United States
Environmental Protection
Agency
Office of Health and
Environmental Assessment
Washington DC 20460
EPA/600/8-82/004F
February T985
Final Report
Research and Development
Health Assessment   Final
Document for         Report
Dichloromethane
(Methylene Chloride)

-------

-------
                             EPA/600/8-82/004F
                                   February 1985
                                     Final Report
Health Assessment Document
      For Dichloromethane
      (Methylene Chloride)
            Final Report
       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

-------
                                    NOTICE
     This document has been reviewed in accordance with the U.S.  Environmental
Protection Agency's peer  and administrative review policies and approved for
presentation and publication.   Mention of trade names or commercial products
does not constitute endorsement or recommendation for use.

-------
                                     PREFACE

      The Office of Health and Environmental Assessment, in consultion with an
 Agency work group, 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;  however,  at the request
 of the Agency 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 are  placed  in perspective  with observed environmental
 levels.
     Any information regarding sources, emissions,  ambient air  concentrations,
 and  public exposure has been included only to give the reader  a preliminary
 indication  of  the  potential  presence  of this  substance in the  ambient air.
 While  the  available information  is presented as accurately as possible,  it is
 acknowledged  to  be limited  and  dependent  in  some  instances  on assumption
 rather than specific data.   This information is not intended, nor should  it be
 used, to support any conclusions regarding  risks to public  health.
     If  a  review of the health information indicates that the  Agency should
 consider regulatory action for  this substance, a considerable effort will be
 undertaken to obtain appropriate information regarding  sources,  emissions, and
 ambient air concentrations.  Such data will provide additional information for
 drawing regulatory conclusions regarding the extent and significance of public
 exposure to this substance.
     In view of the pending release of a National Toxicology Program report on
 long-term animal  studies with dichloromethane, the carcinogenicity conclusions
of this  document are considered  interim and will be updated when the  report
can be evaluated.

-------
                               TABLE OF CONTENTS
LIST OF TABLES	        .
LIST OF FIGURES 	•••••	-....-•     vm

1.   SUMMARY AND CONCLUSIONS 	     1"1

2.   INTRODUCTION 	     2"1

3.   DICHLOROMETHANE:  BACKGROUND INFORMATION	     3-1
    3.1  PHYSICAL AND CHEMICAL PROPERTIES 	     f 1
    3.2  ENVIRONMENTAL FATE AND TRANSPORT 	•     ;f J
         3.2.1  Production	.. — .-.-.".	     * *
         3.2.2  Use	     *_ฃ
         3.2.3  Emissions 	     , ^
         3.2.4  Persistence of DCM	     * ฐ
         3.2.5  Products of DCM  	•••     * If
    3.3  LEVELS OF  EXPOSURE 	•	~	    6~\L
         3.3.1  Analytical Methodology  	    * ฑฑ
         3.3.2  Sampling of Ambient Air and Water	    *ฑ'
    3.4  ECOLOGICAL EFFECTS 	    6  "
         3.4.2  Effects on Plants  	    6~^
         3.4.3  Bioconcentration Potential  	    | ^ฐ
    3.5  CRITERIA,  REGULATIONS,  AND STANDARDS  	    f^/
    3.6  REFERENCES 	    J "

 4.  METABOLISM  AND  PHARMACOKINETICS OF  DICHLOROMETHANE	    4-1
    4.1 ABSORPTION,  DISTRIBUTION,  AND  PULMONARY ELIMINATION 	    4-1
         4.1.1  Oral  Absorpti on  	    J_|
         4.1.2  Dermal  Absorption —	    ฃ *
         4.1.3   Pulmonary  Uptake 	    T ^
         4.1.4 Tissue Distribution 	     T f-^
         4.1.5   Pulmonary  Elimination 	     J~"
     4.2 METABOLISM OF DCM	i"'ป"".*	     1-ia
         4.2.1  Evidence for Metabolism to Carbon Monoxide 	     ^ J-ฐ
         4'.2.2  Evidence for Dose-Dependent Metabolism of DCM:
                 Michaelis-Menten Kinetics 	     J~ฃJ
          4.2.3  Enzyme Pathways of DCM Metabol i sm 	     *[-ซ
     4.3  DCM-INDUCED CHANGES IN HEPATIC ENZYMES  	     4-40
     4.4  COVALENT BINDING TO CELLULAR MACROMOLECULES  	     4-41
     4.5  KINETICS OF CARBOXYHEMOGLOBIN FORMATION  	     4-4.3
          4.5.1  Studies in Humans	     **'
          4.5.2  Studies in Animals 	••••	     ^ ^
          453  Comparison of Kinetics of Humans  and  Rats  	     •*-•ซ
     4 6  MEASURES OF EXPOSURE AND  BODY BURDEN IN  HUMANS  	     4-54
     4.7  SUMMARY AND CONCLUSIONS 	     T .jฃ
     4.8  REFERENCES  	
                                        iv

-------
                         TABLE OF CONTENTS (continued)
5.   HEALTH EFFECTS OF DICHLOROMETHANE
    5.1
    5.2
    5.3
    5.4
                                                          	    5-1
                                                          	    5-1
                                                          	    5-1
                                                          	    5-5
                                                          	    5-7
                                                          	    5-7
                                                          	    5-17
                                                          	    5-20

                                                          	    5-20
5.3.2  Mutagenicity	    5-24
5.3.3  Evaluation of the Carcinogenicity of DCM	    5-45
REFERENCES	    5-112
HUMAN HEALTH EFFECTS 	
5.1.1  Acute Exposures 	
5.1.2  Chronic Effects 	
EFFECTS ON LABORATORY ANIMALS ....
5.2.1  Acute Effects	
5.2.2  Chronic Effects 	
TERATOGENICITY, MUTAGENICITY, AND
5.3.1  Teratogenicity, Embryotoxicity, and Reproductive
       Effects 	
         APPENDIX
                                                                  A-l

-------
                                LIST OF TABLES
Table

3-1
3-2
3-3
3-4
3-5
3-6
3-7

4-1

4-2

4-3

4-4

4-5

4-6

4-7

4-8

4-9

4-10

4-11

4-12
4-13

4-14

4-15
5-1

 5-2
 5-3
 5-4
 5-5
 5-6
 5-7
 5-8
 5-9
 5-10
Synonyms and identifiers for dichloromethane 	
Selected properties of dichloromethane 	
Producers of dichloromethane 	
Consumption of dichloromethane 	
Reaction rate data for OH + CH2C12 	
Ambient air levels of dichloromethane 	
Effects of dichloromethane on freshwater species in acute
  tests	
Pulmonary absorption of DCM by human subjects (sedentary
  conditions)	
Effect of exercise on physiological parameters for volunteers
  exposed to DCM	•	•	
Body burdens of rats after inhalation exposure to 14C-DCM for
  6 hours 	
DCM concentrations in rat whole blood and plasma at apparent
  steady-state conditions after a 6-hour inhalation exposure ...
Tissue concentrations of DCM in rats exposed to 200 ppm DCM for
  4 days for 6 hours daily 	
Distribution of 14C-activity in rat tissue 48 hours after
  6-hour inhalation exposure or oral dosage of 14C-DCM  .	
Comparison of postexposure pulmonary elimination half-times
  of DCM for humans and rats	
Blood carboxyhemoglobin concentrations of rats exposed
  to CO and DCM by inhalation  	
Fate and disposition of 14C-DCM in rats (412-430 mg/kg),
  injected intraperitoneally	
Fate of 14C-DCM in rats after  a single 6-hour inhalation
   exposure  	
 Body  burdens  and metabolized  14C-DCM  in  rats after  inhalation
   exposure  to 14C-DCM	
 Fate  of DCM in rats  48 hours  after  single  oral  doses  	
 In vitro covalent  binding  of  14C-DCM  to  microsomal  protein
   and lipid 	
 Comparative covalent binding  of DCM,  carbon tetrachloride, and
   trichloroethylene  to lipid  and protein in rat hepatocytes  ...
 Blood COHb  and Hb  concentrations in rats exposed to DCM  	
 COHb  concentrations  in nonsmokers exposed to DCM at
   250 ppm (869 mg/m3)  for  5 days 	
 Acute lethal  toxicity  of DCM  	
 Summary of cardiotoxic action of 5% dichloromethane 	
 Mutagenicity  testing of DCM in bacteria  	
 Gene  mutations and mitotic recombination in yeast 	
 Gene  mutations in  multicellular eukaryotes in  vivo  	
 Gene  mutati ons i n  mammali an eel 1s i n  culture  	
 Tests for chromosomal  aberrations 	
 Tests for si ster-chromatid exchange 	
 Analytical  analysis of dichloromethane 	
3-2
3-3
3-5
3-6
3-8
3-13

3-25

4-5

4-7

4-9

4-10

4-12

4-13

4-16

4-21

4-26

4-30

4-31
4-31

4-42

4-42
4-53

5-6
5-8
5-11
5-25
5-33
 5-34
 5-39
 5-41
 5-43
 5-47
                                       VI

-------
                           LIST OF TABLES  (continued)
 Table
 5-11   Cumulative percent mortality of  rats 2-year dichloromethane
         inhalation study	    5-49
 5-12   Summary of total tumor data for  rats administered
         dichloromethane for 2 years by inhalation	    5-53
 5-13   Time-to-tumor, palpable mass, and histopathology data for
         salivary gland region sarcomas in individual male rats
         exposed to dichloromethane by  inhalation for 2 years  	    5-54
 5-14   Summary_of salivary gland region sarcoma incidence in male
         rats in a 2-year inhalation study with dichloromethane ....	    5-56
 5-15   Cumulative percent mortality of  hamsters, 2-year
         dichloromethane inhalation study	    5-57
 5-16   Summary of total tumor data for  hamsters administered
         dichloromethane by inhalation  for 2 years	    5-51
 5-17   Monthly mortality data for male  rats in a 2-year dichloro-
         methane inhalation toxicity and oncogem'city study	     5-66
 5-18   Monthly mortality data for female rats in a 2-year dichloro-     ,_
         methane inhalation toxicity and oncogenicity study 	    5-67
 5-19   Non-neoplastic liver lesions in  male rats ...		    5-68
 5-20   Non-neoplastic liver lesions in  female rats	    5-69
 5-21   Summary of mammary gland tumors  in female rats 	    5-70
 5-22   Group assignment of Fischer 344  rats administered
         dichloromethane in deionized drinking water for 24 months 	    5-72
 5-23   Mean daily consumption of dichloromethane in a 24-month
         chronic toxicity and oncogenicity study in Fischer 344 rats ...    5-72
 5-24   Incidence of hepatocellular tumors in male and female Fischer
         344 rats administered dichloromethane in deionized
         drinking water for 104 weeks	    5-74
 5-25   Historical control  data of liver neoplasia in female
         Fischer 344 rats at Hazelton Laboratories America,  Inc	    5-75
 5-26  Group assignment of B6C3F1 mice administered dichloromethane
         in deionized drinking water for 24 months	    5-76
 5-27  Mean daily consumption of dichloromethane in a 24-month
        chronic toxicity and oncogenicity study in B6C3F1 mice	    5-76
 5-28  Occurrence of convulsions in male and female B6C3F1 mice 	    5-78
 5-29  Number of male mice with Harderian gland neoplasms  	    5-78
 5-30   Incidence of proliferative hepatocellular lesions in male
        and female B6C3F1 mice administered dichloromethane in
        deionized drinking water for 104 weeks	    5-79
 5-31  Pulmonary tumor bioassay in Strain A mice	    5-80
5-32  Chemical  compositions of dichloromethane samples  (ppm)	    5-83
 5-33  Observed and expected deaths,  1964-1980,  1964  hourly male
        dichloromethane cohorts from Kodak 	    5-85
5-34  Malignant neoplasms,  observed  and expected deaths,  1964-1980,
        1964 hourly male  dichloromethane cohorts  from Kodak	    5-86
5-35  Selected dichloromethane chronic animal  studies 	    5-100
5-36  Incidence rates of  salivary gland region  sarcomas in  male
        Sprague-Dawley rats in the Dow Chemical  Company (1980)
        inhalation  study  	    5-101

-------
                          LIST OF TABLES (continued)
Table

5-37
A-l
Relative carcinogenic potencies among 53 chemicals evaluated
  by the Carcinogen Assessment Group as suspect human
  carci nogens	
Estimates of low-dose risk to humans based on salivary gland
  region sarcomas in male rats in the Dow Chemical Company
  (1980) inhalation study derived from four different models
Page



5-105


A-3
                                     vm

-------
                                 LIST OF FIGURES
 Figure
Page
 3-1   The effect of oxygen doping of the carrier gas on the ECD
         response to several halogenated methanes at a detector
         temperature of 300ฐC 	     3-21
/4-1   Inspired and expired air concentrations during a 2-hour,
         100-ppm inhalation exposure to DCM for a 70-kg man, and
         the kinetics of the subsequent pulmonary excretion 	     4-4
 4-2   Plasma levels of DCM in rats during and after DCM exposure
         for 6 hours 	     4-8
 4-3   DCM venous blood levels in rats immediately after a single
         6-hour inhalation exposure to various concentrations of DCM ...     4-10
 4-4   Pulmonary elimination of 14C-DCM from rats following oral
         administration of a single dose of 1 or 50 mg/kg (squares) ....     4-17
 4-5   Carboxyhemoglobin concentrations in male nonsmokers exposed to
         increasing concentrations of DCM for 1, 3, or 5 h day
         for 5 days 	     4-22
 4-6   Carboxyhemoglobin concentrations in rats after inhalation
         exposure to increasing concentrations of DCM for single
         exposures of 3 hours 	     4-23
 4-7   Blood CO content of rats after 3-hour inhalation exposure with
         1000 ppm dichloromethane, dibromomethane,  and diiodomethane,
         respecti vely 	     4-24
 4-8   Rates of production of CO from DCM given to  rats	4-28
 4-9   Enzyme pathways  of the hepatic biotransformation of
         di halomethanes 	     4-34
 4-10  Proposed reaction mechanisms for the metabolism of
         dihalomethanes to CO,  formaldehyde,  formic acid,  and
         inorganic halide 	     4-35
 4-11  Blood COHb  level  in men  during an 8-hour exposure for 5 conse-
         cutive days to 500 ppm and 100 ppm DCM 	     4-44
 4-12  Blood COHb  concentrations in rats during and after a 6-hour
         inhalation exposure to DCM 	     4-43
 5-1   Histogram representing the frequency distribution of the
         potency indices of 53  suspect carcinogens  evaluated by the
         Carcinogen Assessment  Group  	     5-104

-------
                             AUTHORS AND REVIEWERS
The principal authors of this document are:

Steven Bayard, Carcinogen Assessment Group, U.S. Environmental Protection
     Agency, Washington, DC

David L. Bayliss, Carcinogen Assessment Group, U.S. Environmental Protection
     Agency, Washington, DC

Vernon Benignus, Health Effects Research Laboratory, U.S. Environmental
     Protection Agency, Research Triangle Park, NC  27711

I.W.F. Davidson, Department of Physiology and Pharmacology, Bowman Gray
     School of Medicine, Winston-Sal em, NC

John R. Fowle III, Reproductive Effects Assessment Group, U.S. Environmental
     Protection Agency, Washington, DC

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

Bernard H. Haberman, Carcinogen Assessment Group,  U.S. Environmental
     Protection Agency, Washington, DC

Dennis  Kotchmar, Environmental Criteria and Assessment Office, U.S. Environ-
     mental Protection  Agency, Research Triangle Park, NC  27711

Jean C. Parker, Carcinogen Assessment Group,  U.S.  Environmental  Protection
     Agency, Washington, DC

Dharm V.  Singh, Carcinogen Assessment Group,  U.S.  Environmental  Protection
     Agency, Washington, DC

The following  individuals reviewed  earlier drafts  of  this  document  and submitted
valuable  comments.

All Members  of the
Interagency  Regulatory  Liaison Group
Subcommittee  on Organic Solvents

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

Dr. Mildred Christian
Argus  Laboratories
Perkasie, PA  18944

-------
 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. Lawrence Fishbein
 National Center for lexicological Research
 Jefferson, AR  72079

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

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

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

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

Dr. James Withey
Food Directorate
Bureau of Food Chemistry
Ottawa, Canada
                                       XI

-------
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.
Herman J. Gibb, B.S., M.P.H.
Bernard H. Haberman,  D.V.M., M.S.
Charalingayya B. Hiremath, Ph.D.
Robert E. McGaughy,  Ph.D.
Dharm V.  Singh, D.V.M., Ph.D.
Todd W. Thorslund, Sc.D.

Participating Members of the Reproductive Effects Assessment Group

Peter E.  Voytek, Ph.D. (Chairman)
John R. Fowle III, Ph.D.
Carol N.  Sakai, Ph.D.
Vicki Vaughan-Dellarco, Ph.D.
K.S. Lavappa, Ph.D.
Sheila Rosenthal, Ph.D.
Casey Jason, M.D.
Daniel S. Strauss, Ph.D., Consultant
Gary M. Williams, M.D., Consultant

Members of the Agency Work Group on Solvents
Elizabeth L. Anderson
Charles H. Ris
Jean C. Parker
Mark M. 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
Offide 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

-------
                           SCIENCE ADVISORY BOARD
                       ENVIRONMENTAL HEALTH COMMITTEE
Chairman

Dr.  Herschel Griffin
Associate Dean
College of Human Services
San Diego State University
San Diego, CA  92182

Members

Dr.  Herman Collier, President
Moravian College
Bethlehem, PA  18018

Dr.  Morton Corn
Professor and Director
Division of Environmental Health Engineering
School of Hygiene and Public Health
The Johns Hopkins University
615 North Wolfe Street
Baltimore, MD  21205

Dr.  John Doull
Professor of Pharmacology and Toxicology
University of Kansas Medical Center
Kansas City, KA  66103

Dr.  Jack Hackney, Chief
Environmental Health Laboratories
Professor of Medicine
Rancho Los Amigos Hospital
Campus of the University of Southern California
7601 Imperial Highway
Downey, CA  90242

Dr.  Marvin Kushchner, Dean
School of Medicine
Health Science Center
Level 4
State University of New York
Stony Brook, NY  11794

Dr.  Daniel Menzel
Director and Professor
Pharmacology and Medicine Director
Cancer Toxicology and Chemical Carcinogenesis Program
Duke University Medical Center
Durham, NC  27710
                                      xm

-------
Dr.  D.  Warner North, Principal
Decision Focus, Inc.
Los Altos Office Center
Suite 200
4934 El Camino Real
Los Altos, CA  94022

Dr.  William Schull
Director and Professor of Population Genetics
Center for Demographic and Population Genetics
School  of Public Health
University of Texas Health Science Center at Houston
Houston, TX  77030

Dr.  Michael Symons, Professor
Department of Biostatisties
School  of Public Health
University of North Carolina
Chapel  Hill, NC  27711

Consultants

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

Dr. Edward Ferrand
Assistant Commissioner for Science
  and Technology
New York City Department of Environmental
  Protection
51 Astor Place
New York, NY  10003

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

Dr. Bernard Weiss,  Professor
Division  of Toxicology
P.O. Box  RBB
University  of  Rochester
School  of Medicine
Rochester,  NY   14642
                                       xiv

-------
Executive Secretary

Mr. Ernst Linde, Scientist Administrator
Science Advisory Board, A-101
U.S. Environmental Protection Agency
Washington, DC  20460
                                       xv

-------
                                   ABSTRACT

     Dichloromethane  (methylene  chloride, DCM)  is a  volatile  chlorinated
hydrocarbon  used  extensively  in  commercial  and  industrial  solvent
applications. The background  atmospheric  concentration is about 35 ppt.   In
urban areas  levels  can  be one to two orders of magnitude higher.   In surface
water and  drinking  water,  measured concentrations generally have been in the
low parts per billion range.   Available data suggest that DCM is biodegradable
and has low toxicity to aquatic organisms.
     The weight  of evidence  from the available  literature  indicates that
adverse toxicologic (non-genotoxic) effects in humans are unlikely to occur at
ambient air  or water  levels  found or  expected  in  the  general environment  or
even at higher levels sometimes  observed in urban areas.  Available evidence
suggests that the teratogenic potential of DCM for humans is minimal.
     The weight of evidence with respect to mutagenic potential  shows that DCM
is  capable of causing  gene  mutations  and has the potential  to cause such
effects in exposed human cells; additional studies are needed to determine the
strength of evidence for mammalian systems.  With regard to cancer, the weight
of  evidence  from experimental animal  studies  is  limited, according to  the
criteria of the International Agency for Research on Cancer.   When the absence
of  epidemiological  evidence  is considered with the  limited animal evidence,
the  overall  evaluation of DCM,  according to  IARC criteria,  is a Group 3
chemical in  that  it cannot be  classified  as to  its carcinogenic potential  for
humans.  Thus,  additional  testing  would be  warranted  to  clarify  more
adequately the carcinogenic potential of DCM for humans.
                                      xvi

-------
                          1.   SUMMARY AND CONCLUSIONS
     Dichloromethane (methylene  chloride,  DCM)  is a solvent  that  is  widely
used for a variety of purposes.   Annual  production of DCM in the United States
is about 269,000 metric tons.   Approximately 85 percent of the DCM consumed in
the United States  is lost directly to the environment through dispersive use,
largely by evaporation to the atmosphere.  Natural sources  have been proposed
for DCM but none is believed to contribute significantly to ambient  concentra-
tions.   Although ambient  air  and water measurements are  rather scarce, they
indicate that DCM  is  found  in a variety of  urban and non-urban areas  of the
United States and  in  other  regions of the world.   The background atmospheric
concentration is about 35 parts  per trillion  (ppt).  Concentrations of DCM  in
urban areas could  be  one or two orders of magnitude higher than background.
DCM is not expected to accumulate in the atmosphere; estimates of half-life
vary from 20 days to 1 year.  Hydroxyl radical attack is" probably sufficiently
rapid to prevent most, if not all, DCM from reaching the stratosphere.
     DCM has been  detected  in both natural  (fresh and seawater) and municipal
waters in various  geographic  areas of the United States.   Concentrations  in
the low parts-per-billion range have been measured in surface water and drinking
water.   DCM does not appear to be formed to any large extent during the chlori-
nation process.  DCM's short  evaporation half-life from  moving water probably
allows most of  the compound dissolved in water  to  be transported into the
atmosphere.   DCM also  is  readily degraded by bacteria in concentrations up  to
400 parts per million  (ppm).  The  extent to  which DCM enters  groundwater from
surface waters is unknown.  Some DCM is deposited in landfills.   Where leaching
is  possible, the  compound may enter groundwater  systems because  it does not
easily adsorb to clay, limestone,  and/or peat moss,  and  retention  in the soil
is  unlikely.  There is no evidence for significant bioaccumulation of DCM  in
the food  chain.   Data concerning the ecological  consequences of DCM  in the
environment indicate that it is  biodegradable under both aerobic and anaerobic
conditions.  DCM appears to have low toxicity to aquatic organisms.
 Ambient is a term used in this and other documents of this nature to
 refer  to  the  environment and should  not  be construed to include  indoor  and
 occupational settings.

                                     1-1

-------
     As with  other  solvents  of this class, inhalation of DCM in air followed
by lung absorption  is  the most rapid route  of entrance into the body.   DCM
also is well  absorbed  into the body after  oral  ingestion.  Absorption through
the intact skin occurs to some extent but is relatively a much slower process.
Because of DCM's high solubility in water and lipids, it is probably distributed
throughout all  body  fluids  and tissues.   DCM's half-time of elimination from
adipose tissue  (6 to 6.5 hours) is consistent  with  reports  that DCM can be
found in such tissue 24 hours after both  single and chronic exposures.  DCM
readily crosses the  blood-brain  barrier,  as evidenced by its narcotic effect
at higher exposure levels.  DCM also probably crosses the placenta and distri-
butes into the  developing fetus, but studies in experimental animals indicate
that effects  on the  fetus  are unlikely at  levels commonly  experienced by
humans.
     The absorption  of ingested DCM is virtually  complete.   The amount of
airborne DCM  absorbed  increases  in direct proportion to its concentration in
inspired air, the duration of exposure, and physical activity.  At concentra-
tions typical of ambient air,  essentially all   of the compound absorbed  into
the body  is  metabolized.  DCM is  known  to be  metabolized by the  liver to
carbon monoxide (CO).  Carboxyhemoglobin (COHb) is  formed from the interaction
of CO and  hemoglobin;  CO dissociates at the lung  and  is eliminated.   Blood
normally contains about  0.5  percent COHb at all times; therefore, CO formed
from a few ppt  or  ppb of DCM vapor is  of no practical  consequence.   However,
at higher exposure levels (500 ppm and  above),  the  COHb level would be expected
to reach a maximum between 12 and 15 percent in man.   This level  is below that
considered hazardous  for normally healthy  individuals,  but  it could place
additional  stress on people with  compromised  cardiovascular systems or on
pregnant women.  The COHb formed  from  DCM metabolism  is  additive  to  COHb
formed from exogenous  CO.  The results  of animal experimentation by several
investigators indicate that  carbon dioxide (C02)  is  an additional  metabolite
of DCM.   Thus,  at least  two  pathways exist in rat liver for the metabolism of
DCM.
                                                                            /
     The weight of  evidence from  the  available literature  indicates  that
adverse toxicologic  (non-genotoxic) effects in  humans are unlikely to occur at
ambient air and water  levels found or  expected in  the general  environment or
even at higher  levels  sometimes  observed in urban  areas.   In fact,  available
experimental   data do  not indicate  that  any adverse  toxicologic  effects are
                                     1-2

-------
induced in humans at  the recommended Threshold Limit Value (TLV ) of 100 ppm
(347 mg/m3) DCM.  The  TLV  is a recommendation of the American Conference of
Governmental  Industrial  Hygienists  (ACGIH),  1983.   The potential direct  ad-
verse health effect associated  with exposure levels  that  greatly exceed  100
ppm (347 mg/m3) is primarily  neurological.  The lowest concentration  reported
to affect eye-hand coordination was 200 ppm (695 mg/m3).
     Liver and kidney damage are unlikely to occur as a result of DCM exposure
at environmental  levels.   Hepatotoxicity has not been reported  in any  human
case report, even following fatal overexposure.  Only minimal  hepatic changes
were observed  in  animal  studies,  even at doses ranging from the average LD50
(about 2 g/kg)  to near-lethal levels.  Animal studies also indicate that DCM
has low nephrotoxicity.
     Direct cardiac effects of  DCM  in humans  also are unlikely because  of the
low levels of  DCM found in the environment.  Animal  studies  have shown that
acute exposure  levels  exceeding 20,000 ppm  (69,480  mg/m3)  are required  before
a decrease in  myocardial contractility and other effects  on  cardiac  perfor-
mance are observed.
     Available  evidence  suggests  that the  teratogenic  potential of  DCM  in
experimental animals  is  minimal.   However,  embryotoxicity, as manifested by
lowered fetal weights and increases in skeletal variations, has been  reported.
These effects  may be  due to  DCM metabolism.  Since animal studies  have been
conducted  using only  very  high doses, it is not possible to evaluate the
dose-response relationship of DCM.
     The weight of evidence with respect to mutagenic potential shows that DCM
is  capable of  causing gene mutations and has the  potential  to cause  such
effects  in exposed human  cells.    Positive  responses to  DCM exposure  were
observed  in  four different test  systems.   However,  additional  studies are
needed to  determine the  strength of evidence  for mammalian systems.
     The  evidence for the carcinogenic  potential of DCM  is  based  upon six
chronic  (lifetime)  studies in which  DCM was  administered to  rodent species.
Four  studies  involved rats,  one  involved  mice,  and  one  involved  hamsters.
Chronic  inhalation  studies with rats  and hamsters  were  conducted by the Dow
Chemical  Company.   One rat study (Dow Chemical Company, 1980) showed a small
increase in the number of benign mammary tumors compared to controls  in female
rats at  all doses and  in male rats  at the highest dose, as well  as a  statisti-
cally  significant increased incidence of ventral cervical  sarcomas,  probably
                                     1-3

-------
of salivary  gland  origin in male rats.   In  hamsters, there was an  increased
incidence of lymphosarcoma in females only; this increase was not statistically
significant  after  correction  for survival.   The results of the Dow Chemical
Company (1980) study have since been published by Burek et al. (1984).
     In the  second inhalation study in rats by  Dow Chemical  Company (1982)
there were no compound-related increased incidences of any tumor type,  but the
highest dose was  appreciably lower than  previously employed.   A  borderline
hepatocellular adenoma  and/or carcinoma response in female Fischer 344 rats
and B6C3F1 male mice was observed only in the chronic drinking water study,
which was conducted  under the auspices of the  National  Coffee Association.
However, while the response was  significant  with respect to matched controls,
the incidence was  within the range of  historical control values at the per-
forming laboratory.
     The epidemiological  data consist  of two  studies:   Friedlander et al.
(1978), updated by Hearne and Friedlander (1981); and Ott et al.  (1983a, b, c,
d, e).  Although  neither study showed excessive risk, both showed sufficient
deficiencies to prevent them  from being judged negative studies.   The Fried-
lander  et  al.  (1978)  study lacked great enough exposure  (based  on animal
cancer potency estimates) to  provide sufficient  statistical power to detect a
potential  carcinogenic  effect.   The  Ott et  al.  (1983a,  b,  c, d,  e) study,
among other deficiencies, lacked a sufficient latency period for site-specific
cancer.
     Using the criteria of  the International Agency  for.Research  on Cancer
(IARC), the  weight of  evidence for carcinogenicity  in animals  is judged to  be
limited.  This conclusion  is  based upon  the statistically positive salivary
gland  sarcoma  response  in  male  rats (Dow Chemical  Company,  19,80)  and the
borderline hepatocellular neoplastic nodule response in the rat and hepatocel-
lular  adenoma  and/or carcinoma  in  male mice (National  Coffee Association,
1982, 1983).  The  occurrence  of limited  carcinogenic activity is  reason  to
suggest that additional testing is warranted to clarify adequately the carcino-
genic potential.   The  National  Toxicology Program  (NTP)  inhalation bioassay
for DCM, due for  release in  early 1985,  will  clarify the issue.    When the
absence of  epidemiological  evidence  is considered  along with the limited
animal evidence, as  well as the potential for DCM to cause gene mutations in
mammalian systems, DCM is judged to be in  IARC  Group 3 category.   The IARC
characterizes chemicals  in this category as not being classifiable as to their
human carcinogenicity.
                                     1-4

-------
     While th^i qualitative classification for carcinogenicity is IARC Group 3,
there is,  nevertheless,  a positive dose-response trend  in  the  incidence of
salivary gland sarcoma,  and these  data can bemused to derive a  rough estimate
of potency for DCM.   Such an estimate might be of use in a regulatory analysis
of how  severe a problem  DCM exposure would be if  it  really were a carcinogen.
The estimate is  included here only for the  purposes of a possible interim
regulatory analyses  pending the evaluation  of  the  forthcoming NTP report.
That report may clarify the nature of the carcinogenic response.           (
     Note must be mWle  of the aggregate findings for  both  mutagenicity and
carcinogenicity.   A positive  finding  of  gene mutations  is  usually  taken to
support the likelihood that a chemical  is a potential carcinogen.   The mutageni-
city findings,  on the other hand, when considered in their own right,  indicate a
potential human adverse effect, although  further studies are required to clarify
the nature of the genetic damage in humans.
RECOMMENDATIONS FOR FURTHER STUDIES
     Further research is needed in several areas.
research priorities.
This list does not indicate
     (1)  Reproductive Effects:   It would be desirable to have more information
          on the dose-response relationship of DCM.  Also, human epidemiology
          studies in women exposed to DCM would provide  useful information on
          the potential for adverse pregnancy outcomes.
     (2)  Mutagenicity:  Additional  tests  for chromosomal  aberrations and
          aneuploidy should be conducted.
     (3)  Pharmacokinetics:   There is a need to define more fully the pharmaco-
          kinetics so as to explain the carcinogenic responses or lack thereof
          and to  clarify questions  and  hypotheses regarding mutagenicity.
                                     1-5

-------

-------
                               2.   INTRODUCTION
     Dichloromethane is  a high-volume  industrial  chemical widely  used to
remove paint and  clean  metal,  and it is a solvent for aerosol sprays.  This
document provides an evaluation  of the health effects of DCM and a review of
the  relevant available  scientific literature.  To provide  a  perspective  in
evaluating health effects  associated  with  DCM, this  document contains back-
ground chapters relating to analytical methodologies, production, sources and
emissions, and ambient3 air concentrations.
     DCM is released into  the  ambient air as  a result of evaporation during
its production, storage,  and manufacturing,  or during general consumer use.
It is believed  to be  derived from natural  sources,  but such formation is  not
believed to significantly contribute to global concentrations.
     Information on the effects of DCM has  been derived primarily from studies
involving individuals occupationally or accidentally  exposed to DCM.  In such
exposures, the  concentrations of  DCM associated with  adverse effects to human
health were either unknown or greatly exceeded the concentrations measured  in
ambient air.  Controlled exposure  studies have established  that vapor inhala-
tion is the principal  route  by which DCM enters the body.   DCM is eliminated
from the body primarily as the parent compound via the lungs.  DCM is metabo-
lized in  animals  and in humans  to CO,  which  elevates the  COHb  content of
blood.   The distribution,  storage,  and  metabolism of DCM to CO and C02 help
explain its effects upon humans.   Epidemiological studies provide some infor-
mation about the impact of DCM  on human health, but  it is necessary to rely on
animal  studies  to assess  any  indications of potentially harmful effects for
chronic low-level  exposures.
     The primary  concerns  addressed  in  this  document regarding the potential
impact of  DCM  on human  health- are narcosis effects  associated  with  acute
high-level exposures and any mutagenic or carcinogenic  effects  potentially
associated with chronic low-level exposures.
 Ambient is used  in  this  and other documents of this nature to refer to the
 environment and  should not  be  construed to include indoor and occupational
 settings.
                                     2-1

-------
     The U.S.  Occupational Safety and Health Administration (OSHA) time weighted
average health standard  requires  that a worker's exposure to DCM at no time
exceed a time-weighted  average  (TWA)a of 500 ppm in the workplace air in any
work shift of a 40-hr week.   The TLV  for an 8-hr TWA concentration is 100 ppm
with a 15-minute allowable excursion, for an average concentration of 500 ppm.
aTWA  is  the concentration for a  normal 8-hour workday and a 40-hour workweek
 to which   nearly all workers may be exposed  repeatedly, day after day, without
 adverse effect.
                                      2-2

-------
                 3.  DICHLOROMETHANE:  BACKGROUND INFORMATION
3.1  PHYSICAL AND CHEMICAL PROPERTIES
     Dichloromethane  is  one  member of a  family  of saturated aliphatic  halo-
genated  compounds.   Other common names or  synonyms  are shown in Table 3-1.
The Chemical Abstracts Service Registry Number for DCM  is 000075092.  Dichloro-
methane  is  a colorless,  volatile liquid  that  is completely miscible with a
variety  of  other solvents (Anthony,  1979).   Although it has no  measured flash
point, it does possess flammable limits in air.  The important physical proper-
ties of  DCM are  shown in Table  3-2.  For  example,  DCM is  highly volatile
(vapor pressure of 350 torr at room temperature).  Hence, the most common mode
of  entry into the  body  is inhalation.   The ambient air concentration for
compounds of  this nature is  often  expressed in parts per million  (ppm),  parts
per billion  (ppb),  and parts per trillion (ppt).   At standard temperature and
pressure, 1 ppm (v/v) is equivalent to 3.79 mg/m3.   In this document, a tempera-
ture of 25ฐC has been used.  In all calculations, 1 ppm  (v/v) is equivalent to
3.47 mg/m3.
     In the  absence  of  moisture  at ordinary temperatures,  DCM  is relatively
stable compared  with  its congeners,  chloroform and carbon tetrachloride.  In
dry air, DCM  decomposition is  initiated at  temperatures  above 120ฐC  (Anthony,
1979).   As  moisture  content  increases, temperature decreases.  The principal
reaction product is  HC1.   Decomposition under these conditions can be inhibited
by  the  addition  of  small  quantities of  phenolic  compounds.  On prolonged
contact with water,  DCM  hydrolyzes very  slowly,  forming HC1  as the primary
product (Fells and Moelwyn-Hughes,  1958).
     Although anhydrous  DCM is noncorrosive to common metals, the HC1 produced
from hydrolysis  of  DCM  initiates  corrosive action  with aluminum  and iron
(Anthony, 1979).  The addition of epoxides  to consume the HC1 affords protec-
tion against this corrosion (Anthony, 1979).  To minimize the decomposition of
DCM, storage containers  should be galvanized or  lined with  a  phenolic coating
(Anthony, 1979).   Commercial  grades of DCM contain a variety of  stabilizers to
minimize decomposition (McKetta  and  Cunningham,  1979).   Cyclohexane, thymol,
hydroquinone, p-cresol,   and  low-boiling amines  have been used as stabilizers
(De Forest,  1979).   Aerosol  preparations  containing DCM often use propylene
                                     3-1

-------
           TABLE 3-1.   SYNONYMS AND IDENTIFIERS FOR DICHLOROMETHANE

Chemical Abstracts Service registry number:   000075092
Chemical formula:        CH2C12
Structural formula:
Synonyms:
 Dichloromethane
 Methylene dichloride
 Methylene bichloride
 Methylene chloride
     Cl
H -  C
     Cl
                                           - H
oxide as  a stabilizer.   For preparations designed  to  be used in degreasing
applications, special  inhibitor  mixtures  are  used.   One  such  mixture  includes
propylene  oxide,  butylene  oxide,  cyclohexane,  and N-methyl  morpholine  (De
Forest, 1979).
     The  experimentally  determined average  evaporative  half-life of DCM from
water is  18 to 25 min (Oil ling,  1977).  In three separate experiments, Oil ling
used  solutions  of an average depth of 6.5 cm, containing approximately 1 ppm
of  DCM.   The solutions were stirred at 200  revolutions per  minute in  a 250-ml
beaker.   These  experimentally determined half-lives agreed  with  the  value
(20.7 minutes) obtained  by  the following  formula:
                                    0.06391d
                                    (3-1)
where  d  is  the  solution  depth  and  kj  is  the  liquid  exchange  constant  (cm/min).
The  formula is an adaptation  of  the  common equation for  the  half-life  of  a
substance  undergoing  a  first-order  reaction.   The calculated  evaporative
half-life may  not be accurate  for  DCM in natural  aquatic  systems.
                                      3-2

-------
              TABLE 3-2.  SELECTED PROPERTIES OF DICHLOROMETHANE
Molecular formula

Formula weight

Boiling point (760 mm Hg)

Melting point

Vapor density

Density of saturated vapor

Density

Solubility




Explosive limits in oxygen

Flash point

Autoignition temperature

Relative evaporation rate


Vapor pressure
Conversion factors
 (25ฐC; 760 mm Hg)


Concentration in saturated air

Log octane/water partition
   Cri2C I 2

   84.94

   40ฐC (760 mm Hg)

   -95 to -97ฐC

   2.93 (air = 1)

   2.06 (air = 1)

   1.326 g/ml (20ฐC)

   2.0 g/100 ml water at 20ฐC;
   soluble in ethanol,
   ethyl  ether, acetone,
   and carbon disulfide

   15.5 to 67% by volume

   None

   624ฐ to 662ฐC

   14 (water, = 1)
   71 (ether = 100)

   Temp ฐF   Temp ฐC  mm Hg
                                                    50
                                                    68
                                                    77
                                                    86
                                                    95
              10
              20
              25
              30
              35
230
349
436
511
600
1 mg/1 = 1 g/m3 = 288 ppm
1 ppm (v/v) = 3.474 mg/m3 =
   3.474 ug/1

550,000 ppm (25ฐC)

1.25
Sources:   The above information has been compiled from a number of references
          including Hardie (1969), Anthony (1979), Dill ing (1977), DeForest
          (1979), and American National Standards Institute (1970).
                                     3-3

-------
3.2  ENVIRONMENTAL FATE AND TRANSPORT
     Because of its  volatility and dispersive use pattern,  much  of the DCM
produced worldwide is  emitted into the atmosphere.  Almost  all of  the  emis-
sions are from anthropogenic sources.  DCM is also formed from natural sources,
but natural  sources  are  not believed to contribute  significantly to global
concentration (National Academy of Sciences, 1978).

3.2.1  Production
     Dichloromethane is  commercially produced in the United States, predomi-
nantly via the following reaction (De Forest, 1979; Anthony, 1979):
                            FeCl3 or ZnCl
               HC1 + CH3OH  ^Qo _j. nonOp "*  CH3C1
                                                                      (3-2)
               CH3C1 + C12
-*  CH2C12 + HC1.
     In this  vapor  phase reaction sequence, yields  of  95  percent  are  usual.
Dimethyl ether is the secondary byproduct of the hydrochlorination.
     A  less  common  method used to obtain  DCM  is direct reaction of methane
with chlorine  at  485ฐ to 510ฐC  (Anthony,  1979).  Methyl  chloride,  chloroform,
carbon  tetrachloride, and HC1  are coproducts.    However,  both  reactions  are
used in the  chemical  manufacturing industry so  that the HC1  can be  recycled
(Anthony, 1979).
     In the  liquid  phase, DCM can be  produced by  refluxing and distilling a
mixture containing  methanol,  HC1, and zinc chloride at 100ฐ  to 150ฐC.   How-
ever, this method is not widely used (Anthony, 1979).
     According to one  source,  production  in the United  States  is carried  out
by five major companies  at seven  sites (Table  3-3).
     According to statistics  gathered  by the U.S.  International Trade  Commis-
sion, the  United  States annual production of  DCM was estimated to be 256,000
metric  tons  in 1980 (U.S. International  Trade Commission, 1980) and 269,000
metric  tons  in 1981 (U.S. International Trade  Commission,  1982).
     Edwards  et al.  (1982)  estimated 1981 world production at  825,000 metric
tons.
                                     3-4

-------
                   TABLE 3-3.  PRODUCERS OF DICHLOROMETHANE
       Company
     Production in
Million Pounds Per Year
   Diamond Shamrock
   Dow Chemical
   Stauffer Chemical
   LCP Chemical
   Vulcan Materials
          110
          300
           52
          210
 Terminated Production
Source:  Chemical Marketing Reporter, 1983.

3.2.2  Use
     Dichloromethane  is  used  as  a  paint remover, a urethane  foam-blowing
agent, a vapor  degreasing and dip solvent  for  metal  cleaning,  a  solvent  for
aerosol products, a  solvent in the pharmaceutical industry, a solvent in the
manufacture of polycarbonates by polymerization, and an extractant for caffeine,
spices, and  hops.   DCM  is  used in the manufacture  of  plastics,  textiles,
photographic film, and photoresistant coatings, as a solvent carrier  in  the
manufacture of herbicides and insecticides, and as a component of rapid-drying
paints and adhesives, carbon removers, and brush cleaners.  Other minor appli-
cations include  use  as  a  low-pressure  refrigerant,  a low-temperature heat
transfer medium, and an  air-conditioning  coolant  (Ahlstrom and  Steele, 1979).
Distribution of DCM by major use is shown in Table 3-4.
     Dichloromethane is  expected to  retain  popularity  as a paint remover.
Although DCM competes with  trichloroethylene and perch!oroethylene as a sol-
vent, it is preferred  as a paint  remover because of its better performance
(Lowenheim and Moran, 1975).

3.2.3  Emissions
     Emissions from dispersive uses are the major source of DCM  in the environ-
ment.  Of the  total  amount of DCM produced in the Unitsd States, approximately
85 percent is  estimated to be lost into the environment through  sewage treatment
                                     3-5

-------
                  TABLE 3-4.   CONSUMPTION OF DICHLOROMETHANE
          Use
Percent of Production
   Paint remover
   Metal degreaser
   Aerosol  propel 1 ant
   Blowing agent for foams
   Export
   Other
          25
          10
          25
          10
          12
          18
Source:   Chemical Products Synopsis, 1981.

plants and surface waters, deposition on land, or loss to the atmosphere.   The
actual losses during production, transport, and storage are not well  documented,
but it appears that such losses represent only a very small percent of the DCM
entering the environment from product manufacture and use.   Most of the losses
in production, transportation,  and  storage are fugitive (that is, transient)
releases from  leaky  pump  seals, valves, and  joints.  The  dispersive  uses of
DCM are varied and widespread and are distributed geographically approximately
with the industrialized population in the United States.  Although most of the
losses are  to  the atmosphere, DCM  is relatively soluble in water.  The most
effective means of removal of DCM  from water is air stripping, which transfers
the  chemical  from water  to  the atmosphere  (National  Academy of Sciences,
1978).

3.2.4  Persistence of DCM
3.2.4.1  Atmospheric Degradation—Reaction with hydroxyl radicals  (OH) is the
principal process by which many organic chemicals,  including DCM, are scavenged
from  the troposphere (Crutzen  and  Fishman,  1977;  Singh,  1977;  Altshuller,
1980).  These radicals are produced by irradiating ozone (03), and the resultant
singlet  oxygen atoms  then react with water vapor.   The tropospheric lifetime
of  a compound can be  related to the OH-  concentration by the expression:
                                     3-6

-------
                           lifetime  =
                                         k [OH]
                                                                      (3-3)
where k is the rate constant of reaction.

     Available evidence  indicates that the  lifetime  of DCM  in the troposphere
is less than  1 year.   The modelling approaches of Crutzen and Fishman (1977)
and Singh  (1977)  indicate that the range of the  average  OH concentration is
between 2  x 105 and 6 x  10s molecules/cm3.  Using an average concentration of
3 x 10s molecules/cm3,  Altshuller (1980) calculated a tropospheric  lifetime
for DCM of 1.4 years.   The rate constant expression of Davis et al.  (1976) and
a tropospheric temperature of 265ฐK were used.
     Singh et  al.  (1979) computed a 1-year lifetime for  DCM  using the rate
data reported  by  the National  Aeroneutics  and Space Administration  (NASA,
1977) and  the  National Bureau of Standards  (1978) and a temperature  of 265ฐK.
An average OH concentration of 4  x 105 molecules/cm3 was  employed  in the
computation.    More recently,  Singh  et al.  (1983) calculated  an atmospheric
residence   time of 0.9 ฑ 0.3 year,  based upon a  comparison of  atmospheric
budget data with  available  emissions  data.   Since the  mean concentrations in
the northern hemisphere  are 1.8 times the southern hemisphere values, a short
lifetime for DCM is indicated.
     However,  Cox et  al.  (1976) calculated a 0.3-year lifetime in photokinetic
studies in which  DCM  competed  with nitrous acid  as  the target of OH  attack.
The lifetime was derived from a rate constant of 10.4 x 10 14  cm3/molecule •  sec
at 298ฐK.   An  average OH concentration of 1 x 106 molecules/cm3 was  assumed.
Use of 4 x 10s molecules/cm3 for the OH concentration would have resulted in a..
calculated lifetime of  0.76 year;  this  value  is closer  to those  of Singh
(1977) and Altshuller (1980).   Davis  et al. (1976)  calculated a lifetime of
0.39 year  from a rate  constant of 8.7 x 1014 at 265K and an average OH concen-
tration of 9 x 10s molecules/cm3.
     Determination of the rate  constant  for the  reaction of OH with  DCM has
been the focus  of various investigations (Davis  et  al.,  1976;  Cox et al.,
1976;  Howard and Evenson, 1976;  Perry et al.,  1976).  The values are  in general
agreement  (Table 3-5)  with the exception of Butler et al.  (1978).
                                     3-7

-------


















G
DC
O

•f*

^
s_
S-
a>
a.












i
i
i













LD
,
CO
cn
CM














CD

CM
+1

LO

ซ3-
r~H


cn
p— I
*,
c
o
in
e
0)
LU

-a
re

•u

re

o













i
i
i















cn
CM














^j.
^
CO
+1

LO

LO
I — 1





t*O
r*-
C7>


t
r~.
re
.|^>

re
C3




i — i
/^^
•N h-

1 rH
CD CO
x;

cn cn
t*D CD
• rH
Q \ 	 S
+1 1— 1
Is- O.
CNI X
. O)








LO
Is-.
CO
o

LD

CM










LO

CD
+1

UD .

r-4
i
p^



re in
co c-a
Is-. 01-
cn T- re
lฃ) r-i +-> TD
r-~ re c:
cn •- 2: re
, 	 | . ^j

" la r-~
1s-- 4-
r— +J cn o
re cu i— i
3
+j s_ re re
CU CU " CU CO
i— 
re
CM
CD
+1

CU
o
•^
T3
CU
O)
•a
3
"r~3

^"^
cn
o
f-™"

4-
O
•P
re
• r—


-------
 3.2.4.2   Aquatic Biodegradation--Recent evidence indicates that  DCM  is  bio-
 degradable  under both aerobic and anaerobic conditions.  Brunner  and  Lei singer
 (1978)  first reported the isolation of a facultative methylotroph with the
 ability  to  utilize DCM as a sole carbon  source  for growth.  The  organism was
 tentatively  identified as a Pseudomonas species.  Brunner et al.  (1980) observed
 that  utilization of DCM is caused  by  an inducible enzyme, the activity  of
 which  is only partially inhibited  under anaerobic conditions.   Degradation  of
 DCM by  Pseudomonas sp., strain LP  was  also  observed  by LaPat-Polasko et  al.
 (1984).
     Rittman  and McCarty (1980) investigated sewage microorganisms for their
 ability  to  biodegrade DCM.   Bacterial   cultures  were  enriched  from a  seed of
 primary  sewage effluent over  a 12-month  period.   DCM supported bacterial
 growth  when the mineral salts  culture  medium was supplemented with  sodium
 bicarbonate.
     Klecka  (1982)  investigated the fate  and  effects  of DCM in a  system simu-
 lating a municipal waste water treatment plant.   After acclimation, the sludge
 was used in closed-bottle  respirometer studies.  Respirometer studies demon-
 strated  the  disappearance  of  10 mg DCM/1 (14C-label led) within 4 hr, with 49
 percent of the parent compound recovered as 14C-labelled C02 after 50 hr.  At 1
 mg DCM/1, disappearance  resulted within 3  hr, with a  66  percent conversion  of
 the parent  compound  to  C02 after 50 hr.   Rate  constants, calculated  for the
 biodegradation of  1,  10,  and  100 mg/1  by  activated sludge, were  1.42, 1.61,
 and 0.35/hr, respectively.
     In  inhibition  studies  on  the  effect  of  DCM on oxygen consumption and
 organic  carbon degradation by activated sludge,  Klecka  (1982)  found no signi-
 ficant effect  over  a 24-hr period.   Modelling  of a continuously  mixed acti-
 vated sludge  reactor  indicated  that the rate of biodegradation was about 12
 times greater than the rate of volatilization.
     Wood et  al.  (1981)  demonstrated the degradation of DCM under anaerobic
 conditions,  using  sediment-water samples  spiked  with 200 pg DCM.  Degradation
was observed to proceed via methyl  chloride,  although  accumulation in the sam-
 ples was not observed.

3.2.5  Products of DCM
3.2.5.1  Atmospheric Simulation Studies—Pilling et al.  (1976) observed that
DCM was not  very reactive in a chamber atmosphere containing nitric oxide  (NO)
                                     3-9

-------
or nitrogen dioxide  (N02).  Ozone-air mixtures containing 10 ppm (34.7 mg/m )
DCM and 5 ppm NO or 16.8 ppm N02 were exposed to ultraviolet (UV) radiation at
an intensity about 2.6 times that of natural sunlight at noon on a summer day
in Freeport, Texas.  After 21 hr of exposure in the presence of NO, less than
5 percent of the  DCM had disappeared.   Similarly, less  than  5  percent dis-
appeared in an N02 atmosphere after a 7.5-hr exposure.  The effect of varying
UV intensity or concentration on the rate of photodecomposition was not inves-
tigated.  Relative humidity  in  the  photolysis reactor was 35 to 40 percent.
Further investigation of  this  simulated trospheric reaction showed anomalous
behavior, possibly occurring on the chamber walls (Dilling and Goersch, 1980).
Dichloromethane was  judged to contribute to oxidant formation less than other
halogenated compounds investigated,  e.g., tetrachloroethylene, trichloroethylene,
and vinyl chloride.
     Butler et al. (1978) proposed that  the *OH attack on DCM in the presence
of 02 may  result  in  formation of phosgene (COC12) via the following reaction
sequence:
                         CH2C12 + OH •* -CHC12 + H20
                         •CHC12 + 02 •* -CHC1202
                         •CHC1202 •* COC12 + OH.
(3-4)
This pathway was  suggested  to  account  for  a  low  rate  constant  of  the  reaction
of  -OH with  DCM in the presence  and  absence of CO in  the  test atmosphere.
Production of C02 was followed.
     Chlorine-sensitized photooxidation  of DCM in the presence of C12 in dry
air resulted in CO  and  C02  as  the major  carbon-containing products  (Spence  et
al., 1976).  After 5 min of irradiation of 20 ppm DCM and 5 ppm C12 in air, 19
ppm CH2C12 was  consumed.   The product distribution was CO  (5 ppm); HC1 (38
ppm); COC12  (2  ppm);  formyl chloride  (0 ppm); and C02  (12 ppm).   The product
distribution is illustrative of a chain reaction:,
                               •CHC1202 -> CIO + HCOC1
                               HCOC1 + Cl •* HC1 + COC1
                               COC1 + 02 -* C02 + CIO
                               COC1 ^ CO + Cl

                                     3-10
(3-5)

-------
 Chlorine-sensitized photooxidation of  DCM is not expected  to  be  significant
 under real atmospheric  conditions  because Cl will react  with  species other
 than halocarbons (Spence et al., 1976).
      The hydrolysis of  DCM  in natural  waters is expected to be influenced by
 temperature as  well  as  by  acidic  and  basic conditions.  There  is limited
•experimental  evidence on the  rate of hydrolysis of DCM under different condi-
 tions.   Oil ling et al.  (1975) determined an experimental half-life of about
 18 months (at 25ฐC).   The pH was not stated.  Fells and Moelwyn-Hughes (1958)
 reported a hydrolytic half-life of 13.75 days in acidic pollution at tempera-
 tures ranging  from 80ฐ  to 150ฐC.  When  Billing  et  al.  (1975) extrapolated  the
 Fells and Moelwyn-Hughes data  to 25ฐC, the  resultant  half-life was about 680
years.   Dilling  et al.  (1975)  hypothesized that  different  mechanisms of hydro-
 lysis may account for the discrepancy  between  experimental  data at 25ฐC vs.
extrapolated data  from  high  temperature experiments.
      Radding  et al.  (1977)  in  their literature survey reported  a  maximum
hydrolytic  half-life of  704  years (100ฐ to 150ฐC at pH  7).
3.2.5.3   Sorption—Pilling et al.  (1975)  found  that DCM could  be  adsorbed  to
dry  bentonite  clay and  peat  moss when these  absorbents were  added to a sealed
solution  containing DCM.  However, the  DCM that  leaches from landfills  adsorbs
very  little to clay,  limestone, and/or  peat  moss,  so  retention  in the soil  is
unlikely. These  authors  consider evaporation of DCM from water a  more  impor-
tant  process than adsorption.
3.3  LEVELS OF EXPOSURE
     Dichloromethane  has  been detected  in ambient air  and  in surface and
drinking waters at numerous locations throughout the United States.
     Dispersion models  have been  used to  estimate  population exposure to
ambient DCM.   The predicted maximum annual average  to which people may be
exposed is 14.3 ppb  (0.050 mg/m ), resulting from living near DCM production
facilities.   People  living near other DCM  sources  such as organic  solvent
cleaning and paint and  varnish removal operations are expected to be exposed
to concentrations that  do  not exceed 7.1 to 14.3 ppb  (0.025 to 0.050 mg/m3)
averaged over a year's time (Systems Applications, Inc., 1983).
     Average  background mixing ratios  are  approximately 30  to 50 ppt.   In
their review of ambient air data,  Brodzinsky  and Singh (1983)  concluded that
                                     3-11

-------
background levels are  about  50 ppt, with many urban levels one or two orders
of magnitude higher.   Ambient air levels of DCM at various locations are shown
in Table 3-6.  Singh et al.  (1983) reported that the average northern hemisphere
background mixing ratio  is  approximately 38 ppt  and  the global  average is
29 ppt.  The  Washington  State  University  group  reported  free troposphere
values between 30 and  40 ppt (Cronn et al., 1977; Robinson, 1978; Cronn and
Robinson 1979; Grimsrud and Rasmussen,  1975; Rasmussen et al.,  1979).
     Pellizzari and Bunch (1979)  have  compiled a list  of  the  sites in the
United States  at which Pellizzari and coworkers have  identified DCM.  DCM was
                                ฎ
sampled by adsorption onto Tenax  coupled with analysis by high resolution gas
chromatography-mass  spectrometry  (GC-MS).   The  highest  DCM  concentration
reported in New Jersey was at a waste disposal  site in Edison.   A level  of 360
ppb was measured during  an 11-minute sampling period  in  March  1976.  During a
75-minute sampling period (November 1977),  55 ฑ 0.1 ppb  DCM was detected at a
site in Staten Island, New  York.   Longer sampling times (up  to 8 hr) were
required at  sites in Virginia, West Virginia, and Pennsylvania to detect DCM.
During a 7.25-hr period, about 70 ppb DCM  was  detected at a  site in Front
Royal, Virginia (October 1977).   In the southwest, a  level  of  about 1 ppb was
reported at  sites in  Houston,  Texas, during a 3-hr sampling period (October
1977).  During  considerably  longer  sampling periods (up  to 24  hr),  lower
concentrations were reported for  sites  in  Louisiana, e.g., Baton  Rouge and
Geismar.   Sampling during a  48-hr period in Upland,  California (August 1977)
indicated concentrations  of about 12 ฑ 9 ppb DCM.

3.3.1  Analytical  Methodology
     There are four practical  methods  to measure air concentrations  of the
halogenated hydrocarbons.

     1.   Gas chromatography wi.th an electron-capture detector.
     2.   Gas chromatography-mass spectrometry.
     3.   Long-path infrared absorption spectroscopy, usually  with preconcen-
          tration of whole  air and then separation  of  the compounds by GC
     4.   Infrared solar spectroscopy,  using the solar  spectrum  at large
          zenith angles to obtain greatest path lengths through the atmosphere.
                                     3-12

-------


















LU
ฐy*
*ฃ
t—
LU
5^
o
o;
Q

x:
o
t— 1
o
LL.
O
co
_J
LU
^>
LU
i

oi
1—4

O
ฃ
(0
s-

O)
c;

X
•r-
s





"V^
-p
C -O
a> o
ฃ x:
CU -P
i- O)
=3 e
in
ro i—
O> (O
E U

it _n i ^
0 >>
1—
0) ra
•p c
(0 n3
a








cu
-P
• r""
in

q-
o

ai
o.
I—
c
o
•r—
-P
rO
U
O
__j



rH
CO
en
rH
•
n3

-p
cu

x:
O5
c
co

CO
CO
CO
en
.
0


CM en co
to in co
to co en
rH O CO
LO CD O

X <= OX



^^
en

en
rH

*ป
CO

>}
ro


1

CO
CM

r- O
•r- LU
S- 1
Q.CJ
 O5 O> O)
C C C C
co co co co



>=*• >* LO CM
rH en CM CM
O 0 O 0
ซ • • *
0000
+1 +1 +1 +1

r^-ooin cocorH cocn-^- CMCOLO
OOCOLO r>-rOrH CMO-si- OrH-si-
OOO ซtf-Or-H r-HCDO rHOCD
000 OOO 000 OOO

XICICW XICIO)) X|C|0!| X CIO)
fO -r- > rO^r- > rO -r- > tO-r-\>
sls:l -^.
r^ ~ ^^ p*^
en ^f~ r^ f^.
rH p- en
>> CTl rH

r-. s
CM 0
1 CO CO
1 rH
LO 1
rH CM 1
rH CO
CJ r— CJ LO O CM CJ
. LU •!— LU LU LU
C 1 S- 1 >> I >ป 1
rO CJ Q.CJ ro O rO O
•"3CD  32 s:
>> w ro
to  in s-
q- s- cu -P
•r- i— CU O) C
r— i— > T3 >r—
rO *r- T- rO o
cj s: of QQ a.
en en rH
r~- r~- co
en en en
rH rH rH
*i #i n
rO rO rO

•P | ^ j %
cu a> at

x: x: x:
O) O) O5
c c c:
co co co
CO
o
CM
CM r>- CD
LO rH
CO 0 CM

CD CD +1
+1 +\ CO ff r-t
00 rH rH
OOrH OCOCn CMOLO
CMCDCD CO i— 1 CO OCOf->
en o •* ooo ...
t— 1 CD O OOO r-H

X CIM XICIM X|C|O)|
(8 T- > rohr- > ro T- >
s: szl- LO r^- o
en en -sซ>
rH . rH
-P i.
" p ' ป-
r*~ 0) rH
CM CO CM

1 1 1

rH CD en
CM CO
CJ O i — CJ
. LU • LU •!— LU
O) 1 O) 1 i- I
3 CJ 3 CJ O.CJ
 s: =3
ca in
c: cu
CU i—
cu s- cu
in 
-------


































/"->
•a
>

a) ra
-p c
CO CO
ca


O)
•p
•r-
in
0

0)
CL

*
c
o
•r-
4->
CO
U
0




P-.
cn
.c: I-H
u
rH C cn
OO 3 p*.
cn oo cn r—
1 — 1 I — 1 CO
73
C " 4->
co • eu
i— •• r~~
CO *i— CO "i—
S_ S-
•P CO -P CO
(UN O) N
N cn N
* *f~* P** ^" T"
O) i — cn cn i —
C r— rH C i—
•r- CU -i- CU
co a. oo a.
to
r-H in
CO rH
o r-
CD in • t-
CD
•H •=!• C
+1
co cn m +i +
inmm 
csoot— i tnin ocoin Lnซd-Lr
^- CD *^" • • rH CD O tn CD CN

CMOOrH OOO OOC

XI *"" | O3| X CI | Ol| X I C | O3| X I C C
(O IT- 1 > 1 CO *r~ 1 >1 CO |ซr~ 1 > | CO rr" 2
srfeh:! s: s:lซa:l s:ls:l> -P cn
r— CL rH >>
30) CO
o co - s:
r-~-
i i i
i
oo r-- co
CM rH rH
o oo o x: oo
OJ UJ .S CU LU OS
c: i cn i c i s- i
30 30 30 COO
'"O CD ^ CD *~3 CD 2EZ CD





CU
C C .p C
co co o co
r** ft ฃ2 f^
t- t- cu s-
=3 ZD Di =D
0)
CO O)
CO 3
C O
T3 4C CO Qi
C T3 W S- -I-
co cz cO co c/) c:
i — re in S -r- o
^<ฃ r— C -P 3 .p
co CL co cu o co
o => *ฃ <-3 — i ca




o cn
re oo r--
oo cr> cn
r-. I-H rH
cn
rH ** *^
. •
ป, f— ^~
*r— fO rO
5-
ro -P -P
N CU CU
N
r^- CO CJ5
i— C C
CU •!- •/-
O- CO OO

CM
CM CM
s tJD ซ^f- CD
HO rH
O
5 CD CD
+1
1 +1 +1
cn in CM
5 COCOCO CMlฃ>Cn CnrHin
J COCOCO IDrHCO CDOCD

3 OOC3 OOO OOO

•A XIC CTj XICICM X C|0>|
> (0 -r- > CO  • rO -rrr. >
:| S:!E:ซC| S:|E:|, O
!"•* O
*ป ^ v*^
oo cn
•^ rH
r— cu
r*ซซ cz **
cn 3 CD
rH O CM

1 1
rH
CD ^t"
-C OO CO O rH O
us: uj LU
i- i >> i >> i
rO CJ ro O ra o
s CD s: CD s: CD
CU
73
3
-P
• |—
P
p—
CO
c c:
CO CO -C
-O .Q O)
S- S- •!-
=> :D 3:
5-
cu
co tn >
*r- ซp- •(—
t- S- 3 Qi
CO 3 O CO
E O — 1 73 CU
in in co in
•i- in > a>
a) .•!-+•ป aป aป
CD s: co z. ce.
3-14

-------














"O
cu
3
•r-
C
o
u

CD
|
CO
LLJ
_1
GO
H^
























CU
u
c
cu
S-
cu
o>
oi

JD
Q.
Q.
O
re
O)
c
•r—
X
•r-
Z.



^
+-^
C TJ
CU O
ey—
-I—
CU -P
s- cu
3 E
re i—
cu re
E o
•f—
0 >>
cu re
•4-> C
re re
Q
cu
to
o
cu
Q.
r—





C
o
'ฃ
re
o
o
— i

cn
r-l |>ซ.
u
3 re
CQ 00
pv.
•o cn
C,_i_t
I 1
re
f.
•r~~ •[—
s- s-
re re
N N
N N
*r~ *i—
i— r—
cu 'cu
O. Q.
CD
0
CD CO +|

rH
^ ^>
r^^ป r"3
CD
r-l 1

CM CM
CO JZ CO
Q. 1 t! ^
cu o re o
co cs s: o


c c
re re
t ฃ
ZD ..O C
re
cu
u
o
u
nO ti
>j -p •ฃ
CU S- (j
to o re
i- O. Q_
CU CU C
'-JO) 0 JZ
•O 10 +J
cu Z '-5 c3
z CQ uj z:


f** c
r^^ c
O"> t
rH C
00 ปr
**, r*^ ^
CTJ C
I — 1 — I Q
re
rt "C

cu o n
C -r- C
O JO C
1-0 t
C_3 fVT C-
00 CO
0 -3-
0 0
0 0
O CO
co co
O CD
0 0 I
>1 >l x
 1 1
o o o
"V, CJ3 O
r*^* CD r*^ซ
rH CD CD
f"H rH
'C ^ 2
Q. U 3
   r--  r-.
                                       r-^   r^>   i^.  r*.
                                       CD   CD   CD  CD
                                       4->
                                       u
                                       o
     C
     re
     +J
     0
     o
>
o
cu
c
• |W
5-
re
cu
s-
OJ
JZ
Q.
U5
•r—
CU
IT:

c
cu
-C
3
0
CO
s:






CO
0
CM
*='~
o
"^
o
o
cu
^>
o
E
CU
n^





CO
O
o
cn
o
•f-1
o
f^.
r-ซ.
cu
c
*r—
S-
re
y





CO
o
UO
LO
O
•4-5
O
CM
CO
CU

O
ฃ
CO
Qฃ





CO
0
o
CD
O

O

r~ซ
3-15

-------



































<*- \
T3
CO
3
C
'43
c
o
(J
*1*-^


•
10
1
CO

LU
—I
00

C
[x

IE







x^
•P
c -a
CO O
E-e
CO -P
S- CO
3 E
in
10 r—
a> to
E O
•r~~
*t~" -P
O ^>
r—
CO fO
4-J C
to to
Q










CO
-P
in
o

a>
a.
^>
r—







C
O
4J
to
O
O
— 1

en en
f^. f*-*
cn cn
rH rH

ป> ซ\ <•" i
. CO XI
r— r— Cn CO
tO - cn
CO CM CO CO LO ซtf- LO
	 rH . .
rHOO tHC3C5 CMOCO
X|ฃ=|Cซ X C|O)| XlClCrt
tO •!- \>\ \ (0\r- >1
slslccl s:s:l<:l s:|2:lป LLJ \
r^^ i r*^
r^ o r^-
cn cs cn
rH rH

OO
CD E: in
CM II rH
O
• •ป ^3 >
•p r^ "^x. o
o r~ป o z:
o cn r^>
rH Cn i
1 rH
cn
CM CO - CM
CM CM ซ3-
00 CM .00

= i ฃ= >> "clT
3 O 3 tO CO O
r~3 CD *"^ ^* OO CD












tO
X)
S- C C
3 tO (0
(~* o r*>
3 S_ s-
oo rD zj







r--
tO c
* % o* 43
•x c •!- oi cn
co o c c:
in c +J -i- +J •!-
to -i- in cn c JE
X "O 3 S- O in
CO r— O •!- S- 10
I—  LL. ^
^
C CD
co r~-
m cn
in rH
3

in
tO r—
ca to

T3 -P
ฃ= CO
to
c
•a co
3 in
S- LO in
in r-- 3
E cn E
•r- rH in
S- fO
CD o"







LO LO
O CO
O O

O O
1 1 V
X 1 C- 1 cni cn|
sis l-a-l ซ:!






"•x.
LO

cn
rH

f
J3 OO
co s:
U- 1
o

x^
•>!- LO
r-- r-
cn cn
iH rH

• ^" ซ
U 1 >
CO O O
Q CD -Z.













r— r—
to to
S- ฃ_
3 3
I*V fV










C C
1 1
1 — 1 —
3 3
a. a.








































^
CO
00
cn
rH

^
^-
cn
c

oo

•a
c
tO

^^
\x
• in

CO t- *r~
S- -P N
3 co -a
-P E O
Q. O i-
tO S- QQ
0 -P
U <ป-
C CO O
o a.
S- in +J
-P S-
u in o
co m Q.
r- tO CO
CO E S~
>>>> >>
o. a. to
10 to E
S- S- E
cn cn 3
o o in
•p -p
(XJ CO E
E E 0
O 0 S-
S- S- 4-
^Z ^=
U U TD
CO
in in c:
to to T-
cn cn to
-p
II II -Q
o
o oo
LU s: to
1 1 4->
o o m
CD CD C3
to *:
3-16

-------
     Each method has  advantages  and disadvantages and applications for which
it is best  suited.   A major drawback of these techniques is that they do not
allow real-time continuous  measurements  of the halocarbons at ambient levels
in the environment.
     The two  most  widely used systems for identifying and measuring trace
amounts of  DCM  that  occur in ambient air  are  GC-MS and gas chromatography-
electron capture detection  (GC-EC).   Both  systems have a  limit of detection
below 30 ppt.   The GC-EC method  has been reviewed by Pellizzari (1974) and by
Lovelock (1974).   The electron-capture detector'(ECD)  is  specific in that
halogenated hydrocarbons are  quantitated,  while non-halogenated hydrocarbons
do not  respond.  Thus, high background levels  of non-halogenated hydrocarbons
in ambient  air  or  water  samples do not interfere with measurements of halo-
genated hydrocarbons.  In a complex mixture in which  several  compounds  may
have similar  retention times,  alteration  of the operating parameters of the
GC-EC system  will  usually provide separation  of the  components.   Linearity
over a wide concentration range is achieved when the electron capture detector
is used in the constant current mode.   In this mode, the  change  in pulse
frequency  is  linearly related to sample concentration.  Nitrogen  or a 95-
percent argon/5 percent  methane  mixture  is commonly used as the carrier gas.

3.3.2  Sampling of Ambient Air and Water
     Contamination,  absorption,  and adsorption  are common problems of the
methods used to analyze air and water for DCM content.   Four general approaches
are used to collect  samples of  air for analysis of trace  gas  concentration:
(1) cryogenic sampling in which liquid helium  or liquid nitrogen  is used to
cool  a container to extremely low temperatures; (2) pump-pressured samples, in
which a mechanical  pump is used without cryogenic assistance to fill a sampler
to a positive pressure relative  to  the surrounding  atmosphere;  (3) ambient or
subambient  pressure  sampling  in  which an evacuated  container  is simply opened
and allowed to  fill  until  it has reached  ambient  pressure at the sampling
location; and (4) adsorption of selected gases on adsorbants such as molecular
sieves  or  activated   charcoal.   Contamination  and  other problems  are more
serious  in low-pressure sampling  than  in  high-pressure  sampling systems
(National Academy of Sciences, 1978).
     Water  samples are subject  to the same  possibility of contamination and
other problems  that  exist in  air sampling.   DCM aqueous samples must be care-
fully sampled,  transported,  and stored  because of  DCM's volatility and the
                                     3-17

-------
  complexity  of the  samples,  especially those  containing chlorine or other
  oxidants.  A technique that is often used involves filling and sealing a serum
  bottle without  air space and storing it just above freezing (Kopfler et al.,
  1976).  Water samples generally require additional preparation before analysis.
  Normally, such samples may be concentrated by various water analysis techniques,
  but direct aqueous injection is used occasionally in GC analysis.
  3-3.2.1  Sampling  and Detection in Ambient Air—Several  common approaches are
  used to  sample  ambient  air for trace gas  analysis,  including the following
  approaches (National Academy of Sciences,  1978).
      1.
      3.
      4.
 Pump-pressure  samples.  A mechanical pump  is  used to fill a stainless
 steel  or glass container to  a positive pressure relative to  the
 surrounding atmosphere.

 Ambient pressure samples:  An evacuated chamber is opened and allowed
 to  fill  until  it  has  reached  ambient pressure at  the sampling  loca-
 tion.   If filling is  conducted  at high altitude, the sample  may
 become contaminated upon return to ground  level.
 Adsorption on molecular sieves, activated  charcoal, or other sorbents.
 Cryogenic samples:  Air  is  pumped into a  container and  liquefied,
 and the partial vacuum that is created allows  more  air  to enter.
This method allows  collection of  several  thousand liters  of  air.
      Singh et al.  (1979)  have  satisfactorily measured  ambient  levels of DCM by
analysis  with GC-EC.  Samples were  pressurized in stainless  steel vessels,
then  preconcentrated by freezeout on 100/120 mesh glass beads  (use of Tenaxฎ
monomer was  discontinued because oxygen oxidized  the  monomer  and interfered
with  EC).  Separation was performed  on a column containing 0.2  percent Chromo-
sorb  W 1500 80/100 mesh on Carbopack C.. A post-column Ascarite water trap was
used  to  remove  water before  EC.   Dual  detectors were  used  to provide a  coulo-
metric response.
      Harsch et  al.  (1979)  used GC-EC to  identify and  measure the  level  of  DCM
in  samples  of ambient air.  A 500-ml  sample  was  preconcentrated  using the
freezeout concentration  method (Rasmussen et al.,  1979).   Halocarbons  were
desorbed onto a stainless steel column packed with 10 percent SF-96 on  100/120
mesh  Chromosorb W.   The reported  detection limit was 26 ppt.   Good separation
from chlorofluorocarbon 113 (trichlorotrifluoroethane) was reported.
                                     3-18

-------
     Cronn and Harsch  (1979)  reported a GC-MS detection limit of 6 ppt for a
500-ml aliquot of DCM.   Cronn et al.  (1977) also reported a detection limit of
20 ppt for  a  100-ml  aliquot.   Pressurized air  samples  were separated on a
column of Durapack n-octane  on 100/120 mesh  Porasil C.  Cronn et al.  (1976)
have  compared  GC-MS  with GC-EC in terms  of  precision  and sensitivity.  In
general,  GC-MS offered  great  specificity but could not equal GC-EC in repro-
ducibility for the 11 halocarbons studied.  For MS the detection limit for DCM
was 9 ppt with a percent standard deviation of 13.  The detection limit with
temperature-programmed GC-EC was 4 ppt and the  percent  standard  deviation was
7.3.
     Pellizzari and  Bunch (1979) reported an  estimated  detection limit of 200
ppt using a high-resolution GC-MS  system  in which  DCM was  first  adsorbed onto
       ฎ
a  Tenax   GC.   The accuracy of analysis  was  reported as ฑ30 percent.   The
inherent analytical  errors are a function of several factors, including the
following:   (1) the  ability  to accurately determine the breakthrough volume;
(2) the accurate measurement of the ambient air volume sampled;  (3) the percent
recovery of DCM from the sampling cartridge after a period of storage; and (4)
                                                         ฎ
the reproducibility  of  thermal  desorption from the Tenax   cartridge and its
                                                                 ฎ
introduction into the analytical system.  Oxidation of  the Tenax monomer was
not reported.   Singh et  al.  (1982)  have cautioned that the adsorption of
                               ฎ
halogenated compounds on  Tenax  may  not  reliably  reflect  ambient air  levels
because measurements by  some  investigators have been reported  as  less than
background (100 to 200 ppt).
     Difficulties in using a coulometric  approach  to the GC-EC quantification
of  DCM in  air samples  have been reported by  Lillian and Singh (1974).  These
investigators  were unable to  accurately measure DCM with detectors in series
because of  a  greater-than-coulometric  response.  Dichloromethane was  reported
to  have  a  very low  ionization  efficiency.   The observed response might be
attributed to  the  products  of ionization having greater electron affinities
than the reactants.
     Cox et al.  (1976)  reported that polyglycol stationary-phase chemically
bonded to porous glass (Durasil Low Kl) was the only material found to separate
DCM from other halocarbons  during a GC-EC analysis.  Satisfactory separation
and analysis  of  DCM  by GC-MS was  reported  by Grimsrud and  Rasmussen  (1975)
with a 50-ft SCOT OV-101 column.
                                     3-19

-------
     When  the  freezeout concentration method of  Rasmussen  et al.  (1979)  was
applied  to a 500-ml aliquot of  air,  the detection limit for DCM with GC-EC
analysis was 4 ppt  and the percent standard deviation was 26.2.  The GC column
contained  10 percent SF-96 on 100/120 mesh Chromosorb W.
     The National  Institute  for Occupational  Safety and Health (NIOSH, 1974)
method P & CAM 127 is recommended for measurement of DCM in samples in which
the concentration  is  greater than u.05  mg  DCM  per sample.   This method uses
adsorption on charcoal followed by desorption with carbon disulfide.  Analysis
is made  by GC  with flame ionization  detection.   The mean relative standard
deviation  of the method is" 8 percent.  Otson  et al .  (1983)  reported that  this
method is  suitable, under pertain conditions,  for the simultaneous and quanti-
tative determination  of DCM and seven other  chemicals, at and below  their
respective 8-hr TLVฎ values.
     Grimsrud  and  Miller (1979) have reported  an improved GC-EC method  in
which the  detector  response  to DCM is enhanced by the  addition of  02  in  the
carrier gas.   At the  highest 02 doping  (5 ppth) the response of the detector
to DCM (960  ppb) was enhanced 57-fold.   The enhancement is  depicted in Figure
3-1.   A constant-current electron-capture detector was  used.
3.3.2.2  Sampling and Detection  in Water — A gas purging and trapping  method
suitable for DCM has  been described by  Bellar  et al . (1979).  Water samples
are purged by  bubbling  with  helium or nitrogen at 23ฐC.  The halocarbons are
adsorbed onto  a porous  polymer trap as the gas is vented.  Quantification is
made by GC-MS.  Tenax   GC (60/80 mesh) was considered  an effective adsorbent
for compounds  that  boil  above approximately 30ฐC.   A recommended general  pur-
pose column  is an  8-ft  by 0.1-in. inside  diameter stainless steel or glass
tube packed with 0.2  percent Carbowaxฎ 1500 on Carbopackฎ - C (80/100 mesh).
For a sample volume of 5 ml,  the range of the limit of  detection is 0.1 to 1.0
3.3.2.2.1  Sample preservation (water).  Bellar et al. (1979) recommended that
water samples be stored in narrow mouth glass vials.   Vials are filled to zero
                                     @
head space and  covered with a Teflon -faced  silicone  rubber septum.   Screw
caps are suitable seals.  The presence of chlorine in water samples results in
an increase  in  the  concentration of certain halomethanes (not including DCM)
upon storage.
3.3.2.2.2  Soil  and sediment (water).   Dichloromethane  has been  found  in
drinking and surface water at a variety of locations in the United States.   In
                                     3-20

-------
                      60
                  iu
                  tu
                  c
                  IU
                  cc
                      40
                               12345

                                 O2 CONCENTRATION, ppth
Figure 3-1.  The effect of oxygen  doping  of  the  carrier  gas  on the ECD response
             to several halogenated methanes  at  a  detector temperature of 300ฐC.


Source:  Grimsrud and Miller,  1979.
                                    3-21

-------
a recent  National  Academy of Sciences report (1977), DCM formation resulting
from chlorination treatment of water was reported.
     In a survey for volatile organics in five drinking water supplies, Coleman
et al.  (1976)  found that DCM was  common  to all  the cities evaluated (i.e.,
Cincinnati, Miami,  Philadelphia, and  Ottumwa, Iowa).  Concentrations were not
reported.   Analysis was performed using GC-MS.
     In a 1975 survey by the U.S. Environmental  Protection Agency (EPA) (1975a),
DCM was detected  in 9  of  10 water  supplies.   Lawrence, Massachusetts,  had the
highest concentration (1.6 ug/1).  A mean concentration of less than 1 ug/1  in
finished  water  was  reported  in  a  survey of Region  V water  supplies (U.S.
Environmental Protection  Agency, 1975b).   This  survey indicated 8 percent of
the finished-water supplies contained detectable DCM.
     Dichloromethane was not among the major halogenated hydrocarbons detected
by Dowty et al. (1975a) in New Orleans drinking water.   In another report, DCM
was detected in  finished  waters in the New Orleans  area by GC-MS (Dowty et
al., 1975a), and  it was also found in Mississippi river clarifier effluent.
The chlorocarbon was sorbed  onto poly p-2,6-diphenyl phenylene oxide  (35/60
mesh).  Vapors  were desorbed  onto a capillary  chromatographic  column and
quantitated by MS.   Raw water  influent was  purified  after clarifier  treatment
(sedimentation and  some chemical treatment) to  an  extent that DCM  concen-
tration dropped  32  percent.   However,  the  concentration in finished water
increased after chlorination.   Dichloromethane was also  detected  in  commerci-
ally bottled artesian well water.
     A slight  increase  in DCM  concentration in chlorinated finished  water was
observed  by Bellar  et al.  (1974).   Finished water  had  2.0 ug/1  DCM after a
complete  treatment  process of raw  river water containing no detectable DCM.
Water samples were also collected from various locations in a sewage treatment
plant.  Before treatment, the water contained 2.36 ppb DCM (8.2 M9/1)-   Before
chlorination and  after  preliminary treatment, the water  contained 0.8  ppb DCM
(2.9 ug/1).  After  chlorination, the  effluent contained  1 ppb DCM (3.4 ug/1).
These tests show  that  DCM and  other chlorocarbons may have formed as a result
of the  chlorination treatment.   The most  notable  chlorocarbon was chloroform,
for which an increase of 7.1 ug/1 to 12.1 ug/1 was observed after chlorination.
GC-MS analysis  was performed using a headspace  preconcentration  technique.
     Dichloromethane was  detected  at  32 of  204 surface water sites from which
samples were collected  (Ewing  et al., 1977).  Sites  were located  near  heavily
                                     3-22

-------
industrialized river  basins across the United  States.   Concentrations were
reported as greater than  1 ppb (1 ug/1).   Samples  were collected from July
1975 to December  1976.   Of the 204 sites,  91  were  near major rivers and 57
were in tidal  areas  and estuaries.  Samples (125 ml)  were held at 60ฐC and
                                            ฎ
stripped.   Volatiles were  sorbed  onto Tenax  GC and desorbed onto Carbowax
1500 columns and analyzed by MS.
     Dichloromethane was not  among the contaminants detected  by Sheldon and
Hites (1978)  in  raw  Delaware River water collected from August 1976 to March
1977.
     Pellizzari and Bunch  (1979)  reported DCM in untreated Mississippi River
water (Jefferson  Parish,  Louisiana)  at a mean  concentration  of 2.581 ug/1.
The  highest  value reported  was  15.8 ug/1.  Determinations were  made from
February 7 to  August 5, 1977.  A mean  concentration of  0.13 ug/1 was  reported
by Pellizzari and Bunch (1979) in tap water from Jefferson Parish.   The highest
level was 1.1 ug/1.
     Data for  DCM concentrations  in general ambient waters  in EPA's Storet
files,  covering  the period  January  1978 to April  15,  1981,  indicate that
levels ranged from 0 to 120 ug/1  (U.S. Environmental Protection Agency, 1981).
Sediment sampling  detected DCM in 60 of 118 cases.   Concentrations were from
427 to 433 ppb.  Ambient soil concentrations of DCM are unknown (U.S. Environ-
mental Protection Agency, 1981).
     Singh et al. (1983) have detected DCM in seawater samples from the eastern
Pacific Ocean.  Mean surface concentrations of 2 ng/1 were measured.
3.4  ECOLOGICAL EFFECTS
3.4.1  Effects on Aquatic Organisms
     Dichloromethane  has  been tested  for' acute toxicity in at  least  seven
aquatic  species.   The information in  this  section  focuses  upon  observed DCM
concentrations that were reported to result in  adverse effects under laboratory
conditions.  Such parameters of toxicity are not easily extrapolated to  environ-
mental situations.   Test  populations may not be representative  of the entire
species  because  susceptibility to the test substance at different lifestages
may vary considerably.  Guidelines for the  use  of these data in  the development
of criteria  levels for DCM in water  are discussed in the Ambient Water Quality
Criteria for Halomethanes (U.S. Environmental Protection Agency, 1980).
                                     3-23

-------
       The toxicity  of DCM to fish and other aquatic organisms has been gauged
  principally  by  flow-through and  static testing methods.  The  flow-through
  method continuously exposes the organism(s) to a constant concentration of DCM
  while oxygen  is  continuously  replenished and waste products are removed.  A
  static test  exposes  the  organism(s) to the added initial concentration only.
  Results from  both  types  of tests are commonly used as initial  indications of
  the potential of substances to cause adverse effects.
  3'4<1-1  Effects on  Freshwater Species-Results  of flow-through  and acute
  static tests  with  DCM and freshwater species  (fish  and invertebrates) are
  shown in  Table 3-7.
       Alexander et al. (1978) used both flow-through and static methods to in-
  vestigate the acute toxicity of  several  chlorinated  solvents, including  DCM,
  to  adult fathead minnows  (Pimephales  promelas).   Studies were conducted  in'
  accordance  with  EPA  procedures  described by  the Committee on Methods for
  Toxicity  Tests with Aquatic Organisms (1975).   Alexander et al.  reported  that
  the  fish  were observed for the  following effects:   loss of equilibrium, mela-
  nization, narcosis,  and swollen,  hemorrhaging gills.   The  DCM concentration
  that  produced  one  or more of these  observable  effects in 50 percent of the
  fish  (EC50) was  99.0 mg/1 for DCM.   Fish affected during the exposure were
  transferred to static  freshwater  aquaria at the  end of the 96-hour exposure
 period.  Only  the  fish that were severely affected by high concentrations of
 the  chemical  did not  recover.   Short exposures to these  compounds at the
 sublethal  level seem to produce only reversible effects.
      Using flow-through procedures,  Black et al.  (1982) determined the aquatic
 toxicity of DCM to embryo-larval stages of fish and amphibians.   Exposure was
 initiated  within 30  min  of  fertilization for  fathead minnow (Pimephales
 firpmelas),  rainbow  trout  (Salmo  gairdneri).  leopard frog (Rana  pipiens),
 northwestern salamander (Ambystoma gracile),  European common frog  (Rana'
 temporaria),  and  the  African clawed  frog (Xenopus  laevis).   Control and
 experimental  eggs were  simultaneously cultured under  identical  conditions.
 Sample size  ranged from 50 to 125 eggs per exposure chamber.  Evaporation  of
 DCM was  minimized by use  of  test  chambers devoid of an air-water interface.
 Survival of  normal  rainbow trout larvae  at  4 days posthatching was  100, 85,
44, and 9% at exposure  levels of 0.008, 0.41, 23.1, and  36.5 mg/1, respectively.
Teratic larvae  were  observed only at exposure concentrations of 5.55 mg/1  or
greater.   The  order of  increasing species tolerance was  S.  gairdneri, R.
temporaria, A.  sracile,  P.  promelas,  X. laevis,  and R.  pipiens.   Respective
                                     3-24

-------











to
to
1 J 1
L4.I
'


ฃ
o

on

n
O5


ซN
O
m
o
LU




re
ฃ—
Cn
U.

•0

O
^




-P
in
OJ
^_>

4-
O
0)
Q.

1—














to
CD
•i—
O
OJ
Q.
to






CO
en
i-l CO CO
^ 3 S
^*^ i * r^i
"re c.
+J O) • •
1= O) i— r—

S- •!-
•i- +i S_ S-
> u  C C
o re re
• S- X X
to Q- O) Oป
— ^ ^^> ^^ซ



C_3
1 1 1










00 O
o o o
00 0
*t *> V,
fj- CD CO
c\j I-H cn
C\] CO rH




x:
cn
3
0
i.
<•"
U U -P
-P -P 3:
re re o
•p -p ^~
in in 4—


^
3
i.
•i— in in
x: O) oi
U i — r—
o re re
o o. o.
re o>  — <
o ^ 3s
Q. O O
 c c:
— i e s~* c /-
>— ' ?r— W -I— I
E re E r
!_ป. ,_ j_
r— -o  u
C O)
LU -P
o

to a.
^D

CD
CD
CD
#<
•=1-
CM
C\J










1













O
-p
re
4J
in














re
c
-> cn
o re
0 E=

; re

5 c

2. QL
re
Q


































































-------
 LC50 values were 13.1,  16.9,  17.8,  -x-34,  >29,  and >48 mg/1.  LCX  and LC10 values
 could only be calculated  for R.  temporari'a.   The respective values were 69.9
 and 822.4 mg/1.   The LCฑ  and LC10  values suggested to the  investigators that
 developmental  stages of certain  amphibian  species may be affected by concen-
 trations  of 100 |jg/l and  that concentrations at or above 1 mg/1 may produce
 substantial  reproductive  impairment.
      Chronic test data concerning  life  cycle or embryo-larval  tests are not
 available  nor were  data  found  on  the  chronic  toxicity  to invertebrates.
 3.4.1.2  Effects on  Saltwater Species—Static tests with mysid  shrimp resulted
 in  an LCSO value of 256,000  ug/1.   No  data  exist  on  the chronic  effects of
 DCM.
      In a  96-hr  static  test with  Sheepshead minnow (Cyprinodon  variegatus)  the
 LC50  value was  331,000 [jg/1  (U.S.  Environmental  Protection Agency,  1980).

 3.4.2  Effects on Plants
     The 96-hr EC50 values for DCM, based on  chlorophyll a  and  cell numbers of
 the  freshwater  alga,  Selenastrum capricornutum,  were greater than  the highest
 test  concentration  (662,000  ng/1)-   The  96-hr EC50 value  based  on  chlorophyll
 a and  cell numbers  of  the saltwater  alga,  Skeletonema costatum, was  greater
 than the highest test concentration (662,000  pg/l)  (U.S. Environmental  Protec-
 tion Agency, 1980).
     Few studies of the effects on vascular plants  are  available (U.S.  Environ-
 mental Protection Agency, 1981).  Lehman and  Paech  (1972) tested the  effect of
 DCM vapors on the photosynthetic  fixation of  14C02  by alfalfa seedlings.  At a
 very high  concentration (21 percent), DCM reduced photosynthesis by 82  percent.

 3.4.3  Bioconcentration Potential
     Bioconcentration refers  to  increased concentration of  a substance in the
 tissue of  an organism (e.g.,  fish) relative to the  ambient water concentration
 under steady-state conditions.  A measure of  the potential for organic  chemicals
 to bioconcentrate in  the  fatty tissues  of  aquatic organisms is  given by the
 octanol/water partition coefficient  (Neely et a!., 1974).    The log  octanol/
water partition  coefficient for  DCM has  been  measured  at 1.25 (Hansch et a!.,
 1975), but no  steady-state bioconcentration  factor  (BCF) has been measured.
The BCF represents the  ratio  of  the chemical  concentration  in the  organism  to
that in the  water.   An  approximate BCF  for DCM  of 5.2 has  been calculated
                                     3-26

-------
using the relationship Log BCF = 0.76 Log P - 0.23 (Veith et a!., 1979).   This
estimated BCF places DCM in the low range of biconcentration values, suggesting
that the potential  for biconcentration  in  lipids is low.  However}  no infor-
mation is available  concerning DCM levels in  living organisms  (Pearson  and
McConnell,  1975) or the rate of transport of DCM through food chains.
     However, the BCF alone may not be the most useful measure of the overall
fate of a substance in water or its  potential  for  producing toxic effects.
Chemical  and biological degradation of the  substance, volatilization, desorp-
tion, and the  depuration  rate are among  the key  determinants of toxicity.
3.5  CRITERIA, REGULATIONS, AND STANDARDS
     Permissible levels of  DCM in the working environment  have  been estab-
lished in various countries.  The OSHA health standard requires that a worker's
exposure to DCM  at  no time exceed 500 ppm (1,737 mg/m3) TWA in any 8-hr work
day of a 40-hr  week,  with an  acceptable  ceiling concentration of 1000 ppm
(3,474 mg/m3), that should  not exceed 2000 ppm (6,948 mg/m3) for more than 5
minutes in any 2-hr period.   The American Conference of Governmental Industrial
Hygienists (ACGIH) TLV  for inhalation exposure [200 ppm (695 mg/m3)], proposed
for prevention of narcotic  effects and for protection against excessive COHb
formation,  has recently been  lowered to 100 ppm (347  mg/m3).   The 8-hr TWA
value in the  Federal  Republic  of  Germany  is 100  ppm; in the German  Democratic
Republic and  Czechoslovakia  it is 144 ppm, and in Sweden it is 100 ppm.  The
acceptable ceiling concentration  in  the USSR is  14 ppm  (49 mg/m3).   NIOSH  has
recommended that occupational  exposure  to DCM not exceed 75 ppm (261 mg/m3),
determined as  a TWA for up to a 10-hr work day of a 40-hr week, in the absence
of exposure to  carbon monoxide above a TWA of 9 ppm for up to a 10-hr work
day.
     The Ambient  Water Quality Criteria  for  Halomethanes (U.S.  EPA, 1980)
indicates that for the maximum protection of human health from the potential
carcinogenic  effects  caused  by exposure to any  of several halomethanes (in-
cluding DCM, chloromethane, bromomethane, bromodichloromethane, tribromomethane,
dichlorodifluoromethane, and/or trichlorofluoromethane)  through  ingestion of
contaminated  water  and contaminated  aquatic  organisms,  the  ambient water
concentration  should be zero based on the assumption of no threshold for these
chemicals.   However,  zero  level  may not be attainable; therefore, the levels
                                     3-27

-------
that may  result  in  incremental  increase of cancer risk over the lifetime are
estimated at 10-5,  10-6,  and 10-7.  The corresponding  recommended criteria,
based on data for chloroform, are 1.9, 0.19, and 0.019 [jg/1, respectively.   If
the above estimates are made for consumption of aquatic organisms only, exclud-
ing consumption of water, the levels are 157, 15.7, and 1.57 jjg/1>  respectively.
Estimates for water consumption only were not made.
     For halomethanes where  one  criterion  is derived for  an entire class of
compounds, the U.S.  EPA does not state that each  chemical  in the class  is a
carcinogen.   The  intended interpretation of the criterion  is that the  risk is
less than 10 5 whenever the total concentration of all  halomethanes in water
is less than the criterion.   In a  hypothetical case  where all  of the halo-
methanes in a sample are non-carcinogenic, the criterion would  be too  strict.
In most cases where halomethanes are detected, a mixture of compounds occurs
and in calculation of the criterion, the assumption is made that all  components
have the same carcinogenic potency as chloroform.
     The derived  water  quality criterion, based on non-carcinogenic risks and
assuming a daily  water  intake of  2  1  and consumption  of 6.5 grams of fish and
shellfish per day (bioconcentration factor 0.91) would be 12.4 mg/1.
                                     3-28

-------
3.6  REFERENCES
Ahlstrom, R. C. and J. M. Steele.  1979.  Methylene chloride.  In:    Kirk-Othmer
     Encyclopedia of .Chemical Technology.   A.  Standen,  ed.,  Interscience Pub-
     lishers, New York, NY.

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

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

American National Standards Institute, 1970.

Anthony, T.   1979.   Chlorocarbons  and  chlorohydrocarbons.  In:   Kirk-Othmer1s
     Encyclopedia of Chemical Technology.  Wiley-Interscience.

Bellar, T.  A.,  J. J.  Lichtenberg,  and  R.  C.  Kroner.   1974.   The occurrence of
     organohalides  in  chlorinated  drinking waters.   J.  Am.  Water Works Assoc.
     703-706.

Bel Tar, T. A., W. L. Budde, and J.  W.  Eichelberger.   1979.   The  identification
     and measurement  of  volatile organic compounds  in  aqueous environmental
     samples.   Chapter 4.  In:  Monitoring  Toxic  Substances, ACS  Symposium
     Series 94.  D.  Schuetzle, ed., American Chemical Society.

Black, J.  A.,  W. J.  Birge,  W.  E. McDonnell,  A. G.  Westerman, B.  A. Ramey, and
     D. M. Bruser.   1982.  The Aquatic Toxicity  of Organic Compounds  to  Embryo-
     Larval Stages  of Fish and  Amphibians Res. Report No 133, Water Resources
     Institute, University of Kentucky.

Brodzinsky,  R. ,  and H. B.  Singh.   1983.  Volatile Organic Chemicals  in  the
     Atmosphere:  An  Assessment of Available Data.   EPA-600/S 3-83-027.   U.S.
     Environmental  Protection Agency,  June.

Brunner, W., and T.  Lei singer.  1978.  Bacterial degradation of  dichloromethane.
     Experentia 34:1671.

Brunner,  W.,  D. Staub,  and T.  Leisinger.  1980.  Bacterial  degradation of
     dichloromethane.  Appl. Environ. Microbiol.  40:950-958.

Butler,  R. ,  I.  J.  SolomorTT and  A.  Snelson.   1978.   Rate constants for the
     reaction  of OH with  halocarbons  in the  presence  of 02 + N2.   J.  Air
     Pollut. Control Assoc. 28:1131-1133.

Chemical  Marketing  Reporter,  February 1983.   Chemical Products Synopsis, June
     1984.

Chemical Products Synopsis, 1981.
                                      3-29

-------
Coleman, W.  E.,  R.  D. Lingg,  R.  G.  Melton, and  F.  C.  Kopfler.   1976.   The
     occurrence of volatile organics in five drinking water  supplies  using  gas
     chromatography/mass  spectrometry.   Chapter 21.  In:  Identification and
     Analysis of  Organic  Pollutants in Water.   L.  H.  Keith, ed. ,  Ann  Arbor
     Science, Ann Arbor, MI.

Committee on Methods  for Toxicity Tests with Aquatic Organisms.   1975.   Methods
     for acute toxicity  tests with fish, macroinvertebrates, and amphibians.
     Ecol.  Res. Ser.  EPA-660/3-75-009.  U.S.  Environmental  Protection  Agency.
Cox,  R.  A. ,  R.  C.
     Photochemical
     10:305-308.
                   Denwent,  A.  E.  J.  Eggleton,  and J.  E. Lovelock.   1976.
                   oxidation of halocarbons  in the  troposphere.   Atmos.  Environ.
Cronn, D. R., and D. E. Harsch.  1979.  Determination  of atmospheric  halocarbon
     concentrations  by gas chromatography-mass  spectrometry.  Anal.  Lett.
     12(614):1489-1496.

Cronn, D. R., and E. Robinson.  1979.  Determination of trace  gases in  Learjet
     and U-2 whole-air samples collected  during  the intertropical  convergence
     zone study.   Report  submitted to NASA,  August 1978.   In:  1977 Inter-
     tropical Convergence  Zone Experiment.   I. G.  Poppoff,  W.  A.  Page,  and A.
     P. Margozzi, NASA TMX78577.

Cronn, D. R.,  R.  A. Rasmussen, and  E.  Robinson.   1976.   Report for Phase I.
     Measurement of  Tropospheric Halocarbons by  Gas Chromatography-Mass Spec-
     trometry.   Report  prepared for  the U.S.  Environmental  Protection Agency,
     August.

Cronn, D. R.,  R.  A. Rasmussen, and E. Robinson.   1977.  Report for Phase  II.
     Measurement of  Tropospheric Halocarbons by  Gas Chromatography/Mass Spec-
     trometry.   Report  prepared for  the U.S.  Environmental  Protection Agency,
     October.

Crutzen, P.  J.,  and J. Fishman.  1977.  Average concentrations of OH-  in  the
     troposphere and the  budgets  of CH4, CO, H2, and  CH3CC13.  Geophys.  Res.
     Lett.  4:321-324.

Davis, D. D., G. Machado, B. Conaway, Y. Oh,  and R. Watson.   1976.  A temperature-
     dependent kinetics study of the reaction of OH with CH3C1, CH2C12,  CHC13,
     adn CH3.  Br.  J. Chem. Phys.  65(4):1268-1274.

De Forest,   E. M.   1979.   Chloromethanes.   In:  Encyclopedia  of Chemical  Pro-
     cessing and Design.   J.  J. McKetta and W.  A.  Cunningham, eds., Marcel
     Dekker, Inc.
                                                        II.   Evaporation rates
                                                        and  propylenes  from
                                                   theoretical  predictions.
Dilling, W.  L.   1977.   Interphase transfer processes.
     of  Chloromethanes,  ethanes,  ethylenes,  propanes
     dilute  aqueous  solutions.   Comparisons  with
     Environ. Sci. Techno!. 11(4):405-409.

Dilling, W.  L.   and  H.  K.  Goersch.   1980.   Dynamics,  Exposure, and  Hazard
     Assessment of Toxic Chemical.  R. Hague, ed.  Ann Arbor  Science  Publishers,
     Inc. Ann Arbor, Michigan, pg. 111.
                                     3-30

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

 Dilling,  W.  L. , C.  J.  Bredeweg,  and N. B.  Terfertiller.   1976.  Simulated
      atmospheric photodecomposition rates  of  methylene chloride, 1,1,1-tri-
      chloroethane,  trichloroethylene,  tetrachloroethylene,  and other compounds.
      Environ.  Sci.  Techno!.  10(4):351-356.

 Dowty, B. J.,  D. R.  Carlisle,  and J.  L. Laseter.   1975a.   New Orleans drinking
      water  sources tested by  gas chromatography/mass  spectrometry.   Environ.
      Sci. Techno!.  9(8):762-765.

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

 Edwards,  P.  R. ,  I. Campbell,  and G.  S. Milne.  1982.   The impact of chloro-
      methanes  on the environment.   Part 2.   Chemistry  and  Industry,  4 September,
      pp.   619-622.

 Ewing, B. B.,  E. S.  K.  Chian,  J.  C. Cook,  C.  A.  Evans, P.  K.  Hopke, and E. G.
      Perkins.  1977.   Monitoring  to Detect Previously Unrecognized Pollutants
      in  Surface Waters.   EPA-560/6-77-015, U.S.  Environmental  Protection
      Agency, July.

 Fells, I., and E. A. Moelwyn-Hughes.  1958.  The  kinetics of  the hydrolysis  of
      methylene chloride.  J. Chem.  Soc.  (London)  1326-1333.

Grimsrud, E. P., and D. A. Miller.  1979.   A new  approach to  the trace  analysis
      of mono-  and  di-halogenated  organics, an analysis of methyl  chloride in
      the  atmosphere.   In:   National Bureau of Standards  Special Publication
      519, Trace  Organic Analysis:   A  New  Frontier in Analytical  Chemistry,
      Proceedings of  the 9th Materials  Research Symposium, April  10-13,  1978.
      Issued April,  pp. 143-151.

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

Hansch, C. ,  A. Vittoria,  C.  Silipo, and P.V.C. Jow.  1975.   Partition coeffi-
      cient and the  structure-activity  relationships of the anesthetic  gases
      J. Med. Chem.  18(6):546-548.
Hardie, D. W.  F.   1969.   Methylene Chloride. In:
     of Chemical Technology. Vol 5., pp. 111-119.
Kirk-Othmer's Encyclopedia
Harsch, D. E. ,  D.  R.  Cronn, and W. R. Slater.  1979.
     genated hydrocarbons measurable  in ambient air.
     Assoc.  29(9):975-976.
    Expanded list of halo-
    J.  Air Pollut.  Control
Howard, C. J., and K. M.  Evenson.   1976.   Rate  constants  for the reactions of
     OH with  ethane  and some halogen substituted  ethanes at 296K.   J.  Chem.
     Phys. 64:4303.
                                     3-31

-------
 Klecka,  G. M.   1982.   Fate and effects  of  methylene chloride in activated
      sludge.   Appl.  Environ.  Microbiol.  44:701-707.

 Kopfler,  F.  C.,  R.  G.  Melton, R.  D.  Lingg,  and W.  E.  Colman.   1976.   Identifi-
      cation  and analysis of  organic  pollutants in water.   L.  Keith, ed. ,  Ann
      Arbor Science  Publishers,  Inc.,  Ann Arbor, MI,  pp.  87-104.

 LaPat-Polasko,  L.  T. , P. L.  McCarty, and A.  J. B.  Zehnder.   1984.   Secondary
      substrate  utilization of  methylene chloride by  an isolated strain of
      Pseudomonas sp.  Appl.  Environ.  Microbiol.  47(4):825-830.

 Lehmann,  J.  and  D.  Paech.   1972.   Effect of some vaporized lipophilic solvents
      on carbon  dioxide fixation.   Experientia  28(12):1415-1416.

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

 Lovelock,  J.  E.   1974.  The  electron capture detector:   Theory  and  practice.
      J. Chromatogr.  99:3-12.

 Lowenheim, F.  A., and M. K.  Moran.   1975.   Methylene Chloride.   In:  Faith,
      Keyes,  and  Clark's Industrial Chemicals, Fourth Edition, John Wiley, New
      York, pp. 530-538.

 McKetta,  J.  J.  and W. A. Cunningham, eds.   1979.  Encyclopedia  of  Chemical
      Processing  and  Design,   pp. 267.

 National  Academy of Sciences.  1977.   Drinking Water and Health.   National
      Research Council, pp.  743-745.

 National  Academy of  Sciences.   1978.   Non-fluorinated  halomethanes in the
      environment.   Panel on  low-molecular-weight halogenated hydrocarbons.
      Coordinating  Committee  for   Scientific  and Technical Assessments  of
      Environmental Pollutants.

 National  Aeronautics and Space Administration.   1977.   Chlorofluoromethanes
      and  the  Stratosphere.   Robert D. Hudson,  ed.   National  Aeronautics and
      Space Administration Reference Publication 1010.

 National  Bureau  of  Standards.  1978.  Special  Publication 513 "Reaction  rate
      and photochemical data for atmospheric chemistry,"  R. F. Hampson,  Jr.  and
      D. Garvin  (eds.).   National   Bureau  of  Standards,  Washington,  DC, 1978.

National  Institute  for Occupational  Safety and Health  (NIOSH).   1974.  NIOSH
     Manual of Analytical Methods, HEW Publication No.  (NIOSH) 75-121.

Neely, W.B.,  D;B.  Branson,  and G.E.  Blan.   1974.   Partition coefficient to
     measure bioconcentration potential  of organic chemicals  in fish.   Environ.
     Sci.  Techno!. 8(13):1113-1115.

Otson, R., D. T. Williams, and P.  D.  Bothwell.   1983.  Charcoal tube technique
     for simultaneous determination of selected  organics in air.  Am.  Ind. Hyg.
     Assoc. J. 44(7):489-494.
                                     3-32

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

Pellizzari, E. D.   1974.   Electron capture detection  in gas chromatography.
     J.  Chromatogr.  98:323-361, 1974.
Pellizzari, E. 0.  1977.  Analysis  of  Organic  Air Pollutants
     graphy and  Mass Spectroscopy.   EPA-600/2-77-100.   U.S.
     Protection Agency.
                                by Gas Chromato-
                                 Environmental
Pellizzari,  E.  D.   1978a.   Measurement of  Carcinogenic Vapors in Ambient
     Atmospheres.  EPA-600/7-78-062.   U.S.  Environmental  Protection  Agency.

Pellizzari,  E.  D.   1978b.   Quantification  of  Chlorinated Hydrocarbons  in
     Previously  Collected Air  Samples.   EPA-450/3-78-112.   U.S.  Environmental
     Protection Agency.
Pellizzari, E. D.  and J.  E.
     improved  Sampling  and
     EPA-600/2-79-081, May.
Bunch.  1979.  Ambient Air Carcinogenic Vapors:
Analytical  Techniques  and  Field  Studies.
Pellizzari, E. D.,  M.  D.  Erickson, and R. A. Zweidinger.  1979.   Formulation
     of a  Preliminary  Assessment of Halogenated Organic Compounds in Man  and
     Environmental Media.   EPA-560/13-179-006.   U.S.  Environmental Protection
     Agency.

Perry, R.  A.,  R.  Atkinson,  and  J.  N.  Pitts.  1976.  Rate constants for the
     reaction of  OH  radicals  with CHFC12  and CH3C1  over the  temperature range
     298-493K and with CH2C12 at  298K.  J. Chem. Phys.  64:1618.

Radding, S. B. ,  D.  H.  Liv, H. L. Johnson, and T. Mill.  1977.   Review  of  the
     environmental fate of selected chemicals.  EPA-560/5-77-003,  U.S.  Environ-
     mental Protection Agency.

Rasmussen,  R. A., D. E. Harsch,  P.  H.  Sweany, J.  P.  Krasnec, and D. R.  Cronn.
     1979.  Determination  of atmospheric  halocarbons  by a  temperature pro-
     grammed  gas chromatographic freezeout  concentration method.  J.  Air
     Pollut. Control Assoc. 27:579.

Rittman, B. E.  and  P.  L.  McCarty.   1980.  Utilization of dichloromethane by
     suspended  and   fixed-film  bacteria.   Appl.  Environ.  Microbiol.   39:
     1225-1226.
Robinson, E.   1978.   Analysis' of Halocarbons  in Antarctica.
     prepared for the National Science Foundation,,  December.
                                 Report 78/13-42
Sheldon, L.  S.ป  and R. A.  Hites.   1978.   Organic compounds  in  the  Delaware
     River.  Environ.  Sci.  Techno!. 12(10):1188-1194.

Singh,  H.  B.   Atmospheric  halocarbons.   1977.   Evidence in favor of reduced
     average  hydroxyl  radical  concentrations  in the troposphere.   Geophys.
     Res.  Lett. 4(3):101-104.
                                      3-33

-------
  Singh,  H.  B., L.  J.  Salas,  and R.  E.  Stiles.
       genated chemicals in the  air  and oceanic
       88:3675-3683.
   1983.  Selected man-made halo-
   environment.   J.  Geophy.  Res.
  Singh,  H.  B.,  L.  J.  Salas,  A.  J.  Smith,  and
       of some potentially hazardous  organic
       Atmos.  Environ.  15:601-612.
H. Shigeishi.  1981.  Measurements
chemicals in  urban  environments.
 Singh,  H.  B.,  L. J.  Salas,  R.  Stiles, and H. Shigeishi.  1980.  Atmospheric
      Measurements of  Selected Hazardous  Organic  Chemicals.   Second Year Interim
      Report.   Report  prepared for the U.S.  Environmental Protection Agency by
      SRI International, Menlo Park,  California.                       *   y  y

 Singh,  H.  B.   L  J.  Salas,  H. Shigeishi, A.  J.  Smith,  E.  Scribner,  and L.  A.
      Lavanagh.    1979.  Atmospheric distributions,  sources  and  sinks  of  selected
      cnhฃ-ar Sn+' JydrฐCcarb?nS.'  SFe> and N2ฐ"  EPA-600/3-79-107, Final  report
      submitted to the U.S. Environmental Protection Agency by  SRI  International
      November.

 Singh,  H.  B.,  L. J. Salas, and R.  E.  Stiles.   1982.   Distribution  of selected
      gaseous organic mutagens and suspect carcinogens in ambient air.   Environ
      Sci. Techno!.  16(12):872-880.

 Systems Applications,  Inc.  1983.  Human Exposure to Atmospheric Concentrations
      of Selected Chemicals,  Vol   II,  as  amended  by  the U.S.  Environmental
      Protection Agency to reflect public comments.  EPA contract no. 68-02-3066
      National  Technical  Information  Service,  Springfield,  VA, PB83-265249.

 Spence, J.  W.  P  L._ Hanst,  and  B.  W.  Gay,  Jr.  1976.  Atmospheric oxidation
      of methyl  chloride,  methylene chloride,  and chloroform.  J. Air Pollut
      Control  Assoc.  26(10):994-996.

 SRI  International   1980.   Dichloromethane.   In:   Chemical  Economics Handbook
      SRI International,  Menlo Park,  CA.

 U.S.  Environmental  Protection  Agency.   1975a.   Preliminary assessment of
U.S.  Environmental  Protection Agency.   1975b.   Region V Joint Federal/State
      Survey  of Organics and  Inorganics  in  Selected Drinking Water Supplies.

U.S.  Environmental  Protection Agency.   1980.   Ambient Water Quality  Criteria
      for  Halomethanes.   U.S.  EPA.   Available from:  National Technical Infor-
      mation Service, Springfield, VA.   (NTIS  PB81-117624).

U.S.  Environmental Protection Agency.   1981.   Environmental  Risk  Assessment of
      Dichloromethane.  Draft  report.  Office  of Toxic  Substances,  14  September.

U.S.  International  Trade  Commission.   1980.    Synthetic  Organic  Chemicals.
     United States Production & Sales, 1979.  Washington, DC.

U.S.  International  Trade  Commission.   1982.    Synthetic  Organic  Chemicals.
     United States Production and Sales, 1981.  Washington,  DC.
                                     3-34

-------
Veith, G.D.,  K.J.  Maule, and C.J. Petrocelli.   1979.   An  evaluation of using
     partition coefficients  and water solubility to estimate bioconcentration
     factors  for  organic chemicals in fish.   In:  Aquatic Toxicology.  J. G.
     Eaton, P. R.  Parrish, A. C. Hendricks, eds.   American Society  for  Testing
     and Materials, pp 116-129.

Wood, P.  R., F. Z. Parsons, J. DeMarco, H. J. Harween,  R..  F.  Lang,  I. L.  Payan
     and M. C.  Ruiz.   1981.   Introductory study of the biodegradation  of the
     chlorinated  methane,  ethane, and  ethene compounds.    Presented at the
     American Water Works Association Meeting, June.
                                    3-35

-------

-------
             4.   METABOLISM AND PHARMACOKINETICS OF DICHLOROMETHANE
     Dichloromethane is a colorless  liquid with a pleasant smell.  The odor
                                                            3
threshold has  been  reported to  be about  100 ppm  (347 mg/m ) (May, 1966;
                                              3
Leonardos et al. , 1969)  and 250 ppm (868  mg/m ) (Amoore and  Hautala, 1983).
Because of its relatively high  vapor pressure at room temperatures (350 to 500
torr), DCM is  readily  absorbed into the body following inhalation, and most
cases of  severe  poisoning  from this solvent occur from inhalation exposures.
Reports  of poisoning  in man by oral ingestion are  rare  (Llewellyn,  1966;
Stewart  and  Hake, 1976;  Friedlander et al. ,  1978),  and total recovery has
followed the swallowing  of  quite large doses (Roberts  and  Marshall,  1976).
     Dichloromethane has  been  used as a general  industrial  solvent for at
least  six  decades  (Lehmann and  Schmidt-Kehl, 1936),  and its narcotic and
anesthetic properties have  been  known  to clinical  medicine  for over 60 years
(Bourne  and  Stekle,  1923).   However, most of the  available  information on
DCM's metabolism and pharmacokinetics  is  derived from studies that were con-
ducted within  the  last 10 years  due to  the resurgence of  interest in DCM
following the demonstration of  its metabolism to CO (Stewart et al., 1972a,b).
These studies  in animals  and  humans have been carried out at relatively high
oral or inhalation concentrations (50 to 1500 ppm) of DCM.
4.1  ABSORPTION, DISTRIBUTION, AND PULMONARY ELIMINATION
4.1.1  Oral Absorption
     Absorption of  DCM  through the intestinal mucosa after ingestion appears
to be  rapid  and complete.   McKenna and Zempel (1981) obtained virtually com-
                                       14
plete  recovery  (92  to 96 percent) of   C-DCM  radioactivity in  urine,  feces,
and exhaled  air of  rats  after single oral  (gavage)  doses  of 1 or 50 mg/kg DCM
in water.  Pritchard  and Angelo (1982) observed  in B6C3F1 mice a peak blood
concentration  in  less than  10 minutes following an oral  gavage  dose of
50 mg/kg DCM in either water or corn oil.

4.1.2  Dermal Absorption
     Absorption of  DCM  through the skin from direct liquid contact or by the
immersion  of hands  or arms is a  slow  process.   Stewart and Dodd (1964) at-
tempted to quantify the rate of absorption through  human skin by immersing the
                                   4-1

-------
thumbs of volunteers  in  liquid DCM and then determining  the  appearance and
concentration of DCM in the breath.   An estimate of the amount of DCM entering
the body was made  by comparison of these measurements with breath concentra-
tions obtained following  controlled  inhalation exposures.  From these breath
analyses, Stewart and  Dodd concluded that DCM  is very slowly absorbed. They
also suggested that although the amount of DCM absorbed depends on the area of
the exposed skin, the  slow rate of absorption would prevent toxic quantities
of DCM from being taken into the body from direct contact with the skin of the
hands and forearms.   Immersion in DCM was found to be accompanied by an intense
burning sensation within a few minutes.

4.1.3  Pulmonary Uptake
     Inhaled DCM rapidly  equilibrates  across the alveolar endothelium because
of its water and lipid solubility and the very large lung alveolar surface
area.  DCM  is  appreciably more water soluble  (2  g/100  ml),  but less lipid
soluble  than its congeners, chloroform and carbon  tetrachloride;  its lipid
partition coefficient  (olive  oil/water)  is  180 at  37ฐC with a blood/air co-
efficient of  7.9 (Lehmann  and Schmidt-Kehl,   1936;  Morgan et al. ,  1972;
Lindqvist, 1978).
4.1.3.1  Studies in Humans--The magnitude of  DCM uptake into the body (dose,
burden) primarily depends  on  several parameters:  inspired air concentration,
pulmonary ventilation, duration of exposure,  the rates of diffusion into blood
and tissues, and solubility in blood and the various  tissues.  The concentra-
tion of DCM in alveolar air, in equilibrium with pulmonary venous blood content,
asymptotically approaches  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/nvin at rest) provide a means of calculating uptake during exposure:
                            = 
(4-1)
where Q is the quantity absorbed, C is the concentration (inspired and alveolar)
in mg/1, V is the alveolar ventilation rate in 1/min, and T is the duration of
                                   4-2

-------
 exposure  in  minutes.   The  percent retention  is  defined as  (C.     -  C -,  )/C-
                                                           v  insp     alvy   insp
 x 100,  and percent retention  x quantity inspired (V - T ซC.   ) is equal to
 uptake.
      Figure  4-1 (from Riley et al., 1966) illustrates the overall time course
 of absorption and elimination  during  a  2-hour  inhalation  exposure  of 100 ppm
 (347  mg/m )  DCM for a  70-kg man.   During exposure, the  alveolar air concentra-
 tion  of  DCM  can be described  by  an  exponentially  rising curve with  three
 components.   At the beginning  of  exposure,  an  initial rapid rate  of uptake
 occurs  [0 to 50 ppm  (0  to 174 mg/m ) alveolar air],  followed by  a  second
 slower  uptake [50 to  65  ppm (174 to 226  mg/m3)  alveolar air];  finally, a  very
 slow  rate as equilibrium  is approached  at  70 ppm (243 mg/m3) alveolar air
 concentration.   The total  quantity of DCM absorbed  and retained  in the body
 during  exposure is represented by  the area  between the alveolar and  inspired
 environmental air  concentration curves.   Complete equilibrium  or  steady-state
 conditions are  not attained by  the  end of the 2-hour exposure to DCM, as  shown
 by  the  slowly rising  alveolar  concentration  curve.   The three  uptake compart-
 ments of  the exponential alveolar curve  correspond to  equilibrium attained by
 first-order  passive diffusion  of DCM  from blood.  First,  DCM diffuses through
 a vessel-rich group (VRG)  of tissues with high  blood flow (VRG:  brain, heart,
 kidneys,  liver,  and endocrine  and  digestive  systems),  it  then diffuses more
 slowly  through  the lean  body mass  (muscle group  [MG]:  muscle and  skin)  and
 last through adipose tissues (fat group  [FG]).  With the termination of exposure,
 blood and alveolar air DCM concentrations decline in parallel in an exponential
 manner with  three  components  of pulmonary elimination  and desaturation from
 VRG, MG,  and FG body compartments.  The  VRG,  MG,  and FG groups are  mathemati-
 cally derived compartments  used for convenience in analysis.
     DiVincenzo  and Kaplan (1981a) obtained  serial  breath  excretion curves
 from four  to  six volunteers  experimentally exposed to 50, 100, 150,  and  200
ppm (174,  347,  521, and 695 mg/m  )  DCM for 7.5  hr under sedentary conditions.
 Pulmonary uptake was rapid during  the  first  hour  (exposure was interrupted at
4 hr, for 30 minutes).  During  the  post-exposure period, DCM levels decreased
rapidly (i.e., exponentially).   Less than 0.1 ppm (0.347 mg/m3) DCM  was detec-
ted in  the end  tidal  air  of the  individuals  exposed to 50,  100,  or 150 ppm
                       O
 (173,  347, or 521  mg/m ) DCM 7 hr  after exposure was terminated.  For the
200-ppm (695  mg/m ) exposures,  the mean post-exposure end tidal  air  concentra-
tion of DCM decreased  to 1 ppm  (3.47 mg/m ) at  16 hr.  Post-exposure  elimina-
tion of DCM was  less than 5 percent of the amount absorbed.   A  related exposure
                                   4-3

-------
                DURING
               EXPOSURE
          AFTER EXPOSURE
          100
           80
       I
       tif
       Q
       cc
       o
       o
       111
       UJ
       I
       UJ
              RESPIRATORY
               ABSORPTION
                             - ROOM CONCENTRATION
     ABSORPTION AND EXCRETION
      OF METHYLENE CHLORIDE
EXHALED AIR CONCENTRATIONS
                           RESPIRATORY EXCRETION
Figure 4-1.  Inspired and expired air concentrations during a 2-hour,  100-ppm
             inhalation exposure to DCM for a 70-kg man, and the  kinetics  of
             the subsequent pulmonary excretion.

             Source:  Riley et al., 1966.
                                    4-4

-------
study in which volunteers were exposed to 100, 150, and 200 ppm (347, 521, and
        3
695 mg/m )  DCM  for 7.5 hr daily  for 5 consecutive days also indicated that
uptake and elimination were directly proportional to the magnitude of exposure,
thus confirming previous findings by DiVincenzo et al. (1972) and Stewart et al.
(1973).
     The retention  of  DCM as a percentage  of inspired air concentration is
independent of  that concentration at equilibrium.  Retention values for DCM,
reported by different investigators, are shown in Table 4-1.  These values have
a large range and vary with the duration of exposure.   Variation in the values
is also caused  by  differences  in  body  weights of  the  subjects and differences
in body composition (proportion of adipose tissue to lean mass).  For exposures
greater than 1 hour, the mean retention approximates 42 percent of uptake of DCM
or approximately 125 mg/hr for an exposure  of 100 ppm  (347  mg/m3), assuming  a
resting ventilation rate of 6 1/min.
         TABLE 4-1.  PULMONARY ABSORPTION OF DCM BY HUMAN SUBJECTS UNDER
                               SEDENTARY CONDITIONS
Inhalation
concentration,
Investigator ppm
DiVincenzo and Kaplan, 1981a



Lehmann and
Schmidt- Kehl, 1936



Riley et al. , 1966
DiVincenzo et al . , 1972


Astrand et al . , 1975

50
100
150
200

662
806
1,152
1,181
44-680
100
100
200
250
500
Exposure,
hr
7.5
7.5
7.5
7.5

0.3
0.5
0.5
0.5
2
2
4
2
0.5
0.5
Retention,
%
70
60
63
60

74
75
72
72
31
53
41
51
55
55
Engstrom and
  Bjurstrom, 1977
750
34
                                   4-5

-------
      For  short exposures, the quantity (dose) of DCM absorbed into the body is
 theoretically and experimentally directly proportional to the concentration of
 DCM in the expired air (Lehmann and Schmidt-Ken!, 1936; Astrand et al., 1975).
 The body  burden of DCM also increases with exposure duration and with physical
 activity  (increased  ventilation  and cardiac output) at  a  given inhaled air
 concentration (Engstrom  and Bjurstrom, 1977; Astrand et  al.,  1975;  DiVincenzo
 and Kaplan, 1981b).  Astrand et al. (1975) found that physical activity during
 exposure  to  250  and 500 ppm DCM  (868  and  1735  mg/m3)  for  0.5  hour  decreased
 retention from 55 percent in a resting stage to 40 percent during activity but
 that  it  doubled  the  amount  of  DCM  absorbed because  of  a  threefold  increase of
 ventilation rate  (6.9  to 22 ฃ/min).  DiVincenzo and Kaplan  (1981b)  also  found
 that  physical exercise during  exposure increased pulmonary uptake.  Exposure
                                         3
 of three  males  to 100 ppm  DCM  (347  mg/m ) for 7.5 hours, during which they
 exercised on a treadmill for 5 minutes of each 15-minute period, also resulted
 in an estimated doubling of average cardiac output and as much as an eightfold
 increase  in the  average alveolar ventilation rate (Table 4-2).  Effects upon
 blood COHb and conversion of DCM to CO are discussed elsewhere in this chapter.
     One  study attempted to correlate the quantity of DCM absorbed with body
 weight and  fat content  (Engstrom  and Bjurstrom,  1977).   Individuals were
                             3
 exposed to 750 ppm  (2606 mg/m  ) DCM  for  1  hour.  Obese subjects  (average body
 fat 25 percent of body weight) absorbed 30 percent more DCM than lean subjects
 (average body fat 8 percent of body weight).  When fat biopsies of subcutaneous
 adipose tissue of  obese subjects  were taken, DCM concentrations of  about 10
 and 8 mg DCM/kg tissue  weight were  found at 1 and 4  hours post-exposure,
 respectively.   Between 1 and 2 mg/kg were  found in  fat samples  22 hours  after
 exposure.  If confirmed,  such observations would indicate that  elimination of
 DCM from  adipose deposits proceeds at  a  slow rate.  Furthermore, the  residual
 DCM in adipose tissue  is additive  to the next day's exposure, particularly in
 obese people.   However, with multiple daily exposure,  a total body equilibrium
 and constant adipose tissue concentration are eventually achieved with a given
 DCM concentration in inspired air.
 4.1.3.2  Studies in Animals—McKenna et al. (1982) used 250-gram male Sprague-
 Dawley rats to  estimate the total  body  burden  of  DCM  resulting from 6-hour
                                                                        Q
 inhalation exposures of  50, 500,  and 1500 ppm (173, 1737, and 5211 mg/m ) of
 14
  C-DCM.   After 6  hours, the animals were in apparent total body equilibrium
with the  inhaled concentrations of radioactive  DCM, as suggested by  "plateau"
                                   4-6

-------
          TABLE 4-2.   EFFECT OF EXERCISE ON PHYSIOLOGICAL PARAMETERS
                         FOR VOLUNTEERS EXPOSED TO DCM

Work
intensity_
(ml 02 min 1
Volunteer kg b.w. x)
1 4
14
2 4
15
19
3 4
16
28

Aerobic
capacity, %

25

25
45

45
70

Heart
rate,
beats/mi n
56
84
68
96
119
68
123
145
Estimated
average
cardiac
output,
1/min
5
11
5
12
14
5
15
21
Estimated
average
alveolar
ventilation,
1/min
8
18
6
34
45
6
28
46
 Work intensity was estimated by monitoring the heart rate and relating it
 to equivalent 02 consumption measured during pre-exposure aerobic capacity
 testing.   Oxygen consumption at rest was taken as 4 ml 02 min 1 kg body
 weight 1, based on preexposure testing.
Source:   DiVincenzo and Kaplan (1981b).
blood concentrations (Figure  4-2).   The body burden was calculated from the
total radioactivity recovered  from  exhaled air, urine and feces, and carcass
analysis during the first  48 hours  after exposure.   Table 4-3 shows the body
                                                                        14
burden associated with  each  exposure.   The increase in  body  burden of   C-
activity was less than  proportionate to the increments of DCM  inhaled concen-
tration.
4.1.3.3  Blood/Air Relationship—The blood concentration of DCM  during  inhal-
ation and  in the elimination phase after exposure  parallels  alveolar DCM
concentration.   This predictable relationship is defined by the  solubility  of
DCM.  Astrand et al.  (1975) showed that for men exposed to 250 and 750 ppm DCM
                  3
(868 and 2606 mg/m ) for 1.5  hours, the arterial blood (mg/1)  to alveolar air
(mg/1) concentration ratios were constant and averaged 10.3 and  11.1, respec-
tively, over  threefold changes in  alveolar  concentrations.   These ui  vivo
Ostwald coefficients agree with  the value found for blood/air (7.9) and for
water/air (7.2) (Lindqvist, 1978; Morgan et al., 1972) at 37ฐC in vitro.  The
high water/air coefficient suggests that DCM is dissolved in plasma water as
well as in lipid components of blood.

                                   4-7

-------
       10.0
   E
   1
   _•ป
   o
   fซ
   o
   <
   OT
      0.001
                                                           50 ppm
                                                           500 ppm
                                                           1500 ppm
       0.01
                                     4       5      6/0

                                    TIME, hours
Figure 4-2.  Plasma  levels of DCM in rats during and  after DCM exposure
             for  6 hours.   Data points represent  mean ฑ  standard deviation
             for two  to  four rats.

             Source:   McKenna et al., 1982.
                                    4-8

-------
            TABLE 4-3.  BODY BURDENS OF RATS AFTER A 6-HOUR INHALATION
                                EXPOSURE TO 14C-DCM
Exposure
concentration,
ppm
50
500
1500

Number of
rats
3
3
3
Total body burden
mg Eq 14ODCM/kg
ฑ S.D.
5.53 ฑ 0.18
48.41 ฑ4.33
109.14 ฑ 3.15
S.D. = standard deviation.
Source:  McKenna et al. , 1982.

     In the exposure study of DiVincenzo and Kaplan (1981a) described previously
(Section 4.1.3.1), exposure and post-exposure blood concentrations of DCM were
directly proportional to  the  magnitude of exposure.   Blood/air  coefficients
                                                           3
calculated from the data suggest that the 200 ppm (694 mg/m ) exposure concen-
tration may be approaching the level at which saturation of metabolism occurs,
as evidenced by an increased level of DCM in venous blood.
     MacEwen et al. (1972) determined the blood DCM concentration in dogs that
were continuously  exposed for 16  days  to  1000 and 5000 ppm  DCM (3474 and
           3
17,370 mg/m ).  The blood DCM concentrations were 36 and 183 mg/1, respectively,
in  direct  proportion to  exposure  concentration.   Total  equilibrium can be
assumed to have  occurred  in these animals.   Ostwald coefficients of 10.4 and
10.5 for the  two exposure concentrations agree with the above values noted in
man.  Similar values  can  be calculated from the  data  of  Latham and Potvin
(1976); Figure  4-3 shows the proportional  relationship  they found in rats
between DCM blood  and inspired  air concentrations over a  range  of 1,000 to
                               3
8,000 ppm (3,474 to 27,792 mg/m ) during a 6-hour exposure.
     In contrast  to these  findings  of a direct  proportional  relationship
between inspired air  concentration of DCM and blood level  in man and other
animals, McKenna et al. (1982) reported a greater than proportionate increase
of blood concentration with inhalation exposure concentration in rats.  Table
4-4 gives the apparent  steady-state concentration of DCM in plasma and whole
blood of Sprague-Dawley rats exposed for 6 hours each to 50, 500, and 1500 ppm
DCM (173, 1737,  and 5211  mg/m3).   The data indicate that the whole blood and
                                   4-9

-------
          Dl
          CM
         O
          CM
         I
         O
         u_
         O
         _i
         in
          Q
          O
          O
          CO
             0.60
0.40
             0.20
                      INHALATION
Figure 4-3.
   0         1000       2000       4000       8000
               EXPOSURE CONCENTRATION, ppm

DCM venous  blood  levels  in  rats  immediately  after  a  single
6-hr  inhalation exposure to various concentrations of  DCM.
Source:  Latham and Potvin, 1976.
         TABLE 4-4.  DCM CONCENTRATIONS IN RAT WHOLE BLOOD AND PLASMA
    AT APPARENT STEADY-STATE CONDITIONS AFTER A 6-HOUR INHALATION EXPOSURE
Exposure
concentration, Number o1
ppm rats
50
500
1500
3
3
3

Whol
0.22
39.53
DCM concentration
jjg/ml ฑ S.D.
e blood
+ 0.04
ฑ 3.71
Plasma
0.05 ฑ 0.01
2.38 ฑ 0.42
8.94 ฑ 0.39
Plasma/blood
distribution
coefficient
0.23
0.23
S.D. = standard deviation.
Source:  McKenna et al., 1982.

plasma levels  of  DCM  increase disproportionately with an increase in inspired
air  concentration.   Furthermore,  calculation of the blood/air ratio for these
data provide  increasing values of 5.75, 5.97, and 7.59 for 50, 500, and 1,500
ppm  (173,  1737,  and 5211 mg/m3),  respectively.   McKenna et al.  suggest that
the  resultant  increase  in blood DCM  concentration  is  greater  than that  predic-
ted  by increments in  the inspired air because of a rate-limited metabolism of
DCM  in the rat.  Thus,  at low inspired  air  concentrations  (below saturation  of
metabolism),  the  blood/air ratio  is  less  (because of rapid metabolism) than
that  at  high  inspired  air  concentrations  (above saturation  of  metabolism);
                                    4-10

-------
indeed,  the blood/air coefficient  (7.59)  observed at 1,500 ppm  DCM (5,211
mg/m  ) agrees with coefficients observed by others.

4.1.4 Tissue Distribution
      Because of DCM's water solubility, it probably distributes throughout the
body  water,  and  its  lipid solubility  allows  its distribution into  all body
tissues  and  cellular lipids,  particularly into  adipose  tissue.  Engstrom  and
Bjurstrom  (1977)  determined  that  the Ostwald coefficient  for  subcutaneous
adipose  tissue from  human buttocks is 51  at  60ฐC;  this  value  indicates that
the  tissue/blood  partition coefficient may be  about 7 at  body  temperature
(37ฐC).
      DCM readily  crosses  the  blood-brain barrier even at relatively  low vapor
exposure concentrations,  as  evidenced by its impairment of manual and mental
performance at 500 ppm  (1737  mg/m3)  (Winneke  and Fodor,  1976; Winneke, 1981).
DCM also crosses  the placenta and may affect fetal  development (Schwetz et
al.,  1975;  Anders and Sunram, 1982).  Schwetz et al.  (1975) consider the delay
in rat fetal development to be a result of maternal toxicity.
4.1.4.1  Animal Studies—Tissue concentrations  of DCM increase with exposure
concentrations and duration  and for any given  tissue are dependent on the
largely  unknown  tissue  partition  coefficients.   Savolainen et  al.  (1977)
chronically exposed  rats  to DCM [200 ppm (695 mg/m3),  6  hr  daily,  for 5 days]
and determined DCM concentrations  in peri renal   fat and  other  tissues.   The
resulting data, shown in  Table 4-5, indicate that significant amounts of DCM
remained in peri renal fat 18 hr after the previous  exposure  of day 4, and
increased markedly with a 6-hr exposure on day 5. At termination of the last
(fifth daily) 6-hr exposure,  the  ratio of tissue to blood concentrations  was
about one for brain and  liver and 6.6 for peri renal fat.
     Previous work by DiVincenzo  et  al.  (1972)  in man  and  Carlsson  and
Hultengren   (1975)  in rats indicated  that little uptake  by adipose  tissue
occurs.  However,  single,  short exposure periods of 2 hours were used in their
studies,  and of the smaller amount absorbed,  95  percent was probably accomodated
in VRG and MG compartments because the FG compartment  receives only  5 percent
of the cardiac output.
     McKenna and  coworkers  (1981,  1982)  have studied DCM  distribution  in
                             14
tissues of  rats  by observing    C-activity 48  hr  after a single 6-hr inhalation
of 50, 500, or 1500 ppm DCM (174, 1737, and 5211 mg/m3) and after  single oral
                                   4-11

-------
            TABLE 4-5.  TISSUE CONCENTRATIONS OF DCM IN RATS EXPOSED TO
                      200 ppm DCM FOR 4 DAYS FOR 6 HOURS DAILY
Exposure
DCM concentrations
nmol/g tissue wet weight ฑ S.D.
on the fifth
day, hr
0
2
3
4
6
Tissue to
Cerebrum

73
119
57
83
-
ฑ 20
ฑ 33
ฑ 8

blood coefficient
Cerebellum
-
57 ฑ
86
95 ฑ
90
after

20

8

the
Blood

90
79
120
100
fifth
-
ฑ
+
+
ฑ
dai

10
3
10
1
IV
Liver
_
85 ฑ
82 ฑ
101 ฑ
83 ฑ
exposure


2
1
13
10

Peri renal

113
526
537
608
659

fat
ฑ 29
ฑ 94
ฑ 33
ฑ 58
ฑ 77

                0.83:1        0.90:1       1.0:1       0.83:1
6.59:1
S.D. = standard deviation
Source:  Savolainen et a!., 1977.

doses of 1  or 50 mg/kg.  The  results  are  shown  in Table 4-6.  Following the
inhalation exposures  and  the oral dosage, the highest concentrations of   C-
activity were found in the liver, kidney, and lung.   The observed   C-activity
in  epididymal  fat  was consistently lower than that observed  in either whole
                                            14
blood or the  remaining  carcass.   The tissue   C-activity increased with dose,
but it did  not increase in direct proportion to increasing dose.   Intact DCM
was not detected in any of the tissues assayed;  therefore,  the observed radio-
activity is  presumed to  represent nonvolatile  metabolites of  DCM.   These
results contrast with those  of Savolainen et al. (1977),  who found that in
peri renal adipose  tissue,  significant  amounts of intake DCM  remained  18 hr
after the last  of  four  daily 6-hr inhalation exposures  to 200 ppm DCM (695
mg/m3) (Table 4-5).
     The results of  the experiments  of Savolainen et al.  and McKenna et al.
indicate that total  body  equilibrium of DCM to inspired air concentration is
not achieved  in  the  rat within a single 6-hour inhalation  period even though
this response is suggested by the  achievement of a "plateau" blood concentra-
tion as  shown in  Figure  4-2.   Also  of interest is the  evident  difference
                              14
between the amount of tissue   C-metabolites  associated with inhalation and
oral dosage.  A close correspondence exists between tissue metabolites of DCM
                                   4-12

-------
     TABLE 4-6.   DISTRIBUTION OF 14OACTIVITY IN RAT TISSUE 48 HOURS AFTER
              6-HOUR INHALATION EXPOSURE OR ORAL DOSAGE OF 14C-DCM

Exposure method                          mgEq 14C-Activity by exposure
                                          concentrations, tissue ฑ S.D.
Inhalation
Liver
Kidney
Lung
Brain
Epididymal fat
Skeletal muscle
Testes
Whole blood
Remaining carcass
Oral
Liver
Ki dney
Lung
Brain
Epididymal fat
Skeletal muscle
Testes
Whole blood
Remaining carcass
50 ppm
8.4 ฑ 1.5
3.3 ฑ 0.1
1.9 ฑ 0.2
0.8 ฑ 0.3
0.5 ฑ 0.2
1.1 ฑ 0.1
1.1 ฑ 0.2
1.1 ฑ 0.2
1.3 ฑ 0.2
1 mg/kg
0.40 ฑ 0.04
0.15 ฑ 0.01
0.99 ฑ 0.01
0.04 ฑ 0.06
0.02 ฑ 0.002
0.08 ฑ 0.02
0.04 ฑ 0.002
0.06 ฑ 0.003
0.05 ฑ 0.002
500 ppm
35.6 ฑ 7.5
16.2 ฑ 2.4
11.0 ฑ1.3
4.2 ฑ 1.3
6.5 ฑ 0.5
4.4 ฑ 1.9
5.5 ฑ 1.3
8.1 ฑ 1.9
5.6 ฑ 0.9
50 mg/kg
6.67 ฑ 0.69
2.92 ฑ 0.21
1.67 ฑ 0.16
0.63 ฑ 0.10
0.33 ฑ 0.0006
0.86 ฑ 0.04
0.92 ฑ 0.10
1.41 ฑ 0.11
0.99 ฑ 0.60
1500 ppm
44.2 ฑ3.5
30.5 ฑ 0.2
16.5 ฑ1.6
6.7 ฑ 0.2
4.1 ฑ 0.9
7.7 ฑ 0.7
8.1 ฑ 0.5
8.9 ฑ 1.7
8.6 ฑ 1.4






Number of animals in each group =3
Source:  McKenna et al., 1982; McKenna and Zempel, 1981.

for an oral dose of 50 mg/kg and a 6-hr inhalation exposure of 50 ppm, although
                  o
50  ppm (174 mg/m )  provides a  terminal  body  burden  of only  5.5  mg/kg
(Table 4-3).

4.1.5  Pulmonary Elimination
     Pulmonary  excretion  is the mechanism  of elimination of virtually  all
                                                       *
unchanged  DCM  from  the  body.  Less than 2 percent of estimated body doses of
DCM have  been  detected  as unchanged compound  in the urine of human  subjects
exposed to 100 ppm and 200 ppm DCM (347 and 695 mg/m3) for 2 hours (DiVincenzo
                                                                          3
et  al.,  1972)  and in the urine  of  dogs exposed to 5000  ppm (17,370 mg/m )
(MacEwen et al., 1972).
                                   4-13

-------
4.1.5.1  Studies in Humans—Figure 4-1 schematically shows the time-course of
pulmonary elimination  of  DCM after inhalation exposure.  The  parameters  of
elimination equilibration of  the  body are the same as those of assimilation
equilibration.  After  termination  of  exposure,  DCM immediately begins to be
eliminated from the body via the lungs.   Alveolar air equilibrates with pulmo-
nary venous blood whose DCM concentration becomes a function of the first-order
diffusion of  DCM  from  tissues,  the arterial blood flow/tissue mass, and the
relative solubilities  of  DCM in tissues.  Figure 4-1 shows that alveolar DCM
concentration follows  an  exponential  decay curve with three major components
reflecting desaturation of  the  VRG, MG,  and  FG  compartments,  respectively.
The half-times  of  elimination of DCM from these  compartments  have not been
firmly established.  Riley  et al.  (1966) measured expired air concentration
after termination  of exposure and  found  half-times of 5  to 10 min  for  the VRG
compartment,  50 to 60  min for the MG compartment, and 400 minutes for the FG
compartment.  DiVincenzo  et  al.  (1972),  who exposed  subjects to  100 ppm and
200 ppm  DCM (347  and  695 mg/m  )  for  2-  and 4-hr periods,  felt  that "very
little vapor" reached  the fat stores and muscle tissues  under these conditions.
DiVincenzo  et al.  found DCM  to  have  a  half-time value  •(:, olood  of Vu min
following 2  hr  of exposure; prolonging  exposure  to  4 hv had no significant
effect on the half-time.  DiVincenzo and Kaplan (1981a,b) have recently extend-
ed previous studies by following the course of pulmonary elimination in indivi-
duals exposed for 7.5  hr  to  50,  100,  150,  and  200 ppm DCM  (174,  347, 521, and
695 mg/m3)  and  in individuals exposed for 7.5  hr daily'for  5 consecutive days
to the  above three highest concentrations.  The authors concluded that post-
exposure elimination of DCM  in breath is a minor  route of elimination  over the
concentration range used; post-exposure  elimination was  less than  5 percent of
the amount  absorbed.   Pulmonary elimination of DCM was  rapid at  all exposure
                                              3
concentrations.   Less  than 0.1 ppm  (0.347 mg/m ) was  detected in  post-exposure
breath  samples  at 7 hr.  Die-away  of DCM in blood paralleled that observed
with  pulmonary eliminations.   Breath elimination times were  prolonged by
physical  exercise.  Morgan  et  al.  (1972), using  isotopically  labeled DCM,
estimated the half-time of DCM  in  the VRG  compartment as 23 min.   Engstrom and
Bjurstrom  (1977)  reported the half-time of DCM in the MG compartment for lean
subjects  was about 60 min;  these  authors obtained a longer time value  for
obese  subjects.   For  both lean  and obese subjects,  biopsies indicated that
residual  concentrations of DCM existed  in  adipose tissue nearly 24 hr after
                                    4-14

-------
exposure, suggesting that the half-time of elimination from the FG compartment
is fairly long.   From the post-exposure alveolar concentration curves prepared
by Stewart et al.  (1976a) for subjects exposed to inhalation concentrations of
                                      3
50 to 500  ppm  DCM (174 to 1,737 mg/m ) for 1 to 7.5  hr/day for 5 successive
days, the  half-time of elimination for MG and FG can  be estimated as 60 to 80
min, and 240 min,  respectively.   The best guesses from these studies for the
half-times of elimination  from  the VRG,  MG, and FG are 8 to 23 min, 40 to 80
min, and 6 to  6.5 hr, respectively.    The long  half-time  from  elimination  of
the adipose tissue compartment and reports that DCM remains in this compartment
24 hr after  single and chronic exposures indicate  that the concentration  of
DCM  in  adipose  tissue may  only slowly, over a period  of days,  achieve equili-
brium with inspired air concentration, particularly  for  daily exposures  of
short duration.
4.1.5.2  Studies in Animals--The estimates of half-times of pulmonary excretion
of DCM  in  animals are derived from post-exposure alveolar concentration curves.
Blood DCM  decay concentration curves  are a less reliable parameter of pulmonary
excretion  because  these  curves  are influenced by metabolism, the other major
route of  elimination  of DCM.  McKenna et al.  (1982)  determined the pharmaco-
kinetic  half-times for elimination of DCM  from plasma of rats following  a
                                                                     3
single  inhalation  exposure to 500 or 1500 ppm DCM  (1737 or 521 j mg/m ) for 6
hours.  Figure 4-2 shows that two first-order processes governed the disappear-
ance of DCM from plasma with apparent half-times of  2 and 15 minutes for the
rapid and  slow components  of elimination, respectively.  The kinetic parameters
calculated from these curves are summarized in Table  4-7.
     The kinetic parameters  for rats  differ greatly from those of man.  Presum-
ably, the  half-times  for the fast and slow  components correspond  to the  half-
times for  human VRG and MG compartments.  The  half-time  for the FG  or  adipose
tissue  compartment was  not established in  the  rat because plasma  decay was
only followed  for 1.5 hours following inhalation exposure  (Figure  4-2).  The
rates of elimination of  DCM  from plasma in the  rat  are first order  and indepen-
dent of dose,  although there is  considerable  evidence of extensive  metabolism
in  these animals  (Section  4.2).
     McKenna and  Zempel  (1981)  have  also  determined the  kinetics  of pulmonary
excretion  of DCM in rats  after a  single oral dose  (via gavage) of  1 or 50 mg
  C-DCM per kg body weight.  Figure  4-4  shows  the time  course (for 5  hours)
of  DCM  in exhaled air  of  these rats.  Pulmonary elimination of DCM from rats
                                   4-15

-------
         TABLE 4-7.  COMPARISON OF POST-EXPOSURE PULMONARY ELIMINATION
                     HALF-TIMES OF DCM FOR HUMANS AND RATS
Subject
Exposure
 method
    DCM
concentration
                                             % first-order components (minutes)
                                             Alpha       Beta       Gamma
Human      Inhalation    50-500 ppm
Rat        Inhalation     500 ppm (6 hr)
                                             8-23
                                            40-80
                                           360-390

Rat Gavage

1500 ppm (6 hr) 2.4
1.06
1 mg/kg
50 mg/kg
14.8
15.5

12.6
12.6
-

46.5
46.6
Source:  McKenna et al., 1982.

receiving the 1 mg/kg  dose  is characterized by a  two-component  decay curve
representing two first-order  processes with half-times of elimination of 12.6
minutes and 45.6 minutes,  respectively.   The pulmonary excretion of DCM from
rats given the  50  mg/kg dose approximates zero-order kinetics for the  first
hour after  administration (possibly  rate-limited  by absorption)  and then
describes the same  first-order  kinetics  of the 1  mg/kg dose.  These  results
suggest that the half-time  of pulmonary elimination from the FG compartment
for rats approximates  45 minutes, versus 6 to  6.5  hours for man.  However,  as
noted  for rats,  single inhalation or oral exposures  in  man probably do not
result in total  body equilibrium, particularly with the fat compartment, and
elimination from  this compartment may be  longer  than indicated from these
experiments.
     Withey and Collins  (1980)  determined the kinetics of elimination of DCM
from the blood of Wistar rats after intravenous administration of 3, 6,  9,  12,
or 15  mg/kg DCM given intrajugularly in 1 ml  of  water.   For the four lower
doses, the blood decay curves best fitted a first-order two-compartment model,
but the highest dose fitted a first-order three-compartment model.   Withey and
Collins suggested that the  apparent change from a two- to  three-compartment
system occurred as  a consequence of a "difference in biological response  to
                                   4-16

-------
                100.0
                 10.0 —
             0)
             n.
             O
              ซ

             O

             (0
             <
             Q
             01
             ฃ
             CL
             X
             uu
             UJ
             W
             O
             Q
Figure 4-4   Pulmonary elimination of 14C-DCM following  oral  administration
             to rats of a single dose of 1 or 50 mg/kg 14C-DCM (squares).
             Ordinate is percent dose administered  during  0.5-hour collection
             periods with means ฑ SEM for groups of three  rats.   SEM is the
             standard error of the mean.
                                    4-17

-------
the dose either  as  a consequence of the magnitude  of the dose or, in some
cases, due to  the  varying response of different animals  to  the same  dose."
This  interpretation  is probably true because the k  (rate constant for elimi-
nation from  blood out of body, principally  via pulmonary excretion and/or
metabolism) was  relatively  high  for the lower  doses  and  consistent with a
half-time of body elimination of only 4.5 min.  Any significant equilibration
of blood DCM with  the fat compartment was  very unlikely during these  experi-
mental circumstances.  Further support for limited distribution of DCM is the
reported kinetics for the highest dose (15  mg/kg) that fit a three-compartment
system.  In this case, k  was 0.09/min, that is, a half-time of body elimina-
tion  of 7.7 min.   This result reflects the fact that more time was required
for equilibration into fat  to occur.   The half-time  of elimination from the
fat compartment was given as 31.5 min, although the rate constant k^  [movement
of DCM from the  fat  compartment to  the central  compartment (blood)] was given
as 0.006/min,  indicating  a  half-time  of 115 min.  The volume of distribution
(Vd)  of DCM was  calculated  as 48.8 ml or  about 12 percent of body weight,
which was  surprisingly low  for both a water- and lipid-soluble compound that
is known to diffuse into all the major organ systems.
4.2  METABOLISM OF DCM
     DCM is  known  to be metabolized in man to CO and in experimental animals
to CO and COp.  The CO production results in an elevation of blood COHb content,
from which CO dissociates in the lung,  followed by elimination of CO.   Experi-
ments in man and animals have shown that the metabolism of DCM is dose dependent
and limited by hepatic enzyme saturation.

4.2.1  Evidence for Metabolism to Carbon Monoxide
     Before 1972,  most  of the absorbed dose  of  DCM  in  man was thought to be
excreted unaltered in exhaled air, while a small amount was found in the urine
(MacEwen et a!., 1972; DiVincenzo et al., 1972; Heppel et al., 1944).  Metabo-
lism of  DCM  to CO was not  known  to  occur.   However,  in 1972,  Stewart et al.
discovered that  the  COHb concentration in blood increased in persons exposed
to 200  to  1000 ppm DCM  (695 to  3474  mg/m3)  for 1 to  2 hours.   The COHb levels
continued  to  rise  after cessation of exposure and decreased more slowly than
when similar  levels  of COHb were induced  by breathing CO.   Stewart and his
                                   4-18

-------
associates (1972a,b) proposed that  CO  was the end product of DCM metabolism.
In the following year, Fodor et al. (1973) showed that COHb blood levels from
DCM exposure were  further  elevated by concomitant exposure to diiodomethane
and dibromomethane, thus indicating that these dihalomethanes also are metabo-
lized to  CO.   At  first,  this unique halocarbon metabolism was not generally
accepted  because the increased COHb levels might  reflect  a change in  the rate
of endogenous  CO production  or  excretion associated with heme degradation by
the microsomal hepatic heme oxygenase system in man (Coburn, 1973; Tenhunen et
al., 1969).   However,  Stewart  et al.  (1972a,b, 1973) observed no evidence of
an enhanced metabolism of hemoglobin in their subjects exposed to DCM, and the
subjects  did  not excrete  increased amounts  of urobilinogen in  their urine
either during or after exposure.
     A second  hypothesis  for the origin of the excess COHb postulated a DCM-
induced  conformational  change in  hemoglobin, which increased CO affinity.
Hence, more  COHb  would be formed, with  a longer biological half-life, from
endogenous  and exogenous  CO  sources  at  the same ambient  concentrations.
Settle (1971)  had  shown that xenon and,  to a much larger degree, cyclopropane
bind to  myoglobin  and  increase CO  affinity.   Following this observation, Nunes
and  Schoenborn (1973) used  X-ray  diffraction to demonstrate that DCM also
binds  to sperm whale myoglobin  and suggested that this binding might increase
CO  affinity.   Settle (1975)  then investigated the CO binding to  human hemoglo-
bin    He observed that CO binding to  human hemoglobin in vitro  at  37ฐC  is
                                                       o
increased in the presence of DCM  (1000  ppm,  3474 mg/m  ).  The amount of COHb
formed at a given  CO  concentration is  doubled.   Also,  measurement of the  P5Q
values of hemoglobin  (partial pressure  of 0ฃ or  CO at  which  50 percent satura-
tion  occurs  at 37ฐC)  in the  presence  and absence of  DCM (1000  ppm) indicated a
 six-fold increase  in  CO affinity.  From  these  observations,  Settle (1975)
 suggested that the increased COHb  seen in-vivo may  be  caused by  an  increase  in
 CO affinity  and not  by metabolism.   More recently, Collison  et al. (1977)
 determined the Haldane affinity constant for CO for both human  and  rat blood
 equilibrated at 37ฐC with air containing only CO and CO plus DCM (10,000  ppm).
 No difference in  the  Haldane constants  for  human blood (mean value  of 227) or
 rat blood (mean value of  179) was found.  Negative results also were obtained
 when the absolute  affinity  of  CO was measured  in a nitrogen-DCM atmosphere.
 Dill  et  al.  (1978) redetermined the P5Q value for human and rat blood in the
 presence and  absence  of  CO  (2500 ppm)  and  DCM  (800 ppm).  DCM was  found to
                                    4-19

-------
 have  no  effect on the Pgo values;  therefore,  Dill  concluded  that DCM does  not
 significantly  increase the affinity of hemoglobin for CO.
      The biotransformation of DCM to CO and C0ฃ has now been confirmed by many
 metabolism  studies.   The  hepatic  metabolism  of DCM has been  unequivocally
 shown to be the origin of the  CO  responsible for increased COHb blood con-
 centrations.   Independently, several groups have shown by administering 13C-DCM
 or   C-DCM to  rats that labeled CO subsequently appears in COHb with essential-
 ly the same specific activity (Carlsson and Hultengren, 1975;  Miller et al.,
 1973; Kubic et al.,  1974;  Zorn, 1975).   Furthermore,  Fodor and coworkers
 (1973, 1976) and  Kurppa  et al.  (1981) have demonstrated that rats exposed to
 CO and DCM  singly and in combination showed an additive increase in blood COHb
 levels (Table 4-8).  In addition, many investigators have shown a dose-response
 relationship between  injected or inhaled  DCM and increased blood COHb levels
 in both  experimental  animals and men (Figures  4-5 and 4-6)  (DiVincenzo et al.,
 1972;  Astrand  et  al.,  1975;  Fodor et al.,  1973; Ratney et  al., 1974; Stewart
 et al.,  1973,  1976a,b; Forster  et al.,  1974; Roth  et  al.,  1975; Ciuchta et
 al.,  1979;  Hake et al.,  1974).
      The  extensive metabolism of halogenated hydrocarbons to CO  is apparently
 unique to the dihalomethanes.  This metabolism is  not observed  to any signifi-
 cant  extent  with  chloroform,  carbon tetrachloride, methyl chloride,  methyl
 iodide,  trichlorofluoromethane,   dichlorodifluoromethane,  carbon  disulfide,
 formaldehyde,  formic  acid, or methanol  (Miller  et al. , 1973;  Kubic  et  al.,
 1974;  Fodor  and  Roscovanu,  1976;  Rodkey  and Collison,  1977).   Fodor and
 Roscovanu (1976) state, without  giving data,  that  chloroform,  bromoform, and
 iodoform  are metabolized to CO in the rat,  thus increasing  blood COHb levels;
 this  has  not been the observation of other  investigators.   According to Fodor
 and Roscovanu (1976), of the dihalomethanes,  the bromo-iodo-halides  are  more
 extensively  metabolized  to CO than  is DCM  (Figure 4-7), thus increasing  COHb
 to  a  significantly greater extent  than DCM metabolism (Miller  et al. , 1973;
 Kubic  et  al., 1974; Fodor and Roscovanu, 1976;  Rodkey and Collison,  1977).

 4-2.2  Evidence for Dose-Dependent Metabolism of DCM:  Michaelis-Menten Kinetics
 4.2.2.1  Studies in Humans—McKenna  et al.  (1980) have  reported  in abstract
 form kinetic studies carried out  in six healthy male volunteers exposed to 200
                                  Q
 and 350 ppm DCM (695 and 1216 mg/m ) during each of two 6-hour exposure periods.
These investigators reported  that the metabolism of  DCM was  dose-dependent,
                                   4-20

-------
         TABLE 4-8.  BLOOD CARBOXYHEMOGLOBIN CONCENTRATIONS OF RATS
                     EXPOSED TO CO AND DCM BY INHALATION
Exposure concentration, ppm
DCM CO
100 (0.5 - 2)a
1000 (0.5 - 2)a
0 100
100 100
1000 100
None None
100
1000
1000 100
COHb
saturation
%
6.2
12.5
10.9
16.4
19.0
0.7 ฑ 0.2b
8.8 ฑ 1.9b
6.2 ฑ 0.9b
14.6 ฑ 1.3b
aAmbient air CO concentration.
 Mean ฑ standard deviation, N= 5/group.
Source:  Fodor et al., 1973; Kurppa et al. ,  1981.
based on  comparison  of the kinetics of the  two  doses during and  following
exposure.   They compared the DCM blood levels and of the concentrations of DCM
in expired air.  Blood COHb levels and exhaled CO concentrations were less than
those expected for the 350 ppm (1216 mg/m ) exposure.   Calculations made of the
rate of DCM metabolism during exposures were consistent with Michaelis-Menten
kinetics for DCM metabolism.
     More recently,  DiVincenzo and Kaplan  (1981a) evaluated the conversion of
DCM to COHb and  CO in sedentary, nonsmoking individuals (11 males and 3  fe-
males) exposed to  DCM levels  of 50, 100,  150, or 200 ppm (174, 347, 521,-or
695 mg/m ),  for 7.5 hr, or for 7.5 hr daily to 100,  150,  or 200 ppm (347,  521,
           3
or 695 mg/m )  for  5  consecutive days.   Between 25 and 34 percent of the  ab-
sorbed DCM was excreted  in expired air as CO during and after exposure.   The
                                   4-21

-------
                 O
                 O
                     2 -
                             100      200     300

                                   INHALED AIR, ppm
400
500
Figure 4-5.    Carboxyhemoglobin concentrations in male nonsmokers exposed to
              increasing concentrations of DCM for 1, 3, or 5 h/day for 5 days.
              Pre-exposure values averaged 0.8%, but with 3- and 7.5-hour expo-
              sures the pre-exposure values were above this baseline value on
              the mornings following exposure.

              Source:  Stewart and associates (1972, 1973, 1974).
                                     4-22

-------
                  I
                  o
                  o
                       4  -
                      2  -i
                              200      400     600     800

                                    INHALED AIR, ppm
1000
Figure 4-6.    Carboxyhemoglobin concentrations In rats after  inhalation  exposure
              to increasing concentrations of DCM for single  exposures of  3  hours.
              The values are corrected for pre-exposure COHb  concentration and
              calculated from the data of Fodor et al., 1973.
                                     4-23

-------
65
60
55
50
I 45
-j 4ฐ
LU
2; 35
0 30
0
8 25
o
ฃ 20
15
10
5
.
-
-
-
.



.
-
-
-
t











•


















_


••








-
-
-
_

-
-
-
-
-
-
f A I A I A I
                           CH2CI2 CH2Br2
Figure 4-7.  Blood CO content of rats after 3-hour  inhalation  exposure with
             1000 ppm dichloromethane, dibromomethane,  and diiodomethane,
             respectively.

             Source:  Fodor and Roscovanu, 1976.
                                   4-24

-------
authors estimated that as much as 70 percent of the inhaled vapor was absorbed;
less than 5 percent was expired as unchanged DCM.   Materials balance indicates
that as  much  as  70 percent of the  amount  absorbed may be converted to CO,,.
Blood COHb  levels  increased  directly with the magnitude  of exposure and did
not reach  a plateau after 7.5 hr of  exposure.   At 200 ppm (695 mg/m ), the
                                                                 ฉ
peak blood COHb level  was 6.8 percent.   At  the  recommended TLV  for  DCM
(100 ppm;  347 mg/m3)  for 7.5  hr, COHb  levels were  about  3 percent  less than
the increase  in  blood COHb levels produced by an  exposure to  CO  at  its  recom-
mended  TLVฎ of  35  ppm.   DiVincenzo and Kaplan  (1981b)  also  observed that
physical exercise resulted in higher blood levels than those found with seden-
tary individuals.   Physical exercise  (Table 4-2)  increased absorption  of DCM,
the biotransformation of DCM to CO, blood COHb levels, and pulmonary excretion
                                                          3
of  CO.   Individuals were exposed to 100 ppm DCM  (347 mg/m ) for 7.5 hr.  The
authors  concluded  that workers performing physical exercise while exposed to
DCM at  the recommended TLVฎ are  unlikely  to  exceed the  COHb  biological TLV
suggested  by the National  Institute of Occupational Safety and Health (NIOSH).
4.2.2.2  Studies in Animals—DiVincenzo and Hamilton (1975) provided the first
information on the  extent  to which DCM  is  metabolized in  rats.  These investi-
gators  injected  rats  intraperitoneally with   C-DCM in corn oil and determined
fate and disposition  of  radioactivity in exhaled  air, urine,  feces, and carcass
1,  8,  and  24 hr after  single  doses ranging  from 412 to  930  mg/kg.   Volatile
compounds  in  exhaled air were collected,  identified,  and quantified by GC  and
radiotracer assay.   Recovery of radioactivity was  essentially 100 percent 24
hr  after administration.   About  98  percent of total  radioactivity was  eliminated
in  exhaled air, and  less than 2 percent  was eliminated in urine or  feces
(Table  4-9).  Some 90 percent  of injected  DCM was eliminated  unmetabolized in
exhaled air.  Most  of  this  elimination (85  percent)  occurred within  2  hr.  Only
2 percent  of  the dose was  metabolized to CO,  3 percent was metabolized to C02,
and 1.5 percent was  metabolized  to an  unidentified volatile compound (Table
4-9).   These  results  indicate  that  less than  7 percent of the dose  is  metabol-
 ized  in the rat.
      Rodkey and Collison  (1977) considered that the small proportion of meta-
bolized DCM found  by  DiVincenzo  and Hamilton (1975) could be  caused by the
 high  dose  of DCM used,  i.e.,  metabolic  transformation  of DCM may be limited by
 the more  rapid  excretion.  Rodkey  and  Collison  carried  out  balance  studies
with  small doses  of  14C-DCM  administered  to  rats (17  mg/kg)  by inhalation  or
                                    4-25

-------
              TABLE 4-9.  FATE AND DISPOSITION OF 14C-DCM IN RATS
                 (412-430 mg/kg) INJECTED INTRAPERITONEALLY
                                            Percent of dose (averages)
                                      2 hr
                 8 hr
               24 hr
Breath

 Unchanged 14C-DCM
 14CO
 14C02
 Unidentified 14C
 Total
84.5
 0.14
 0.55
 0.40
85.59
94.0
 1.43
 1.53
 0.80
97.76
91.5
 2.15
 3.04
 1.49
98.18
Urine

 Unidentified 14C
<0.01
<0.01
 1.06
Feces

 Unidentified 14C

Carcass

 Unidentified 14C
 (mainly in liver)
<0.01
 3.09
<0.01
 2.06
 0.07
 1.53
Source:  DiVincenzo and Hamilton, 1975.


intraperitoneal injection.  The  animals  were placed in a closed rebreathing
system that trapped  C02 and CO after  conversion to CO™ by passing through a
catalyst  bed  of  Hopcalite  .   About 76  percent of    C-radioactivity  was
             14                        14
recovered as   CO  (46.9 percent) and   C09  (28.9 percent). The remaining 24
                                             14
percent could have been exhaled as unchanged   C-DCM,  because no radioactivity
was  recovered  in  carcass tissues.   Their results also showed  that  DCM is
directly metabolized to CO without isotopic dilution.  The extent of the conver-
sion was  surprisingly  great at this low  dose,  and  it is independent of the
mode of administration.   For  each mole of DCM metabolized,  about 0.5 mole of
CO and 0.3 mol  of CO™ were produced.
     In a second experiment, Rodkey and Collison (1977) investigated the rela-
tionship between dose  and extent  of metabolism.  DCM was administered to rats
                                   4-26

-------
 by  intraperitoneal  injection  or by vaporization of  the  dose  in  their closed
 rebreathing system with a CO trap.  DCM doses from 6.8 to 69 mg/kg were given.
 CO  production  and  DCM disappearance were calculated  from  the change in gas
 phase composition,  as determined  by GC.  A  control period was  used to measure
 the endogenous rate of CO production.   When DCM was added to the system, there
 was an immediate increase in the initial rate of CO production (about 35 times
 endogenous for all  doses) and a rapid disappearance of DCM  from  the  gas phase
 (90 percent in 30 minutes).   Carbon monoxide continued to be produced for more
 than 2 hr  after  nearly complete disappearance  of  gaseous  DCM.  Figure 4-8
 shows the rates of  CO production for various doses of DCM.  The  initial rates
 (about 25 umol hr/kg  body  weight) are similar  for all  doses,  but for lower
 doses they progressively decrease  after 1  to 2 hr to the endogenous  rate as
 the DCM dose is metabolized.   For a very high  dose of DCM (69 mg/kg), CO  was
 produced  at a  nearly  constant  rate over a  6-hr period  (COHb,  44 percent).
 These observations  suggest a saturation  of  the metabolizing enzymes even at
 the lowest dose (6.8 mg/kg), giving initially zero-order kinetics followed by
 first-order  kinetics as the DCM concentration in the  inhaled air is decreased
 below enzyme  saturation.   The total amount  of  CO produced was  related to  the
 dose of DCM.   For  lower  doses,  the moles of CO  produced  per mole of  DCM were
 similar and  averaged 0.48;  at a high DCM dose  (69  mg/kg), the  ratio  was 0.62
 after 10.5  hr of exposure,  suggesting  that  substrate-induced enzyme formation
 may occur with  long exposure  to high doses  of DCM.   Similar  results were
 obtained with germ-free rats, obviating  intestinal bacteria  as  a  source of CO.
 In  normal  rats, DCM inhibited  methane  production by intestinal  bacteria.   The
 same  results  were  also obtained with dibromomethane, dichloromethane, bromo-
 chloromethane,  and  diiodomethane in respective  order  of  magnitude and rate of
 CO  production.
      Rodkey and Collison's (1977) important finding that  the metabolism of DCM
 to  CO in rats is rate-limited by enzyme saturation to about 25  umol/hr/kg body
weight  explains the seemingly  low conversion  observed  by  DiVincenzo  and
Hamilton  (1975)  in  this species.   Recalculation of their data (Table 4-5)
gives  a  DCM metabolism to  CO  of about 19 pmol/hr/kg  body  weight.   Yesair
et al. (19?7)  obtained comparable results for mice.   They administered 1.0 and
100  mg/kg   C-DCM in corn oil by  intraperitoneal  injection. Exhaled  14CO,
  C02, and  unmetabolized  14C-DCM  were  trapped   (14CO  after  oxidation with
Hopcalite   to    C02 in aqueous potassium hydroxide and  14C-DCM on coconut
                                   4-27

-------
            200
          en
         .*

         J3)

          o

          E
          =ฃ.
         111
         o
         o
         Q
         Z
         IU
         UJ

         O  100
         CO
         <

         o
         o
             50
                                                         793
                              I
I
                             2             4



                         TIME AFTER CH2CI2 ADDITION, hours
Figure 4-8.  Rates  of production of CO from DCM  given to rats.  Each curve

             represents changes above endogenous CO  rate after the dose

             (in  (jmoles/kg body weight) was given by inhalation.


             Source:   Rodkey and Collison, 1977.
                                   4-28

-------
charcoal) and quantified by GC and radiotracer assay.  The 1 mg/kg dose (11.76
(jmole/kg)  was  quantitatively metabolized  to  CO  (0.45 mol/mol DCM)  and  CO,,
(>0.50  mol/mol  DCM).   In  the  exhaled air collected for 12 hr,  the larger  dose
(11.76  umol/kg)  yielded 4.7  |jmol/kg of  unmetabolized   C-DCM (40  percent
dose),  0.20  mol    CO,  and  0.25  mol  14C02.   Hence,  in mice under  these experi-
mental  conditions,  a  12 [jmol/kg dose of  DCM  (1  mg/kg) does  not  saturate  the
metabolizing enzymes,  whereas 1200  umol  (100  mg) DCM/kg  saturates the enzymes
and is  metabolized at  a constant rate of about 20 umol/hr/kg body weight.  The
remainder of the dose  is excreted unchanged in the exhaled air.
     More  recently, McKenna  et  al.   (1982)  exposed  rats  to 50, 500,  and  1500
    14                               3
ppm   C-DCM (174, 1737, and 5211 mg/m ) for 6 hours.   They also found that the
net uptake  and  metabolism of DCM to CO  and C02 did  not  increase in direct
proportion to the  incremental increase of  DCM exposure  concentration (Table
4-10).   Furthermore,  increasing  amounts  of unchanged  DCM in exhaled air were
found with  increasing  exposure  concentration  (Table  4-10).   At the end of the
6-hour  exposure, the body burden of  DCM was calculated from total radioactivity
recovered during the  first 48 hours following exposure; the body burdens had
not increased to the incremental increase of DCM exposure concentration (Table
4-11).  McKenna et al. estimated the amount of DCM metabolized during each in-
halation exposure by  subtracting the unchanged  DCM  recovered  in  expired  air
(Table  4-10) from the  calculated body burdens.   The  results  are  summarized  in
Table 4-11.
     Of particular  interest  is  the   finding that the percent of  body burden
metabolized decreased  with the  increase of body burden  or increase of the
level  of  DCM exposure.  McKenna et  al.  plotted  the data in  Table  4-11  in
accordance with the following linear form of the Michaelis-Menten equation:
                          dC =
                          dt    "m
dC /
dt/
S + V
                            (4-2)
                                               max
where dC/dt is the |jg/kg DCM metabolized during the fixed time of the experiment
and S  is  the  exposure concentration in parts per million.   Estimates of V
                                                                          max
and K , as determined from the y-intercept and slope, were used with a computer
program of the  Michaelis-Menten  equation to derive the values V    = 65.55 ฑ
                                                                ITI3X
2.54 mg/kg DCM  metabolized  and K  = 493 + 57 ppm DCM.  Deviation from first-
                                 m                _
order kinetics occurred at about 250 ppm (868 mg/m ) or one-half K .   Therefore,
                                   4-29

-------
           TABLE 4-10.   FATE OF 14C-DCM IN RATS AFTER A SINGLE 6-HOUR
                              INHALATION EXPOSURE

Parameter
measured
Expired CH2C12
Expired C02
Expired CO
Urine
Feces
Carcass
Skin
Cage wash
% body

50 ppm
5.42 ฑ 0.73
26.20 ฑ 1.21
26.67 ฑ 3.00
8.90 ฑ 0.39
1.94 ฑ 0.19
23.26 ฑ 1.62
6.85 ฑ 1.62
0.75 ฑ 0.33
burden (x ฑ S.D.

500 ppm
30.40 + 7.10
22.53 ฑ 4.57
18.09 ฑ 0.81
8.41 ฑ 0.90
1.85 ฑ 0.68
11.65 ฑ 1.87
6.72 ฑ 0.13
0.24 ฑ 0.23
, n=3)

1500 ppm
55.00 + 1.92
13.61 + 1.20
10.23 ฑ 1.68
7.20 + 0.74
2.33 + 0.05
7.24 + 0.65
3.97 ฑ 0.15
0.43 ฑ 0.15
 Source:  McKenna et al.,  1982.

 zero-order  kinetics  with saturation of DCM  metabolizing capacity can be ex-
 pected  at  two to three  times K  , or between 1000 and 1500 ppm of  inhaled DCM
                      o        HI
 (3,474  and  5,211 mg/m ).  These values correspond  to  a body burden of about
 100 mg/kg for the rat  (Table 4-11); therefore, they are  in general  accord with
 the observations  of  Rodkey and Collison  (1977) for the  rat  and Yesair et al.
 (1977)  for the mouse.
     Methods  of  determining the kinetic  behavior of DCM metabolism during or
 after  inhalation  exposure  are  subject to unknown  error from the  indirect
 methods  of estimating both body burden and  amount or rates  of metabolism.
 McKenna  and  Zempel  (1981) investigated the kinetics of DCM  metabolism in the
 rat after doses of 1 or 50 mg/kg 14C-DCM.   Table 4-12 gives  the disposition of
 DCM after single oral doses in terms of   C-equivalent.
     The results of this study  clearly indicate that the metabolism of DCM  in
 rats after oral dosing is dose dependent.   Rats given a 1 mg/kg dose metabolized
 a greater percentage of the oral dose  (88 percent) than those given a 50 mg/kg
 dose (28 percent).   Moreover, the rates of pulmonary elimination of unmetabol-
 ized DCM (Figure 4-4)  and of the metabolites C0ฃ and CO were first order and
were essentially  unaffected by  the dose despite large  differences in the
amounts  of  DCM  and of metabolites  excreted.   Therefore,  the dose-dependent
fate of  DCM  is caused  by the saturation of metabolic pathways.  Comparison of
the near-saturating inhalation  body burden (100 mg/kg)  (Table 4-11)  shows a
fair correspondence.
                                   4-30

-------
             TABLE 4-11.  BODY BURDENS AND METABOLIZED 14C-DCM
                IN RATS AFTER INHALATION EXPOSURE TO 14C-DCM
Exposure
concentration
50 ppm
500 ppm
1500 ppm
Total body burden ,a
mgEq 14DCM/kg
5.53 ฑ 0.18
48.41 ฑ 4.33
109.14 ฑ 3.15
Metabolized 14DCMa
mgEq 14DCM/kg
5.23 ฑ 0.32
33.49 ฑ 0.33
49.08 ฑ 1.37
Metabolized
14C-DCM, %
94.6
69.2
45.0
 Values are mean ฑ standard deviation; number of animals in each group = 3.

Source:  McKenna et al., 1982.




      TABLE 4-12.  FATE OF DCM IN RATS 48 HOURS AFTER SINGLE ORAL DOSES
Parameter
Measured
                                           Percent 14C-DCM dose by dose
                                                  concentration
   1 mg/kg
   50 mg/kg
Expired CH2C12

Expired C02

Expired CO

Urine metabolites

Feces

Carcass

Skin

Cage wash
12.33 ฑ 1.43

35.01 + 0.85

30.92 ฑ 1.67

 4.52 ฑ 0.05

 0.93 ฑ 0.02

 5.84 ฑ 0.24

 1.56 ฑ 0.05

 0.53 ฑ 0.04
72.09 ฑ 0.07

 6.33 ฑ 0.39

11.87 ฑ 0.07

 1.96 ฑ 0.05

 0.25 ฑ 0.02

 2.40 ฑ 0.24

 1.15 ฑ 0.06

 0.08 ฑ 0.01
 Values are mean ฑ standard deviation.   Number of animals in each group = 3.

Source:  McKenna and Zempel,  1981.
                                   4-31

-------
     In summary, the metabolism of DCM is dose dependent and follows Michaelis-
Menten kinetics in the rat and mouse.   In these species, saturation of metabolic
capacity with  zero-order  kinetics (about 25 nmol/kg/hr) occurs at about 50 to
100 mg/kg orally or 1000 ppm DCM (3,474 mg/m ) in inspired air for an inhalation
period of 6 hours.  At oral doses of 1 mg/kg or at 50 ppm (174 mg/m ) inhalation,
90 percent is  metabolized, and at enzyme saturation doses 30 to 45 percent is
metabolized.    Dose-dependent metabolism  of  DCM also occurs in man; at 100 to
200 ppm  (347  to 694 mg/m3) inhalation concentration,  50 to 60 percent of the
body burden is metabolized, as judged  from retention values of DCM determined
in man  (Table  4-1).   Experimental  studies in  man suggest that as much as 95
percent  of  the absorbed  dose  may be metabolized at these  exposure levels
(DiVincenzo and Kaplan, 1981a).
4.2.2.3  Effect of Dosing Vehicle—Pritchard  and Angelo (1982) have  described
a  physiologically based pharmacokinetic model (Bishoff model) for mice  and
have used it  to simulate  the distribution, metabolism,  and elimination of DCM
after both acute  and chronic  dosing.   Preliminary  results indicate  that the
kinetics depend on the route and vehicle used for administration.   Administra-
tion of DCM in water by oral gavage or by intravenous injection yields similar
blood and tissue profiles; administration in 50 percent polyethylene glycol/water
shows a rapid  blood elimination and a slow liver elimination, while oral  gavage
with corn oil  as  a vehicle shows a slower rate of tissue clearance than that
found with a water vehicle.
     Withey et al.  (1983) have investigated the absorption of DCM in fasting
rats following oral gavage of equivalent doses (125 mg/kg) in 4 ml of water or
corn oil.  The post-absorptive  peak blood concentration averaged three times
higher  for a  water vehicle than for  corn  oil (121 ug/ml versus  44  ug/ml),
while the time to peak blood concentration  averaged  three times  longer for
corn oil than for the water vehicle  (16.3  min versus 5.2 min).   Aside from
these differences in vehicle-mediated absorption, absorption was  apparently
rapid with both vehicles  and occurred to a comparable extent because the ratio
of the  areas under the blood concentration curves averaged 1.25 for water:oil.
The post-absorption  kinetics  of blood elimination of DCM associated with the
corn oil vehicle  also was extended (tj  was  49 min  for  oil  versus 32 min  for
                                       'T.
water).  These vehicle-related differences in  absorption, kinetics of elimina-
tion, and  tissue  clearance are probably related  to altered transfer of DCM
from oil or water across  the  gastrointestinal (GI)  tract  mucosa.   In contrast
                                   4-32

-------
to aqueous  absorption  into the portal system and then to the liver, corn oil
and other lipids are extensively transported via the mucosal lymphatic system,
which slowly drains by way of the left lymphatic thoracic duct at the junction
of the  internal jugular  and subclavian veins and  hence  into  the systemic
circulation via the superior vena cava.  The absorption, first-pass metabolism,
tissue distribution, and elimination kinetics are probably greatly affected by
the volume  of  lipid vehicle as well as  its nature when introduced into the
stomach  of  a rat.   While these considerations are  unlikely to  affect the
pharmacokinetics of DCM in man in any practical way, they are of importance in
relation to the modes of dosing employed in long-term carcinogenicity tests of
DCM and other lipophilic compounds.

4.2.3  Enzyme Pathways of DCM Metabolism
     Figures 4-9 and 4-10 summarize current knowledge of the enzyme pathways
involved in the biotransformation of DCM and other dihalomethanes.  The scheme
is based on studies  i_n vivo  and i_n  vitro with hepatocytes and with microsomal
and cytosolic preparations.  The preponderance of evidence from ijn vivo experi-
ments indicates that these enzyme pathways are unique to the dihalomethanes
and give rise to both CO  and C02  in  nearly  equimolar amounts.  However, CO  is
an end product of microsomal oxidation, while C02 is an end product of cytosolic
enzyme systems  via the metabolism  of  formaldehyde  and formic acid (Figure
4-9).
4.2.3.1  Microsomal Oxidation—The  primary reaction of  the dihalomethanes
appears to  be an oxidative dehalogenation first described by Kubic and Anders
(1975).   These workers found that DCM and  other dihalomethanes were metabo-
lized by rat  liver microsomal  fractions to CO with inorganic halide release.
The system  required  both  nicotinamide  adenine dinucleotide phosphate (NADP),
reduced  (NADPH), and molecular 02 for maximal activity.   These  experiments
were carried out in  a  closed vessel,  substrates were  added without carrier
solvent,  and CO was  determined by the gas  chromatographic headspace method.
With dibromomethane as the substrate,  3.6 mol of bromide  were  produced per
mole of CO.   In  the absence of NADPH,  microsomal  fractions dehalogenated the
methanes  without CO formation.   Anaerobic conditions substantially reduced the
rate of conversion, although some CO formation (20 percent maximal) occurred.
Equimolar substrate  concentrations  of dichloromethane,  bromochloromethane,
dibromomethane,  and  diiodomethane added to microsomes produced  the  least
                                   4-33

-------
                                     CH2CI2
                                     CH2Br2
                                     CH2BrCI
                                     CH2I2
                       MICROSOMAL
                       MIXED
                      FUNCTION
                     OXIDASE
                    NADPH
                              CYTOSOL
                               GLUTATHIONE
                                 TRANSFERASE
                                  BINDING TO CELLULAR
                                   MACROMOLECU LES
                                    HCHO
          COHb
CO
                                        PULMONARY
                                        ELIMINATION
Figure 4-9.  Enzyme pathways  of  the  hepatic  biotransformation of dihalomethanes.
                                    4-34

-------
Microsomal Pathway
               MFO
NADPH
— H
-x
\
Covalent
OH
Nonenzymatic
rearrangement
r
, H Spontaneous
.r '
llpid 0 Decomposition
protein -H, -X
                                                       CO
                    formyl halide
Cytosolic Pathway
    X - CH
       GSH   	
       transferase
 GS - CH2X
S-halomethylglutathi one
                                          HOH
                                   Nonenzymatic
                                   hydrolysis
                                          GS  -  CH2OH
                                         S-hydroxymethyl glutathione
                            NAD
GS - C
                    formaldehyde
                   dehydrogenase
             H2C=0 + GSH
            formaldehyde
        0
S- formyl glutathione

  HOH   S-formyl  glutathione
      y   hydro lase
  HC "     + GSH
    ^  OH

    formic  acid
 Figure  4-10.
Proposed reaction mechanisms for the metabolism of dihalomethanes
to CO, formaldehyde, formic acid, and inorganic halide.

Source:  Ahmed et al., 1980.
                                    4-35

-------
amount of  CO for dichloromethane, while diiodomethane yielded  the greatest
amount (seven times DCM).  Liver microsomes were 5 times more active than lung
microsomes and  30  times more active than  kidney microsomes.   Hogan et al.
(1976) also found that DCM was converted to CO by rat liver microsomes requir-
ing aerobic  conditions  and an NADPH generating  system.  These workers noted a
high correlation between iji vitro CO production and microsomal  cytochrome P^Q
content.
     Further evidence of the participation  of the  P.rQ mixed-function oxidase
system in  the metabolism of dihalomethanes is  the observation  that dibromo-
methane and  dichloromethane  added to  microsomal cytochrome P45Q preparations
produce type 1  binding  spectra (Kubic and  Anders, 1975;  Cox et al. ,  1976).
However,  Cox et al.  (1976)  found that the  affinity  for P45Q is  less  for DCM
(Ks, 10 mM)  than for chloroform  (Kg, 3 mM) or  carbon tetrachloride (K$, 1.5
mM) although carbon  tetrachloride and chloroform do  not give rise  to  signifi-
cant amounts  of CO  iji  vivo  (Miller  et al., 1973; Kubic  and Anders,  1975;
Rodkey and Collison,  1977).   Both Kubic and Anders  (1975)  and Hogan et al.
(1976) found that  jji vivo phenobarbital pretreatment  induced  additional  ijn
vitro  CO  production, while  cobaltous  chloride, which  depletes microsomal
cytochrome PซKQ>  reduced microsomal CO production.   Furthermore,  SKF 525A,
ethylmorphine,   and hexobarbital  (type 1 substrates)  inhibited iji vitro micro-
somal conversion of dibromomethane to CO (Kubic and Anders, 1975).
     Kubic and Anders and coworkers (Kubic  and Anders, 1978; Ahmed and Anders,
1978;  Stevens and  Anders, 1978,  1979) have  studied the mechanism of the reac-
                                     -i o
tion of DCM  with deuterium and with   02-  The reaction shows a  prominent
deuterium  isotope  effect with  a  comparative rate of about 12 percent of that
of  the hydrogen isotope, indicating that carbon-hydrogen  bond breakage is  the
                                  18
rate-limiting step.   Studies with  02 showed that the  oxygen appearing  in CO
is  derived from molecular oxygen rather than from water.  On the basis of
these  studies,  Kubic and Anders  and coworkers proposed  the  reaction mechanism
shown  in  Figure 4-10.   P45Q-mediated hydroxylation  of  dihalomethanes yields
the intermediate, hydroxydihalomethane (X2CHOH), which spontaneously rearranges
to  a  formyl  halide (XCHO) with  the loss of one halogen  atom.   The resulting
formyl halide is known to readily decompose to yield CO.
4.2.3.2   Cytosolic Pathways--In   addition to metabolism  to CO, DCM is also
converted  to formaldehyde,  formic acid,  inorganic halide, and CO,,.   Kuzelova
and Vlasak (1966) detected formic acid in the urine  of DCM-exposed workers and
                                   4-36

-------
 suggested that DCM was metabolized via formaldehyde to formic acid.  Originally,
 Heppel and  Porterfield  (1948)  reported the conversion of dibromomethane  and
 bromochloromethane  to  stoichiometric amounts  of  formaldehyde and inorganic
 halide by a 9000-g supernatant fraction of rat liver and by liver slices and
 homogenates.  The system did not require 02> but it was glutathione dependent.
 Kubic and Anders  (1975)  more recently confirmed these findings and localized
 this metabolic pathway to the cytosol.  Ahmed and Anders  (1976, 1978) extended
 these findings and showed that the cytosol  system is a glutathione transferase
 that is found only in the liver; it requires no cofactor  other than glutathione
 (cysteine is not  a substitute)  and is not  inducible  by  phenobarbital  or by
 repeated exposure  to  DCM or dibromomethane.   Furthermore,  the  reaction was
 inhibited by  reagents  that  react  with sulfhydryl groups,  such  as diethyl
 maleate and parachloromercuribenzoate, as well as known substrates for gluta-
 thione transferases.  The substrate  order  of activity is diiodo > dibromo =
 bromochloro  > dichloromethane;  this order  is the  same  as  that found for oxida-
 tive dehalogenation by Kubic and  Anders  (1975).   This pathway probably does
 not  contribute to CO production  via metabolism of formaldehyde  to CO because
 formaldehyde administration  does  not produce an increase  of COHb  in animals  or
 humans (Kubic et  al.s  1974;  Rodkey  and Collison, 1977;  Kasselbart and Angerer,
 1974).
      The  cytosolic pathway has been investigated in  detail  by Ahmed and Anders
 (1976,  1978),  who have proposed the  reaction  sequence shown  in  Figure 4-9.
 These workers observed that both dibromomethane and bromochloromethane have
 identical  kinetic constants, suggesting that  the  initial and rate-limiting
 step  is  a displacement of halide,  with  glutathione interaction,  to give the
 conjugate S-halomethylglutathione.   This  conjugate is postulated  to  undergo
 rapid nonenzymatic hydrolysis to produce S-hydroxymethylglutathione and thus
 to yield  formaldehyde and regenerate  glutathione.   Because the addition of
 nicotinamide  adenine dinucleotide  (NAD+)  to incubation mixtures  decreased
yields of formaldehyde (Ahmed and  Anders, 1976), the postulated  intermediate,
S-halomethylglutathione,  a known substrate of the hepatic cytosolic enzyme for-
maldehyde dehydrogenase,  also undergoes conversion to S-formylglutathione, and
with  enzymatic  hydrolysis (S-formyl  glutathione hydrolase) yields  formic acid
and regenerated glutathione.  Ahmed and Anders (1978)  found that  removal of
formaldehyde dehydrogenase from  the cytosolic fraction by precipitation with
ammonium  hydroxide  resulted  in  production of only  formaldehyde.   Thus, the
                                   4-37

-------
proposed  reaction  sequence for the metabolism of dihalomethanes accounts for
the  experimentally observed  stoichiometric  ratio of two  inorganic  halides
formed to  one formaldehyde and the requirement  (but  nonconsumption)  of  gluta-
thione.   Furthermore,  the sequence serves as a  detoxification  mechanism  for
dihalomethanes that  depend on cellular availability of  glutathione,  and  the
cellular  availability  of  NAD   determines  the  ratio of the  products  (formalde-
hyde/formic acid) formed.
4.2.3.3   Carbon Dioxide Formation—The  cytosolic glutathione  transferase
system for  dehalogenation of  dihalomethanes with the production of formalde-
hyde and  formic  acid appears  to be the major source of C0? as an end product
of the metabolism  of DCM  in  the  intact  rodent  (Rodkey and Collison, 1977;
DiVincenzo  and Hamilton,  1975; Yesair et al.,  1977; McKenna  et al. , 1982).
Neely  (1964)  has demonstrated  that formaldehyde is  almost quantitatively
metabolized to C02.  When Neely injected    C-formaldehyde  (14CH20)  intraperi-
toneally into  rats  at  dose levels  of  0.25  and  2.5  pmol/kg body weight,  he
recovered 82 percent of the dose as C02 in exhaled air collected over a 24-hour
period.  Peak  concentrations  of   CO- in exhaled air occurred  1 hour after
administration.  Neely  suggested  that the  formation of COp  occurred from
formaldehyde entering the 1-C metabolic  pool  with transfer to glycine by the
folic acid  cycle to  give  serine.  Transamination of serine to  pyruvate pro-
vides entry to the  tricarboxylic acid cycle  and  completes the  oxidation to
CO
                                                            14,
  2.  In  support  of  this  metabolic  route,  small  amounts  of    C-labeled  serine
and methionine were found in the urine.  However, when   C-DCM was administered
             14
to  rats,  no   CH20 was detected  in the breath, serum,  or  tissues 2 hours
later, although substantial  changes in tissue  formaldehyde  content were  noted
(Rodkey and Collison, 1977; DiVincenzo and Hamilton, 1975).
     An additional source of C02 production from the  metabolism of dihalo-
methanes is the j_n vivo oxidation of CO to C02-  Carbon monoxide is metabolized
to C02 by  various animal  tissues (Fenn, 1970).  Luomanmaki  and Coburn (1969)
have also demonstrated that   C09 exists in the expired air of humans breathing
14
  CO during a  4-hour period in a closed rebreathing system.  Carbon monoxide
is metabolized by combining with the reduced form of tissue cytochrome oxidase
in the presence of a low 02 tension,  and  is released  as C02  (Coburn, 1970).
However,  the amount  of C02  generated by oxidation of CO  is  very  small because
the rate of conversion  is less than 5 percent endogenous CO from heme catab-
olism (Tzagoloff and Wharton, 1965).  On the other hand, the metabolic rate of
                                   4-38

-------
conversion of CO to CO™ appears to increase as a function of body stores of CO
and thus as  a  function of blood COHb  concentration.   In dogs, at  10 to 15
percent COHb, the metabolic conversion rate equals the formation of endogenous
CO (Luomanmaki and  Coburn,  1969).   Increased CO production and elevated COHb
that results from catabolism of DCM may stimulate the metabolic production of
COp from CO, thus  contributing to the total  C02 produced by DCM metabolism.
4.2.3.4  Pathway Utilization Ratio—The  microsomal  oxidative  dehalogenation
and cytosolic  glutathione transferase dehalogenation  systems (Figure 4-9)
account for the CO and CO,, generated from the metabolism of the dihalomethanes.
Because the microsomal system is apparently saturated and rate limiting at low
doses  (Section  4.2.2), the  relative  molar amounts of CO and  C02  produced
should provide an index of the activity of the two pathways.   However, Yesair
et al. (1977)  found nearly equimolar amounts of CO and CO,, with both low and
high saturating  doses of DCM  in mice.   For  low doses in rats,  Rodkey  and
Collison (1977) found 1.6 times as much CO produced as C0?,  suggesting greater
metabolism (at low  doses)  by the microsomal   oxidative pathway.  McKenna and
Zempel  (1981) observed a  CO:CO?  ratio of 0.9  for  a  low oral  dose of DCM (1
mg/kg) to rats and a ratio of 1.9 for high saturating dose (50 mg/kg), indica-
ting a greater utilization of  the microsomal  oxidative pathway at "saturating
doses" (Table  4-12).   However, McKenna et al.  (1982)  found no significant
preference for either  pathway  (Table  4-10) for a low inhalation dose of DCM
                    3
(50 ppm or 174 mg/m )  and a metabolic  saturating inhalation dose (1500 ppm or
5211 mg/m ).   Clearly, the important factors  of hepatic content of glutathione
and of Pป5Q  play  major roles in the relative utilization of the two pathways
of metabolism.
     Though it has  been  observed that equimolar doses of dibromomethane and
diiodomethane produce  COHb concentrations  greater,than those  produced by DCM
in rats (Fodor and  Roscovanu,  1976; Roth et  al. , 1975;  Rodkey and  Collison,
1977),  no information is  available on the ratio of CO to C02 produced by these
compounds in microsomal preparations  (Kubic  and Anders,  1975) and in cytosol
preparations (Ahmed and Anders,  1976).  The  use of isolated hepatocytes may
prove useful and  avoid many  of the difficulties  inherent with whole animal
experiments.
                                   4-39

-------
4.3  DCM-INDUCED CHANGES IN HEPATIC ENZYMES
     The metabolism of the dihalomethanes  by the microsomal oXidative dehalo-
genation pathway  (but apparently not the cytosolic pathway) can be modulated
by inducers of the microsomal mixed-function oxidase system.  Pretreatment of
animals with phenobarbital was  found by some workers to  increase  blood COHb
levels and microsomal production of  CO  (Kubic et al., 1974; Kubic  and Anders,
1975; Hogan et al., 1976; Stevens et al., 1980), but other investigators found
no effect or observed  a decrease (Miller  et al. ,  1973;  Roth et al., 1975).
Roth et al.  (1975) suggested that enhanced metabolism may be initially induced
by phenobarbital, but the resulting  increase in local microsomal levels of CO
may  be sufficient to  inhibit cytochromic  P45Q oxidative  dechlorination.
Heppel and Porterfield  (1948) also reported that  repeated  administration of
bromochloromethane to rats led to an increased rate of dehalogenation.   However,
Haun et al. (1972) found that continuous exposure  of mice to 100 ppm DCM (347
mg/m ) for 4 to 12 weeks decreased the hepatic cytochrome P45Q content.
     Daily exposures for shorter periods do not appear to  influence hepatic
P/icn content,  but they do modify the activity of other enzyme systems.   Kurppa
 4ou                                                                       3
and Vainio (1981)  exposed  rats  at 500 and 1000 ppm DCM (1737 and 3474 mg/m )
6 hr/day for 5  and 10 days.   They reported no change in  hepatic P45Q content
but they observed a  marked decrease of NADPH-cytochrome  c (35 percent) and a
twofold increase  in UDP-glucuronosyl transferase.   The  hepatic glutathione
content remained  unchanged. Toftgard et al. (1982) exposed  rats to 500, 1500,
and 3000 ppm DCM  (1737,  5211, and  10,422 mg/m3) 6  hr/day  for 3  days  and found
no change  in  hepatic P45Q content, but a dose-related increase of microsomal
metabolism of  biphenyl   and  of  benzopyrene was noted.   These  changes were
postulated to be caused  by a  change in the proportions of different  cytochrome
p     isozymes  resulting  from DCM-inducing effect  on  some specific  forms of
 tOU
cytochrome Pป5Q-   In  contrast to these observations, Pritchard et al. (1982)
recently administered DCM to  male  mice  for 3 days  by gavage (5, 50,  100, 250,
500, 1000 mg/kg doses in corn oil) and found no significant changes  in hepatic
weight, microsomal  protein,   P450> cytochrome bg  contents,  or activities of
aminopyrene N-demethylase and biphenyl 4-hydroxylase.  Furthermore,  28 days of
administration of DCM  in drinking  water, providing daily doses  ranging  from 5
to 1000 mg/kg,  also did not  affect  these  parameters or  hepatic glutathione
content.
                                   4-40

-------
 4.4   COVALENT BINDING TO CELLULAR MACROMOLECULES
       The  likelihood of  significant covalent binding  of reactive metabolites
 from  the metabolism of DCM to cellular macromolecules depends on  the postulated
 reactive  intermediates:   formyl  chloride from  microsomal  oxidative metabolism
 and  S-chloromethyl  glutathione  and  formaldehyde from  cytosolic  metabolism
 (Figure 4-10).   These  compounds  may be  capable of acylating cellular nucleo-
 philes.  S-chloromethyl glutathione  is  structurally similar to  the reactive
 bis-halomethyl ethers.
      Anders et al.  (1977)  have studied  the extent and  pattern of binding  to
 microsomal lipid and protein after aerobic incubation of rat hepatic microsomes
 with   C-DCM.   Table 4-13 shows that metabolites  of DCM become  covalently
 bound to both microsomal protein and Tipid under conditions optimal for meta-
 bolism of DCM to CO.  Furthermore,  microsomes  from rats pretreated with pheno-
 barbital  showed increased  binding.   Thus, the  formyl  chloride intermediate  may
 either acylate macromolecules or  decompose to  CO.
      Cunningham et  al.  (1981)  have  investigated covalent binding of  inter-
 mediates  from the metabolism of  14C-DCM  by rat hepatocytes.   Rat hepatocytes
 in suspension  have been  shown to  metabolize DCM and other  dihalomethanes  to CO
 (Stevens  et al. , 1980).   The results  of  irreversible binding of  14C-DCM to
 cellular macromolecules  in this system, in comparison  to 14C-carbon tetrachlo-
 ride,  are  given  in  Table 4-14.
      The  covalent binding  of carbon tetrachloride  to  lipids and protein was
 greatly enhanced  in the absence  of  02  and was  almost eliminated in the pre-
 sence  of  02 (Table 4-14).   In contrast,  binding of DCM  and trichloroethylene
 was enhanced in the  presence of oxygen; this is consistent with the  microsomal
 oxidative metabolism of these compounds to an epoxide and formyl chloride,  re-
 spectively. Jlutathione-depleted hepatocytes showed markedly decreased covalent
 binding of   C-DCM to both lipids and protein, suggesting inhibition of cytosolic
 DCM-glutathione conjugation with  a decreased production  of reactive S-chloro-
 methyl glutathione and of formaldehyde.   However, at least part of the decrease
 in binding  may be  caused by diethylmaleate inhibition  of microsomal  oxidation
 (Stevens et al. ,  1980)  as  well  as by diethyl  maleate depletion of cellular
glutathione.  Phenobarbital pretreatment  also markedly decreased binding  from
  C-DCM hepatocyte metabolism.  The explanation of this  effect is  not readily
apparent because phenobarbital  is  not  known to  deplete cellular glutathione,
but it does increase DCM microsomal oxidative metabolism to CO in  hepatocytes
                                   4-41

-------
               TABLE 4-13.   IN VITRO COVALENT BINDING OF 14ODCM
                       TO MICROSOMAL PROTEIN AND LIPID
Conditions
  nmol 14C bound/mg/min ฑ SD
 Protein                Lipid
Normal rat microsomes

Microsomes from pheno-
  barbital-treated rats
0.24 ฑ 0.02
0.57 ฑ 0.03
                                                                 0.27 ฑ 0.02
1.97 ฑ 0.19
S.D. = standard deviation.

Source:  Anders et a!., 1977.
TABLE 4-14.  COMPARATIVE COVALENT BINDING OF DCM, CARBON TETRACHLORIDE (CC14),
      AND TRICHLOROETHYLENE (TCE) TO LIPID AND PROTEIN IN RAT HEPATOCYTES
Atmosphere3
(02/N2)
Substrate
CCฃ4
TCE
DCM
Protein
0.09
9.63
8.53
Lipid
0.06
5.08
11.96
Glutathipne
Depletion0, %
Protein
100
74
44
Lipid
100
74
38
Phenobarbitalc
induction, %
Protein
145
288
44
Lipid
120
238
40
 aRatio of covalent binding observed under oxygen and nitrogen atmosphere.
 Values are the mean of three to six determinations for each condition.

 bThirty minutes prior to hepatocyte isolation,  rats were treated  intraperi-
 toneally with 0.6 ml/kg of diethylmaleate.  Values are expressed as the mean
 percent of binding observed in hepatocytes  from untreated  rats,  with  N=5.

 cRats were pretreated with three consecutive daily doses of phenobarbital
 (80 mg/kg, intraperitoneally) beginning 4 days before isolation  of hepatocytes.
 Values are expressed as the mean percent of binding observed in  hepatocytes from
 untreated rats, with N=5.

 Source:  Cunningham et al., 1981.
                                    4-42

-------
 (Stevens et  al.,  1980).   Perhaps  the most  important observation made by
 Cunningham et al.  (1981)  was that 14C-DCM metabolism by isolated hepatocytes
 did not result  in  the alkylation of the  nucleic  acids RNA or DMA, whereas
 metabolites of  carbon tetrachloride and  trichloroethylene labeled nucleic
 acids under the conditions of the experiment.
      Reynolds  and Yee  (1967)  studied labeling patterns of  14ODCM  and 14C-
 formaldehyde in rat liver after in vivo injection.   They found similar patterns
 of labeling of DCM  and its metabolite, formaldehyde.  Binding occurred most
 often at the ami no acid locus corresponding to serine and on the .acid-soluble
 cell  constituents;  smaller amounts  occurred  in  lipid  and  nucleic acids.
 However,  formaldehyde or formic acid can  directly  combine  with tetrahydrofolic
 acid  and consequently  be  incorporated  into  de novo nucleic acid  synthesis;
 therefore,  the association of 14C-activity with nucleic acids does not neces-
 sarily  indicate  covalent binding.
 4.5   KINETICS OF  CARBOXYHEMOGLOBIN  FORMATION
      Blood  COHb  accumulates  when  the  amount  of endogenous or  exogenously
 derived  CO  in  the  body exceeds that of pulmonary elimination.   Pulmonary
 elimination  of  CO  is  a first-order  process that  involves the exchange of
 hemoglobin-bound  CO with oxygen and  the diffusion of CO across  capillary and
 alveolar  endothelium into  alveolar lung spaces.  In humans, the half-time of
 elimination of  CO is 4 to  5 hours  (NIOSH,  1972;  Lambertsen,  1974), and in  the
 rat  the  time  is 1.8 to  2.5 hours  (McKenna et  al.,  1982; McKenna and Zempel,
 1981).  The  half-time  of pulmonary elimination  is  independent of the blood
 COHb  concentration  but  is  dependent on factors such as  lung function and
 dysfunction, pulmonary  rate and amplitude, regional blood  flow,  and cardiac
 function.
     Ordinarily,  the sole  endogenous source of  CO,  and hence COHb,  is the
 physiologic catabolism of heme by the hepatic microsomal heme oxygenase pathway
 (Coburn,  1973;  Tenhunen  et al. , 1969).   The endogenous rate of CO production
 from this source  in the normal  human is about 20 pmol/hr, producing a blood
 COHb concentration  of  approximately 0.4 percent.  A close linear correlation
 exists between the molar rate of CO production  and the percent COHb saturation;
 only 10 to 15 percent of the  total  body CO  is not associated with  hemoglobin.
Most of this 10 to  15 percent is bound to  hemoproteins  such as myoglobin  and
                                   4-43

-------
       O)
       a.
       UJ

       UJ

       JQ
       X
       O
       O
                                                            CHoCIo, ppm
                                                            O—O 500
                                                            O	O 100

                                                                I I
                                                                I I
                                                                I
                                   EXPOSURE, days
Figure 4-11.
Blood COHb level in men during  an  8-hour exposure for 5 consecu-
tive days to 500 ppm and 100  ppm DCM.   COHb percent saturation is
equal to ug CO per ml blood divided  by 2.5.

Source:  Fodor and Roscovanu, 1976.
                                     4-44

-------
heme cytochromes;  about  1 percent is dissolved  in  body water.   The rate of
endogenous CO production from heme catabolism is markedly inhibited by exogenous
CO  sources  producing COHb  levels of approximately 12 percent saturation,
suggesting that blood levels of 12 percent saturation are sufficient to inhibit,
by  CO  binding,  hepatic  microsomal  oxidase  systems involved in hemoglobin
degradation to CO (Coburn, 1970).   The half-life of COHb (4 to 5 hours) arising
from heme  catabolism or  exogenous  environmental  CO (e.g., ambient-air CO,
tobacco smoking)  is  decreased  by  increased alveolar ventilation or increased
inspired partial pressure of oxygen (Lambertsen, 1974).
     Since Stewart and his associates (1972a,b) reported the remarkable increase
of  blood COHb  (up to 15 percent  from 0.6  percent pre-exposure) in persons
acutely exposed by  inhalation  to  DCM vapor, numerous  investigations  of the
phenomenon have been  made in experimental  animals  and  humans.  Studies have
sought to determine  the  dose-response relationship of  blood COHb level with
DCM air concentration, with duration of exposure, with the time course of COHb
blood concentration  rise  and decline,  and with the magnitude of exposure  in
the industrial  setting.   Because CO generated from DCM is additive to exogenous
environmental CO,  DCM exposures at high levels could pose an additional health
burden.   Of  particular concern  are smokers who maintain significant constant
levels  of COHb, i.e.,  4.6 to 5.2 percent  (Stewart  et al., 1974a,b; Kahn et
al., 1974) and  others who may have  increased sensitivity to CO toxicity, such
as  pregnant  women  and persons  with cardiovascular  disease or pulmonary dys-
function.   Indeed, Stewart  et  al.  (1972a,b) have noted that exposure to con-
                                                        S)                   O
centrations of DCM that do not exceed the industrial TLV  (100 ppm,  347 mg/m )
may yield COHb  levels  exceeding those allowable from exposure to CO (35 ppm,
         3
40.1 mg/m ;  about 5 percent COHb blood concentration).
     As previously discussed,  the recent findings  of  DiVincenzo  and Kaplan
(1981a,b) with  sedentary  individuals  and"those engaged in physical exercise
while exposed to  DCM  indicate that blood levels of  COHb are increased  but  are
within  the biological TLVฎ  recommended  by the National Institute for Occupa-
tional  Safety  and Health (1976).    The  National  Institute for Occupational
Safety  and Health  has recommended  that the time-weighted average (TWA) exposure
to  DCM should not produce COHb levels that exceed 5 percent.   DiVincenzo and
Kaplan  (1981a)  found  that an 8-hour exposure to  150  ppm  DCM (521 mg/m3) in
sedentary individuals is  equivalent to an 8-hour exposure to 35 ppm CO; either
exposure has been shown to result in blood levels of COHb of about 5 percent.
                                   4-45

-------
 In their  companion  study in  which individuals either exercised  or  smoked
 cigarettes during exposure,  DiVincenzo and Kaplan  (1981b)  observed an additive
 increase in blood COHb concentrations.   Individuals  were  exposed for  7.5  hours  to
 100 ppm DCM (347 mg/m ).  The effects of exercise on physiological parameters
 are presented in  Table  4-2.   Moderate to heavy workloads  produced about a
 twofold increase in blood COHb  levels compared to the  increase  for sedentary
 individuals.
      From  a review of the literature, an important observation of DCM-induced
 COHb  is that the  blood  concentration,  although it is DCM-dose dependent  and
 exposure-time  dependent, never exceeds 10 to  12 percent in either humans or
 animals in usual  circumstances of  free breathing and normal pulmonary function
 (Figures 4-5, 4-6, 4-11).  This  limiting  blood  concentration  is  clearly deter-
 mined by the resultant of first-order pulmonary elimination of CO  and zero-order
 kinetics of hepatic  DCM metabolism to  CO (Section 4.2.2).   Because  COHb  is
 essentially  confined to the  blood compartment,  a one-compartment open kinetic
 model with zero-order input  can be used  to describe  the time course  of blood
 COHb as follows:
                              V
                                                                      (4-3)
where V  is  the  volume  of the  hemoglobin  compartment (90  percent  CO  is  distri-
buted in this compartment) (Luomanmaki and Coburn, 1969), k  is the zero-order
rate  of  COHb formation,  kg is  the  first-order rate constant for pulmonary
elimination  of  CO  from COHb,  and C is the the concentration at any time t of
COHb formed. Then,
                           _  ko
                     COHb,
Vk
                                           -k  t
1-e
(4-4)
This equation  (Wagner, 1975) describes the  time  course of  rising COHb concen-
tration with zero-order  formation  of COHb.   With long periods of exposure to
DCM (6 to 8 hours), the relation becomes
                          COHb,
    = VVke
                                                                      (4-5)
                              -   . 4-46

-------
That  is,  a steady-state plateau, concentration  of blood COHb is reached (see
Figure  4-12).   Therefore,  maximal  blood COHb  is  mandated when K ,  V,  and k
                                                                 o          e
are constants.

4.5.1   Studies  in  Humans
      In  a series  of  studies, Stewart and  his  associates (Stewart et al. ,
1972a,b,  1973,  1974a,b;  Forster et al. , 1974;  Hake et al. ,  1974) have shown
that blood COHb levels achieved  in response to DCM  exposure are  related to the
inhaled  concentration  and  to the duration of exposure.   Male  nonsmokers were
exposed to DCM  for 1, 2, and  7.5 hours daily, for 5 days  per week.  Blood COHb
levels  were  determined for daily pre-exposure  and post-exposure times.   Their
data, which  are replotted  in part in  Figure 4-5,  indicate  that maximum  COHb
concentrations  occur with 400 to 500 ppm DCM (1390  to 1737 mg/m3) exposure and
increase  with  duration of  exposure.   Similar results  were  reported in women
nonsmokers exposed 1, 3, and  7.5 hours to 250 ppm DCM (868 mg/m3) for 5 consecu-
tive  days  (Hake et al. , 1974) and in male  volunteers  exposed  to 100 and  500
                           3                                        i
ppm DCM (347 and 1737 mg/m ) daily for  5  days (Fodor  and  Roscovanu,  1976)
(Figure 4-11).   In each of these studies,  the  time course of decay of COHb
levels  to  pre-exposure  levels occurred within 24  hours so  that a consistent
increment in COHb with daily  exposure was not observed  (Figure 4-11); however,
the half-time  of  COHb  disappearance  was significantly  longer  than  expected.
The longer  apparent half-time  of  pulmonary elimination  of  CO produced by
hepatic metabolism of  DCM  results  from storage of DCM  in adipose tissue with
conversion to  CO  continuing subsequent to termination  of exposure.   In these
circumstances,  the biological  half-life  of COHb derived  from  DCM (10  to 15
hours)  is proportional  to  the body burden of DCM,  in contrast  to the constant
half-life of 4 to 5 hours from CO inhalation (Stewart et al., 1972a,b,  1976a,b;
Fodor et  al.,  1973;  Peterson, 1978;  Peterson and  Stewart,  1970).  However,
when workplace  exposures were simulated,  the biologic  half-life for COHb was
similar to that for CO (DiVincenzo  and Kaplan,  1981a).
     DiVincenzo and Kaplan  (1981a) found that blood COHb  saturation in seden-
tary male volunteers  is  not attained during a  7.5-hour exposure to DCM at
concentrations  ranging from  50  ppm  (174 mg/m ) to 200 ppm (695 mg/m3).  Peak
blood COHb during  the 200-ppm exposure was 6.8 percent.   At the  TLVฎ for DCM,
the authors  estimated  that an 8-hours exposure would  produce  a blood COHb
level  of  about  3 percent.  A  linear relationship was found between  blood COHb
                                   4-47

-------
   100.0
e>
u
I
6
1
    10.0 —
'   I   '   I   '   I  '   I   '   I   '=
     0.1
                      •EXPOSURE'
                                                 A 50 ppm
                                                 • 500 ppm
                                                 • 1500 ppm
                                      i   .   i  i  i  .  I   .   I
        0123456/01234

                                  TIME, hours

        Figure 4-12. Blood HbCO concentrations in rats during and after a
        6-hour inhalation exposure to DCM. Each data point is the mean
        ฑ standard error for two to four rats.
        Source:  McKenna et al. (1982).
                                 4-48

-------
and DCM exposure concentrations.   Repeated exposure to DCM levels of 100,  150,
                                 3
or 200 ppm (347, 521, or 695 mg/m ) for 7.5 hours daily for 5 consecutive  days
produced slightly higher  blood  COHb levels than those found for single expo-
sures.  Following  the weekend, blood  COHb  levels returned to pre-exposure
levels.  Blood  COHb declined at  a slower rate than DCM  in  expired breath
during post exposure.   About  24 hours were required for blood COHb to return
to preexposure values.
     The findings in  a  companion  study by DiVincenzo and Kaplan (1981b) with
smokers and individuals engaged  in physical activity indicated" that exposure
                         3
to 100 ppm  DCM  (347 mg/m ) may result  in slightly higher blood COHb than  in
sedentary nonsmokers.   The investigators concluded  that  workers  performing
                                                           3
physical  exercise while exposed to  DCM  at 100 ppm (347 mg/m  ) are  unlikely to
                               ฎ
exceed the  COHb biological TLV   recommended by the National Institute for
Occupational Safety and Health (1976).
     Ratney et  al.  (1974)  studied the blood COHb  levels  (calculated from
alveolar concentrations)  of a group of young male adults in their workplace
where  large quantities of DCM were  used (a  plastic-film plant).  Workroom  air
                                                3
concentrations of DCM averaged 200  ppm  (695 mg/m  ) with no measurable CO.   At
the beginning of the  work day, blood COHb levels averaged 4.5 percent.  After
an 8-hour exposure,  COHb  levels rose to about  9  percent and then declined
exponentially to 4.5  percent  by the next working  day (16 hours later) with an
apparent half-time  of CO  pulmonary elimination of 13 hours.   These prolonged
half-times  for  CO  in alveolar air  and for COHb in  blood are  difficult to
interpret in  light  of the contrasting  results of  Stewart et  al. (1972a,b)  and
DiVincenzo  and  Kaplan (1981a,b).    Ratney et al.  (1974)  reported that  pre-
exposure levels of CO in expired air and COHb in blood were elevated above the
values expected  for nonsmokers.   Three of  the  seven individuals that  were
monitored were not previously exposed to DCM; therefore,  it is surprising  that
the CO levels  in  their  alveolar air and  blood  COHb values were as high as
reported in the paper.   Ratney et al.  (1974) reported a mean pre-exposure CO
concentration in expired air of about 29 ppm.  Normal levels of CO in alveolar
air are expected to range between 1 and 5 ppm.
     The studies of Stewart and associates, Fodor and Roscovanu, and Ratney et
al. are consistent  in their findings and lead  to the  following conclusions.
     1.   COHb derived  from CO  of DCM metabolism is additive to COHb derived
from CO of exogenous sources.
                                   4-49

-------
     2.   Blood COHb concentration attained from DCM metabolism is a function
of both DCM  inhalation concentration and of exposure duration, with a maximal
attainable concentration of 10 to 12 percent COHb.
     3.   A steady-state blood COHb concentration in sedentary individuals is
not attained within 8 hours for single exposures of DCM.
     4.   The half-time of first-order pulmonary elimination of COHb is 4 to  5
hours, although a longer pseudo half-time of 10 to 15 hours is observed because
of continued post-exposure  metabolism  of DCM, from solvent stored in the fat
compartment during exposure.
     5.   Blood COHb  concentrations  from  multiple  daily exposures reach a
steady-state level within  3 to 5 days, and the level is only slightly higher
than that after a single 8-hour exposure.
     These observations on the kinetics of COHb formation during DCM inhalation
exposure are consistent  with Michaelis-Menten kinetics for DCM metabolism to
CO (Section  4.2.2).  The data plot of Stewart  and  associates of Figure 4-5
(7.5-hour exposures) illustrates  dose-dependent formation of COHb and  is of
the form of the Michaelis-Menten equation:
                                dC
                                dt
               V    S
                max
(4-6)
where dC/dt  is the  rate  of  COHb  formation  and  S  is  the  exposure  concentration
in parts per million.
the data in accordance with the linear form of the equation.
Estimates of V     and  K  were  obtained  from  plotting
                                     K
              m
          V    + S
           max
                                                max
(4-7)
                         1/dC
                           dt
This form  provides  values of V    =  15% COHb/7.5 hr and K   =  200  ppm  (695
    A                          'iTlcLX                         Ml
mg/m ) DCM.   These  values indicate a maximum  obtainable  COHb blood level  in
humans caused by  DCM exposure  in  air  of  about  25  percent, and a saturation of
                                                                      3
hepatic  metabolism  of DCM to  CO  in  humans at 400 ppm  DCM  (1390 mg/m ) in
inhaled air (2 x KJ.
     The COHb blood  levels resulting from DCM  inhalation are  dependent on pul-
monary function status as well as on metabolism of DCM to CO.   Physical activity
or exercise during exposure to DCM markedly increases pulmonary absorption and
body retention  (Section  4.1.3) but tends  to diminish  the maximum blood  COHb
                                   4-50

-------
level achieved by, the  end of the exposure period, although higher blood COHb
levels are attained  3  to 4 hours after  exposure  when compared with control
sedentary subjects (Stewart  and  Hake,  1976; Astrand et al.,  1975; DiVincenzo
and Kaplan, 1981b).  These findings may  be  explained  by the decrease in half-
life of pulmonary  CO elimination affected by the  increased pulmonary ventila-
tion rate  and increased  cardiac  output associated with  physical activity
(Lambertsen,  1974).  Conversely, compromised  pulmonary function (e.g., adult
respiratory distress  syndrome,  emphysema,  and  asthma) can be expected to
increase COHb  blood  levels  by  decreasing  pulmonary  elimination  of CO.   A
comprehensive kinetic model describing the parameters of absorption, distribu-
tion, and elimination  of DCM  in humans,  as  well as metabolism  to  CO and COHb,
would be valuable  in evaluating the effects of physical  activity,  pulmonary
and cardiovascular diseases,  and concomitant environmental xenobiotic exposures,
including drugs  such as alcohol and barbiturates,  of which  very little  is
known.  Hake  (1979)  has developed a computer simulation  model, based  on  the
Coburn-Forster-Kane  equation, describing the kinetics of  COHb  formation using
the experimental data of Stewart and his associates.  He was able to show that
physical activity  during exposure  to DCM led to  a  lower  blood COHb than  the
sedentary level  because while increased ventilation  increased DCM  uptake, it
also increased the pulmonary elimination rate constant (k ) for CO.

4.5.2  Studies in  Animals
     The kinetics  of COHb formation from DCM exposure elucidated  in humans
have  largely  been confirmed  in animals.    Figure 4-6 shows the relationship
between exposure concentration  and COHb levels in  rats  reported  by Fodor et
al.  (1973).   For a 3-hour exposure,  maximal levels  of COHb  (12.5  percent) are
achieved with  about  1000 ppm DCM  (3,474 mg/m  ).   Fodor  and Roscovanu  (1976)
                                                        3
noted that  a  3-hour  exposure with 200 ppm  DCM (695 mg/m  ) in  humans produced
COHb levels of about 4.3  percent but they found nearly twice this level in the
rat  (Figure  4-6); this  difference  suggests that metabolic capacity for  CO
formation  is  greater in  rats than in humans.  Hogan  et al. (1976)  found  that
                                       Q
rats  exposed  to 440  ppm  DCM  (1529 mg/m  ) for 3 hours had maximal COHb levels
                                                        3
of about 7 percent,  and exposure to 2300 ppm (7,990 mg/m  ) produced no further
increase.  Pretreatment of the animals  with phenobarbital increased the  rate
of  rise of COHb levels  and  the time the maximum  level was maintained, but it
did  not increase the highest level.  These investigators suggest that endo-
genous  CO  inhibition of  the  P.™  metabolizing  system by CO binding to  the

                                   4-51

-------
 cytochrome may  occur  at very high levels  of  DCM exposure.   However, their
 observations are also  consistent with the Michaelis-Menten kinetics of hepatic
 CO formation from DCM,  with saturation of metabolic conversion from exposure
 to 400 ppm  (1,390 mg/m  ).   Kurppa and Vainio (1981) exposed rats to 500 and
 1000 ppm DCM (1737  and  3474 mg/m ),  6 hr/day for 5 days/week for  1  and  2
 weeks.   The test results  are  given in Table 4-15.  Since maximal blood COHb
 levels  of 8 to  9  percent occurred with 500 ppm  (1737 mg/m3) exposure and no
 further increase was obtained with 1000 ppm (3474 mg/m3) or with longer dura-
 tion of exposure,  these  results  also  indicate  that saturation  of  metabolism of
 DCM to  CO in rats occurs at a DCM exposure concentration of  500 ppm (1737
 mg/m )  or less.
      McKenna et al.  (1982)  exposed rats  to 50,   500, and  1500  ppm  DCM (174,
 1737, and 5211 mg/m ) for  6 hours;  during testing, steady-state conditions
 were reached between  production  of CO, maintenance of  a  given circulation
 blood COHb concentration,  and  pulmonary CO  excretion.  These results are shown
 in  Figure 4-12.   The  blood percent  COHb  at  steady-state was   not  linearly
 related to exposure  concentration  but reached a maximum  of about  12.5  percent.
 Kinetic parameters  for  the production of CO and formation  of COHb can be
 estimated on the clear assumption of  dose-dependent Michaelis-Menten kinetics
 (Section  4.2.2).  Estimates  calculated  from the data in  Figure  4-12  are V    z
                                                              o          niax
 13.5 percent COHb/6-hr exposure and Km  * 170 ppm  DCM (591  mg/m  ). These values
 indicate  a saturating DCM exposure  concentration  of  about  350 ppm  (1,216
 mg/m )  (2 x Km)  for  the  rat.
     The  determination  of Michaelis-Menten kinetics of  DCM  metabolism to  CO
 and  an  upper limit to  the attainable  blood COHb concentration  provides an  ex-
 planation of  the observations  of  Roth et  al. (1975) and  of Haun et  al.  (1971,
 1972).  Roth  et al.  found that concentrations  of  COHb  in the blood  of rabbits
 after very  short exposures  (20  minutes)  to DCM  inhalation concentrations
 ranging from 2,000 to 12,000 ppm (6948  to 41,688  mg/m3) were a  linear function
 of DCM  exposure concentration.  COHb  levels were  approximately  5.5  percent  at
 2,000 ppm (6,948 mg/m3) and 13 percent at 12,000  ppm (41,688 mg/m3).  However,
with 4 hours of exposure at about 7,000 ppm (24,318 mg/m3), steady-state blood
COHb concentrations  of  14 percent were attained.   Phenobarbital pretreatment
of the rabbits decreased the blood COHb level  achieved with a given DCM exposure,
although  in parallel experiments,  phenobarbital   stimulated rabbit microsomal
benzene  hydroxylase and benzphetamine N-demethylase activity.   Therefore,  Roth
                                   4-52

-------
 TABLE 4-15.  BLOOD COHb AND HEMOGLOBIN CONCENTRATIONS  IN  RATS  EXPOSED TO  DCM'
Exposure
(ppm)
None
500
1000
Carboxyhemogl obi n, %
Five days
0.4 ฑ
8.1 ฑ
9.2 ฑ
0.1
0.6
0.6
Ten days
0.4 ฑ
8.1 ฑ
8.5 ฑ
0.2
0.7
0.6
Hemoglobin, q/1
Five days
155.2 ฑ 5.5
144.0 ฑ 21.2
151.5 ฑ 6.0
Ten days
156.6 ฑ 6.2
155.4 ฑ 3.5
158.0 ฑ4.5
aT. , . .
Source:  Kurppa and Vainio, 1981.

et al. (1975) suggested that increased CO production from DCM at the microsome
in response  to  a  phenobarbital-induced  increase of the cytochrome  P.,.,,  system
                                                                   . ^3U
inhibits further  DCM  metabolism.   A more likely explanation of these results
is that the short exposures did not provide sufficient time for the establishment
of body equilibrium to inhaled DCM; therefore,  even at high inhalation concen-
trations,  the  DCM presented to  the  hepatic-metabolizing system was on  the
linear portion of the Michaelis-Menten curve.
     In a  comparison  of  dogs and monkeys continuously exposed  to  25 and 100
ppm DCM  (87  and 347 mg/m3) for  6 to 13 weeks,  Haun et al.  (1971)  found  that
steady-state levels  of blood COHb were maintained throughout the exposure
period, and  these  concentrations were  directly proportional to the exposure
concentration.  However, these concentrations  result from low-level exposures;
therefore,  they are expected to be on the linear portion of the Michaelis-Menten
function.  Haun et al. noted that dogs  had higher steady-state blood concentra-
tions of DCM than monkeys  at these inhalation concentrations, but  monkeys  had
the higher COHb blood levels,  suggesting that monkeys have a greater hepatic
capacity for CO formation.

4.5.3  Comparison of Kinetics of Humans  and Rats
     The kinetics of DCM metabolism to  CO and of COHb  formation are strikingly
similar for humans and rats.  Both species demonstrate Michaelis-Menten  dose-
dependent kinetics with similar estimates for Vm (15 and 13.5 percent COHb per
time unit,  respectively) and  Km  (200 and 170 ppm DCM,  respectively).   Hence,
the saturating DCM  inhalation concentration  for the metabolism of DCM to  CO
                                   4-53

-------
 and formation of COHb are comparable for the two species (400 versus 350 ppm;
 1390 versus 1216 mg/m3).
      The metabolism of DCM  involves  two pathways:   the microsomal oxidative
 pathway leads to CO, and the cytosolic pathway using glutathione converts DCM
 to formaldehyde, formic acid,  and  C02>   Comparison of  the  kinetic parameters
 estimated above for the CO  pathway in  the  rat  with those obtained by McKenna
 et al.  (1982)  for the  total  overall  (both pathways) metabolism of DCM in  these
 rats (Section 4.2.2) reveals that  the  Km s 400 ppm (1390 mg/m3) for overall
 metabolism is about twice that for the CO pathway alone.   Furthermore,  this
 figure  is an average overall  value,  suggesting  that the cytosolic glutathione-
 dependent pathway has  a much higher  Km value than  that of the CO pathway.
 Andersen (1981) has called  the CO  metabolism a high-affinity,  low-capacity
 pathway and the glutathione-dependent pathway low-affinity  but high-capacity.
4.6  MEASURES OF EXPOSURE AND BODY BURDEN  IN HUMANS
     In  the  controlled laboratory setting, estimating  the  DCM absorbed into
the  body by  comparing inspired  and alveolar air concentrations,  or by measur-
ing  blood levels  of DCM and then extrapolating these parameters  to body dose,
is an  imprecise task.  However, the  goal  of this research is to  develop a
sufficient data base  and knowledge of the  kinetics of absorption,  disposition,
elimination, and  metabolism of  DCM to enable assessment of  the body burden of
DCM  from acute  or chronic  exposure in the  industrial  or ambient  setting where
air concentrations and exposure period vary widely.  Air monitoring, an  impor-
tant control measure, cannot be used as a reliable index of body  burden.  At
present,  monitoring  levels  in blood or alveolar air and measuring blood COHb
levels  are  the available approaches  to  estimating human body burdens  from
recent exposures.  However,  in addition to' the lack of complete pharmacokinetic
knowledge necessary to interpret these determinations as accurate  and reliable
measures of body burden, the results are also subject to unknown inter-individual
variation from factors such as anthropometric differences, metabolism and work
load, age and sex, and modifications from drugs and environmental xenobiotics.
     Stewart and associates (1976a)  advocate the use of  breath  analysis to
monitor DCM exposure because it is a noninvasive method and avoids the problems
associated with the multiple blood sampling required for determinations of
blood DCM and COHb levels.   In addition,  breath samples  can be readily collected
                                   4-54

-------
with little  inconvenience  in the immediate post-exposure  period  and  also  at
later periods.  Analysis  of DCM in those samples by infrared spectroscopy or
gas liquid chromatography provides both an identification and a measurement of
the magnitude of exposure.
     Stewart and  his  associates have constructed a "family" of post-exposure
breath decay curves  spanning 20 hours from controlled  and known inhalation
exposures of volunteers  in the laboratory.  The  concentration  of DCM in the
alveolar  air during and after  exposure  is  directly related to the average
inhalation  exposure  concentration.   When the duration  of  exposure  and work
intensity are  known,  the total body burden can  be  estimated  by reference  to
"standard"  breath  concentration curves.   Because of large variations between
individuals exposed under identical conditions, only very approximate DCM body
doses can be estimated.   The 2-hour period following exposure  appears  to  be
the most  reliable breath sampling time  for estimating  the TWA DCM exposure
concentration.  Stewart  et  al.  (1976a)  provide  little information on the
statistical  reliability  or  the inter-individual variation expected  in the
predictive  values  for exposure burdens,  although they  point  out the desir-
ability of  constructing  "individualized" breath  decay curves  for  every  person
expected  to be exposed to  DCM.  Peterson (1978), using the same  experimental
exposure  data  as  Stewart et al. (1976a),  has  developed empirical equations
relating  post-exposure  breath  concentrations  to exposure time, duration, and
blood COHb  levels.
4.7  SUMMARY AND CONCLUSIONS
     The metabolism and pharmacokinetics of DCM have been extensively studied.
At  ambient  temperature,  DCM is a  volatile  liquid with high lipid solubility
and  modest  solubility in water; therefore, the  principal  routes of entry to
the  body  are by pulmonary and oral absorption.   Comparatively fewer data  are
available on the metabolism and pharmacokinetics of absorption  and excretion
of  DCM in humans,  but these  kinetics  have  been  extensively studied  in  the
rodent.   Absorption from the GI tract is  rapid  and complete, occurring by
first-order  passive processes in the  rat; the kinetics of  peroral absorption
and  post-absorption disposition and elimination  are influenced  by the dosing
vehicle.  Pulmonary absorption also occurs  by first-order diffusion processes
in  both humans  and rats.  There  are  three  distinct components whose rate
                                   4-55

-------
 constants  correspond to tissue loading of at least three major body compart-
 ments.   At equilibrium with inspired air  concentration,  a blood/air partition
 coefficient of about 10.5 at 37ฐC has been observed.
     Distribution  of DCM in the  tissues  is consistent with  its  lipophilic
 nature  and modest water solubility.  The chemical readily crosses  the blood-
 brain and  placental  barriers.   Concentrations occurring  in  all major  tissue
 organs  are dose  related  to inspired air concentration  or  to oral dosage.
 During  inhalation  exposure, the quantity  of DCM  absorbed is  also  dependent on
 body weight and fat  content of  the body; the adipose tissue/ blood  coefficient
 at  37ฐC is about 7  and is  about 0.8 to 1.0 for brain and liver tissues.  The
 rate of tissue  loading with a  given  inspired air concentration is increased
with physical  activity and with  exposure duration, with steady-state  body
equilibrium requiring more  than 6 hours.
     The kinetics  of elimination  of DCM from the body are complex and  are
dominated  by two  major and parallel processes:    (1) pulmonary elimination of
unchanged  DCM,  and  (2)  hepatic metabolism  of  DCM.   Pulmonary elimination
follows  first-order  kinetics,  is  independent of  body  dose,  and  exhibits at
least three distinct body compartments with half-times for humans of about 8
to  23 minutes, 40 to 80 minutes, and 360 to 390  minutes, and  half-times  for
rats of 1  to  2 minutes, 12 to 16 minutes, and 47 minutes, respectively.  The
longest half-time is associated with the lipid and adipose tissue body compart-
ment.   The differences between humans and rats for the half-times  of pulmonary
elimination from body compartments are presumed to be caused by differences in
pulmonary  and  cardiovascular functions.   Hepatic metabolism  is  body-dose
dependent  and  saturable,  and  follows  nonlinear  Michaelis-Menten  kinetics.
Body dose, as given  by  oral  dosing, also is  subject to the kinetics of  first-
pass hepatic metabolism and first-pass  pulmonary  excretion.  The  estimated K
for overall metabolism of DCM in the rat,-  as obtained from inhalation exposures,
is  about 400  ppm.   This  inhalation concentration for 6  hours produces an
estimated body dose of approximately 50 mg/kg.   For the body dose  of 50 mg/kg,
70 percent of the DCM is metabolized, and for a 5.5 mg/kg body dose resulting
from 50  ppm for a 6-hour exposure, 95 percent is  metabolized.  The  inhalation
concentration  saturating overall metabolism of DCM is about 800 ppm (2 x  K ).
     The hepatic metabolism of  DCM  occurs by two enzymatic pathways:   (1) an
inducible cytochrome P^g-mediated microsomal oxidative dehalogenation to CO,
and 2)  a noninducible cytosolic glutathione transferase dehalogenation system
                                   4-56

-------
yielding formaldehyde  and  formic acid, which are further metabolized to CO,,.
The  activities  of these  two pathways are approximately  equal  at low body
burdens of DCM, and both pathways are saturable at high body burdens. Covalent
binding to cellular macromolecu!es  (proteins :and  lipids)  by  putative reactive
intermediates of metabolism, formyl chloride from microsomal oxidative metabolisr
and.S-chloromethyl glutathione and formaldehyde from cytosolic metabolism, has
been  shown  to occur  i_n vitro with rat  hepatic  microsome preparations and
intact isolated rat hepatocytes  and jj\ vivo after   C-DCM injections to  rats.
No evidence was found  in these studies for the occurrence of covalent binding
to DNA.
     The occurrence  of increased  blood  COHb levels  in  humans  and  animals
exposed to DCM is a consequence  of hepatic microsomal  oxidative metabolism of
DCM to CO.   The COHb produced is additive to COHb formed from exogenous CO.   A
functional  relationship exists  between  the  DCM  inhalation  concentration,
duration of exposure,  and  the time course and peak  blood COHb level.   The
blood COHb level achieved  is  determined  by the nonlinear  kinetics  (Michaelis-
Menten kinetics) of hepatic metabolism of DCM to CO  and  the parallel  linear
(first-order) kinetics  of  pulmonary  elimination  of CO from circulating COHb.
Because hepatic metabolism  of DCM to  CO  is saturable,  the zero-order kinetics
of CO  production  constrains  blood  COHb accumulation  to  an  upper,  limited
concentration of 12 to  15 percent COHb,  as observed  experimentally.   However,
human studies have demonstrated  that exposures to DCM at levels  up  to about
150 ppm (521 mg/m ) are unlikely to exceed the biological  TLVฉ for blood COHb
(5 percent) recommended by  the National  Institute for  Occupational Safety and
Health (1976).  Kinetic parameters  for the production of CO and formation of
COHb, calculated from experimental DCM exposure data  of blood COHb concentra-
tions in humans and rats,  have similar values.   Estimations of V     for humans
                                                                max
and rats are  15 and  13.5 percent COHb per 7-hour exposure, respectively, and
estimates  of Km are 200 and 170 ppm, respectively.   These findings  indicate
that the CO pathway is saturated with an inspired air concentration of approx-
imately 400 ppm DCM (2 x Kffl) in both humans and rats.
                                   4-57

-------
4.8  REFERENCES
Ahmed, A. E.,  and M.  W. Anders.  1976.  Metabolism of dihalomethanes to  for-
     maldehyde and inorganic chloride.  Drug Metab. Dispos. 4:357-361.

Ahmed, A. E.,  and M.  W. Anders.  1978.  Metabolism of dihalomethanes to  for-
     maldehyde and  inorganic halide.  II.   Studies on the mechanism  of the
     reaction. Biochem. Pharmacol. 27:2021-2025.

Ahmed, A. E., V.  L.  Kubic, J. L. Stevens, and M. W. Anders.  1980.  Halogenated
     methanes:  metabolism and toxicity.  Fed.  Proc. 39(13):3150-3155.

Amoore, J. E., and E.  Hautala.  1983.  Odor as  an  aid to chemical  safety:  odor
     thresholds  compared  with threshold limit  values  and volatiles for  214
     industrial chemicals in air and water dilution. J. Appl. Toxicol.  3:272-290.
Anders, M. W.  and  J.  M.  Sunram.
     thane and carbon monoxide.
          1982.  Transplacental passage  of  dichlorome-
          Toxicol.  Lett. 12:231-234.
Anders, M. W.,  V.  L.  Kubic and A.  E.  Ahmed.
     methanes  and macromolecular  binding.
     1:117-121.
                      1977.   Metabolism  of  halogenated
                      J.  Environ.  Pathol.  Toxicol.
Astrand, I., P. Ovrum, and A. Carlsson.  1975.   Exposure  to  methylene  chloride.
     I.  Its concentration  in alveolar air and blood during rest and exercise
     and its metabolism.  Scand. J. Work Environ.  Hlth. 1:78-94.

Bourne, W.,  and R.  L.  Stekle.  1923.   Methylene chloride in anesthesia.  Can.
     Med. Assoc. J. '13:432-433.

Carlsson, A., and M. Hultengren.  1975.  Exposure  to  methylene  chloride.   III.
     Metabolism  of 14C-labeled  methylene  chloride in rat.   Scand.  J. Work
     Environ. Hlth. 1:104-108.

Ciuchta, H. P., G. M.  Savell, and R.  C. Spikes.  1979.  The  effect of  alcohols
     and toluene  upon  methylene chloride induced carboxyhemoglobin in rat and
     monkey.  Toxicol. Appl.  Pharmacol. 49:1-20.

Coburn, R. E.  The carbon monoxide  body stores.  1970.   In:   Biological  effects
     of carbon  monoxide.   Proceedings of  a Conference,  New York  Academy  of
     Science, New York, January 12-14, 1970.   Ann.  N.  Y.  Acad.  Sci.  174:11-22.
Coburn,  R.  E.   1973.
     24:241-250.
Endogenous carbon monoxide metabolism.  Ann.  Rev. Med.
Collison,  H.  A.,  F.  L.  Rodkey, and J. D. O'Neal.  1977.  Effects of dichloro-
     methane  on hemoglobin  function.   Biochem.  Pharmacol.  26:557-558.

Cox, P. J., L. J.  King,  and D.  V.  Parke.   1976.   The binding of trichlorofluoro-
     methane  and other  haloalkanes to  cytochrome P-4&0 under  aerobic and
     anaerobic conditions.   Xenobiotica 6:363-375.
                                    4-58

-------
Cunningham, M.  L. ,  A.  J.  Gandolfi/  K.  Brendel, and  I.  G.  Sipes.  1981.
     Covalent binding of  halogenated  volatile solvents to subcellular macro-
     molecules in hepatocytes.  Life Sci. 29:1207-1212.

Dill, D. C. ,  P.  G.  Watanabe, and  J.  N.  Morris.  1978.  Effect of methylene
     chloride on the oxyhemoglobin dissociation curve  of  rat and  human  blood.
     Toxicol.  Appl.  Pharmacol. 46:125-129.

DiVincenzo, G.  D. ,  and M.  L.  Hamilton.   1975.  Fate  and disposition of
     14C-methylene chloride in the rat.  Toxicol. Appl. Pharmacol. 32:385-393.

DiVincenzo, G. D. and C. J. Kaplan.  1981a.  Uptake, metabolism, and elimination
     of  methylene  chloride  vapor  by humans.   Toxicol.  Appl.  Pharmacol.
     59:130-140.

DiVincenzo, G.  D. and C. J.  Kaplan.   1981b.  Effect of exercise or smoking on
     the  uptake,  metabolism, and  excretion of  methylene chloride  vapor.
     Toxicol.  Appl.  Pharmacol. 59:141-148.

DiVincenzo, G.  D. ,  F.  J.  Yanno, and  B.  D.  Astill.   1972.  Human and canine
     exposures to methylene chloride vapor.  Am. Ind. Hyg. Assoc.  J. 33:125-135.

Engstrom,  J. ,  and R.  Bjurstrom.   1977.   Exposure  to  methylene  chloride.
     Content  in  subcutaneous  adipose  tissue.  Scand.  J.  Work Environ.  Hlth.
     3:215-224.

Fenn, W. 0.   1970.   The burning of CO in tissues.   Ann.  N.  Y. Acad.  Sci.
     174:64-71.

Fodor, G.  G. , and  A.  Roscovanu.  1976.  Increased  blood CO content in humans
     and animals caused by  incorporated halogenated hydrocarbons.   Zbl.  Bakt.
     Hyg. 162:34-40.

Fodor, G.,  D. Prajsnar, and H-W.  Schlipkoter.   1973.   Endogenous  CO-Bildung
     durch  inkorporierte   Halogankohlenwasser-Staffe  der  Methanreihe:
     Staub-Reinhalt. Luft 33:258-259.

Forster, H. V.,  S.  Graff,  C. L. Hake, R. Soto, R.  D. Stewart.  1974.  Pulmo-
     nary- hemato logic studies on humans during exposure to methylene chloride.
     Report No.   NIOSH-MCOW-ENVM-MC-74-4.   The  Medical College  of  Wisconsin.
     Department of Environmental Medicine, Milwaukee, WI, 17 pp.

Friedlander,  B.   P.,  T.  Hearne,  and S. Hall.  1978.   Epidemiological investi-
     gation of employees chronically exposed to methylene chloride.  J. Occup.
     Med. 20:657-666.

Hake, C.  L.  1979.   Simulation studies  of blood carboxyhemoglobin levels
     associated  with  inhalation exposure  to methylene chloride.   Toxicol.
     Appl.  Pharmacol. 48:A56 (abstr).
                                   4-59

-------
Hake, C.  L. ,  R.  D.  Stewart, H. V. Forster, A. J.  Lebrun, J.  E.  Peterson,  and
     A.  Wu.  1974.  Results of the controlled exposure of human  females  to the
     vapor  of  methylene chloride.   Report  No.   NIOSH-MCOW-ENVM-MC-74-3.
     Milwaukee, Wis.  The Medical College of Wisconsin,  Department  of  Environ-
     mental Medicine, Milwaukee, WI, 22 pp.

Haun, C.  C.,  E.  S.  Harris, and K. I. Darmer.  1971.  Continuous  animal  expo-
     sure to  methylene' chloride.   AMRL-TR-71-120, #10,  In:   Proceedings 2nd
     Conference Environmental Toxicology, Wright-Patterson Air Force Base,  OH,
     pp. 125-135.

Haun, C. C., E. H. Vernot, K. I. Darmer, and S. S. Diamond.   1972.  Continuous
     animal exposure  to  low  levels  of  dichloromethane.   AMRL-TR-130,  #2.  In:
     Proceedings  of  the  3rd  Conference on  Environmental  Toxicology,  Wright-
     Patterson Air  Force Base,  OH,  Aerospace Medical  Research Laboratory, pp.
     199-208.

Heppel,   L.  A., and V. T.  Porterfield.   1948.  Enzymatic dehalogenation of
     certain brominated  and chlorinated compounds.  J. Biol.  Chem.  176:763-769.

Heppel,   L. A.,  P. A.  Neal, T.  L.  Perrin,  N. L. Orr, and V.  T.  Porterfield.
     1944.  Toxicology of dichloromethane  I.   Studies on effects  of  daily
     inhalation. J.  Ind.  Hyg. Toxicol.  26:8-16.

Hogan,  G. K., R. G. Smith, and H. H. Cornish.  1976.  Studies on the  microsomal
     conversion of CH2C12 to CO.  Toxicol. Appl. Pharmacol.  37(1):112.

Kahn, A.,  R.  B.  Rutledge, G.  L.  Davis,  J.  A. Altes, G.  E.  Gantner,   C.  A.
     Thornton,  and  N.  D. Wallace.   1974.   Carboxyhemoglobin sources  in the
     metropolitan St.  Louis population.   Arch.  Environ. Hlth.  29:127-135.

Kasselbart, V. ,  and  J.  Angerer.  1974.  Influence of dichloromethane on the
     disappearance rate  of ethanol in the blood of rats.  Int. Arch. Arbeitsmed.
     33:231-236.
Kubic, V. L, and M.
     monoxide.  II.
W. Anders.  1975.
 In  vitro studies.
Metabolism of dihalomethanes to carbon
 Drug Metab. Dispos. 3:104-112, 1975.
Kubic, V.  L.  and  M.  W.  Anders.   1978.   Metabolism in dihalomethanes to carbon
     monoxide.  III.   Studies on  the mechanism  of the reaction.   Biochem.
     Pharmacol. 27:2349-2355.

Kubic, V.  L. ,  M.  W.  Anders,  R.  R.  Engel, C.  H.  Barrow,  and W.  S. Caughey.
    .1974.   Metabolism  of dihalomethanes to  carbon monoxide.  I.   Iji  vitro
     studies.  Drug Metab. Dispos. 2:553-557.

Kurppa,  K.  and H. Vainio.   1981.  Effects of intermittent dichloromethane
     inhalation on blood carboxyhemoglobin concentration  and drug metabolizing
     enzymes in rat.   Res. Comm.  Chem.  Path.  Pharmacol. 32:535-544.

Kurppa,  K. ,  H. Kivisto, and  H.  Vainio.  1981.   Dichloromethane  and carbon
     monoxide  inhalation:   carboxyhemoglobin addition, and  drug  metabolizing
     enzymes in rat.   Int. Arch.  Occup.  Environ.  Hlth.  49:83-87.
                                   4-60

-------
Kuzelova^ M. , and R. Vlasak.   1966.   Der  Elnfluss  von  Methylenchlorld auf die
     Gesundheit der Arbeiter bei der Herstellung von Filmfolien und Ermlttlung
     der Ameisensaure als  Stoffwechselproduct des Methylenechlorids.  Praco.
     Lek. 18:167-170.

Lambertsen, C. J.   1974.   Effects  of  excessive  pressures  of oxygen,  nitrogen,
     carbon dioxide, and carbon monoxide.   Implications in  aerospace,  undersea,
     and industrial  environments.   In:   Medical Physiology, ed. Mountcastle,
     V.  B.  Vol.  2,  13th ed., C. V.  Mosby Co., Saint Louis,  MO.

Latham,  S.  ,  and  M.  Potvin.  1976.   Microdetermination  of dichloromethane in
     blood with  a  syringe!ess  gas  chromatographic injection system.   Chemos-
     phere 6:403-411.

Lehmann, K.  B. ,  and L. Schmidt-Kehl.   1936.   (The thirteen most  important
     chlorinated aliphatic hydrocarbons  from  the standpoint  of  industrial
     hygiene.)  Arch. Hyg.  116:131-268, (Ger).

Leonardos,  G. , D.  Kendall, and H.  Bernard.  1969.  Odor  threshold determina-
     tions  of 53 odorant  chemicals.   J.  Air  Pollut. Contr. Assoc.  19:92-95.

Lindqvist,  T.  1978.   Partition coefficients of some commonly used solvents.
     Ind.  Environ.   Xenobiot.  Proceedings International  Conference,   1977.
     Prague, J.   R.  Fouts,  and I. Gut., eds., pp.  165-167.   Excerpta  Medica,
     Amsterdam-Oxford.

Llewellyn,  0.  P.   1966.   Halogenated hydrocarbons used  as  solvents.   Ann.
     Occup. Hyg.  9:199-208.

Luomanmaki, K.,  and  R.  E.  Coburn.  1969.   Effects  of metabolism and distribu-
     tion of  carbon  monoxide on blood and body stores.   Am. J. Physio!.  271:
     354-363.

MacEwen, J. D.,  E.  H. Vernot, and C.  C. Haun.  1972.  Continuous animal exposure
     to  dichloromethane.   Report AMRL-TR-72-28.  Aerospace Medical  Research
     Lab.  Wright-Patterson Air Force Base, OH, pp. 33.

May, J.   1966.  Odor thresholds of solvents for assessment  of  solvent  odors in
     the air.  Staub-Reinhalt.  Luft 26:34-38.

McKenna, M.  J. ,  and J.  A.  Zempel.  1981,   The  dose-dependent metabolism of
     14C-methylene  chloride following  oral  administration  to rats.   Food
     Cosmetics Toxicol.  19:73-78.

McKenna, M. J.,  J.  A. Zempel, and W.  H. Braun.  1982.  The  pharmacokinetics of
     inhaled methylene  chloride in rats.   Toxicol. Appl. Pharmacol. 65:1-10.

McKenna, M. J.,  J.  H. Saunders, W.  R.  Boeckler, R. J. Karbowski, K. D. Nitschke,
     and M.  B.  Chenoweth.   1980.  The pharmacokinetics  of inhaled methylene
     chloride in human  volunteers.   Paper #176, 19th Annual Meeting,  Society
     of Toxicology, Washington, D.C.,  March 9-13.

Miller, V.   L. ,  R.  R. Engel, and M. W.  Anders.   1973.   In vivo metabolism of
     dihalomethanes  to carbon monoxide.   Pharmacologist 15:190  (abstr).
                                   4-61

-------
 Morgan,  A.,  A. Black, and D. R. Belcher.  1972.  Studies on the absorption of
      halogenated hydrocarbons and their excretion in breath using 38C1 tracer
      techniques.   Ann.  Occup.  Hyg.  15:273-282.

 National  Institute for Occupational Safety and Health.  1972.  Criteria for a
      Recommended Standard for Occupational  Exposure  to Carbon Monoxide.   HSM
      73-11000.

 National  Institute for Occupational Safety and Health.  1976.  Criteria for a
      Recommended Standard in Occupational Exposure to Methylene Chloride, HEW
      No.  76-138.

 Neely,  W. B.  1964.  Metabolic  rate of formaldehyde-  14C  intraperitoneally
      administered  to  the  rat.   Biochem.  Pharmacol.  13:1137-1142.

 Nunes,  A. C.  and  B.  P.  Schoenborn.   1973.   Dichloromethane  and  myoglobin
      function. Mol. Pharmacol.  9:835-839.

 Peterson,  J.  E.   1970a.   Post-exposure relationship  of  carbon monoxide in
      blood and expired  air.   Arch  Environ. Hlth.  21:172-173.

 Peterson, J.  1978.   Modeling the  uptake metabolism and excretion  of dichloro-
      methane by  man.  Am.  Ind. Hyg.  Assoc. 39:41-47.

 Peterson, J.  E., and  R.  D. Stewart.   1970b.   Absorption and elimination of CO
      by inactive young men.   Arch.  Environ.  Hlth.  21:165-171.

 Pritchard, A.,  and M. J.  Angelo.   1982.  Comparative  pharmacokinetic studies
      of methylene  chloride in oil  versus drinking water.   Report commissioned
      by National Coffee Association  of U.S.A. Inc.  Available  August.

 Pritchard, A.,  B.  Doonan  and  R. Rourke.  1982.   Methylene  chloride:   effects
      on microsomal  enzymes in mice.   Study  commissioned by National  Coffee
      Association of U.S.A. Inc.

 Ratney, R. S. ,  D.  H.  Wegman, and H. B.  Elkins.   1974.   In  vivo conversion of
      methylene chloride  to carbon  monoxide.   Arch.  Environ. Hlth.  28:223-226.

 Reynolds, E. S. ,  and  A. G. Yee.   1967.   Liver parenchyma!  cell injury.  V.
      Relationships  between patterns of  chloromethane-14C  incorporation into
      constituents  of  liver in vivo  and  cellular injury.   Lab.  Invest.  16:
      591-603.

 Riley, E. C. ,  D.  W.  Fassett,  and  W.  L.  Sutton.  1966.   Methylene chloride
     vapor in  expired air of human  subjects.   Am.  Ind. Hyg.  Assoc.  J.  27:
     341-348.

Roberts,  C.  J.  C. ,  and F. P.  F.  Marshall.   1976.  Recovery  after "lethal"
     quantity of paint remover.  Brit. Med. J. 6000:20-21,  January 3.

Rodkey, F.  L., and H.  R. Collison.   1977.   Biological oxidation of 14C-
     methylene chloride  to carbon monoxide  and carbon dioxide by the rat.
     Toxicol. Appl. Pharmacol. 40:33-38.
                                   4-62

-------
 Roth,  R.  P., R.  T.  Drew, R. J. Lo, and J.  R.  Fouts.   1975.   DichToromethane
      inhalation,  carboxyhemoglobin concentration, and drug metabolizing enzymes
      in,rabbits.   Toxicol.  Appl.  Pharmacol.  33:427-437.

 Savolainen,  H.,  P. Pfaffli, M. Tengen, and  H.  Vainio.   1977.   Biochemical  and
      behavioral  effects  of inhalation exposure  to tetrachloroethylene and
      dichloromethane.   K.  Neuropath.  Exptl.  Neurology, 36:941-949.

 Schutz,  E.   1958.  Effect  of  polyethylene glycol 400 on percutaneous absorp-
      tion  of active  ingredients.   Arch.  Exp.  Path.  Pharmakol. 232:237-238.

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

 Settle, W.   1971.   Ph.D.  thesis, University  of  California.

 Settle, W.   1975.  Role of  changes  in carbon monoxide-hemoglobin binding  in
     methylene  chloride toxicity.   Fed.  Proc.  Fed. Am. Soc.  Exptl.  Biol.
     34:229  (Abstr.).

 Stevens, J.  L.,  and M.  W.  Anders.   1978.   Studies on the mechanisms of metab-
     olism  of
     297-298.

Stevens, J.  L.,
     monoxide.
     Pharmacol.
haloforms to carbon  monoxide.   Toxicol. Appl.  Pharmacol.  45:
  and M. W.  Anders.   1979.   Metabolism of haloforms to carbon
 III.   Studies  on  the mechanism  of the  reaction.   Biochem.
 28: 3189-3194.
Stevens,  J.  L. ,  J.  H.  Ratnayake, and M.  W.  Anders.
     dihalomethanes  to  carbon monoxide.  IV.   Studies
     cytes. Appl. Pharmacol.  55:484-489.
                                        1980.  Metabolism  of
                                        in  isolated rat hepato-
Stewart,  R.  D. ,  and H. C.  Dodd.   1964.   Absorption of carbon tetrachloride,
     trichloroethylene,  tetrachloroethylene, methylene  chloride,  and 1,1,1-
     trichloroethane  through the  human  skin.   Am.  Ind. Hyg.  Assoc.  J.  25-
     439-446.

Stewart,  R.  D. ,  and C. L.  Hake.   1976.   Paint-remover  hazard.  JAMA J.  Am.
     Med. Assoc.  235:398-401.            •

Stewart,  R.  D. ,  C.  L. Hake, and  A.  Wu.   1976a.  Use  of breath analysis to
     monitor  ethylene chloride exposure.  Scand. J.  Work Environ. Hlth.  2:
     57-70.

Stewart,  R.  D. ,  R.  S. Stewart, W.  Stamm,  and R. P.  Seelen.   1976b.   Rapid
     estimation of carboxyhemoglobin level in fire  fighters.   JAMA J.  Am.  Med.
     Assoc. 235:390-392.

Stewart,  R.  D.,  H.  V. Forster, C. L. Hake, A. J. Lebrun, and  J. E. Peterson.
     1973.   Human  responses to  controlled exposure  of methylene chloride
     vapor.  Report No.  NIOSH-MCOW-ENVM-MC-73-7.  Milwaukee,  WI,  The Medical
     College of Wisconsin,  Department of Environmental  Medicine,  December., 82
     pp.
                                   4-63

-------
       ™4*u*  SVu -T  V'  Forster>  c- L- HaKe, A. J. Lebrun and J. E. Peterson.
       1974b.  Methylene chloride:    Development  of  a biologic standard for the
       industrial  worker by breath analysis.   Report No.  NIOSH-MCQW-ENVM-MC-74-9
       Milwaukee,  WI,  The Medical  College  of Wisconsin,  Department of Environ-  '
       mental  Medicine,  December.

 Stewart,  R.  D.,  T. N.  Fisher, M. J. Hosko, J.  E.  Peterson, E.  D. Baretta, and
       H. C. Dodd.   1972a.   Carboxyhemoglobin elevation after exposure to dichloro-
       methane.  Science 176:295-296.

 Stewart   RD.,  T. N.  Fisher, M.  J.  Hosko,  J.  E.  Peterson,  E.  D.  Baretta and
       H. C.  Dodd.   1972b.   Experimental  human exposures  to methylene chloride
       Arch. Environ. Hlth.  25:342-348.

 Stewart   R.  D    E D.  Baretta,  L.  R.  Platte, E.  B.  Stewart, J.  H.  Kalbfleish,
       B. Van  Yserloo,  and  A.  A.  Rimm.  1974a.   Carboxyhemoglobin  levels in
      American blood donors.  JAMA  J.  Am. Med. Assoc.  229:1187-1195.

 Tenhunen,  R   H.  S.  Marver, and R. Schmid.  1969.  Microsomal  hemeoxygenase-
      characterization of the enzyme.  J. Biol.  Chem.  244:6388-6394.

 Toftgard,  R., 0.  G.  N. Nilsen,  and J.  S.  Gustafsson.   1982.   Dose-dependent
      induction of rat liver microsomal cytochrome P-450 and microsomal  enzymatic
      activities  after inhalation of toluene and dichloromethane.  Acta  Pharmacol
      Toxicol. 51:108-114.

 Tzagoloff, A., and D.  C.  Wharton.  1965.   Studies  on the electron transfer
      system.   LXII.  The reaction  of  cytochrome oxidase with carbon  monoxide
      J.  Biol. Chem. 240:2628-2632.

 Wagner,  J. G. 1975.   Fundamentals of Clinical  Pharmacokinetics.  Druq  Intelli-
      gence Publications, Inc., Hamilton, IL.

 Winneke, G.   1981.  The neurotoxicity of dichloromethane.   Neurobehav. Toxicol
      Teratol.  3:  391-395.

 Winneke, G.  and G.  G.  Fodor.   1976.  Dichloromethane produces narcotic effects
      Int.  J.  Occup. Hlth.  Safety  45(2):34-37.

 Withey,  J.^R  and  B. T.  Collins.  1980.  Chlorinated aliphatic  hydrocarbons
      used  in  the  foods  industry:   the  comparative  pharmacokinetics of methylene
      chloride, 1, 2-dichloroethane, chloroform,  and trichloroethylene  after
      I.V.  administration in the rat.  J. Environ.  Pathol.  Toxicol. 3:313-332.

 Withey,  J.  R., B.  T.  Collins,  and P. G.  Collins.  1983.   Effect of vehicle on
      the pharmacokinetics and  uptake of  four halogenated hydrocarbons  from the
      gastrointestinal  tract of the rat.   J.  Appl.  Toxicol. 3(5)-249-253
Yesair, D.  W.,  P.  Jaques, P.  Shepis,  and R. H.
     pharmacokinetics  of  14C-methylene chloride
     Exp.  Biol. 36:998 (abstr).
 Liss.   1977.
in mice.   Fed.
Dose-related
Proc.  Am.  Soc.
Zorn, H.  1975.  In:  Bericht uber die 14 Jahrestagung der  Deutschen  Gesellschaft
     fur Arbeitsmedizin  e.  V.,  G. Lehnert,  D.  Szadkowski  und H.  J.  Weber
     eds.,  p. 343, Gentner Verlage, Stuttgart.
                                   4-64

-------
                       5.   HEALTH EFFECTS OF DICHLOROMETHANE
                 >
 5.1  HUMAN HEALTH EFFECTS
 5.1.1  Acute Exposures
 5.1.1.1   Nervous System  and Behavior—Virtually  all  experimental  exposure
 studies of OCM  using humans have studied acute exposures and neural or behav-
 ioral dependent variables.   Using between three and eight subjects, Stewart et
 al.  (1972b) reported that  subjects  had  symptoms  of "lightheadedness"  and had
 difficulty with speech articulation  at  DCM levels  higher than  868  ppm (3,012
 mg/m3)  for more than  1 hour.   They  also reported anecdotal  data about increased
 amplitude  in certain visual  evoked potentials  at  about the same exposure  levels;
 however,  no statistical analyses were possible because of the small number of
 subjects.   In  a later  study, they reported  that 50,  100,  or  250 ppm (173,  347,
 or  867 mg/m3) DCM had  no effects on  the Romberg  equilibrium test,  vigilance,
 coordination,  or arithmetic  skill  (Stewart  et  al.,  1973).
      Winneke  (1974) and Winneke  and  Rodor (1976)  exposed  female human  subjects
 to  0, 300, 500, and 800  ppm (0,  1042, 1737, and  2,779 mg/m3) DCM for  up  to  3
 hours.   They measured  performance on a  series  of hand-eye coordination tasks
 including  tapping pursuit rotor and  hand steadiness.   They  also measured  the
 critical flicker fusion (CFF)  frequency  and performance on an auditory signal
 detection  task.  Concentration-related decrements in signal  detection, beginning
 at 300 ppm (1042 mg/m3),  were observed.   CFF frequency also  decreased  in a con-
 centration related  fashion.   Many motor  decrements  were  observed at 800  ppm
 (2,779 mg/m3),  the  only concentration at which these  tests were conducted.
 Fodor and  Winneke (1971)  reported a similar study in which 300  and 800 ppm (1,042
 and 2,779  mg/m3) both  decreased CFF  frequency  and  impaired  signal  detection
 behavior;  however, the  two  DCM levels did  not produce effects  significantly
 different  from each other.
     Gamberale et al.  (1975) investigated the effect of DCM on psychomotor and
 cognitive  performance  in  14 men, ages 20 to 30 years,  divided at random into
 two groups.  The first  group was  initially  exposed  to  progressively increased
 concentrations of DCM—250,  500, 748 and 997 ppm (869,  1,737, 2,599, and 3,464
mg/m3)—through  a face  mask.   Seven  days later they were observed under con-
trol conditions.  The second group was studied under identical conditions, but
 in reverse order.  The  subjects were exposed to each concentration for 30 minutes
with no break in exposure; total exposure time for each individual  was 2 hours.
                                     5-1

-------
At each concentration, a peak plateau of alveolar DCM occurred in about 10 min-
utes. Heart  rate showed no change.  During the two hours of exposure to DCM,
no statistically significant impairment in any measured performance was observed,
although reaction time appeared to be more irregular during exposure than during
control conditions.   Numerical  ability  and  short-term memory were  unaffected.
The experiment did not consider any latency effects.
     Putz et al. (1976) exposed six men and six women (ages 18 to 40 years) to
201 ppm (698 mg/m3)  of 99.5 percent pure DCM or  to 7 ppm  (8 mg/m3)  CO  for 4
hours in an  8-m3 chamber with unspecified dynamics.  Alveolar concentrations
were measured  hourly,  and  peripheral blood  from a finger was taken before and
after exposure.  Peak alveolar CO after 4 hours of DCM exposure was about 17 ppm
(19.5 mg/m3),  and  COHb  was 5.1 percent.  Alveolar CO after CO exposure for 4
hours was 4  ppm (4.6 mg/m3), and  COHb was 4.85 percent.  Thus, there was COHb
and alveolar CO equivalence between the two compounds.   Eye and hand coordina-
tion at the  end of the exposure  period was depressed in subjects  exposed to
either compound, compared with controls, with DCM depression being greater than
CO depression.   The authors concluded that CO may have been primarily responsi-
ble for the decreases in performance after exposure to both compounds.
     These studies can only be considered a preliminary survey of possible CNS
and behavioral  consequences  of DCM exposure.   Only one study (Winneke, 1974)
reported concentration effects data on CFF and signal  detection,  both possibly
involving the  alertness  of the subjects.   CFF is a primitive measure of CNS
activation.   The lowest concentration of DCM reported to have affected behavior
was 200 ppm  (695 mg/m3) (Putz et  al.,  1976).   They also reported vigilance
decrements in addition to compensatory tracking decrements.  In spite of these
few preliminary  findings,  the  results are consistent with  the hypothesis that
DCM effects  are depressant and also agree well with the available laboratory
animal data, reported elsewhere in this chapter.
     Data are  required  on  the  effects  of DCM upon  other behaviors.  No data
were found regarding cognitive  skills,  and  no concentration effects  data were
available for motor skills or hand-eye coordination.  No quantitative tests of
electrophysiological effects of DCM  in humans were found.  Finally, no data
regarding the effects of DCM in combination with other pollutants or medicinals
were found.
     Putz et al.  (1976) concluded  that  DCM  produced its effects via  its prin-
cipal metabolite, COHb.  Stewart et al.  (1972a) had suggested this possibility.
                                     5-2

-------
Winneke (1974) presented evidence suggesting that DCM is more deleterious than
equivalent levels of CO.  He compared subjects exposed to 50 or 100 ppm CO for
5 hours to subjects exposed to 300, 500, and 800 ppm (1042, 1737, and 2779 mg/m3)
DCM for 4  hours.  DCM-exposed  subjects  showed more  effects  on the  same tasks.
Comparison with  data  presented by PUtz et al.  (1976) reveals that equivalent
COHb levels were produced by 70 ppm CO and 200 ppm (695 mg/m3) DCM.  Examination
of the behavioral results of the  study  by  Putz et al. reveals that DCM always
produced greater decrements than  did CO.   Logically DCM should be more toxic
than CO,  since it not only produces COHb,  but,  because of its lipophilic quali-
ties, it probably has some primary neural  effect.  Additional data are required
to explore this issue.
5.1.1.2  Other Experimental DCM Effects—Stewart et  al.  (1972a,b)  reported no
effects of exposure to 500 or 1,000 ppm (1,737 or 3,474 mg/m3) DCM for 2 hours
on various hematological,  hepatic, or renal measures.  Continuous electrocardio-
graph!' c monitoring during  one  or two exposures to 1000 ppm (3,474 mg/m3) was
not reported  to  cause arrhythmias.  Gamberale et al. (1975)  reported that DCM
exposure up to 1,000 ppm (3,474 mg/m3)  produced  no  alterations  in  heart  rate.
Based on laboratory  animal  exposures, it  is not  reasonable to  expect other
internal  organ system involvement in healthy humans with low-level acute expo-
sures.
     Stewart and Dodd (1964) studied groups of three to five men and women (25
to 62 years old) who immersed their  thumbs in various chlorinated aliphatic
solvents.   Within the first 2 minutes of immersion in DCM, all subjects reported
an intense burning sensation on the dorsal  surface of their thumbs.  Within 10
minutes,  a feeling of coldness or numbness, alternating with the burning sensa-
tion, was  reported;  the slightest movement of the  thumb triggered waves of
searing and excruciating pain.   Following removal from the solvent, their thumbs
were described as "numb and cold" or "asleep."   A very mild erythema and a hint
of white scaling were the only overt signs of irritation.  Within 1 hour, both
the erythema and paresthesia subsided.
     Studies of  human  blood ijn vitro from individuals homozygous  for sickle
cell anemia (Matthews et al.,  1977, 1978)  have shown that DCM caused hemolysis,
preferentially of sickle and other abnormal cell  types.   These reports suggest
that acute exposure  to  relatively high concentrations of  DCM may result in
anemia, particularly  in individuals who are predisposed  to  hemolytic anemias,
e.g., persons  with sickle cell anemia  or  glucose-6-phosphate dehydrogenase
                                     5-3

-------
 deficiency.  However, more  compelling evidence is required to ascertain that
 such an effect exists.
 5.1.1.3  Accidental Exposure—A number  of case reports have implicated expo-
 sure to exceedingly high levels of DCM as one factor in human fatalities (Stewart
 and Hake,  1976; Kuzelova et a!., 1975;  Moskowitz and Shapiro, 1952; Bonventre
 et al., 1977; Winneke et a!.,  1981).   However, systematic conclusions  from acci-
 dental  exposure data  are  difficult.   Exact exposure levels and  durations are
 unknown.   Simultaneous exposure to other substances is  often the case.   Serious
 disturbances of the CNS and  cardiovascular system in accidental overexposure
 situations have not been reported  in  any human case report, even following fatal
 exposures.   The only evidence  of human nephrotoxicity resulting  from DCM over-
 exposure  was a finding of  congested kidneys following a fatal  exposure.   Ocular
 toxicity  other than eye irritation and congested conjunctivae  has  not  been
 reported  in humans  exposed  to  DCM.
 5.1.1.4  Occupational  Exposure—Ott et al.  (1983a,b,c,dse)  in  a  series  of studies
 examined  various health  evaluations of employees  occupationally  exposed  to DCM.
 This  involved a mortality study, clinical  laboratory  evaluation  study, metabolism
 data  and  oxygen half-saturation pressure study, and a 24-hr electrocardiographic
 monitoring  study.   The mortality study  examines  general  mortality,  ischemic
 heart disease,  mortality,  and  cancer  mortality, and is  discussed in  length in
 Section 5.3.3.2.2  as part  of the section on evaluation  of the carcinogenicity
 of  DCM.   No mortality effects  attributable  to  DCM  exposure  were demonstrated
 in  the  study, but given qualifications of  low  statistical power  and  confounding
 factors,  these  results do not  exclude  the possibility of  increased health  risks
 in  the study  population.
     Ott  et al.  (1983a,c)  studied  six serum constituents that had the poten-
 tial to detect possible liver  injury,  in relation to DCM exposures that ranged
 from 60 to  475 ppm (208 to  1650 mg/m3).   The  exposed group consisted of 313
 individuals; the reference group consisted of 321.  A dose-related rise in serum
bilirubin was observed for both men and women.   A consistent positive associa-
tion between  total  bilirubin and DCM  exposure  was  found in  three of  four sub-
groups by exposure.  The meaning of this  and  the  sensitivity  of changes  and
possible  associated  liver  damage are  not clear and tests which could clarify
the situation  (direct  bilirubin)  were not performed.   Women in  the  subgroup
exposed to  475  ppm  (1650 mg/m3) DCM showed  an  increase in red cell  counts,
                                     5-4

-------
 hemoglobin, and hematocrit, but men did not.   These findings are suggestive of
 a hematopoietic effect.   As expected, the carboxyhemoglobin concentration showed
 an increase and a possible association with DCM exposure for all four subgroup
 exposures.   Two standard liver function tests were not performed (SCOT,  SGPT),
 and these might have proved to be more sensitive indicators of liver function.
      Twenty-four-hour electrocardiographic monitoring was conducted by Ott et al.
 (1983d) in a study involving 50 employees, 24 of whom were occupationally exposed
 to DCM.  All subjects were white males between the ages of 37 and 63 years.
 The results showed neither an increase in ventricular or supraventricular ectopic
 activity nor episodic ST-segment depression to be associated with exposure to
 DCM ranging from a time-weighted average DCM  exposure of 60 to 475  ppm (208 to
 1650 mg/m3).   This study may  have  lacked adequate power to detect  expected
 changes.   Other  critical  factors that  may limit the meaning of the  study
 include choice  of  subject,  exercise  regimen,  and  control  possible in
 laboratory  studies but difficult for  field studies.
      A  further  study by Ott  et al.  (1983) of 136  DCM-exposed and  132 non-
 exposed employees produced  a calculated decrease in oxygen half-saturation
 pressure  between 2 and 4  mm  Hg among  persons previously  exposed to >  300 ppm
 DCM for an  8-hour work day.   The  expected  dose-response  increase, in the  level
 of  carboxyhemoglobin and carbon  monoxide,  were  produced from DCM exposure.
 Physiological adaptations  may have occurred,  but risk of ischemic events was
 not demonstrated  in the associated Ott et al.  (1983b) study  on  mortality,
 discussed in more  detail in Section 5.3.3.2.2.
 5.1.2   Chronic Effects
 5.1.2.1   Experimental  Exposure—Stewart and  his  colleagues extended  their
 studies of  the  effects  of DCM  on human subjects  to include longer  exposure.
 In  one  study (Hake et al., 1974), male volunteers were exposed 5 days/week for
 5 weeks  to  DCM  concentrations of 50  ppm (174 mg/m3)  in  week 1,  250 ppm  (868
 mg/m3)  in week 2,  250 ppm (868.mg/m3)  in week 3, 100 ppm (347 mg/m3) in week 4,
 and 500 ppm (1730 mg/m3) in week 5.  Three subjects were exposed for 1 hr/day,
 three subjects were  exposed for 3 hr/day, and four subjects were exposed for
 7.5 hr/day.  This complex experimental design was further confounded by attempts
 to  separate  smoking  and  nonsmoking populations.   The information that can be
 gathered from this report  is  summarized  in Table  5-1.  Alongside is a  summary
of a companion report from Stewart's group (Forster et al., 1974),  which  exposed
 nine female  subjects  to  250 ppm  (868 mg/m3) DCM  for  1, 3,  or  6.5 hr/day  for
5 days.
                                    5-5

-------
            TABLE 5-1.   COHb CONCENTRATIONS IN NONSMOKERS EXPOSED
                    TO DCM AT 250 ppm (869 mg/m3) FOR 5 DAYS
Exposure Time
hr/day
Pre-exposure
1
3
7.5
Mean of Daily Maximum
Male (n)a
0.9 (5)
3.3 (1)
7.0 (1)
9.6 (3)
Observed Average COHb (%)
Female (n)
1.4 (8)
3.5 (3)
5.1 (1)
10.1 (4)
aHake et al.,  1974.
bForster et al., 1974.

     Carboxyhemoglobin levels  up to 10 percent were  reported  in 10 men  (20 to
39 years old) and  9 women (20 to 41 years old) exposed to DCM concentrations
between 40 and  500 ppm (139 and 1,737 mg/m3) by inhalation (Peterson, 1978).
Exposure durations were between 1 hr and 7.5 hr/day for not more than 5 succes-
sive days.
5.1.2.2  Occupational Exposure—Toxic encephalosis due to occupational exposure
to DCM was diagnosed in a  chemist  (Weiss,  1967).  Other etiologies,  including
exposure to other  chemicals,  were not considered.  No liver or kidney damage
or ECG alterations were reported.  Erythema and fissures appeared on the hands
and forearms.
     Collier  (1936)  reported  four  cases  of occupational exposure to  paint re-
mover containing approximately 96 percent DCM.  The men, all of whom were pro-
fessional painters,  had  been  exposed to  lead  for 5  to  14 years.  During one
autumn, while they were removing paint, the workers complained of loss of appe-
tite, dullness,  faintness,  and giddiness while using the  remover and during
the following few  hours.   The author  diagnosed the symptoms  as  resulting from
slight chronic lead  intoxication and acute DCM-induced toxemia.
     Barrowcliff (1978)  and Barrowcliff  and Knell (1979)  reported that an in-
dividual exposed to 300  to 1,000 ppm  (1,032  to 3,474 mg/m3)  DCM for 3 years
developed  bilateral  temporal  lobe degeneration.   This was  thought  to result
from chronic  CO intoxication  as a result  of exposure to DCM.
                                     5-6

-------
 5.2   EFFECTS ON  LABORATORY ANIMALS
      Much  of  the experimental  research on the effects of DCM has been done on
 laboratory animals.  Such work can be  used to elucidate  the  general principles
 of DCM action and to discover which  target  organs or systems are involved.
 Generalization  of laboratory animal  findings to  humans,  however,  is  not
 straightforward.   Human  health  effects must,  in  the final  analysis,  be
 assessed in man.
 5.2.1 Acute Effects
                                                                         &
 5.2.1.1  Lethality—Only  preliminary  work has been done on  the  lethality  of
 DCM.   No  systematic data  exist  which would make  it  possible to specify a
 time-by-dose-by-lethality function.
      Table 5-2  is a  summary of  lethality  data.   The inhalation data  are
 remarkably consistent between  rats and mice.  The  one report on guinea pigs
 suggests that they may  be  more sensitive  to  DCM but procedural variation can-
 not be ruled  out.   Data on  shorter exposures  are  required since incidental
 exposures  and  substance   abuse   frequently  involve  high  concentration,'
 short-term profiles. A  curve of  LC50  for a  range  of  exposure times would  be
 valuable.
      Oral  and intraperitoneal injection LD50 data appear to be remarkably con-
 sistent across investigations and species.  Exceptionally low values (Zakhari,
 1977)  and  high  values  (Ugazio et al.,  1973)  are  possibly due to procedural
 differences.   The  mean  LD50  from  Table 5-2,  excluding the  low and h^gh  values
 mentioned,  for  injection  and oral doses in  rodents is near  2000 mg/kg.  No
 data  for other  sites  of injection were found.  Oral and intraperitoneal data
 cannot be  compared with inhalation data because of the  "first pass effect"
which occurs  following oral or intraperitoneal administration.
     Morbidity resulting after short-term exposure includes effects on several
different organs.  Ocular damage, liver damage,  kidney damage, changes in car-
diac parameters, and increased .pancreatic bile duct flow all  occur.
     The course  of events  during lethal exposures to  DCM via inhalation was
described by Berger and Fodor (1968)  who exposed rats to DCM concentrations
 ranging from 2,800 to 28,000 ppm (9.7  to 97.3 g/m3).   In these  exposures an
 initial period of  excitation was  followed by a deep narcosis  accompanied by  a
decrease in muscle tone and  a reduction in brain electrical  activity.  Rats
exposed to  concentrations of 25,000 to 28,000 ppm  (86.9 to 97.3 g/m3) ceased to
exhibit electroencephalographic  (EEC)  activity after  only 1.5 hours;  those
                                     5-7

-------




0)
0
CD
0)
ซ[^-
>rore •.- -a ^^re
Q. Q_ i<;cj3i^C3tOQQOQOQ
"re re
> >
^*> >00000 0
cs S- ;*s S- m in m m m /~s /~\ m
g3C330QQQQre^OQ
rHWCMW-J-l-l-l-J^-'^-' M-
E
cn
E
in
i- CO
i i i i i 1/1 in 3 in in
1 s- s- o S- S- II
3 3 -C 3 3
O O O O E
jc. jc LO -c ^: a.
• Q-
ID 1 — rH ID ID
in
r—
re
E E E
a. a. ••-
a. a. <=
re
0 0
O LO r—
cn cn cn cn cn E E - - E .--re
\s •v' J^^^Q-Q-OOOOO-
~^ •>,, -^,^,-^Q.a.CMrHO--C
cn cn cn cn cn i i tr~
* ^ SEEOOOOO ^
O LO OOOrHrHlDrHLO P
co cn cncniD ซซป>•.•< -r-
co cn cncncNj^iOLOiDrH >
rH rH rHrHrHrHrHlTMrHrH T-
p
U
re
V)
cn i—
•r- re
Q. U
re s-
Q) P
m a) a)a)u) in
re i"
•— OJ
re o
c ^
t-H s*->
5-8

-------
exposed  to  concentrations  between 16,000 and 18,000 ppm (55.6 and 62.5 g/m3)
ceased to exhibit EEG activity after 6 hours.
5.2.1.2  Nervous System  and Behavior—The  nervous  system is a likely  target
organ because DCM is lipophilic and thus is concentrated in myelin.  Rat brain
tissue has  been  shown  to have high concentrations  of  DCM  following  exposure
(Savolainen et al.,  1977;  Bergman, 1978; see also Section 4.1.3).  Pankow et
al. (1979) showed that nervous tissue function is affected by DCM dosage. They
demonstrated  a  linear  decrease in sciatic  nerve  conduction velocity, using
intraperitoneal injections of DCM at dose levels of 1 to 6 mmoles/kg.
     EEG and  rapid  eye movement (REM) in rats have been used to characterize
sleep during DCM inhalation.  These data form a concentration-related continuum
from 500 to 3000 ppm (1,737 to 10,422 mg/m3) in which REM sleep was reduced in
duration beginning  at  about 500 ppm (1,735 mg/m3) (Fodor and Winneke, 1971).
At 3,000 to  9,000  ppm  (10,410 to  31,230 mg/m3), sleeping  time was increased
with a further decrease  in REM sleep (Berger and Fodor, 1968).   Higher concen-
trations eventually  produced  a flat EEG after coma (Berger and Fodor, 1968).
The above may  be  viewed as a  continuum  of effects  beginning with REM sleep
reduction at  500 ppm (1,737 mg/m3) and ending in brain death after 1.5 hours
at 27,000 ppm (93.8 mg/m3) or 6 hours at 17,000 ppm (59 mg/m3).
     Measures of  general activity level are  commonly  used to characterize
behavioral  effects.   However,  general  activity is not a simple or unidimensional
measure.   Frequently, some measures of activity level are affected though others
are not.  A  concentration  of  5,000 ppm (17,370 mg/m3)  DCM administered for 1
hour to male rats was sufficient to decrease running activity (Heppel and Neal,
1944).   Running activity increased after exposure,  but was lower than activity
in the same male rats when they had not been exposed to DCM.  Similarly, Thomas
et al.  (1971) reported that the spontaneous activity of mice was decreased by
a 3-hour exposure to 1,000 ppm (3,474 mg/m3) of DCM.  Weinstein et al.  (1972)
exposed female mice in  groups  to about 5,000 ppm (17,370 mg/m3)  DCM for 24 hours
and observed progressive decreased spontaneous activity.
     In an experiment designed to determine whether the passive-avoidance con-
ditioning task was appropriate for measuring impairment of learning ability in
mice,  Alexeff and Kilgore  (1983)  exposed groups of Swiss-Webster mice (male)
to 168 mg/1  DCM  until  loss of righting reflex (-v 20 sec).   Groups of 10 mice
in age ranges of 3,  5,  and 8 weeks were tested in the task described by Essman
and Alpern (1964) and modified by  Reiter et al. (1973).  Recall of the condi-
tioning task was tested from 1 to 4 days following exposure.  In all  three age
                                     5-9

-------
groups, the  proportion  of  animals that  learned the task was consistently less
in the  groups  exposed to DCM.  The  3-week-old mice exhibited the most consis-
tent impairment  in  learning.   When  recall latencies of this  age group were
compared to  latencies of  the other groups by the Kruskal-Wallis test,  recall
was significantly lower (p = 0.01).   Learning impairment was observed particu-
larly in those animals  tested  at 1  or 2 days following training.  The percen-
tage of mice in  the control group  that recalled the task was significantly
higher  than  the  percentage of DCM-exposed mice  (p  = 0.024).   The  learning
impairment observed was not reported by the authors to be related to differences
in observable  behavior, motor  activity,  or analgesia.  In the analgesia test,
morphine was used as a positive control.
     Little can be said about the effects of acute DCM exposure upon the nervous
system and behavior of laboratory animals.  The effect appears to be consistent-
ly depressive  above 500 ppm (1,737  mg/m3).  However, to say that an  effect  is
"depressive" is  to  oversimplify  matters.  Other than the report by  Alexeeff
and Kilgore (1983), no work has been found on the effects of acute DCM exposure
upon motor  tasks, sensory  discrimination,  schedule-controlled  behavior,  or
response acquisition in laboratory  animals.   Similarly,  no research has been
published concerning the effects of acute DCM exposure on any electrophysiolog-
ical variable  other than nerve conduction velocity,  and  secondarily,  brain
electrical activity.  There have also been no follow-up studies to show whether
acute exposure effects were (or were not) reversible.
5.2.1.3  Cardiovascular Effects—The use of DCM in the study of cardiovascular
effects generally has been at a very high level (> 5,000 ppm;  17,000 mg/m ) in
short-term exposures (^ 5 minutes).   While knowledge of effects of such levels
would be  important  for  accidental  or substance abuse exposures, they are not
otherwise particularly  useful  for  assessment of health concerns relating to
environmental  exposures.   A summary of the effects of high level exposure in
rabbits and dogs is shown in Table 5-3 and draws upon the work of Taylor et al.
(1976) and Aviado et al. (1979).
     Several reports here  indicated that exposure of animals to  levels of DCM
                                 3
exceeding 5,000  ppm (17,000 mg/m )  caused cardiac arrhythmias (Aviado, 1975;
Aviado, 1977;  Zakhari,  1977; Belej  et  al.,  1974;  Aviado and Belej, 1974).
Additional cardiovascular  effects  of exposure to exceedingly high  levels of
DCM were  reported by  Loyke (1973),  Douglas et  al.  (1976),  Wilkinson et al.
(1977), Adams  and Erickson  (1976), and  Pryor et al.   (1978).
                                     5-10

-------
                TABLE  5-3.   SUMMARY OF CARDIOTOXIC ACTION OF 5% DCM
Function8
HR
MAP
LVP
CVP
LVEDP
LVdP/dt
CO
SV
VR
Taylor et al
1% t
0%
0%
0%
9% t
17% 4-
14% 4-
14% +
25% t
. (1976)
(NS)



(NS)




Aviado et al.
11% 4-
4% 4-
4% 4-
—
125% t
22% 4-
36% 4-
33% 4-
44% t
. (1977)b
(NS)
(NS)
(NS)


(NS)



 Comments:
          Rabbits

Pentobarbital-anesthetized
Spontaneously breathing
Close-chested
1-min. exposure (except
   1.5 min.  for CO)
Static dose (5%)
                                                                Dogs

                                                     Pentobarbi tal-anestheti zed
                                                     Artificially ventilated
                                                     Open-chested
                                                     5-min. exposure
                                                     Increasing  doses  (0.5, 1.0,
                                                       2.5, and 5.0%)
  Key:         HR - Heart Rate
              MAP - Mean Arterial Pressure
              LVP - Left Ventricular Pressure
              CVP - Central Venous Pressure
            LVEDP - Left Ventricular End-Diastolic Pressure
          LVdP/dt - Left Ventricular Rate of Time-Dependent Pressure Change
               CO - Cardiac Output
               SV - Stroke Volume
               VR - Vascular Resistance (Peripheral or Systemic)
               NS - Not significantly different from control values at p <0.05

  ""Results presented are for the highest dose (5%) of dichloromethane.
5-2.1.4   Hepatic,  Pancreatic,  and Renal  Effects—Klaassen and  Plaa  (1966)

injected Swiss-Webster mice (25 to 35 g) intraperitoneally with analytical grade

DCM.  Doses  of  13,300 mg/kg had  no  effect  on  Bronisulphalein  (BSP)  retention

or on serum glutamic pyruvic transaminase (SGPT) activity 24 hours after injec-

tion.  No  histopathological  change was seen upon  examination  of the liver.

     In the dog, however, effects on the liver by DCM were reported by Klaassen

and  Plaa  (1967).   They derived an ED5Q (effective dose  in 50% of  animals)  for

SGPT elevation of 798 mg/kg (the LD5Q was 1,260 mg/kg).   Histopathology showed

moderate neutrophilic infiltration in the sinusoids and portal areas.  Necrosis
was not seen.  At "near lethal" doses, there was vacuolization of centrilobular
hepatocytes.

                                     5-11

-------
     Reynolds and Yee  (1967)  gavaged fasted male Charles River rats (100 to
300 g) with  doses of 14C-DCM  up to 2,210 mg/kg dissolved in mineral oil.  The
rats were sacrified 24 hours  later.  No liver necrosis was seen and no change
in glucose-6-phosphatase activity was observed.   Labelled DCM (14C) was found
in hepatic lipids and proteins, minimally in the lipids and in high concentra-
tions  in  proteins.   Similar patterns of  incorporation  were  found in liver
microsomes.
     Weinstein et al.  (1972)  exposed female ICR mice (13 to  20 per group,  23
to 27 g) continuously to approximately 4,893 ppm (17,000 mg/m3) vaporized  tech-
nical grade DCM in Thomas Domes, at pressures slightly below  ambient.   Tempera-
ture, humidity, C0?, and air flow were monitored and controlled.   During a
24-hour period,  there  was a  progressive  decrease of spontaneous activity.
Body weight  decreased,  liver  weight  increased absolutely and  as  a ratio  to
body weight, liver triglycerides increased (indicating liver  toxicity),  glyco-
gen decreased,  and  protein synthesis was reduced as  shown by reduction of
3H-leucine incorporation.
     Many histological  changes  appeared.   Fatty infiltration after 24 hours
involved the entire lobule; centrilobular hepatocyte nuclei became smaller and
denser, and ballooning  developed.   Staining showed glycogen reduction.   Progres-
sive changes began  after 12  hours of exposure.   Smooth (SER) and rough (RER)
endoplasmic  reticulum showed  changes:  polysomes  broke down, ribosomal parti-
cles detached from RER  in the centrilobular cells, RER membranes broke up  into
vesicles, perinuclear cisternae dilated, and lipid  droplets increased.  Mito-
chondria, however, were unaffected.   The authors concluded that the pattern of
liver  damage was  similar  to  that  following carbon tetrachloride exposure.
     Morris et al. (1979) exposed male Hartley guinea pigs weighing 500 to 750
g for 6 hours to a 5,181 ppm (18,000 mg/m3) vapor concentration of reagent grade
DCM.  Animals were sacrificed immediately after exposure and  liver samples were
taken.    Liver  and serum were analyzed for  triglycerides, which  increased
markedly in the former  and decreased in the latter.   Liver phospholipids showed
no change, neither did  serum-free fatty acid.  Liver slices were incubated with
14C-palmitic acid.  The uptake of 14C was similar to controls.   Uptake of  14C-
leucine did not differ between controls and exposed samples.
     Differences in protein  synthesis  in the above two studies may be due to
different periods of exposure and the use of different animal species.
                                     5-12

-------
     Harms et  al.  (1976)  cannulated the  bile  duct  of male  Sprague-Dawley  rats
(350 to 450 g) after pretreatment with intraperitoneal injections of 670 mg/kg
DCM dissolved  in  corn oil.   Tritiated 3H-insu1in was instilled into the duct
24 hours later.  DCM seemed to induce an increase in pancreatic bile duct flow,
which was unrelated to any observed hepatic effect.  Hamada and Peterson (1977),
in a  follow-up study,  intraperitoneally injected 860 mg/kg DCM (dissolved in
corn oil) in male Sprague-Dawley rats (280 to 320 g).   The bile duct was cannu-
lated, and bile  duct pancreatic flow and its contents were measured and com-
pared with controls.  DCM induced increased pancreatic bile duct flow, decreased
protein concentration, and  increased chloride,  sodium, and potassium.  Bicar-
bonate was unaffected.   There was no statistically significant difference in
wet weight of  the  pancreas  or in  total bile flow;  this may indicate a  reduced
hepatic bile flow.   An  experiment with secretin indicated that these changes
were  not  related to that substance  or,  after an ancillary experiment with
atropine, to any cholinergic effect.
     Plaa and Larson (1965) injected 10 male Swiss mice (18 to 30 g) intraperi-
toneally with 1330 mg/kg of DCM (source and quality unidentified) dissolved in
corn oil.   No glycosuria or proteinuria was detected 24 hours later.   Two sur-
viving mice of 10 injected with 1,995 mg/kg DCM had proteinuria but not glyco-
suria.  No renal  histopathology was seen after the lower  dose.  Proteinuria
following the  higher  dose suggested that there might have been some tubular
damage.
     Klaassen and  Plaa (1966)  also  injected male Swiss-Webster mice (25 to 35
g) intraperitoneally with  13.3 g/kg analytical grade DCM.   These mice showed
no glycosuria, proteinuria, or changes in BSP  excretion 24 hours after injec-
tion.   Two groups of 10 mice each were gavaged for 3 days with 5 g/kg 60 percent
ethanol and equicaloric solutions of dextrose.  All animals were then injected
with DCM, and  urine was collected and analyzed  24  hours later.  BSP excretion
remained within control limits-, indicating the  absence of  a renal lesion, but
the authors reported, "A few kidneys exhibited hydropic degeneration with mini-
mal necrosis of the convoluted tubules."
     Kluwe et al.  (1982) have investigated the effects of DCM on renal  tubular
cells of adult male Fischer 344 rats by injecting DCM (1300 mg/kg),  i.p., in
corn oil.  Renal  proximal  tubular swelling was observed in the cortex and in
the outer medulla.  Tubular cell functions measured included organic ion trans-
port,  ability to maintain a potassium gradient, and the rate of oxygen utiliza-
tion.   None of these functions was altered by DCM.   Rats were sacrificed 2,  12,
                                     5-13

-------
24, 48, or 96 hours after injection, the kidneys were removed, and thin slices
were prepared.  DCM was not observed to alter glomerular, distal tubular, inner
medullary or papillary cell morphology.  Although DCM did cause an increase in
the fraction  of water in  renal  tissue,  the  lack of  effect on  other parameters
suggested to  the  authors  that  little or  no functional  cell disturbance had
occurred.
5.2.1.5  Other effects— Ballantyne et al.  (1976) studied the effects of liquid
DCM on various ocular parameters by instilling 0.1 and 0.01 ml of DCM in rabbits'
eyes.   Lachrymation persisted for a week, inflammation of the lids and conjunc-
tivae for 2  weeks,  conjunctival edema  for  a week,  sloughing  for 3 days, and
increased cornea! thickness  for 9 days.   Iritis  and keratitis appeared within
6 hours and lasted for 7 and 14 days, respectively.   Intraocular pressures in-
creased also.  Increased cornea! thickness  developed in rabbits exposed to DCM
vapor at concentrations of 504 and 5,040 ppm (1,750 and 17,500 mg/m3).
     Sahu and  Lowther (1981) observed that inhalation  of DCM by 2-month-old
Sprague-Dawley rats  led to pulmonary injury, presumably rupture  of  type II
alveolar cell  membranes and  release of celj contents into the airways.   Rats
were exposed  to about 4000 ppm  (13,896 mg/m3)  for  5 hr/day,  5 days/wk,  for
4 weeks.  Pulmonary  secretions were obtained  by lung lavage.  The  soluble
supernatant of the lung homogenate and the  cell-free lung lavage were used for
determination of lung lipid peroxidation.   Lipid peroxidation was significantly
elevated (P < 0.05).
5.2.1.6   Summary of Effects  of Acute Exposure  on Laboratory Animals—Of  all
the organ systems studied,  the central nervous system appears to be affected
by DCM  at levels ranging  from  500 to 1,000  ppm  (1,737 to 3,474 mg/m3).  While
only short-term exposures were  used, levels of  over 20,000  ppm (69,000 mg/m3)
were required  before  cardiac function changes were  produced.  Hepatic effects
were reported at exposure levels as low as 5,000 ppm (17,370 mg/m3).   While it
is difficult  to  compare  results of injection studies to those of inhalation
exposures, it  appears  that half the LD50 is required  to produce hepatic or
renal  changes.  The above comparisons are based upon the lowest level reported.
In two cases, however, the behavioral data were obtained for levels of exposure
at or below 1,000 ppm.  The weight-of-evidence  for  short-term exposures indi-
cates that the CNS is the primary target organ for DCM.
     The concentrations of DCM necessary to depress cardiac function (25,000
to 50,000 ppm) in acute experiments are so  high that chronic long-term exposures
                                     5-14

-------
of humans to levels considerably in excess of 250 ppm (865 mg/m3) would be un-
likely to have  any effect.   However, comparatively  little research has been
done on acute DCM effects on laboratory animals.
5.2.2  Chronic Effects
5.2.2.1  CNS Effects--In early studies of DCM chronic exposure (Heppel et al.,
1944; Heppel and  Neal,  1944),  dogs, monkeys, rabbits, guinea pigs, and rats
were exposed 4 hours/day, 5 days/week for 8 weeks to 5,000 or 10,000 ppm (17,370
or 34,740 mg/m3) DCM.  Unfortunately, only qualitative observations of behavioral
"symptoms" were reported.   No  symptoms were noted in groups exposed to 5,000
ppm  (17,300  mg/m3).   At 10,000 ppm (34,740 mg/m3) all species were affected
but in different ways.  Dogs became excitable and hyperactive.   Monkeys became
progressively more  inactive and, by the end of  each  daily exposure, lay pros-
trate with barely  perceptible  respiration.   Rabbits first were excitable and
then became  inactive  toward the end of each daily session.  Guinea pigs and
rats simply became more inactive.  All species appeared well within 1 hour after
cessation of exposure.   Only  monkeys (n = 2) appeared to develop  behavioral
tolerance.  No other cumulative effects were reported over the course of the 8
weeks.
     Weinstein et  al.  (1972)  exposed female mice to 5,000 ppm (17,300 mg/rn3)
DCM  continuously  for  7 days.   Only qualitative  behavioral observations were
reported.  For  the  first few hours of exposure, the animals exhibited an in-
creased activity level and increased food and water intake.   This was followed
by decreased activity,  "hunched" posture, dehydration, and the appearance of
roughened yellow  coat.   By the fourth day of exposure, many of  the mice had
adapted so that by the end of the study they were virtually normal.
     Thomas et  al.  (1972)  studied  activity level  in  mice continuously exposed
to 25, 100,  or  1,000 ppm (87, 347, or 3,474 mg/m3) for 14 weeks.  The lowest
exposure  level  elevated activity level.  "There was no effect at 100 ppm (347
mg/m3); at 1,000  ppm  (3474  mg/m3),  there  was reduced activity level.  The  in-
creased activity level in the 25 ppm (87 mg/m3) exposed group was, if replicable,
probably  due to the increased sensory stimulation due to  the odor of DCM.
     Savolainen et  al.  (1977)  exposed  male rats to 500 ppm  (1,737  mg/m3) DCM,
6 hr/day  for 4 days.  Exposed rats engaged in more grooming behavior than con-
trols during the first exposure hour but by the seventeenth exposure hour (third
day) exposed rats no  longer differed  from  controls.  Other behaviors were
observed, but by  their absence in  the  results summary, it may be assumed that
                                     5-15

-------
only grooming was significantly affected.  Biochemical analysis of tissue from
the right cerebral  hemisphere  showed  no  difference  in protein,  RNA, or gluta-
thione levels as  compared with levels in control animals.  Relatively small
increases in acid  proteinase and nonspecific cholinesterase activities were
reported, but the determinations for treated animals were made by only two assays
and may therefore be of questionable significance.
     Since only one study of activity level is available, it is difficult to
conclude much about neurobehavioral effects of chronic exposures.  As in acute
exposure studies,  no other  behaviors,  such as  sensory  disturbance,  motor
response acquisition, or  schedule-controlled behaviors,  were studied.  Where
effects were seen, no follow-up studies were conducted to determine the irrever-
sibility of effects.
     Suggestions  of species  differences  in DCM  sensitivity  occurred  in  the
studies by Heppel  et al.  (1944) and  Heppel  and  Neal  (1944).   Based on these
qualitative observations,  one  could conjecture that DCM  behaves  in a manner
similar to other  anesthetics:   There  is  an  early excitatory phase  followed  by
progressive depression.   If  some species had different sensitivities to DCM,
they might remain in the excitatory phase or progress more rapidly into depres-
sion.
     Several  reports mentioned an increased tolerance to DCM over the duration
of chronic exposures (Weinstein et al., 1972; Savolainen et al., 1977).   Heppel
et al. (1944) also  reported  monkeys to have adapted somewhat.   Probably adap-
tation, like sensitivity, is a function of species,  exposure level, and duration.
     Because of the paucity of data and the qualitative approaches used,  these
conclusions must  be regarded as tentative.   CMS  effects  have been  reported  at
exposure levels as low as 1,000 ppm (3,474 mg/m3) but not lower.  The one case
of increased activity level at 25 ppm (87 mg/m3) must be discounted.
5.2.2.2  Hepatic  and Renal Effects—Many of the  "high-dose"  chronic animal
studies with DCM  have revealed a certain degree of liver and kidney  involve-
ment as  target  organs.  The  magnitude  of this  involvement increases  in such a
way that extremely  high doses  of DCM  (depending upon dosage and duration of
exposure) can produce  toxic  .effects upon these organs.   Heppel et al. (1944)
observed moderate centrilobular congestion and fatty degeneration of  the liver
in dogs  and  guinea pigs exposed to 10,000 ppm (34,740 mg/m3), 4 hours/day, 5
days/wk for 8 weeks.  Weinstein et al. (1972) reported identical findings after
exposing mice  to 5,000  ppm  (17,370 mg/m3) continuously  for  7 days.   High
                                     5-16

-------
mortality was observed  by  MacEwen et al.  (1972) where 14 weeks of continuous
exposures at 1,000 or 5,000 ppm (3,474 or 17,370 mg/m3) resulted in severe toxic
effects  and  a high degree  of  mortality  in mice, rats,  dogs,  and monkeys.
     More important, chronic mouse studies by Weinstein and Diamond (1972) and
Haun et  al.  (1972)  have revealed that continuous exposure even to levels of
DCM as  low  as 100 ppm  (347 mg/m3) can effect changes  in both  liver function
and cell architecture.
     Weinstein and  Diamond  (1972)  exposed ICR mice (17 to 25 g) continuously
for 3 days to 10 weeks to 100 ppm (347 mg/m3) of chemical grade DCM in a Thomas
Dome with specified dynamic characteristics and at 96.66 Pa (725 mm Hg) pressure.
Twelve groups of  16 mice each  were used.   Except for one instance at week 2,
body weights were comparable to controls.   Liver weights followed body weights,
and in  all  cases,  liver to body weight ratios  were within control limits.
Triglycerides increased approximately threefold at week 2,  and nearly fourfold
by week  3, but then decreased  to about double at week  4.  Four mice withdrawn
at 3 days showed no abnormalities, but at 7 days, centrilobular fat accumulation
was seen, accompanied by a decrease in liver glycogen.  These abnormalities
persisted to the termination of the experiment at 10 weeks.   During this time,
the nuclei enlarged.  No other histopathology was seen under the light microscope.
Under the electron  microscope, autophagic vacuoles  containing  debris appeared
in the hepatocytes.   The smooth and rough endoplasmic reticulum showed no changes.
     Haun et al.  (1972) exposed mice, rats, dogs, and monkeys to 25 and 100 ppm
(87 and  347  mg/m3)  reagent grade DCM for  continuous  exposure periods up to
100 days in  Thomas  Domes at ambient  pressure.  This is the longest continuous
exposure study reported.   Strain,  sex,  and size of the test animals were not
specified.   Exposure  to the lower concentration  had no observable effect, but
exposure to the higher concentration resulted in positive fat stains and vacuo-
lization.  The rats showed nonspecific  tubular degeneration and regeneration
in the kidneys,  but no changes in organ-body weight ratios.
     Norpoth  et al. (1974)  tested DCM in studying  the possibility of  enzyme
induction in  inhalation of  hydrocarbon solvents.  Male SPF Wistar rats  (80 to
100 g) were  exposed 5 hours/day to vapors containing 0,  500,  or 5,000  ppm (0,
1,737, or 17,370 mg/m3 DCM) of the hydrocarbon solvents for 10 days or 250 ppm
(869 mg/m3 DCM) of the solvents for 28 days.  There were 50 animals in the con-
trol group and six animals  in each of the exposed groups.  At the end of these
                                     5-17

-------
exposure periods, the animals were killed and the concentration of liver cyto-
chrome P4so and microsomal aminopyrine N-demethylase activity were determined.
     Following the 10-day exposure, a significant increase in liver cytochrome
P450 in animals exposed to 500 ppm (1,737 mg/m3) but not in animals exposed to
5,000 ppm (17,370 mg/m3), of the compound was reported.  In contrast, aminopyrine
N-demethylase activity was  not elevated in animals exposed to 500 ppm (1,737
mg/m3) DCM but was substantially elevated in those exposed to 5,000 ppm (17,370
mg/m3) DCM.  After 28 days of exposure to DCM at 250 ppm (868 mg/m3), no changes
were noted in liver enzymes, liver weight, or histologic appearance.
     These results  indicate  that differential enzymatic  induction  can be pro-
duced by exposure  to DCM. , The inverse  dose-response  relationship noted for
cytochrome P450 may be due to the combined effect of DCM and CO.  Acclimatiza-
tion may have  occurred  by the 28th day of exposure to ameliorate the effects
seen at 10 days.   In addition, the doses given in the second exposure period
(28 days) were much smaller than during the first period.
     Loyke (1973) induced chronic renal hypertension in 13 Sprague-Dawley rats
(100 g, sex not specified).   Both poles of one kidney were ligated and the con-
tra! ateral  one was removed.   After maintenance of high systolic pressure for 3
months, 11 experimental  rats and 3 controls were  injected  subcutaneously with
2 mg/kg unspecified DCM biweekly for 15 doses.  Two hypertensive rats were used
for positive controls and 11 normotensive rats as naive controls.   DCM reduced
the systolic blood  pressure  of the hypertensive  rats  from 200  to  160 mm Hg.
The positive controls remained hypertensive.  DCM was  ineffective  in reducing
blood pressure in  normotensive rats.   No changes ascribable to DCM were seen
in the liver.
     In the Dow Chemical  Co.  (1980) chronic inhalation study in rats, exposure
to 500, 1500,  or  3500 ppm (1,737,  5,211, or  12,159 mg/m3),  for 6  hours/day,
5 days/ week for 2  years resulted in exposure-related non-neoplastic hepatic
lesions in both males and females.  Grossly,  the  effect was  most prominent in
females exposed to 3500 ppm and consisted of increased numbers of dark or pale
foci.   The percentages of total rats with any degree of vacuolization were 17
percent, 38  percent,  45  percent,  and 54 percent  in the males of the 0, 500,
1,500, and 3,500 ppm exposure groups, respectively, and 34 percent, 52 percent,
59 percent, and 65 percent in the females, respectively.   The degree of severity
tended to  increase  with  the  dose.   Further information relating to protocol
and additional  observations are discussed in Section 5.3.3.1.1.
                                     5-18

-------
     In the Dow Chemical  Co.  (1980) 2-year inhalation study in hamsters at the
same levels described  above  for rats,  a variety of gross and histopathologic
observations were recorded for hamsters sacrificed at the 6-, 12-,  or 18-month
interim points.  Histopathologically,  exposure-related  differences consisted
of hamsters with amyloidosis of the liver, kidney, adrenals, thyroid, and spleen.
Further observations are discussed in Section 5.3.3.1.3.
                                                                          t
     In the Dow Chemical  (1982) 2-year inhalation study in Sprague-Dawley rats
(Spartan substrain) exposed to 50, 200, and 500 ppm (174, 695, and 1,737 mg/m3),
no definite exposure-related histopathologic findings were noted.   An exception
occurred with  females  sacrificed at 15 months.   Some had  a  focus  or foci of
altered liver cells.  There were significant increases in non-neoplastic liver
lesions (i.e., hepatocellular vacuolization and multinucleated hepatocytes) in
female rats at 500 ppm (1,737 mg/m3).   Further observations are discussed in
Section 5.3.3.1.4.
     Histopathologic changes in liver cells of Fischer 344 rats exposed to vari-
ous  levels  of DCM  in drinking water over a  2-year exposure  period have  been
reported by the  National  Coffee  Association  (1982).   Details are described  in
Section 5.3.3.1.5.
5.2.2.3  Morbidity and Mortal ity—Heppel et al. (1944) conducted two inhalation
experiments.   One  used 489 ppm (1,700 mg/m3)  commercial  DCM for 7 hr/day,  5
days/week for  6 months.  The other involved exposure for 4 hr/day, 5 days/week,
for 7 to 8 weeks at a concentration of 9,789 ppm  (34,000 mg/m3).  Dogs, rabbits,
guinea pigs,  and rats were used in each experiment.  Two monkeys were added to
the  experiment at  the  higher concentration.   Animals were exposed  together  in
a  single  chamber.   Temperature and humidity were  uncontrolled.  At  the lower
dose, 3 of 14 male guinea pigs died.  They had fatty degeneration of the  liver
and pneumonia.  No other animals'  deaths  attributed to DCM exposure were  reported.
     At the higher dose, the dog experiment was terminated after six exposures
because of  the continuing Stage II excitement  reactions.   Three  rabbits and
one  rat  died  during the course of the experiment.   Each animal had  extensive
pulmonary congestion.  Clinical observations  in the  dogs at  the lower dose  showed
no changes  in blood pressure,  blood chemistry, or liver  function tests, and at
autopsy, organ weights of  liver,  kidneys,  heart,  lungs,  and  spleen were similar
to controls.   No lesions due  to DCM were found.   All animals  showed clinical
effects  after exposure to the higher concentration.   Aside from the responses
in dogs noted above, the  other animals  showed progressive  signs of depression,
                                      5-19

-------
 usually  becoming  prostrate at the end  of  each daily exposure period.  All
 animals, including  dogs,  recovered rapidly upon  removal  from  the chamber, and
 fed well.  At  autopsy,  dogs and guinea pigs showed fatty degeneration of the
 liver.   Pulmonary congestion was  found in the  rabbits.   Monkeys  and rats
 showed no lesions related to DCM exposure.
 5.2.2.4  Summary of Effects of Chronic Exposures on Laboratory Animals—Chronic
 exposure to  high  levels  of DCM has been reported  to produce alterations in
 behavior and in  hepatic  and renal function.  When deaths occurred, they were
 frequently due to pulmonary congestion.  Apparently these effects begin at about
 500 to 1,000 ppm with behavioral effects in evidence early, followed by changes
 in internal  organs.   No  data are available to determine if the effects seen
 were reversible after cessation of chronic exposures.   This issue  could best
 be addressed through additional  research.
 5.3  TERATOGENICITY, MUTAGENICITY, AND CARCINOGENICITY
 5.3.1  Teratogenicity, Embryotoxicity, and  Reproductive Effects
      It is  not possible,  on the  basis  of limited available  data,  to define the
 full  potential of DCM to produce adverse teratogenic or reproductive effects.
 Human epidemiological studies that evaluate the  effects of DCM on the exposed
 population  are difficult to conduct.   Each of the available mammalian studies
 had methodological drawbacks that  do  not allow  for conclusive evaluation of
 the ability of DCM to produce a  teratogenic response over a wide  range of doses,
 which should include doses  high enough  to  produce signs of maternal  toxicity
 and lower doses that  do  not produce  this  effect.   Other studies in  chicken
'embryos have  indicated that DCM  disrupts embryogenesis  in a dose-related manner
 (Elovaara et al., 1979).   However, since administration of DCM dfrectly into
 the air space  of  chicken  embryos  is not comparable to  administration of dose
 to animals  with a  placenta,  interpretation  of  this  result related to  the poten-
 tial  of DCM to  cause adverse human reproductive effects is  not possible.   Another
 preliminary study  in rats  indicated that adverse  effects on offspring behavior
 may occur after maternal exposure to  high levels of DCM.  However,  additional
 behavioral  studies have  not been conducted  to  evaluate  this effect  more  fully.
 5.3.1.1  Animal Studies—The following discussion subscribes  to basic viewpoints
 and definitions of the terms  "teratogenic" and "fetotoxic" as summarized by
 the Office  of Pesticides and Toxic Substances  (U.S.  EPA,  1980):
      Generally,  the term "teratogenic"  is  defined as  the  tendency  to  produce
 physical  and/or  functional  defects  in offspring iji utero.   The  term "fetotoxic"

                                      5-20

-------
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, reduc-
tions 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
categories.  Rather,  the  spectrum of adverse embryonic/fetal effects  is con-
tinuous, 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  necessarily 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 Environmental  Protection
Agency (EPA) 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  subcategory would
be teratogenesis  and  would encompass those changes (structural  and/or func-
tional) which are induced prenatally, and which is 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 jjj utero exposure.  The third cate-
gory would be embryo or fetal  toxicity as comprised 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.

     Two major problems with a definitional  scheme of this nature must be pointed
out, however.   The first is that the reversibility of any phenomenon is extreme-
ly 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 is performed on the neonate,  however, no conclusion may be drawn
concerning permanent  organ  functional  changes.  This same uncertainty as to
possible long-lasting  after effects  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.


5.3.1.2  Mice—Swiss-Webster mice  (30 to  40) were  exposed via inhalation to
1,250 ppm (4,342 mg/m3) of DCM (97.9  percent pure)  for  7 hr/day  during days 6

through  15  of gestation  (Schwetz et a!., 1975).   Two  control  groups  were
similarly exposed to filtered room air.   Day 0 of gestation was determined when
a vaginal plug was observed.   Caesarean sectioning of  dams  was  performed on
day 18 of gestation.

     Dams were  evaluated for  body weight gain and various  organ weights.
Maternal body weights  were  recorded on days 6,  10, and 16 of gestation,  as
well as  on  the  day of caesarean section.   Maternal COHb level determinations

                                     5-21

-------
were performed on blood samples collected via orbital sinus puncture immediately
following the third and tenth (last) exposure.  Following caesarean sectioning,
fetuses were weighed,  measured (crown-rump length), sexed, and examined for
external malformation.  One-half  of the fetuses in each litter were examined
for soft-tissue malformations (free-hand sectioning) and one-half were examined,
following staining, for skeletal  malformations.  One fetus  in each  litter was
randomly selected and evaluated using histological  techniques following serial
sectioning.
     In this study, maternally toxic effects of DCM exposure were  observed,
consisting of a significant increase in body weight, a significant increase in
absolute liver weight, and significant increases in COHb values with return to
control levels after 24 hours.  On the basis of the maternal liver weight obser-
vations, a minimal  toxicity may have occurred.  The cause  of the increased
maternal weights is unknown.
     Of the 12 litters examined, a statistically significant number of litters
contained fetuses that had a single extra center of ossification in the sternum.
This common variation  in  mice is thought  to  reflect the degree of  embryonic
development.   It is not  known if this observation resulted from ah accelera-
tion in development or was  a  chance  occurrence.  The litters  in exposed group
were heavier than  control  fetuses (5.74 g  vs.  5.42 g);  however this may be
caused by the  average  slightly smaller litter  size compared  to controls (10
vs. 12).  The  litters  in  this treatment group  also had a lower incidence of
delayed ossification of the sternebrae (17 vs.  23 percent), split sternebrae
(8 vs. 18 percent), and  ossification of the skull  bones (25 vs.  36 percent).
Cleft palate and "rotated kidney" were observed in two (17 percent) of the DCM
exposed fetuses, but  not  in any of  the control litters.  Because of the low
incidence of these effects, these effects may reflect spontaneous malformation
rates.
5.3.1.3  Rats—A study using the same design as that used for the mice (Section
5.3.1.2) was performed  in  Sprague-Dawley rats  (Schwetz et  al., 1975).  Rats
inhaled DCM at  1,250  ppm  (4,342 mg/m3) for 7 hr/day on days  6 through 15 of
gestation, with day 0 being the day spermatozoa were observed in vaginal  smears.
Dams were Caesarean sectioned on day 21 of gestation.  All other procedures
were identical  to  those performed in the mouse  study with the exception that,
in the rat study, food consumption was monitored.
     No effect  on  maternal  body weight  or  food  consumption  was observed.  The
average absolute maternal  liver weight was significantly increased in compari-
son with the control values but there was  no  effect on the  relative weight  of
                                     5-22

-------
the liver.   Carboxyhemoglobin values in the dams .increased significantly during
exposure but returned to control levels within 24 hours.
     In the 19 litters evaluated, there was no effect on the average number of
implantation sites per  litter,  litter size, number of resorptions, or fetal
sex ratio  and  body weight.   The incidence of dilated renal pelvis was signi-
ficantly increased, but this observation might indicate a slight but reversible
delay in development similar to delays in sternal ossification.   However, since
this study evaluated only one dosage level, it is not possible to firmly estab-
lish the significance of this effect or its reversibility.
     Hardin and Manson  (1980)  used Long-Evans rats to evaluate the effect of
exposure to DCM (97  percent pure) via inhalation at 4,500 ppm (15,633 mg/m3)
for 6 hr/day,  7 days/week to determine whether  exposures before and during
gestation  were more  detrimental to the developing  conceptus  than exposures
before  gestation  only.   Minimal maternal  toxicity, consisting  of increased
absolute and relative liver weight and elevated COHb levels, was bbserved. The
litters of  rats exposed to DCM during gestation also had significantly lower
fetal body weights than controls.  No other significant deleterious effect was
observed.   Twenty  animals were used to evaluate teratogenicity.   Ten were used
in a separate evaluation of behavioral toxicity.
     Bornschein et al.  (1980)  reported on the behavioral teratogenic effects
in the Long-Evans  rats exposed to DCM from the Hardin and Manson (1980) study.
Ten rats were  evaluated for general activity at  5,  10,  45, and 108 days of
age, avoidance learning at approximately 4 months of age, and activity follow-
ing avoidance  learning  at approximately 5  months  of age.  Fetuses  delivered  by
Caesarean  section  had  lower fetal  body weight than those that were naturally
delivered.   Treatment-related effects were reported for animals in the general
activity tests as  early as 10 days of age (both sexes) and were still demon-
strable in  male rats at 150  days of age.  -No adverse effects were observed  in
growth  rate,  long-term  food  and water  consumption, wheel  running  activity,  or
avoidance learning.
     In the Bornschein  et al.  (1980)  study, the  number of rats per test  group
was small,  usually one  male  and one female per litter.   Therefore, this  study
should  be  regarded as preliminary,  and additional studies are needed  to  fully
confirm these  effects.   Also,  the  entire field of behavioral teratology  is  in
its early  stages  of  development (Buelke-Sam and  Kimmel,  1979), and the signi-
ficance of  alterations  in behavior to human risk assessment  is not clearly
defined.
                                     5-23

-------
5.3.2  Mutagem'city
     Dichloromethane has been tested for mutagenic activity in bacteria, yeast,
insects, nematodes,  mammalian cells jjn  vitro,  and  rodents.  These  studies are
discussed below and are summarized in Tables 5-4 to 5-9.
5.3.2.1  Gene Mutations
5.3.2.1.1  Bacteria.   There  are 14 reports in  the literature concerning the
mutagenic potential  of DCM in  bacteria; the  Salmonella histidine reversion
assay was  used in  all of these studies (Simmon  et al., 1977;  Simmon  and
Kauhanen, 1978; Kanada and Uyeta,  1978; Jongen et al., 1978;  McGregor, 1979T*
Snow et  al.,  1979;  Green, 1980; Rapson et  al., 1980;  Barber et al.,  1980;
Nestmann et al., 1980; Green, 1981; Gocke et al., 1981; Nestmann et al., 1981;
and Jongen et al.,  1982).  Kanada and Uyeta (1978) also tested DCM in the B._
subtil is rec assay.  DCM tested positive in all studies using Salmonella without
or with metabolic activation in strains TA100, TA1535, or TA98 when assays were
performed in sealed gas tight exposure chambers.  Negative responses were report-
ed by Rapson  et al.  (1980) and Nestman et al. (1980) in standard assays, but
these tests are judged to be inadequate because DCM was  added directly  to the
agar medium, and no precautions were taken to prevent excessive evaporation of
the test material.  The tests were carried out at 37ฐC, which is very close to
the boiling point of DCM  (39ฐC);  thus,  it  is likely excessive evaporation
occurred.  Data were presented in many of the reports, and a clear dose-related
response is apparent for each,  with 10-fold or greater increases in revertants
observed at the highest doses compared to negative controls.   The doses employed
and the responses observed are summarized in Table 5-4.
     The purity of the test material was not  given in  any report.  Because of
this, most positive  responses must be viewed  with  caution; they may be  caused
by substances other than DCM.   For instance, formaldehyde, a metabolite of DCM,
could form nonenzymatically in the aqueous solutions used for biological testing
by hydrolysis  of  DCM (March,  1977).   Because  of the consistency of positive^
responses with several  different samples, it  is more likely the DCM itself is
mutagenic.   For instance,  in  their tests of DCM,  Barber et al.  (1980) used a
redistilled sample estimated to be >99.9 percent pure,  containing  only  traces
of 1,1- and 1,2-dichloroethane,  chloroform,  chloromethane, and an unidentified
CgH-,Q aliphatic material (Dr.  E. Barber, Eastman Kodak, personal communication).
Nestmann et al. (1981) reported that their sample was "gas chromatographically
pure," and Gocke  et  al. (1981)  checked their sample for the "correct melting
                                     5-24

-------



















r-H
Cฃ
UJ



5
s:

o
LL.
O

i

H-
CO
LU

>;
(—4
CJ
t-H

UJ
g

s:
i
in
LU
CQ
ซ^
































(A
•P
O












w
•p

u
•• 0)
0

-p
(O
S-
.p
c
o>
u
c
o
CJ














c
•r~ E
4-1 Q)
ro -p
> W

•P CO
JJ
 U ro 3
S- -i- S- O
0) S- -P 0.
W OJ W E
.O G- O


OJ 1/1 M— M—
•P -P TD 0
S- C Qป -D
o to ซr—  4- -P U tO
OJ S- -r- C S- C
s- ai u a> 3 o
> QJ (/) . o a.
j_> aป Q_ fli Q t/> o>
o $- in fc. o at
C Q,rH TD t_
<ฃ -P 
•r- S- C C r^TJ-r-
u tt) o ro 4J -i- -P
•r- J2 O fO J= -r- > •ซ-
0 3 
Q. O ^NS fO
to ai CM i — u

x tOio o in
^>C rH CO ~O








rH
r~ซ
ro
•P
0)

c
o
i
—
CO

•D C
C ซP-
3
O r—
a. o i
0 P *>,
• u c ฃ
-a OP
a> ^- u aj
•P O O
t- vs.
O 0) > O
O. U -r- r--
aป t- +3 j=
ฃ-3 .r-0
o to i
•P Ul O CM
o a.
c -a 4-
C . ro O






e.
response.
>, TO T3 +J CD
•P Q) UJ O) CO >
•r™" >ป"O CQ C E •ป"
U P *r- -r- t~ -J-J
•r- ซr- > T3 p
x s- o a> tn
(_ Q_ (^^3 4J

. .ซ .Jg
rH CM C CO 4-ป
^|oo ^D "ej1 CO
•P ko PO r*. in
C rH rH rH rH

o|


"O
10 a>
•P -P CO ซ* O CM
o c to oo ซ3- ro rH
o co a> to ro co en
rH P I_ rH

ฃ ^^
aj u i + i +
E 
-------
s.
                                                 - "O Oป 3 -r-

                                                 > 0) > OT W)
                  •  E *r-       O-ซX  O     •*•> 03 C T- :
                  >  a) .a o    
                   4-1     -M    t-     tj -r- tn 
           . ZJ  I- -r
          01 0) -r-  O r— t.
                       o ฃ ••- i
                4-> •*-! T3 O >
                                                   o at (n  c
                                                   O..C C -i- 4
                                                   in en o  ra
             o iH c  in oo
        > o
        > o
                         5-26

-------









































^
-o
OJ
3
C


C
0
u


^
1

LLJ
m

I —


















































tn
•4-1
c
OJ
o
O



















•p

tn
0)

"^
o
re
S-
•P
C
0)
u
c
o
0















o

•P fl)
re -P
•r- >>
-PCO
*
c
're
i-

to






OJ
-P
(A
3f

tn
O)
F—


O>
U
cu
S-


0>







.•c





1


.u
c
c

1
q-
0

>
-M

Q-



HI
(A
C
o
a.
IA
ฃ

0)


-U

&•

t-iซsj



c:
0

.p
(A
ฃ
5
S-
OJ
>
OJ
reoe
•p
c:
01
o
c
o
CJ

•P
re
n_

*S
o
*>
re


c
01
u
o
CJ
o

s~
tu
-C
a
U)
o








aป
c
2


C-O
3

1—





to

ซ

C
o
E

00

Ol
( —
3

s-
o-
O)
C3






o







re
3

U





r_
re

•P
0)
s_
o
 O
r-l CM CSJ in Pซป 00






S



C •ซ*• CT1 O CM C
CM m 
S-
o
a.
>


















•d

c
01
(A


-a

re
•a
c
re to to
•P re -P a)
tn O) c in
3 re
c: c: o a>
•r- -r- U 5-
0) >*-
O.i- i. . tt) t


o
c
re
1C
a

iH






































•a
a>
S- ฃ- iD
V QJ 4)


S-f- -c: oim o
tn U C rH rH

3E • O. -P r— f— (—
tn 013 S- S~
• CD • *r~ O O O
CM -P ro -P o ซป- **-








































^a,
•o
•^
1
ฃ_
0
U
ฃ






QO
cn
i— t
*
*re










in
c
a>


















in

3
in
a>















(U
in
0











0

•P 0)


4j tO
*
re
s.

to








(A


V)



a>
u
ฃ

iซ-

06






ป
1

u
jj


z
^
tf-
O


'ฃ
3
O-

>.
•P
^

1
(0
>

3
_g


O



O

C
o
z

rHCM





CO
OT

t^







•K
•K
|
H-







*"^
OJ

re


^
n.


ซ
C
0

u


:o
i




en
in
+





ff^

i

01
LO
+
















ai
(A
c
S.
IA

0)

Uป*P-
-P-P
3 V)
m o
QJ Q_

ooo


CrtrH VOrHCM





CO r-- CTป CM CTi
•o^nocnro





r> CM ^h co CM

rH CM ^f iO

A (*-. CO CM CM

rH ^ tO CH








C3 a o o o
oooo








c
re
S-

i— t





I

•P (J
W) -r-
*•"> c c:



^.^
ai
01
a
f


ฃ2


O)
5
C
o


i

•p
o
u

















































O)
in
0
•o

tn
OJ
re

Q.

O)
cu
S-

E
o
(t-

re
re

c:
re



*


5-27

-------
































•a
aj
13
C
C
O
0




*J-
in
LU
ง


























































1
i
u















ฃ

Ul
at
cc




c
0



-f-1
c
at
u
o





c



> in

*3 W
U


aป

*r-
at






u
s_
4-) ฃ
in ui
f> Q)
(O E
C S
"- M-
•a o
aป
M Ul
•ป- 4->
t- r-
S at
in
01
in >
4-> •!-
*3 •.-
m ui
a> o
o= . a-
E
• O •





aป
>
0) 4J
> ปr-
•r- C CO
4-> •!- O



d
a<
4->
ฃ3?
V) 0)
o in
o.
J
Oitn 4->1
1C (O tO|
ai *>> in cr
4-> Ul'r-
J- r—
O -CTT-
OJ 0.0
&-.Q 3
Ul
Hoi




T3
a>

S-
ง.
at
s-

o
rE


4J ai

Ul
•a o
ai s-
u u
3 •!—




ca >
OT- cn
Q. r— tO





ll




to
**ซs. U)
ro 'f

at +J
C J3
ง3
Ul
i— T3
ra c .







CO
•or*.
COrH

Is"
cr at

^ฃ ^


c=
o
a
S-
0







































s*
in cn
Ul C

u in
a a<
S- 4-1
















JZ
Ol

4J
Ul
g,
C


o
1
o ซ
u in
O)
II
Ul X
O) OJ
I— CX
in
t,
* a)
m
ui
01
•a
at TJ
4-> a>
•r- 4-1
-a t-
•r- O
-C CX
SO)
t-
cu 4^ a)
tn o ui
o c cr
•a . o
4-1 ro CJ u>
tn > a a*
OJ •<- U
^r- > 4-
cn t- o at
•r- 3 >
.cr ui >!••—
4-P+A
cr co =i o
o^o-a.
.1 . .
rH CXCM 4J CO ซJ-

S5
cn


cn




\
* ?
CD
CD
rH

?T
to
*






CO
O
rH
X
t
CX




ra
.*ฃ "S
•s1-
rO-O
JQ at
o u
C 3

-ฃ= C
a. -i-




0
coo
Cn rH
iฃfS


0)

tO 3
•^x ui
03 O
r— CX
r- X
a> a>
o t.
E 0
r— CX

to >




„

'ra

01

O1CO

ocn

O Csl rH CM
+j +j !f| !+} +[ 5|
cn ^ป- to to to CD
— * ^* in to cn rH

O CM


i-H ^ ^* co co cn
CM to r- cn CM ^~
rH rH


CM CM Pป* ซ^ cn **•
rH co "et* in co cn
cn co pt! CM co S
rH CM ^- in IO I-*


rH
Sr-. to c\j CM cn
co r-. co co rH
•H 4-1 -H +| 4-1 +
M cn in p- in rH
ft CM rH in tO CD
rH co in r*- co CM
rH







r-- **• rH CO O
CD in rH tf CM r-ป
rH rH CM in




a>

0
in
2
u
'5
X
t-'l-
a> s


i — CO






























































































.
01
CA
3

Ul
a*
ro
CX
0)





42

I

t_
at .
CX

at
at
at
s-
**
i
S-
4-
tn

3
Ul


-X








Ul
•M
c
J






o cn
O CO
1-1 +
p5


3 o,
ra to

cZ
tn
c
ro co cn
4-> cn to


a>
Cฃ.
cn
CO
i

m
men
m to
rH 4.

(—


CO
I

•K
0)
(0

•e
Ul
at
o

a.






t_
0
ง•

1





a>

Ul

jj
Ul
a>






a>
u
0)

<ซ_
a>




s: a) *"•
o ui "a! i:
ฐ •ง -Su, TJ.?
4— O) +J rO S- 4J
o 4-> s- u cn ro
Ul O) o> T3 in
O> f O) S ex C crro
ป— "oio ^'^ J2 OT
s T-CD ฃ o> in cr
rO JI rH c *r-
"O rtJ o ui Ul
a) ~ . at at cr
r— O) CT CD 4-> Ul O

•*•: *9 "a*- o S*
**j e - cr o ex CD
.!2 ^g""*- S'-
I Sisซo"iป:'-S
t- * cr CD o a) a) *r™
Oซ 3 t-coo *" rl'-P'In
4JQ.aซinra^r(ao .
W>rH *4-> O Cป CX fi-

* .CD jr; 4-> e
cn t- CD cn u> ra
^A
0 t-
E 0



-Q
S-
ra
ca


a.

fO
CX
01
cr
i
o
tr-

OI
^r
+j
c
o
1
c

I































.
Ul

•5
•j;
tu
CX


o

Ol
u>
5
CX

Ul
3
O
O)
3
cr
c
•r"






j?
•rj
at
ฃ_
3
Ul
ra
a>
E
O
ra
t-
cr
0)
^
o
(J
CJ

•K
5-28

-------











































^
o>
3
C


C
o
u




?"
m
LU

^




















































V)
c
n
,ฐ
















IS>




01
oc

c
0

1o
t.
 in




c

(0
S-

co



10



-M
to







a)
u
c
O)
i—
^

ฃ s-oi
JD OJ to 1
E r— > C O)
re m i o o u
.c c a* EO.
u o fc. a* tn • -4->
•r- S- 0) T3 C
t! ซ e *-> -S'ra
3 -(-> *i- C fc. S- CL
(A 3 fO -r- (0 O
o E a. re r- a. t- •
Q_ CX'*™ CU O ^*
x a> co E s- M~ o
ai .c a> •*- a
•+J 4— c w •*•> at
C O O O IA O
•r- O Ol C C +•>
+J 01 t- > O
501 o m ac a. >> ป*- 0)0 W 3
ฃ 3 CCM S-
"4- U O S *t- >> ~O
o a> o s- o a>i— •—
C-+J ^ 0) > 0) 0
ui >*ป- JM: c
O> **- ซ ^ -r- ปr- r— ,C
a>a)(U(nre 3 o in rH uj
— 1 -4-ป 1 t-C3 - Q_ Q_ fi- "^s.
re ai a> >> a> o
.-j'S-g iculco^ i ^^ 1


> •(-*
ai aป

CD
s_
U)
O
s-
UJ











s
o












re
_2
ra
to
ง
S+, t
CM re >

^^s; s
00) H-
r— 0 t. U)
U 3  • ' - s-
en s- ai
CO 3 4->
31 =
^ X
4) Q)

ง5-
O
r^ Q.














•a
0)
S-
3
(O
(0

u
•o
0)
c •*->
tฃ> re

X 3
ra u
ra
0
J3
•s
O) CJJ
E 




CO
en
i —


1
r—

in
co
in
rH
H-


(U
N

CT O

•*-^ re


Oi
S 0
i
cC





tt











1 1 1 1 1
I 1 1 1 1
1 1 1 1 1

I


OOOrH
V V



oooo
1 ....
i co co in 10





in in en ^^
i ....
i m m en in





P^ Cn rH CM


1 rH CM *$* CO






tn i— i CM 10 o



^" rH CO CD CO




CD CM ^- CO rH
rHCSI rHCM CO



1 •sj-rHCncO

rH CM ^*- Cn





ocnoom
CM cor*. ซf
r-4


S-
O)
•P >
re a)
Q_ S-










i en r-ป rH r*-
i ....
1 OOrHCM
V


in m en co

i OOOCM
1 V V



m ir> o LT>

! rHCMงcn





CO CO CO CD
1 ....
1 CM in O rH
1 rH CM in 0




CM Cn CD rH
I ....

i rH CM in cn






CM CO CO CO *$•
t-H i-H


^- co rH en *f
m CD o co co
rH



co in **• m •*•
rH r-4 CM CM CO



—ป 1 rH CM *3- CO
i — 1 ....
ฃ 1 OOOO





rHCM -3- GO
O * * * >
OOOO

z\
CJ r— 1 	 1
LO O O r—
CO
in > re c
• •^.c re
XO > •*-> JC
•i- cn ^x. cu •*->











i
i
i




i
i
i




i
i
i






i
i
i





i
i
i






s
—1


O




CO
CM



t
1
l




rH
O
O
0

Is c"i


re U> *r- to -r-
S- CUr- O> S-
3 CD 3 d.4-> CU











C 
S1
Q.
01
.1
-2

4—
0)
•*-*


Q


ง
C
1
o
^J














O CJ
4— C3



S- C
3 O
U
cn
S- C
0)-r-
•o a
C 3
3 (A
in
re as
0)
ra eu
C -r-
ra s_
0
E a.
o re
j_ >
t-
•o c:

•|I
QJ E •
-4J O "O
5t|
TD IO
. a> re
X-N4J w'
^ 1 •




to O
s- re
*t- OJrH
re cn
U)
a> c c
3 O ••—
> ra ~o
ฃ_ 0)
OJ C •!—

S- (J O
aป c a.
> o ra

/— N
re
5-29

-------











(A

C
CU











CO
-p

IA
CU
Cฃ






CU
CO
o
o








c
o

re -P
^> IA

u

c

-p
CO



cu
•p
CO

CO*
^j
CO


CU
CJ
I
cu
•+-
o>
oc







t
re
-P
0>


c
re
re

E
T3
C
re

*re

S
u
re
00

a
>) 1
r-* re >,
re i -P i s-
•PCO 310 -P
re ex o t. c
U E U CU
• re •!- re
CU CM C E C CU
O O *r- .Q ปf- •ป- > CO
•r- t- O •(—
4J cz c , a) 4- cn
*"•" re r-* CU > -C E -P O CU rH

E O fc- *~ S— O O •ป•" >>*r~
"* 3 E "c SI "™ > *tl *Lo CU
E >> o o -r- c_> eu . X E 4J S-
• O . O-C-r- . O O 3 . >O
I-H J3 CM a U-PCO c= co cxซ3- un s-









en to co CD CD
CA COIC3 CO CM CD
•P i Cncor-cn
c:
re
•P
t-
cu

cu
OS
















in

i
t-
0
L)
O



cu
•p- in
r- CU

re co o
ฃ- O CO
t- o
"O tj +J
C0*f- >ป
U E t?
3
•a --a
c cn c
•r-co re



cu

3
CO
i.
X
cu
J_
o
1"
















I_
o


J5
1
S-
0)
>
2

CO
3
O
CU
c:















o

So
c o
o -P
a.
CO 0

o


S-
o cu
^- ซA
C
TJ . 0
a> ca.
•P aป IA
c S cu
CU -r- S-
CO O)
CU CU
J- 4J >
CX 0*i-
C 4J
C -r-
0 Z in
r- CJ O
•P C3 Q.
>o
->,^^
^fc! 5
o */- 3 cr
Z (JQ.UJ

rH X
• 


-P
re
f_
r-ป






CO
co ro co CD
S


CO
^

cu
Q

re
CO





cu



u
o
CJ
S
cu

3
(A
O
0.
X
cu
S-
0
o.
re
>




30

i— 1




re

re
>
r-
-P





cn
CO
S-
0)
>


re
ฃ-



0)
S-
3
(A
O
O.
X
cu
^_
0
I"






cn
rH




re
oo o o
LO CO r-i rH
in i^. oo CD



0000
in rH CM CO
co ^" ^* ro


CD o o in
i-H ซ* CM rH
CM CM CM CM

Z> CD CD CD


(A

1 r— 0
i cn o cn
1 CO CO O
o s-
f -P U
i >>*^:
i O E



J t
(A O
CU 
-------
point (sic) and elementary analysis."   From this  information we conclude that
the  consistent  positive responses in Salmonella  can  be attributed to DCM.
Barber et al.  (1980) conducted their tests in a chemically inert,  closed incu-
bation system and  analyzed  the concentrations of DCM in the vapor-phase head
space and  in  the  aqueous phase of a test plate by gas-liquid chromatography
(Barber et al., 1981).   Based  on  this information, the  mutagenic  responses at
the  highest dose  (i.e.,  115 umoles/plate) for TA1535,  TA98,  and  TA100 were
0.0006, 0.006, and 0.03 revertants per pmol, respectively, indicating DCM is a
weak mutagen for Salmonella under the conditions of the test.
     The results  discussed  above  clearly  show that  DCM is mutagenic  in
Salmonella. However,  questions have  been raised  about  the  applicability of
these results to predicting mutagenicity in other species, especially mammals.
DCM  is metabolized,  apparently via mutagenic intermediates, to CO and COp in
both rodents  and humans  (see Chapter 4).  CO  is produced  by oxidative dechlo-
rination of DCM by  the microsomal  P*™  mixed  function oxidase system.  Formyl
chloride is believed to be an intermediate in this pathway.   A second cytosolic
glutathione transferase system dehalogenates DCM to produce formaldehyde, which
is further oxidized to  CO^.   This pathway  is  thought  to proceed  via an S-
chloromethyl  glutathione intermediate (Ahmed and Anders, 1978;  Kubic and Anders,
1975).   Formyl  chloride and S-chloromethyl glutathione  are  highly reactive
alkylating agents.   Salmonella also  metabolizes  DCM to COp and CO apparently
by reaction pathways similar to those occurring in mammals (Green, 1980,  1981).
Because of the reactivity of formaldehyde,  formyl  chloride,  and S-chloromethyl
glutathione,  and the proximity of bacterial  DNA to bacterial cytoplasmic enzymes,
it has been hypothesized that  these chemical  substances are more  effective as
mutagens when they are formed by bacterial  metabolism than when they are formed
outside the bacterial cell  by rat liver fractions (Green, 1980,  1981).  The
basis for this hypothesis is that rat liver fractions used for metabolic acti-
vation have little effect on increasing the mutagenicity of methylene chloride
in the Ames test.   The  implication is  made that  as organismic complexity is
increased,  there is less likelihood that DCM will  cause mutations. It is argued
that compartmentalization of DNA into the nucleus protects the genetic material
from exposure to the mutagenic metabolites of DCM (i.e., they would react with
other cellular constituents  first) and  thus there is little or no mutagenic
risk.  The positive results using eukaryotes, discussed in the following sec-
tions,  argue against this hypothesis.
                                     5-31

-------
5.3.2.1.2  Yeast.   Call en  et al.  (1980) studied  the  ability  of  DCM  obtained'
from Fisher Scientific Company (purity not reported) and six other halogenatJSS
hydrocarbons  to  cause gene  conversion,  mitotic recombination,  and  reverse-
mutations in  Saccharomyces cerevisiae  (Table  5-5).  Strain  D7  log  phase celjl
were incubated for 1 hour in culture medium containing 0, 104, 157, and 209 Ifflf
DCM.  The percent survival for these doses were 100, 77, 42, and <0.1, respec-
tively.  Due  to  the toxicity of the  compound,  the genetic end  points  were  not
measured at the  highest  dose.   The response for the other doses (0, 104,  and
157 mM)  expressed per  10   survivors  were:  gene conversion  at  the  trp-5 locus
(18, 28, and 107); mitotic recombination for ade-2 (310, 190,  and 4,490);  total
genetic alterations for ade-2 (3,300, 3,900,  and 14,000); and reverse mutations
for ilv-1 (2.7,  4.4, and 5.8).  A greater than twofold dose-related increase
over negative controls was observed for each endpoint measured.  The magnitude
of recombinogenic response at the ade locus may have been overestimated in this
study because the treatment regime used for estimating the recombinants overlaps
that used for estimating  the number of trp-5   convertants.   No exogenously
applied metabolic activation was  used in these experiments, which indicates
that yeast metabolizes DCM intracellularly to a mutagenic intermediate(s)  thaf
reaches nuclear  DNA.   In another  genetic study employing yeast,  Simmon et  al.
(1977) reported  that DCM  (source  and purity  not  given, but stated to be the
highest available purity)  did  not increase mitotic recombination in  strain  D3
                                               Q
of Saccharomyces cerevisiae  when  cells (1 x  10 )  in  suspension  culture .were
exposed for 4 hours at 30ฐC (Table 5-5).   The doses used and the actual  expeff-
mental values  obtained for mitotic recombination  were  not reported.   The dis-
crepancies between  the work  by Call en et al.  (1980) and Simmon et al. (1977)
may be due to a number of factors including  the  different strains used (D3
versus D7), exposure time  differences  (4 hours versus  1 hour), or  differences
in the incubation temperature (30ฐC versus 37ฐC).   Callen et al.  (1980) repfrtecj
that an  increase in the  treatment time of D7 cells with DCM from 1 hour to 4
hours significantly  reduced  the  level  of genetic activity.   Other variables,
such as  a lower  level  of P.5Q  enzymes  in strain D3, could conceivably account
for the  discrepancy in the results.   At this time, DCM is considered to be a
positive mutagen in yeast.
5.3.2.1.3  Drosophila.  Two  reports  are available concerning  the  ability  of
DCM to induce sex-linked recessive lethal  mutations in Drosophila melanogaster
(Table 5-6).    Abrahamson and Valencia  (1980)  reported  negative results, while
                                     5-32

-------
         r- U1
        +•> c
         o> o
       •  fO •*->
       . 4J,—
       .  o 
-------
I
                  ui a* a.
         . a o c
         J   D.C
         >+ป H e '
         > c p-r- :
; aj o
I t-'i-
               ' t- ฃ- in o
                tU ZJr— 0)
                CL U O *r-j
        C CX-t-> O)  O 4J -f-
     en ro co o t,
         JT3-O->-3
         j d) 01 c
     U_ *+- O CO
      ซ.ซ
                                    •r- C   ซJ
                                           ป
                                          oo *—/   csj^-'
                               5-34

-------
             01
             >

             Ol
  0)
  tfl
  C
  O

  8-   ซ,2?
      2p- c
      o o

  OJ   4J &.
                  Q_QJ-r-
                  tO  S-r—
        cn Q o> SI *i
        o t— > uj ^
        ฃ^ flj *i—  *i
        (0  O ID


                C\j C3 00
c -
(U I
CJJ  O O O
                      I  I  I
                      sss
            X U 4^

            in $- f—
  2
  0)
          5-35

-------
a positive response was reported by Gocke et al. (1981). Abrahamson and Valencia
(1980) conducted their  sex-linked recessive lethal tests using two routes of
administration:  adult  feeding  and injection.   Due to  the  low solubility of
DCM in aqueous  solutions,  high concentrations of the test substance were not
used in these experiments, which may account for the negative response observed.
In the feeding study, male Canton S flies were placed in culture vials contain-
ing glass microfiber paper soaked with a saturated solution of 1.9% DCM in a
sugar solution  (224  mM  DCM)  for 3  days.  The feeding solution was added twice
daily to compensate  for evaporation of the  compound.  At this dose, there was
no evidence  of  toxicity.   After mating, chromosomes from the 14,682 offspring
of treated parents  and  chromosomes from the 12,450 offspring  of concurrent
control parents were assessed for  recessive lethal mutations.  No evidence of
mutagenicity was observed.   DCM gave  a level  of 0.204% lethal  mutations com-
pared to 0.215% for  controls.   However, because of the  volatility and  insolu-
bility of DCM,  the  actual  dosing to the animals may have been much less than
expected.
     In the  injection study, 0.3 |jl of an  isotonic solution containing 0.2%
DCM was administered to male flies.  This exposure level resulted in 30% post-
injection mortality.  However,  the post-injection mortality observed for the
controls was  not reported.   Because the mortality  observed  in studies  such as
this is due not only to the test chemical administered, but also to the damage
caused by injection, concurrent negative controls upon which to base conclusions
concerning toxicity  of  the test chemical are necessary.-  After mating, 8,262
chromosomes  from the offspring  of treated parents and 8,723 chromosomes from ,
the offspring of control parents were assessed for recessive lethal mutations.
No evidence  of mutagenicity was  observed  by  this route of administration.
Flies injected with 0.2% DCM had 0.157% lethals compared to 0.206% for controls.
     Gocke et  al.  (1981)  also tested DCM  (Merck,  Darmstadt, FRG, purity not
given) for  its ability to  induce  sex-linked  recessive lethal  mutations in
Drosophila.   Two solutions,  125 mM and 625 mM  in  2% DMSO and 5% saccharose,
were fed to  wild-type Berlin K male flies  for  an  unreported period of time.
The higher dose (625 mM)  is reportedly close to the LDcn-  These males were
then mated to  Base  females.  Three broods  were scored  (i.e., offspring from
virgin females  mated to treated males on days 1 through 3,  4 through 6, and 7
through 10 after exposure).  There were  significantly more  lethals  (24 out of
                                     5-36

-------
 4,845 chromosomes scored) for brood 1 (0.50% lethal mutations) compared to the
 negative controls, (19 lethals out of 7,130 chromosomes, 0.27% lethal mutations),
 P <0.05.   Elevated,  but  not  statistically significant increases in  lethal
 mutations were  noted in broods 2  (0.16 percent compared to 0.14% lethals) and
 3 (0.47 percent compared  to 0.39% lethals) of the treated flies compared to
 the controls.   As noted in Table 5-6 the incidence of lethals is dose-related.
 The incidence of lethals for the high dose in brood 2 is elevated nearly three-
 fold over th;e corresponding negative control value (0.41 versus 0.14  lethals,
. respectively),  but this may not be a significant increase because of the small
 sample size (735 vials).   This  test indicates DCM is  mutagenic to  sperm in
 Drosophila (a multicellular eukaryote).  The  discrepancies  in results between
 Abrahamson and  Valencia (1980)  and  Gocke et al. (1981) may be due to stock-
 specific differences (Canton  S. versus  Berlin K)  in  the metabolic activation
 of DCM or more  likely to the larger  doses of  DCM employed by  Gocke et al.  (1981).
 5.3.2.1.4  Nematodes.   In another sex-linked recessive lethal test,  Samoiloff
 et al.  (1980) tested DCM  for  its  ability to mutate the nematode Panagrellus
 redivivus  (Table 5-6).   Individual females homozygous  for the X-linked mutation
 b7 (coiled phenotype  in liquid medium) were grown  for  120 hours  in the presence
 of several concentrations of DCM ranging from 10~3M to 10~8M.  They  were then
 washed  and mated to S-15 males who carry an X-chromosome crossover suppressor
 extending  at least 15  recombination  units to  either side of  b7.  One  hundred
 female  progeny  were collected and mated  to wild-type (C-15)  males and their
 progeny  scored  for the presence of the b7 phenotype.   The absence of  b7 male
 progeny  indicates lethality of the X-chromosome marked with  b7 derived from  a
 female grown on  DCM.  Three  replicate experiments were  performed.  A non-dose-
 related  increase in the level of  lethals  was observed  in the progeny  of DCM-
 treated worms compared  to  the  negative controls.  For worms treated with 10~8
   "6-4                                            c.                   '
 10  , and  10  M  DCM, the corresponding lethal mutations/10  loci were  6.0, 10.1,
 and  9.8, respectively,  compared to an estimated spontaneous mutation frequency
 of 2.2 x 10   mutations/locus.  Some of the positive controls tested concurrent-
 ly,  such as proflavine, yielded a positive response  (12.5,  10.0, and 28.6
 lethals/10  loci  at 10~8,  10~6, and 10~4M, respectively).  But others, such as
 aflatoxin  B  and  ethyl  methanesulfonate (EMS), did  not cause  an increase in
 lethal mutations.   The  investigators  suggest that DCM  is mutagenic  in nema-
 todes, but firm  conclusions  cannot be made because  the  assay  is  not  validated
                                     5-37

-------
and, more importantly, because of the negative responses obtained with some of
the positive controls.
5.3.2.1.5  Mammalian cells in culture.  Jongen  et  al.  (1981) tested DCM for
its mutagenic potential  in several mammalian  cell culture tests.  Testing for
the induction of forward mutations at the HGPRT locus is described here (Table
5-7).   Testing  for the ability of DCM  to  cause sister chromatid exchanges
(SCE), unscheduled  DNA synthesis  (UDS), and inhibition  of DMA synthesis (IDS)
will be  discussed  later  in the section on other  indicators of DNA damage.
     In their testing of the ability of DCM to cause forward mutations, Jongen
et al. (1981)  incubated  log phase CHO  and V79  cells with 1, 2, 3, 4,  and 5%
DCM or 1, 2, 3, and 4% DCM, respectively, at 37ฐC for 1 hour in a closed glass
container without  exogenous  S9  mix.   DCM was obtained from Merck (analytical
grade).  The cells  were  exposed to gaseous DCM and  then DCM in solution for
15-minute intervals  each by alternately tilting the plates then placing them
horizontally.  After  growth  to  allow for an  8-day  (CHO cells) or 6-day (V79
cells) expression  period,  mutant  cells  were selected in thioguaninecontaining
medium.  DCM failed to increase the mutation  frequency  of either cell  line  at
any dose compared  to controls.  However, DCM  was not very cytotoxic to either
cell line.   At  the highest dose,  survival  decreased  only 20 percent.  It would
be appropriate to repeat the experiment using higher doses of DCM.  EMS yielded
a positive, dose-dependent increase in mutation induction in V79 cells, but it
was not tested in CHO  cells.
     Based  on  the  positive responses in bacteria, yeast, and Drosophila, and
the suggestive  positive  response  in the nematode,  DCM  is judged  to be  capable
of  causing  gene mutations.   Metabolic activation to  highly  reactive mutagenic
metabolites  apparently accounts  for  this  response,  and although these are
thought  to  be  unstable,  they seem to be capable of  interacting  with  genetic
material of both prokaryotes and  eukaryotes.
5.3.2.2  Chromosomal Aberrations—Three studies on the  ability of DCM  to cause
chromosomal aberrations  were evaluated.  Burek  et al. (1984) subjected 4 groups
of  10  Sprague-Dawley albino rats  (Spartan  substrain, SPF-derived,  5 males  and
5 females)  to 0, 500,  1,500, or 3,500 ppm  (0, 1735,  5205, 12,145 mg/m3) DCM by
inhalation  6 hr/day,  5  days/week for  6 months.   The  animals were  then
sacrificed,  bone marrow  cells  were  collected,  chromosome preparations were
made,  and  slides were coded and  analyzed.  Two hundred metaphases per animal
were  scored and aberrations were tabulated  (See  Table 5-8).   No increase
                                      5-38

-------
    ^ O
    0X3
    CM rtJ
  C 
  Q.I— in
  in 3 oj-o
  o> i/> in a)
  t- o> o 4->

  O)       OJ
  > >,S-^i
 ^- "O  . O
 3 O3 S- -f-
 CT-i- 3 U
 LLIIC W)-^
er> co cxi a\ i~\ in

rH i-H i-i O Csj CSJ
O i—I CM OO ซ
— V) Ot r—
    5-39

-------
In the total  frequency of abnormal cells or in the frequency of any specific
type of aberration was  noted  in the treated animals compared to the controls.
There were 1.1 ฑ 1.3,  0.6 ฑ 0.7, 0.8 ฑ 1.2, and 1.1 ฑ 0.9% cells with aberra-
tions in animals treated with 0,  500, 1,500,  and 3,500 ppm (0, 1735, 5205,
12,145 mg/m ) DCM,  respectively.
     Thilagar and Kumaroo (1983) treated CHO cells grown  in either plastic  or
glass culture flasks with 0, 2, 5, 10 and in one experiment 15 pi/ml (i.e., 0,
31, 78, 156, and 234 mM) DCM for 2 hours with or 12 hours without 59 mix derived
from Aroclor-induced rat livers.   DCM was obtained from Fisher Scientific (cer-
tified A.C.S., lot no. 713580).  After the exposure period, the cells were washed,
refed, and  allowed  to grow before being arrested at metaphase  with colcemid
and harvested for chromosome preparation.  Slides were coded and read "blind;"
100 cells were  scored for each dose level  (50  cells/duplicate flask). DCM  in-
duced a dose-related increase  in chromosome aberrations (see Table 5-8) ranging
from 0.02 aberration/cell  in  the negative controls to 1.44 aberrations/ cell
at 15  ul/ml(234 mM).   The response was  not dependent on  the presence of the
exogenous metabolic activation system.
     Gocke et al. (1981) assessed the ability of  DCM (Merck, Darmstadt; purity
not given) to cause micronuclei in polychromatic  erythrocytes (PCE).  Two male
and two  female  NMRI mice were used for  each of  three dose  levels  (425, 850,
and 1,700 mg/kg per intraperitoneal injection).   The highest dose approximated
the LD,-n  for mice.   Intraperitoneal injections of each dose v/ere  given at 0
       ou
and 24 hours, the animals were sacrificed at 30 hours, bone marrow smears were
made,  and 1,000 PCEs per animal were scored for  the presence of micronuclei.
An  increase  in  PCEs with micronuclei was observed at the two  highest doses,
but the  response was not dose-related and was not double the  control value.
Thus, the results are  considered suggestive of a  positive response but are  not
conclusive.   (There  were 0.19% micronuclei  in  the untreated controls  compared
to 0.35% micronuclei  in the animals receiving two injections of 850 mg/kg,  and
0.28% micronuclei at  the highest dose).
     Based  on the  positive response reported  by  Thilagar and Kumaroo (1983),
DCM  is tentatively judged to  be  capable of causing chromosomal aberrations.
The  rn vivo negative  responses reported by  Dow Chemical Company  (1980) and
Gocke  et al. (1981) are not  readily  comparable with the j_n vitro results.
5.3.2.3  Other  Indicators of  DNA Damage
5.3.2.3.1   Sister chromatid exchange  (SCE).  Two  papers  have been  published on
the  ability of DCM to  induce SCEs (Table 5-9).   Jongen et al.  (1981) tested
                                      5-40

-------






















to
z
o
*— t
u.
f
LU
00
> Ul
in c re 01
^ *•* o s • s-
ui m .a 7
" — r— Ul O O)
ro r~ o i — i — >
E tti -P i- T-
•r- U 0) CZ -p
a • oi u o) co
re o ui tn ai 01
0 r- 0 5 .0 01
in • CM re Q o z
at E t- OP
Ul -I- S- >
• o • c . re ro •
rH "O CM re CO f= .C ^
0
O O -H O

rH
0

CM
^|
d
0 O O +1
CM
O





CO O O O




CM CM CM CM
^ *^I ฐ? "^
O O O O
•M +1 -H +1
CM CM rH CM
O O O O
01 rH r*ป oo

0 CO CD O
+1 +1 -rl' -H
01 in in r^-
O O O O

o o o o
o o o
in in m
r-i CO


c
o
fO
'ซ
c
H- 4




i— • >\ O
E •— (_
*2E
•a i ai
C Ol C
ro a o
O1.Q
01 re \
r- t 4J
re a. re
S 00 t
'io
o re
Q.O
d O rH





Ul
•P
C
01
E
O
o
^ง
Ul •!-
ป— re
01 ฃ-
0 S-
ai
*c



"0!
o
O Ul
f- O
0) *r~
"E re
3 &.
•z. '-
Ol

*
Ol
Dl
C
re
u
LU



-o
o
,c
u
o
tn
tn
Ol
00 ••-
|
CJ

<5
o
ce.

Ol
Ul
o
a






!„}
Ul
Ul
01
1—

Reference

ai
Ul Ul
C P 1
o c ai
a. 01 s-
tn E
01 -r- S.
s. s- a
01 r-
O) ฃ^*r~
> X E
•p~ O) , tn
CM ID IO 00 10
CM CO tD












CM IO ซ*• ID U5
o o co in en

o o o o o



O CM 00 O CM







O O "!^ tt" C3
rH CO CO





CM •* 00 CM CM
rH CM

ซJ- CO I--
O 00 in VO
O OS f^ \O Z
rH

I
3.O CM in O Dl
v^ rH E O.
LU
g
O

o
u
•a
01
'n 'a!
o u
o
a ^
i— CO
r- TJ 00
ฃ C 01
1— re rH






















































5
|
O)
"cu
u
Ol
re
cl
II
8





in
c
o
CJ
in
OJ
Ol
c
ro
f—

L^




.1
a:



tn
O

s_
c,
0)
o
o


0)
E
tn
o
E
g
-C
CJ
tn
re
oa -a

re
5

01
ul E
O CL.
a a.




o s-
Ol Ul
•P O
2 a.
O X
Qฃ LU





01
2
Ul
Ul
ฃ
^x
c
2
•P
CO

Reference
> -r- O) O O
•*- T3 in S- r—
•P C C S-
•i- -^ O (0 C
in Q. E Q*
O ui tn  a) A
O ฃ- C
a> -c o at
4 ซ S -ฐ 8
t!'ฐ^2-c
0) •* (0 >ป
01 aป 01 ซ m
01 w a) in E
3 C C O
co o a 
-------
the ability of  0.5,  1.0,  2.0, 3.0, and 4% DCM (i.e., 58, 118, 235, 353, and
471 mM)  to  induce SCEs in V79 cells.   Log phase cells were incubated at 37ฐC
for 1  hour  in a closed glass  container.  The cells were exposed to DCM in the
gaseous  phase and in the  medium by tilting the  plates  for 15 minutes,  then
placing  them horizontally.  The experiment was conducted seven times and each
yielded  a dose-related increase  in SCEs/cell,  which  approached but  did not
exceed a twofold increase above the control level.  An analysis of variance of
effects  of different doses within experiments showed the increases in frequency
of SCEs  to  be  statistically significant (p <0.001).   Increasing the exposure
time to  2 hours or 4 hours  or using S9 from rat  liver did not alter the shape
of the  dose-response curve, which plateaued at  1% DCM.  The authors suggest
that this phenomenon is due to a saturation of the metabolic activation system
of V79 cells.
     Thilagar and Kumaroo (1983)  exposed CHO cells to 0, 2, 5, 10, and  in one
experiment 15 ul  DCM/ml of medium (0, 31, 78, 156, and 234 mM DCM) for 2 hours
with and 24 hours without metabolic activation.  The cells were grown for 24
hours  in BrdUrd followed  by a mitotic shake off, fixing,  and staining by a
fluorescence-pius-Giemsa  technique,  and then  the coded slides were scored
"blind." Slight dose-related  elevations  in SCE  values  were noted  (see  Table
5-9), but they  never exceeded a 50% increase at  the highest dose.   The authors
judged  their test to be negative,  but  the  concentrations were  lower  than  used
by Jongen et al.  (1981).
     McCarroll  et al.  (1983)  reported in an abstract  that consistent and dose-
related  increases  were observed in SCEs in CHO cells  following 24-hour exposures
to 1,  3.6,  5.4, and  7.0%  atmospheres  of  DCM.   A 7%  atmosphere  was  required  to
elicit  a statistically significant increase.   Based  on the reports of Jongen
et al.  (1981),  Thilagar and Kumaroo (1983), and McCarroll et al. (1983), DCM
is capable  of causing  DNA damage, that results  in SCEs.
5.3.2.3.2   DNA  repair assays.   In their study of the genotoxic potential  of
DCM, Jongen et al.  (1981)  also measured UDS and IDS  in V79 cells and primary
human  fibroblasts (AH cells).  These experiments were  conducted  by exposing
105  cells  attached to glass  covers!ips  (UDS assay)  or to  glass petri  plates
(IDS assay) to 0.5,  1.0, 2.0,  3.0,  and 5.0% DCM (58, 118,  235,  353, and 471
mM,  respectively) without metabolic activation.   UDS experiments  were  done in
duplicate,  and at least  25 nuclei  of  non-S phase cells were  scored for the
number of  silver  grains/nucleus at each  dose  level.   DCM had no detectable
effect on  UDS  in  either  cell  line.   In the IDS assays, the relative rate of
                                     5-42

-------










































UJ
CD
:
LU
a

r-
SI

Oฃ
U

ce
LU
i—
CO
I— 1
CO


o
u.

1 —
en
I 	



en
i

UJ
ca
j—




















































to
•P
c
OJ



































to

r—
3

QJ

















0}
to
CD



E
at
-p
to

CO
•p
i/j
 O
O 0)
to u
J2 C
0 CD
-o
0) '1-
to • u
C C . C
O) O /""N't—1

• CO
s_
3
O •
J= O)
LO
rH C
O
0) CX
CL.C
to-^ a>
HI Q (ft
t, J= (0
I if) 0)
QJ ฃ-
UJ P U
O 0 C
•a c T-

E tn^%4- to P
•r- OJ rH 0 -P 0
P S-
0) 0)
s_ >
3 T-

0 -r-
a. to
X O
UJ 0_


o to c
001*0
. o.^-'rj
0 >) 'r-
v -P CD ~a
i
Q- 
o e

UJ ฃ
CJ 0)
co a.
X
UJ rH












E
Q


to


Of 0)
CJ S-
3
en -p
> 's
\ u
LU
CJ C










-
"ฃ
+J
;






0)
o
CD












3J
in

CO

-p
to
0)
I—


O)
(J
c:
o>
J-
OJ
4-
O)

















































































i
'a.
E
CJ
CD








0
-c

•a
at
s_

•P


0


s_
to
O)
fO


JZ
r—
to t-
O) (0
10 i— >, Ul
tO *i- J2 0) >
0) E U)
t- •!- -P O ฃ
U to UJ T3 E
c at
•r- -a -P -P rH
a> to r-
P ^ f" (U ซ^
C r- 4-> JT
lO O> -r- OJ -
U -r- J T- .

•r- lO C QJ ปi—

OJ .C . -P 0)"^
•i- ' a* • to j= s_
to • m:>: -r- X 0>
•rr -i .W -P
+J S- ' C /— s fO
O - . 0) ' O rH Ol
C ."ป 'Q. CJ 00 S-
, X c en O)
+J OJ •.- rH
3 •-'•••' **^s 10
J2 s- +J at

f— p m -P
m o • to
c to -P -P at
•r- • CJ CJ r— CJ Ot ซ
O) to a> to 3 i- *-^
rocjjz o a) OI-P e.
E; co H- Q.Qฃ OJ
(/> C tO CO
. c .  CO
co co CM ro



CO CD CO CO
CM co m co
O rH CM CM
rH rH rH rH





O CM in O
rH

















UJ


a>
u



0
o
-a s-
C fO CO
(0 E CO
3 en
iฃ rH
5-43

-------
DNA synthesis was  determined  radioisotopically  immediately after DCM exposure
and 0.5, 1.5, and  3.5  hours later.  The  average of duplicate  samples revealed
that DCM inhibited DNA synthesis in V79 and AH cells at all  dose levels com-
pared to controls but that synthesis recovered with time after exposure in all
cases.  This  is  unlike  the persistent  inhibition of DNA  synthesis by the posi-
tive control 4-nitroquinoline-l-oxide.   The authors  conclude  that DCM was not
inducing genetic damage in cells but was inhibiting DNA synthesis by an effect
on cell metabolism.
     Perocco and Prodi (1981) also performed a UDS assay using DCM.   They col-
lected blood samples from healthy individuals for their studies, separated the
lymphocytes, and cultured 5  x 10  cells in 0.2-ml medium for 4 hours at 37ฐC
in the presence or absence of DCM (Carlo Erban, Milan, Italy or MerckSchuchardt,
Darmstadt,  FRG, 97 to  99 percent pure).  The tests were conducted both in the
presence and in the absence of PCB-induced rat liver S9 mix.   A comparison was
made between treated  and  untreated cells for  scheduled DNA synthesis (i.e.,
DNA replication) and  UDS.   No difference was  noted  between  the groups with
                                                         3
respect to scheduled  DNA  synthesis measured as  dpm  of  [  H]  deoxythymidylic
acid (TdR) after 4 hours  of  culture  (2,661 ฑ  57 dpm in untreated cells com-
pared to 2,356  ฑ  111 dpm in cells treated with 5 ul/ml  [78 mM] DCM).   Subse-
quently, 2.5, 5, and  10 ul/ml (39, 78,  and  156 mM) DCM was  added to cells
cultured in 10 mM hydroxyurea to suppress scheduled DNA synthesis.   The amount
of unscheduled  DNA synthesis  was estimated by  measuring dpm from incorporated
[3H]TdR 4  hr later.   At 10 ul/ml DCM, 532 ฑ 31 and 537 ฑ 39 dpm were counted
without and  with exogenous  metabolic  activation, respectively.  Both values
were lower than corresponding negative controls  of 715 ฑ 24 and 612 ฑ 26 dpm,
respectively.   No  positive controls were run to  ensure that  the system was
working properly,  although testing of chloromethyl  methyl ether (CMME) with
activation resulted in a doubling of dpms  over the corresponding negative
control values  (1,320  ฑ 57  at 5 ul/ml CMME versus 612 ฑ 26 untreated).  The
authors calculated  an  effective  DNA repair value (r) for each chemical  based
on the  control  and experimental  values with and without metabolic activation.
DCM was evaluated  by  the  authors as  negative  in the test, but they did not
state  their  criteria  for  classifying a  chemical  as  positive.  None of the
experimental values from cells treated with DCM had higher dpm values than the
controls.
     Based on these experiments  there is no evidence that DCM specifically in-
hibits DNA synthesis or causes UDS.
                                     5-44

-------
5.3.2.4   Summary  and  Conclusions.   Dichloromethane has  been  tested for its
ability to cause gene mutations (in Salmonella, yeast, Drosophila, Panagrellus,
and  cultured  mammalian cells), chromosomal aberrations  (in  rats,  mice, and
cultured mammalian cells), and other indicators of DNA damage in cultured cells
(sister chromatid  exchange,  unscheduled DNA synthesis, and inhibition of DNA
synthesis).
     Commercially available samples of DCM gave.positive results in Salmonella.
yeast, and Drosophila.  The responses were weak under treatment conditions used
and were  obtained without  the  addition  of  metabolic activation  systems  (e.g.,
S9 mix).   The data suggest that DCM may be metabolized j[n vivo  to  a^mutagenic
metabolite(s).  Some negative results have been reported for gene mutation tests
in fungi (Saccharomyces) and mammalian cells in culture, but these may represent
false negative results because of the treatment conditions used.  DCM has also
been reported  to  induce chromosomal  aberrations in cultured mammalian  cells
but not in bone  marrow cells from animals exposed jji vivo,  perhaps because a
sufficient dose of DCM did not reach the bone marrow.  DCM causes a weak in-
crease in  SCEs but has not been shown to cause UDS or inhibit DNA synthesis.
     Mutagenicity tests of DCM have given positive responses in four different
organisms  based on the weight of available evidence.  DCM is judged to be a
mutagen with  the  potential of  inducing  gene mutations in exposed human  cells.
A positive response in cultured mammalian cells indicates that DCM also causes
chromosomal aberrations, but additional  testing in another j_n vivo or in vitro
chromosomal aberration assay is needed to confirm the available data.   If such
tests are  conducted,  care  should  be taken to ensure  that the test cells are
exposed to sufficiently high doses of DCM, otherwise  false negative responses
may be obtained.

5.3.3  Evaluation of the Carcinogenicity of DCM
     The purpose  of this section is to provide an evaluation of the likelihood
that DCM  is  a human carcinogen and,  on the  assumption that it  is  a  human
carcinogen, to provide a basis for estimating its public health impact,  includ-
ing a potency evaluation in relation to other carcinogens.  The evaluation of
carcinogenicity depends heavily on animal  bioassays and epidemiologic evidence.
However, other factors,  including  mutagenicity, metabolism (particularly in
relation to interaction with DNA),  and pharmacokinetic behavior, are important
to the qualitative and quantitative assessment of carcinogenicity.   The avail-
able information  on  these subjects  is  reviewed in other sections  of  this
  ;                                   5-45

-------
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 section dealing with all of the relevant
aspects  of  the carcinogenicity  of DCM.    Further,  the  National  Toxicology
Program  (NTP)  rat and  mouse gavage bioassay draft technical report (1982) on
DCM was  cancelled because of data discrepancies at their contract laboratory
(memo from John A.  Moore dated July 25, 1983).
5.3.3.1  Animal Studies
5.3.3.1.1  Dow Chemical Company  (1980) inhalation study in  rats.   A  total  of
1,032 male and female Sprague-Dawley rats (129/sex for each exposure concentra-
tion) were exposed by  inhalation to DCM at  0,  500, 1,500, or 3,500 ppm (0,
                            o
1,735, 5,205, or 12,145 mg/m ) for 6 hr/day, 5 days/week (excluding holidays),
in a  2-year  toxicity and oncogenicity study.  Approximately 95 rats of each
sex  for each exposure concentration were  part  of  the chronic toxicity and
oncogenicity portion of the study.  This  number also included those animals
that died spontaneously, were killed moribund during the study, or were killed
at the  end  of the  2-year exposure.  The remaining animals were sacrificed as
part  of  the  cytogenetic studies  or for one of the interim kills at either 6,
12, 15,  or  18 mo of exposure.  The rats were received at 6 to 7 weeks of age
(males weighed 220 to 250 g; females weighed 170 to 200 g) from Spartan Research
Animals, Inc., Haslett, Michigan, and were individually marked for identifica-
tion  with metal  ear  tags.  All  rats were  maintained  on a  12-hour  light/dark
cycle.  They were observed daily, including weekends and holidays, for general
health status and signs of possible toxicity.
     Dichloromethane representative of technical  grade material was obtained
from  Dow Chemical Company,  Plaquemine,  Louisiana, and was  used throughout  the
exposure.  Fourteen  different  samples  of DCM were analyzed during  the  2 years
of animal exposure;  each sample showed 99% pure DCM, with a few trace chemical
contaminants that varied  slightly from sample to sample,  as  shown in Table
5-10.   The  concentration  of DCM vapor in chambers was considered well within
the range of expected variability.  Hematologic determinations, serum clinical
chemistry,  urinalysis,  bone marrow collection,  and blood carboxyhemoglobin
(COHb)  determination were done in animals sacrificed at 6, 12, 15, and 18 mo
(interim kills).  Plasma estradiol determination was done at the 12- and 18-mo
interim  kills.   This included samples from  six  controls/sex  and  four high-
exposure animals (3,500 ppm)/sex from the 12-mo kill, which were pooled
                                   5-46

-------




















UJ
z
1—
UJ
o
a:
o
1— 1
a

o

CO
t/>


t— 1
^

1
3
1
in

LLJ
cn



CM


r-
CO
CO

S
CO

CO
CM
CO
CO
in


rH
CM
CO

1
1




00






o
in
in




o
in
in

CO
o





CO
CO
CM

t-H
in





CD
m


oT
c
0)
>^

+J
o>
a
t-
o
!c
u
T3
1
CM^
r-T
in
c e
m CL
CM CO

CM
in

ง' i-H
CM

SCO
CM

CM CM
CO
CM
CO CM
CM


CD CM
CO
CM

aป CM
cn
CO



in in
CO
CO





ซ* i-H
r**. r— t
CO




m oo
CO
CO

r-ป rH
CO rH





r- in
•=*•
CM

in cn
CO





to co
CO
CO






s
a. -
a. ซ
oT 'ฃ
1 S
OJ U
O r—
u 1*
>> -^
O UJ
rH rH




rH rH


rH rH


rH rH


rH rH


rH rH



rH rH




I 1
1 1






1 1





i i
I i


1 |






I |
1 i


J j






rH CO
V CM






& i.
a. a.
O) o>
•o -a
1 1
U
•5, !•
c •>->
.1- a>
=> s:
CM
rH



O
CM

S


CO
rH

rH


m
t-H


rH




1
I






1
1





(
1



1





1
1


1






CM
V



1


O)
T3
i tetrachlor
o '
•e
ซ
o












































^
0)
N
>>
S
ซ
 o)
•a TJ
g1" -u •!->
0 0
O 1=1 1
ซ Z 1
5-47

-------
together  (two  animals/sample) to  give  three control samples  and  two  high
exposure  (3,500  ppm) samples/sex.   Ten  individual  samples/sex (not pooled)
from the  high  exposure  and control groups were also  sent from  the  18-mo kill.
     All  animals that either  died  spontaneously, were killed in moribund con-
dition, or were  killed  at the interim or terminal  kills  were subjected to
complete  gross and microscopic pathological  examinations by a veterinary path-
ologist.   Liver  samples for  possible electron microscopic evaluation were
collected.
     In females  exposed to  3,500 ppm, there was a  statistically significant
increase  of  mortality  from  the 18th through the 24th mo that may be exposure
related.   The  remaining treated groups  in males  or females did not differ
significantly  from  the  controls  (Table 5-11).   There was no exposure-related
difference  in  body  weights of either male  or female rats  exposed to  500,
1,500, and 3,500 ppm DCM.
     Although  some hematologic values were increased and others were decreased,
the mean  values were within the normal range of biological variability.  Serum
glutamic  pyruvic transaminase (SGPT), blood urea  nitrogen  (BUN),  and  serum
alkaline  phosphatase (AP)  values were in the normal range.  It is noted that
the females  had  significantly increased  (P <0.025)  plasma  estradiol  levels at
18 mo,  which may be related  to the higher incidence of mammary tumors in  the
exposed (3,500 ppm)  group.  Urinalysis findings were in the normal range, with
the exception  of a few  statistically  significant values in specific gravity in
males  exposed  to 1,500  ppm  at 6  mo and males and  females  exposed  to  3,500  ppm
at 12  mo.  Rats  exposed to 500, 1,500,  or 3,500 ppm had elevated  COHb values
but with  no  evidence of either dose-response or increased values with prolonged
exposure.
     5.3.3.1.1.1   Gross and histopathologic  observations  of rats  from  the  6-,
12-. 15-,  and  18-month  interim kills.   Numerous gross  and  histopathologic
observations were recorded for  control  and DCM-exposed  rats  at each time
period, and  most were  typical of  spontaneous  or  naturally-occurring lesions
normally  seen  in rats  of this  strain.   There  were many palpable  masses  in
males  and females.  Some palpable masses  appeared to  be  abscesses of the prepu-
tial or clitoral glands, while others were cyst-like lesions of the skin.  The
                                                      3
total  number of  masses  in the  3,500  ppm (12,145 mg/m  ) group of  males was
significantly  increased over the  controls at  15,  IB,  and 21  mo,  but  not  at
                                    5-48

-------
               TABLE 5-11.   CUMULATIVE PERCENT MORTALITY OF RATS
                    2-YEAR DICHLOROMETHANE INHALATION STUDY
DCM concentration, ppm
Month of
study
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24

0
0.8
0.8
0.8
0.8
0.8
0.8
0.9
0.9
1.7
1.7
1.7
2.6
2.7
3.6
10.9
15.2
23.8
32.4
44.2
48.4
56.8
63.2
80.0
85.3

500
0
0
0
0.8
0.8
1.6
2.6
3.4
3.4
4.3
6.0
6.8
10.9
10.9
11.8
16.2
20.0
36.2
45.3
52.6
56.8
65.3
73.7
85.3
Males
1,500
0
0
0
0
0
0.8
0.9
0.9
0.9
1.7
3.4
4.3
5.5
10.0
13.6
20.0
31.4
37.1
49.5
63.2
74.7*
83.2*
89.5
93.7
Females
3,500
0
0.8
2.3
3.9
4.7
4.7
6.0
6.0
6.0
6.0
6.8
8.5
9.8
15.2*
21.4
29.0*
33.6
43.0
54.6
58.8
72.2*
80.4*
87.6
92.8
0
0
0
0.8
0.8
0.8
0.8
0.9
0.9
3.4
5.1
5.1
6.0
7.2
9.0
13.5
14.2
23.6
26.4
36.5
42.7
49.0
62.5
70.8
78.1
500
0
0
0
0.8
0.8
0.8
1.7
2.6
3.4
5.1
6.0
6.8
8.2
8.2
10.9
15.2
20.0
29.5
42.1
50.5
58.9
64.2
65.3
74.7
1,500
0
0
0
0.8
0.8
0.8
1.7
2.6
1.7
2.6
4.3
5.1
8.2
8.2
10.0
13.3
20.0
29.5
46.3
55.8
65.3*
73.7
81.1
86.3
3,500
0
0
0
0
0.8
0.8
0.9
0.9
2.6
2.6
4.3
6.0
12.5
12.5
19.6
20.6
29.9
42.1*
57.7*
68.0*
81.4*
86.6*
90.7*
95.9*
*Significantly different from control by Fisher'.s Exact Test, p <0.05.

Source:  Dow Chemical Company, 1980.
                                   5-49

-------
23 months.  Female rats exposed to 500, 1,500, and 3,500 ppm showed an exposure-
related increase in total number of masses.  There was also a trend of increased
benign mammary  tumors  in females exposed  to  1,500 and  3,500 ppm.  The total
numbers of  animals  with benign mammary gland tumors were 9/28 (0 ppm), 10/29
(500 ppm), 11/29 (1,500 ppm), and 14/27 (3,500 ppm), whereas the total numbers
of benign mammary  gland tumors were 17/28 (0 ppm),  17/29 (500 ppm), 28/29
                          -A.                                      -~\ n
(1,500 ppm, p = 9.23 x 10 ^),  and  37/27  (3,500 ppm,  p = 2.33 x 10  1U).  These
observations were apparent  only when the  cumulative results of the 6-, 12-,
15-, and 18-mo  kills were evaluated.
     A few  other observations  appeared to  reflect exposure-related lesions.
The liver was  the only organ  that  exhibited  definite  exposure-related non-
neoplastic effects  in  both  males and females at all exposure concentrations.
Grossly, the effect was most prominent in females  exposed to  3,500 ppm and
consisted of increased numbers of dark or  pale foci.   The control  group had an
incidence of 0/28,  while  the 3,500  ppm DCM-exposed female  rats had a  signifi-
cantly greater  number  of  foci  (11/27).   Some  rats from  the 3,500 ppm  exposure
group had mottled  livers  or had an  accentuated  lobular pattern to the liver
(0/29 control males compared to 6/27 exposed males).  Because of the limited
number of rats  at each interim kill, this latter  change may be related to
exposure but may also be due to biological variability.
     Histologically observed,  exposure-related  lesions  were present  in  the
livers of both  males  and females exposed  to  500,  1,500, or 3,500 ppm.  In
males, the total number of animals with any degree of vacuolization consistent
with fatty  changes  were 5/29 (0 ppm), 19/29 (500 ppm, p =  2.05 x 10~4), 21/29
(1,500 ppm, p = 2.47  x 10~5),  and  23/27  (3,500  ppm, p = 2.81 x 10"7).   The
livers of female rats  also  had alterations considered  to  be  related to DCM
exposure.    The  total   numbers  of females  with any  degree  of  vacuolization
consistent with fatty  changes  were 13/28  (0 ppm),  20/29 (500 ppm,  p = 7.16 x
10~2), 20/29 (1,500 ppm,  p  =  7.16  x 10"2), and  22/27 (3,500 ppm,  p = 7.16 x
10  ).  Because of  these  effects,  it may  be  considered that this  experiment
was performed at the maximum tolerated dose (MTD).
     5.3.3.1.1.2  Gross and histopathologic observations of rats killed
moribund or dying spontaneously during the study and those from terminal
sacrifice (24 months).
     Non-neoplastic observations—The liver was  affected  in both  males  and
females exposed to  500,  1,500, or 3,500  ppm.  The  percentage  of total rats
                                   5-50

-------
with any degree  of vacuolization was 17, 38, 45, and 54% in the males of the
0, 500, 1,500 and 3,500 ppm exposure groups, respectively, and 34, 52, 59, and
65%  in  the females, respectively.  Also, the  degree  of severity tended  to
increase with the  dose.   The males in all exposure groups had fewer cases of
grossly observed mottled  and/or enlarged adrenals.  These gross  alterations
appeared to  correspond  to the histologically observed decrease in the number
of cases of  adrenal cortical necrosis, but the nodular hyperplasia incidence
(unilateral  or bilateral)  was increased:   18/95 (0 ppm), 30/95 (500 ppm, p =
3.07 x 10~2), 31/95 (1,500 ppm, p = 2.2 x 10"2), and 24/97 (3,500 ppm).
     Tumor or tumor-like lesions—The  Sprague-Dawley  strain  of rats used  in
this study historically has had a high spontaneous incidence of benign mammary
tumors.  The incidence varies  slightly  from study to  study,  but normally
exceeds 80%  in females  and about  10%  in males  by  the  end  of  a  2-yr  study.  The
mammary gland tumors  have  been  classified,  based  on their predominant  morpho-
logical cellular pattern, as fibromas, fibroadenomas, or adenomas.
     The benign  mammary tumor response was present in males and, to a lesser
extent, in females.  There was a non-statistically significant increase in the
number of male rats with a benign mammary tumor exposed to 3,500 ppm (14/95 as
compared to  7/95,  3/95, and 7/95 in  the  0, 500, or 1,500 exposure groups).
There was  a  slight increase in the total number  of benign mammary  tumors  in
males exposed to 0  ppm  (8/95), 500 ppm (6/95), 1,500 ppm (11/95), or 3,500 ppm
(17/97, P = 4.6 x 10"2).
     The total  number  of  female  rats with a benign  mammary tumor  did not
increase in  any  exposure  group  (0,  500,  1,500,  or 3,500 ppm  groups  had totals
of 79/96,  81/95, 80/95, and  83/97 benign  mammary  tumors,  respectively).   How-
ever,  the  total  number of benign mammary tumors increased in an exposure-
related manner, with 165/96 in the controls, and 218/95, 245/95, and 287/97 in
the  females  exposed to 500,  1,500, and  3,500  ppm, respectively.   Expressed
another way,  the average  number of benign  mammary tumors per tumor-bearing
female  rat increased  from 1.7 in the control rats, to 2.3 in rats exposed to
500  ppm, to  2.6  in those  exposed  to 1,500 ppm,  and to 3.0 in those  exposed to
3,500  ppm.   This effect is exposure related, and a dose-response  relationship
was  apparent.  There  was  no  indication of an  increased  number  or  incidence of
malignant  mammary tumors in either males or females.
                                   5-51

-------
     The number of malignant tumors (Table 5-12) increased in male rats exposed
to 3,500 ppm.   This increase did not appear to correlate clearly with an in-
creased number  of any one tumor type or location.   However, this observation
led Dow Chemical  Co.  (1980)  to  re-evaluate the  gross  and  histopathologic data
on all tumors arising in  or  around  the  salivary glands.   Table  5-13  lists the
specific individual  animal data for these salivary  gland  area tumors,  showing
the palpable mass data, specific histopathologic diagnoses,  and the  number of
sarcomas with metastases.   Table 5-14  summarizes the incidence of salivary
gland region sarcomas in male rats.
     Grossly, these tumors were  large  (several centimeters in diameter),
cystic, necrotic, or hemorrhagic.  They  appeared  to invade all  adjacent
tissues in  the  neck region,  and often completely replaced the normal salivary
gland tissue. Histologically, all were  sarcomas.  They were  composed of cells
that varied from  round to spindle-shaped,  but that appeared to be of mesenchymal
cell  origin. Mitotic figures were frequently observed, as were necrosis and
local  invasion  into adjacent tissues.  Most  tumors had remnants of normal
salivary acini  or ducts  that were  caught up  in the cellular proliferation.
Two were relatively small masses, and appeared  to be  arising in the  intersti-
tial  and capsular tissue of the salivary glands.
     One tumor  of this type was found in the  controls (1/93) compared  to 0/94
in the  500 ppm exposure  group,  5/91  in males  from the 1,500  ppm exposure
group, and  11/88 in males from the 3,500  ppm  exposure group  (P = 0.002).
Historically, a spontaneous  incidence  (0  to 2%) of this  tumor  type has been
observed in  Dow's laboratory.   Therefore,  the  12.5%  incidence  (11 of  the 88
rats,  Table  5-14) found in the  males from the 3,500 ppm group was  higher than
the corresponding  controls of this  study,  and was higher than expected based
on historical control  data  for male rats of  this  strain.  Also, the  males
exposed to  1,500  ppm had  five of these  tumors,  which  was  also slightly higher
than expected,  but  was not statistically significant.  Therefore,  this effort
appeared to be exposure related in the males exposed to 3,500 ppm.
     The total  number  of  male rats with malignant  tumors was similar  in the
control,  500 ppm,  and 1,500  ppm exposure groups.   Males exposed to 3,500 ppm
had an  increase in  this  category,  since 69  of the 124 rats had malignant
tumors compared to 57, 59, and 57 in the 0,  500, and 1,500 ppm exposure groups,
respectively.
     In 1984, Burek et al.  published the results of the Dow Chemical Company
(1980) study.
                                   5-52

-------






o
_1
-r*
5E
l — l

>—
CO
to
UJ

CM
rv
O
u_

LU
z
1—
LU

o

O
_J
"r*
O
l— 1
a
a
LL!
UJ

to
1 — 1
•z.
1 — 1
a
"^
to

^f
tx,

r**
O
LL.

?
a
oi

^-
1—
o
1—

LL.
o

>_
rv
^C
s:

— ^
to


,
CM
rH •
1
LO

LLJ
_J •
CQ

1—











01
•!— in
-P r—
re to
r— -P
=! 0
e -P
o
uj re
CU C i —

're E •'r-
s s- -^
a) cu
LL. 1—

•3 -0 T3
o c -a c
a; re cu =i
C i— J3
re to i — ซr—
•P r* ปi— J_
C -P J* O
O to E
a. cu
to -o
a>
>
•i— U)

re "re

3 o
E -P
3
0


ui re
CU C i—

lo 'E ^
s: s- -^
0)
l—

to
i3 -a -a
o c T3 c:
a> re cu n
C i— J2
re in i — T-
•P -C -i- S-
C -P Jii O
o re E
Q. CD
10 T3


S_ r—
CD •!-
-P ^
1— 1
c
O

'-P
re E
S- Q.
•P Q.
c
O) C
U •!-
c
o
o

























^ ^ ^. ^
CM CM CM CM
rH rH rH rH





rH •* CO •=!•
CM CM rH






LO rH CM CO
r-- r-ป oo cn








^t* ^~ ^~ ^~
CM CM CM CM
i-H rH rH rH








^^ ^^ iฃ> r^^
rH rH








i-H rH Cn O
OO 00 00 Cn







cn cn cn r^
CM CM CM CM






CD CD CD CD
O O O
LO LO LO
i-H CO








10
-P
re
s-

i-
o

S-
ai
^Q
E
rt T3
c ai

1 	 •! —
to E
-p re
o x
1— 0)

CM r~- to cn
0 0 O 0
rH i-H rH rH





rH ซ3" CO ซ3"
CM CM rH






r- co cn I-H
to to P- cn








LO CM CM tD
f^ป r^ co co









co ro to r^-
r-H rH








CO CM f~~ CO
LO LO tO tD







CT> ^ cn i — I
I— 1






0000
000
LO LO LO
i-H CO








m

re
S-

>tซ
o

i- S-
*
to to r-. oo








O CV) ID i-H
LO ^ LO LO









i— 1 i-H tO LO
rH rH








co cn CM r^*
ro CM  13
re -P
S-
•p
<(- C
o re
c
S- CD
CU *f~ •
J3 i—
E re
13 E
C

i —
re sz
•p -P
O •!-
h- 3





























































0
co
cn
rH

n
>j
C
re
a.
E
0
o

I—
to
U

E
cu
.c
C_5

3
O
CD



CU
U
ฃ-
Z3
0
to
5-53

-------




—I
1

t— *
s
t— 4

H-4


1

s
ง
>-i CO
C3 Cฃ
ฃ2
^-
J-+I
ZCM
3 os
u o
tu

ce z
|S

co <ฃ
ttl
O >-ซ
Lu

< CQ

< UJ
a o
I-H
>- Cฃ.
C3 O
g ^


t- UJ
< z
a. LU
o —i

co 3:
t— ซ r—
a: uj
ง0
ซE r—

-Q
CO UJ
CO CO

ฃ a.

UJ UJ



SS
n ^J
_J

rv* •?
^3

•2
i
o
I—
1
UJ
t— 4
r-

co
rH

UJ

ง
l~






14-
O Ul
+j ai
Ol C Ul
u re re
CZ -P -P
Of U) Ul
•a •<- re
•<- TJ -P
> Ol
UJ E





re ui
U -r-
•r- Ul
r- D)
O 03





^^

S- U
O x_y
งป
4J in
o.
t- 0
O t.
u
Ol Ol
N C
•f—
CO P
re


._
15?
•p a.
Ul O
i.
1- U
o ai
c

4-> C.
C O
O 1-
s:


O E
m P u
3 Ul •**•

•r- -D
It- It- 01
o >
1= S-
ai to 01
IM J= Ul
•i- ฃ J3
CO O
*^
•Q
3 -P
•P Ul
Ul f- T3
•r- Ol
*->!->
O t-
S- 01


C 3 O
i*1
"S1
o.
01 a.
ฃ• ^rf'
3
ui a.
O 3
a. o
X i-
LU D)

r- U
re ai
E -Q

'ฃ 3
<: c



01
c
o




J_
•r- ซ
•a E
e o
3 U
i re
Ul
Ul
3 -a
o 01
01 

> c C re re c: > Dl re re CO E X •*• X in [^ CO CO r-l. ง in rH CO CO r-. CO i CO 01 c o z re o c 1 C re •a s C -C re u 1 — U) >i jg re c > Dl •r- ปf— re re CO E •a Ol Dl re T3 i— C TJ c re 0> Ol r— •P Dl 0 >> 01 f- >> •p *> C 01 j= re •a DI > o ui re z ^ ui S i •a 01 ^, 01 Ul J3 O O Z ง in 1-1 en o CO CO i CO Ol o z i 14- re •5 o c c 3 C re 41 re "DI 01 ฃ".™ re P > c: •i- 0) r— S- re oi CO 1- CM X CO X ^* CM in i-i CM O s rH CM CM P- CO 1 CO ai c o z •o c 3 O i. I ui re Ol U c s- re re •P Ul 3 O i— 3 01 CO U C 01 '5)^> Ul ai E N re •r- S- Ul Dl O CO ~yr ^^ rH CM s 0 in iH CO CO 1 CO I-- 01 c 0 z •a c 3 O i- 1 ui re 3 E O O re re 3 u, U I— J3 i— 3 01 CO U X •=*" X in 0 CM CO CO rH O 0 in i-i en ^^ CO co Ol c 0 z i T3 C ,DI O ฃ> *j re ui •>ฃ r- -Q re •!- CO *r— "re re CO U CO X CO X fn. in r-l CO 3 o o in CO CO en CO CO i CO Ul Ol o 1 S ui re 3 U] 0 0 01 i. c .a re -r- 3 O u s- J3 3 3 01 CO C in X CO X en CM CM rH CM O 0 in CO o CO in CO 1 co f- Ul Ol 01 Dl re a. Dl 1 C ui re ^ 3 E 0 00 •— ai u i— C i. 0 re re t- •P Ul 3 O Ol US- J= .Q J3 P 3 -r- 00 t- C o •o Ol 3 rr +3 C C Ol O > u 'DI Ol N >l— Ul o z rH CM CM 8 o o in CO CO CO CO CO i CO r- 5-54


-------



































T3
01
3
C
C
O
u



CO
rH

in

UJ
cd
*~







































O in
•P Ol
01 c in
U 10 10
ot tn in
"O v- (O
•i- -a -p
> 0)
UJ E







n) in
U •!-
•i- in
O) O
o c
r— 0)

in ~o
•r-


S- U
O *~s
3 >,
•P in
a.
if. o
O i.
u
O) 01
N C
tr>
to -P
(0
•ง•>,
3 in
•P Q.
in o
t- U
O 01
c
.c
•p i-
c o
=•*•
fc. X-s
0 E
ง4-> U
0) V- •
•P S.

*- "ป- 0)
o >
C ฃ-
01 01 01
N jz in
•r- S J3
to o
•^
•a
3 -P
•P Ul
in i. -a
•r- 0)
"*->ป->
0 S-
S- 01
jz o in
•p E .n
C 3 0
O +ป
^*

^
a.
Ol Q.
fc. s^^
3
in a.
0 3
a. o
x t-
LLJ D)

i— S-
10 01
E XI

C 3
•a: c



01
c
o
•z.






•o

1 -p
ID
U) •!-
3 P
0 C
Ol Ol
C S-
rO 01
4-> 11-
3 <(-
U -r-
-Q "O
3 C
to 3


,3-

X

^>

X

p^.






rH













to









rH
CM









0
O
in
CO



CM
00
ro
1
to
r^ป









(0
E
O

ฃ„
<0
in

^""
01
u
•o
c
3
O
f-



































































c
o
z





•o


i to

U) -P
3 C
O 01
01 t-
C 01
(0 "ป- (0
•P  3 in

in
CO

X

^

X

pv.






to
rH












00









10
rH









0
O
in
CO



00
r—
in
CO
i
to
i**



0)
c
o
•z.



ro
E
O
u
i.
i as
in
U)
3 U
O •!-
ai s:
c a.
ro c.

u o
J2 01
3 i—
to a.


CO

X

^ป

X

to






00
rH










in

CM









CO
rH









O
O
in
CO



.00
S
CO
1
10




01
c
o
•z.



10
E
o


i as
in
in
3 U
O •!-
C Q.

3 E
U O
.a at
3 r—
to a.


CO

X

in

X

in






CO
rH












^>









f^
rH









O
0
in
CO



rH
CM
to
CO
1
to




in
01








I

in
3
O
f

3
U
J3
3
l/>


^

X

1C

X

CO






to
rH












CO









in
r-i









o
0
in
CO



to
to
to
CO
1
IO
r^








as
E
O


10
in

u

Q_

E
O
01
r-
a.



































































01
c
o
z





10


1 U

in n)
3 U)
o o
01 t-
C J3
(O v
-p 1ป-
3 O

ฃi 3
3 ai
to c





in

CO









,4.
rH












1




•o
01
2
(U
in

O


i






o
0
in
CO



rH

CO

IO
(X,



S
z








1

in n)
3 E
ง8
C i.
10 10
H-> in
3 0
U C-

"ง •"?
to *-

in
CO

X

f*^

X

fปx.






ai
rH












to









CT>
rH









•o
O
in
CO



^
a>
in
CO

to
r^.































































o
CO
on
rH


^^
C
IO
Q,
E
O
O

-------
    TABLE 5-14.   SUMMARY OF SALIVARY GLAND REGION SARCOMA INCIDENCE IN MALE
            RATS IN A 2-YEAR INHALATION STUDY WITH DICHLOROMETHANE
Dose
0 ppm
500 ppm
1500 ppm
3500 ppm
Incidence*
1/93
0/94
5/91
11/88
(1%)
(0%)
(5.5%)
(12.5%)
Fisher1


(P =
(P =
s exact
—
—
0.10, N.
0.002)
test


s.)

*Cochran-Armitage test for linear trend, p <0.0001.
N.S. = Not significant.
Source:  Dow Chemical Company, 1980.

5.3.3.1.2  Dow Chemical Company (1980)  inhalation study in hamsters.  A total
of 866 Golden Syrian hamsters [Ela: Eng (syr) strain; Engle Laboratory Animals,
Inc.,  Farmersburg,  Indiana]  (107  to 109/sex per exposure concentration) were
exposed by inhalation  to  0,  500, 1,500,  and  3,500  ppm (0, 1735, 5205, and
          o
12145  mg/m ) of  DCM.   The materials and  methods for experimental design are
the same as mentioned previously in the rat portion of the Dow Chemical  Company
(1980) study.  The body weights of the hamsters were 61 to 70 g when they were
received.  The hamsters were marked by unique toe clips for group identification
and by ear punch for individual identification within the cages.
     The mortality  data  for  males and  females  are  presented  in Table 5-15.
Female hamsters  exposed  to DCM at  3,500  ppm  had a  statistically significant
decreased mortality  from  the 13th through the 24th mo of the study.  Females
exposed to 1,500 ppm also had statistically  significant  decreased  mortality
from the 20th through the 24th mo.  This decreased mortality in females exposed
to  3,500 and 1,500 ppm was considered to be exposure related.  The remaining
exposure groups  of male  (500,  1,500,  and 3,500 ppm)  and female  (500 ppm)
hamsters had no  exposure-related differences in mortality.  Some hamsters in
each  group  had  alopecia  at  5.5 mo into  the  study, but this alopecia was
secondary to a mange mite (Demodex species)  infection.  This  parasite did  not
result in increased mortality or  morbidity.   No treatment-related differences
were  observed  in the body weights of either males or females exposed to 500,
1,500, or 3,500 ppm of DCM.
                                   5-56

-------
             TABLE 5-15.  CUMULATIVE PERCENT MORTALITY OF HAMSTERS
                    2-YEAR DICHLOROMETHANE INHALATION STUDY
DCM concentration, ppm
Month of
study
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
21
22
23
24
Male
0
0
0
0
0
0
0
1.0
1.9
1.9
2.9
3.8
5.1*
6.4
10.6
14.9
23.4
24.5
31.9
36.0
37.1
41.6
56.2
61.8
82.0
500
0
0
0
0
0.9
1.9
1.9
3.8
5.8
5.8
5.8
. 6.7
10.1
14.1
17.2
18.2
19.2
24.2
29.8
43.6
51.1
61.7
63.8
78.7
1,500
0
0
0
0
0
0
1.0
1.9
1.9
2.9
2.9
3.9
10.2
13.3
14.3
17.3
19.4
24.5
31.2
41.9
54.8
68.8
75.3
88.2
3,500
0
0
0
0.9
3.7
3.7
5.8
5.8
6.7
9.6
11.5
12.5
17. 2f
24.2
26.3
33.3
34.3
37.4
42.6
55. 3f
60.6
69.1
74.5
85.1
0
0.9
0.9
1.9
1.9
2.8
3.7
3.9
4.9
4.9
5.8
9.7
13.3*
22.6
26.9
32.3
36.6
41.9
52.7
63.6
71.6
80.7
88.6
94.3
100.0
Females
500
0
0
0.9
1.9
1.9
2.8
4.9
4.9
5.8
9.7
10.7
11.7
16.3
24.5
26.5
30.6
32.7
38.8
59.1
63.4
70.0
82.8
91.4
95.7
1,500
0.9
0.9
1.9
1.9
2.8
3.7
4.8
7.6
8.6
8.6
9.5
13.3
15.0
20.0
24.0
29.0
32.0
40.0
50.5
55. 8t
61.lt
68. 4t
75. 8t
89. 5t
3,500
0
0
0.9
0.9
0.9
0.9
2.9
3.9
3.9
4.9
4.9
4.9
7. It
11. 2t
15. 3t
23. 5f
27. 6t
30. 6t
40. 9t
45. 2t
53. 8t
72. Ot
80. 6t
90. 3t
*Five males and five females died due to food deprivation.  These animals were
 subsequently deleted from mortality calculations.
tSignificantly different from controls by Fisher's Exact Test, P <0.05.
Source:  Dow Chemical Company, 1980.

     Based on  the  information available to the  Carcinogen  Assessment Group
(CAG), it is very difficult to conclude whether the MTD was used.  Dow Chemical
Company has not submitted a 90-day dose-finding study, but a 30-day inhalation
study  has been  reported in a letter from Dr.  J.  Burek to Dr.  D. Singh, dated
May 1, 1981.
     "The study was  conducted prior to the two-yr  study, but has not been
reported.  CD-I mice,  Golden  Syrian hamsters, Sprague-Dawley and CDF (F-344)
                                   5-57

-------
rats  were exposed to 0,  2,500,  5,000,  or 8,000 ppm DCM  vapor 6 hr/day, 5
days/week,  for  a total  of 20, 19,  20 or 6-2/3 exposures,  respectively,  in
21-29  days.   Body weight data was  obtained  throughout the study.   Clinical
chemistry parameters  were measured.   All  animals  underwent gross pathological
examination  at  the termination of the  experiment.  The weights  of  the  liver
and kidneys  were recorded from animals  in the  0,  2,500, and 5,000 ppm groups,
and organ/body  weight ratios were calculated.  Animals exposed  to  8,000 ppm
DCM exhibited anesthetic  effects,  increased blood urea nitrogen levels  in
Sprague-Dawley  rats,  and decreased  body weights in rats.   The  animals exposed
to 5,000  ppm showed slight anesthesia,  decreased  body weight  in male rats,
increased SGPT  values in female mice and Sprague-Dawley  rats  and increased
liver weights in  female mice,  hamsters and rats.  Animals exposed to 2,500 ppm
DCM appeared to scratch more than controls and therefore appeared to be affected,
but showed no other effect attributable to exposure.  The target organ in this
study was the liver.  Because  of the results obtained, 8,000 and 5,000 ppm DCM
were considered  to be too high a dose level  for the two-year study, and 2,500
ppm did  not appear to have produced a severe enough response over the 30-day
period. Therefore,  concentrations of  3,500 ppm of DCM  was  [sic]  chosen  as the
top dose for the two-year study."
     Dow Chemical Company (1980) conducted a subchronic dose-finding study for
a perfeid  of only 30 days  rather  than for the  90-day period that is usual in
most animal  bioassays.   Further,  body weight or mortality  rate in the experi-
mental group of  hamsters  does not decrease as  compared to the controls.
Hematologic  determinations,  serum clinical chemistry,  urinalysis, bone  marrow
collection,  and  blood COHb determination were  done in  animals  at the 6-,  12-,
18-, and  24-mo  interim  kills.   No treatment-related effects were observed in
any of the parameters evaluated in male or female hamsters after 6,  12,  18,  or
24 mo of exposure to 500, 1,500,  or 3,500 ppm of DCM,  respectively.
     Carboxyhemoglobin determinations were performed on the blood of male and
female hamsters  following 22 mo  on  test.   Males and females exposed to  DCM at
500, 1,500,  or  3,500  ppm all  had significantly elevated COHb  values.   There
was a  slight trend in a  dose-response relationship in  females, since the mean
  i
CpHb value for those exposed to 500 ppm was 23.6%, while the values  for females
exposed to 1,500 or 3,500 ppm were 30.2% and 34.6%, respectively.  However,  an
apparent dose-response relationship was not observed in males.  Dow Chemical
Company (1980)  indicated that  these data, when compared to those in the rat
                                   5-58

-------
 study,  suggested that hamsters had a  greater degree of metabolism of DCM to
 carbon  monoxide.  Furthermore, the  apparent dose-response  in females was
 surprising.   As  a result, additional  hamsters  were  exposed to a single 6-hr
 exposure,  and their COHb values were  determined.   There was no apparent sex
 difference,  and  the dose-response relationship observed  in  females after 22
 months  could not be verified.  Since  there was no dose-response  relationship
 in  male and  female  rats or  male  hamsters,  and since the  female hamsters
 exposed  to a single 6-hr exposure did not show a dose-response relationship,
 the  apparent trend for an exposure-related increase in female hamsters at 22
 months may be a  cumulative effect.
     5.3.3.1.2.1 Gross and histopathologic observations of  hamsters from the
 6-,  12-, and 18-month interim kills.    A  variety of gross and histopathologic
 observations were  recorded  for hamsters that were sacrificed at  the 6-, 12-,
 or  18-mo  interim  kills.   Histopathologically,  exposure-related  differences
 were present,  and  consisted of  decreased numbers of  hamsters with amyloidosis
 of the  liver,  kidney,  adrenals, thyroid, and spleen.  A  few animals  in each
 group may  or may not represent the trend of amyloidosis in males.
     5.3.3.1.2.2  Gross and histopathologic observations of  hamsters killed
 moribund or dying spontaneously during the study and those from terminal
 sacrifice  (24-months).
     Neoplastic and non-neoplastic observations—Gross  and  histopathologic
 examinations were  conducted on  all hamsters that died or were  killed moribund
 during the study and  on all  surviving hamsters at the end of the study.  The
 histopathologic  observations  for males and females are presented in the Dow
 Chemical Company report  (1980,  tables  124 and  127).  The observations  shown
 include  all  the  neoplastic and non-neoplastic lesions  recorded  for these
 hamsters.  Most observations  were  within the normal or expected  range for
Golden Syrian  hamsters,  as indicated  by Dow  Chemical  Company.   The female
 hamsters had  increased incidences  of lymphosarcoma in the experimental  group.
The  incidences were 1/96,  6/95, 3/95,  and 7/97 (p  = 0.033) in the 0,  500,
1,500,  and 3,500 ppm  groups,  respectively.   A re-evaluation of lymphosarcoma
data of  female hamsters  by  the Carcinogen  Assessment Group (CAG) resulted in
the  following  incidences:  1/91,  6/92, 3/91,  and 7/91 (P =0.032)  in the 0,
500, 1,500,  and  3,500  ppm groups,  respectively. The differences  between the
denominators above reflect the CAG's  use of actual  numbers of animals examined
(which did not include animals  that were severely cannibalized, autolyzed, or
                                   5-59

-------
missing), whereas the  Dow denominators included the total number of animals.
Also, only  a small  number  of mammary gland tissues were  examined,  and no
lesions were  found.  Table  5-16 summarizes the total tumor data.  The  total
number of hamsters  with benign tumors increased  significantly  in  females  at
3,500 ppm;  the total  number of  hamsters  with malignant tumors increased
significantly in males at 1,500 ppm.
     In 1984, Burek et al.  published the results of the Dow Chemical Company
(1980) study.
5.3.3.1.3  Summary of the Dow Chemical Company (1980) rat and hamster inhala-
tion studies. Based  on all  the data evaluated, the four following points are
considered to be major findings in the rat and hamster studies:
     1.  Male rats exposed to 1,500 or 3,500 ppm appeared to have an increased
number of sarcomas  in  the ventral midcervical area near the salivary glands.
There were  1/93, 0/94,  5/91,  and 11/88 (P =  0.002)  sarcomas  in male  rats
exposed  to  0, 500,  1,500,  or  3,500  ppm,  respectively.   Based  on  routine
sections,  special   stains,  and ultrastructural  evaluations,  these  tumors
appeared  to  be of  mesenchymal cell  origin;  however,  a  myoepithelial  cell
origin of these  cells  could  not  be ruled  out.  These tumors had some  areas
that  morphologically  resembled one  cell  type  (i.e.,  neurofibrosarcoma,
fibrosarcoma),  and  still  other tumors  had  cell types  that were  undif-
ferentiated  or pleomorphic.   In some, one  cell type was  predominant,  while in
others,  areas  of all of  the  above  cell  types were  present,  depending on the
area  of  the  tumor examined.   Furthermore, the origin of each of these  tumors
remains  questionable.   All  appeared to be arising  in the midcervical region,
and all  involved the salivary glands.  Only two tumors were small enough to be
localized within the salivary gland.  The rest were  larger tumors that  clearly
involved  the salivary  glands as well  as adjacent tissues,  and  could have been
growing  either into or  out  of the salivary glands. However, they probably
arose  within the  salivary  glands based on the  two localized small  tumors
described above.
     Therefore,  there  was an apparent association between the  increased inci-
dence  of sarcomas  in the salivary  gland region of male  rats and  prolonged
exposure  via inhalation to 1,500 or  3,500  ppm DCM.  There were no  salivary
gland  sarcomas  in female rats or in hamsters  of either sex.   Further, it will
be  of interest to find  out what  kind of lesions  are present or  absent  in  the
ongoing  National Toxicology  Program inhalation study.
                                    5-60

-------











c/5

•a:
LU


CM


0
u_

•^
O
t-H
1—
3

•i- in
•p.—
to to
1— -p
3 0
E-P

CJ



t—
re
cz. —

E-i-
S- ^
in co
CO I—

"to
E
co in
LL. 3 T3
O C TJ
co ca -a c
CZ CO 3
ca in i — .a
-P -Ci — •!-
(= +J-i- S-
o ro -^ o
Q- 01 E
oo -a


E
•r- in
S-i—

-P-"^





CO
> in

-P ro
i— 0
3 +->
E

C-]







C i —
E -^

aj
•

in
CO
f — in
to 3 -a
•Z. o cz -o
co ra *o cz
CZ CO 3
•P -Czi— •!—
C -P -i- S-
o ra -^ o
O- Ol E
oo-o




E
•r- in

CO i —

C -^




cz
o


ro^->
S- E
•P 0.
CvS
u

o



















LO CM i— ILO












o ซ3- o cn
I-l










cn co So oo








LO O O ซ*•
r-HrHi-HrH








CD CD O CD
(— t i— 1 i~H r— 4











UD O i— 1 ซ3-
t-H CMrH i— 1










UDซ*- CMOO
p~r~- OOP~











LO O CD LO












O CD O O
000
LO LO LO
I— 1 CO






cn

O-r-
S--X
S- 3-D
co -a o

E in s-
3 S- CO
cz 
1—,

>^
crt t-H cn CM
r-) CM r-ICO












1 r— 4 4~ CO











i— ICMrH CM








CM CD CM CM









LO CD 1 — CT>
CM CM CM CM












ro cn oo i^.










oo r*- r*- oo
r-l i-lr-( r-H











•<3- ro CM ^*












CD O CD
LO LO LO








O -CZ
S-T-
0) 3:

E in
3 S- i.
CZ CO O
__ -P S

"to E -P
•P ro
O -ฃZ ro
1—

1,
ro cnro m
i— 1 r-ICM












i i— Icoro











i-icocneM
i— 1 CM








CM OrHrH









CD cn oo cn












CM r^ LO in










•3- CD in CD
r-H rH i— 1











ซ^- CM CM CO












o oa
LOLO LO
r— t OO






CO
t—
0 J=

^'* i

E in -P
3 S-
CZ CU CZ
•p cn
i — in •(—
ro E cz
•P ro CO
O .C JQ
1 —


ooror-- o
T-H T-H












1 O CMO











oo ro *^* crt








CD CD i — 1 i — I









1 — CO LOO












t-l-cfrOrH










m oo CM co
1—1











CD i — t CD i — 1












CD CD CD
LO LO LO
i— ( ro






ra i-

0 J= E

CO 3

E in c:
3 i- ro
cz co c:
-P O)
i — in *i—

0-ฃ= E
1—



in
- ro
o 3
in
Sal
o
• o
-a j=
CO -P
M CO
•r- 0.
i — O
ra -P
J3 in

CZ J=
cz
ro O
U CZ

>> CO
i — in


CO U
> co
co .a
in

ฃ. f—
O •!-
-
CO •<-
Nl S-

r— -P r^t
0 C CZ
-P-1- 0
CO -CZ +J
-P U
>>cz ro
i — o a)
CO S Cฃ
s_ i
CO UD CZ
> o
CO CO S-
in -ฃ: I-H

CO CO
s- e 3
CO O i—
3 5-OQ

•P CZ
to in ca
^Z S-T-
-P co in
-P in
in in 3
s- e s-
co co a.
+J s:
to tn

_^ 'ta "si

S- CU E
O 
H— i— 1 S-
CO
•a s- >
CO O-i-
ฃD- i —
ra E
u a. ca
in o.
CO 5-
o o
•p 0 <<-
CO LO
JZ -P
-p co a.
•D CO
in 3 o
t- i — X
CO U CO
•P CZ
in •!— in
E i —
to .P ro
-cz o E

CO CZ
"U in ra
3 CO
i — O CO
u -a in
CZ CO
•1-1 — -CZ
ra -p
•P -P
0 0 CZ
CZ -P O

in in co
co-i- c:
O -ฃZ O
CD -P -0
•X











































f
LO
CD
CD
V

Q_

ซ,
•p
in
CU
1 —

-P
u
to
X
LU

in
-^_
CU

in

LL.

_Q
-a
CO
Nl

,^
ra
cz
to

cz
CO

3

in

O
i.
-P
cz
o
u


o
S-

-l->
cz
a>
s_
CO



T3


JJ
c;
ro
CJ

•,ซ

cn
oo


















































































CO

r~\
to
(J


CL CD
a. co
ro cn
!— I
-P
o

cz
t- ra
O O-
E
-a o
CO CJ
cz

"E "to
ra u
X '!-
CO E
CO
CO -C
CZ CJ
i 3
0
in CD.
CO
•p
ro
U CO
•r- U
•a s-
CZ 3
1— ( O
1 OO
5-61

-------
     2.  Male and  female rats exposed to DCM had increased numbers of benign
mammary tumors as  compared to control values.   Female  rats  exposed to 500,
1,500, or 3,500 ppm of  DCM had  increased  numbers of benign mammary  tumors per
tumor-bearing rat  in  comparison with the controls.   The increase was evident
in the palpable mass  data and the gross  necropsy  findings,  which were con-
firmed by the  histopathologic examination.   The total  number of  female rats
with benign mammary tumors was not statistically increased  in  any exposure
group (0, 500, 1,500, and 3,500 ppm groups had a total  of 79,  81, 80, and 8-3
animals with benign mammary tumors,  respectively).   Sprague-Dawley rats have
very high incidences of spontaneous mammary tumors.   However, the total number
of benign  mammary tumors  has  increased  in  an exposure-related manner with
165/92 in the  controls  and 218/90, 245/92, and 287/95 in the females exposed
to 500, 1,500, or 3,500 ppm, respectively.  Expressed another way, the average
number of  benign  mammary  tumors  per female rat increased from  1.7 in the
controls, to 2.3 in those exposed to 500 ppm, to 2.6 in those exposed to 1,500
ppm, and to  3.0  in those exposed to 3,500 ppm.  This increase was considered
to be exposure-related and dose dependent.
     A mammary tumor  response was present in male rats also, but to a lesser
extent than  in females.   The number of  rats  with  benign mammary tumors  in
males exposed to  3,500  ppm increased but this increase was not statistically
significant.   The  total  number of benign mamary tumors in males exposed to
1,500 or 3,500 ppm increased slightly.   As was  the  case in females,  these
effects in males exposed to 1,500 or 3,500 ppm were considered to be exposure-
related.
     There were no mammary gland tumors in male or female hamsters.  Also only
28/92, 44/93, 30/94,  and 27/93 mammary gland tissues were examined in the 0,
500,  1,500,  and 3,500  ppm groups, respectively.  Not  a single  lesion was
recognized in the mammary gland tissues examined.  The CAG feels that a greater
number of  hamster  mammary gland tissues  should  have  been  examined  to better
evaluate the true incidence of mammary tumors.
     3.  There was an increased incidence of lymphosarcoma in female hamsters.
The  incidence was  1/96, 6/95, 3/95, and  7/97 (p = 0.033) in the 0-,  500-,
1,500-and  3,500-ppm  groups,  respectively Dow Chemical  Company  (1980).   A
re-evaluation of  lymphosarcoma data  of female hamsters  by  the CAG resulted  in
the  following  incidences:   1/91, 6/92, 3/91,  and 7/91  (P  =  0.032)  in the 0,
500,  1,500,  and  3,500 ppm groups, respectively.  The differences between the
                                   5-62

-------
 denominators  above reflect the CAG's  use of actual  numbers  of animals  examined
 (which did not include animals that were severely cannibalized, autolyzed, or
 missing),  whereas the Dow  Chemical Company (1980)  denominators included the
 total  number  of animals.   Dow Chemical  Company  (1980)  reported that  the  females
 exposed to 3,500 ppm had better survival (statistically significant) than the
 controls  and thereby had  a greater chance to develop  these  tumors.   After
 correction for survival  (1/39  versus 7/63) by  the CAG, these  data  are  not
 statistically significant  (P  = 0.12).
     4.   There appears  to be a question as to whether the doses given the rat
 and  hamster were at or  near the MTD.   The body  weights of male rats  increased
 particularly  toward  the  latter part of  the  experiment,  whereas  the body  weights
 of  female rats were unaffected  in any  experimental group.  Exposure to  3,500
 ppm  resulted  in an  increased  mortality rate  in female  rats during the last 6
 months,  but the male rats  were unaffected at any  concentration.  On  the  other
 hand,  decreased mortality was  observed  in female hamsters exposed to 1,500 and
 3,500  ppm,  while mortality in male  hamsters was unaffected at 500, 1,500,  and
 3,500  ppm based on only a  30-day  rat and  hamster  inhalation  (dose-finding)
 study.  Based on this information, it is difficult  to judge whether the  animals
 were given  a dose equal to  the MTD.
 5.3.3.1.4   Dow Chemical Company (1982) inhalation  toxicity and oncogenicity
 study  in rats.  A  2-yr  inhalation study of DCM with  Sprague  Dawley  rats and
 Golden  Syrian hamsters  by  Dow Chemical Company  (1980) has been described
 earlier  in  this  document.   In that study,  animals  were exposed 6 hr/day, 5
 days/week  for  2  yr to DCM  at 0, 500,  1,500, and 3,500 ppm.  The carcinogenic
 response was  positive in rats but negative in hamsters.   In  rats, the liver
 appeared to be the target organ affected by exposure.   Hepatocellular vacuoli-
 zation, consistent with  fatty change, was  observed in  male and female rats
 inhaling 500,  1,500,  or  3,500 ppm DCM.  There was  an  increased incidence-of
multinucleated hepatocytes  upon  exposure to. 500,  1,500, or 3,500 ppm, and an
 increased number of  foci  and areas of  altered hepatocytes  at 3,500  ppm in
female rats.  Benign mammary tumors were increased in male rats inhaling 1,500
or 3,500  ppm, and  in female rats inhaling 500, 1,500, or 3,500  ppm DCM.  Male
rats exposed  to 1,500 or 3,500  ppm DCM  had  an increased number  of sarcomas in
the region  of  the  salivary gland.   Female rats  inhaling 3,500 ppm DCM had an
 increased mortality  rate.   Carboxyhemoglobin levels  in the  blood of rats
exposed to  DCM were  higher than control levels;  however,  no differences  were
                                   5-63

-------
observed in COHb  levels  of animals inhaling DCM at  500  ppm versus animals
inhaling it at  3,500  ppm.   The objective of this second  study was to further
investigate the toxicity of DCM at concentrations far below  those that may
cause saturation of the metabolic processes in rats.
     A total of 360 male and 492 female Sprague-Dawley rats (Spartan substrain,
6 to 8 weeks old) were used in this study.  Groups of 90  rats/sex were exposed
by inhalation to  0 (control), 50, 200,  and  500  ppm  (0,  173,  692, and 1,735
mg/m3)  DCM  (technical grade,  lot  #TA 05038, with purity  of  at  least 99.5
percent) 6 hr/day, 5 days/week, for 20 (males) or 24 mo (females).  Occasional-
ly an  exposure  was shorter than 6 hr, due to vapor  generation or mechanical
problem.  In addition, 30 extra female rats, identified as 500/0, were exposed
to 500  ppm  DCM  for the first 12 mo  of the study and were housed as control
rats for the  duration of the  study  (last 12 mo).   Another 30 female  rats,
identified  as 0/500,  were  housed in  the same manner as  control  rats  for  the
first  12 mo of  the study and  were exposed  to 500 ppm DCM for the remaining
12 mo  of the  study.   To determine the rate of DNA synthesis in the liver, 18
female  rats were  included  in each group.   After  6, 12, 15,  and  18 mo of
exposure, five  rats  of each  sex at  each exposure  level  were  sacrificed.   In
addition, five  female rats  from each  of  the 500/0 and 0/500 groups were sacri-
ficed  at the 18-mo interim  necropsy.
     Clinical laboratory tests for chemistry, plasma hormone levels, and DNA
were made on interim  sacrificed rats.  Gross and microscopic examinations were
made of animals at interim  and terminal  sacrifice, as well as of  animals dying
spontaneously and  those that  were  killed moribund during the study.
     As reported  by the authors,  the nominal and analytical  concentrations of
DCM  in the  chambers were in close agreement, indicating  no detectable loss or
decomposition of  test material during vaporization.   Approximately 2 mo after
the  initial exposure  to DCM,  symptoms consistent with sialodacryoadenitis  (SDA
virus)  were observed  in  male and female rats  in each experimental group,
including control  groups.   The rats  from all  exposure groups appeared to be
equally affected,  and the  symptoms were  not  apparent 3 weeks  after the  initial
observation.
      No significant difference in body  weight gain  was  noticed  in male rats,
but  the mean body weights of  female rats at 50,  200, or 500 ppm were signifi-
cantly higher throughout the study period in comparison  with  controls.  Although
the  authors of this study consider  this to be  a reflection of  biological
                                    5-64

-------
 variability,  the CAG considered that the highest dose was not the MTD because
 the same strain of  rat  tolerated  a dose of 3,500  ppm by inhalation in the
 previous study at  the  same  laboratory  (Dow  Chemical  Company,  1980).
      No  increase in  mortality  rate from  that of  the control group  was observed
 in  male  or  female  rats.   According to  the authors,  "...due  to the  high  mortality
 rate  in  all groups of male rats, the terminal  necropsy for male rats occurred
 during the  21st month of exposure to  methylene chloride to  ensure  adequate
 numbers  of  surviving animals for pathologic evaluation."  This  is  not consistent
 with  the findings  in Tables 5-17 and 5-18.
      No  significant  effect on absolute  or  relative organ weight was seen in
 male  or  female rats.  Blood COHb levels  were  significantly elevated  (P  <0.05)
 above controls  in all  experimental groups of rats.   Incorporation  of 3H-
 thymidine into hepatic DNA was unaffected  in  female rats  at 6 and 12 mo.   DNA
 synthesis in  rats  was  not determined at  15  mo, as  originally  scheduled  in the
 protocol, due  to the number of  mammary  tumors observed in female  rats  at 200
 and 500  ppm.
      Results from  the  6-, 12-,  15-, and 18-mo interim necropsies showed no
 definite exposure-related gross  or  histopathologic  findings in male  and female
 rats  from any  of  the  interim  sacrifices.   An  exception  was the interim
 sacrifice of female  rats at 15  mo, where 1, 3, 4, and 5 females inhaling DCM
 at  0,  50, 200, or  500  ppm,  respectively, had a focus  or foci  of altered cells
 in  the liver.   This  effect was  not apparent in  female rats from 6-, 12-,  or
 18-mo interim  sacrifices,  nor  was  it apparent from rats dying spontaneously,
 killed moribund  during the study, or terminally sacrificed.
     There were  no significant histological lesions observed  in  other organs,
with  the exception of the  liver.   Data  on the  liver  lesions are  given in
Tables 5-19 and  5-20.  In males, the incidence of hepatocellular vacuolization
 increased slightly (Table 5-19).  The liver lesions in female  rats were signi-
ficantly increased for foci of altered cell, hepatocellular vacuolization, and
multinucleated hepatocytes at  500  ppm (Table  5-20) as compared  to controls.
The significance of these alterations in the liver is not known.  Further, the
number of liver  lesions appeared to be increased in the 500/0  group at terminal
sacrifice, combined sacrifice,  and death as compared to controls, but this was
not the case in the 0/500 ppm group.  There were no significant differences in
any tumor type for liver, kidney,  spleen,  brain, salivary gland, lung,  skin,
pancreas, and  mammary  gland  in male and female  rats,  with  the exception  of
mammary gland tumors, which were significantly higher in females (Table  5-21).
                                   5-65

-------
              TABLE 5-17.   MONTHLY MORTALITY DATA FOR MALE RATS IN
    A 2-YEAR DICHLOROMETHANE INHALATION TOXICITY AND  ONCOGENICITY STUDY (%)


                             DCM concentration (ppm)
Month
of
study
1
2
3
4
5
6
7
8
9
10
11
12
13
14
15
16
17
18
19
20
Terminal sacrifice
0
0*
0
0
0
0
0
0
0
1(1/85)
1(1/85)
4(3/85)
4(3/85)
5(4/80)
15(12/80)
20(16/80)
33(25/75)
37(28/75)
47(35/75)
59(41/70)
70(49/70)
74(52/70)
50
0
0
1(1/90)
1(1/90)
1(1/90)
1(1/90)
2(2/85)
2(2/85)
4(3/85)
4(3/85)
4(3/85)
5(4/85)
9(7/80)
13(10/80)
18(14/80)
20(15/75)t
37(28/75)
45(34/75)
64(45/70)
70(49/70)
73(51/70)
200
0
0
0
1(1/90)
1(1/90)
1(1/90)
2(2/85)
4(3/85)
4(3/85)
4(3/85)
5(4/85)
7(6/85)
11(9/80)
14(11/80)
23(18/80)
31(23/75)
37(28/75)
53(40/75)
69(48/70)
79(55/70)
81(57/70)
500
0
0
0
0
0
0
0
0
1(1/85)
1(1/85)
1(1/85)
1(1/85)
4(3/80)
8(6/80)
13(10/80)
23(17/75)
25(19/75)
41(31/75)
50(35/70)
66(46/70)
73(51/70)
^Percent mortality (number dead/original number of animals minus animals
 sacrificed for an interim necropsy).
tSignificantly different from control value by Fisher's Exact Test.
Source:  Dow Chemical Company, 1982.
                                   5-66

-------
                  TABLE  5-18.   MONTHLY  MORTALITY  DATA  FOR  FEMALE  RATS  IN
          A  2-YEAR DICHLOROMETHANE  INHALATION  TOXICITY AND ONCOGENICITY STUDY  (%)


                              PCM concentration (ppm)
Month
of study
1
2
3
4
5
6
7
8
9
0
1
2
3
4
5
6
7
8
9
10
11
12
13
Terminal
sacrifice
(after 24
months)
0
0*
0
0
0
0
0
0
0
1(1/85)
1(1/85)
2(2/85)
4(3/85)
4(3/80)
5(4/80)
9(7/80)
12(9/75)
15(11/75)
19(14/75)
30(21/70)
36(25/70)
43(30/70)
53(37/70)
60(42/70)



64(45/70)
50
0
0
0
0
0
0
0
0
0
0
0
2(2/85)
5(4/80)
5(4/80)
9(7/80)
15(11/75)
15(11/75)
17(13/75)
27(19/70)
36(25/70)
46(32/70)
53(37/70)
63(44/70)



76(35/70)
200
1(1/90)
1(1/90)
1(1/90)
1(1/90)
1(1/90)
1(1/85)
1(1/85)
2(2/85)
2(2/85)
2(2/85)
2(2/85)
2(2/85)
5(4/80)
5(4/80)
10(8/80)
13(10/75)
15(11/75)
21(16/75)
30(21/70)
34(24/70)
49(34/70)
54(38/70)
64(45/70)



67(47/70)
500
0
0
0
0
0
1(1/90)
1(1/85)
1(1/85)
1(1/85)
1(1/85)
1(1/85)
4(3/85)
6(5/80)
9(7/80)
15(12/80)
17(13/75)
19(14/75)
28(21/75)
37(26/70)
39(27/70)
47(33/70)
50(35/70)
54(38/70)



61(43/70)
0/500
0
0
0
0
0
0
0
0
0
0
0
0
3(1/30)
3(1/30)
3(1/30)
7(2/30)
7(2/30)
17(5/30)
24(6/25)
32(8/25)
36(9/25)
48(12/25)
60(15/25)



72(18/25)
500/0
0
0
0
0
0
0
0
0
0
0
0
0
0
0
0
ot
Of
3(l/30)f
8(l/25)t
8(2/25)t
16(4/25)f
36(9/25)f
48(12/25)t



52(13/25)
^Percent mortality (number dead/original number of animals minus animals sacrificed
 for an interim necropsy).

tSignificantly different from control value by Fisher's Exact Test.

Source:  Dow Chemical Company, 1982.
                                       5-67

-------
            TABLE 5-19.  NON-NEOPLASTIC LIVER LESIONS IN MALE RATS
                   Exposure level
                       Cppm)
                   Foci of
                altered cells
                Hepatocellular
                 vacuolization
Terminal kill
Moribund and
  0
 50
200
500
1/18
3/19
5/13*
7/19*
3/18
9/19*
7/13*
8/19
death



Combi ned



0
50
200
500
0
50
200
500
6/52
6/51
6/57
4/51
7/70
9/70
11/70
11/70
19/52
9/51
14/57
20/51
22/70
18/70
21/70
28/70
*Fisher's Exact Test, P <0.05:

Source:  Dow Chemical Company, 1982.
                                    5-68

-------
             TABLE 5-20.  NON-NEOPLASTIC LIVER LESIONS IN FEMALE RATS
                Exposure level
                     (ppm)
Moribund and
 spontaneous
                     500
                     0/500
                     500/0
   Foci of
altered cells
Hepatocellular
vacuolization
Multinucleated
  hepatocytes
Terminal kill


0
50
200
9/25
5/17
10/22
23/25
15/17
21/22
4/25
4/17
5/22
    17/27*
     5/7
     8/12
   23/27
    6/7
   12/12
     16/27*
      1/7
      6/12*
death





Combined





0
50
200
500
0/500
500/0
0
50
200
500
0/500
500/0
12/45
12/53
17/48
14/43
7/18
5/13
21/70
17/70
27/70
31/70
12/25
13/25
i 18/45
27/53
23/48
30/43*
9/18
4/13
41/70
42/70
44/70
53/70*
15/25
16/25
4/45
2/53
7/48
11/42*
2/18
3/13
8/70
6/70
12/70
27/70*
3/25
9/25*
*Fisher's Exact Test, P <0.05.

0/500 = rats exposed to 500 ppm DCM for first 12 months.

500/0 = rats exposed to 500 ppm DCM for last 12 months.
Source:  Dow Chemical Company, 1982.
                                     5-69

-------
          TABLE 5-21.   SUMMARY OF MAMMARY GLAND  TUMORS  IN  FEMALE RATS

Rats with a
benign mammary
tumor
(adenoma,
fibroadenoma,
or fibroma)
Total number
of benign
mammary tumors
(adenomas,
fibroadenomas,
and fibromas)
Exposure
level
(ppm)
0
50
200
500
0/500
500/0
0
50
200
500
0/500
500/0
Number
of rats
70
70
70
70
25
25
70
70
70
70
25
25
Moribund and
spontaneous
35
43
41
32
17
11
69
97
91
68
35
27
Terminal
kill
17
15
20
23
6
12*
36
36
44
79
15
33
Cumulative
52
58
61*
55
o o
23
23
105
133
135
147
ป- f\
50
60
^Significantly different from control when analyzed by Fisher's Exact
 Test, P <0.05.
tData could not be analyzed by Fisher's Exact Test.
0/500 = rats exposed to 500 ppm DCM for first 12 months.
500/0 = rats exposed to 500 ppm DCM for last 12 months.
Source:  Dow Chemical Company, 1982.
                                    5-70

-------
      In summary,  there were  significant  increases  in non-neoplastic liver
 lesions (i.e.,  hepatocellular vacuolization and multinucleated hepatocytes) in
 female rats  at 500 ppm.   There was an increase  in  benign mammary tumors
 (adenoma,  fibroma, and fibroadenoma)  in  female rats.   The number  of  benign
 mammary tumors/tumor-bearing rats observed  in  female  rats was 2.0, 2.3,  2.2,
 and 2.7 in rats inhaling  DCM at 0,  50,  200,  and 500 ppm,  respectively.   Female
 rats of group  500/0 showed  effects  that were similar to rats exposed to 500
 ppm for 24 months, but the 0/500 group did not differ from the control group.
      In conclusion, the results of  this study  offer very limited evidence  of
 the carcinogenicity of DCM.   However,  the highest dose  in this  study  is  half
 of the lowest doses in both the NTP gavage study (1982 draft) and the ongoing
 NTP inhalation  study.
 5-3.3.1.5   National Coffee Association  (1982a.b) study in rats.   On August  11,
 1982,  Hazelton  Laboratories  America,  Inc.,  reported  on  a  chronic  study  in
 Fischer 344 rats administered DCM in deionized  drinking  water for 24 mo.  This
 study  was  sponsored by the  National  Coffee  Association   (NCA), and  utilized
 1,000  animals in 7  different  dose groups or  regimens (Table 5-22).   The actual
 mean daily consumption  levels  of DCM  in the  drinking water were similar to  the
 expected target levels  (Table  5-23).
     Interim  sacrifices were performed at 26,  52, and 78 weeks  of treatment
 with 5,  10, or  20  rats of  each sex  from each dose group, respectively, with
 the  exception  of the  animals  in Group 7 (the recovery group, which  were only
 sacrificed with the remaining terminal  animals  at  104  weeks).  The  effects  of
 compound administration were evaluated using the following criteria:  survival,
 body weight gains, total food consumption, water consumption, clinical observa-
 tions, ophthalmoscopic  findings, clinical pathology,  organ and tissue weights,
 and gross  and microscopic pathology.
     No compound-related  findings were  reported for either survival, clinical
 observations,  opthalmoscopic findings, gross  necropsy findings,  or organ
weight  data.   Throughout  the study,   small but  significantly  decreased body
weight gains and water consumption were reported for both male and female rats
 in Groups  5,  6, and 7.  Food  consumption  also  decreased,  but  this  criterion
was  only  monitored for the  first 13 weeks.  These decreased  effects  were
attributed to DCM administration.   Low-magnitude statistical increases in mean
hemotocrit, hemoglobin, and  red blood cell counts were noted in both male and
female animals  of  Groups  4,  5, and 6.  In most cases, these were within the
range of historical control  values of Fischer 344 rats.
                                   5-71

-------
        TABLE 5-22.   GROUP ASSIGNMENT OF FISCHER 344  RATS  ADMINISTERED
           DICHLOROMETHANE IN DEIONIZED DRINKING WATER FOR 24 MONTHS

1.
2.
3.
4.
5.
6.
7.
Group number
Control
Control
Low-dose
Mid-dose 1
Mid-dose 2
High-dose
High-dose/recovery
(78 weeks/26 weeks)
Number
Males
85
50
85
85
85
85
25
of animals
Females
85
50
85
85
85
85
25
Target dose,
mg/kg/day
0
0
5
50
125
250
250
Source:  National Coffee Association, 1982a.
     TABLE 5-23.  MEAN DAILY CONSUMPTION OF DICHLOROMETHANE IN A 24-MONTH
          CHRONIC TOXICITY AND ONCOGENICITY STUDY IN FISCHER 344 RATS
Group
3
4
5
6
7
Target level,
mg/kg/day
5
50
125
250
250*
Males
5.85
52.28
125.04
235.00
232.13
Females
6.47
58.32
135.59
262.81
268.72
*The designated recovery group  (Group 7) mean  is  for the  first 78 weeks only.
Source:  National  Coffee Association, 1982a.
                                    5-72

-------
      Histopathological  alterations were  described in the livers of  rats  of
 both sexes in Groups 4, 5, 6, and 7.  These changes consisted of an increased
 incidence of foci/areas of cellular alteration in Groups 4,  5,  6,  and 7 and of
 fatty changes in Groups 5 and 6 after 78  and 104 weeks of treatment.  The
 incidence of  neoplastic  nodules and/or hepatocellular  carcinomas  in  female
 Fischer 344 rats (Table 5-24)  was  derived  from the data presented  in  Volumes
 I-IV of the August  11,  1982,  NCA report (National Coffee Association 1982a)
 and in the Addition  to  the Final NCA Report  of  November 5, 1982 (National
 Coffee Association  1982b).  Male rats  did  not show an  increased  incidence  of
 liver tumors in treated animals versus controls (Table 5-24).   These statist-
 ically significant  increases in the incidences of liver tumors in female rats
 were within the range of  historical  control values  at  this  laboratory (Table
 5-25),  as presented  in a  letter from Dr.  John Kirschman of General Foods  to
 Dr.  Dharm Singh of  the CAG,  dated  February 17, 1983.   Therefore,  based on a
 review of the NCA study,  DCM  administered  in  deionized water at  doses up  to
 250  mg/kg/day was borderline for carcinogenicity  to  Fischer  344 rats.
 5.3.3.1.6   National  Coffee Association  (1983)  study  in  mice.  In December
 1983,  Hazelton  Laboratories of America, Inc.  reported  on a  chronic study  in
 B6C3F1  mice administered  DCM  in deionized  water for 24 mo.  This  study,
 sponsored  by  the National  Coffee Association  (NCA), utilized 1,000 mice (650
 males  and  350  females)  in  six  different dose groups or  regimens (Table 5-26).
 The  actual mean daily consumption levels of DCM in drinking water were similar
 to the expected target levels (Table 5-27).   Each animal was housed individual-
 ly and identified by  a unique number.
     Adjusted survival for male mice at 104 weeks was 88.3% for Group 1, 76.6%
 for  Group  2,  81.5%  for Group 3,  81.0%  for  Group 4, 82.8% for Group 5,  and
 81.5%  for  Group  6.   For female mice,  adjusted  survival  at 104 weeks was  69.4%
 for  Group  1,  78.0%  for Group 2,  73.0%  for  Group  3,  84.0% for Group 4, 76.0%
 for  Group  5,  and 91.8% for Group 6.  No compound-related significant differ-
 ences between groups were found, but survival was slightly increased in Groups
4 and 6.
     There were  some exceptional  clinical findings,  i.e., convulsions  charac-
terized by  quaking,  extension  of the head,  and lowering of the pinnae of the
ears (peyer reaction).  In some cases the behavior progressed to full  seizure.
There was  no correlation to any increased mortality.  These  convulsions were
                                   5-73

-------






1—
ce.

5
coco
OS LU
LU LU
%^
CO "Cf
t-t O
U_ rH
I t 1 Qฃ
— 1 O
—4
=> z;
r- 0
i — t
OS LU
-< LU
CJ OS
Z LU
•— 1 1 —
CO
1 — 1
. z
^J* H"H
CM S
tn ซc

UJ
i
CO







S-
0)
0
u
CU
o;
O
in
CM



o
in •
CM



1? CM
"O rH
O)
^^
****.
cn
E
"**'
Q. 0
3 in
o
s-
cn

.p
ฃ1
a)
fcฃ
ro
CU in
l_



0






o










CO
•r—
CO
O
^™
cn
rO
Q











CD /•— . in
rH CD rH in
*^s ^_s \^^ป CM
•=f CD •^-



CM CM •?)-
rH rH CM LO
^^> v^^ ^*-^ oo
i-H rH CM



in '*^ m
CD
oo ^-s oo m
*~s CD ^— ' CO
CO CO




S-*. /— • N
•X  . in
CM O CM OO
CM" o" CM



S ^ rH 0
S^^ S^^ N*^ LO
tn c\j r^




*^™N ^""r** ^T*
LO
*^ c\j r^ oo
^J- 00 UD





fO
z i -o
— ^ ._. QJ

^^ S- CJ -r-
cu re) a: e
i — O ro
3 T3 X
T3 S- C CU
O (0 ro
C i — CO
3 Z S-
U i — Z CU
•r- i— >
-P CU T3 T—
CO O CU i —
rO O '""^ c
•— -P CJ -i- •—
o.  — ' CD **-J
CM CM


	 *^
t-~ <=t- rH
ซ^t* CM r*^ m
^ , 	 , v_^ oo
^J- CM CD



CM CM
l-i CD rH in
****s *^— ^ s**— ^ CO
rH 0 rH



_._
OJ /*~"N /-""I <^~^
r- ซ* >=1- 00
ro
E CM CM ซ* CO
co ^~s --- ' -~-^ oo
U_ CM CM -sf




/^-> /^^s
CM CM
s-*, • in
rH O rH 00
rH O rH



^ ^ _
CD CD CD 
CD CD CD





,— , / — , ^^ LD
CD CD CD OO
CD O O





ro
S
1 J "S
s^^ซ tj C
S- CJ •!-
CU ro ZC E
i — <_) rO
3 T3 X
-O i- c: cu
o re) ro
c ) — co
3 Z S-
0 i — z cu
-p CU ~O •<-
CO U CU i —
ro o ป — - c:
1— -P 0 T- r—
r*> rO in ซ* ro
o a.^—' e -P
cu cu o o
z 3: cj i —




t
1
CO
III]
J— ]
CD
O
CU
0
cu
T3
0
C
p^
o
S-
ง
u
"S
. g
c: -r-
O XI
•r- E
CO O
cu u
ro
re) CD
i — C.
3 -r—
' CJ CO
•i— 3

i- -P
ro co
Q. CO
CU
-p o
ro
i- X
O UJ
CO
cu -
c. cu
CU -C
"O co
• r- T~ *
O ' ' o

•r-' ai ro
JZ CM
-p -p oo
c: cn
CU >, rH
(_) X5
CU /— N ฃZ
Q. in O
CD *i—
•P • -P
•co ro
HI V •!-
CO CJ
CU Q- O
S- ซ — ' CO
Q. CO
CU -P , o
c ro rO
•r- U Z
•i—
co -P
S- CO ..
CU •!- CU
ja -p u
II E ro S-
3 +J 3
Z OO 0
•X •*<- 00
5-74

-------









OO
t—
ce
^J.
OO
ce
LU
3:
o
00
1 — 1
1 1
LU
— 1
LU
|__1 ,
o
1— 1 1 — 1
ซar
I-H  Cฃ
F— 1 O

LU
	 1
CO
^r—
T3 (O
13 4->
+-> 0
00 (—

-OM



l-H|rH







C3|i-H


CM

U_

i— 1



LU|i— 1


f—^> II-H




CJjrH



OQ|i— 1



CM

ซ=C


r-H




* • • •
•งปง•
3 0
+J S-
OO O











un CM

CD Cft "N|" LO I — 1
rH i-H
^*


"3" oo
oo CM LO i— i r-^
l-H I-H

00
r-. CM rH 1 I
i-H rH 1 1


O I I i i
*3" 1 1 1 I


cn i i i i
oo i i i i

10 i-H
ซ3~ "xf oo •! — i r^
i— 1 CM

i— i r- 1
i — 1 rH O*> i-H O">
i-H

OO OO
OO i-H CM rH CM
UD
OO 1 1 r-l CNJ
OO I 1



f— t III |
<• 1 1 1 1



^> 111 1
00 1 1 1 |

p^
o ^- ir> i i


oo
i i
r-* ir> oo i i
un


•a
Ct) Ol
C OJ
^ ^
n3 ^3 ^
X 0 (B
OJ C i —
0) 13 ro
(J U i— S  O 1— ( Q_ O ฃZ
i— i ai cu i— i
— i "z. 5-5 IE ^3





















































,
OO
oo
CD
i— 1
„
C
•d ro
,3 ^
s- u
O in
Q- S_

-------
   TABLE 5-26.   GROUP ASSIGNMENT OF B6C3F1 MICE ADMINISTERED DICHLOROMETHANE
                   IN DEIONIZED DRINKING WATER FOR 24 MONTHS
Group number
Control
Control
Low-dose
Mid-dose 1
Mid-dose 2
High-dose
Number
Males
60
65
200
100
100
125
of animals
Females
50
50
50
50
50
50
Target dose
(mg/kg/day)
0
0
60
125
185
250
Source:  National Coffee Association, 1983.
     TABLE 5-27.  MEAN DAILY CONSUMPTION OF DICHLOROMETHANE IN A 24-MONTH
             CHRONIC TOXICITY AND ONCOGENICITY STUDY IN B6C3F1 MICE
Group
3
4
5
6
Target level
(mg/kg/day)
60
125
185
250
Males
mg/kg/day ฑ S.D.
60.55 ฑ 7.680
123.61 ฑ 14.356
177.48 ฑ 19.214
234.29 ฑ 26.495
Females
mg/kg/day ฑ S.D.
59.46 ฑ 8.413
118.19 ฑ 16.799
172.41 ฑ 22.950
237.76 ฑ 29.329
Source:  National Coffee Association, 1983.
                                    5-76

-------
demonstrated in each  group  including the controls, at  least  once,  and were
noted during body  weight procedure when animals were  placed  in pans.  The
occurrence of convulsions in B6C3F1 mice is presented in Table 5-28.
     An increased  number  of Harden'an gland neoplasms were also observed in
males in Groups 4 and 6.   The other groups were similar to the controls (Table
5-29).  The increase  in  Groups 4 and 6 may be a result of normal biological
variation as suggested by the sponsors.
     No compound-related  findings were  observed  for either survival,  body
weight changes, water consumption, clinical observation, leukocyte counts,  and
gross necropsy  findings.   Food consumption was not done  in this experiment.
Histomorphologic alterations  of the  liver were observed in  both  male and
female mice in  the high-dose group  (containing greater amounts of oil  Red 0
positive  material).   Hepatocellular  lesions were observed in  all  groups  of
both  sexes, including controls.   The lesions in the treated female group were
comparable to controls (Table 5-30).   In males, there was also a statistically
significant increase  in  the incidence of hepatocellular adenomas and/or car-
cinomas in  Groups  4 and 5  and  a borderline significant increase in Group 6
(the  high-dose group).
      Based  on  the  results of this study, DCM administered in deionized water
at doses  up to  250 mg/kg/day  can be  considered borderline for  carcinogen!city
to B6C3F1 male mice.
5.3.3.1.7   National Toxicology  Program (1982 draft) gavage study in rats and
mice.  The  National  Toxicology Program (NTP) conducted a 2-yr carcinogenesis
bioassay  of food-grade DCM  and reported  on their  results  in a  draft  technical
report dated September 22,  1982  (National Toxicology Program, 1982).   Dichloro-
methane was administered in corn oil by  gavage  to male and female  Fischer
344/N rats  and B6C3F1 mice.  The DCM was  more than 99.5% pure, with vapor
phase chromatographic analysis detecting the presence of vinylidene chloride
and  trans-dichloroethylene  up  to 0.4%.   A 13-week  dose  finding study was
conducted to  evaluate the  toxicity  of the compound.  Based on survival, body
weight gain,  and histopathological  examination,  doses of 500 and 1,000 mg/kg
by gavage were selected  for male and female rats  and mice for the  2-yr study.
      The  NTP announced on July  25, 1983 that the draft NTP report would not be
issued as a final  report due  to discrepancies in experimental data that com-
promise a clear interpretation.  NTP further allowed that pending  the results
                                   5-77

-------
         TABLE 5-28.  OCCURRENCE OF CONVULSIONS IN MALE AND FEMALE B6C3F1 MICE
                Males
                                                 Females
                                                                    4
86.8   89.8
 5.4    6.3
       Percent with convulsions at least once
80.4   74.1   76.8   77.2      100.0   89.7   90.7   88.1
            Mean frequency of occurrence
 4.1    4.3    5.2    5.0        8.6    7.6    8.0    8.0
94.7   91.1
 7.1    8.3
Source:  National Coffee Association, 1983.
             TABLE 5-29.  , NUMBER OF MALE MICE WITH HARDERIAN GLAND NEOPLASMS

^Harderian gland
Adenoma
Carcinoma
Percent
Adenoma
Carcinoma
Combined
Group: 1
Number examined: 60

3
, 0

5
0
5
2
65

2
2

3
3
6
3
200

13
0

7
0
7
4
100

9
0

9
0
9
5
99

5
0

5
0
5
6
121

11
0

9
0
9
     Source:   National Coffee Association, 1983.
                                        5-78

-------
o
co
o
CD
CO

LU co

 UJ
—I l*s|
—I I—I
LU Z.
O O
O I-H
f— UJ
 z

H- 3C
LU •&,
LU O


JO
O -J

CL. U
   l—i
LL. O
O

UJ LU
cj ca.
Q co
i—ซi—)
o z
   a
   ซ3C
CD
CO
 I
in

LU

en

                   o
                   o
           o
           S-
+J

i
+J
ro
cu
                fo
                   o
                CM
                   in
                   
               cu

              i
S1
o
iH

co
iH
g
rH
NM*'

O
iH
9


^r




ฃ$
r^*>

"^s

ซ^j*
rH




f*^\
<5^
O^

**fc^*

CD




/*"""N
*Mฃ
pซ-ป

V**ซ/


CO

\**t

ir>




















rd
ps
0
c
•I~
u
s-
ro
0

i-
(0
p—
3
r—
r—

CO
cu
f-
+J
u
(0
X
UJ

S-
cu
t~
CO
u.
fe? ฃ? S?"
CO CM <^-


co rH CM

i 9 S

V-^ >ta^ SM^

0 CM CO

g g g

*c ^^^
CU rH rH rH
r-~
(0
E
CU
fcS S^ ^


"ซซ— ^ Sw*' S*^

CO CO iH





^*^s ^™*\ /"™N
S^ Sซ &9
0 •* CM

VH^ Sซ*iซ^ *SM*1'

O CM rH




*•*"% c*"*% ^*N
^? ^ ^S
CM CM S#
rH
V-^ ^^-^ X—X

CD rH CM




















ro
ps
ro o
E ฃ=
O •!—
C U
CU i-
TJ (0
rO U

S~ i-
ro ro
f~~ f—
ro 3 3
•I— r— r—
CO I— < i —
ro cu cu
r- ซJ O
CL O 0
S- -P -P
co ro ro
CL Q. O.
>> CU CO
O
CO


CO

i
rH
V**/

LO

?


CM




&•?
ซ^

SM^

•at"





/^"S
5*^
Vฃ>

V^

co




/""N
&?
CD

v^^

CO




















ro

o

*r—
U
i-
ro
u

S-
o

TJ
C
ro

ro
1
c
cu
TJ



















•
C
0
•r-
10
CU


S-
ro
r™
3
U
*^_
•P
S~
CO
CL

cu
•P

S-
o
<<-

CD
u
c
cu
TJ

U
c
*lป
4->
c
cu
u
ฃ-
CO
O-

c
cu
to
cu
^.
CL
CU
i.

4
r— •
r—
ro
o

4->
CO
+J
ro
-P





















































CO
CO
cn
rH

ซrt
C
0
•r-
^^
ro
•r-
(J
O
to
to

-------
of an in-depth audit, select and relevant information from these gavage studies
might be incorporated into the future draft technical report for DCM inhalation
studies.
5.3.3.1.8  Theiss et al.  (1977).   A pulmonary  tumor bioassay  in  mice was
reported by Theiss et al. (1977).  Groups of 20 male strain A mice were injected
intraperitoneally three  times a  week with 0, 160, 400, or 800 mg/kg DCM for a
total of 16  or 17 injections.   Mice were sacrificed 24 weeks after the first
injection,  and the lungs were  examined under  a  dissecting microscope for
surface adenomas. Some adenomas were confirmed by histology.
     Tumors were found at all three dose levels; however, due to poor survival
and the small  number of  animals, the increase in  tumors  did not reach  statis-
tical significance at the two highest dose levels (Table 5-31).   At the lowest
dose, a highly significant  increase in the number of tumors was observed (P =
0.013).  Therefore,  this  study was marginally positive  for carcinogenicity.

             TABLE 5-31.   PULMONARY TUMOR BIOASSAY IN STRAIN A MICE
Dose
(mg/kg)
0
60
400
800
Total dose
given
0
2,720
6,800
12,800
No. of mice
at beginning
20
20
20
20
No. of mice
examined
for tumors
15
18
5
12
Tumors/
mouse
0.27
0.94
0.80
0.50
Significance*
—
<0.013
>0.1
>0.1
Source:  Adapted from Thiess et al., 1977.
*The test of significance used is the exact test of ratio of two
 Poisson parameters.

5.3.3.1.9  Heppel et al. (1944).  Heppel  et al.  (1944) exposed  dogs, rabbits,
guinea pigs, and  rats  to DCM by  inhalation at levels of 5,000 ppm (17,350
    3                                           3
mg/m) for 7 hr/day and 10,000 ppm (34,700 mg/m ) for 4 hr/day, 5 days/week,
for 6 months.   No tumors developed in any animals.
5.3.3.1.10  MacEwen et al. (1972).  MacEwen et al.  (1972) exposed dogs to DCM
by inhalation  at  500 ppm (1,735 mg/m  )  for 14  weeks;  no  tumors  were reported,
but edema of the  meninges of the brain occurred.  Neither this study nor the
                                   5-80

-------
Heppel et al.  (1944) study could have detected a carcinogenic response because
of the shortness of the observation times and the fact that these studies were
not originally designed to test for carcinogenicity.
5.3.3.1.11  Other animal studies in progress.  The National Toxicology Program
sponsored a 2-yr  inhalation  study in male and  female  Fischer 344/N rats and
B6C3F1 mice at  exposure concentrations of 0, 1,000,  2,000, and 4,000 ppm (0,
3,470, 6,940,  and 13,880 mg/m3)  for  rats  and 0, 2,000, and 4,000  ppm (0,
6,940, and 13,880  mg/m )  for mice.  The animals  have  been  sacrificed  (April
1983), but the pathology report is not yet available.
5.3.3.1.12  Cell transformation studies.   Price et al.  (1978) exposed  Fischer
rat embryo cell cultures (F1706, subculture 108) to DCM liquid at concentrations
of 1.6 x 102  and 1.6  x 10   uM for 48 hr.   Dichloromethane was diluted with
growth medium  to  yield the appropriate doses.  The DCM sample, obtained from
the  Fisher  Scientific  Company,  was  >_99.9% pure.  The  cells were grown in
Eagles minimum  essential  medium  in Earle's salts  supplemented with  10  percent
fetal bovine  serum,  2 mM  L-glutamine,  0.1  mM nonessential  amino  acids,  100  ug
penicillin, and  100  ug streptomycin/ml.  Quadruplicate cultures were treated
at 50 percent confluency with each dose.  After treatment, cells were cultured
in growth medium  alone at 37ฐC.  Transformation of cells treated with either
dose  level of DCM was  observed by 23 and 30 days of incubation, and was charac-
terized  by progressively growing foci composed  of cells lacking contact inhibi-
tion  and orientation.   There was  no  transformation  of cells grown  in  medium
alone or in  the presence of a  1:1,000 acetone concentration,  even after  a
subculture. Twenty  and 27 microscopic foci per three dishes with the  low and
high  DCM doses respectively, were  found in dishes inoculated with 50,000 cells
from  cultures treated  four subcultures earlier  and held for 4 weeks at 37ฐC in
a humidified C00  incubator before  staining.
                                                          2
      Subcutaneous injection of cells treated with 1.6 x 10  uM DCM  five subcul-
tures earlier produced local fibrosarcomas  in  5/5  newborn Fischer 344 rats
within  60 days following treatment.   The  ability of cells grown  in  growth
medium alone to induce local fibrosarcomas was  not determined; however, negative
responses were obtained with cells grown in  the presence of a 1:1,000  concentra-
tion  of acetone.   Exposure of  cells  to 3.7 x  10~   uM 3-methylcholanthrene
produced 124 microscopic  foci per  three dishes  in the  inoculation test described
above by 37 days of  incubation,  and local fibrosarcomas  in 12/12 rats by 27
days  following subcutaneous injection of cells.  The exposure  of 3-methylcho-
lanthrene was attained by initial dilution  in  acetone to  1 mg/ml followed  by
                                   5-81

-------
 further dilution in growth medium  to  0.1 pg/ml  (personal  communication from
 Or.  P.  J.  Price).
     Dr.  Price wrote a  letter to the CAG  dated Nov. 14, 1980, saying that "the
 analysis  of methylene chloride showed a purity of 99.9 percent.   The  original
 study  was done in quadruplicate and  in each case the Fischer rat cells were
 transformed.   Since  the publication,  the same batch of methylene chloride was
 sent to Andy Sivak at  Arthur  D.  Little to be run against  the Kakunago clone
 A31  of BALB/c 3T3.  It  did  not transform his cells.  We  then  repeated the
 study  in  Fischer rat cells and  at the  same time tested methylene chloride sent
 to  us  by  the National  Coffee  Producers  Association.  The test was run in
 triplicate.  The  Fisher methylene chloride again  transformed the cells, while
 the  National  Coffee  Producers'  (supplied by  Diamond  Shamrock) was negative."
 The  differing responses  in the  two  experiments may  have been due to differences
 in the  levels of impurities present in  the samples  used.   The chemical  composi-
 tions of  DCM  samples from different suppliers are given in Table 5-32.
     The  Fischer rat embryo cell line contains  the genome of the  Rauscher
 leukemia  virus,  but  there is no basis for minimizing the positive results.
 Since  the mode of action of DCM is not known, this transformation may  be due
 to activation of the virus.
 5.3.3.2   Epidemiologic  Studies
 5.3.3.2.1  Friedlander  et al. (1978),  Hearne  and  Friedlander (1981).  Fried!ander
 et al.  (1978) performed mortality  analyses  of  male Eastman-Kodak employees
 exposed to low levels of DCM.   This study was updated by Hearne and Friedlander
 (1981).   Measurements  from  1959 to 1975  ranged from  30 to 120 ppm (104 to
        2
 416  mg/m  ).   Both  the  original  1978 analysis  and  the  1981  update  found no in-
 crease  in neoplasms,  heart  disease, or any other  cause of  death  in  comparison
with the  two control  groups, which  were composed  of other  Kodak  employees and
 of New York State males.  The population was  relatively stable and the workers
were rotated throughout the work area,  and thus exposure was averaged among all
the workers.  Dichloromethane had been  used for 30 years as the primary solvent
 in this Eastman-Kodak operation.
     Two  separate  mortality  analyses  were done.   In  the earlier paper, one
 approach  used the proportionate mortality ratio to  examine  334 deaths of
 DCM-exposed workers during 1956 to 1976.  Seventy-one neoplasms were found;  73
were expected based on other Kodak employee mortality ratios.   Furthermore,  no
 single site was over-represented.
                                   5-82

-------
     TABLE  5-32.   CHEMICAL  COMPOSITIONS  OF  DICHLOROMETHANE  SAMPLES (ppm);
                          Fisher D-123
                            Lot 761542
Diamond Shamrock
                                                                      Dow
Methyl chloride
Vinyl chloride
Ethyl chloride
Vinylidine chloride
Carbon tetrachloride
Chloroform
Trichloroethylene
1,2-Di chlorethy1ene
Methyl bromide
Cyclohexane
                                 0.5

                                 0.8
                               329
                                 3.6
                          3
                         33
                               369
                                                    26
      20
                                                   300
 <5

 86
 11
305
Presented by Drs.  Sivak and Kirschman at the Science Advisory Board
 Meeting, Sept.  4-5, 1980.
bFisher sample used by Dr.  P.  Price in two series of cell transformation
 studies.
cThis material is under test in NCA's chronic rat study on DCM in drinking
 water.  It was also used in Dr. Price's follow-up study.
dThis material, also analyzed by NCI, with the same results as given by
 Drs. Sivak and Kirschman,  was used in the following tests:
     NCA's 90-day studies in rats and mice
     Dr. Sivak's neoplastic transformation assay
     Dr. P. Price's follow-up series of cell transformation tests
     NCI's chronic bioassay studies in rats and mice presently under way
                                   5-83

-------
      A second approach, included in  the  first paper and used exclusively in
 the  update,  was a cohort mortality study of all 751 employees who were in the
 DCM  work  area in  1964.   The  results of the  update are shown  in Tables  5-33 and
 5-34,  taken  from  that paper.  There were  110 deaths  during the 16-year follow-
 up (retrospective).  Two control groups were used:   other Kodak employees, and
 New  York  State males.    The  expected  numbers  of deaths  in the exposed groups
 based upon the control  group experiences were  105  and 168,  respectively.   The
 differences  between the observed and  the  expected deaths based on the  controls
 are  either not statistically significant  (other Kodak employees) or the expec-
 ted  deaths based on New York State males are  significantly  increased  over the
 observed  numbers.
      The  results  show  that malignant neoplasms  accounted  for 24 of the 110
 deaths  in the study cohort,  which was  less than the 28.6 or 38.5  expected
 malignancies  based  on  the control data.  Nine of  these 24  deaths were from
 respiratory  cancer  (7.6 and  13.6 were expected based on the control  groups)
 and  seven were from cancers  of  the  digestive organs (less  than  expected).
 Only the  two deaths associated with  brain  or nervous tissue represented  a
 higher than  expected total (SMR = 169 and SMR =  227  versus two control groups),
 but  these  SMRs  (standard mortality ratios) were  not  statistically significant.
      Further  stratification  of  the cohort focused on  the  252 males with 20
years  or  more of exposure  who were employed in 1964.   In this group 59 deaths
 occurred:   13 were due to malignant neoplasms  (17.8  and  24.7  expected  based on
 the  two control groups)  and  32 were due to circulatory diseases (37.9  and 59.7
 expected).
     A further analysis of the 252 males shows  that this cohort had a median
 age  of approximately 54 years in 1964.  With this group the more common cancers
would  have to be markedly increased for there to be a reasonable probability
 of detecting the  increase.   For example, following  the  cohort for 16  years,
 cancer mortality  at  this age would require 10 deaths from respiratory cancer
to detect a  significant result at the P  =  0.05 level.  This  represents  an
 increase  of  at least 100 percent over that  expected,  the expected probability
 !*
 least 100  percent over  that  expected, the expected probability of lung cancer
death for this cohort  being  0.018 over  the 16 years, based  on other  Kodak
employees' rates.  The  statistical power to detect 100-percent increases (at a
= 0.05, one-sided) is about 95 percent for all malignancies and 45 percent for
respiratory  cancer deaths.   The  remaining 499  males with less than 20 years
                                   5-84

-------


^
•a:

ฃ2
**^
^T
o
Oฃ
U_
l/}
K—

0
3Z
0
O
LU
*3>ป
<:
3Z
K-
LU
2:
o
0ฃ
3
3:
i — i
a
LU
_i

^—
r^
:D
o
JC

S
CD
rH

*t
0
00
en
I— 1
1
s
cr>
,H

*i
to

ee
LU
O

a
LU
r—
O
LU
Q_
X
LU

1=1
•z.

ce
LU
to
CO
o


,
00
CO

ir>

LU
_i
CO
i
o
CM

E
3
E

c
• ^
ฃ
-a
01
in
O
a.
X
ai

s-
o
f""
o
CJ








































. 13
O OI
ja 01
to' a.
>- X
Z OI




13
. o>
O -P
a*
ro Q.
a. x
iฃ Ol




13
0)

O i-
•z. 01
in
JO
O




• TD
O OI
C "tJ
r> 01
to Q.
>- X
Z OI







T3
• 01
O -P
C L)
01
ซ Q.
a, x
^ o>






•Q
at
. >
0 S-
Z 01
in
J3
0

















Q.
3
o
S-
cn

u

^J>
in
o
c:
D)
ro

0


O I
•51"
Lf)
00







^^*
VD
oo
CM









^*
CM







JD I
CM
1 —
,3-
CM










r--

r-.
rH











CO
rH

















in
E

ro

a.
o
OI
c

•p

jrj
f"
O)

r—
ซ



J I
O
<7>
O







f— 1
"H
cn
LO









CM
UO







J I
CO
f^~
cr>
ur>










i— i

r—
CO











CM
CO
















in
01
in
rO
01
in

'•S

>>
s-
o

(rj
P—
3
U
S-
•r—
f ^


J

CT>
ID







CO
"*
CO
ซ*•









00
^^







J
o
in
Lf)
•=1-










*4~
CO
1 —
CM











r^.
CM












OI
in
ro
OI
in
't-
is

•P
S-
(O
Ol


U

E

r"
O

(— l




U

rH O  00 00
CM rH rH JO
rH rH O
rH









P*** ^f" LO P*"* *^t~
m  co CM en
o ro CM *d~ LO
rH ID











in un CM r*"> cr>
in
















in
>) 0)
s- in
o ro
•P 01 in
ro in a)
i — •!— in
3"O 3
O ro
s- >> u
•r- S- in
U O in S- 01
-P -P o> in
S- (O C Jd 3
Ol t- Ol -P (0
jz: .r- -a o u
•P CL-r-
o in u i — i —
O) O i — i —
rv* ^fj ซ^ ซ^































CD
rH
U3

rH

vx
S-
(O
Q_
sx
ro
•o
O
~*^*

*+—
o

w
o>
1-

<*
f~~
Ol
E

r— •*
S-
3
O
JC

if—
O

>^
1 ^
•r-
t—
ro

S-
0
E

u

14—
•r"
(J
01
a.
in

X
o>
in
i
0)
O)
m

<™
o

•o
ai
in
ซ
OQ
ro .






.
in
en
iH

Co

en
rH

^
LO
en
rH

U)
S-
ro
Ol
^>^

Ol
f-

S-
o
s
•r*
O
^
S-
o


O)
z

O)
(Z
•f"
•a
Z3
f—
U
O)
\ซ/

a>
40
(0
^>
to
VX
S-
o
>—

3E
01
Z

>^
4^
• r—
r—
ro
+J
l-
0
E

U
•t—
4-
• r—
U
0)
D.
in

X
ai
m
i
O)
o>
ro

c:
o

13
CU
m
ns
CO
Q


















































,
/ 	 N
ID
CO
.
0
V 1
Q_
v^^1

4J
c:
ro
U
• r—
M-
• !—
C
O)
ซr-
in

in
01
u
ฃ=
Ol
S-
Ol
If —
4ซ
• r—
•a

•o
01

u
Ol
a.
x
ai
i
-a
01

S-
01
in
ja
o
o
































































,
rH
oo
cr>
i— i

•ป
j_
O)
13
C
ro
r—
13
ai
•r-
S-
u_

-a
(~*
ro

ai
c:
s-
ro
O)
~T"


, .
CU
o
L
3
o
to
5-85

-------
ง
en
rH
 I

ID **"

rH a
   O
 ac
rvป Q
LU _ J
tO -E
ca o
oป—<
   o

tO LU
s: _i
(X >-
o —J
LU CU


^1
CO
 I
LO

LU

ca

f^




40
ซ.
o
o
u
^
o
u

s
cn
rH
'co
•P
0
'







in
s-
<0
CD
O
CM
ฃ
3
•r-
C
•r-
T3
01
in
o
Q.
X
4J
S-
0
J=
o
o




























o 01
C I *
u
ja 01
to a.
>- X
Z 01


T3
O -P
C( *
U
CD
co a.
Q- X
*ฃ Ol


T3
0)
0 S_
z 01
in
JD
O
. T3
O CD
C -P
-Q CD
to a.
>- X





T3
• 0)
O -P
(Z U
fl3 f^f
Q. X
s^ Q)





•a
01

o s-
Z 01
in
o










a.

o
S-


u
•ป—

in
o
!=
co
CO
a
u
oo^f^'CM'tf'COLor*-'*
iDCMซ4'VDcncoซ*cnLO
rHCMcnco'st-ococooo
rH rH CO






COVOr^tDCOCOOOlD"*-
CncOLOLOCMrHlDOlD
OCMCOI~ซCOrHCMCOOO
rH CM





f^COซ3-cnrHCMrHซi'ซ3"
CM



u a
COOOLOt^-CO-*OOCM
ps, ^" (^j ijQ ซ^ ^^ cj <^" I^H
r^l-HlOOOCOOCMCM-sl-
CM








co^i"'5}'OcncMi<5t''^r^*
OCOr~.lDrHtDซ!fCOr~
|-^t—lLOซ^-CMC3rHrHI^
rH









LOCMCOซ3-OrHOCOCO
rH-


0
+J U) W
01 E E
in -r- 
3 0 CO CO
o a.!— i—
> o a. Q.
(-•POO
CD It) CD CD
C E C C
in ai
C S- J= +J P
co CD c c:
CO ^>-C *O CO CO
s- s- p c c c
O >) CO O CO CO CO
U> S- C T~ -r-
aico o -i- ia o i— i—
>OI +JS-C-i-cO(O
•1-S-S-C03CO-PEE
H-> O CD S- O CO
in cj=T-4-> cj= s-i—
CD ro P C3.-1- T- a. 01 co
coa. o u) c >+-> O
Q Qฃ O CO — 1 O 1—


ID

cn
j_4
1
ID
cn
rH
*2
s-
rO
OL.

_V^
CO
T3
O

co
03
co
LO
cn
rH
cn
rH
LO
ID
cn
rH
in
(0
O)
O)
.ฃ=
i-
o
<4-
/•^
-P*

S-
o
>-ซ

S
Ol
z

Cn
•5

u
X
Ol

cu
•p
CO
•p
1/5
v^
s-
o


25
Ol
z
-J^
•r—
lo
s-
o
E

O
•r—
M-
U
CD
O.
in

X
01
in

CD
co
co
=
o
•o
CD

CO
03
_Q






















•
LO
O

O
VI
Q.
^^

•p
C
CO
U
•r-
*4—
•r—
C
•r*

O
CD
Q.
X
Ol
73
Ol
S-
Ol
in
_o
O
o



































•
00
cn
rH
n
0)
T3
,C
to
r-*
"O
0)
il
u_

T3
C
CO

Ol
c
S-
CO
Ol
mz

oi
U
S-
3
O
to
                              5-86

-------
 exposure were  significantly younger  (median  age  about  36 years), and  a  follow-up
 of this cohort for 16 years  mortality might fail to  detect  even  a moderate
 effect, since  expected cancer mortality in this  age  group  is  so  low.
 5.3.3.2.2   Ott et al.  (1983a,b,c,d,e).  Ott et  al.  (1983a,b,c,d,e)  reported
 the results of a health evaluation of employees of one fiber production plant
 who were  exposed to DCM  as part of  a solvent mixture  consisting of this  sub-
 stance plus methanol and  also acetone in a separate  container.   A  second  fiber
 production  plant utilizing only  acetone, but  similar  to the first in  other
 respects,  was  selected as a  referent population.   The investigation  focused
 primarily  on health effects  occurring to the  cardiovascular system stemming
 from the metabolism of DCM to COHb in the body.   In  addition  to  a  retrospective
 cohort mortality study,  the  authors  examined several  other health end  points
 in the still-employed group.   These  involved clinical  evaluations, electrocar-
 diographic   monitoring, metabolism tests, and  evaluation  of oxygen  half-
'saturation  pressures.
                                                                        q
      Environmental  exposure  in the plant varied from  140  ppm (486 mg/m ) in
                                            3
 areas of  low DCM use to 475 ppm (1,748 mg/m )  in areas of  high DCM use, based
 on an  8-hr time-weighted average (TWA).  Methanol  was present in smaller
 quantities  by  a  factor of ten,  while  acetone ranged  from 100  to  1,000 ppm (347
 to 3,470  mg/m3) TWA in both  plants  (Ott et al., 1983a).   Industrial  hygiene
 surveys were conducted from September 1977 to February 1978.
      To qualify for inclusion  in  the cohort,  production employees (both men
 and women)   had to  have worked a minimum of 3 months  in  the preparation or
 extrusion  areas of either plant  during January 1, 1954,  to January 1,  1977.
 In the DCM plant, both cellulose diacetate  (acetate) and cellulose triacetate
 (CTA) fibers were made side  by side.  Although acetone was  present  in both
 plants, it  was the  solvent of choice  for making  acetate fibers in  the referent
 plant.  The exposed cohort consisted of 1,271  persons versus 948 persons in
 the referent plant, as follows:
    White men
        women
PCM plant
    487
    615
plant
 696
 248
 Nonwhite men
        women
              Total
     64
    105
   1271
   1
 	3
 948
                                    5-87

-------
Persons were  followed through June of 1977.   The  authors noted that vital
status was  not  available for 226 (18 percent) of the DCM-exposed cohort and
112  (12  percent) of  the companion plant.  The authors  commented  that  few
deaths would  be added from this  last group based on "previous experience with
the  social  security follow-up mechanism."  Such a  statement is subject to
question, however, without knowing the age distribution of this group.
     The authors  found no excessive mortality  from any cause in the "exposed"
or the "referent" population, either when the  group with  unknown vital status
was  presumed  lost to  follow-up  at the time lost,  or was presumed alive  until
June  of  1977; i.e., person-years would cease  accumulating  at  the time the
person was  lost to  follow-up in the former instance, but in the latter  case,
person-years  would  continue to  accumulate  until  the end of the follow-up
period,  as  if the individual were still  alive.  Altogether in the exposed
category for  white  men, 37 observed deaths out of 487 were  seen versus 34.9
expected based  on the latter definition,  or 30.4 expected based on the former
definition  regarding  the group  with unknown vital  status.   For white women,
only  11  deaths  were observed versus either 15.9 or 13.5, depending upon the
choice of the first or second definition above.  For malignant neoplasms  in
white men,  there were five observed deaths versus either  6.3 or 5.6 expected.
In white women, two observed deaths were seen, versus either 5.2 or 4.5  expec-
ted.  No cancer deaths were reported in nonwhite males and females,  probably
due  to  the small size of these select subgroups.   Since the  authors were
mainly  interested in  cardiovascular  effects,  they  examined further only
ischemic heart  disease in terms  of duration of exposure  and  length of follow-
up.   Even when  one  considers only individuals with a minimum of 10 years  of
exposure who .were followed for a minimum  of 10 years, only 2 male deaths were
observed versus  1.7 expected based On this cause.  Ischemic  heart disease  was
not  observed  as a cause of death in women.   No corresponding data are avail-
able  for any form of cancer by latency or duration of employment.
      Several qualifications limit-the possible use of this study as a sensitive
indicator of mortality.  Foremost among these  is the relatively low and unusual
distribution  of mortality  among the members of the  cohort.   Because of the
excessive numbers of observed and expected deaths from external causes compared
to the numbers  of observed and expected deaths from malignant neoplasms, and
the  fact that deaths  from accidents are  the  leading cause of death in young
males nationwide, it  is quite likely that the  cohort is  a relatively youthful
                                   5-i

-------
group.  This  is further evidenced by  the  surprisingly few deaths observed
overall compared to  the size of the cohort—less than 10 percent  In all  In-
stances (white  men,  nonwhite men,  and white females).  No age breakdown  was
provided to evaluate this observation.
     Additionally,  only 310 of the  exposed  males  have had a chance  to be
followed at least 17.5 years.  Some 241 exposed males entered the cohort after
1960 and could not possibly have been followed 17.5 years.   Because of the 15-
to 20-year  latency period  involved with most human  cancers,  cancer  effects
attributable to DCM exposure would probably not have been expected to manifest
themselves prior to  the 17th year.   Thus, the power  of this study  to detect a
statistically significant elevated  risk of cancer (as well  as ischemic heart
disease) is low.
     Another possible problem deals with the extent of follow-up of the cohort.
Almost 18 percent (226) of the exposed cohort was lost to follow-up as of June
1977.  Although the  authors discount that as unimportant,  it should be  of
concern that if the ages of the lost-to-follow-up group are relatively advanced,
the  likelihood  is  great that enhanced mortality will  be  in  the higher age
groups.  Whenever  extraordinary means  are employed  to determine the vital
status of a  subgroup of the cohort  for which all other methods of follow-up
have failed, the residual deaths found as a result of this endeavor are usual-
ly overly represented by sudden deaths due to heart failure or accidents.   The
opportunity for  leaving a  record  of their deaths is minimized because of the
nature of the deaths;  hence, it becomes less likely  that the vital status can
be determined.
     Still another problem with this study is the "healthy worker" effect.  In
most studies of this kind,  workers at the time of their employment and shortly
thereafter are  generally somewhat  healthier  than the population from whence
they  came.   Ill or  infirm  persons  do  not usually choose jobs that  may be
detrimental to their health.  This tendency usually results in as much as a 20
percent deficit of mortality compared to that expected.
     In summary, this  study is inadequate to assess cancer mortality in  the
described cohort for the reasons stated.  The study results do not exclude the
possibility of  increased health  risk  in the study  population.   The study
focuses mainly on heart disease as a consequence of DCM exposure.
5.3.3.3  Quantitative  Estimation.   This quantitative section  deals with  the
unit risk for  DCM  in air and  water  and the potency  of DCM relative to  other
                                   5-89

-------
carcinogens that the CAG  has evaluated.  The unit risk estimate for an air or
water pollutant is defined as the incremental  lifetime cancer risk occurring in
a hypothetical population in  which all individuals are exposed continuously
                                                                    o
from birth throughout  their lifetimes to a concentration  of 1 ug/m  of the
agent in the  air  they breathe or  to  a concentration  of 1 pg/l in the water
they drink.  This calculation provides a quantitative estimation of the impact
of the  agent  as  a carcinogen.   Unit  risk  estimates  are used to compare the
carcinogenic potency of  several  agents with each other and  to give a crude
indication of  the  population  risk that might be associated with air or water
exposure to these agents, if the actual exposures are known.
5.3.3.3.1   Procedures for the determination of unit risk for animals.   The
data used  for  the  quantitative estimate are taken from  one  or both  of the
following:  1)  lifetime animal  studies,  and 2)  human  studies where excess
cancer risk has been associated with exposure to the agent.  In animal studies
it is assumed, unless  evidence exists  to the contrary, that  if a carcinogenic
response occurs at the dose levels used in the study,  then responses will  also
occur at all  lower doses, with an  incidence determined  by an extrapolation
model.
     There is  no  solid scientific basis for any mathematical extrapolation
model that relates  carcinogen  exposure to cancer risks at the extremely low
concentrations that must  be dealt with in evaluating environmental hazards.
For practical  reasons,  such low levels of risk  cannot be measured directly
either by animal  experiments or by epidemiologic studies.   We must, therefore,
depend  on  our  current  understanding of the mechanisms of-carcinogenesis for
guidance as to which  risk model to use.   At  the present time, the dominant
view of the carcinogenic process involves the concept that most cancer-causing
agents  also  cause  irreversible damage to  DNA  and  are mutagenic.   There is
reason to expect that the quantal type of biological  response, which is charac-
teristic 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  higher doses,  there can be an
upward  curvature,  probably  reflecting the effects of multistage processes on
the mutagenic  response.   The  linear non-threshold dose-response relationship
is also consistent with  the relatively few epidemiologic  studies  of cancer
                                   5-90

-------
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).   Also, some evidence from animal experiments  is
consistent with the linear non-threshold model  (e.g., liver tumors induced in
mice by 2-acetylaminofluorene  in  the large scale EDQ-, study at the National
Center for lexicological  Research, and the initiation stage of the two- stage
carcinogenesis model  in rat liver and mouse skin).
     Because its scientific basis, although limited, is  the best of any of the
current mathematical  extrapolation models, the  linear non-threshold model  has
been adopted  as the  primary  basis for risk extrapolation  in  the  low-dose
region of the  dose-response  relationship.   The risk estimates made with this
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.
     The mathematical  formulation  chosen to describe the linear non- threshold
dose-response  relationship at  low doses  is the linearized multistage model.
This model employs enough arbitrary constants to be able  to  fit almost any
monotonically  increasing  dose- response data,  and  it incorporates a procedure
for estimating the largest possible linear slope (in the  95 percent confidence
limit sense) at low extrapolated doses that is consistent with the  data at all
dose levels of the experiment.
     5.3.3.3.1.1  Description of the low-dose animal extrapolation  model.   Let
P(d) represent the lifetime  risk (probability) of cancer at dose  d.   The
multistage model has the form
P(d) = 1 - exp [-(q
                              Q
where
Equivalently,
where
                                              qd
      Pt(d) = 1 - exp [-
                                        - P(0)
                           qi > 0, i = 0, 1, 2, ..., k.
                                        + q2d2 + ...  + qRdk)]
is the extra  risk over background rate at dose d or the effect of treatment.
                                   5-91

-------
     The point  estimate of the coefficients q.. ,  i  = 0, 1, 2,  .. . ,  k,  and
consequently the extra  risk function Pt(d) at any given dose d, is calculated
by maximizing the likelihood function of the data.
     The point  estimate and the 95% upper confidence limit of the extra risk,
P,(d), are calculated by using the computer program GLOBAL79,  developed  by
 o
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 qr  When-
ever q, > 0, at low doses  the extra risk Pt(d)  has  the approximate form P^Cd)
= 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. The
upper limit, qฃ,  is  calculated by increasing qI to a value q| such that when
the log- likelihood is remaximized subject  to this  fixed value, qฃ,  for the
linear  coefficient,  the resulting maximum  value of the  log- likelihood 1^
satisfies the equation
                               2 (l_0 - L^) = 2.70554

where 2.70554  is  the  cumulative  90%  point of the  chi -square distribution with
one degree  of  freedonij  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 con-
 o
fidence  limit  for the extra risk calculated at low doses is always linear.
This  is conceptually  consistent with  the linear  non-threshold concept dis-
cussed  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 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, data at
the highest dose are  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
                                   5-92

-------
                     X2 =
 h
 I
1=1
                                                        .th
is calculated where  N.  is the number of animals in the i   dose group, X. is
                               f- h
the number of animals  in the i   dose group with a tumor response, P. is the
                                    J_ L                                '
probability of  a response  in  the i   dose group  estimated  by fitting the
multistage model to  the data,  and  h  is the number of  remaining  groups.   The
                              p
fit is unacceptable whenever X  is  larger than the cumulative 99% point of the
chi-square distribution  with  f degrees of freedom, where f equals the number
of dose groups minus the number of  non-zero multistage coefficients.
     5.3.3.3.1.2  Selection of  data.   For some  chemicals,  several  studies
using different  animal  species,  strains,  and sexes and  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.  The pro-
cedures used  in evaluating these data are  consistent  with the approach  of
making a maximum likely-risk estimate.  They are listed 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  significantly higher statistically
than the control for at least one test dose level or where the tumor incidence
rate shows a statistically  significant trend with  respect to dose  level.   The
                                                                             *
data set that gives the highest estimate of the lifetime carcinogenic risk, q-,
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  relation-
ship 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 assess-
ment. The geometric mean of numbers A-,, A,,, ..., A  is defined as
                            (A-, x
                                                vl/m
                                   5-93

-------
     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.
     5.3.3.3.1.3  Calculation of human equivalent dosages.  It  is appropriate
to correct for  metabolism differences between species and absorption factors
via different routes  of  administration.   Following the suggestion of Mantel
and Schneiderman (1975),  it is assumed that mg/surface area/day is  an equivalent
dose between species.   Since the surface area is approximately proportional  to
the two-thirds power of the weight, as would be the case for a perfect sphere,
the exposure in mg/day per two-thirds power of the weight is also  considered
to be  equivalent exposure.   In an animal experiment,  this equivalent dose is
computed in the following manner.   Let
     L. =
     1  =
     m
     W  =
duration of experiment
duration of exposure
average dose per day in mg during administration of the agent
(i.e., during 1 ), and
average weight of the experimental animal.
Then, the lifetime average exposure is
                                     1  x m
                                d=
     Inhalation—When exposure  is via  inhalation, the calculation of dose can
be considered  for  two cases where 1) the carcinogenic agent is either a com-
pletely 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 that reaches an equilibrium between the air breathed and the body
compartments.  After  equilibrium is  reached,  the rate of absorption of these
agents is  expected to be proportional  to  the metabolic rate, which in turn is
proportional to the rate of oxygen consumption, which in turn is a function of
surface area.
     Case  1~Agents  that are  in the form of particulate matter of virtually
completely absorbed  gases,  such as sulfur dioxide, can reasonably be expected
                                   5-94

-------
 to be  absorbed proportionally to  the breathing  rate.   In this  case  tne exposure
Jn ing/day may be expressed as

                                   m =  I x v x  r

                                        3          3
 where.  I  = inhalation rate per  day in  m ,  v  = mg/m  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 sur-
 face 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.
                              o
 For humans,  the  value of 20 m /day* is adopted as  a standard  breathing rate
 (International Commission on Radiological Protection, 1977).
                                     2/o
     The equivalent  exposure in mg/W '   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  kilogram per  day,  i  = I/W,  based upon the pre-
viously  stated relationships, are tabulated as follows:
                   Species
                     Man
                     Rats
                     Mice
  W
70
0.35
0.03
i = I/W
  0.29
  0.64
  1.3
Therefore, for particulates  or completely absorbed gases, the equivalent ex-
              2/3
posure in mg/W    is
     *From "Recommendation of International Commission on Radiological Protec-
                                                 7   3
tion," page 9.   The  average breathing rate is 10  cm  per 8-hour workday and
      7   3
2 x 10  cm  in 24 hours.
                                   5-95

-------
                                       - lWvr -
                                                     wr
                                                     vr.
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.
     Case 2— The dose in mg/day of partially soluble vapors is  proportional  to
                                                      O /O
the 02 consumption, which in turn is proportional to W    and is also propor-
tional to the solubility of the gas in body fluids, which can be expressed as
an absorption  coefficient,  r, for  the gas.  Therefore, expressing  the  02
consumption as 02 = k W2/3, where k is a constant independent  of species,  it
follows that

                               m = k W2/3 x v x r
or
                               d = -JL- = kvr.
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 absorp-
tion is equal by either exposure route.
     5.3.3.3.1.4   Calculation of the unit risk from  animal  studies.   The  95
percent  upper-limit risk  associated  with -d  mg/kg '  /day  is obtained from
6LOBAL79, and for most cases  of interest to risk assessment, can be adequately
approximated by" P(d) = 1  - exp (-qjd).  A "unit risk" in units X is the risk
corresponding to an  exposure  of X = 1.  This  value is estimated by finding the
number of mg/kg2/3/day that corresponds to one unit of X and substituting this
value  into  the relationship  expressed above.  Thus,  for example,  if X  is  in
                                    5-96

-------
units of pg/m  in the air, we have for case 1, d = 0.29 x 701'3 x 10~3 ug/kg
                                          o
per day,  and for case 2, d = 1, when pg/m  is the unit used to compute para-
meters 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)
molecular weight (air)
mg/m .
Note that an equivalent method of calculating unit risk would be to use mg/kg/day
for the  animal  exposures,  and  then  to  increase  the jth polynomial  coefficient
by an amount
                          (Wh/Wa)j73   j = 1, 2, ..., k
and to  use  mg/kg/day equivalents for the  unit  risk values.   In the section
calculating the  unit risk  for animals, the final q? will always be the upper-
limit potency estimate for human risk based on animal  data.
     5.3.3.3.1.5  Interpretation of quantitative estimates.  For several  reasons,
the unit risk estimate based on animal bioassays is only an approximate indica-
tion of the absolute risk in populations exposed to known carcinogen concentra-
tions.  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 regarding carcinogenic response.   Finally, human populations are
variable with  respect to  genetic  constitution, 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.  The  comparative potency
of different agents  is  more reliable  when the comparison is based on studies
in the  same test  species,  strain, and sex,  and  by the same route of exposure,
preferably inhalation.
     The quantitative aspect of  carcinogen risk assessment is included here
because it  may  be of use  in the  regulatory decision-making process, e.g.,
                                   5-97

-------
setting regulatory priorities, or evaluating the adequacy of technology-based
controls.   However, with  present  technology,  only imprecise estimations are
possible concerning cancer risks  to humans at  low  levels  of exposure.  At
best, the  linear extrapolation model used here provides a rough but plausible
estimate of the  upper limit of risk, and while the true risk is probably not
much more  than the estimated risk,  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
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.
     5.3.3.3.1.6  Alternative methodological approaches.  Methods used  by the
CAG for quantitative assessment are consistently conservative,  i.e.,  they tend
toward high estimates  of  risk.   The most important  part  of the methodology
contributing to this  conservatism  is the linear non-threshold extrapolation
model.   There are  a variety  of other extrapolation models that could be  used,
all of which would give lower risk estimates.   These alternative models, the
one-hit, Probit, and  Weibull  models,  have  not been  used  by the CAG in  the
following  analysis,  but are included  for comparison in the appendix.    The
CAG's position is  that, given the limited  data  available  from these animal
bioassays, especially  at  the high-dose levels required for testing,  almost
nothing is  known about the  true shape of  the  dose-response curve at  low
environmental  levels and that the risk estimates obtained by use of the linear
non-threshold model represent plausible upper limits only.
     Extrapolation from animals to  humans could also be done on the basis of
relative weight rather  than  on  the basis of relative surface area.   Although
the  latter approach,  used here,  has more  justification in  terms  of human
pharmacological responses, it is not yet  clear which of the two approaches  is
more appropriate for  the  assessment of carcinogenicity.  In the  absence of
information on this point, it seems appropriate to use the more conservative
basis for extrapolation, the relative surface area.
5.3.3.3.2  Unit risk potency estimates and  relative  potency.   Neither  of the
two  available epidemiologic  studies  provided positive results from which to
derive a quantitative  risk estimate  for DCM exposure.  With respect to  animal
data, three chronic  studies  are relevant to this discussion:   two inhalation
studies and one drinking water study.  These are summarized in Table 5-35.  In
                                   5-98

-------




















co
LU
1— (
O

?
co

i
 O
•r- O
i— S-
(0 (0
CO (A


*t
O
o
in
rH
O
"O
o in
o co
in
•a
- c
O 10




vX > 2t ป-


cu

o
•a

cu
(A
x^
in
P
(0
s-

m
en



in
+3
(0
s-

^k
cu
r—
^:
(0
/*^
1
CU

O)
(0
S-
Q.
co

0
00
cr^
r™f
^^

2t
o








1
1









M
o
o
in
rH
O
-O
o in
o ro
m
-a
- c
O (0




NX (J)
C CU S-
O CU (0
•r- >\ 3t CU
-P CO •V. >,
18 "O IA
•— -x, >, (A
> 3t x>  o w>
f— v^ J>ป .^1 ^^ . _{••
f8 S- (0 CM P
J= j= T3 S- C
C O i- O
HH'VO m.t- o E






X
cu
(A
•x.
(A
P
ro
S- CU

O 0
en -a



IA
P
(p


^^
a?

"s
fO
Q

CU

O)
<0
t.
a.
co

CM
00
cn
rH


U
0
o
in
1 CO CU
O CU J= r—
d} r^ P (0
C =3 E
•a <ป- cu
a; o o M-
c c
•i- CU C
i— O Ift -f-
S- ••-* C
aป -P o s-
•o o> a. cu
s- 
O r- CO rr^
CO ,Q. t- r-

ซ\
in
CM
rH
/•"V
cs'o t-
in m ,cu
CM >
o
in - u
o 

S-
CU O
^_> f"
(0 P
2s c
T3 0
CU 05 S
H C
•r- *i— <^-
C JซJ CM
0 C
•r- v- t.
0) S- O
Q 73  o
"X. f) (A O
in rH r-
4J O CU

S- O) P (0
(ACE
in o o co
.00 T3 0 <ป-






(A
j ^
(0
.s-

^^ซ
t^L
co

S-
cu

o
SA
•r-
u_
*v
ro
CM
00

rH


^C
CJ

5-99

-------
the two inhalation studies (Dow Chemical Company 1980, 1982) only the salivary
gland regions in male Sprague-Dawley rats in the 1980 study showed statistically
significant increased cancers.  These results are discussed in Section 5.3.3.1.1.2
and are presented  in Tables 5-12 through 5-14.  In the drinking water study
(NCA 1982), the increase over untreated controls in combined neoplastic nodules
and hepatocellular carcinomas in female rats was the only statistically signi-
ficant finding (Table 5-24).  However, when compared with historical controls,
these results lose their statistical significance.
     5.3.3.3.2.1  Unit risk (ug/m3) for inhalation studies.  The data used for
estimates of the  unit risk for inhalation are presented in Table 5-36, which
shows the positive salivary gland region sarcomas for the inhalation bioassay.
Exposure was 6 hr/day, 5 days/week,  for  2  years.   Equivalent dosages were
determined for  humans from  the  animal  dosages  utilizing the equivalent dosage
methodology presented previously.   As described in the section on  pulmonary
uptake  and  distribution, DCM  is  readily absorbed  into the body following
inhalation and it equilibrates rapidly across the alveolar epithelium.  There-
fore, the CAG  considers it a virtually completely absorbed gas especially at
low doses and  determines equivalent human exposure as explained under Case 1
of the  inhalation  section.  As  presented  in  Table  5-36, the nominal  exposures
are nearly 15 times the human lifetime equivalent exposures.  This difference,
24/6 x  7/5 = 5.6,  is partly due to  the use  of  a continuous equivalent dosage.
There is an additional  factor of  about 2.6,  however,  which is  attributable to
the nature of  the method used  for  determining human equivalent dosages for
inhalation studies.   Put another way, if DCM had  been determined  to-be a
partially soluble  vapor,  the  unit risk slope  would be lower by a factor of
about 2.6.
     When the incidence data given above were  fitted with  the continuous human
equivalent exposures,  the linearized multistage model yielded  the  following
value for the 95 percent  upper  limit of risk:

                       q* = 1.8 x 10"7(|jg/m3)~1

     5.3.3.3.2.2   Unit  risk  (mg/kg/day) and (ug/1) for oral studies.   This
unit risk  estimate should be used  only under  the assumption that  DCM is  a
potential human carcinogen.  As discussed in the qualitative section, there is
only limited animal  evidence  and  no  human evidence  to  support that  assumption.
                                   5-100

-------
       TABLE 5-36.  INCIDENCE RATES OF SALIVARY GLAND REGION SARCOMAS IN
                        MALE SPRAGUE-DAWLEY RATS IN THE
          DOW CHEMICAL COMPANY (1980) DICHLOROMETHANE INHALATION STUDY
Continuous human
(animal nominal)
ppm and
0 (0)
34 (500)
103 (1,500)
240 (3,500)
equivalent
exposures
pg/m
0 (0)
1.2 x 105 (1.8 x
3.6 x 105 (5.2 x
8.4 x 105 (1.2 x


106)
106)
107)
Incidence rates
Number of rats with tumors/
total rats examined (%)
1/93 (1%)
0/94 (0%)
5/91 (5%)
11/88 (12%)
* 1 ppm x 1.2 x 103 pg/m3 x 84.9 = 3.5 x 103 pg/m3
                            2875
                                                   o
An exposure of 500 ppm DCM in air expressed as pg/nr is
          500 ppm x 3.5 x 103 pg/m3 = 1.8 x 106 [jg/m3
                          ppm
Since animal exposure was for 6 hr/day, 5 days/week, the animal continuous
lifetime exposure equivalent was
           1.8 x 106 pg/m3 x  6 x 5 = 3.2 x 105 pg/m3
                             24   7
The human equivalent dose for DCM is calculated first by determining the amount
actually breathed by the rats.  As presented in the inhalation dose equivalence
section, 350 g rats breathe
           I = 0.105 (0.350/0.113)2/3 = 0.223 m3 (air)/day.
Thus continuous animal dose was 3.2 x 105 pg/m3 x 0.223 m3/day = 7.15 x 104 pg/
day, or, for a 350 g rat, 7.15 x 104 pg/day/0.350 kg = 2.0 x 105 pg/kg/day.
The human equivalent dose is
             2.0 x 105 pg/kg/day -=- (70/0.350)173 = 3.5 x 104 pg/kg/day
  or
3.5 x 104 pg/kg/day x 70 kg = 1.2 x 105 pg/m3.
                      20 m3
                                   5-101

-------
     Since publication of  a final  NTP report on gavage studies for rats and
mice on DCM has been cancelled, there is no suitable oral study from which to
estimate a unit risk.
     5.3.3.3.2.3   Comparison of animal and human inhalation studies.    The
study of  Kodak  employees,  yielding negative cancer results, is compared with
the positive tumor results  of the  rat inhalation study (Dow Chemical Company,
1980).   In the latter study, the salivary gland region tumors in male  Sprague-
                                                                  ~7      3 "1
Dawley rats led to a 95% upper-limit slope estimate of qฃ=1.8 x 10   ((jg/m )  .
If this slope factor is applied to the human inhalation study, in which time-
weighted  average  exposure  was  estimated  as ranging from approximately 30 to
120 ppm,  the expected  impact of exposure can be estimated.  For this purpose,
1 ug/m3 of DCM  is equivalent to 2.9 x 10   ppm.   Thus, the upper-limit slope
in ppm is

      q* = 1.8 x 10"7 (pg/m3)'1 x 3.45 x 103 ug/m3 = 6.2 x 10"4 ppm"1.
                                        ppm

Since this upper-limit slope is based on continuous lifetime equivalent exposure,
the exposure range of  30 to 120 ppm time-weighted average must be adjusted  to
lifetime  equivalence as follows:
        30 ppm x 20 years x 240 days x  8 hr = 1.88 ppm
                 70         365"        2?
and 7.52  ppm lifetime  equivalence  for  the  120  ppm  exposure  group,  assuming  20
years  of  exposure for  the 252 long-term  exposure workers.   Based on this
upper-limit  slope  factor and the range of exposure, the group of 252 workers
could expect an additional lifetime risk of between

          R = 6.2 x 10~4 ppm"1 x 1.88 ppm = 1.2 x 10"3 and 4.7 x 10~3.
For these 252 workers, this would translate to an upper limit of between 252 x
1.2 x  10~3  = 0.3 and  1.2  excess lifetime cancer deaths.   Based  on  the  total
expected Kodak  employee  deaths of 65.9 (Table 5-33) for this 20-yr minimally
exposed  cohort,  we  would expect 26% of the cohort to die  in the  17-yr follow-
up period.  Transposing  this 26% to the expected excess lifetime cancer deaths,
an upper limit  of between 0.1  and  0.3 excess cancer deaths can be expected
                                   5-102

-------
.during the 17-yr follow-up period.   The  power to detect this  increase  from
 17.77  to 18.1 cancer deaths  is  less than 10%. If these excess cancer deaths
 were from cancers of one  specific  site,  the power would be greater, but not
 great  enough to declare this  a  negative  study.   Even  for a rare  cancer,  such
 as  a liver cancer, the  expected number of cases would  be  much less than 1.
 Since  only deaths can be  observed,  the power  to  observe one death  from  liver
 cancer in this  cohort is quite small.
     Based on the analysis presented above, the study  of Kodak  workers exposed
 to  DCM, showing no increase  in  cancer, cannot be judged as having negative
 results because  of  its low power, which is related to  low exposure  from  a weak
 animal  carcinogen.
     5.3.3.3.2.4  Relative potency.   One  use of  the unit  risk concept is to
 compare the relative  potencies  of carcinogens.   To estimate relative potency
 on  a per mole basis, the unit  risk slope  factor is multiplied by  the molecular
 weight and the  resulting  number is  expressed in terms  of  (mmol/kg/day)"1.
 This is called the  relative potency  index.
     Figure  5-1  is  a  histogram representing  the  frequency distribution of the
 potency indices  of 53 chemicals evaluated by the CAG  as suspect  carcinogens.
 The actual data  summarized  by  the  histogram  are presented in Table 5-37.
 Where  human data are  available  for  a compound, they have been  used  to calcu-
 late the index.   Where   no human data  are available, animal oral  studies and
 animal  inhalation  studies  have been  used, in that order.  Animal  oral  studies
 are selected over animal inhalation  studies because most of  the chemicals  have
 been subjected  to animal  oral studies; this  allows potency  comparisons  by
 route.
     The  potency index for DCM,  based on  salivary gland  region  tumors  in male
 Sprague-Dawley rats in the Dow inhalation study  (Dow Chemical Company, 1980),
            _p               -T
 is  5.3 x 10   (mmol/kg/day)   .  This is  derived as follows:  The slope esti-
                                                 in converted units of 6.3 x
mate from the  Dow study,  1.8 x 10~7  (pg/m3)'1
10    (mg/kg/day)   ,  is  multiplied by the molecular weight  of 84.9 to give a
potency  index  of  5.3 x 10  .   Rounding off of the nearest order of magnitude
gives a  value  of  10   ,  which  is the  scale presented on the  horizontal axis of
Figure 5-1.  The  index of 5.3 x 10~2 is the least potent of the 53 suspected
carcinogens.  Ranking of the relative potency indices is subject to the uncer-
tainty of  comparing estimates  of potency of different  chemicals  based on
different routes  of  exposure  to different species using studies of different
                                   5-103

-------
20
                                         4th         3rd         2nd        1st
                                      QUARTILE   QUARTILE   QUARTILE    QUARTILE
                          1      23456
                               LOG OF POTENCY INDEX
7
8
      Figure 5-1.  Histogram representing frequency distribution of the potency indices of 53 suspect
      carcinogens evaluated by the Carcinogen Assessment Group.
                                       5-104

-------






Q.

0

o
z-
LU
to
to
LU
CO
to
•a:
LU
C3
O
•z.
cj
-
CQ

O
LU
<
ID
—1 to
<: z
LU O
O
to z
_l 1— 1

LO 1C
C3 1—
Z CJ
O LU
ฃ! Q.

S- +J rHO
0) ••- 0) C
73 C O •!-
t- O3r—
O to v- '




U X
C O)
CU 73
O *r—
Q_




ฃ-
10 +J
D- co
U T-
CU 0)
o5


rH
'S
cu to
0-73
O \
i — cn
to ^t
cn
v^



en c to
CO •!-
•r- 0 S.
Q-73 Q; CO
D- cu 
CU U)
c:
f— tO
O E
^



ฃ-
cu
^2
z
to

o















73
C
O
O-
E
O
CJ



rH




\

rH
CD
rH
X
rH






rH
00






/— N

CM
O






J

CJ
^

5



co






co
rH
X
CM






00
CD








LO
I-H






rH





I — i







to





CM
00
00
O
ซ^-


















u
•r-
c
OJ

•a:



00






oo
CD
rH
X
co






co
CM
LO
CM








r-i
i-H






CQ
CM





to







1 — I





OO
1
CM
CO
1
CD
LO




















Q-
l — i
ro
i 	 i
CQ



rH






CD '
CD
rH







CO



t — s
3

CM
1
O
i-H
X
CM
LO






rH





t/1







to





CM
1
CO
•=*•
1
rH



















CU
C
CU

c
cu
CQ



LO rH






•* rH
+ +
O CD
i — 1 i — 1
X X
•3- CM






CM
^- CD







f — s
s
•* ID
OO
CM CM






i-H CM





to to







to _J





LO r^
i i
f-^ rH
00 •=!-
1 1
CM CD
CD <3-
•=(-
















c E

CU •!-
73 i—
N ">>
C. i-
O) CU
CQ CQ



00






CM
0
P-J
X
CO






CM
rH
rH








3:
00
^.






CM





to







	 1





CD
co
CD
ซ^-


















E

'E
73
to
CJ



rH






rH
CD
, — |
X
• CM






00
co
LO
rH




, — |

CD
rH
X
CD
CO
r-i






CQ
CM





to







1 — t





in
i
CO
CM
1
tD
LO








CU
73

ฃ_
O
^
u
2
1 %
cu


c.
o
-Q
S-
. to


^^
OO O)
+ cn
co
Q_

cn
f—

3
CM 0
•r r—
O r—
rH O

CU

-f^

r—
Q
CO
73
CD 0)
CD 3
•sf C
^
g
^





rH
to
r-i





CU
O
co a
cu
73
CU
CU

to
—i cr
cu

to
c



1 — 1 1 — 1

cu
(J
cu
73
CD >
1 CU
1 CU
r- -P
LO •!-
E

	 1
II

CU
u
cu
73


CU

c
0)
CJ

cu t— .
C 4—
ro u
73 to
0 II

x: to
cj to
5-105

-------























TJ
CU
3

C.
O
u
*"•*'

r-
CO
i
CO

UJ

CO
 rHCt
cu -i- cn c
•o ซ= o -r-
J- 0)r-

S

?? b^
c cu
CU T3
•P C
O -r-
0-

i.
re -P
i — x:
3 cn
O T-
CU CU
"o
^-


i-H
1
f— N
o re
a. -a
o-^s.
r— Ol
CO -^
•S

cn cz re
CO -r-
•i- 0 ฃ-
Q.*O Cฃ CU
3 CU  T3
CJT-
	 1 >
cu in
cz
f- re
0 E
3
-1-


S-
CU
x>
CO
CJ




Compound


•+ 0+"+





i—]
00 + 0
O CD O C)
rH rH rH i-H
X X X X
r-~ co co co


cr> r** cn ^J-
oo CD r-^ co
cn co co co
CM rH rH





CM CM
CM 1 1
1 CD CD
O rH rH
rH X X
X CM CD CO
o^ *^ CM r^**
CD i-H CD LO



CD
CM CO CO CO







co -a —i —I







I-H 1 — II — II — 1




CM rH LO LO
1 1 1 1
CO CM ซ* CD
CD r-~ co CD
1 1 1 1
r-- I — cn cn
o co r^ r~-
rH


cu
ฃZ
re
f
-P cu
Chlorinated ethanes
1,2-dichloroethane
hexachloroethane
1,1,2,2-tetrachloroe
1,1,2-trichloroethar


rH






CD
O
i-H
X
oo


^t-
s
rH






CM
CD
rH
r-



cn
CM







CO







1 — 1




co
1
CD
CD
r-
CD





Chloroform


?





co
X



o
rH







t — s
^ — '
\ — 1



rH







co







co




co
r--
i
CD





Chromium


CM





CM
+0
i-H
X •
rH


LO
CO
CO







co
ฐ"



CO
CM







co












co
i
cn
CM
o
CO





a
a


co






CD
i-H


rH
CO
LO
CM







cn
CO
rH



CO
CM







CO












rH
1
cn
rH
cn





Dichlorobenzidine


r-l





, — |
CD
3



f-ป
cn




/ — *
i — i
rH^
1
O
rH
X
r-.
<-
r-i



co







_i












LO
co
1
LO
r^





1,1-dichloroethylene


•* CM
+ +





ซd- rH
+0 +0
i-H rH
X X
i-H CO


cn
CD' CM
00 00
CO rH







co
CO CD



ca ca
CM , CM







co co












rH CM
1 1
LO rH
1 1
0 rH
CD CM
i-H





Dieldrin
2,4-dinitrotoluene


CM CD





CM rH C
+o 'o
3 3
rH CD


LO
O CM
co at





co
i
CD
rH
r~- x
r^* cn
CD cn


t
ca ca
CM CM







CO CO












f — OO
1 1
CD cn
CD CO
1 1
CM CD
CM O
rH rH





Diphenylhydrazine
Epichlorohydrin


CM CO





NJ CD
*o +o
l-H rH
X X
CM rH



co tn
rH l-H






ฃT
•* o
i-H cn



CO
CM rH







CO CO












•* i-H
4 i
it- 00
1 1
rH CM
i-l "3-
rH LO



i.
CU S-
x: cu
Bis(2-chloroethyl)et
Bi s(chl oromethyl )eth

— _
cu
cn
re
Q_

cn
c
•r™
o
o
t—
cu
+J
cz
o
-o
cu
c
j S
cz
o
u






t
cu
u
c.
cu

Ir_
2>
cu
cu
re

cr
cu

re

i — i

ii


cu
u
cu
•o
cu
T3
CU
-p
'i
— 1
II
— 1
cu
u
cz
cu
aS = Sufficient evid
5-106

-------






































/— -s
•o
 e
U •!—
i— C C
cu cu  "D
CU •!-
CU U)
M- tO
o e
3




S-
O)
-Q
E
3
^^

oo
3- rH rH rH




CO CO
CM CO CM








OO — 1 — 1






1 — II — II — 1






ID r~ en
i i i
•3- LO CD
CO 00 CO
1 1 1
CD CD CO
I— 1 rH Ln
co co




cu
c
03
X
CU
-C CU
O T3
i— 03
U S- S- S-
>, cn cu s- co
u E cu E
O i— O E O
i- 03 1/5 o in
O U •!- •!-

[E E 03 *~ 03
u j= -c: 03 e
03 U Q.-P E
X CU i— CU 03
cu +-> 03 .a 03
3Z



ID i-H CM
+ 1 +





ID CM rH

CD CD CD
rH rH rH
XXX
CM un rปป



CD P^

rH 
X X LO
CM CO rH
< • •
ID ID r-l




CO , 'cu
03 -C 3ฃ
X •)-> U
CU CU *i—
1C EZ ^



CO
+





co
+
CD
i— 1
X
CM



rH

^~
r^ป



/ 	 N
*!-H
• cr
ฃ

•p
O
C
s^^/
CD
.
LO
CM




CO
CM








oo






I — i






CD
1
LO
r-

CM
ID






CU
t~~
• r-
E
03
in
o
^_
in +->
CU •!-
c c
*r- r—
H >>
03 ^=

O CU
S- E
•P -r-
•r- Q




•3- co
-1- 4*





CO CM

O CD
rH i-H
X X
•3- CD



rH CM

CM 00
CD Ln
i— 1 rH


/•""*%
*r-H
a-
-Q

•p
O
CI
\^^
Ln co
• *^"
co .
•^- LO




CQ CQ
CM CM








oo oo






1 — 1 1 — 1






LO CO
1 1
OO ID
i— 1 rH
1 l
LO Ij"
LO CM
CD






CU CU
C C
•i — ซr-
E E
03 03
in in
o o
S- S-
-P 4-*
•r- •(—
!= C
r— r—
^> ^}
r- 4_J
4^ 3
CO JD

a (5




cu
D)
03
Q_

C
's
o

1 —
o
^
cu
jC
c
o

•a
cu
3
C
'43
0
u














ซ
cu
u
c
cu
"O
•!~
cu
CO
+J
OJ
3
cr
cu
T3
03
C
1 — 1
II
1 — 1


cu
(J
cu
-a
'>
cu
-a
cu
i|..*
., —
.E
II
	 i
cu
(J
c
cu
•a
•f_
>
cu

4-5
C
cu

o
•r-
t-
M-
3
oo

II

OO
03
5-107

-------
*f- <1) <->
o -a x
3 OO)
t- 4-> rHa
O) -r- CD C
•o c o -r-
t. O^f*
O nj *•— '
c

>^
U X
C 0)
QJ "O
.j C
O 'i—
a.



-
(5 ii *
r— <"T
3 O)
U T-
QJ CL>
,- IS
O
,_.
•b.




|~~'
*^_BI^
^^^
oj S?
cx.-o
i— cn
to J*:
—
Dl
E

r~^
a> cn c to
3 C 0 •!-
C -i- C_> 5-
•r- Q.T3 a; a>
+> 3 QJ 
CO r—
1 CO CO
in a) ฃ
U T-
UJ r— C C
5s > "o ^^
^ Q) •r—
ai i/>
tl 5
^^ 2
O c
^•j
•"T*





S-
1
z:
to
^r
^~*













•a
3
o
i-
o
CJ

CM **• <• O CO
•ซ• + + +






NJ CO ซ* CO
+ + + 0 +
CD CD O O O
t-H i-H rH r-H rH
X X X X X
CM ^- CO rH rH





CM l-H rH
• * *
o r~- co co ซd-
CD i — I CD Cn CM
rH i-H i-H rH CO









CO
1
CD
ซ3
co cn . CM ซ*
rH • CM Cn CO
. CM O .
CM CO CO ^i" "!j-



CQ CO CO CO CO
CM CM CM CM CM




oo to oo oo to




. ^^^ t l l
t—i 1 r*~l 1— 1







CM cn in ID co
1 1 1 1 1
in co co o to
in r~- cn co co
CD cn <* ID ID
co in oo oo co
cn r- uo co






fO C

QJ OJ S- E
C S- 3 CO
"5 i— *>>'>i
•i— >>.C C
i — .C -P Q)

S- QJ E O-
S- 1 1 ••-
O- 1 1 1
o o o o
in i/> i/) i/>
o o o o
i- i- s- i.
•r— ปr^ ปr~ •ป"•
c. c c c. at
ฑฑฑฑ S

i-H
-r






CD
CD
rH

•^





ซ^-
•
r^.
cn
rH









CM
CD
rH
X
cn
cn
rH



CO
CM




oo













CM
1
to
CD
1
00
oo







f
o
c
Jr
Q.
o

.c.
u
•r—
s-
-p •
in i
r— ID
o •>
OJ -
x: CM
a-

OO rH CO CD O
+ + "1"



c n:
o
0 T3 QJ
r~- CM CD T3 C t.
+ 0+T-HO QJCO CO
CD O O X CD ซ
1-HrHrHinrH CO-.C i —
X X X • X -Q *""^ ' ,
r-r- D.T3 i— -P
3 X r— in C
OO "5t* in U CD O *r-
* * " ' , "^ ~* ^\
CM in ^" rH CM CO f"™ in -C +^
CvJ t^ rH CO IซO tj CO CU ซP S~
CO rH "^" i 1 C ^ ^^ w
S- •!- 4-> C
CO -P C -r- 4-
CO O >> O
>,0.<= r—
ai 3 o- a>
x: o s- T- o)
^— v h- U CO -P 5-
h-H O Q) r— O)
v_/ C 3 QJ
LO CM . C -i- E 13
+ CM CM 1 i — CO i —
0 1 10 0) E >> 0)
rH CD O r-H 13 3 CD _Q E
XrH rHX OJZXT CO
ID X CO X in E'^-'-P 13 m
"1 "l1"!0^1^ Q)3c+o a)
l-HOOrHrHrH CnOCO-C
CO *• i~ -P
in c CD U -P
•i- o in i— c:
-P *t— ro co CU
a> 3 co a)
co 03 U EI — mwS-
CMCOCMCOrH C COCD-r- O.
03 T3 X: -P CD
13 O) C CO 13 S-
'> •!- "~ •!- CO  -P
CO -r- C -P >>
3 r— co c co in
OO — lOO — lOO O" > — "r- 13 • •<-
T3 .c: H-H o- D) c: -P
(O 4J Jซi 3 O)
c: >, — I QJ-PCD > — ' -P CO
incoa.
couo ex: QJCO
aj xi •*— i — •(— -P in
u "O in QJ t/>
c we aj 4- t. CD
tU QJ-r-C Q-O Q.E
•o o. ca o o
•i— OQJEi — -P inu
f i i i i QJ *uj o a: cn -r- ij
iH CO in i-H rH -C J- ••- U
OrHCOCDCD T3 4J-P M-CDCCU
, , | , | QJ -g^^OS^JU

SI-HCD E i — t-s-ojco O.T3
rH OO •!- 1 3 13 i— -p-
_1 S--PWC3U>
CD O. O 3 OJ -r- CD
II Q- CD Q- O QJ C
	 1 3 X QJ O CD CD
QJ CO E O C
6-S S- C
u cn w -p •!— x: uco
C QJ CO -P S-
CD QJ *' — ^ X CO CD
~& fc- "O _QJ >i OJ O)
QC '> 4->CCQ)CO
ivi oj QJ a) in in -r- T-rH x; x:
C^-N r- C QJ -^ 1 -P 0)

o i-~i x: r— ai c oco-i—oj^t-o
.^C-J-P >>T3 OJ •— S-S-CCOO-P
•tjl — Q) jC'i— *t- • l/)OT3QJ'O
OS_x 0 4-J S- U -P V.. •— 4^
s_ j_ cuo *r- i — i — cocni — u
OCT O CD O " — '.4— COCOCOO-^^COQ)
r— T-i— C S-X: <<- ..EEE "V. -1-3
x:x x: CD o u 3 in -i— -i— 3 CD cn 4^ xj
oo u x: r— oo _^ccx:x:E03
cO-r- CO O.X: r- S-eCCO'— '(— ^-'Zin
t--as-cocj>) u co
-P i +J x -r- c: E
QJ p. OJ O S- T— CO w • • •
f— [_(_|_>.a3 Qฃr-4 CSI CO
5-108

-------
quality.   Furthermore, all the indices are based on estimates of low-dose risk
using linear extrapolation  from  the observational range.   Thus these indices
are not valid for the comparison of potencies in  the experimental or observa-
tional range if  linearity does not exist there.  The potency  index for DCM,
furthermore, is  valid only  under the  assumption that DCM is a  potential human
carcinogen.   The evidence for that is limited.
     5.3.3.3.2.5  Summary of  quantitative estimation.   No  positive  epidemic-
logic studies exist  from which to estimate a unit risk for exposure to DCM.
Only one animal  data set has shown increased cancers from which a unit risk
assessment could be  estimated.   In the Dow Chemical  Company (1980)  rat inha-
lation study, there  were increased sarcomas in the salivary gland region of
male rats; this yielded an upper 95% limit of the potency estimate of q? = 1.8
x 10"7 (Mg/m3)'1, equivalent  to q* = 6.3 x 10"4 (mg/kg/day)'1 by the oral
route.  However, no  positive  data exist with which to  provide a direct oral
route estimate.   In total, there is only limited evidence that DCM is a poten-
tial human  carcinogen.   The unit risk estimate is valid only  if one accepts
that limited evidence.   Further, if one chooses to express the potency of DCM
relative to that of  53 chemicals evaluated  as suspect carcinogens, DCM is the
weakest,  ranking last.
5.3.3.4  Summary.  Six chronic studies of  DCM administered  to animals have
been  reported:   four in  rats, one  in mice, and  one in hamsters.   The Dow
Chemical  Company (1980)  reported the  results of chronic inhalation studies  in
rats and  hamsters.   The  rat study showed a small increase in the number of
benign mammary tumors compared to  controls  in female rats at all doses and  in
male rats at the highest dose, as well as a statistically significant increased
incidence of ventral  cervical  sarcomas, probably  of salivary gland origin,  in
male  rats.   The  response pattern of  the  salivary gland tumors is unusual,
consisting  of sarcomas only and appearing  in males but not in females.   In
hamsters, there  was  an  increased incidence of lymphosarcoma in females only,
which was not  statistically significant after correction for  survival.  The
results of  the Dow Chemical Company  (1980)  study  have since been published  by
Burek et  al. (1984).  The second  inhalation study in rats by the Dow Chemical
Company (1982) reported  that  there were no compound-related increased inci-
dences of any tumor  type, but that the  highest dose was far below that of the
previous Dow inhalation  study in rats.  The National  Coffee Association (1982a,b)
drinking water study in  Fischer 344 rats reported that the incidence of neo-
                                   5-109

-------
plastic nodules  and/or  hepatocellular  carcinomas  in  female  rats was  increased
significantly with respect to matched controls, but their incidence was within
the  range  of historical  control  values at  that  laboratory.   The National
Coffee Association  (1983)  drinking water study in B6C3F1 mice also  showed a
borderline response of combined neoplastic nodules and hepatocellular carcino-
mas.  The  National  Toxicology Program (1982) draft  gavage  study  on  rats and
mice will  not be  published as a  final  report due to data discrepancies.
Selected information  from the gavage  studies  may be incorporated into the
future NTP inhalation  bioassay,  pending the results of  the in-depth audit.
     There are some  other inadequate animal studies in  the literature.  One
study (Theiss et al.,  1977) reported a marginally positive pulmonary adenoma
response in  strain  A mice injected intraperitoneally with DCM.   Two negative
animal  inhalation  studies  were inadequate because they  were  not  carried out
for a full  lifetime (Heppel et al., 1944; MacEwen et al., 1972).
     One carcinogenicity study of DCM in animals is currently in progress:   an
NTP  2-year bioassay by  inhalation in Fischer 344/N rats and B6C3F1 mice.
     Positive results in a rat embryo  cell transformation study were reported
by Price et  al.  (1978).   The  significance of  their  findings  with regard to
carcinogenicity is not well understood at the present time.
     The epidemiologic data consist of two studies:  Friedlander et al.  (1978),
updated by Hearne  and  Friedlander (1981), and Ott et  al.  (1983 a,b,c,d,e).
Although neither study  showed excessive risk, both  showed  sufficient  defi-
ciencies to  prevent them from  being judged negative  studies.  The Friedlander
et al.  study (1978) lacked great enough exposure (based on  animal  cancer
potency estimates) to provide  sufficient statistical power to detect a poten-
tial carcinogenic effect.  The Ott et  al. study (1983 a,b,c,d,e), among other
deficiencies, lacked a sufficient latency period  for  site-specific  cancer.
     Only one animal data  set has shown increased cancers from which a unit
risk assessment  could be  estimated.   In the Dow Chemical Company (1980) rat
inhalation study, there were increased sarcomas in the salivary gland  region
of male rats; this  yielded an upper 95 percent limit of the potency estimate
                                                          -4            -1
                               equivalent to q* =• 6.3 x 10   (mg/kg/day)   by
of q* =  1.8 x 10"7 (ug/m3)'1
the oral route. This upper bound unit risk estimate should be used only under
the assumption that  DCM is  a potential human carcinogen.  As discussed pre-
viously, there is only limited animal evidence and no human evidence to support
that assumption.
                                   5-110

-------
5.3.3.5  Conclusions.  Animal  studies  show a statistically positive salivary
gland sarcoma response in male rats (Dow Chemical Company, 1980) and a border-
line hepatocellular  neoplastic nodule  response in the  rat (National  Coffee
Association, 1982).   There is also evidence  that  DCM is weakly mutagenic.
According to the  criteria of the International Agency for Research on Cancer
(IARC),  the  weight of evidence  for carcinogenicity  in animals is  limited.
     There was an absence of epidemiologic evidence for the carcinogenicity of
DCM in a well-conducted epidemiologic study having long-term exposure.  However,
on the basis  of animal data, the level of exposure to the individuals in the
study was too  low to produce  an observable  increase  in  cancer.  The  overall
evaluation of DCM, based on IARC criteria, is Group 3, meaning that the chemical
cannot be classified as to its carcinogenicity for humans.
     The upper-limit  unit risk for DCM, based on  a rat  inhalation  study,  is
estimated to be roughly  1.8  x 10~  for a  lifetime  exposure to  1 pg/m   in air.
The above is true only under the assumption that DCM is a potential human car-
cinogen, although the likelihood  of  this  is highly uncertain  given the
currently assessed data  base.   Even under  that assumption, the potency of  DCM
is the lowest of the 53 chemicals that the CAG has evaluated as suspect carci-
nogens.  The  Environmental Health Committee of EPA's  Science Advisory Board
(SAB)  indicated that  the  unit risk  estimation was "scientifically"  unsup-
portable because  of  the  questionable value of the  animal  study upon which  the
unit  risk  estimation was based, and that  consideration should be  given to
deleting the  analysis from this document.  The  SAB noted, however, that the
unit risk  estimates  may be useful for risk management purposes.  In  light of
the potential  need for risk management analysis, the risk estimation  section
has been retained.
     Preliminary  results  of  a new  NTP  study in mice  indicate that  DCM induces
a  high  incidence of lung and  liver tumors in both sexes.  When the study  is
completed and  available,  the results will  be evaluated, and the qualitative
and quantitative portions of this assessment updated.   For  this reason the
present  report  is regarded as  interim.
                                    5-111

-------
5.4  REFERENCES
Abrahamson, S., and  R. Valencia.  1980.   Evaluation  of  substances  of interest
     for genetic damage  using Drosophila  melanogaster.   Prepared for FDA
     Contract  No. 233-77-2119.

ACGIH (American Conference of Governmental  Industrial Hygienists).   1980.
     Threshold Limit Values for Chemical  Substances  and Physical Agents in  the
     Workroom  Environment with Intended Changes  for  1980.   Cincinnati, OH:
     ACGIH, p. 23.

Adams, J.D., and H.H. Erickson.  1976.  The effects  of  repeated  exposure  to
     methylene chloride  vapor.  Prep. Annu. Sci. Meet.,  Aerosp.  Med.  Assoc.
     61-62.

Ahmed, A.E., and M.W. Anders.  1978.  Metabolism of  dihalomethanes  to formal-
     dehyde and inorganic halide.   II.  Studies  on the  mechanism of the re-
     action.   Biochem. Pharmacol. 27:2021-2025.

Alexeeff, G. V., and W.W. Kilgore.  1983.   Learning  impairment in mice follow-
     ing acute exposure  to dichloromethane and carbon tetrachloride.  J.
     Toxicol.  Environ. Health 11:569-581.

Aviado, D.M.,  and M.A. Belej.  1974.  Toxicity of aerosol propellents on  the
     respiratory and circulatory systems.   I. Cardiac arrhythmia in the mouse.
     Toxicol.  2:31-42.

Aviado, D.M.,  S.  Zakhari, and T. Watanabe.  1977.  Non-fluorinated  propellants
     and solvents for aerosols.  CRC Press, Cleveland.

Aviado, D.M.   1975.   Toxicity of aerosol  propellants in  the respiratory and
     circulatory systems.  X.  Proposed Classification.  Toxicol.  3:321-332.
Ballantyne, B., M F. Gazzard, and D.W. Swanston.  1976.
     cology of dichloromethane.  Toxicol. 6:173-187.
The ophthalmic toxi-
Balmer, M.F., F.A. Smith, L.J. Leach, and C.L. Yuille.  1976.  Effects in the
     liver of methylene chloride inhaled alone and with ethyl alcohol.  Amer.
     Ind. Hyg. Assoc. J. 37:345-352.

Barber, E.D., W.H. Donish, and K.R. Mueller.  1981.  A procedure for the
     quantitative measurement of volatile liquids in the Ames Salmonella/
     microsome assay.  Mutat. Res. 90:31-48.

Barber, E.D., W.H. Donish, and K.R. Mueller.  1980.  Quantitative measurement
     of the mutagenicity of volatile liquids in the Ames Salmonel1 a/microsome
     assay.  Abstract.  P-39, Environmental Mutagen Society llth Annual Meeting.

Barrowcliff, D.F., and A.J.  Knell.  1979.  Cerebral damage due to endogenous
     chronic carbon monoxide poisoning caused by exposure to methylene chloride.
     J. Soc. Occup. Med. 29:12-14.
                                    5-112

-------
Barrowcliff, D.F.  1978.  Chronic carbon monixide poisoning caused by methylene
     chloride paintstripper.  Med. Sci. Law. 18(4):238.

Belej, M.A., D.G. Smith, and D.M. Aviado.  1974.  Toxicity of aerosol propel-
     lants  in the respiratory and circulatory systems.  IV.  Cardiotoxicity  in
     the monkey.  Toxicol. 2:381-395.

Berger, M., and G.G. Fodor.  1968.  CNS disorders under the influence of air
     mixtures containing dichloromethane.  Zentralbl.  Bakteriol.  (Ref.) 215:
     517.

Bergman, K.  1978.  Application  of whole body autoradiography to  distribution
     studies of organic solvents.  Int. Symp. Control  Air Pollut. Work Environ.
     (Proc.) Part 2, 128-139.

Bonventre,  J., 0. Brennan, D. Jason, A. Henderson, and M. L. Basots.  1977.
     Two deaths following accidental inhalation of dichloromethane and 1,1,1-tri-
     chloroethane. J. Anal. Toxicol. 1(4):158-160.

Bornschein, R.L., L. Hastings, and J.  Manson.  1980.   Behavioral  toxicology  in
     the offspring of rats following maternal exposure to dichloromethane.
     Toxicol. Appl. Pharmacol. 52:29-37.

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

Burek, J.D., K.D. Nitschke, T.J.  Bell,  D.L.  Wackerle,  R.C.  Childs, J.E. Beyer,
     D.A.  Dittenber, L.W.  Rampy, and M.J. McKenna.   1984.   Methylene Chloride:
     A Two-year  Inhalation Toxicity and Oncogenicity Study  in Rats and Hamsters.
     Fund.  Appl.  Tdxicol.  4:30-47.

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

Clark, D.G.  and  D.J. Tinston.  1973.   Correlation  of the cardiac sensitizing
     potential  of halogenated hydrocarbons  with  their physiochemical properties.
     Br.  J.  Pharmacol.  49(2):355-357.
 Collier,  H.   1936.
      of two  cases.
Methylene dichloride intoxication in industry — a report
Lancet 1:594-595.
 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.

 Crump,  K.S.,  and W.W.  Watson.  1979.   GLOBAL79.   A Fortran program to extrapo-
      late dichotomous animal carcinogenicity data to low dose.  Natl. Inst. of
      Environ.  Health Science Contract No. l-ES-2123.

 Douglas, B.H., J.S.  Wilkinson, R.F.  Williard, and W.L.  Williams.  1976.
      Effect of dichloromethane on the blood pressure of spontaneously hyper-
      tensive rats (abstract).  IRCS Med. Sci. Libr. Compend. 4:507.
                                     5-113

-------
 Dow Chemical  Company Report,  1979.

 Dow Chemical  Company.   1980.   Methylene chloride:  a two-year inhalation toxicity
      and oncogenicity study in rats and hamsters.   FYI-OTS-0281-0097.   Follow-up
      response A.  Washington,  D.  C.:   U.  S.  Environmental  Protection Agency,
      Office of Toxic Substances.

 Dow Chemical  Company.   1982.   Nitschke,  K.D.,  T.J.  Bell,  L.W.  Rampy, and M.J.
      McKenna.   Methylene chloride:   A two-year inhalation toxicity and onco-
      genicity study in rats.   Toxicology Research  Laboratory,  Health and
      Environmental  Sciences,  Dow Chemical  Company,  Midland,  MI  (October 11,
      1983).
 Dripps,  R.D.,  J.E.  Eckenhoff,  and L.D.  Vandam.   1977.
      thesia; the  Principles  of Safe  Practice.   5th ed.
      W.B.  Saunders  Co.,  pp.  121-123.
                                       Introduction to Anes-
                                       Philadelphia, PA:
Elovaara,  E.,  K.  Hemminki,  and  H.  Vainio.   1979.   Effects  of methylene chloride,
     trichloroethane,  trichloroethylene,  tetrachloroethylene and toluene on
     the development of  chick embryos.  Toxicology 12:111-119.

Environmental  Criteria and  Assessment Office.   1979.   Air  Quality Criteria for
     Carbon Monoxide.  EPA-600/8-79-022,  U.S.  Environmental  Protection Agency,
     Washington,  DC, October.

Essman, W. B., and H.  Alpern.   1964.  Single trial  conditioning:  Methodology
     and results  with  mice.   Psychol. Rep.  14:731-740.

FASEB.  1974.  Biological data  books.  2nd. ed., Vol.  III.   Edited by  Philip
     L. Altman and Dorothy  S. Dittmen.  Federation  of  American  Societies for
     Experimental Biology,  Bethesda, MD.   Library  of Congress No.  72-87738.
Filippova, L.M.
     8:134-137.
1967.  Genetic activity of germinal systems.  Genetika
Flury, F., and F. Zernik.  1931.  Harmful Gases, Vapors, Fogs, Smoke and  Dust.
     Berlin:  Julius Springer, pp. 311-312.

Fodor, G.G., and A. Roscovanu.  1971.  Increased blood CO content  in humans
     and animals experimentally exposed to industrial solvent vapors in England.
     HM (ed.):  Proceedings of the 2nd International Clean Air Congress.  New
     York:   Academic Press, pp. 238-243.

Fodor, G.G., and A. Roscovanu.  1976.  Increased blood CO content  in humans
     and animals caused by incorporated halogenated hydrocarbons.  Zbl. Bakt.
     Hyg. 162:34-40.

Fodor, G.G., and G. Winneke.  1971.  Nervous system disturbances in men and
     animals experimentally exposed to industrial solvent vapors in England.
     Proceedings of the 2nd International Clean Air Congress.  New York:
     Academic Press,  pp. 238-43.

Fodor, G.G., D.  Prajsnar, and H.W. Schlipkoter.  1973.  Endogenous CO formation
     by incorporated halogenated hydrocarbons of the methane series.   Staub-
     Reinhalt Luft. 33:260-261.
                                    5-114

-------
Forster, H.U.,  S.  Graff, C.L. Hake, R. Soto, and R.D. Stewart.  1974.
     Pulmonary - Hematologic studies on humans during exposure to methylene
                Report No. N10SH-MCOW-ENVM-MC-74-4.  The Medical College of
                Dept. of Environmental Medicine, Milwaukee, Wis., 17 pp,
chloride.
Wisconsin
April.
Freireich, E.J., E.A. Gehan, D.P. Rail, L.H. Schmmidt and H.E. Skipper.  1966.
     Quantitative comparison of toxicity of anticancer agents in mouse, rat,
     hamster, dog, monkey, and man.  Cancer Chemother. Rep.  50: 219.

Friedlander, B.R., F.T. Hearne, and S. Hall.  1978.  Epidemiologic investiga-
     tion of employees chronically exposed to methylene chloride.  J. Occup.
     Med. 20:657-666.
Gamberale, F., B.A. Annwall, and M. Hultengren.  1975.
     chloride — II.  Psychological functions.  Scand.
     Health 1(2):95-103.
                                                   Exposure to methylene
                                                  J. Work. Environ.
Gocke, E., M.T. King, K. Eckhardt, and D. Wild.  1981.  Mutagenicity of cos-
     metics ingredients licensed by the European communities.  Mutat. Res.
     90:91-109.

Gradiski, D., J.L. Megadur, M. Baillot, M.C. Daniere, and M.B. Schuh. 1974.
     Comparative toxicity of the principle chlorinated aliphatic solvent. J.
     Eur. Toxicol. 7:247.

Green, T.  1980.  The metabolism and mutagenicity of methylene chloride.
     Abstracts of papers, Society of Toxicology, Inc. 19th annual meeting,
     Wash., D.C., March 9-13, 1980.

Green, T.  1981.  The metabolic activation of dichloromethane and chlorofluoro-
     methane in a bacterial mutation assay using Salmonella typhimurium.
     Unpublished Manuscript.  Imperial Chemical Industries, PLC, Central
     Toxicology Laboratory, Alderly Park, Macclesfield, Cheshire SK104, United
     Kingdom.

Hake, C.L., R.D. Stewart, H.U. Forster, A.J. Lebrun, J.E. Peterson, and A. Wu.
     1974.  Results of the controlled exposure of human females to the vapor
     of methylene chloride.  Report. No. NlOSH-MCOW-ENVM-MC-74-3. Milwaukee,
     Wis.  The Medical College of Wisconsin, Dept. of Environmental Medicine,
     22 pp, March.

Hamada, N. and R.E. Peterson.  1977.  Effect of chlorinated aliphatic hydrocar-
     bons on excretion of protein and electrolytes by rat pancreas.  Toxicol.
     Appl. Pharmacol. 39:185.

Hardin, B.D., and J.M. Manson.  1980.  Absence of dichloromethane teratogenicity
     with  inhalation exposure in rats.  Toxicol. Appl. Pharmacol.  52:22-28.

Harms, J.S., R.E. Peterson, J.M. Fujimoto, and C.P.  Erwin.  1976.  Increased
     "bile duct-pancreatic fluid" flow in chlorinated hydrocarbon-treated
     rats. Toxicol. Appl. Pharmacol.  35:41-49.
                                     5-115

-------
 Haun,  C.C.,  E.S.  Harris,  and K.I.  Darmer.   1971.   Continuous animal exposure
     to  methylene chloride.   Am.  RL-TR-71-120,  #10.   In:  Proceed.  2nd Conf.
     Environ.  Toxicol.  Wright-Patterson AFB,  Ohio,   pp 125-135.

 Haun,  C.C.,  E.H.  Vernot,  K.I.  Darmer,  Jr.,  and S.S.  Diamond.   1972.  Continuous
     animal  exposure to low levels of  dichloromethane AMRL-TR-130, paper No.
     12.   In:   Proceedings  of the 3rd  Annual  Conference on Environmental
     Toxicology,  Wright-Patterson Air  Force Base,  Ohio, Aerospace  Medical
     Research  Laboratory, pp.  199-208.
Hearne,  F.T.,  and  B.R.  Friedlander.
     study.  J.  Occup.  Med.  23:660.
1981.   Follow-up of methylene chloride
Heppel,  L.A.,  and  P.A.  Neal.   1944.   Toxicology of dichloromethane (methylene
     chloride) ~  II.   Its  effect  on  running activity in the male rat.   J.
     Ind. Hyg. Toxicol.  26:17-21.

Heppel,  LA.,  P.A. Neal,  T.L.  Perrin,  M.L.  Orr,  and V.T.  Porterfield.   1944.
     Toxicology of dichloromethane (methylene chloride).   I.  Studies on effects
     of  daily  inhalation.   J.  Ind.  Hyg.  Toxicol.  26:8-16.

Hogan, G.K., R.G.  Smith,  and  H.H.  Cornish.   1976.   Studies on the microsomal
     conversion of dichloromethane to  carbon monoxide.   Toxicol.  Appl.  Pharmacol
     37:112.
Hughes, J.P.  1954.  Hazardous exposure  to  some  so-called  safe  solvents.
     Med. Assoc. 156:234-237.
                                     J.
International Commission on Radiological  Protection  (ICRP).   1977.   Recom-
     mendation of the International Commission on  Radiological  Protection.
     Publ. No. 26, adopted Jan. 17, 1977.  Pergamon  Press, Oxford,  England.

Johnson, E.M.  1981.  Screening for teratogenic  hazards,  nature of  the  problems.
     Ann. Rev. Pharmacol. Toxicol.  21:417-429.

Jongen, W.M.F., G.M. Alink, and J.H. Koeman.  1978.  Mutagenic  effect of
     dichloromethane on Salmonella typhimurium.  Mutat. Res.  56:245-248.

Jongen, W.M.F., E.G.M. Harmson, G.M. Alink, and  J.H. Koeman.  1982.  The
     effect of glutathione conjugation and microsomal oxidation on  the  muta-
     genicity of dichloromethane in S^_ typhimurium.  Mutat.  Res. 95:183-189.

Jongen, W.M.F., P.H.M. Lohman, M.J. Kettenhagen,-G.M. Alink,  F.  Berends, and
     J.H. Koeman.  1981.  Mutagenicity testing of  dichloromethane in short-term
     mammalian test systems.  Mutat. Res. 81(2): 203-213.

Kahn, A., R.B. Rutledge, G.L.  Davis, J.A. Altes, G.E. Gantner,  C.A. Thornton,
     and N.D. Wallace.  1974.   Carboxyhemoglobin sources  in  the metropolitan
     St. Louis population.  Arch. Environ. Health  29:127-135.

Kanada, T., and M.  Uyeta.  Mutagenic screening of  organic solvents  in microbial
     systems.  Mutat. Res. 54:215, 1978.  (Abstract)
                                    5-116

-------
Kimura, E.T., D.M. Ebert, and P.W. Dodge.  1971.  Acute toxicity and  limits of
     solvent residue for sixteen organic solvents.  Toxicol. Appl. Pharmacol.
     19:699-704.

Klaassen, C.D.  and G.L. Plaa.  1966.  The relative effects of various chlori-
     nated hydrocarbons on liver and kidney function in mice.  Toxicol.  Appl.
     Pharmacol. 9:139-131.

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

Kluwe, W.M., F.W. Harrington, and S.E. Cooper.  1982.  Toxic effects  of organ-
     ohalide compounds on renal tubular cells J_n vivo and i_n vitro.   J. Pharm.
     Exper. Therap. 230(3): 597-603.

Kubic, V.L., and M.W. Anders.  1975.  Metabolism of dihalomethanes to carbon
     monoxide.   II. In vitro studies.  Drug Metab. Dispos.  3:104-112.

Kuzelova, M., J. Cerny, S. Havova, M. Hub, V. Kunor, and A. Popler.   1975.
     Lethal methylene chloride poisoning with severe chilblains (Czech.).
     Prac. Lek. 27(9):317-319.

Loyke, H.F.  1973.  Methylene chloride and chronic renal hypertension.  Arch.
     Pathol. 95:130-131.

MacEwen, J.D.,  E.H. Vernot, and C.C. Haun,  1972.  Continuous animal  exposure
     to dichloromethane.  AMRL-TR-72-28, Systems Corporation Report No. W-71005.
     Wright-Patterson Air Force Base, Ohio, Aerospace Medical Research.
Mantel, N., and M.A. Schneiderman.  1975.  Estimating "safe"  levels:
     dous undertaking.  Cancer Res. 35:1379-1386.
a hazar-
Matthews, R.M., D.H. Martin, I.D. Kuntz and T.L. James.  1977.  Antisickling
     behavior of dichloromethane Jji vitro (abstract).  Blood 50(5):113.

McCarroll, N.E., T.A. Cortina, M.J. Zito, and M.G. Farrow.  1983.   Evaluation
     of methylene chloride and vinylidene chloride in mutational assays.
     Environ. Mutagen 5(3):426-427.

McGregor, D.B.  1979.  Practical experience in testing unknowns ni  vitro.
     Chapter 4.  Pages 53-71 in G.E. Paget, ed. Topics in Toxicology.   Muta-
     genesis in submammalian systems:  Status and significance.  Baltimore,
     MD: University Park Press.

Morris, J.B., F.A. Smith and R.H. Garman.  1979.  Studies on methylene  chloride
    .induced fatty liver.  Exp. Mol. Pathol. 30:386-393.

Moskowitz, S., and H. Shapiro.  1952.  Fatal exposure to methylene  chloride
     vapor.  Arch. Ind. Hyg. Occup. Med. 6:116-123.

National Coffee Association.  1982a.  24-month chronic toxicity and oncp-
     genicity study of methylene chloride in rats.   Final report.   Prepared by'
     Hazelton Laboratories America, Inc., Vienna, VA.  (August 11,  1982).
     Unpublished.

                                    5-117

-------
National Coffee Association.  1982b.  24-months chronic toxicity and onco-
     genicity study of methylene chloride  in rats.  Addition to the final
     report.  Prepared by Hazelton  Laboratories America,  Inc., Vienna, VA
     (Nov. 5, 1982).  Unpublished.

National Coffee Association.  1983.  24-month oncogenicity study of methylene
     chloride in mice.  Final Report.  Prepared by Hazelton Laboratories
     America, Inc., Vienna, VA.  Unpublished (Nov. 30, 1983).

National Toxicology Program (NTP).  1982.  Draft technical report on the
     carcinogenesis bioassay of dichloromethane (methylene chloride), gavage
     study.  Research Triangle Park, NC, and Bethesda, MD.

Nestmann, E.R., E.G.-H. Lee, T.I. Matula,  G.R. Douglas, and J.C. Mueller.
     1980.  Mutagenicity of constituents identified in pulp and paper mill
     effluents using the Salmone11 a/mammalian microsome assay.  Mutat. Res.
     79:203-212.

Nestmann, E.R., R. Otson, D.T. Williams, and D.J. Kowbel.  1981.  Mutagenicity
     of paint removers .containing dichloromethane.  Cancer Lett. 11:295-302.

Norpoth, K., U. Witting, M. Springorum and C. Witting.  1974.  Induction of
     microsomal enzymes in the rat  liver by inhalation of hydrocarbon solvents.
     Int. Arch. Arbeitsmed.  33(4):315-321.

Ott, M. G., L. K. Skory, B. B. Holder, J. M. Bronson, and P. R. Williams.
     1983a. Health evaluation of employees occupationally exposed to methylene
     chloride. General study design and environmental considerations.  Scand. J.
     Health 9(Suppl. l):l-7.

Ibid.  1983b.  Health evaluation of employees occupationally exposed to methy-
     lene chloride.  Mortality.  Scand. J. Work Environ. Health 9:8-16.

Ibid.  1983c.  Health evaluation of employees occupationally exposed to methy-
     lene chloride.  Clinical Laboratory Evaluation.  Scand. J. Work Environ.
     Health 9:17-25.

Ibid.  1983d.  Health evaluation of employees occupationally exposed to methy-
     lene chloride.  Twenty-four hour electrocardiographic monitoring.  Scand.
     J. Work Environ.  Health 9:26-30.

Ibid.  1983e.  Health evaluation of employees occupationally exposed to methy-
     lene chloride.  Metabolism data and oxygen half-saturation pressures.
     Scand. J. Work Environ. Health 9:31-38.

Pankow, D., R. Gutewort, W. Glatzel, and K. Tietze.  1979.  Effect of dichloro-
     methane on the sciatic motor conduction velocity of rats.  Experentia
     35:373-374.

Perocco, P., and G. Prodi.  1981.  DNA damage by haloalkanes in human lympho-
     cytes cultured in vitro.  Cancer Lett. 13:213-218.
                                    5-118

-------
Peterson, J.  1978.  Modeling the uptake, metabolism and excretion of dichloro-
     methane by man.  Am. Ind. Hyg. Assoc. 39(l):41-47.

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

Price, P. J., C. M. Hassett, and J. I. Mansfield.  1978.  Transforming activi-
     ties of trichloroethylene and proposed industrial alternatives.  Iji Vitro
     14:290-293.

Pryor, G. T., R. A. Howd, R. Malik, R. A. Jensen and C. S. Robert.  1978.
     Biomedical studies on the effects of abused inhalant mixtures.  Quart.
     Prog.  Rep. #7. Contract #271-77-3402.  Rockville, MD.  Nat.  Inst. Drug.
     Abuse.

Putz, V.  R., B. L. Johnson and J. V. Setzer.  1976.  A comparative study of
     the effect of carbon monoxide and methylene chloride on human performance.
     J. Environ. Pathol. Toxicol. 2:97-112.

Rail, D.P.   Difficulties in extrapolating the results of toxicity studies  in
     laboratory animals to man.  1969.  Environ. Res.  2: 360-367.

Rapson, W.H., M.A. Nazar, and V.V. Butsky.  1980.  Mutagenicity produced by
     aqueous chlorination of organic compounds.  Bull. Environ. Contam. Toxicol.
     24:590-596.

Reiter, L.,  G,  Talens, and D. Woolley.  1973.   Acute and subacute parathion
     treatment.  Effects on cholinesterase activities and learning in mice.
     Toxicol. Appl. Pharmacol. 25:582-588.

Reynolds, E. S., and A. G.  Yee.  1967.  Liver parenchymal cell injury.  V.
     Relationships between patterns of chloromethane C14 incorporation into
     constituents of liver in vivo and cellular injury.  Lab. Invest. 16:591-
     603.

Sahu, S.C.,  and O.K. Lowther.  1981.  Pulmonary reactions to inhalation of
     methylene chloride:  Effects on lipid peroxidation in rats.  Tox. Lett.
     8:253-256.

Samoiloff,  M.R., S. Schulz, Y. Jordan, K. Denich, and E. Arnott.  1980.  A
     rapid simple long-term toxicity assay for aquatic contaminants using  the
     nematode Panagrellus redivivus.  Can. J.  Fish Aquat. Sci. 37:1167-1174.

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

Schutz, E.   1958.  Effect of polyethylene glucol 400 on percutaneous absorption
     of active  ingredients.  Arch. Exp.  Pathol. Pharmakol. 323:237-238.

Schwetz, B.  A., B. K. J. Leong, and P. J. Gehring.  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.

                                    5-119

-------
Simmon, V.F., and K. Kauhanen.  1978.  In vitro microbological mutagenlclty
     assays of 2-chlorethyl chloroformate.  Final report, Contract No.
     68-03-11-74.  Prepared for U.S. Environmental Protection Agency, National
     Environmental Research Center, Water Supply Laboratory, Cincinnati, OH
     45268.

Simmon, V. F., V. Kauhanen, and R. G. Tardiff.  1977.  Mutagenic activity of
     chemicals identified in drinking water.  Dev. Toxicol. Environ. Sci.
     2:249-258.

Skory, L.K.  1980.  Health Surveillance of Employees Occupationally  Exposed to
     Methylene Chloride.  V.  A Review of Effects on Oxygen Transport.  Scand.
     J. Work. Environ. Health (in press), 1980.  Cited in Burek et al.

Skory, L.K., M.G. Ott, P.R. Williams, J.M. Bronson and B.B. Holder.  1980.
     Health Surveillance of Employees Occupationally Exposed to Methylene
     Chloride.  III. Clinical Pathological Evaluation.  Scand. J. Work  Environ.
     Health (in press), 1980a.  Cited in Burek et al.

Skory, L.K., M.G. Ott, P.R. Williams, J.M. Bronson and B.B. Holder.  1980.
     Health Surveillance of Employees Occupationally Exposed to Methylene
     Chloride.  IV. 24-Hour EKG Monitoring.  Scand. J. Work Environ. Health
     (in press), 1980b. Cited in Burek et al.

Snow, L., P. McNair, and  B.C. Castro:  1979.  Mutagenesis testing of methylene
     chloride and 1,1,1-trichloroethane in Salmonella strains TA-100 and
     TA-98.  Personal Communication from Northrop Services, Inc., P.O.  Box
     12313, Research Triangle Park, NC, 27709, September 19

Stewart,  R. D., and C. L. Hake.  1976.  Paint remover hazard.  J. Med.  Assoc.
     235(4):398-401.

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

Stewart,  R. D., H. V. Forster, C. L. Hake, A. J. Lebrun, and J. E. Peterson.
     1973.  Human responses to controlled exposure of methylene chloride
     vapor.  Report No. NIOSH-MCOW-ENVM-MC-73-7.  Milwaukee, WI, Dept.  Environ-
     mental Medicine, December.  82 p.

Stewart,  R. D., T. N. Fisher, M. J. Hosko, J. E. Peterson, E. D. Baretta, and
     H.  C. Dodd.  1972a.  Carboxyhemoglobin elevation after exposure to dichlo-
     romethane. Science 176(4032):295-296.

Stewart,  R. D., T. N. Fisher, M. J. Hosko, J. E. Peterson, E. D. Baretta, and
     H.  C. Dodd.  1972b.  Experimental human exposure to methylene chloride.
     Arch. Environ. Health 25:342-348.

Svirbely, J. L., B. Highman, W. C. Alford, and W. F. Von Oettingen.  1974.
     The toxicity and narcotic action of mono-chloro-mono-bromo-methane with
     special reference to inorganic and volatile bromide in blood, urine and
     brain.  J. Ind. Hyg. Toxicol. 29:383-389.
                                    5-120

-------
Tariot, P. N.  1983.  Delirium resulting from methylene chloride exposure:
     Case report.  J. Clin. Psychiatry 44:340-342.

Taylor, G. J., R. T. Drew, E. M. Lores and' lY A. Clemmer:  1976.  Cardiac
     depression by haloalkane propellants, solvents, and inhalation anesthetics
     in rabbits. Toxicol. Appl. Pharmacol. 38:379-387.

Theiss, J. C., G. D. Stoner, M. B. Shimkin, and E. K. Weisburger.  1977.  Test
     for carcinogenicity of organic contaminants of United States drinking
     waters by pulmonary tumor response in strain A mice.  Cancer Res. 37:
     2717-2720.

Thilagar, A.K. and V. Kimaroo.  1983.  Induction of chromosome damage by
     methylene chloride in CHO cells.  Mut. Res. 116: 361-367.

Thomas, A. A., M. K. Pinkerton, and J. A. Warden.  1972.  Effects of low level
     dichloromethane exposure on the spontaneous activity of mice.  AMRL-TR72-
     130, paper No. 14.  In:  Proceedings of the 3rd Annual Conference on
     Environmental Toxicology, Wright-Patterson Air Force Base, Ohio, Aerospace
     Medical Research Laboratory, pp. 223-227.

Thomas, A. A., M. K. Pinkerton, and J. A. Warden.  1971.  Effects of methylene
     chloride exposure on the spontaneous activity of mice.  AMRL-TR-H-120.
     Paper No. 13.  In:  Proceedings of the 2nd Annual Conference on Environ-
     mental Toxicology, Wright-Patterson Air Force Base, Ohio, Aerospace
     Medical Research Laboratory,  p. 185-189.

Ugazio, G., E. Budino, 0. Danni and Milillo:  Hepatotoxicity and lethality of
     haloalkanes.  1973.  Biochem. Soc. Trans. 1:968-972.

U.S. Environmental Protection Agency.  1976.  Interim procedures and guidelines
     for health risk and economic impact assessments of suspected carcinogens.
     Federal Register 41:21402 (May 25).

U.S. Environmental Protection Agency.  1978.  Proposed Guidelines for Regis-
     tering Pesticides in the United States.  Fed. Regist. 43(163):37382-37888,
     August 22.

U.S. Environmental Protection Agency.  1979.  Halomethanes:  Ambient Water
     quality criteria.  NTIS PB-296797.  National Technical Information Service,
     Springfield, VA.

U.S. Environmental Protection Agency.  1980.  Determination not to initiate a
     rebuttable presumption against registration (RPAR) of pesticide products
     containing carbaryl:  Availability of decision document.  Fed. Regist.
     45:81869-81876, December 12.

U.S. Environmental Protection Agency.  1979.  Proposed Health Effects Test
     Standards for Toxic Substances Control Act Test Rules and Proposed Good
     Laboratory Practice Standards for Health Effects.  Fed. Regist. 44(145):
     44089-44092, July 26.

U.S. EPA (U.S. Environmental Protection Agency).  1980a.  Ambient Water Quality
     Criteria for Halomethanes.  U.S. EPA.  Available from:  National Technical
     Information Service, Springfield, VA (NTIS PB81-117624).

                                    5-121

-------
U.S. EPA (U.S. Environmental Protection Agency).  1980b.  Guidelines and
     Methodology for the Preparation of Health Effect Assessment Chapters of
     the Ambient Water Quality Criteria Documents.  U.S. EPA, Environmental
     Criteria and Assessment Office; Office of Health and Environmental Assess-
     ment; Office of Research and Development, Cincinnati, OH, November 28.

U.S. EPA (U.S. Environmental Protection Agency).  1981.  Advisory Opinion for
     Dichloromethane (Methylene Chloride).  Office of Drinking Water, U.S.
     EPA, Washington, DC.  Draft.

Valencia, R.:  Contract report to FDA, 1978.

Von Oettingen, W. F., C. C. Powell, N. E. Sharpless, W. C. Alford, and L. J.
     Pecora.  1949.  Relation between the toxic action of chlorinated methanes
     and their chemical and physiochemical properties.  NIH Bulletin 191.

Weinstein, R. S., and S. S. Diamond.  1972.  Hepatotoxicity of dichloromethane
     (methylene chloride) with continuous inhalation exposure at a low-dose
     level.  AMRL-72-130, paper No. 13.  In:  Proceedings of the 3rd Annual
     Conference on Environmental Toxicology, Wright-Patterson Air Force Base,
     Ohio, Aerospace Medical Research Laboratories, pp. 209-222.

Weinstein, R. S., D. D. Boyd., and K. C. Back.  1972.  Effects of continuous
     inhalation of dichloromethane in the mouse—morphologic and functional
     observations.  Toxicol. Appl. Pharmacol. 23:660-79.

Weiss, G.  Toxic encephalosis as an occupational hazard with methylene chloride.
     1967.  Zentrabl. Arbeitsmed. 17:282-85

Wilkinson, J. S., B. H. Douglas, R. F. Williard and W. L. Williams:  Reduction
     of blood pressure in the spontaneously hypertensive rat (SHR) with methy-
     lene chloride (CH2CL2) (Abstract).  Clin. Res. 25:48A, 1977.

Winneke, C.L., W.D. Collom, and F. Esposito.  1981.  Accidental methylene
     chloride fatality.  Forensic Sci. Intern. 18:165-168.

Winneke, G.  1974.  Behavioral effects of methylene. chloride and carbon monoxide
     as assessed by sensory and psychomotor performance.  In:  C. Xintaras, B.
     L. Johnson, and I. DeGroot, eds., Behavioral Toxicology—Early Detection
     of Occupational Hazards, Publication HEW No. (NIOSH) 74-126.  U.S. Dept.
     of Health, Education and Welfare, Public Health Service, Center for
     Disease Control, National Institute for Occupational Safety and Health.
     p. 130-144.

Winneke, G. and G. G. Fodor:  Dichloromettiane produces narcotic effects.
     Occup. Health Safety.  Mar/Apr:34-37, 1976.

Zakhari, S.  1977.  Methylene chloride.  Chapter 2  In: Non-fluorinated propel-
     lants and solvents for aerosols.  L. Golberg, ed.  CRC Press, Cleveland.
                                     5-122

-------
                                   APPENDIX
             COMPARISON OF RESULTS BY VARIOUS EXTRAPOLATION MODELS
     The estimates  of  unit  risk based  on  animal  studies  presented  in  the  body
of this  document  are all calculated by the  use  of  the linearized  multistage
model.  The  reasons for  its use have been detailed  herein.   Essentially,  this
model is part  of  a methodology that estimates a conservative linear slope at
low extrapolation  doses  and is  consistent with the  data  at  all  dose levels of
the experiment.  It is a nonthreshold model which holds that the upper-limit of
risk predicted  by  a linear extrapolation  to  low levels  of  the  dose-response
relationship is the most plausible upper limit for the risk.
     Other models  have also  been used for  extrapolation,  and  include the
three nonthreshold  models presented  here:  the one-hit, the log-Probit,  and
the Weibull.   The one-hit  model  is  characterized by a continuous downward
curvature,  but is linear at low doses.   It can be considered the linear form or
first stage of the multistage model because of its functional form.  Because of
this and its downward curvature, the one-hit model will  always  yield estimates
of low-level  risk that are at least as  large as those of the multistage model.
Further, whenever  the  data  can  be fitted  adequately by means of the one-hit
model, estimates from the two procedures will be comparable.
     The other  two  models,  the  log-Probit and the Weibull,  are often  used to
fit toxicological  data in  the observable range,  because  of their general  "S"
curvature.   The low-dose upward curvatures of these two  models  usually yield
lower low-dose  risk estimates than those of the one-hit  or multistage model.
     The log-Probit model  was  originally proposed for use  in  problems of
biological  assay,  such as the assessment of potency of toxicants  and drugs,
and has usually been  used  to estimate such values as percentile lethal dose
or percentile effective  dose.   Its development was strictly empirical, i.e.,
it was  observed that several log dose-response  relationships  followed the
cumulative  normal  probability distribution function.   In fitting  the cancer
bioassay data,  assuming an  independent  background, this  becomes:

          P(D;a,b,c) = c  +  (1-c) * (a+blog10D)   a,b <  c  <  1
                                    A-l

-------
where P  is the proportion responding at dose D, c is an estimate of the back-
ground rate,  a is an estimate of the standarized mean of individual  tolerances,
and b is an estimate of the log dose-Probit response slope.
     The one-hit model  arises  from the theory that  a  single molecule of a
carcinogen has a probability of transforming a single  noncarcinogenic cell
into a carcinogenic one.  It has the probability distribution function:

                    P(D;a,b) = l-exp-(a+bd)   a,b > 0

where a  and  b are the parameter estimates.  The  estimate a represents the
background or zero  dose  rate,  and  the parameter estimated by b represents the
linear component or slope of the dose-response model.  In discussing the added
risk over background, incorporation of Abbott's correction leads to

                       P(D;b) = l-exp-(bd)   b > 0

Finally, a model  from the theory  of carcinogenesis  arises from the multihit
model applied to  multiple target cells.   This model has been termed here the
Weibull model.  It is of the form

                       P(D;b,k) = l-exp-(bdk)   b,k > 0

For  the  power of dose only, the restriction  k >  0  has been placed on this
model. When  k >  1,  this model yields low-dose estimates  of risks usually
significantly lower  than either the multistage or  one-hit models, which are
linear at low doses.  All three of these models usually project risk estimates
significantly higher at the low exposure levels than those from the log-Probit.
     The estimates  of added risk for low  doses  for  the  above models are given
in Table A-l for the DCM inhalation study.  Both  maximum  likelihood estimates
and  95%  upper confidence  limits are  presented.  All estimates incorporate
Abbott's correction for  independent background rate.
                                    A-2

-------
  :3
  : LU ซ
      cu
      +J
      fU
      S-
O
(_} LU
a: ce -o
- a a.
o: LU m
   a s-
      O
O l/> •!-
LU    +->
i/> z u

      •U
O "• O
r- O J3
   CO J3
ie: en - 
LO LU It)
•X.    -P
ฃ 3: in
1-1 O CD
r- a
to    i—
LU LU r-
             •i—  in
              0) i—
             XI  10
             •r-  C
             It-  O
              C T-
              O 4->
              U T-
              

.a •—
O 01
t- TJ
Q- O
1 E
O)
0




, 	
'a "a!
.a -a

3S

•P
•r- i—
f CD
1 "O

O

CD
05

in CD
't— TJ
•P 0
3 S


^_>


So
Q. TJ
1 O
0) E
O
-1





jH , 	
3 CD
.0 IT
•<- O
CD E








ฃ "CD
i -a
CD O
f= E
O



ai
ai
ID
in CD
•>- TJ
•P 0

"3
Z.











CO

3.

O
rH


in
f-4
1
O
rH
X

m

CM

to
i
o
rH
X

rH
10

1
O
rH

X
O
CM
in
i
0

X

00
1-1
to
H
1
o
rH
X

rH

i-i


00
1
0

X

o

in




in

o
X

CO

r-i



to

0

X

CM

CM







CO


0)


o
o
rH



a>
t
0
rH
X

CO

rH

in

o
i-H
X

O
CM

1
O
rH

X
O
CM


0

X

CO
rH

t-i
1
O
rH
X

O

LD


to
1
O

X

CM

CM




•9"

O
X

CO

i-i



in
i
0

X

CM

CM






CO
m


-^

o
o
o
rH



i-l

0
rH
X

*4"

CM

V
1
0
1-1
X

rH
1C
CO
1
o
rH

X
o
CM
CO

o

X

00
rH

1C
1
o
i-H
X


.
rH


in
i
O

X

m

ai




CO
i
o
X

CO

r-i



^t
1
0

X

CM

CM





CO
s^
O)


o
o
o

o
rH





































,
1C
CO
in

01

JD
nj
r-
c

•a
CD

cz
CD
in
aj
i-
O.

in
03
in
CD
D)
ID
in
o

t—
CD

1o

3
CT
CD

m


f—

4j
in
c

-------

-------

-------

-------

-------

-------

-------

-------

-------
3"  9
-r  =i
•ฃ  CD
S  ฐ>
™  CO
CD


cn
CD
CO
O
O
m
•Q
i
o
o
oo

oo
N)
1
o
o
4=-
~n















c Q. =;
2 2<
^ " ฐ
2 o c
IP-Q.
|ซ|
ZT O O
III
-* *"
O -r D"
S_, ^
w o
3 n -ป
ฎ O CD
^ < O
CD ID
0) P
Q. ?



C -i
P
O "j
— w
3"
CO O
o. rn
Q. O
(3 ^
S x
" m
IP

^ Q, "U
•1-S iT
' Q Q)
IT O Ol
ซ 3-0
o^? 3
ฐงl
S? 2.

71 ซ 3
i-S?
i CO
CD O
1 -1
o S-
— 0)
3" D
CD (Q

Q."
Q. O
S 3
W 2.
U) J

D Q)
Erg"
0 ง
c ฐ
2 01
m cr
W5 CO
> m  c

ซ  32.

If  i

    3  ซป
    CD  01
    3  ซ
    0)  C/)
                          CD
                          O
                          6'
                             o
                           ,  CD
                      3  m  -*1

                      =r. o  ?

                      O •'  rn
                      I     =


                      or     o
                      NJ    "3
                      05     3
                      OS     CD
                             D

                             S)
                             0
                             cn
                             0)

                             o
                              -

-------