-purity material did not
produce a detectable response under- experimental conditions, as did the lower-
purity materials  (i.e., Perchlor 200 and 230).   Therefore, the increases in
revertants observed may be due to a contaminant(s).  However, the high-purity
Perchlor appeared to be more toxic (zero revertants observed at 2.5%) than the
other test materials, and it is possible that a weak mutagenic response may
have been masked by its toxicity.  A weak response was observed for high-purity
material at 1% in TA1535 (with S9 mix), but was not repeatable.  Also,  it
should be pointed out that concurrent negative and positive controls
were not used in  this study.  This weakens the negative conclusions for the
high-purity material and makes interpretation of the magnitude of the responses
in  the presence of S9 mix for the lower-purity materials difficult.
     Margard  (1978) examined tetrachloroethylene (provided by Detrex Chemical
Industries, Inc.) in the Ames/Salmonella test to determine whether- mutagenic
activity of technical grade samples is the result of added stabilizers.
Precautions to prevent  escape of material were taken (Margard, personal
communication 1981), but they were not specified in writing.  Tester strains
TA1535, TA1537, TA1538, TA98, and TA100 were used.  Tests were conducted in

                                    8-7

-------
                                                   TA153S
                                 300


                                 mo


                                 100

                                  0
                           ut
                           a.

                           i
                           4
                           ec
                           UJ
                           >
                           UJ
                           CC
                           C
                           uj
                                                                       TA1535
                                                     300


                                                     no

                                                     100
               2.6
                                               1041
CD
 I
CXI
GOO


500


400


30O


200


100
                  TA 100
300


2OO


100
                                    0.1           2.8         5.0        7.6         10.0

                                      TETRACHLOROETHYLENE. Stabilized (%V/V)
                  TA100
                                                         0.1  1.0    2.6         5.0         7.5         10.0

                                                          TETRACHLOROETHYLENE. Low-Stabilized (%V/V)
                                Figure 8-1.  Oose-response curves for Perchlor 200 (low-stabilized tetrachloroethylene)  and  Perchlor 230
                                            (stablllz tetrachloroethylene) using Salmonella typhlmurlum tester strains  TA100  and TA1535 in the
                                            presence of (O--O) and 1n the absence of  (•—•) 59 mix (150 ul of Aroclor-lnduced rat liver).
                                            Each data point  represents the geometric mean of triplicate plates from one experiment.  (Williams
                                            and Shimada 1983)

-------
the presence and absence of S9 mix (prepared from livers of Aroclor-1254 induced
rats).  Nonstabilized and stabilized materials were added directly to the
petri plates at 0.01, 0.05, and 0.1 ml  per plate rather than vapor exposure.
The nonstabilized test material was described as purified tetrachloroethylene
that contained no detectable epoxides or other stabilizing components, and the
stabilized material was identified as an industrial degreasing grade of
tetrachloroethylene that contained 0.07% (by weight) epichlorohydrin, 0.007%
N-methylmorpholine, 0.07% beta-hydroxypronitrile, and 0.01% hydroquinone monoethyl
ether (written communication from L. Schlossberg, Detrex Chemical Industries
Inc., 1981).  Nonstabilized test material was not detected as positive in the
presence or absence of S9 mix.  But stabilized material at 0.1 ml per plate
(equivalent to 160 mg*) caused a weak response in the absence of S9 mix;
twofold increases were observed for TA1538, TA98, and TA100.  In the presence
of S9 mix, greater increases in the number- of revertants for- tester- strains
TA1538 (17-fold increase), TA98 (10-fold increase), and TA100 (1.7-fold increase)
at 0.1 ml per plate, were found.  A clear dose-response was not observed for
any of the tester strains.  The toxicity of the test material was reported as
greater than 90% killing at concentrations which caused increases in the number
of revertants.  The method used to determine toxicity may be inaccurate, as
discussed previously for the Shimada and Williams  study (1983).  Negative
results were reported for stabilized material in TA1535 and TA1537.  Although
the stabilized test material contained epichlorohydrin, which has been shown
to be strongly mutagenic in Salmonella,  it is primarily active in base-pair
substitution-sensitive  strains  (i.e., TA1535 and TA100; McCann et al. 1975,
Anderson et al. 1978),  and thus it does  not seem likely that this agent can
solely account for the  activity observed for stabilized material in  the
frameshift-sensitive strains TA1538 and  TA98.  Nevertheless, it appears
that  the mutagenic activity is  due to the presence  of  a contaminant(s).
      Other-  investigators have  conducted  studies  in  which precautions were
taken to prevent escape of test material during  testing.  SRI Inter-national
(1983 EPA-sponsored  test)  reported tetrachloroethylene  (99+% purity,
      Calculation  based on  the density of tetrachloroethylene as 1.586 g/ml.
                                    8-9

-------
Aldrlch) to be negative when tested as a vapor In sealed desiccators using
TA1535, TA1537, TA98, and TA100 with or without S9 mix prepared from livers of
female and male Aroclor-1254 induced rats and mice.  Cells were exposed at
37°C for 8 hours in desiccators, and then incubated at 37°C for an additional
42 hours.  The concentrations tested were 0.05, 0.1, 0.5, 1.0, 1.5 ml  added to
a petri plate at the bottom of the desiccator chamber in one experiment and
0.025, 0.05, 0.1, 0.5, and 1.0 ml added to a plate at the bottom of the desiccator
in another experiment.  A toxic response (reduction or absence of bacterial
lawn) was found at 1.5 ml/per desiccator.
     The National Toxicology Program (NTP 1983) sponsored Salmonella testing
on tetrachloroethylene (technical grade, source Fischer) and obtained  negative
results.  Four standard tester strains were used:  TA98, TA100, TA1535, and TA1537.
A preincubation protocol  was followed in which the cells, S9 activation system,
and chemical are preincubated in test tubes for 20 minutes at 37°C before
addition of the top agar and plating in petri plates.  Two different S9 systems
were used;  S9 mix derived from livers of Aroclor-1254 induced rat liver and
Aroclor-1254 induced hamster liver.  Tests were also conducted in the  absence
of S9 mix.  Six concentrations (0, 3, 10, 33, 100, 333 ug/plate) were  evaluated.
(In TA100, up to 10,000 ug/plate was evaluated.)  Although incubation  was
carried out in capped tubes, it is possible that evaporation and some  escape
may have occurred.  However, it should be noted that toxic levels were tested
as indicated by absence or reduction of bacterial lawn.
     Bartsch et al. (1979) investigated the mutagenicity of tetrachloroethylene
(99.7% purity, Merck-Darmstadt) using the standard Ames/Salmonella plate
test in which precautions to prevent escape of test material are not taken.
Negative results were obtained using tester strain TA100 in the presence of
mouse liver  microsomes with and without cofactors.  The authors indicated that
toxic concentrations (above 82.9 mg/plate) were tested, but did not indicate
their- criteria for determining toxicity.  Interpretation of these negative
conclusions is limited by the amount of data presented and because only one
tester  strain was evaluated.
     In an abstract, Cerna and Kypenova (1977) reported that in a spot test
protocol of the Ames/Salmonella assay, tetrachloroethylene (purity and source
                                    8-10

-------
not reported)  induced both base-pair substitution  and frameshlft mutations in
Salmonella typhimurium.   The results were obtained in the absence of exogenous
activation.  These authors also reported that in a host-mediated assay using
female ICR mice,  tetrachloroethylene induced significant increases in the
number of revertants in  tester strains TA1950,  TA1951,  and TA1952 at dosage
levels reported as representing the 1059 and one-half of the 1059.  These
results were reported as not being dose-dependent.  Because this report was in
abstract form and did not provide details of the protocol nor present the
data, the acceptability  of the test results is  indeterminate.  Also, the
possibility of mutagenic contaminants must be considered.
     Beliles et al. (1980) used a host-mediated assay to evaluate the effects
of tetrachloroethylene (91.43% pure from North  Strong Division Chemicals) in
the presence of whole mammal metabolism.  In this  study, Salmonella tester
strain TA98 was used as  the indicator organism, and male and female mice (strain
CD-I) were the hosts.  The animals were exposed by inhalation 7 hours per day
for 5 days to either 100 ppm or 500 ppm.  Bacteria were injected intraperitoneally
into the mice after the  last exposure.  The bacteria were removed from the mice
3 hours later.  At the 100 ppm dose level, increases in the number of revertants
were observed for males  (approximately twofold increase) but not for females.
At 500 ppm, a positive response was reported for females (approximately fourfold
increase) but not for- males.  Because of the lack  of a dose-response and the
weak responses, these findings should be viewed with caution.  Also, the material
used was of low-purity,  and the responses may have been due to contaminants
and/or added stabilizers.  In addition, parallel in vitro plate tests using
Salmonella were not conducted.  The parallel plate tests are important the
determination of the requirement of whole-mammal activation.
     Studies on tetrachloroethylene have also been conducted in Escherichia coli.
Henschler (1977)  reported that tetrachloroethylene was not mutagenic when tested
in E. coli K12 with metabolic activation (liver microsomal fraction prepared
from phenobarbital-induced mice).  This report, however, is difficult to evaluate
because actual revertant count data (experimental  and control number of revertants)
and the details of the protocol are not provided.   It appears that the conclusions
presented in this report are actually based on data derived from the study by
Grelm et al. (1975).

                                    8-11

-------
     Greim et al. (1975) reported that negative results were obtained when
tetrachloroethylene (purity reported as analytical grade; Merck-Darmstadt)
was assayed in the multi-purpose test system of Escherichia coli  K12/343/113 (>).
Tests were conducteo in the absence and presence of metabolic activation
(phenobarbital-induced mouse liver microsomal fraction plus TIADPH cofactors)
at a concentration of 0.9 mM and a treatment time of 2 hours in  liquid suspension
at 37°C.  These treatment conditions resulted in 99 + 1% survival.   The genetic
markers evaluated for mutation induction were the missense marker arg+ and
the frameshift marker nad+ for reverse mutation, and gal+ and MTR for forward
mutation.  Deficiencies in the study design and reporting of the  results
reduce the weight of the negative conclusion.  These deficiencies are as
follows:  1) only one concentration was evaluated, 2) adequate exposure may
not have been achieved, as indicated by the high survival, 3) there was no
reporting of revertant count data (experimental and control), and 4) there
was no reporting of the number of replicate plates used or the number of the
tests conducted.
     The bacterial tests discussed above do not clearly demonstrate that
tetrachloroethylene Itself is mutagenic.  The positive responses  found may
be due to contaminants and/or added stabilizers.  The induced increases in
revertants did not require exogenous metabolic activation and were observed in
both frameshift and base-pair substitution-sensitive tester strains.  The
positive findings were not considered strong in that large amounts of material
(estimated vapors at 2.5% v/v and at 160 ing/plate) were needed for  the detection
of mutagenicity.  Also, there was a very narrow range of effective concentrations
because of toxidty; thus dose-response relationships were not established.
When tested, highly purlfed samples were not detected as mutagenic under the
conditions 1n which technical samples caused increases 1n the number of revertants.
Although some technical tetrachloroethylene samples were positive, there were
other samples that were not detected as positive.  The available  results provide
suggestive evidence that certain technical samples of tetrachloroethylene are
weakly mutagenic 1n Salmonella and that the positive responses may be due to
impurites and/or added stabilizer's.
8.1.2  Drosophila
     Bellies et al. (1980) used the sex-linked recessive lethal  assay in
DrosophUa melanogaster to test tetrachloroethylene (91.43% purity, North Strong
Division Chemicals) and reported negative results.  Adult male flies were
exposed for 7 hours by Inhalation at 100 ppm and 500 ppm.  Treated males were

                                     8-12

-------
mated to nontreated females at various times (2-3-3-2 day mating scheme)  to
test specific germ cell stages.  No significant increases (P < 0.05) over the
background values were observed.  However,  only a small  sample size was examined.
A total of 3804 chromosomes for the 100 ppm dose and 3956 chromosomes for the
500 ppm dose were evaluated.  This sample size was only  large enough to
exclude the induction of an approximately fourfold increase in mutation frequency
(Kastenbaum and Bowman 1970).  Ideally, at least 7000 chromosomes at each dose
level should be screened to preclude a doubling in mutation frequency, which is
generally considered to be the increase of biological significance.  Survival
was not reported in this study, and thus it is uncertain whether a sufficient dose
was given.  These deficiencies prevent a judgment regarding the mutagenic
activity of tetrachloroethylene in Drosophila.
     The ability of tetrachloroethylene to cause gene mutations in an eucaryotic
organism has not been adequately examined.   The only available study was a sex-
linked recessive lethal test in Drosophila in which tetrachloroethylene was not
properly evaluated.

8.2  CHROMOSOMAL ABERRATION TESTS
8.2.1   Whole-Mammal Bone Marrow Cells
     Rampy et al. (1978) examined bone marrow cells for  chromosome aberrations
from male and female Sprague-Dawley rats after tetrachloroethylene exposure.
Animals were exposed to 300 ppm (2.03 mg/1) or 600 ppm (4.07 mg/1)
tetrachloroethylene* by inhalation 6 hours/day, 5 days/week, for one year.
Three animals per dose were examined.  The authors reported "zero" chromosomal
aberrations pet cell for both females and males.  The data for females, however,
are inadequate  for a clear interpretation because of the very low number of
metaphases scored (less than 25 cells per animal).  In male rats, 150 cells
were scored (50 cells per animal).  The negative controls were also reported
as "zero" aberrations.  This observation is very unusual because, in general,
most laboratories have reported about 1 to 2% total aberrations for background
values.   In this study, it is not known whether the highest exposure level was
near the maximum tolerated dose (MTD) for females because no weight loss and
no mortality was observed.  In males there was no weight loss, but significant
increases in mortality above control values were observed at the highest dose
      *Formulation  (liquid  volume percent):   trichloroethylene, 3 ppm; hexachloro-
 ethane,  <  12  ppm;  carbon tetrachloride, 2 ppm; 4-methyl morpholine, 44 ppm; non-
 volatile residue,  2  ppm; and  tetrachloroethylene balance.

                                     8-13

-------
tested, and therefore an MTD may have been approached.   It is not apparent that
the Investigators determined the toxicity of the test material  to arrive at an
MTD for this study, because the dosage levels used were based on the threshold
limit value of 100 ppm for tetrachloroethylene.
     A rat bone marrow assay was also performed  by Bellies et al. (1980) and
reported as negative.  Ten males and ten females [CRL:COBS CD(SD)BR] were
exposed to an acute dose of 100 ppm and 500 ppm  tetrachloroethylene (91.43%
purity, North Strong Division Chemicals) by Inhalation for 7 hours.  Bone
marrow cells were harvested 6, 24, and 48 hours  later.  No increase in aberrations
was found for females, but for males, weak clastogenic effects  (breaks, fragments,
deletions, and aneuploid cells) were observed.  At 500 ppm, 3.3% cells with
aberrations versus 0.7% in the control were found at the 24-hour kill.  A
subchronic study (five exposures, 7 hours per day) at 100 ppm and 500 ppm was
also conducted.  Animals were killed 6 hours after the last exposure.  No
increase in abnormalities was found in males, and only a slight increase at
100 ppm (1% cells with aberrations) was found in females.  This response was
not dose-related.  The female subchronic control group had a very low background
(0.3% cells with aberrations).  Although isolated incidences of increases in
chromosomal aberrations were observed, the lack  of dose-responses precludes an
unequivocal positive conclusion.  On the other hand, a negative conclusion
cannot be drawn because the authors did not discuss the criteria used to select
the dosage levels, and thus, there is the possibility that a toxic dose may
not have been evaluated.
     Cerna and Kypenova (1977) reported in an abstract that mice (ICR)
given an acute intraperitoneal dose one half of the 1050 of tetrachloroethylene
or dosed intraperitoneally for five applications in 24 hour intervals (dose of
one injection equalled 1/6 LDso) did not show cytogenetic effects in the bone
marrow cells.  Details of the protocol and the cytogenetic data were not available
for an evaluation.  Hence, the negative conclusion of the authors cannot be
considered definitive.
8.2.2  Human Peripheral Lymphocytes
     Ikeda et al.  (1980) studied chromosomal aberrations, sister chromatid exchanges
(SCEs), and variation 1n the mitotic index of peripheral lymphocytes cultured from
ten workers (seven males and three females) occupationally exposed to technical grade
tetrachloroethylene  (impurities not reported).  The workers were divided into
high (Group 1) and low (Group 2) exposure groups.  Group 1 consisted of six
workers (five males and one female aged 20 to 66 years) from a  degreasing workshop.

                                     8-14

-------
These workers had a geometric mean exposure of 92 ppm (range 30 to 220 ppm).  The
five males of Group 1 had work histories of 10 to 18 years, whereas the one female
had worked in the degreasing shop for only 1 year.  Group 2 included four
workers (two males and two females aged 17 to 31 years) from a support department
with a shorter work history (3 months to 3 years) and with an exposure range
of 10 to 40 ppm.  The control group consisted of six males and five females.
The authors did not indicate if this was a matched control, and did not indicate
the medical histories of the subjects (e.g., recent illnesses, radiation
exposures).  There were no statistically significant differences (P > 0.05)
in the incidences of chromosomal aberrations (structural and numerical) and
SCEs between the exposed workers and control group.  The mitotic index was
similar for both exposed and control groups.
8.2.3  Drosophila
     In addition to performing the sex-linked recessive lethal assay discussed
earlier, Bellies et al. (1980) also conducted a sex chromosome loss assay on
low-purity (91.4%, North Strong Division Chemicals) tetrachloroethylene for
nondisjunction  in Drosophila melanogaster.  Males were exposed to 100 and 500 ppm
of tetrachloroethylene for- 7 hours.  The phenotypic classes used allow for
detection of losses of the entire X or- Y chromosome and the short or- long arm of
the Y chromosome.  Although marginal increases, which were not dose-related,
were observed after tetrachloroethylene treatment, they are not considered
sufficient to judge the data as positive or- negative.
     The cytogenetic tests discussed above using mice, rats, Drosophila, and
exposed humans  have been reported as negative.  Although  these studies are
not considered  to be a thorough evaluation of the ability of tetrachloroethylene
to cause chromosomal aberrations, the data collectively indicates that
tetrachloroethylene is not strongly clastogenic.  However, there have been  no
adequate studies on the ability of tetrachloroethylene to cause chromosome
nondisjunction  (aneuploidy).

8.3  OTHER TESTS INDICATIVE OF DNA DAMAGE
8.3.1  DNA Repair
     Unscheduled DNA synthesis  (UDS) is measured by repair of DNA lesions,
which  is indicative of DNA.damage.  Bellies et  al. (1980) assessed  the ability
of tetrachloroethylene (91.43%  purity, North Strong Division Chemicals) to
cause  UDS  in human fibroblast (WI-38) cells.  Because WI-38 cells have little
if any enzyme activation capability, the tests  were conducted with  an exogenous

                                     8-15

-------
source of metabolic activation (i.e., S9 mix).   The test material  was examined
at 0.01, 0.05, 0.1, and 0.5% (v/v) using conventional  liquid phase exposure.
Scheduled DNA synthesis was blocked by treatment with  hydroxyurea, and DOS  was
measured by liquid scintillation counting of incorporated tritiated thymidine
(C3H]-TdR) into DNA.  A very slight increase was seen  at 0.01% (v/v)
tetrachloroethylene both in the presence (1.5-fold of  control) and absence
(1.35-fold of control) of Aroclor rat liver S9  mix. No increases  occurred  at
0.1 and 0.5%.  A toxic response was reported at 0.5% (i.e.,  a decrease in
total amount of DNA as well as in the Incorporation of [3H]-TdR).   An increase
in total amount of DNA at 0.01% was found, suggesting  that more cells are entering
the S phase.  Cells accelerated into S phase would account for the increases
seen in [3H]-TdR incorporation at 0.01%.  Therefore, it is uncertain  whether
tetrachloroethylene induced a weak UDS response in this system.  Also, other
problems were found in this study.  The positive controls gave weak responses.
N-methyl-N'-n1tro-N-nitrosoguanidine elicited a weak response (1.8-fold
increase), but at a toxic concentration (5 ug/ml) as indicated by  a decrease
1n total DNA (6.27 ug of DNA versus 23.76 ug of DNA in solvent control).
Benzo[a]pyrene gave a response of 1.16-fold above background.  This is an
equivocal response, particularly because there  was also an increase found in
the total amount of DNA.
     Williams and Shimada (1983, sponsored by PPG Industries, Inc.) evaluated  two
different samples of tetrachloroethylene, Perchlor 200 (low-stabilized, 99.93%
purity) and Perchlor 230 (stabilized, purity 99.80%),  for their ability to
cause UDS 1n the hepatocyte primary culture (HPO/DNA  repair test.  The target
cells in this test system have a capability to  metabolize xenobiotics.
Williams and Shimada measured UDS by autoradiographic  determination of the
amount of [3H]-TdR (10 uCi/ml) incorporated into nuclear DNA.  Hepatocytes
were isolated from adult male Fischer 344 rats.  Cells were treated for 18  hours
or 3 hours.  For the 3-hour exposure, cultures  were incubated another 15
hours in the absence of tetrachloroethylene to  allow for DNA repair synthesis.
The criterion that was used for a positive result was  a net nuclear grain count
of 5 in triplicate coverslips.*  Negative results were reported for both Perchlor
230 (at concentrations of 0.001, 0.01, 0.1, and 1.0% v/v) and Perchlor 200  (at
     *Nuclear grain counts were reported as the mean +_ standard deviation.
Cytoplasmic grain counts 1n three nuclear  size areas adjacent to the nucleus
were determined.  The highest cytoplasmic  grain cbunt was subtracted from the
nuclear count.  This value is referred to  as "net" grain count.

                                     8-16

-------
concentrations of 0.0001, 0.001, 0.1, and 1.0%)  when tests were conducted
using conventional liquid-phase exposure in which tetrachloroethylene was
added to the culture medium.
     For both materials, positive responses were reported when testing was
performed using vapor-phase exposure in gas tight chambers.  Testing was conducted
at 0.1, 1.0, and 2.5% v/v (desired air concentration) for 3 and 18 hours.
Perchlor 230 at 0.1% caused an increase in UDS when the cells were exposed for
3 hours (6.2 +_ 4.9 net nuclear grain count, 50% cells showing toxic effects*)
and for 18 hours (15.9 +_ 1.6 net nuclear- grain count, 25% cells showing toxic
effects).  Perchlor 200 caused an increase in UDS at 0.1% v/v for the 3-hour
treatment (10.8 +_ 6.1% net nuclear grain count, 75% cells showing toxic effects)
but not for the 18-hour treatment.  At 1.0% and 2.5% for both materials at
both treatment times, nearly 100% of cells showed toxic effects.  It should be
pointed out that the background control values were taken from the conventional
exposure experiment.  This makes intrepretation of the results difficult,
particularly because the conventional-phase media was different from the gas-
phase media.  Because these data are based on one test in which there were no
concurrent positive and negative controls, the results are considered only
suggestive of a positive effect.  To validate these findings it is necessary
to repeat experiments using appropriate concurrent controls and a concentration
range to demonstrate a dose-response.  Although the possibility exists that an
impurity(ies) may be responsible for- the observed effects, the authors, did not
examine high-purity Perchlor (99.98+%) as they did in the Salmonella tests
discussed earlier-.  If high-purity material had tested negative under the same
experimental conditions under which  the lower-purity materials tested positive,
as in the Salmonella test, a stronger  argument could be made that impurities were
causing the effects.
     Williams (1983) conducted HPC/DNA repair tests using hepatocytes from male
B6C3F1 mlce and ma1e Osborne-Mendel  rats on tetrachloroethylene (99+% purity,
Aldrich Chemical Company).  Conventional liquid-phase exposure conditions
were used.  Negative results were reported for both rat and mice hepatocytes
when tetrachloroethylene was added to  the culture medium at 0.00001, 0.0001,
      'Toxicity  identified by the absence of S phase cells and general cellular-
morphology.  This  is not an accurate method of determining toxicity.
                                    8-17

-------
0.001, 0.01, and 0.1% (v/v) for 18 hours.  The material  was reported as "toxic"
at 0.01% and higher.  The highest cytoplasmic grain count was subtracted from
the nuclear count.  This reduces the possibility of false positives, but the
chance of missing a weak UDS inducer would be increased, especially if the
cytoplasmic grain count is high.  This consideration also applies to the Williams
and Shimada conventional-phase exposure study discussed previously.  The cytoplasmic
grain counts were not reported in any of the HPC/DNA repair studies.
8.3.2  Mitotic Recombination
     Call en et al. (1980) evaluated the ability of tetrachloroethylene (purity
not reported, stabilized 0.01% thymol, Eastman Kodak) to cause mitotic gene
conversion (nonreciprocal recombination) at the trp-5 locus,  mitotic crossing-
over (reciprocal recombination) at the ade-2 locus, and gene mutation (reversion)
at the ilv-1 locus in log phase cultures of Saccharomyces cerevisiae D7.
Cells were incubated for one hour in culture medium containing 0, 4.9, 6.6, and
8.2 mM tetrachloroethylene.  No exogenous source of metabolic activation was
used in these studies.  At 4.9 mM (84% survival), no significant increases
in the frequency of gene conversion (1.9 convertants/lO^ survivors versus 1.4
convettants/lO^ survivors in background control) and mitotic crossing-over
(5.3 mitotic recombinants/lO4 survivors versus 3.3 mitotic recombinants/104
survivors in background control) occurred.  As shown in Figure 8-2, when the
concentration of tetrachloroethylene was increased to 6.6 mM (58% survival),
increases in mitotic gene conversion and mitotic crossing-over  did occur (8.3
convertants/105 survivors and 52.6 mitotic recombinants/104 survivors,
respectively).  Mitotic recombinational activity was not determined at 8.2 mM
because less than 0.1% survival was found.  No significant increases in gene
mutations were observed at 4.9 mM (3.8 revertants/106 survivors versus 2.9
revertants/106 survivors in control).  The reverse mutation frequency was
not determined at 6.6 mM.  Therefore, the induced number of revertants is too
low to be indicative of a positive result, but the high values for the
recombinational events do indicate a positive effect at 4.9 mM test material.
The possibility that the effects were caused by a mutagenic impurity(ies)   . .
should be considered.  It should be pointed out that in the study by Callen et
al. the Ade* recombinants were estimated from a total of 30 plates, ten of
which contained minimum medium (plus adenine and isoleucine)  used for estimating

                                    8-18

-------
                            4            6

                     TETRACHLOROETHYLENE (mM)
10
Figure  8-2.  Induction of mitotic recombination  by  tetrachloroethylene in
            Saccharomyces cerevisiae D7.  The frequency of mitotic crossing-
            over (•—•} and  gene conversion (A---A) was determined.  Log-
            phase yeast cells were treated with test chemical for one hour
            without the addition of an exogenous metabolic activation system.
            (Adapted from Call en et al. 1980)


                                        8-19

-------
the number of trp-5 convertants.   Because mitotic crossing-over- and gene conversion
are not necessarily distinct events, in that they probably depend on a common
inducible mechanism, the number of Ade+ recombinants may have been overestimated
by including those Ade+ that were already Trp+.   There is experimental support
that these recombinational  events are inducible  by a common factor(s) (see Fabre
Fabre and Roman 1977, Fabre 1978).  The positive findings of Call en and coworkers
for mitotic recombination should be confirmed by repeating the assay using
appropriate selection conditions.  In addition,  because the responses were
observed within a narrow "window," at least one  additional concentration between
the low and high doses used should be tested to  demonstrate a clear dose-response.
     Bronzetti et al. (1983) also used the yeast S. cerevisiae D7 to evaluate
the effects of tetrachloroethylene (99.5% pure,  stabilized with 0.01% thymol,
Carlo Erba Co.) at the trp-5, ade-2, and ilv-1 loci.  In this study, however,
negative results were obtained after a 2-hour treatment at 5, 10, 20, 60, and
85 mM in the presence or- absence of S9 mix (Aroclor-1254 induced rat liver).
Differences in the experimental protocol from that used by Call en et al. may
explain these negative findings.   Bronzetti et al. used cells in the
stationary phase of growth rather than in the log phase of growth.  For some
chemicals it has been observed that stationary cells are more refractory than
log cells to mutagenic treatment (Mayer and Coin 1980, Shahin 1975).
Bronzetti et al. were able to test higher concentrations than Callen and
coworkers.  In the absence of S9 mix, Callen et al. reported less than 0.1%
survival at 8.2 mM, while Bronzetti et al. did not observe complete killing
until 85 mM.  Another difference between these two studies is the source of
tetrachloroethylene.  Bronzetti et al. purchased tetrachloroethylene from
Carlo Erba Co. (Milan, Italy) and Callen et al.  obtained the test agent from
Eastman Kodak Co. (Rochester, N.Y.); thus, it is possible that the test samples
may have contained different impurities.
     Bronzetti et al. (1983) also obtained negative results in an intrasanguineous
host-mediated assay using _S. cerevisiae 07 as the indicator organism and CD-I
mice as the host.  Stationary yeast cells (4 x 10^ cells) were injected into
the retro-orbital sinus of mice.  After injection of the yeast, an acute oral
dose of 11 g tetrachloroethylene/kg body weight (b.w.) was given.  A subacute
dose of 2 g/kg b.w. given 5 days a week for a total of 12 administrations was

                                    8-20

-------
also used (the last test dose was 4 g/kg b.w.;  therefore,  the total  dose was 26
g/kg b.w.).  In the subacute study, yeast cells were injected after  the last
dose of tetrachloroethylene (i.e., 4 g/kg b.w.).  Four hours after injection of
yeast, in both the acute and subchronic study,  the cells were recovered from
the liver, lungs, and kidneys of three animals.  No concurrent positive control
chemical was used in these studies to ensure that the system was functioning
properly.
     The positive responses discussed above for mitotic recombination in yeast
and DMA repair synthesis in mammalian cells provide suggestive evidence that
certain technical samples of tetrachloroethylene may be active in damaging DMA.
However, toxic concentrations of material were needed to elicit these responses.
The possibility that impurities and/or added stabilizers caused the increased
effects should be considered.

8.4  DMA BINDING STUDIES
     Chemical adduct formation is a critical step in certain types of mutagenesis.
Schumann et al. (1980) reported that there was no detectable binding of ^4C-
labeled tetrachloroethylene  (99% purity) to DNA when mice were treated by inhala-
tion at 600 ppm for 6 hours or when they were given an acute dose of 500 mg/kg
orally.  The specific activity of the 14C-label (26,593 dpm/umol in the
inhalation study and 133,273 dpm/umol in the oral study) is too low, however,
to preclude the possibility of very low levels of DNA binding.  For example, at
the specific activities used under the experimental conditions, at an assumed
binding level of 10~5 alkylations per nucleotide (Stott and Watanabe 1982)
there would be 5-10 dpm per-  sample.  This is at the limit of practical
detection.  Therefore, the possibility of binding at slightly less than 10~5
alkylations per- nucleotide cannot ruled out.  These negative findings, however,
are consistent with the negative and weak results reported in the mutagenicity
tests discussed above.

8.5   STUDIES INDICATIVE OF MUTAGENICITY  IN GERM CELLS
     An  important aspect of  a mutagenicity evaluation is to assess the potential
of the  chemical to reach the germinal tissue of humans and cause mutations
that may contribute to the genetic disease bur-den.  This assessment is almost

                                    8-21

-------
always based on animal  experimentation.  The ability of tetrachloroethylene to
cause genetic damage in germinal  tissue has not been well  studied.   The only
test results available were from a dominant lethal  study in rats [CRL:  COBS
CD(SD)BR] and a sperm morphology assay in both rats and mice (strain CD-I).
Tetrachloroethylene (91.43% purity, North Strong Division  Chemicals) did not
cause an increase in dominant lethals in rats when  unexposed females were
mated during a 7-week period to exposed adult males given  an acute  dose of 100
ppm and 500 ppm by inhalation for 7 hour's per day for 5 days (Beliles et al.
1980).  The dominant lethal assay is generally thought to  measure gross chromosome
damage (Bateman and Epstein 1971).  Also, this test is not considered a sensitive
assay because of the high spontaneous level of lethal events (Russell and
Matter 1980), and thus, negative results do not necessarily indicate that the
chemical does not reach and damage the germ cell DMA.
     Beliles et al. (1980) also examined tetrachloroethylene for altered sperm
morphology in treated rats and mice.  After- dosing  at 100  ppm and 500 ppm by
inhalation 7 hours per  day for 5 consecutive days,  groups  of four animals
were killed at the end of 1 week, 4 weeks, and 10 weeks to examine  effects on
various germ cell stages.  Sperm was collected from the cauda epididymis, and
at least 500 cells were examined.  Negative results were reported in rats;
the mice, however-, showed positive responses.  At 500 ppm, 19.7% abnormal
sperm were observed (versus 6.0% in the negative controls) during the fourth
week after exposure (corresponding to the spermatocyte stage).  By  themselves,
these positive findings alone are not sufficient to conclude that tetrachloroethy-
lene alters germ cell DMA because this assay is only an indicator of chemical
effects on sperm and does not provide definitive evidence  that a chemical
reached germinal tissue and damaged DNA.  Therefore, because limited information
was provided, it is not clear whether tetrachloroethylene  (or impurities)
reaches the germ tissue.  However, if tetrachloroethylene  reached germ cell
DNA, there may be no serious risk of mutation because of the largely negative
or marginal results found in mutagenicity tests discussed  previously.

8.6  MUTAGENICITY OF METABOLITES
     Trichloroacetic acid (TCA) is a known human metabolite of tetrachloroethylene.
The formation of TCA is thought to occur through the formation of an epoxide,
tetrachloroethylene oxide, and its subsequent rearrangement to trichloroacetyl
                                    8-22

-------
chloride or trlchloroacetaldehyde,  which then rapidly hydrolyzes to TCA.   (See
chapter 5  on metabolism for a more detailed discussion.)   These intermediates are
considered relevant in assessing the mutagenicity of tetrachloroethylene.
     Tetrachloroethylene oxide, which is considered to be  the biologically active
intermediate of the parent compound tetrachloroethylene, was assayed for'  mutagenicity
1n the absence of exogenous metabolic activation in several bacterial  tests
(Kline et al. 1982).  Tetrachloroethylene oxide increased  the number of revertants
in a dose-dependent manner in Salmonella tester strain TA1535 when assayed by
a preincubation liquid protocol (20 minutes at 37°C), but  did not cause an
Increase in revertants using Escherichia coli WP2 uvr A.   In Salmonella,  a
14-fold increase occurred at 2.5 mM and a 20-fold increase occurred at 5mM.
Tetrachloroethylene epoxide was toxic at 25mM.
     This epoxide was also evaluated by Kline et al. (1982) in the E.  coli pol
A assay.  Positive effects were observed at 0.04, 0.09, and 0.44 mM/ml as
measured by differential growth inhibition of a DMA polymerase-deficient strain
in comparison with its polymerase-proficient parent.
     Waskell (1978) tested TCA at 0.45 mg/plate in Salmonella TA98 and TA100
and obtained negative results.  There are other intermediates which have not
been identified in humans but are thought to occur- (e.g.,  trichloroethanol
chloral hydrate).  Waskell (1978) obtained negative results for trichloroethanol
1n Salmonella TA98 and TA100 up to a dose of 7.5 mg/plate.  Gu et al.  (1981),
however-, reported suggestive evidence that trichloroethanol weakly induced
sister chromatld exchange (SCE) formation in primary cultures of human lympho-
cytes.
     Chloral hydrate was reported to be marginally mutagenic (less than twofold
increase over- a dose range of 0.5 to 10 mg per plate) in Salmonella TA100  (Waskell
1978).  Gu et al. (1981) also provided suggestive evidence than chloral hydrate
at 54.1 mg/1 caused a weak increase in SCEs in cultured human lymphocytes.
Chloral hydrate has also been shown to block spindle elongation in insect
spermatocytes (Ris  1949).  Data on metabolites of tetrachloroethylene
suggest that if the parent compound was biotransformed, its metabolites may be
genotoxic;  these data are limited, however, and additional studies are needed
on metabolism and on the mutagenicity of metabolites to reach a clear- conclusion.
                                    8-23

-------
8.7  SUMMARY AND CONCLUSIONS
     Tetrachloroethylene itself has not been clearly shown to be a mutagen.
Certain commercial and technical  preparations have elicited positive responses
in the Ames bacterial test, a yeast recombinogenic assay, a host-mediated
assay using Salmonella, and DMA repair assays.  In general, the responses were
weak, and eliciting them required rather high toxic concentrations of
tetrachloroethylene.  No dose-response relationships were established in
these studies.  The positive findings may be explained by the presence of
mutagenic contaminants and/or- added stabilizers.  Highly purified tetrachloro-
ethylene has only been evaluated in the Ames/Salmonella test, where negative
results were obtained.
     Several other- tests of commercial and technical samples have been reported
to be negative.  In addition, The National Toxicology Program (NTP) has recently
sponsored mutagenicity testing (a modified Ames Salmonella test, a sex-linked
recessive lethal test in Drosophila, sister chromatid exchange formation and
chromosome aberrations in Chinese hamster ovary cells in vitro on a technical
sample of tetrachloroethylene; the preliminary results were negative.  (These
studies are in the process of being peer-reviewed, and were not discussed in
this chapter, except for the Ames test.)  The inconsistencies of available
results on different samples of tetrachloroethylene may be a function of the
toxicity of the test material, of exposure conditions used for testing this
volatile chemical, or of differences in sample contaminants and/or added stabil-
izers.  Information on chemical composition of the tetrachloroethylene test
samples was scarce.
     Although tetrachloroethylene itself has not been shown to be mutagenic,
it should be emphasized that the negative results are not wholly unequivocal.
Appropriate concurrent controls, adequate sample sizes, and exposure conditions
were sometimes not used, and in some cases the available data are not sufficient
to determine whether an adequate test was conducted.  Also, there have been no
reliable studies investigating the ability of tetrachloroethylene to cause
chromosome nondisjunction, which would result in aneuploidy, a significant
genotoxic effect.
     Because the epoxide of tetrachloroethylene was mutagenic in bacterial studies,
the concern should be raised that it may pose a mutagenic hazard.  It should be

                                    8-24

-------
noted,  however,  that the parent compound was  assayed  in  the  presence  of  several
types of metabolic activation systems (i.e.,  liver  homogenates,  intact hepatocytes,
and whole mammals) and the results were largely  negative or  weakly  positive.
Therefore, it is uncertain whether these negative or  weak findings  were  the
result of limitations of the activation systems,  the  epoxide not being produced
in sufficient quantities, or the epoxide possessing too  short a  half  life to
cause a detectable mutagenic response.
     In conclusion, inadequate information exists to  war-rant a provisional
classification of tetrachloroethylene either  as  nonmutagem'c or-  mutagenic.   If
tetrachloroethylene is a mutagen, the evidence available thus far indicates
that it is only weakly so.  (Because of insufficient  information, this conclusion
is not made with regard to its potential for  causing  chromosome  nondisjunction.)
Certain commercial and technical preparations of tetrachloroethylene  may contain
mutagenic impurities and/or added stabilizers.  Although there may  be mutagenic
agents in certain preparations of tetrachloroethylene,  usually large  amounts  of
material (at toxic levels) were required to elicit  weak  responses.
                                    8-25

-------
8.8  REFERENCES

Bartsch, H., C. Malaveille, A. Barbin,  and G.  Planche.   1979.   Mutagenic  and
     alkylating metabolites of halo-ethylenes, chlorobutadienes and dichlorobutenes
     produced by rodent or  human liver tissues.  Arch.  Toxicol. 41:249-277.

Bateman, A.J. and S.S. Epstein.  1971.   Dominant lethal  mutations in mammals.   In:
     Chemical mutagens:  Principles and methods for their  detection, Vol.  2
     (A. Hollaender, ed.) Plenum Press, New York, pp. 541-568.

Beliles, R.P., D.J. Brusisk, and F.J. Mecler.   1980.  Teratogenic mutagenic
     risk of workplace contaminants:   trichlorethylene,  perchloroethylene, and
     carbon disulfide.  Contract No.  210-77-0047.  Litton bionetics, Inc.,
     Kensington, Maryland.

Bronzetti, G., C. Bauer, C. Corsi,  R. Del  Carratore, A.  Galli,  R. Nieri,  and
     M. Paolini.  1983.  Genetic and biochemical studies on perchloroethylene
     in vitro and in vivo.   Mutat.  Res. 116:323-331.

Callen, D.F., C.R. Wolf, R.M. Philpot.   1980.   Cytochrome P450  mediated genetic
     activity and cytotoxicity of seven halogenated aliphatic  hydrocarbons in
     Saccharomyces cerevisiae. Mutat. Res. 77:55-63.

Cerna, N., and H. Kypenova.  1977.   Mutagenic  activity of chloroethylenes
     analyzed by scr-eening system test.  Mutat. Res. 46:36 (Abst.).

Fabre, F.  1978.  Induced intragenic recombination in yeast can occur- during
     the GI mitotic phase.   Nature 272:795-798.

Fabre, F., and H. Roman.  1977.  Genetic evidence for- inducibility of recombination
     competence in yeast.  Proc. Natl.  Acad. Sci. USA 74:1667-1671.

Greim, H., G. Bonse, Z. Radwan, D.  Reichert, and D. Henschler.   1975.  Mutagenicity
     in vitro and potential carcinogenicity of chlorinated ethylenes as a  function
     oT metabolic oxirane formation.   Biochem. Phar-macol.  24:2013-2017.

Gu, Z.W., B. Sele, P. Jalbert, M. Vincent, C.  Marka, D.  Charma, and J. Faure.
     1981.  Induction d'echanges entre les chromatides soeurs  (SCE) par- le
     trichloroethylene et ses metabolites.  Toxicological  European Research
     3:63-67.

Henschler, D.  1977.  Metabolism and mutagenicity of halogenated olefins:  a
     comparison of structure and activity.  Environ. Health Per spec. 21:61-64.

Ikeda, M., A. Koizumi, T. Watanable,  A. Endo,  and K. Sato.  1980.  Cytogenetic
     and cytokinetic investigations on lymphocytes from workers occupationally
     exposed to tetrachloroethylene.   Toxicol. Letters.   5:251-256.

Kastenbaum, M.A. and K.O. Bowman.  1970.  Tables for determining statistical
     significance of mutation frequencies.  Mutat. Res.  9:527-549.
                                      8-26

-------
Kline, S.A., E.G.  McCoy,  H.S.  Rosenkranz,  and  B.L.  Van  Duuren.   1982.   Mutagenicity
     of chloralkene epoxides in bacterial  systems.   Mutat.  Res.  101:115-125.

Margard, W.  1978.  In vitro bioassay of chlorinated hydrocarbon solvents.
     Battelle Laboratories.   Unpublished proprietary document for Detrex Chemical
     Industries.

Mayer, V.W., and C.T.  Coin.   1980.   Induction  of mitotic  recombination  by
     certain hair-dye chemicals in  Saccharomyces cerevisiae.   Mutat.  Res. 78:243-252.

McCann, J., E. Choi, E. Yamasaki,  and B.N. Ames.  1975.  Detection of carcinogens
     as mutagens in the Salmonella/microsome test:   Assay of  300 chemicals.   Proc.
     Natl. Acad. Sci.  USA 72:5125-5139.

NTP.  1983.  Unpublished data on tetrachloroethylene:   on Salmonel1 a/mi crosome
     preincubation test provided by Or.  E. Zeiger.

Rampy, L.W., J.F.  Quast,  M.F. Balmer, B.K.J. Leong, and P.J.  Gehring.   1978.
     Results of a long-term inhalation toxicity study.   Perchloroethylene in  rats.
     Toxicology Research Laboratory.   Health and Environmental  Research.  The Dow
     Chemical Company.  Midland, Michigan.  Unpublished.

Ris, H. 1949.  The anaphase movement of chromosomes in  the spermatocytes of the
     grasshopper.   Biol. Bull. 96:90-106.

Russell L.B., and B.E. Matter.  1980.  Whole mammal mutagenicity tests.  Evaluation
     of five methods.   Mutat. Res.  75:279-302.

Schumann, A.M., J.F. Quast, and P.G.  Watanabe.  1980.  The pharmacokinetics and
     macromolecular interactions of perchloroethylene in mice and rats  as related  to
     oncogenicity.  Toxicol. Appl.  Pharm.   55:207-219.

Shahin, M.M.  1975.  Genetic activity of niridazole in  yeast.  Mutat. Res.
     30:191-198.

SRI International.  1983.  Salmonella test results on tetrachloroethylene.
     Prepared for- U.S Environmental Protection Agency,  Dr. Harry Milman, project
     officer.  Unpublished.

Stott, W.T. and P.G. Watanabe.  1982.  Differentiation  of genetic versus epigenetic
     mechanisms of toxicity and its application to risk assessment.  Drug Metabolism
     Reviews 13:853-873.


Schlossberg, L.   (Detrex Chemical  Industries,  Inc. Detroit, Michigan)  January
     5, 1981.  Memorandum to Dr. V. Vaughan-Dellarco of the U.S. Environmental
     Protection Agency, Reproductive Effects Assessment Group.

Waskell, L.  1978.  A study of the mutagenicity of anesthetics and their
     metabolites.  Mutat. Res. 57:141-153.
                                      8-27

-------
Williams, G.M.  1983.   DMA repair tests of 11  chlorinated  hydrocarbon  analogs.
     Prepared for ICAIR Life systems,  Inc. TR-507-18 and U.S.  Environmental
     Protection Agency.  Dr. Harry Mllman, project officer.   Unpublished.

Williams, G.M., and T. Shimada.   January  1983.   Evaluation  of several  halogenated
     ethane and ethylene compounds for genotoxicity.  Final  report  for PPG
     Industries, Inc., Pittsburgh, Pennsylvania.   Unpublished.
                                      8-28

-------
                             9.0   CARCINOGENICITY

     The purpose of this section is to provide an evaluation of the likelihood
that tetrachloroethylene (perchloroethylene)  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 carcinogenic!ty depends  heavily on animal
bioassays and epidemiologic evidence.   However, information on mutagenicity and
metabolism, particularly in relation to interaction with DNA, as well  as to
pharmacokinetic behavior, has 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 tetrachloroethylene.

9.1  ANIMAL STUDIES
     Two long-term animal bioassays have been performed  to assess the carcino-
genic potential of tetrachloroethylene.  In one study involving exposure of rats
and mice to tetrachloroethylene by gavage, the National  Cancer Institute (NCI)
(1977a) reported the induction of hepatocellular carcinomas in male and female
mice, but determined that the test with rats  was inconclusive because of excessive
mortality.  In the other study, in which Sprague-Dawley  rats were exposed to
tetrachloroethylene by inhalation, the Dow Chemical Company (Rampy et al. 1978)
reported no evidence for- the carcinogenicity  of the chemical.  However, limita-
tions in this study make it difficult to assess the carcinogenic potential of
tetrachlor oethylene.
9.1.1  National Cancer Institute Bioassay (1977a)
     The tetrachloroethylene sample used in this bioassay was purchased from
the Aldrich Chemical Company, Milwaukee, Wisconsin.  Analysis by gas-liquid
chromatography and infrared spectroscopy yielded results indicating a purity
of > 99% with at least one minor impurity not identified in the report.
Identification of the impurities in the test sample was  not made (personal
communications with the NCI and the Aldrich Chemical Company).

                                    9-1

-------
     The carcinogenicity of tetrachloroethylene was tested in Osborne-Mendel
rats and B6C3F1 mice.  The initial  age of the weanling animals was 25 days for
the mice and 35 days for the rats.   Two treatment groups consisted of 50 males
and 50 females, and matched vehicle (corn oil) and untreated control  groups
comprised 20 animals of each sex.  Selected dosage levels were those  determined
to be maximally tolerated in an 8-week subchronic study, i.e., a dosage that
was not fatal and/or did not reduce body weight gain more than approximately
20%, and one-half maximally tolerated in an 8-week subchronic toxicity test.
Time-weighted average doses (mg/kg/dose) in the chronic study were 941 and 471
for- male rats, 949 and 474 for female rats, 1,072 and 536 for male mice, and
772 and 386 for female mice.  Tetrachloroethylene was administered to the animals
by gastric intubation in corn oil once each day, 5 days/week, for 78  weeks.
During the final  26 weeks of treatment, doses were administered to rats in a
cyclic pattern of 1 week without treatment followed by 4 weeks with treatment.
Body weights and food consumption were obtained weekly for the first  10 weeks
and monthly thereafter.  Mice and rats were permitted to survive an additional
12 and 32 weeks after treatment, respectively, until sacrifice.
     Each animal  was submitted to extensive gross and microscopic examinations.
Specified organs, plus any other tissue containing visible lesions, were fixed
in 10% buffered formalin, embe'dded in paraplast, and sectioned for slides.
Hematoxylin and eosin staining (H and E) was used routinely, but other- stains
were employed when needed.  Diagnoses of observed tumors and other  lesions were
coded according to a modified Systematized Nomenclature of Pathology  (SNOP)
originally developed by the College of American Pathologists in 1965.
     Tetrachloroethylene was found to be carcinogenic in mice in this study.
Results summarized in Table 9-1 indicate that tetrachloroethylene induced highly
statistically significant (P < 0.001) increases in the incidence of hepatocellular
carcinomas in both sexes of mice in both treatment groups as compared to untreated
controls or vehicle-controls.  The microscopic appearance of carcinomas was
variable, with some tumors composed of well-differentiated hepatocytes arranged
in rather uniform hepatic cords, and other lesions consisting of anaplastic cells,
often with inclusion bodies with vacuolated, pale cytoplasm.  Mitotic figures
were often present.  In male mice,  the first hepatocellular carcinomas were
detected at 27 weeks in the low-dose group, 40 weeks in the high-dose group,

                                    9-2

-------
       TABLE  9-1.   INCIDENCE OF HEPATOCELLULAR CARCINOMAS IN B6C3F1 MICE
                           FED TETRACHLOROETHYLENE
                       (National Cancer  Institute 1977a)
Dose (nig/kg/day)3
Male
untreated
vehicle-control
536
1072
Femal e
untreated
vehicle-control
386
772
Hepatocellular carcinomas

2/17 (12%)
2/20 (10%)
32/49 (65%)
27/48 (56%)

2/20 (10%)
0/20 (0%)
19/48 (40%)
19/48 (40%)
P valuesb



P < 0.001
P < 0.001



P < 0.001
P < 0.001

«nme-weignted^ average doses.
^Probability level  (P-values)  for  the  Fisher  Exact Test  comparison  of  dose
 groups with vehicle-control  group.
and 90 and 91 weeks in vehicle-control  and  untreated  control  groups.   In  female
mice, the first hepatocellular carcinomas were  observed  at week  41  in  the
low-dose group, week 50 in the high-dose group,  and week 91 in the  untreated
control  group.  Metastases of hepatocellular  carcinomas  occurred in the kidneys
of one untreated control  male and in the lung of three low-dose  males, one
low-dose female, and one high-dose female.
     Toxic nephropathy in mice was apparent in  40/49  low-dose males, 45/48  high-
dose males, 46/48 low-dose females,  and 48/48 high-dose  females. Control animals
did not exhibit this lesion.   Chronic murine  pneumonia was also  a frequently
observed finding.  A low incidence of bloating  or abdominal distension was

                                    9-3

-------
noted 1n treated animals during the second year of the study.   Body weight gain
was comparable between groups (Figure 9-1).  Median survival  times were greater
than 90 weeks in control males, 78 weeks in low-dose males,  and 43 weeks in
high-dose males (Figure 9-2).  Median survival  times for females were greater
than 90 weeks in control females, 62 weeks in low-dose females, and 50 weeks  in
high-dose females (Figure 9-2).
     In rats, toxic nephropathy, not found in control  animals,  was detected in
43/49 low-dose males, 47/50 high-dose males, 29/50 low-dose  females, and 39/50
high-dose females.  Figure 9-3 indicates that treated rats gained less weight
than controls, though the difference was slight, with maximum reduction being
13% during the first year and 19% during the second year.   Clinical signs
apparent in treated animals included a hunched appearance and urine stains on
the lower abdomen.  Respiratory abnormalities, characterized by dyspnea, wheezing,
and/or  reddish nasal discharge, were noted with increased incidence in all  groups
during aging of the animals, and chronic murine pneumonia was diagnosed in >^ 62%
of the animals in each group.  As indicated in Figure 9-4, median survival  times
were greater than 88 weeks in the control groups, 68 weeks in low-dose females,
66 weeks in high-dose females, 67 weeks in low-dose males, and 44 weeks in
high-dose males.  The survival was not adequate to support any conclusions
about the cardnogenicity of tetrachloroethylene in rats.
     In an attempt to characterize impurities present in the tetrachloroethylene
product used in this study, documented chemical analyses of  the test samples
performed at the carcinogenicity testing laboratory of the NCI bioassay program
were examined by the analytical department of the Diamond Shamrock Corporation
(interoffice memorandum from E. A. Rowe to G. K. Hatfield, Diamond Shamrock
Corporation, October 8, 1979, obtained with the documented chemical analyses
from G. K. Hatfield, January 28, 1981).  As indicated in the interoffice memo-
randum, tetrachloroethylene samples used in the National Cancer Institute
bioassay (NCI 1977a) were not available for- analysis at Diamond Shamrock;
however, the analytical method, with the same type of instrument and column
used at the carcinogenicity testing laboratory, was reproduced.  Evaluation of
the analytical method led to the conclusions presented in the previously mentioned
memorandum that the method could not distinguish epichlorohydrin from trichloro-
ethylene, and that the tetrachloroethylene product used in the NCI (1977a)

                                    9-4

-------
                                                                          -40
                                                                          - 30
                                                        UNTREATED CONTROL
                                              	•	VEHICLE CONTROL

                                              • •••••••< LOW DOSE

                                              — —	HIGH DOSE
                                                                          -20
                                           - 10
45      60       75

TIME ON TEST (WEEKS)
                                                        90
                                                                105
120
3U —
_ 40-
cc
0
- 30-
I
O
UJ
§2°-
m
2
UJ
5 10-

0-

/x
-^t^"*™--«Sr^ — ^C^J^^'^^r
^t+gsJ*^' ^^*nT.. ^^


•— • — 	 • VEHICLE CONTROL
• «•••••• LOW DOSE
FEMALE MICE 	 HIGH DOSE
i 1 i 1 i 1 i 1 i I i 1 i 1 i
j la jG **5 6^ /o SO iuj "A
— su
-40
— 30


- 20
- 10


^
                               TIME ON TEST (WEEKS)
Figure 9-1.   Growth curves for male  and  female mice  in  the tetrachloroethylene
              chronic study.   (National  Cancer  Institute  1977a).
                                    9-5

-------




0.8-
_J

>
>
OC
V) 06 —
u.
O
>
h-
_j
5 0.4-
<
CD
O.

0.
0.2 -


.0 —
C


*M """a1 "^
1 •• 1 1
I ••••••••"•*• ^~ I
1 *** *— — 1

1 • **: L

t| *•• • • • •« • ^
**• ^
n« i^^* "
^ ^
_ • •
•^ •
• ••



	 VEHICLE CONTROL "I — —
• •••••• LOW DOSE
— 	 HIGH DOSE
1 1 ' 1 ' 1 ' 1 ' | ' 1 ' I
) 15 30 45 60 75 90 105 12

1.0


— 0.8


-
-06





- 0.4



-0.2


0
                                    TIME ON TEST (WEEKS)



0.8-
^
^
—

-------
    750-T-
                                                            UNTREATED CONTROL

                                                 	—	VEHICLE CONTROL

                                                 • ••••••••  LOW DOSE

                                                 — — — -  HIGH DOSE
                 I
                 15
 I
30
 I
45
60
 I
75
 I
90
 I
105
                                                                               750
                                                                             — 600
                                                                             — 450
                                                                             — 300
                                                    - 150
                                                   120
                                  TIME ON TEST (WEEKS)
     750
                                                                               750
                                                                              — 600
                                                                              — 450
                                                   	VEHICLE CONTROL

                                                   • ••«•••  LOW DOSE

                                                   — — —  HIGH DOSE
                                                                              - 300
                                                    - 150
                 15
                                                                    105
                                                                            120
                                  TIME ON TEST (WEEKS)
Figure 9-3.   Growth curves for male  and female  rats in the  tetrachloroethylene
              chronic  study.  (National  Cancer  Institute  1977a).
                                       9-7

-------

        0.8-
      cc
      3

      u.
      O
      03
      <
      m
      O
      QC
      Q.
0.6-
0.2-
        o.o
               •••      1
                 • •••••
                                                                       MALE RATS
              UNTREATED CONTROL
 	VEHICLE CONTROL


      LOW DOSE


— — HIGH DOSE
     I    'I
    15       30
                                      45
                                               1^
                                              60
                                                I
                                               75
 I
90
                                                                 105
                                                                           -1 0
                                                                           L-0.8
                                                                           -0.6
                                                                                   -0.4
                                                                                  — 0.2
                                                                                    •0.0
                                                                                 120
                                      TIME ON TEST (WEEKS)

        0.8 -J
     _j
     <

     >
     

1
     =i 0.4
     m

     CO
     O
     cc
     0.
       0.2-1
       0.0-

• ••••• * —1 j 1
"***7}i— . ~~" *i i
••i_ l j_
••• ~1"~i ~~
- •••» '^
•-..^
• • • • ^ •
u.%
•j
-


_


_ 	 VEHICLE CONTROL
• •••••• LOW DOSE
— — — HIGH DOSE


FEMALE RAlb

1
•1 ' 	 1
^\ \
• ••• "^
"1 '• 	 	 ^
L ••*_..

~~ "^" ~~ t_ **i*
h__^
M «^» M^ •




i n

_
OQ
— 06

-


— 04

—
- 0.2


n n
            15       30        45       60       75


                              TIME ON TEST (WEEKS)
                                                                90
                                                                        105
                                                                                 120
Figure  9-4.   Survival  comparisons  of male and  female rats  in the  tetrachloroethylene
               chronic  study.   (National  Cancer  Institute  1977a).
                                           9-E

-------
bloassay could have contained one or both of these compounds  as impurities.
The documented chemical  analyses show contaminant levels of 0.055%,  0.041%,
and 0.010% in tetrachloroethylene samples analyzed at the beginning  of the
bloassay, at 1 year into the bloassay, and at 2 years into the bloassay,
respectively.  The conclusion stated in the memorandum 1s that the contaminant
was probably eplchlorohydrln, since:  1) epichlorohydrln was  a commonly used
stabilizer for tetrachloroethylene at that time; 2) a reported analysis by  a
competitor of Diamond Shamrock showed a maximum of 0.015% trlchloroethylene in
any tetrachloroethylene product manufactured 1n the United States, and 3) the
decreased amount of impurity found in the tetrachloroethylene samples as  the
bloassay progressed suggests some decomposition 1n that, according to the
experience of Diamond Shamrock, eplchlorohydrln but not trlchloroethylene
levels would decrease during storage.  Nonetheless, although  the evidence
provided by Diamond Shamrock indicates that eplchlorohydrln was present 1n  the
tetrachloroethylene product used in the NCI (1977a) bloassay, the quantity  of
epichlorohydrln in the test material used in this study remains uncertain.
Furthermore, the different levels of unknown material shown 1n the documented
chemical analysis may be due to the use of the indicated different lots,  possibly
containing unequal amounts of impurities.
     An estimate of the likelihood that the eplchlorohydrln impurity in the
tetrachloroethylene material used in the NCI experiment could have been large
enough to account for the positive results can be obtained by considering the
results of Laskin et al. (1980), who exposed rats to epichlorohydrln via inhalation.
They found that 1/100 rats exposed to 30 ppm (0.115 mg/1 air, equivalent to
13.2 mg/kg/day) of epichlorohydrln for 6 hours/day, 5 days/week, for 730 days
developed squamous cell nasal carcinomas, compared with 0/50  in control animals.
     The eplchlorohydrln impurity 1n the NCI tetrachloroethylene high-dose  male
mice experiment was approximately 1,032 mg/kg/day x 0.041% =  0.42 mg/kg/day, from
the discussion above.  This is only about 0.42/13.2 ~ 0.03 times the dose that
gave an incidence of only 1/100 in the Laskin et al. rat experiment.  Therefore,
it 1s unlikely that epichlorohydrln impurities at the level estimated to be present
1n the NCI experiment could have contributed appreciably to the positive response.
     In a second cardnogeniclty bloassay sponsored by the National  Toxicology
Program (NTP 1983, draft), tetrachloroethylene was given orally to B6C3F1 mice

                                    9-9

-------
and to four strains of rats (Sherman, Fischer 344, Long-Evans, and Wistar).  The
tetrachloroethylene sample used in this ongoing carcinogenicity bioassay did
not contain detectable amounts of epoxide contaminants.  In the study with
female B6C3F1 mice (NTP 1983, draft), groups of 100 females received 25, 50,
100, or 200 mg of more than 99% pure tetrachloroethylene per kg body weight in
corn oil  by gavage for 103 weeks, 5 days/week.  Vehicle-control and untreated
control groups of 100 mice each were used.  Survival was not affected significantly.
Doses of 50 mg/kg or- more produced a dose-related cytomegaly of the kidneys.
Other signs of liver toxicity appeared during the study as increases of sorbitol
dehydrogenase activity, and increases in relative liver weight and lipid levels
were associated with increasing dose levels.  Doses of 50 mg/kg or more produced
time- and dose-related increases in the incidence of hepatocellular adenomas
and carcinomas bearing no causal relationship to the observed renal damage.
The first adenoma appeared at week 46; the first carcinoma at week 58.  The
NTP intends to conduct an audit of the raw data of this study before the final
technical report is prepared.  The findings of the audit will determine the
validity of the study.  If the audit reveals no major problems, the NTP expects
to present the report to a peer review group no sooner then early 1984 (letter
from Dr.  Schetz, NTP, to Dr. Haberman, CAG, September 29, 1983).
9.1.2  Dow Chemical Company Inhalation Study (Rampy et a!. 1978)
     Two groups of weanling Sprague-Dawley (Spartan substrain) rats, each
composed of 96 males and 96 females, were exposed to 600 ppm (4.07 mg/1 air)
or 300 ppm (2.03 mg/1 air) of tetrachloroethylene 6 hours/day, 5 days/week, for
52 weeks.  An untreated group of 192 males and 192 females served as controls.
Controls were not put in inhalation chambers, but were in the treatment room
during exposure.  At the end of the treatment period of 52 weeks, animals
were allowed to survive until sacrifice at 31 months.  The composition of the
test material (Lot A12282D) by gas chromatographic analysis was as follows:
trichloroethylene, 3 ppm (liquid volume %); hexachloroethane, < 12 ppm; carbon
tetrachloride, 2 ppm; 4-methyl morpholine, 44 ppm; nonvolatile residue, 2 ppm;
tetrachloroethylene, balance.  Exposure was done in 3.7 m3 inhalation chambers
with a dynamic airflow system.  Analyses of tetrachloroethylene levels in the
inhalation chambers during the treatment period revealed analytical concentrations
(mean +_ standard deviation) of 310 +_ 32 ppm (244 analyses) and 592 +_ 62 ppm

                                    9-10

-------
(1,245 analyses).  Analytical  concentrations by infrared analysis within jf 10%
of nominal levels were achieved on 89.3% (low-dose)  and 97.1% (high-dose) of
the exposure days.  Animals were evaluated for clinical  toxic signs,  body
weight changes, urinalysis at 24 months, and hematology at 12 and 24  months.
Survivors and decedents were given gross and histopathologic examinations.
Bone marrow samples were taken from three males and  three females in  each group
sacrificed at 1 year for cytogenetic evaluation.
     Clinical signs of toxicity were not observed with the nominal concentrations
of tetrachloroethylene used in this study.  Mean body weight gains were similar
among groups.  Hematology and urine analyses showed  no treatment-related effects
of tetrachloroethylene.  The mortality patterns exhibited in the study are des-
cribed in Table 9-2.  Mortality in high-dose males was slightly greater than that
of controls during months 5 to 24; the earlier- onset of chronic renal disease in
this treatment group was considered to be a contributing factor in increased
mortality.
     No carcinogenic effects of tetrachloroethylene  were observed from pathologic
examination of the animals.  Statistical analysis of the data showed  numerous
non-neoplastic abnormalities that occurred spontaneously and were within the
normal variation encountered in lifetime studies with this strain of  rat.  With
respect to tumor findings, statistical analysis of the data did not reveal a
definite  increased tumor incidence in animals exposed to tetrachloroethylene.
Tumors or tumor-like changes in the kidney were found in 1/189 control, 2/94
low-dose, and 4/94 high-dose males during gross necropsy; however, light micro-
scopic examination of kidney lesions did not show a  statistically significant
tumor- incidence compared to controls.  Although many tumor- types were found in
treated and control animals, there was no statistically significant (P > 0.05)
increase  in tumor incidence at any anatomical site.
     The  results of this study do not indicate a definite carcinogenic effect
of tetrachloroethylene in Sprague-Dawley rats; i.e., the tumor incidence between
control and treated rats was similar.  However, this study has the following draw-
backs:  1) the period of exposure was only 12 months rather than  the lifetime of
the animals, which would have been a more appropriate duration for- carcinogenicity
studies;  and 2) the dose levels in this study do not appear to have been high
enough to provide maximum sensitivity.

                                    9-11

-------
       TABLE 9-2.  CUMULATIVE SURVIVAL OF SPRAGUE-DAWLEY RATS EXPOSED TO
                       TETRACHLOROETHYLENE FOR 12 MONTHS
                        (adapted from Rampy et al. 1978)
Month of Study        0 ppm
                  Male     Female
    300 ppm
Male       Female
    600 ppm
Male       Female
Initial
6
12
18
24
31
189
187
183
155
44
1
189
188
185
151
70
12
94
94
91
74
26
1
91
91
91
77
37
6
94
88a
84*
55a
13*
1
94
94
91
863
493
5

dP < 0.005 by the FisnerTxact Test.
9.1.3  Intraperitoneal Administration Study (Theiss et al.  1977)
     Thelss et al. (1977) tested tetrachloroethylene for cardnogenicity in the
strain A mouse pulmonary tumor induction system.  The test  sample,  a  product of
the Aldrich Chemical  Company, was reagent grade with a purity exceeding 95 to 99%.
Strain A/St male mice, 6 to 8 weeks old, were used in this  assay.  The maximum
tolerated dosage, defined as the dosage which five mice tolerated after- six
intraperitoneal injections over  a 2-week period followed by a 4-week  observation
period, was determined and used in the bioassay.  In the main test, 20 mice per
treatment group received three intraperitoneal  injections of 80,  200, or 400 mg/kg
of tetrachloroethylene weekly until total dosages of 1120,  4800,  and  9600 mg/kg,
respectively, were achieved.  Survivors were sacrificed at 24 weeks after the
first injection, and the number of surface adenomas was counted.   Results were
compared with findings in vehicle (tricaprylin) and untreated controls by the
Student t test.  Tetrachloroethylene did not statistically  increase (P > 0.05)
the incidence of pulmonary tumors in this study (Table 9-3).  This  strain was
sensitive to the positive control chemical urethan, as shown in Table 9-3.
                                    9-12

-------
          TABLE 9-3.   PULMONARY TUMOR RESPONSE TO TETRACHLOROETHYLENE
                       (adapted from Theiss et al. 1977)
                                            No.  survivors/        No.  lung
Compound               Dosage (mg/kg)        No.  animals           tumors/mouse
Tricaprylln
Tetrachl oroethyl ene
—
80
200
400
46/50
15/20
17/20
18/20
0.39 +_ 0.063
0.27 + 0.07
0.41 + 0.10
0.50 T 0.12
Urethan                  1,000 mg/kg            20/20              19.6^2.4
                        {1 injection)
aMean +_ S.E.

     A negative result in this assay is not considered conclusive, since several
chemicals known to be carcinogenic in chronic rodent bioassays induce no response
in this assay.
     The strain A mouse pulmonary tumor assay is relatively insensitive to mouse
carcinogens for which the effect is confined to the liver (Theiss et al. 1977).
For example, chloroform, 2-chloroethyl ether, and hexachlorocyclohexane induce
tumors of the liver (not other sites) in mice (NCI 1976, 1977c; Innes et al.
1969) but were not carcinogenic in the assay by Theiss et al.  (1977).  The
reasons for the negative lung response are not understood, but it may be due to a
smaller concentration of activating enzymes in the lung than in the liver.
9.1.4  Skin Painting Study (Van Duuren et al. 1979)
     A carcinogenicity study of purified tetrachloroethylene in ICR/Ha Swiss
mice was described by Van Duuren et al. (1979).  Maximum tolerated dosages were
determined in range-finding studies 6 to 8 weeks in duration,  and were
selected as dosages that did not affect body weight gain or- produce clinical
signs of toxicity.  This study included the following experiments:  1) 30
females were treated topically on the dorsal skin with a single application of
163 mg tetrachloroethylene followed 14 days later by the applications of 5.0 ug
phorbol myristate acetate (PMA) to the skin three times weekly until termination
of the study at 428 to 576 days; median survival time was 428 to 576 days.  2)
30 females were given thrice weekly topical applications of 54 mg tetrachloro-
ethyl ene for the duration of the test (440 to 594 days) with a median survival
time of 317 to 589 days.  A vehicle (acetone) control group of 30 mice and an
untreated control group of 100 mice were included in these experiments.
                                    9-13

-------
     In the initiation-promotion experiment,  210 mice treated with PMA alone were
also on test.  The mice were 6 to 8 weeks old at the beginning of the study, and
were housed six to a cage.  Test sites on the skin were shaved as necessary and
were not covered; however, it was the authors' impression that tetrachloro-
ethylene was immediately absorbed and that evaporation from test sites was
minimal (personal communication, B. L. Van Duuren, New York University).   The
animals were weighed monthly, and each animal was examined by necropsy.  Tumors
and lesions as well as skin, liver, stomach,  and kidneys were examined histo-
logically.
     Tetrachloroethylene did not show initiating activity in the initiation-
promotion experiment; the number of mice with skin papillomas (squamous cell
carcinomas) was:  4 (0) initiated with tetrachloroethylene, 15 (3) treated
with PMA alone, and 0 (0) in the control groups.  The study involving repeated
application to the skin produced lung and stomach tumors in 16 and 0 tetrachloro-
ethylene-treated mice, respectively; 11 and 2 vehicle-treated controls,
respectively; and 30 and 5 untreated controls, respectively.
     The negative results for tetrachloroethylene in mouse skin as an initiator
and as a complete carcinogen can be reconciled with the positive mouse liver-
response in the NCI study if it is hypothesized that the skin does not have the
necessary enzymes to convert tetrachloroethylene to an active metabolite,
whereas the liver does have this capability.
     The lack of sensitivity of skin application tests, as compared with tests
using other routes of exposure, is apparent from the results of Van Duuren et
al. (1979) with 1-chloroprene, cis-l,3-dich1oropropene, and 2-chloroproponal.
They found that none of the three compounds induced a response as initiator's in
initiation-promotion experiments or with repeated topical application on the
skin.  However, they did observe a statistically significant increase in the
incidence of forestomach tumors in female HatlCR Swiss mice dosed by gavage
with 1-chloroprene (P < 0.0005) and 2-chloroproponal (P < 0.05) and in the
Incidence of local sarcomas In female Ha:ICR Swiss mice treated with cis-1,3-
dlchloropropene (P < 0.0005) by subcutaneous injection.
                                    9-14

-------
9.2  EPIDEMIOLOGIC STUDIES
     There are six epidemiologic studies that relate to tetrachloroethylene
exposure.  Only one of these studies,  however, has actually identified workers
exposed to tetrachloroethylene.  Because tetrachloroethylene has been used in the
dry-cleaning industry, the present discussion also includes three proportionate
mortality studies of decedents who had worked in the dry-cleaning industry, as
well as two case-control studies in which the cases and controls were asked
about their employment histories, including employment in the dry-cleaning
industry.
9.2.1  Kaplan (1980)
     Kaplan (1980) did a retrospective cohort mortality study of dry-cleaning
worker-s exposed to tetrachloroethylene for at least one year prior to 1960.
The study was performed under contract to the Biometry Section of the National
Institute for Occupational Safety and Health (NIOSH) Industry-Wide Studies Branch.
     In a preface to the discussion of the study, Kaplan reported that levels
of tetrachloroethylene exposure were "much higher" for- cleaners (machine opera-
tors) than for- other employees of dry-cleaning establishments.  A geometric
mean time-weighted average for tetrachloroethylene was 22 ppm for the machine
operators.  For- all other jobs, the highest corresponding value was reported
to be 3.3 ppm.  These data were provided through a NIOSH industrial hygiene
survey of dry-cleaning facilities.
     The study cohort, selected from records maintained by several labor unions,
consisted of 1,597 individuals employed more than 1 year- prior to 1960 in  dry-
cleaning establishments.  The primary solvent was tetrachloroethylene.  Efforts
were made by the author to exclude all persons with previous occupational  exposure
to carbon tetrachloride or- trichloroethylene.  By September- 30, 1977, the  end of
the study period, 1,058 individuals were found to be alive, 285 were deceased,
and 254 were of unknown vital status.  The extent of follow-up varied by sex;
8% of the males and 20.4$ of the females remained lost to follow-up.  Race was
known only for deceased workers, and was obtained from death certificate data.
Because of the lack of information regarding race, observed deaths by cause
were compared to expected deaths by means of a standardized mortality ratio
(SMR) for whites, an SMR  for blacks, and a composite point estimate SMR for both.
Based on the assumption that every member of the cohort was white, expected

                                    9-15

-------
deaths for whites were derived by multiplying the person-years accumulated for
the entire cohort by the white death rates within 5-year age groups (separately
for males and females and then with the two combined).  Expected deaths for
blacks were similarly generated by assuming that all  cohort members were black.
Composite expected deaths were calculated by weighting the total accumulated
person-years for  the cohort in each 5-year age group by the proportion of
person-years attributable to the deceased blacks and the proportion attributable
to deceased whites, and then multiplying each total  separately by the corresponding
death rates for whites and blacks, and finally adding across age groups to get
the "composite" expected deaths.
     Because death certificates could not be located for all of the deceased, it
was assumed that those deaths for which no death certificates could be found had
the same distribution by cause as those for which death certificates were
available.  Thus the SMRs for each cause of death were corrected to reflect
the missing death certificates.  Using the SMR for- deaths from malignant
neoplasms of the colon in whites as an example, this correction was made in the
follow.ing manner:
                     (11/247 x 38) + 11  x  11  x 100 = 182
                             TT                ~
where:  247 is the number of deaths in the cohort with death certificates;
        38 is the number- of deaths in the cohort without death certificates;
        11 is the observed number of colon cancer deaths identified by death
           certificates;
        6.98 is the expected number- of white colon cancer deaths;
        100 is a constant used in calculating SMRs (by convention, SMRs are
            expressed as a factor of 100); and
        182 is the corrected white colon cancer SMR.

No significance tests of any of the SMRs were done by the author.  However, the
author called attention to the elevated SMR from malignant neoplasms of the colon
as possibly related to occupational exposure (6.98 white expected deaths, 6.77
black expected deaths, and composite SMR = 182).  Using an observed number- of
colon cancer cases corrected for the loss of death certificates, the Carcinogen
Assessment Group (CAG) found that the SMR for either whites or- blacks would be
statistically signficant (P < 0.05).

                                    9-16

-------
     The author points out that because the expected numbers of deaths were cal-
culated using U.S. rates, which include a higher socioeconomic  class than tKe
dry-cleaners in this study, and because higher socioeconomic class is associated
with a risk of colon cancer, the risk of colon cancer from exposure to tetra-
chloroethylene found in this study is probably underestimated.   Furthermore,
the expected number of colon cancer cases in this study was calculated using
mortality rates for neoplasms of the intestine, except rectum.   Although most
of the deaths expected using mortality rates for neoplasms of the intestine
would be deaths from malignant neoplasms of the colon, some deaths would be
from neoplasms of the small intestine.  Since all of the observed deaths were
from malignant neoplasms of the colon, the comparison of the observed to expected
colon cancer deaths in this study would also tend to underestimate the colon
cancer risk from exposure to tetrachloroethylene.
     It should also be noted that although it could be argued that the number
of observed colon cancer deaths in each of the four- union locals in this study
was small, an elevated colon cancer SMR did exist in each of the four locals.
Finally, it should be noted that the colon cancer- SMR appeared to demonstrate a
positive correlation with the length of the follow-up period.  This last finding
must be viewed with a great deal of caution, however, because of the author's
difficulty in defining length of exposure and hence length of follow-up.
     In addition to colon cancer, the SMRs for cancer of various other sites
were also elevated.  These included rectum, pancreas, respiratory system,
urinary organs, and "other and unspecified sites (major)."  None of these were
significant at the P < 0.05 level when tested using an observed number- that was
corrected for lost death certificates; however, the SMRs for- cancer of three of
these sites [respiratory system, urinary organs, and "other and unspecified
sites (major-)"] could be considered borderline significant (0.10 < P < 0.05).
     Perhaps the major weakness of this study with regard to evaluating
tetrachloroethylene as a carcinogen is that the history of solvent exposure
prior to 1960 was unknown for nearly half of the union member- shops.  Because
the majority of dry-cleaning establishments in the United States used petroleum
distillates as the primary cleaning agent prior to 1960, it is quite likely
that most of the shops in this study used petroleum distillates as the cleaning
solvent prior- to changing to tetrachloroethylene.

                                    9-17

-------
     Other  Important confounding variables were also not controlled.   For  example,
smoking is a major confounding variable to be considered when  evaluating a
potential  risk for respiratory or bladder cancer,  both of which  were  found in
excess in this study.  Socioeconomic status,  as has been discussed,  is a con-
founding variable for colon cancer.
     Another weakness of this study  is that 16 percent of the  study cohort
was lost to follow-up.  Currently, NIOSH is attempting to improve the percentage
of follow-up as well as to add to the length  of follow-up.  In addition, NIOSH
has identified other individuals who were exposed  to tetrachloroethylene for
at least one year prior to 1960, so  that the  size  of the cohort  has also been
increased.
     In summary, this study appears  suggestive that dry-cleaning workers exposed
to tetrachloroethylene are at an elevated risk of  colon cancer mortality.
Potential  exposure to petroleum distillates for- approximately  half of the  cohort,
however, limits any conclusions with regard to the carcinogenicity of tetra-
chloroethylene in humans.
9.2.2  Blair et al. (1979)
     Blair et al. (1979) reported, as the preliminary results  of a cohort  study
of 10,000 laundry and dry-cleaning workers, a proportionate mortality analysis
of 330 of the workers who had died during the period 1957-1977.   Deaths were
identified from the mortality records of two union locals in St. Louis, Missouri.
The distribution by cause of death among the 330 was compared  to that expected
based on the proportionate mortality experience of the United  States.  Only
279 of the 330 had worked exclusively in dry-cleaning establishments; however,
the authors did not report the proportionate mortality analyses  of these 279.
Furthermore, the dry-cleaning agent used by the dry-cleaners in  this  study is
unknown.  Among the 330 deaths, deaths from cancer of the lung,  cervix, and
skin contributed to the finding of a significant (P < 0.05) excess proportion
of deaths from cancer at all sites.   For lung cancer, there were 17  deaths
observed versus 10 expected (P < 0.05); for cervical cancer, 10  observed deaths
versus 4.8 expected (P < 0.05); and for skin cancer, 3 observed  deaths versus
0.7 expected (P < 0.05).  On the other hand,  a significant deficit of deaths
occurred in the cause category identified as "all  circulating  diseases," with
100 observed versus 125.9 expected (P < 0.005)—a  finding which  could account
for the excess proportion of cancers observed.
                                    9-18

-------
     Only limited conclusions can be drawn  from this  study as to the potential
carcinogenicity of tetrachloroethylene.   This is true for several  reasons.   First,
the study did not report the distribution of deaths for  dry-cleaners alone.
Second, the dry-cleaning agent these workers used is  not known.   Lastly, certain
possibly confounding variables were not considered in the analysis of the results.
These variables include smoking (with regard to the lung cancer  excess), socio-
economic status (with regard to the cervical cancer excess),  and sunlight
exposure (with regard to the skin cancer  excess).
9.2.3  Katz and Jowett (1981)
     Katz and Jowett (1981) analyzed the death certificate records of 671 white
female laundry and dry-cleaning workers who had died  during the  period 1963-
1977.  The records of dry-cleaning workers were not studied separately from
those of laundry workers, since both groups of workers shared the same occupa-
tional code.  Furthermore, it is not known whether those decedents who were
dry-cleaners used tetrachloroethylene as a dry-cleaning agent.  During the
1940s, petroleum derivatives were the predominant dry-cleaning agents used in
the United States.  A shift to the use of tetrachloroethylene began in the
late 1940s and gained momentum in the 1950s and 1960s.  By 1977, approximately
75% of the dry-cleaning plants in the United States were using tetrachloro-
ethylene.  However, in the period before 1960, petroleum distillates were
still the dominant solvents in use (Kaplan 1980).
     In this study, cause-specific proportionate mortality for the 671 deceased
laundry and dry-cleaning workers was compared to that for the deaths of all
other working females in Wisconsin during the same period, and to deaths of
females in "lower-wage occupations" in Wisconsin during the same period.
Significantly elevated proportionate mortality ratios (PMRs)  were found for
deaths from cancer of the genitals (unspecified) (P < 0.01) and for deaths from
cancer of the kidney (P < 0.05) when deaths among women of all occupations and
deaths among women of "lower-wage occupations" were used as the comparison
groups.  In summary, this study, although suggesting an association of employment
in the dry-cleaning industry with excess risks of certain types of cancer, cannot
be said to demonstrate conclusively that such an association exists, because of
the possible confounding influence of other dry-cleaning agents, and the fact
that no distinction was made between laundry and dry-cleaning workers.

                                    9-19

-------
9.2.4  L1n and Kessler (1981)
     Lin and Kessler (1981) did a case-control  study of 109 pancreatic cancer
cases diagnosed during the period 1972-1975 from 115 hospitals in five metro-
politan areas.  The control group was composed of subjects who were free from
cancer but who were similar to the study group in age (+_ 3 years), sex, race,
and marital status, and who were selected at random from among contemporaneous
admissions to the same hospital.  Cases and controls were asked about demographic
characteristics, residential history, toxic exposures, animal contacts,
smoking habits, diet, medical history, medications, and family history.  Males
were asked about sexual practices and urogenital conditions.  Females were
questioned on these topics and also on their- marital, obstetric, and gynecologic
histories.  Among other statistically significant associations, an association
was found between pancreatic cancer- and employment either as a dry-cleaner or
in a job involving exposure to gasoline.  It is not known, however, how many
individuals with pancreatic cancer were employed as dry-cleaners and how many
were employed in occupations involving exposure to gasoline.  Furthermore, the
dry-cleaners may have used a variety of dry-cleaning agents other than tetra-
chloroethylene.  Thus this study, although suggestive of an association between
employment as a dry-cleaner and an excess risk of pancreatic cancer, cannot be
said to demonstrate an association between pancreatic cancer and tetrachloro-
ethylene exposure.
9.2.5  Asa! (personal communication 1983)
     In two epidemiologic studies conducted by Dr. Nabih Asal of the University
of Oklahoma, an excess of cancer among dry-cleaners was found (personal communi-
cation 1983).  These studies are as yet unpublished.  One of the studies was
a kidney cancer- case-control study in which Asal found an association between
kidney cancer and the dry-cleaning occupation.  The second study was a propor-
tionate mortality study of dry-cleaning workers in Oklahoma.  Asal reported
that results from the latter study suggest an increased mortality from kidney
and lung cancer among the workers.  As in the Blair et al., Katz and Jowett,
and Lin and Kessler studies previously discussed, exposure in the studies by
Asal was not specific for- tetrachloroethylene.  Moreover, the conclusions of
these studies cannot objectively be evaluated until published descriptions of
them are available.
                                    9-20

-------
9.3  QUANTITATIVE ESTIMATION
     This quantitative section deals with estimation of the unit risk for
tetrachloroethylene as a potential  carcinogen in air  and water,  and with the
potency of tetrachloroethylene relative to other carcinogens that have been
evaluated by the CAG.  The unit risk for an air or  water  pollutant is defined
as the lifetime cancer risk to humans from daily exposure to a  concentration of
1 ug/m3 of the pollutant in .air by inhalation,  or to a concentration of 1 ug/1
in water  by ingestion.
     The unit risk estimate for tetrachloroethylene 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
practical reasons the correspondingly low levels of risk cannot be measured
directly either by animal experiments or- by epidemiologic study.  Low-dose
extrapolation must, therefore, be based on current understanding of the mechanisms
of carcinogenesis.  At the present time the dominant view of the carcinogenic
process involves the concept that most cancer-causing agents also cause
irreversible damage to DMA.  This position is based in part on  the fact that a
very large proportion of agents that cause cancer are also mutagenic.  There
is reason to expect that the quantal response that is characteristic of
mutagenesis is associated with a linear 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 high 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 is
also consistent with the relatively few epidemiologic studies of cancer responses
to specific agents that contain enough information to make the  evaluation
possible (e.g., radiation induced leukemia, breast and thyroid  cancer-, skin
cancer induced by arsenic in drinking water, liver cancer induced by aflatoxins
in the diet).  Some supporting evidence also exists from animal  experiments
(e.g., the initiation stage of the two-stage carcinogenesis model in rat liver
and mouse skin).
                                    9-21

-------
     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  extrapolation
to low levels of the  dose-response  relationship.   The risk estimates made with
such a model should be regarded as  conservative,  representing the most plausible
upper  limit for the risk; i.e., the true risk is  not  likely  to  be higher than
the estimate, but it  could be lower.
     For several reasons, the unit  risk estimate  based on animal  bioassays  is
only an approximate indication of the absolute risk  in populations exposed  to
known carcinogen concentrations.  First, there are important species differences
in uptake, metabolism, and organ distribution of  carcinogens, as  well as species
differences in target site susceptibility,  immunological responses,  hormone
function, dietary factors, and disease.  Second,  the  concept of equivalent
doses for humans compared to animals  on a mg/surface  area basis is virtually
without experimental  verification as  regards carcinogenic response.   Finally,
human populations are variable with respect to genetic constitution  and  diet,
living environment, activity patterns, and other  cultural factors.
     The unit risk estimate can give  a rough indication  of the  relative  potency
of a given agent as compared with other carcinogens.   Such estimates are, of
course, more reliable when the comparisons are based  on  studies in which the
test species, strain, sex, and routes of exposure are similar.
     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 representations  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.

                                    9-22

-------
9.3.1  Procedures for the Determination  of Unit Risk
9.3.1.1  Low-Dose Extrapolation Model--The mathematical  formulation  chosen  to
describe the linear nonthreshold dose-response relationship  at  low doses  is the
linearized multistage model.   This model  employs enough  arbitrary  constants to
be able to fit almost any monotonlcally  increasing dose-response data,  and  it
incorporates a procedure for  estimating  the largest possible linear-  slope (in
the 95% confidence limit sense) at low extrapolated doses that  is  consistent
with the data at all dose levels of the  experiment.
     Let P(d) represent the lifetime risk (probability)  of cancer  at dose d.
The multistage model has the form
                 P(d) = 1 - exp [-q0 + qj^d + qjd2 + ...+ qkdk)]
where
                          q,  _> 0, 1 = 0, 1, 2, ..., k
Equivalently,
                   Pt(d) = 1 - exp [qjd  + q2d2 + ...  + qkdk)]
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 qi, 1 = 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,
P^(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 qi > 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.

                                    9-23

-------
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 coef-
ficient, the resulting maximum value of the log-likelihood LI satisfies the equation
                             2 (L0 - l\) = 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 P^(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

                               2
                              X  =
                                       N,P,
is calculated where N^ is the number of animals in the i"1 dose group, X^ is
the number of animals in the ith dose group with a tumor- response, Pn- is the
probability of a response in the itn dose group estimated by fitting the
multistage model to the data, and h is the number of remaining groups.  The
fit is determined to be unacceptable whenever X2 is larger than the cumulative
99% point of the chi-square distribution with f degrees of freedom, where f
equals the number of dose groups minus the number of non-zero multistage
coefficients.
                                    9-24

-------
9.3.1.2  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 car-
cinogenic risk, q*, 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.
     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]_, A2»  ••., Am is defined as
                            (Aj x A2 x  ... x A,,,)1/"1.
     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.
9.3.1.3 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.
                                    9-25

-------
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
Then, the lifetime exposure is
                                     l_e x W2/3

9.3.1.3.1  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,
1n 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 « ppm x W    x r
or
                                   jn    a ppm.
                                   rW2/3

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
                                    9-26

-------
of drinking water requited 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:
                                            Fraction of body
                                           weight consumed as
                   Species        W        ffood      fwater
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 1n mg/W^/3 1s
                   rc   = PP") * F = ppm x f x W = ppm x f x W1/3
                 rW2/3
When exposure is given in terms of mg/kg/day = m/Wr = s, the conversion is
simply
= s x
                                           Wl/3.
9.3.1.3.2  Inhalation.  When exposure is via inhalation, the calculation 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 proportionally 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 compart-
ments.  After equilibrium is reached, the rate of absorption of these agents
is expected to be proportional to the metabolic rate, which in turn is propor-
tional to the rate of oxygen consumption, which in turn is a function of surface
area.
9.3.1.3.2.1  Case 1.  Agents that are 1n the form of particulate matter  or
virtually completely absorbed gases, such as sulfur dioxide, can reasonably be
                                    9-27

-------
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 I = 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, I = 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           VI          i = I/W
                        Man           70            0.29
                        Rats          0.35          0.64
                        Mice          0.03          1.3
Therefore, for- particulates or- completely absorbed gases, the equivalent
exposure in mg/W2/3 is
                              m  = Ivr  = iWvr = iv/l/3vr
                                "
     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.
9.3.1.3.2.2  Case 2.  The dose in mg/day of partially soluble vapors is pro-
portional to the 02 consumption, which in turn is proportional to W2'3 and is
also proportional to the solubility of the gas in body fluids, which can be
     *From "Recommendation of the International Commission on Radiological
Protection," page 9.  The average breathing rate is 107 cm3 per 8-hour workday
and 2 x 107 cm3 in 24 hours.
                                    9-28

-------
expressed as an absorption coefficient,  r,  for the gas.   Therefore,  expressing
the 62 consumption as 02 = k W^/3,  where k  is a constant independent of species,
it follows that
                               m =  k W2/3 x v x r
or
                                 d  =   m  = kw
                                     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
assumption is made, unless there is pharmacokinetic evidence to the contrary,
that absorption is equal by either-  exposure route.
9.3.1.4  Calculation of the Unit Risk from Animal Studies—The risk associated
with d mg/kg2/3/day is 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
corresponds to one unit of X, and substituting this value into the above
relationship.  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 lO'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 jtn polynomial coefficient
by an amount
                         (Wh/Wa)J/3  j = 1, 2, ..., k,
and to use mg/kg equivalents for the unit risk values.

                                    9-29

-------
9.3.1.4.1  Adjustments for  Less Than Llfespan Duration of 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  multiplying
a factor (L/Le) .   We assume that if the average dose d is continued,  the age-
specific rate of cancer will continue to increase as a constant function of the
background rate.  The age-specific rates for humans increase at least  by the
third power of the age and often by a considerably higher power, as demonstrated
by Doll (1971).  Thus, it is expected that the cumulative tumor rate would increase
by at least the third power of age.  Using this fact, it is assumed that the
slope 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* [or g(d)] would have been
increased by at least (L/Le) .
     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)].
9.3.1.5  Model for Estimation of Unit Risk Based on Human Data—If human epidemic-
logic studies and sufficiently valid exposure information are available for the
compound, they are always used in some way.  If they show a carcinogenic effect,
the data are analyzed to give an estimate of the linear dependence of  cancer
rates on lifetime average dose, which is equivalent to the factor B^.   If they
show no carcinogenic effect when positive animal evidence is available, then it
is assumed that a risk does exist, but it is smaller than could have been observed
in the epidemiologic study, and an upper-limit to the cancer incidence is calculated
assuming hypothetically that the true incidence is below the level of  detection
in the cohort studied, which is determined largely by the cohort size.  Whenever
possible, human data are used in preference to animal bioassay data.
     Very little information exists that can be utilized to extrapolate from high
exposure occupational studies to exposures at low environmental levels.  However,
if a number of simplifying assumptions are made, it is possible to construct a
crude dose-response model whose parameters can be estimated using vital statistics,
epidemiologic studies, and estimates of worker exposures.

                                    9-30

-------
     In human studies, the response is measured in terms of the relative risk of
the exposed cohort of individuals as compared with the control  group.   The
mathematical  model employed for the low-dose extrapolation assumes that for
low exposures the lifetime probability of death from cancer, PQ, may be
represented by the linear equation
                                  P0 = A + BHx
where A is the lifetime probability in the absence of the agent, and x is the
average lifetime exposure to environmental levels in units such as ppm.  The
factor  BH is the increased probability of cancer associated with each unit
increase of x, the agent in air.
     If it is assumed that R, the relative risk of cancer for exposed workers
as compared to the general population, is independent of length of exposure or
age at exposure, and depends only upon average lifetime exposure, it follows that
                           R = P  = A + BH (*i + *2)
                               FO   A + BH xf
or
                             RP0 = A + BH (X! + x2)

where xj = lifetime average daily exposure to the agent for the general population,
X2 = lifetime average daily exposure to the agent in the occupational  setting,
and PQ = lifetime probability of dying of cancer- with no or- negligible exposure.
     Substituting PQ = A + BH xj and rearranging gives
                               BH = PQ (R - D/X2
To use this model, estimates of R and x2 must be obtained from epidemiologic
studies.  The value PQ is derived by means of the life table methodology from
the age- and cause-specific death rates for- the general population found in
U.S. vital statistics tables.

9.3.2  Unit Risk Estimates
9.3.2.1  Data Available for Potency Calculation—The data on hepatocellular
carcinomas in B6C3F1 female mice, as reported in the NCI gavage study (1977a),
constitute the only information available for use in estimating the carcinogenic
potency of tetrachloroethylene.  Because of the high mortality and early tumor-
occurrence in the animals treated with tetrachloroethylene in the above study,
the data on times-to-death cited therein have been used in the present report

                                    9-31

-------
for potency calculations.  These data,  along with the results of the calculations,
are presented in Appendix B.  For comparison, the study's incidence of hepato-
cellular carcinomas (Table 9-4) is also utilized, employing four different
extrapolation models.
 TABLE 9-4.  INCIDENCE RATE OF HEPATOCELLULAR CARCINOMAS IN FEMALE B6C3F1 MICE
                                  (NCI 1977a)
Experimental
dose (mg/kg/day)
0
386
772
Human equivalent
dose (mg/kg/day)a
0
18.02
36.04
Tumor
incidenceb
0/20
19/48 (40%)
19/45 (42%)

 aThe human equivalent dose is calculated by d x (5/7) x (78/90) x (0.03/
  70)1/3 = 4.67 x 10~2 x d, where d is the experimental dose given to animals 5
  days per week for 78 weeks.  The lifespan of mice is assumed to be 90 weeks.
  The factor (0.03/70)1/3 is the cubic root of the ratio of body weights between
  animals and humans.  The use of this factor assumes that the doses in mg per
  body surface are equally effective between mice and humans.  For comparison,
  the potency is also calculated when mg/kg/day is assumed to be the equally
  effective dosage between mice and humans.
 ^Denominators are the numbers of animals that survived beyond 40 weeks.  The
  time to the first tumor is 41 weeks.
     There 1s no sound human epidemlologic data presently available that can be
used to calculate the cancer risk of tetrachloroethylene.  However, existing
human data can be used for- arriving at rough estimates of the magnitude of
human cancer  risk from exposure to tetrachloroethylene, and for comparison
with potency estimates made on the basis of animal data.  The colon cancer
mortality data presented in the NIOSH report (Kaplan 1980) have been used for
this purpose.  A detailed discussion of that study, focusing particularly on

                                    9-32

-------
its investigation of colon cancer,  was presented above in  the epidemiology
section of this chapter.
9.3.2.2  Assumption About Equally Potent Doses—To convert animal  doses to
human doses, it is assumed that mg/surface area/day is equivalent  among species.
This assumption is suggested by Mantel and Schneiderman (1975) and is supported
empirically in comparing  the toxicity of anticancer agents in various animal
species and man (Freireich et al. 1966, Rail  1977).  Under this assumption,
the animal dose d, in mg/kg/day, is multiplied by a factor (Wg/W^)1'^ to obtain
the human equivalent dose, where Wa and Wn are, respectively, the  animal and
human body weights.  Another approach to interspecies extrapolation that has
been used in risk assessment is direct extrapolation on the basis  of body
weight (i.e., mg/kg/day).  In the present report, the risks calculated on the
basis of body weight are  also provided.  The carcinogenic  potency  per- mg/kg/day
calculated on the basis of body surface area is always higher than when
calculated on the basis of body weight.  However, the fact that the calculated
potency expressed in a particular unit, (mg/kg/day)"^, is  higher- on the
basis of surface area does not necessarily imply that humans are assumed to be
more sensitive than animals.  This would be the case if doses in mg/kg/day
were in reality equally potent between humans and animals.
9.3.2.3  Choice of Low-Dose Extrapolation Models--In addition to the multistage
model currently used by the CAG for low-dose extrapolation, three mote models,
referred to as the probit, the Weibull, and the one-hit models, are employed
for- purposes of comparison (Appendix A).  These models cover almost the entire
spectrum of risk estimates that could be generated from existing mathematical
extrapolation models.  Generally statistical in character, these models are not
derived from biological arguments, except for the multistage model, which has
been used to support the somatic mutation hypothesis of carcinogenesis  (Armitage
and Doll 1954, Whittemore 1978, Whittemore and Keller 1978).  The main difference
among these models is the rate 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 than the multistage model because of the
difference of the deer-easing rate in the low-dose region.   However, it  should
be noted that one could always artificially give the multistage model the same
(or even greater) rate of decrease as the probit model by making some dose

                                    9-33

-------
transformation and/or by assuming that some of the parameters in the multistage
model are zero.  This, of course, is not reasonable without knowing, a priori,
what the carcinogenic process for the agent is.  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 experi-
mental exposure levels.  Furthermore, point estimates at low doses extrapolated
beyond 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 multistage 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 estimate 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-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.
9.3.2.4  Calculation of the Carcinogenic Potency of Tetrachloroethylene
9.3.2.4.1  Calculation on the Basis of Gavage Data.  As indicated previously,
data on hepatocellular carcinomas in female mice are analyzed by using either
the data of time-to-event or- the data of incidence rate.
     The time-to-event data and the calculations of potency are presented in
Appendix B.  In this calculation, it is assumed that all animals that died and
were found to have hepatocellular carcinomas died from these tumors.  Strictly
speaking, one knows only that the tumor occurred before the death of an animal,
and that a tumor was observed.  However, the carcinogenic potency estimates
based on other- assumptions do not differ greatly when the multistage model with
a time factor tk is used for low-dose extrapolation.  This conclusion is generally
true for life-threatening tumors, as observed by Krewski et al. (1983) using
simulated data.
                                    9-34

-------
     The data on hepatocellular carcinoma incidence rates are used to fit four
different extrapolation models.  The maximum likelihood estimates of the parameters
in each of the four models are presented in Table A-l in Appendix A.  Table 9-5
presents the upper-bound and point estimates of the risk at 0.01, 0.05, 0.1,
0.5, and 1 mg/kg/day.
     Both the probit and the Weibull models estimate much higher risk at these
dose levels than the multistage and the one-hit models.  The potency q* = 3.5 x
10~2 mg/kg/day, calculated by the multistage model with a time factor, will be
used for calculating the unit risk of tetrachloroethylene from drinking water.
This potency is obtained when the dose in mg/surface area/day is assumed to be
equally effective among species.  If this assumption is made, then q* = 2.6 x
10-3 mg/kg/day should be used for- this calculation.
9.3.2.4.2  Calculation on the Basis of Human Epidemiologic Data.  No sound
epidemiologic data are available that could be used to estimate the carcinogenic
potency of tetrachloroethylene.  However-, one study by Kaplan (1980) for- NIOSH
may be used to provide an estimation of the possible range of its potency.
This is the retrospective cohort mortality study on dry-cleaning workers exposed
to tetrachloroethylene.  The author of this report concluded:  "Based on the
study results, cancer of the colon, which accounted for 11 observed deaths,
appears to be the cause of death most likely to have resulted from occupational
exposure in our cohort."  For this reason, data on colon cancer- have been
selected by the CAG for the pur-pose of calculating the carcinogenic potency of
tetrachloroethylene.  In the present study, the standardized mortality ratio
(SMR) for colon cancers is estimated to be 182, which is statistically
significant at P < 0.05.  There are no data on the concentration of tetra-
chloroethylene to which the cohort workers were exposed.  Based on an industrial
hygiene study by NIOSH (Kaplan 1980), Campbell et al. (1980) estimated that
the average annual tetrachloroethylene exposure in dry-cleaning plants that
used the chemical ranged approximately from 1 ppm for "coin-op" workers to 7
ppm for machine operators.  The annual average exposure was calculated under
the assumption that a worker- worked 40 hours a week  for 50 weeks in a year.
     As an approximation, 0.8 ppm and 6 ppm will be used as lower limits and
upper limits of lifetime exposures by the cohort workers.  If exposure were
0.8 ppm, the carcinogenic potency would be:

                                    9-35

-------
          TABLE 9-5.  UPPER-BOUND (POINT) ESTIMATES OF RISK AT VARIOUS DOSE LEVELS,  BASED ON THE DATA OF
                               HEPATOCELLULAR CARCINOMAS IN FEMALE MICE (NCI 1977a)
Assumption of
human equivalent
dose3

Dose with
surface
corrections



Model s
Multistage with
time factor
Multistage

Problt
Wei bull
One-hit
0.01
3.5 x lO'4
(1.8 x 10-4)
2.6 x lO'4
(2.0 x 10-3)

1
(1.6 x 10-1)
1
(1.6 x 10-M
2.6 x lO'4
(2.0 x 10-3)
Risk at Dose Levels (mg/kg/day)
0.05 0.1 0.5
1.7 x 10-3
(8.8 x lO'4)
1.3 x 10-3
(1.0 x 10-3)

1
(2.0 x 10-1)
1
(2.0 x 10-1)
1.3 x 10-3
(1.0 x 10-3)
3.5 x 10-3
(1.8 x 10-3)
2.6 x 10-3
(2.0 x 10-3)

1
(2.2 x 10-1)
1
(2.1 x 10-M
2.6 x 10-3
(2.0 x 10-3)
1.7 x 10-2
(8.8 x 10-3)
1.3 x ID'2
(1.0 x 10-2)

1
(2.7 x 10-1)
1
(2.5 x 10-1)
1.3 x 10-2
(1.0 x 10-2)
1
3.5 x ID'2
(1.8 x ID'2)
2.6 x 10-2
(2.0 x 10-2)

1
(2.9 x 10-1)
(2.7 x 10-1)
2.6 x ID'2
(2.0 x 10-2)


Dose without
surface
correction



Multistage with
time factor-
Multistage
Problt
Welbull
One-hit
2.6 x 10-5
(1.3 x 10-5)
2.0 x 10-5
(1.5 x lO-5)
1
(1.1 x 10-1)
1
(1.4 x 10-1)
2.0 x 10-5
(1.5 x lO-5)
1.3 x 10-4
(6.6 x lO-5)
1.0 x lO'4
(7.5 x 10-5)
1
(1.4 x 10-1)
1
(1.6 x 10-1)
1.0 x lO'4
(7.5 x 10-5)
2.6 x 10-4
(1.3 x 10-4)
2.0 x 10-4
(1.5 x 10-3)
1
(1.5 x 10-1)
1
(1.8 x 10-1)
2.0 x 10-4
(1.5 x lO'4)
1.3 x 10-3
(6.6 x lO'4)
1.0 x 10-3
(7.5 x lO'4)
1
(1.9 x 10-1)
(2.1 x 10-1)
1.0 x ID'3
(7.5 x 10-4)
2.6 x 10-3
(1.3 x 10-3)
2.0 x lO-3
(1.5 x ID'3)
(2.1 x 10-1)
1
(2.3 x 10-1)
2.0 x 10-3
(1.5 x 10-3)
*Doses with surface correction assume that doses 1n mg/surface area/day  are equally potent in producing cancer  1n
 humans and animals.  Doses without surface correction assume  that doses 1n mg/kg/day are equivalent between
 humans and animals.

-------
                B  =  (1.82  -  1)  x  1.6  x  IP"2  =  1>6  x  10-2/ppm.
                 *•               0.8
     If exposure were 6 ppm, the carcinogenic  potency  would be:
                B  = (1.82 - 1)  x 1.6 x IP"2 = 2.2 x 10-3/ppm.
                 £               6

These estimates will be used to  evaluate  the reasonableness of  the tetrachloro-
ethylene potency estimations calculated on the basis of animal  data.
9.3.2.5  Pharmacokinetic Data  Relevant to Quantitative Risk Assessment—AIthough
a large amount of pharmacokinetic data is available  on experimental  animals, the
uses of these data  for risk  extrapolation are very limited because of the lack
of corresponding human data.  Based on the information in Table 9-7,  taken from
Pegg et al.  (1979)  and Schumann  et al.  (1980), it is suggested  that the rate of
tetrachloroethylene metabolism for- mice is much  higher- than that for rats when
animals are exposed via inhalation.  However,  the comparative metabolic rates
of mice and rats do not appear to be different when  the animals are exposed via
gavage.  The percentage of tetrachloroethylene that  was expired unchanged from
rats is consistent with that which was observed in humans when  tetrachloro-
ethylene was given by the inhalation route.  No Information is  available on the
human absorption rate of tetrachloroethylene via the oral route.  For the oral
route, if it is assumed that humans and rats are similar- with respect to absorption
rate, then the absorption rate for mice would be also similar to that of humans,
because, on the basis of available information,  mice and rats have a similar-
rate of absorption by this route of exposure.

       TABLE 9-6.  EXPIRED TETRACHLOROETHYLENE UNCHANGED AS PERCENTAGE OF
                            RECOVERED RADIOACTIVITY
                    (Pegg et al. 1979, Schumann et al. 1980)


                           Oral                          Inhalation
                  1 mg/kg       500 mg/kg         10 ppm          600 ppm
Rats
Mice
72%
NA
90%
83%
68%
12%
88%
NA

NA =OJot avail able.
                                    9-37

-------
  9.3.2.6   Risk Associated With 1 ug/liter of Tetrachloroethylene In Drinking Water—
  As  discussed above,  it  Is  reasonable to assume that both humans and mice have
  similar  absorption rates for tetrachloroethylene.  Therefore, the carcinogenic
—potency  q* = 3.5 x 10~2/(mg/kg/day), estimated from the mice gavage data, is used
  herein to calculate  the unit risk of tetrachloroethylene in drinking water.  Under
  the assumption  that  dally  water consumption for a 70-kg person is 2 liters, the
  risk from drinking water containing 1 ug/Hter of tetrachloroethylene is estimated
  as  follows:  P  = 3.5 x  10-2/(mg/kg/day) x 10-3 (mg/ug) x 2 (liter/day) v 70 kg
  = 1 x ID'6.
  9.3.2.7   Risk Associated With 1 ug/m3 of Tetrachloroethylene in Air--The
  carcinogenic potency q* =  3.5 x 10~2/(mg/kg/day) estimated from the data in the
  NCI mice gavage study (1977a) is used herein to calculate the carcinogenic
  risk from tetrachloroethylene by inhalation.  The relative absorption rate of
  tetrachlorothylene by inhalation and by gavage is not known.  As an approximation,
  it  is assumed that the  effective dose by inhalation in mice 1s one-sixth of
  that by  gavage. This assumption is based on the pharmacokinetic data presented
  in  Table 9-7 and on  the observation that absorption rates for tetrachloroethylene
  by  the oral route are similar- between mice and rats, since humans are assumed to
  be  similar to rats with respect to absorption rate.  To calculate the potency of
  tetrachloroethylene  1n  terms of ug/m3, it is assumed that the daily air intake
  for  a 70-kg person is 20 m3.  Thus, for 1 ug/m3 of tetrachloroethylene in air,
  the corresponding dose  in  mg/kg/day is
  (1/6) x  (1 ug/m3) x  (20 m3/day) x  (1/70 kg) x (10'3 mg/ug) = 4.76 x 10-5 mg/kg/day.
  The carcinogenic potency of tetrachloroethylene by inhalation is calculated as
  follows:
              q*  = (3.5 x 10-2) x (4.75 x 10-5) = 1.7 x 10-6/{ug/m3).

  The risk associated  with 1 ug/m3 of tetrachloroethylene in air is thus estimated
  as  1.7 x 10-6.  since 1 ppm of tetrachloroethylene is equivalent to 6,908
	ug/m3, the potency q* in terms of  ppm is estimated as

                     q* = 1.7 x 10-6 x 6,908 = 1.0 x 10-2/ppm.
                                       9-38

-------
 This  estimate  does  not  appear  to  be  inconsistent with  the  crude  estimates  that
 have  been  calculated  on the  basis of human  data.   These  range  from  2.2  x 10-3/ppm
 to 1.6  x 10'2/ppm.
 9.3.3  Comparison of  Potency With Other  Compounds
      One of the uses  of quantitative potency  estimates is  to compare  the
 relative potencies  of carcinogens.   Figure  9-5  is  a  histogram  representing the
 frequency  distribution  of potency indices for 53 suspect carcinogens  evaluated
 by the  CAG.  The actual data summarized  by  the  histogram are presented  in  Table
 9-7.  The  potency  index is derived from  q*  the 95%  upper  bound  of  the  linear
 component  in the multistage  model,  and is expressed  in terms of  (mMol/kg/day)'1.
 Where no human data were available,  animal  oral studies  were used  in  preference
 to animal  inhalation  studies,  since  oral studies have  constituted  the majority
 of animal  studies.
      Based on  available data concerning  hepatocellular carcinomas  in  female
 mice, the  potency  index for  tetrachloroethylene has  been calculated as  6 x 10^.
-This  figure is derived  by multiplying the slope q* = 3.5 x 10~2/(mg/kg/day) and
 the molecular  weight  of tetrachloroethylene,  165.8.  This  places the  potency
 index for  tetrachloroethylene  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 comparison  of  potencies if
 linearity  does not exist at the low-dose range, or if  comparison is to  be  made
 at the  high-dose range.  If the latter is the case,  then an index  other than
 the one calculated above may be more appropriate.

 9.4  SUMMARY AND CONCLUSIONS
 9.4.1  Qualitative
      Tetrachloroethylene induced  a statistically  significant  increase in the
 incidence  of hepatocellular  carcinomas in both  high- and low-dose  male  and female
 B6C3F1  mice when administered  by  gavage  for a period of  78 weeks.   The  tetra-
 chloroethylene used was over 99%  pure, but  was  estimated to contain epichlorohydrin
                                   9-39

-------
                                  4th
                                quartile
                                       •4-
                       3rd
                     quartile
                            -»-
 2nd
quartile
       -»•
  1st
quartile
                                     1 x 10*'    4x 10+i   2x10


                              NO


                                        co
           -2
i
0
                              246
                              Log of Potency Index
       r
       8
Figure 9-5.   Histogram representing the  frequency  distribution of
              the  potency indices of 53 suspect  carcinogens evaluated
              by the Carcinogen Assessment Group.
                             9-40

-------
TABLE 9-7.  RELATIVE CARCINOGENIC POTENCIES AMONG 53 CHEMICALS EVALUATED BY
     THE CARCINOGEN ASSESSMENT GROUP AS SUSPECT HUMAN CARCINOGENS1.2.3
Slope
Compounds (mg/kg/day)"l
Acrylonitrile
Aflatoxin B^
Aldrin
Allyl Chloride
Arsenic
B[a]P
Benzene
Benzidene
Beryllium
Cadmium
Carbon tetrachloride
Chlordane
Chlorinated ethanes
1,2-dichl oroethane
hexachl oroethane
1,1, 2 ,2- tetrachl oroethane
1 , 1 , 1-trichl oroethane
1 , 1 ,2-trichl oroethane
Chloroform
Chromium
DDT
Dichlorobenzidine
1,1-dichloroethylene
Dieldrin
0.24(W)
2924
11.4
1.19x10-2
15(H)
11.5
5.2xlO-2(W)
234(W)
4.86
6.65(W)
1.30x10-1
1.61
6.9x10-2
1.42x10-2
0.20
1.6x10-3
5.73x10-2
7x10-2
41
8.42
1.69
1.47x10-1(1)
30.4
Molecular-
weight
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
104
354.5
253.1
97
380.9
Potency
i ndex
1x10+1
9xlO+5
4x10+3
9x10-1
2x10+3
3x10+3
4x10°
4xlO+4
4x10+1
7x10+2
2x10+1
7x10+2
7x10°
3x10°
3x10+1
2x10-1
8x10°
8x10°
4x10+3
3x10+3
4x10+2
1x10+1
lxlO+4
Order of
magnitude
(Iog10
index)
+1
+6
+4
0
+3
+3
+1
+5
+2
+3
+1
+3
+1
0
+1
-1
+1
+1
+4
+3
+3
+1
+4
                                    9-41

-------
                            TABLE  9-7.   (continued)
Slope Molecular-
Compounds (nig/kg/day)'! weight
Dinitrotoluene
Diphenylhydrazine
Epichlorohydrin
B1s(2-chloroethyl )ether
B1s(chloromethyl ) ether
Ethyl ene di bromide (EDB)
Ethyl ene oxide
Heptachlor
Hexachlorobenzene
Hexachlorobutadiene
Hexachl or ocycl ohexane
technical grade
alpha Isomer
beta Isomer
gamma Isomer
Methyl ene chloride
Nickel
N1trosam1nes
Dimethyl ni tr osami ne
D1 ethyl nitr osami ne
Di butyl nitrosamine
N-ni tr osopyr rol 1 di ne
N-ni troso-N-ethyl urea
N-ni troso-N-methyl urea
N-ni troso-di phenyl ami ne
0.31
0.77
9.9xlO-3
1.14
9300(1)
8.51
0.63(1)
3.37
1.67
7.75x10-2
4.75
11.12
1.84
1.33
6.3xlO-4
1.15(W)
25.9(not by q*)
43.5(not by q*)
5.43
2.13
32.9
302.6
4.92xlO-3
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
Potency
index
6xlO+1
1x10+2
9X10'1
2xlO+2
lxlO+6
2x10+3
3X10+1
1x10+3
5x10+2
2xlO+1
1x10+3
3x10+3
5x10+2
4x10+2
5x10-2
7xlO+1
2x10+3
4xlO+3
9x10+2
2x10+2
4x10+3
3xlO+4
1x10°
Order of
magnitude
nog10
index)
+2
+2
0
+2
+6
+3
+1
+3
+3
+1
+3
+3
+3
+3
-1
+2
+3
+4
+3
+2
+4
+4
0
PCBs
4.34
324
1x10+3
+3
                                    9-42

-------
                         TABLE  9-7.   (continued)
Compounds
Phenols
2 ,4 ,6-tHchl orophenol
Tetrachlorodloxin
Tetr achl oroethyl ene
Toxaphene
Trichloroethylene
Vinyl chloride
Slope
(mg/kg/day)'1
1.99xlO-2
4.25x10$
3.5xlO-2
1.13
1.9x10-2
1.75x10-2(1)
Order- of
magnitude
Molecular Potency (logjo
weight index index)
197.4 4x10° +1
322 lxlO+8 +8
165.8 6x10° +1
414 5xlO+2 +3
131.4 2.5x100 0
62.5 IxlOO 0

Remarks:
1. Animal slopes are

95% upper-limit si

lopes based on the linearized multi-
2.
3.
stage model.  They are calculated based on animal  oral  studies, except
for those indicated by I  (animal  inhalation),  W (human  occupational
exposure), and H (human drinking water- exposure).   Human slopes are
point estimates based on  the linear  non-threshold  model.

The potency index is a rounded-off slope in (mMol/kg/day)'1 and is cal-
culated by multiplying the slopes 1n (mg/kg/day)'1 by molecular weight
of the compound.

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.
                                 9-43

-------
concentrations of less than 500 ppm.   It is unlikely that this response could be
attributed to the low concentration of epichlorohydrin.
     No carcinogenic effect was observed in lifetime studies of Osborne-Mendel
rats given tetrachloroethylene by gavage or in Sprague-Dawley rats exposed via
inhalation for 12 months followed by 12 months of observation.  However, because
of excessive dose-related mortality in the gavage experiment, and because of
the low dose level in the inhalation study, no conclusions can be made about
the carcinogenicity of tetrachloroethylene in rats.   A gavage study in four rat
strains, now in progress at the National Toxicology  Program (NTP), should
clarify the nature of the rat response.
     Intraperitoneal injection of tetrachloroethylene in strain A mice induced
no statistically significant incidence of pulmonary  adenomas.  In mouse skin
initiation experiments, tetrachloroethylene did not  initiate skin tumors, nor
did it induce skin tumors when applied alone three times per week for the
lifetime of the animals.  However, because of inherent limitations in these
assays, the negative results they showed do not detract from the positive
findings of the National Cancer Institute (NCI) mouse experiment.
     In a cohort study of dry-cleaning workers exposed to tetrachloroethylene,
1t was found that these workers were at an elevated  risk of colon cancer
mortality; however, it was also the case that as many as one half of these
workers may have been exposed to petroleum distillates earlier in their working
history.  Other studies either completed or currently in progress of dry-cleaners
or of cancer cases for which employment in the dry-cleaning occupation was
found to be a risk factor- did not attempt to Identify workers by tetrachloro-
ethylene exposure.
9.4.2  Quantitative
     Data on hepatocellular carcinomas in female mice from the NCI (1977a)
gavage study on tetrachloroethylene have been used herein to calculate the risk
associated with drinking water contaminated with tetrachloroethylene.  Although
the available pharmacokinetic data suggest that humans absorb less tetrachloro-
ethylene than mice via inhalation exposure, the same conclusion cannot be made
concerning exposure via the oral route.
     Under the assumption that absorption rates for-  humans and mice are similar,
the upper-bound cancer risk due to drinking water containing 1 ug/liter of
tetrachloroethylene is estimated to be 1 x 10~6.
                                    9-44

-------
     The upper-bound cancer  risk associated with 1  ug/m3 of tetrachloroethylene
in air is estimated to be 2  x 10~6.   This is calculated on the basis of data
obtained from the NCI gavage study in mice (1977a), under the assumption that
the effective dose by inhalation in  mice is one-sixth of that by gavage, and
the consequent assumption that tetrachloroethylene  in air is about six times
more potent in mice than in  humans.
     The resultant estimate  of the carcinogenic potency of tetrachloroethylene
in air is 1.0 x 10~2/ppm.  This does not appear to  be inconsistent with the
potency estimate based on the NIOSH  epidemiologic study (Kaplan 1980), which is
a crude risk estimate ranging from 2.2 x 10"3/ppm to 1.6 x 10~2/ppm.
9.4.3  Conclusions
     Tetrachloroethylene has been demonstrated to induce malignant tumors of
the liver in both male and female mice of the B6C3F1 strain.  This constitutes
limited evidence that tetrachloroethylene may be carcinogenic in humans.  It
should be recognized that there is a substantial body of opinion in the scientific
community to the effect that the mouse liver- overreacts to chlorinated organic
compounds in contrast to that of the rat, and that  the induction of liver-
cancer in the mouse represents only a promoting action for- spontaneous liver-
tumors which normally occur with substantial incidence.
     According to the criteria of the International Agency for- Research on
Cancer (IARC), the data supporting the carcinogenicity of tetrachloroethylene
must be classified as "limited."  Since existing human epidemiologic data for
tetrachloroethylene is inconclusive, its overall IARC ranking should be Group 3,
corresponding to the conservative scientific view that tetrachloroethylene is
probably carcinogenic in humans.
                                    9-45

-------
                                   REFERENCES


Armitage, P., and R. Doll.  1954.   The age distribution of cancer  and  a
     multistage theory of carcinogenesis.   Br.  J.  Cancer 8:1-12.

Blair, A., P. Drople, and D. Grarrman.  1979.  Causes of death among laundry
     and dry-cleaning workers.   Am. J. Public Health 69:508-511.

Campbell, G.L., D. Cohan, and D.W.  North.   1980.   The application  of decision
     analysis to toxic substances:   Proposed methodology and  two case  studies.
     EPA Contract No. 69-01-6054.   Prepared for the U.S. Environmental Protection
     Agency, Office of Toxic Substances.

Cox, C.B.  1972.  Regression model  and life tables.  J. Roy.  Stat  Soc. B
     34:187-220.

Crump, K.S., and W.W. Watson.  1979.  GLOBAL79:  A Fortran program to  extrapo-
     late dichotomous animal carcinogenicity data to low doses.  Natl. Inst.
     Environ. Health Science Contract No.  l-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
     carcinogenicity data.  Biometrics 33:437-451.

Daffer, P., K. Crump, and M. Master-man.  1980.   Asymptotic theory  for
     analyzing dose-response survival data with application to low-dose
     extrapolation problem.  Mathematical  Biosciences 50:207-230.

Doll, R.   1971.  Weibull distribution of cancer:   Implications for models  of
     carcinogenesis.  J. Roy. Stat. Soc. A 13:133-166.

Dripps, R.D., J.E. Eckenhoff, and L.D. Vandam.   1977.  Introduction to
     anesthesia:  The principles of safe practice.  5th ed.
     Philadelphia PA:  W.B. Saunders Company, 121-123.

Federation of American Societies for- Experimental  Biology (FASEB).  1974.  2nd
     ed. Vol. III.  Philip L. Altman and Dorothy S. Dittmen (eds.).  Bethesda,
     MD.  library of Congress No.  72-87738.

Freireich, E., E. Gehan, D. Rail,  L. Schmidt, and E. Skipper.  1966.   Quantitative
     comparison of toxicity of anticancer agents in mouse, rat, hamster,  dog,
     monkey, and man.  Cancer Chemotherapy Reports 50(4):219-244.

Howe, R.B., and K.S. Crump.  1982.   GLOBAL 82:   A computer- program to  extrapo-
     late quantal animal toxicity data to low doses.  OSHA Contract No.  41  USC252C3.

Howe, R.B., and K.S. Crump.  1982.   A computer program, WEIBULL82.  Science
     Research System, Inc., Ruston LA.

International Commission on Radiological Protection (ICRP).  1977.  Recommendation
     of the ICRP, Pub. No. 26, adopted Jan. 17, 1977.  Oxford, England:   Pergamon
     Press.
                                      9-46

-------
 Replacement Page 9-45  for  EPA-600/8-82-005B,  "Health Assessment Document
                       for  Tetrachloroethylene  (Perchloroethylene)
      The upper-bound cancer  risk  associated with  1 ug/m3 of  tetrachloroethylene
 in  air is estimated  to be  2  x  10~6.   This  is  calculated on the basis  of data
 obtained from the NCI  gavage study  in mice (1977a),  under the assumption .that
 the effective dose by  inhalation  in mice is one-sixth of that by gavage, and
 the consequent assumption  that tetrachloroethylene 1n air is about  six times
 more potent in mice  than in  humans.
      The resultant estimate  of the  carcinogenic potency of tetrachloroethylene
 in  air is 1.0 x 10'2/ppm.  This does  not appear to be inconsistent  with the
 potency estimate based on  the  NIOSH epidemiologic study (Kaplan 1980), which is
 a crude risk  estimate  ranging  from  2.2 x 10~3/ppm to 1.6 x 10'2/ppm.
 9.4.3  Conclusions
      Tetrachloroethylene has been demonstrated to induce malignant  tumors  of
 the liver in  both male and female mice of  the  B6C3F1 strain.  This  constitutes
 limited evidence that  tetrachloroethylene  may  be  carcinogenic in humans.   It
 should be recognized that  there is'a  substantial  body of opinion in the scientific
 community to the effect that the  mouse liver  overreacts to chlorinated organic
 compounds in  contrast  to that  of  the  rat,  and  that the induction of liver
 cancer in the mouse  represents only a promoting action for spontaneous liver
 tumors which  normally  occur  with  substantial  incidence.
      According to the  criteria of the International  Agency for Research on
 Cancer (IARC), the data supporting the carcinogeniclty of tetrachloroethylene
 must be classified as  "limited."  Since existing  human epidemiologic  data  for
 tetrachloroethylene  is inconclusive,  its overall  IARC ranking should  be Group 3,
w:*:*:*:*x*x*:-xtt^^
 meaning that  there is  inadequate  evidence  for  classifying tetrachloroethylene :*:*:*
                                    9-45                            1/25/84

-------
Innes,  J.R.M.,  B.M.  Ulland,  M.G.  Valeric,  L.  Petrocelli,  L.  Fishbein,  E.R. Hart,
     A.J. Pallota,  R.R.  Bates,  H.L.  Falk,  J.J.  Gart, M. Klein,  I.  Mitchell,  and
     J. Peters.  1969.   Bioassay  of  pesticides  and  industrial chemicals  for
     tumorigenicity in  mice:   A preliminary  note.   J.  Natl.  Cancer Inst.  42:1101-
     1114.

Kaplan, S.D.  1980.   Dry-cleaners workers  exposed to perch!oroethylene:   A retro-
     spective cohort mortality study.   NIOSH Contract  No. 210-77-0094.

Katz, R.M., and D.  Jowett.   1981.  Female  laundry and  dry-cleaning workers in
     Wisconsin:  A mortality analysis.   Am.  J.  Public  Health 71:305-307.

Kovar,  J., and D. Krewski.   1981.  RISK81:   A computer- program  for low-dose
     extrapolation of quantal  response  toxicity data.   Department  of Health
     and Welfare, Canada.

Krewski, D., K.S. Krump, J.  Farmer,  D.  Gaylor,  R. Howe, C. Portier, D. Salsburg,
     R. Sielhen, and J.  VanRyzin.  1983.   A  comparison of statistical  methods
     for low-dose extrapolation utilizing  time-to-tumor data.   Fundamental and
     Applied Toxicology 3:140-180.

Laskin, S., A.R. Sellakumar, M. Kuschner,  N. Nelson, S. La Mendola, G.M.  Rusch,
     G.V. Katz, N.C. Dulak,  and R.E. Albert.  1980. Inhalation carcinogenicity
     of epichlorohydrin in  non-inbred Sprague-Dawley rats.   J.  Natl. Cancer  Inst.
     65(4):751-755.

Lin, R.S., and I.I. Kessler.  1981.   A multifactorial  model  for pancreatic cancer
     in man.  J. Am. Med.  Assoc.  245:147-152.

Mantel, N., and M.A. Schneider-man.  1975.   Estimating  "safe" levels, a hazardous
     undertaking.  Cancer  Res. 35:1379-1386.

National Cancer Institute  (NCI).   1976.  Report on  carcinogenesis  bioassay on
     chloroform.  Natl.  Tech. Info.  Serv.  No. PB-264018.

National Cancer Institute  (NCI).   1977a.   Bioassay  of  tetrachloroethylene for
     possible carcinogenicity. DHEW Pub.  No. (NIH) 77-813.   U.S.  Department of
     Health, Education and  Welfare,  Public Health Service, National Institutes
     of Health.

National Cancer- Institute  (NCI).   1977b.   Bioassay  of  trichloroethylene  for
     possible carcinogenicity. DHEW Pub.  No. (NIH) 76-802.   U.S.  Department of
     Health, Education and  Welfare,  Public Health Service, National Institutes
     of Health.

National Cancer- Institute  (NCI).   1977c.   Bioassay  of  lindane for  possible
     carcinogenicity.  DHEW Pub.  No. (NIH) 77-814.   U.S.  Department of Health,
     Education and Welfare,  Public Health  Service,  National  Institutes of Health.

National Toxicology Program (NTP).  1983.   Bioassay of tetrachl oroethylene  in
     female B6C3F1 mice.  Draft (not peer-reviewed).
                                      9-47

-------
Pegg, D.G.,  J.A.  Zempel,  W. H.  Braun,  and P.G.  Watanabe.   1979.   Disposition  of
     tetrachloro(14C)ethylene following oral  and inhalation exposure  in  rats.
     Toxicol. Appl.  Pharmacol.  51:465-474.

Rail, D.P.  1977.   Species differences  in carcinogenesis testing.   In:   Origin of
     Human Cancers,  Book C, pp.  1383-1390.

Rampy, L.W., J.F.  Quast, M.F. Balmer, B.K.J.  Leong,  and P.J. Gehring.   1978.
     Results of a  long-term inhalation  toxicity study on rats of  a perchloro-
     ethylene (tetrachloroethylene) formulation.  Toxicology Research  Laboratory,
     Dow Chemical  Company, Midland, Michigan.

Schumann, A.M., J.F. Quast, and P.G. Watanabe.   1980.  The pharmacokinetics and
     macromolecular interactions of perchloroethylene in mice and rats as
     related to oncogenicity.  Toxicol. Appl. Pharmacol. 55:207-219.

Theiss, J.C., G.D.  Stoner, M.D.  Shimkin, and  E.K. Weisburger.  1977.   Tests for
     cardnogenicity of organic  contaminants  of United States drinking waters  by
     pulmonary tumor response in strain A mice.   Cancer- Res. 37:2717-2720.

Van Duuren, B.L.,  M. Goldschmidt, G. Loewengart, C.  Smith, S. Melchione, I. Seidman,
     and D. Roth.   1979.  Carcinogenicity of  halogenated olefinic and  aliphatic
     hydrocarbons  in mice.  J.  Natl. Cancer Inst. 63(6):1433-1439.

Whittemore, A.  1978.  Quantitative theories  of carcinogenesis.   In:   Advances in
     cancer research,  Vol. 27.   New York, NY:   Academic Press, 55-58.

Whittemore, A., and B. Keller.   1978.  Quantitative  theory of carcinogenesis.
     Society for Industrial and Applied Mathematics  Review 20:1-30.
                                      9-48

-------
                                   APPENDIX A

                COMPARISON AMONG DIFFERENT EXTRAPOLATION MODELS

     Four models used for low-dose extrapolation,  assuming the independent
background, are:
Multistage:         P(d) = 1 - exp [-(q1d+ ... + qkdk)]
where qj are non-negative parameters.

                                  A + B ln(d)
Probit:                  p(d) =  /    f(x) dx
                                    oo

where f(.) is the standard normal probability density function

Weibull:                 P(d) = 1 - exp [-

where b and k are non-negative parameters

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.
                                    A-l

-------
     TABLE  A-l.   MAXIMUM LIKELIHOOD  ESTIMATE OF THE PARAMETERS FOR EACH OF THE FOUR EXTRAPOLATION MODELS BASED ON
                             HEPATOCELLULAR CARCINOMAS  IN FEMALE MICE  (doses in mg/kg/day)
Basis of  interspecies
   extrapolation
   Multistage
    Probit
Weibull
   One-hit
Body surface area
q1 = 2.00 x 10

q2 = 0
                                            -2
A = - 0.55
                                                       B = 9.80 x 10
                                                                     -2
b = 0.33

k = 0.12
b = 2.00 x 10
                                                                                -2
Body weights
q1 = 1.5 x 10

q2 = 0
                                           -3
A = -0.80

B = 9.80 x 10
                                                                     -2
b = 0.25
                                                                             k = 0.12
b = 1.51 x 10
                                                                                -3

-------
                                    APPENDIX B

                  TIME-TO-EVENT DATA AND CALCULATIONS OF POTENCY
   TABLE B-l.   TIME-TO-DEATH IN WEEKS (H INDICATES THAT AN ANIMAL DIED OR WAS
            SACRIFICED AND WAS FOUND TO HAVE A HEPATOCELLULAR CARCINOMA.   ALL
                     ANIMALS WERE SACRIFICED AT 91 WEEKS).
Control  group (vehicle):  20 animals

   90, 90, 90, 90, 90, 90, 90, 90, 90, 90, 90, 90, 90,  90,  90, 90, 90, 90
   90, 90, 90, 90, 90, 90, 69, 90, 90, 90, 90, 90, 90,  90,  90, 90, 90, 90
   90, 90, 90, 90, 90, 90, 90, 90, 90, 90, 90, 90, 90,  90,  68.
Low-dose group:  50 animals
91(H),
58,
66,
57,
65(H),
50,
91(H),
49(H),
62,
57,
66,
50(H),
63,
50,
91(H),
45,
61(H),
47,
66(H),
91(H),
62,
48,
87(H),
44
61,
23,
64,
91(
62,
48,
52,

                                                  60,        59
                                                  91(H),      83
                                                  62,        62
                                                  68,        67
                                                  55,        21
                                                  52,
                                                  41(H),      91(H)
                                                  51,        51(H)
High-dose group:  50 animals
91(H),
49,
50(H),
87(H),
50,
56,
56(H),
48,
91(H),
46,
49,
56,
49,
56,
53,
24,
91(H),
46,
46,
55(H),
46,
54(H).
53,
26,
63,
91(H),
45,
55(H),
91,
50(H),
52(H),
17
53,
91(H),
23,
52,
58(H),
49,
50(H),

50,
72(H),
22,
50,
56,
49,
49,

50(H)
50
91(H)
50
56(H)



The dose-response function at a time t is assumed to have the form


                         P(d/t) = 1 - exp [-q(d) x f(t)]

where

                              q(d) = q0 + q^

and

                                   f(t) = tk.
                                       B-l

-------
The lifetime cancer risk 1s calculated when t 1s set equal  to  90  weeks.   When
the dose unit 1s mg/kg/day, the maximum likelihood estimates of the  parameters
are qQ = 0, qj = 1.6 x 10'9, q2 = 2.3 x NT11, and k = 3.6.  At t =  90  weeks, the
95% upper-bound estimate of the linear component of the dose-response model  1s
q* = 3.5 x 10~2 mg/kg/day.  These estimates are calculated  using  the computer
program WEIBULL82, developed by Howe and Crump (1982).  In  the calculation,  only
the tumors that were observed before week 91 are considered to be the causes of
death for those animals in which the tumors were observed.
                                    B-2

-------