2,3,7,8-TETR ACHLORO -
DIBENZO-p-DIOXIN
                                   r
                                   c
Agency for Toxic Substances and Disease Registry
U.S. Public Health Service
                                   C.
                                   c
                                   3

-------
                                                       ATSDR/TP-88/23
           TOXICOLOGICAL PROFILE FOR
      2,3,7,8-TETRACHLORODIBENZO-p-DIOXIN
              Date Published — June 1989
                    Prepared by:

             Syracuse Research Corporation
            under Contract No. 68-C8-0004

                         for

Agency for Toxic Substances and Disease Registry (ATSDR)
              U.S. Public Health Service

                 in collaboration with

      U.S. Environmental Protection Agency (EPA)
       Technical editing/document preparation by:

            Oak Ridge National Laboratory
                       under
     DOE Interagency Agreement No. 1857-B026-A1

-------
                         DISCLAIMER

Mention of company name or product does not constitute endorsement by
the Agency for Toxic Substances and Disease Registry.

-------
                                FOREWORD

     The Superfund Amendments and Reauthorization Act of 1986 (Public
Law 99-499) extended and amended the Comprehensive Environmental
Response, Compensation, and Liability Act of 1980 (CERCLA or Superfund)
This public law (also known as SARA) directed the Agency for Toxic
Substances and Disease Registry (ATSDR) to prepare toxicological
profiles for hazardous substances which are most commonly found at
facilities on the CERCLA National Priorities List and which pose the
most significant potential threat to human health, as determined by
ATSDR and the Environmental Protection Agency (EPA).  The list of the 100
most significant hazardous substances was published in the Federal
Regiscer on April 17, 1987.

     Section 110 (3) of SARA directs the Administrator of ATSDR to
prepare a toxicological profile for each substance on the list.  Each
profile must include the following content:

     "(A)  An examination, summary, and interpretation of available
     toxicological information and epidemiologic evaluations on a
     hazardous substance in order to ascertain the levels of significant
     human exposure for the substance and the associated acute,
     subacute, and chronic health effects.

     (B)  A determination of whether adequate information on the health
     effects of each substance is available or in the process of
     development to determine levels of exposure which present a
     significant risk to human health of acute, subacute, and chronic
     health effects.

     (C)  Where appropriate, an identification of toxicological testing
     needed to identify the types or levels of exposure that may present
     significant risk of adverse health effects in humans."

     This toxicological profile is prepared in accordance with
guidelines developed by ATSDR and EPA. The guidelines were published in
the Federal Register on April 17, 1987. Each profile will be revised and
republished as necessary, but no less often than every three years, as
required by SARA.

     The ATSDR toxicological profile is intended to characterize
succinctly the toxicological and health effects information for the
hazardous substance being described. Each profile identifies and reviews
the key literature that describes a hazardous substance's toxicological
properties. Other literature is presented but described in less detail
than the key studies. The profile is not intended to be an exhaustive
document; however, more comprehensive sources of specialty information
are referenced.

-------
Foreword
     Each toxicological -profile begins with a public health statement.
which describes in nontechnical language a substance's relevant
toxicological properties. Following the statement is material that
presents levels of significant human exposure and,  where known,
significant health effects. The adequacy of information to determine a
substance's health effects is described in a health effects summary.
Research gaps in toxicologic and health effects information are
described in the profile. Research gaps that are of significance to
protection of public health will be identified by ATSDR, the National
Toxicology Program of the Public Health Service, and EPA.  The focus of
the profiles is on health and toxicological information; therefore, we
have included this information in the front of the document.

     The principal audiences for the toxicological profiles are health
professionals at the federal, state, and local levels, interested
private sector organizations and groups, and members of the public. We
plan to revise these documents in response to public comments and as
additional data become available; therefore, we encourage comment that
will make the toxicological profile series of the greatest use.

     This profile reflects our assessment of all relevant toxicological
testing and information  that has been peer reviewed. It has been
reviewed by scientists from ATSDR, EPA, the Centers for Disease Control,
and the National Toxicology Program. It has also been reviewed by a
panel of nongovernment peer reviewers and was made available for public
review. Final responsibility for the contents and views expressed in
this toxicological profile resides with ATSDR.
                                    James 0. Mason, M.D., Dr. P.M.
                                    Assistant Surgeon General
                                    Administrator, ATSDR
 iv

-------
                                CONTENTS

FOREWORD 	

LIST OF FIGURES 	     ix
LIST OF TABLES 	     xi

 1.   PUBLIC HEALTH STATEMENT 	     1
     1.1  WHAT IS DIOXIN? 	     1
     1.2  HOW MIGHT I BE EXPOSED TO 2,3,7 ,8-TCDD? 	      1
     1.3  HOW DOES 2,3,7,8-TCDD GET INTO MY BODY? 	     2
     1.4  HOW CAN 2,3,7,8-TCDD AFFECT MY HEALTH? 	     3
     1.5  IS THERE A MEDICAL TEST TO DETERMINE IF I  HAVE
          BEEN EXPOSED TO 2,3,7,8-TCDD? 	     3
     1.6  WHAT LEVELS OF EXPOSURE HAVE RESULTED IN
          HARMFUL HEALTH EFFECTS? 	     4
     1.7  WHAT RECOMMENDATIONS HAS THE FEDERAL GOVERNMENT
          MADE TO PROTECT HUMAN HEALTH? 	     7
 2.   HEALTH EFFECTS SUMMARY 	     9
     2.1  INTRODUCTION 	'.'.'.'.'.     9
     2.2  LEVELS  OF SIGNIFICANT EXPOSURE 	'. .'.    10
          2.2.1  Key Studies and Graphical  Presentations 	    10
                 2.2.1.1  Inhalation 	    11
                 2.2.1.2  Oral 	    11
                 2.2.1.3  Dermal 	    18
          2.2.2  Biological Monitoring as a Measure  of Exposure
                 and Effects 	    20
          2.2.3  Environmental Levels as Indicators  of Exposure
                 and Effects 	    22
                 2.2.3.1  Levels found in the environment 	    22
                 2.2.3.2  Human exposure potential 	    22
     2.3  ADEQUACY OF DATABASE 	    23
          2.3.1  Introduction 	    23
          2.3.2  Health Effect End Points 	    24
                 2.3.2.1  Introduction and  graphic summary 	    24
                 2.3.2.2  Descriptions of highlights of graphs  ....    27
                 2.3.2.3  Summary of relevant ongoing research  ....    28
          2.3.3  Other Information Needed for Human
                 Health Assessment 	    28
                 2.3.3.1  Pharmacokinetics  and mechanisms
                          of action 	    28
                 2.3.3.2  Monitoring human  biological samples  	    31
                 2.3.3.3  Environmental considerations 	    31
 3.   CHEMICAL AND PHYSICAL INFORMATION 	    35
     3.1  CHEMICAL IDENTITY 	    35
     3.2  PHYSICAL AND CHEMICAL PROPERTIES  	    35

-------
Contents

 4.   TOXICOLOGICAL DATA  	   3
     4.1  OVERVIEW 	' ' '  .   3.
     4.2  TOXICOKINETICS  	   43
          4.2.1 'Abs'orption 	   43
                 4.2.1.1  Inhalation 	   43
                 4.2.1.2  Oral 	   43
                 4.2.1.3  Dermal 	   44
          4.2.2  Distribution 	   44
                 4.2.2.1  Inhalation 	   44
                 4.2.2.2  Oral 	   44
                 4.2.2.3  Dermal 	   45
          4.2.3  Metabolism 	   46
          4.2.4  Excretion 	   46
                 4.2.4.1  Human 	   46
                 4.2.4.2  Animal 	   47
     4.3  TOXICITY 	   48
          4.3.1  Lethality and Decreased Longevity 	   48
                 4 3.1.1  Inhalation 	   48
                 -3.1.2  Oral 	   48
                 4.3.1.3  Dermal 	   49
          4.3.2  Systemic/Target Organ Toxicity 	   49
                 4.3.2.1  Chloracne 	   49
                 4.3.2.2  Wasting syndrome 	   51
                 4.3.2.3  Hepatic effects 	   52
                 4.3.2.4  Immunotoxicity 	   54
          4.3.3  Developmental Toxicity 	   5'
                 4.3.3.1  Inhalation 	   5
                 4.3.3.2  Oral 	   56
                 4.3.3.3  Dermal 	   57
                 4.3.3.4  General discussion 	   57
          4.3.4  Reproductivity Toxicity 	   58
                 4.3.4.1  Inhalation 	   58
                 4.3.4.2  Oral 	   58
                 4.3.4.3  Dermal 	   59
                 4.3.4.4  General discussion 	   59
          4.3.5  Genotoxicity 	   60
                 4.3.5.1  Human 	   60
                 4.3.5.2  Nonhuman	   61
                 4.3.5.3  General discussion 	   61
          4.3.6  Carcinogenic ity 	   61
                 4.3.6.1  Inhalation 	   61
                 4.3.6.2  Oral 	   64
                 4.3.6.3  Dermal 	   64
                 4.3.6.4  General discussion 	   68
     4.4  INTERACTIONS WITH OTHER CHEMICALS 	   68

 5.   MANUFACTURE, IMPORT, USE, AND DISPOSAL 	   69
     5.1  OVERVIEW 	   69
     5.2  PRODUCTION  	   69
     5.3  IMPORT 	   69
     5.4  USE  	   69
     5.5  DISPOSAL/STABILIZATION 	   6
vi

-------
                                                                Contents

 6.  ENVIRONMENTAL FATE  	    71
     6.1  OVERVIEW 	    71
     6.2  RELEASES TO THE ENVIRONMENT 	    71
          6.2.1  Production and Use of Certain Herbicides and
                 Chlorophenols and Bleaching Process in
                 Pulp and Paper Industry 	    71
          6.2.2  Photochemical Reactions 	    72
          6.2.3  Thermal Reactions 	    72
          6.2.4  Improper Disposal of Chlorinated Chemical Wastes  .    73
     6.3  ENVIRONMENTAL FATE 	    73

 7.  POTENTIAL FOR HUMAN EXPOSURE 	    75
     7.1  OVERVIEW 	    75
     7.2. LEVELS MONITORED OR ESTIMATED IN THE ENVIRONMENT 	    76
          7.2.1  Air 	    76
          7.2.2  Water 	    77
          7.2.3  Soil 	    77
          7.2.4  Other 	    78
     7.3  OCCUPATIONAL EXPOSURES 	    83
     7.4  POPULATIONS AT HIGH RISK 	    83

 8.  ANALYTICAL METHODS  	    85
     8.1  ENVIRONMENTAL MEDIA 	    86
          8.1.1  Air, Water,  Soil, and Food 	    86
     8.2  BIOMEDICAL SAMPLES 	    86
          8.2.1  Fluids/Exudates and Tissues 	    86

 9.  REGULATORY AND ADVISORY STATUS 	    93
     9.1  INTERNATIONAL  	    93
     9.2  NATIONAL 	    93
          9.2.1  Regulations 	    93
          9.2.2  Advisory Guidance 	    93
                 9.2.2.1  Air 	    93
                 9.2.2.2  Water	    93
                 9.2.2.3  Food 	    94
          9.2.3  Data Analysis 	    94
                 9.2.3.1  Reference doses (RfDs) 	    94
                 9.2.3.2  Carcinogenic potency, q * 	    94
                 9.2.3.3  Carcinogenic potency, methods used by
                          other agencies 	    95
     9.3  STATE 	    95
10.  REFERENCES 	    97

11.  GLOSSARY 	   125

APPENDIX:  PEER REVIEW 	   129
                                                                     vii

-------
                            LIST OF FIGURES
1.1  Health effects from ingesting 2,3,7,8-TCDD 	    5
1.2  Health effects from skin contact with 2,3,7,8-TCDD 	    6
2.1  Effects of 2,3,7,8-TCDD--oral exposure 	     ..    12
2.2  Effects of 2,3,7,8-TCDD--dermal exposure 	     13
2.3  Levels of significant exposure for 2,3,7,8-TCDD--oral 	   14
2.4  Levels of significant exposure for 2 , 3,7,8-TCDD--dermal  	   15
2.5  Availability of information on health effects of
     2,3,7,8-TCDD (human data) 	   25
2.6  Availability of information on health effects of
     2,3,7,8-TCDD (animal data) 	   26

-------
                             LIST OF TABLES
2.1  Research in progress on 2,3,7,8-TCDD 	   29
3.1  Chemical identity of 2,3,7.8-TCDD 	    36
3.2  Physical properties of 2,3,7,8-TCDD 	   37
4.1  Recommended TEFs for CDDs and CDFs 	   42
4.2  Genotoxicity of 2,3,7,8-TCDD in vitro 	   62
4.3  Genotoxicity of 2, 3, 7, 8-TCDD in vivo 	   63
4.4  Summary of the oral carcinogenicity bioassay of
     Kociba et al. (1978a,b) 	   65
4.5  Other oral studies supporting the conclusion that
     2,3,7,8-TCDD is an animal carcinogen 	   66
7.1  Levels of 2,3,7,8-TCDD in soil from different locations  	   79
8.1  Analytical methods for environmental samples 	   87
8.2  Analytical methods for biomedical samples 	   90
                                                                      XL

-------
                      1.  PUBLIC HEALTH STATEMENT

1.1  WHAT IS DIOXIN?

     The chlorinated dibenzo-p-dioxins are a class of compounds that are
loosely referred to as dloxins. There are 75 possible dioxins. The one
with four chlorine atoms at positions 2, 3, 7 and 8 of the dibenzo-p-
dioxin chemical structure is called 2,3,7,8-tetrachlorodibenzo-p-dioxin
(2,3,7,8-TCDD). It is a colorless solid with no known odor. 2,3,7.8-TCDD
does not occur naturally nor is it intentionally manufactured by any
industry, except as a reference standard. It can be inadvertently
produced in very small amounts as an  impurity during the manufacture of
certain herbicides and germicides and has been detected in products of
incineration of municipal and industrial wastes. At the present time,
2,3,7,8-TCDD is not used for any purpose other than scientific research.

1.2  HOW MIGHT I BE EXPOSED TO 2,3,7.8-TCDD?

     The main environmental sources of 2,3,7,8-TCDD are:

   • Use of herbicides containing 2,4,5-trichlorophenoxy acids (2,4,5-T)
   • Production and use of 2,4,5-trichlorophenol in wood preservatives
   • Production and use of hexachlorophene as a germicide
   • Pulp and paper manufacturing plants

   • Incineration of municipal and certain industrial wastes

   • Small amounts formed during the burning of wood in the presence of
     chlorine

   • Accidental transformer/capacitor fires involving chlorinated
     benzenes and biphenyls

   • Exhaust from automobiles powered with leaded gasoline

   • Improper disposal of certain chlorinated chemical wastes

     Although 2,4,5-T, 2,4,5-trichlorophenol and hexachlorophene are no
longer produced commercially (except  for certain medical purposes),
disposal sites of past production wastes are still sources of present
exposure. 2,3,7,8-TCDD has been found in at least 28 of 1,177 hazardous
waste sites on the National Priorities List (NPL).  Very low levels of
2,3,7,8-TCDD have been detected in ambient air. Detection of 2,3,7,8-
TCDD in drinking water has not been reported. 2,3,7,8-TCDD has not been
detected in most rural soils examined, but it can be present at trace
levels in urban soils. The highest concentration of 2,3,7,8-TCDD was
detected in a waste-oil-contaminated  soil In Missouri that contained a
2,3,7,8-TCDD level more than one million times higher than soils from
normal urban areas. 2,3,7,8-TCDD was detected in fish obtained from the

-------
2   Section 1
contaminated sections of Lake Ontario, Saginaw Bay, the Michigan rivers
and several watersheds including those from Maine,  Wisconsin, and
Minnesota. In human milk, minute amounts of 2,3,7,8-TCDD have been
detected in the United States and in several European countries.
Consumer sources are:
   • Skin contact with surfaces such as soil or vegetation contaminated
     by the chemical
   • Skin contact and inhalation of wood dusts from use of
     pentachlorophenol-treated woods
   • Inhalation of air near improperly maintained dump sites or
     municipal incinerators
   • Consumption of fish and cow's milk from contaminated areas
   • Consumption of breast milk containing 2,3,7,8-TCDD by babies
   • Minute exposure from the use of paper towels,  napkins, coffee
     filters, computer papers, and other contaminated paper products
Workers at risk of contacting 2,3,7,8-TCDD are:
   • Workers who have been involved in the production or use of
     trichlorophenol and salts, hexachlorophene,  and 2,4,5-T or other
     herbicides containing this chemical.  The production of 2,4,5-T and
     2,4,5-trichlorophenol, however, has been discontinued in the United
     States.
   • Workers in the pump and paper industry
   • Workers at certain municipal and industrial incinerators
   • Workers involved in the high-temperature/pressure treatment of
     woods with pentachlorophenol
   • Workers at certain hazardous waste sites
   • Workers involved in the cleanup of certain accidental
     capacitor/transformer fires and in the salvaging of transformers
   • Workers who have been involved in spraying of phenoxy herbicides
     such as Agent Orange
1.3  HOW DOES 2,3,7,8-TCDD GET INTO MY BOOT?
   • Absorption through skin from contaminated soils and other materials
   • Ingestion of 2,3,7,8-TCDD through the consumption of contaminated
     fish, cow's milk, foodstuffs, and, in the case of small children,
     soil
   • Breathing contaminated ambient air. This may contribute very small
     amounts Co total body intake; however, particulates such as fly ash
     from municipal and industrial incineration may constitute a major
     source of exposure.
   • Intake of 2,3,7,8-TCDD from the consumption of drinking water
     should be negligible

-------
                                             Public Health Statement   3

   • According to one. estimate of ambient exposure, breathing air
     constitutes 2%, drinking water less than 0.01%, and consuming foods
     98% of the total human exposure to 2,3,7,8-TCDD. No estimate of
     relative intake of 2,3,7,8-TCDD due to skin absorption is
     available.

1.4  HOW CAN 2,3.7,8-TCDD AFFECT MY HEALTH?

   • In humans, 2,3,7,8-TCDD causes chloracne, a severe skin lesion chat
     usually occurs on the head and upper body. Unlike common acne,
     chloracne is more disfiguring and often lasts for years after the
     initial exposure.
   • There is suggestive evidence that 2,3,7,8-TCDD causes liver damage
     in humans, as indicated by an increase in levels of certain enzymes
     in the blood, although these effects might also have resulted from
     the concomitant exposure to the chemicals contaminated with
     2,3,7,8-TCDD or to the solvents in which these chemicals are
     usually dissolved. Animal studies have demonstrated severe liver
     damage in some species.
   • There is suggestive evidence that 2,3,7,8-TCDD causes loss of
     appetite, weight loss, and digestive disorders in humans, although
     these effects might also have resulted from the concomitant
     exposure to the chemicals contaminated with 2,3,7,8-TCDD or to the
     solvents in which these chemicals are usually dissolved. Animal
     exposure to 2,3,7,8-TCDD results in severe loss of body weight
     prior to death.
   • Although not demonstrated in humans, in animal studies 2,3,7,8-TCDD
     produced toxicity to the immune system. This toxicity can result in
     greater susceptibility to infection.
   • Although not demonstrated in humans, in some animal species
     exposure to 2,3,7,8-TCDD resulted in adverse reproductive effects
     including spontaneous abortions. The monkey is very sensitive to
     this toxic property of 2,3,7,8-TCDD.
   • Although not demonstrated in humans, in some animal species
     exposure to 2,3,7,8-TCDD during pregnancy resulted in malformations
     in the offspring. Low levels of 2,3,7,8-TCDD have been detected in
     human milk, but the effects on infants and children are unknown.
   • The human evidence for 2,3,7,8-TCDD alone is inadequate to
     demonstrate or reflect a carcinogenic hazard, although certain
     herbicide mixtures containing 2,3,7,8-TCDD as an impurity provide
     limited evidence of causing cancer in exposed humans. Based on the
     positive evidence in animal studies, 2,3,7,8-TCDD is probably
     carcinogenic in humans.

1.5  IS THERE A MEDICAL TEST TO DETERMINE IF I HAVE
     BEEN EXPOSED TO 2,3,7.8-TCDDT
     There is no common medical test available to demonstrate
convincingly that you have been exposed to 2,3,7,8-TCDD. It is believed
that a blood test to detect certain enzymes indicating liver damage may
be helpful in determining whether exposure has occurred. These  tests do

-------
4   Section 1

not indicate with certainty that you have been exposed to 2,3,7,8-TCDD.
since other chemicals, as well as drinking alcohol, can cause similar
results. When tests for these enzymes have been performed, changes in
these enzymes were demonstrated only in some of the people suspected of
2,3,7,8-TCDD exposure.

     Other tests are available that are not commonly conducted by a
physician but appear to more adequately indicate that you have been
exposed to 2,3,7,8-TCDD. One test consists of removing a small piece of
body fat by a simple surgical procedure; the fat is then analyzed for
the presence of 2,3,7,8-TCDD. In another recently developed test, blood
serum is obtained and analyzed for the presence of 2,3,7,8-TCDD.  The
initial study appears to indicate that the method is sensitive enough to
detect extremely low levels of 2,3,7,8-TCDD. If the levels of 2,3,7,8-
TCDD are higher than the determined background range for people in the
United States, the test indicates that you have probably been exposed to
more 2,3,7,8-TCDD than the average population, or exposure has occurred
more recently than that of the comparison group. In addition, detection
of 2,3,7,8-TCDD in mother's milk would also indicate exposure; the level
of 2,3,7,8-TCDD in the milk may provide some indication of whether
exposure was to background levels or if additional exposure had
occurred. This method has not been-used widely to evaluate human
exposure.

1.6  WHAT LEVELS OF EXPOSURE HAVE RESULTED IN HARMFUL HEALTH EFFECTS7

     The graphs on the following pages show the relationship between
exposure to 2,3,7,8-TCDD and known health effects. In the first set of
graphs labeled "Health effects from ingesting 2,3,7,8-TCDD" (Fig. 1.1),
exposure is measured in milligrams of 2,3,7,8-TCDD per kilogram of body
weight (mg/kg)• In the second set of graphs (Fig. 1.2),  the same
relationship is represented for the known "Health effects from skin
contact with products containing 2,3,7,8-TCDD." Exposures are again
measured in milligrams of 2,3,7,8-TCDD per kilogram of body weight
(mg/kg). In all graphs, effects in animals are shown on the left side
and effects in humans on the right side.

     The levels marked on Fig. 1.1, which are dose estimates associated
with minimal risk for health effects other than cancer, in humans, are
based on information from animal studies; therefore, some uncertainty
still exists. For cancer, the U.S. Environmental Protection Agency (EPA)
has estimated that lifetime exposure to 1 nanogram of 2,3,7,8-TCDD per
kilogram per day would result in 1,560 or 1,560,000 additional cases of
cancer in a population of 10,000 or 10,000,000 people, respectively. It
should be noted that these risk values are plausible upper-limit
estimates. Actual risk levels are unlikely to be higher and may be
lower. (One nanogram is one-billionth of a gram.) EPA is in the process
of reviewing its risk assessment of 2,3,7,8-TCDD.

     There was not enough information to prepare a graph for exposure by
breathing.

-------
                                                    Public Health Statement    5
           SHORT-TERM EXPOSURE
       (LESS THAN OR EQUAL TO 14 DAYS)
                                    LONG-TERM EXPOSURE
                                   (GREATER THAN 14 DAYS)
     EFFECTS
        IN
     ANIMALS
  DOSE
(mg/Kg/day}
   DEATH.
  0001
   oL
   T
DEVELOPMENTAL <
EFFECTS
                  00005
                  0.0001
                    «
                  0.00008
                  0.00004
                  0.00002
                 0 000008
                 0.000006
                 0.000004
                 0.000002
                 0 000001.
EFFECTS
   IN
HUMANS
EFFECTS
   IN
ANIMALS
  DOSE
(mg/Kg/day)
                                           DEATH
                                                          0.00001
                                         0.000005
                          REPRODUCTIVE      I
                          TOXICITYAND       RJ
                          CHLORACNE	|
                                       r  0000001
                                        0.0000008
                                          LIVER DAMAGE ^
                                                         0.0000006
                                                         0.0000004
             MINIMAL RISK
             FOR EFFECTS
             OTHER THAN
            .CANCER
                                                         0 0000002
                                        00000001
                                           1
                                           T
                                        0.00000005
                                                         0 00000001
                                                            T
                                                         0 000000005
                                                         0000000001.
EFFECTS
   IN
HUMANS
                                                       MINIMAL RISK
                                                       FOR EFFECTS
                                                       OTHER THAN
                                                      .CANCER
                  Fig. 1.1.  Health effects from ingesting W.8-TCDD.

-------
   Secclon  1
      SHORT-TERM EXPOSURE
  (LESS THAN OR EQUAL TO 14 DAYS)
                                    LONG-TERM EXPOSURE
                                   (GREATER THAN 14 DAYS)
EFFECTS
   IN
ANIMALS
DEATH.
  DOSE
(mg/kg/day)
               1.0
               0.8
               0.6
               0.4
               0.2
EFFECTS
   IN
HUMANS
                         QUANTITATIVE
                         DATA WERE
                         NOT AVAILABLE
EFFECTS EFFECTS
IN DOSE IN
ANIMALS (mg/kg/day) HUMANS



0
0.
0.
0
CHLORACNE 	
0.
QUANTITATIVE
DATA WERE
NOT AVAILABLE
01
308
)06
304

)02
              Fig. 1.2. Health effects from skin contact with 24,7,8-TCDD.

-------
                                             Public Health Scacemenc   /

1.7  WHAT RECOMMENDATIONS HAS THE FEDERAL GOVERNMENT MADE TO
     PROTECT HUMAN HEALTH?

     Both the EPA and the International Agency for Research on Cancer
(IARC) have concluded that 2,3,7,8-TCDD causes cancer in animals and
probably causes cancer in humans.

     The EPA calculated health advisories (HAs) for 2,3,7,8-TCDD in
drinking water, that is, estimates of levels below which adverse health
effects are not expected to occur. The 1-day HA Is 1 part of 2,3,7,8-
TCDD per trillion parts of waste (1 ppt) for a child; the 10-day HA is
0.1 ppt for a child. The longer-term HA is 0.01 ppt for a child and
0.035 ppt for an adult; the lifetime HA is also 0.035 ppt for adults.
These are very small amounts. The EPA also calculated the amount of
2,3,7,8-TCDD in ambient water (lakes and rivers) that would be
associated with increases in one additional incidence of cancer over
background cancer incidence in a population of 1,000,000 to be 0.013
parts per quadrillion, an extremely small amount. This calculated
measurement takes into account that 2,3,7,8-TCDD concentrates in fish;
hence, exposure may occur through both the drinking of water and the
eating of fish. With regard to advisories based on EPA's cancer risk
estimate for 2,3,7,8-TCDD, it should be noted that the Agency is in the
process of revising this risk estimate.

-------
                       2.  HEALTH EFFECTS SUMMARY

2.1  INTRODUCTION

     This section summarizes and graphs data on the health effects
concerning exposure to 2,3,7,8-TCDD. The purpose of this section is to
present levels of significant exposure for 2,3,7,8-TCDD based on key
toxicological studies, epidemiological investigations, and environmental
exposure data. The information presented in this section is critically
evaluated and discussed in Sect. 4, Toxicological Data, and Sect. 7,
Potential for Human Exposure.

     This Health Effects Summary section comprises two major parts.
Levels of Significant Exposure (Sect. 2.2) presents brief narratives and
graphics for key studies in a manner that provides public health
officials, physicians, and other interested individuals and groups with
(1) an overall perspective of the toxicology of 2,3,7,8-TCDD and (2) a
summarized depiction of significant exposure levels associated with
various adverse health effects. This section also includes information
on the levels of 2,3,7,8-TCDD that have been monitored in human fluids
and tissues and information about levels of 2,3,7,8-TCDD found in
environmental media and their association with human exposures.

     The significance of the exposure levels shown on the graphs may
differ depending on the user's perspective. For example, physicians
concerned with the interpretation of overt clinical findings in exposed
persons or with the identification of persons with the potential to
develop such disease may be interested in levels of exposure associated
with frank effects (Frank Effect Level, FEL). Public health officials
and project managers concerned with response actions at Superfund sites
may want information on levels of exposure associated with more subtle
effects in humans or animals (Lowest-Observed-Adverse-Effect Level,
LOAEL) or exposure levels below which no adverse effects (No-Observed-
Adverse -Effect Level, NOAEL) have been observed. Estimates of levels
posing minimal risk to humans (Minimal Risk Levels, MRL) are of interest
to health professionals and citizens alike.

     Adequacy of Database (Sect. 2.3) highlights the availability of key
studies on exposure to 2,3,7,8-TCDD in the scientific literature and
displays these data in three-dimensional graphs consistent with the
format in Sect. 2.2. The purpose of this section is to suggest where
there might be insufficient information to establish levels of
significant human exposure. These areas will be considered by the Agency
for Toxic Substances and Disease Registry (ATSDR), EPA, and the National
Toxicology Program (NTP) of the U.S. Public Health Service in order to
develop a research agenda for 2,3,7,8-TCDD.

-------
10   Section 2

2.2  LEVELS OF SIGNIFICANT EXPOSURE

     To help public health professionals address the needs of persons
living or working near hazardous waste sites,  the toxicology data
summarized in this section are organized first by route of
exposure-- inhalation, ingestion, and dermal--and then by toxicological
end points that are categorized into six general areas — lethality,
systemic/target organ toxicity, developmental  toxicity, reproductive
toxicity, genetic toxicity, and carcinogenicity. The data are discussed
in terms of three exposure periods--acute,  intermediate, and chronic.
     Two kinds of graphs are used to depict the data. The first type is
a "thermometer" graph. It provides a graphical summary of the human and
animal toxicological end points (and levels of exposure) for each
exposure route for which data are available.  The ordering of effects
does not reflect the exposure duration or species of animal tested. The
second kind of graph shows Levels of Significant Exposure (LSE) for each
route and exposure duration. The points on the graph showing NOAELs and
LOAELs reflect the actual doses (levels of exposure) used in the key
studies. No adjustments for exposure duration or intermittent exposure
protocol were made.
     Adjustments reflecting the uncertainty of extrapolating animal data
to humans, intraspecies variations, and differences between experimental
versus actual human exposure conditions were considered when estimates
of levels posing minimal risk to human health were made for noncancer
end points. These minimal risk  levels were derived for the most
sensitive noncancer end point for each exposure duration by applying
uncertainty factors. These levels are shown on  the graphs as a broken
line starting from the actual dose  (level of exposure) and ending with a
concave-curved line at its terminus. Although methods have been
established to derive these minimal risk levels  (Barnes et.al. 1987),
shortcomings exist in the techniques that reduce the confidence in  the
projected estimates. Also shown on  the graphs under  the cancer end  point
are risks (10*^ to 10'7) estimated by EPA. In addition, the actual  dose
(level of exposure) associated  with tumor incidence  is plotted.

2.2.1  Key Studies and Graphical Presentations
     It  is difficult  to assess  the  risk to humans from exposure to
2,3,7,8-TCDD. There are many species differences in  toxicity;  the monkey
and guinea pig are apparently  the most sensitive and the hamster is the
least  sensitive.  It  is not known how sensitive  humans  are  to 2,3,7,8-
TCDD.  In addition, 2,3,7,8-TCDD strongly adsorbs to  materials  such  as
soil,  which may significantly  affect the bioavailability and toxicity  of
this compound. Animal studies  have  generally been conducted with
2,3,7,8-TCDD  administered  in oily vehicles from which the  compound is
readily  bioavailable. There are insufficient data available  to consider
the effect of bioavailability  on  the toxicity  studies used to  define the
risk from exposure to 2,3,7,8-TCDD. Both bioavailability  and species
differences  in sensitivity should be considered when evaluating  the
remainder of  the  data presented.

-------
                                              Health Effaces Summary   11

      Oral and dermal'NOAELs and  LOAELs are presented on "thermometer"
 graphs in Figs. 2.1 and 2.2, respectively. Although some qualitative
 data are available for humans, quantitative data were insufficient to
 graphically present inhalation effects.

      Levels of significant exposure are depicted in Figs.  2.3 and 2 4
 for the oral and dermal routes. There were insufficient human and animal
 inhalation data for graphical representation. The intermediate minimal
 risk level for oral exposure was calculated by EPA (1985a) from a
 three-generation reproductive toxicity study in the rat.  with effects
 observed in the fetuses. Since the effects were attributed to in utero
 fetal exposure,  which chronologically represents an exposure of
 relatively short duration, as well as to the chronic exposure of the
 dams,  the same minimal risk level for chronic exposure was calculated
 from these data.

 2.2.1.1  Inhalation

      No studies  are available on the inhalation toxicity of
 2,3,7,8-TCDD.  Exposure through inhalation,  however,  may also have
 occurred in  the  population exposed to chemicals contaminated with TCDD
 in accidental  releases or  in the  workplace  (especially herbicide
 spraying).

 2.2.1.2  Oral

     Lethality and  decreased longevity.   There  have  been no reports of
 death  in  humans as  a  result of oral  exposure  to 2,3,7,8-TCDD.

     2,3,7,8-TCDD is highly toxic to all  laboratory  animals tested, even
 though  there is a large  difference in species sensitivity.  LDso  values
 range  from 0.6 MgAg  in  male  guinea  pigs  (Schwetz  et al. 1973) to 5,500
 AigAg  in hamsters (Henck et al. 1981). These values  are plotted  in
 Figs. 2.1 and 2.3 for  the  lethality  of acute oral  exposure.  Death
 usually occurs 13 to 18  days  after a single exposure.  Extended exposure
 in  a 90-day feeding study  in  guinea  pigs  resulted  in an estimated 50%
 mortality after consumption of  a  total of 0.8 /*g of  2,3, 7,8-TCDDAg
 (0.008  /JgAg/day) (DeCaprio et  al. 1986), whereas  deaths did not occur
 at  0.0006 MgAg/day (NOAEL) .  Five  of eight female  monkeys  that ingested
 approximately 0.01 /*g of 2,3,7,8-TCDDAg/day for 9 months  died (Allen  et
 al. 1977). These FELs  in guinea pigs and monkeys and the NOAEL in guinea
 pigs are shown in Figs. 2.1 and 2.3.

     Target organ/systemic  toxicity.   Four major toxic effects
 characteristic of 2,3,7,8-TCDD are chloracne, the wasting  syndrome,
hepatotoxicity, and immunotoxicity.  The latter  three effects have  been
clearly demonstrated only  in laboratory animals. Chloracne,
 immunotoxicity, hyperpigmentation, hyperkeratosis, hirsutism of  the
skin, possible hepatotoxicity, hypertriglyceridemia  and
hypercholesterolemia, aching muscles,  loss of appetite, weight loss,
digestive disorders, headaches, neuropathy, insomnia, sensory changes,
and loss of libido have been observed  in humans  exposed to chemicals
contaminated with 2,3,7,8-TCDD. These  reported effects may have  been the
result of 2,3,7,8-TCDD exposure, or  exposure to  the chemicals of which
2,3,7,8-TCDD is a contaminant or to  the solvents in which  these

-------
12    Section 2
     ANIMALS
     (n»*g/<»y)
                                                                              HUMANS
   10000
    1.000
     100
     01
    001
   0001
  00001 «—
           •  HAMSTER L0» SINGLE DOSE
           •  MOUSE. MONKEY CHLORACNE SINGLE OOSE
- •  MOUSE. DEVELOPMENTAL TOXRrTV 10 DAYS. CONTINUOUS
  •  GUINEA PIG. LO» SINGLE DOSE


  O  MOUSE. DEVELOPMENTAL TOXKfTY 10 DAYS. CONTINUOUS

  •  RAT. DEVELOPMENTAL TOX1CI7Y <0 OAYS. CONTINUOUS
- •  GUINEA PK). UVERTOXICITY. SINGLE OOSE

  •  GUINEA PtO. IMMUNOTOXICrrV I WEEKS. INTERMITTENT
  O  RAT. DEVELOPMENTAL TCUOCfTV. 10 DAYS, CONTINUOUS
        -  •  MONKEY. DEATH. 9 MONTHS. CONTIM
                                                  KEY CHLORACNE. 7 MONTHS. CONTINUOUS
                                                                     "*««TWUOUS
               --------- ..... ------ . - ....——.
              GUINEA PKX DEATH. 90 DAYS. CONTINUOUS
              GUINEA PKLIMMUNOTOXICITV. 8 WEEKS. INTERMTITENT
              GUINEA PW. WASTING SYNDROME UVERTOXICITY. 90 DAYS. CONTINUOUS
           •  MONKEY. REPRODUCTIVE TOJOCITY 7 MONTHS CONTINUOUS
              RAT. REPRODUCTIVE TOXICITY 3 GENERATIONS. RAT. UVER TOXICITY 2 YEARS. CONTINUOUS
              GUINEA PIG. WASTING SYNDROME. UVER TOXICITY 90 OAYS  CONTINUOUS
              GUINEA PKJ. DEATH. 90 OAYS. CONTINUOUS
                • LOAEL
                O NOAEL
                                                                                  QUANTITATIVE DATA
                                                                                  WERE NOT
                                                                                  AVAILABLE
                          Fig. 2.1.  Effects of 23,7,8-TCDD—oral exposure.

-------
 ANIMALS
(Wjfcg/day)

 1000 r-
  100
  10
   I i-
        •  RABBIT LD,o. SINGLE OO6E
        •  MCXJSE. DERMAL LESIONS 4 WEEKS INTERMITTENT
                                      LOAEL
                                                          Health Effects Summary     13
                                                                   HUMANS
QUANTITATIVE DATA
WERE NOT AVAILABLE
                    Fig. 2.2.  Effects of 2,3,7,8-TCDD—dermal exposure.

-------
14    Section 2
         10000


          1000


           100


            10


            1


           01


          001


         0001


        00001


       000001


      0 000001


     0 0000001


     0 00000001


    0000000001


   0 0000000001


  o 00000000001


 0 000000000001


0 0000000000001 I-
                             ACUTE                       INTERMEDIATE              CHRONIC
                            (SI4 DAYS)                     (15-364 DAYS)             (2365 DAYS)


                             DEVELOP-  TARGET  DECREASED  REPRO-   TARGET   TARGET
                    LETHALITY  MENTAL   ORGAN   LONGEVITY   DUCTION  ORGAN   ORGAN    CANCER
• s
                     k. m (SKIN)
• g
r
r
                   • g (LIVER)
                           • k
                                                • g (BODY WEIGHT
                                                I  LIVER)
                                                O        • r (LIVER)
                                                              10-*-,


                                                              10-5-





                                                              io-7-l
                                                           ESTIMATED
                                                           UPPER-
                                                           BOUND
                                                           HUMAN
                                                           CANCER
                                                           RISK
                                                           LEVELS
                 MINIMAL RISK LEVEL
                 FOR EFFECTS OTHER
                 THAN CANCER
              g  GUINEA PIG
              k  MONKEY
             m  MOUSE
              r  RAT
              S  HAMSTER
                    ILOAEL AND NOAEL
                    IN SAME SPECIES

                  • LOAEL FOR ANIMALS
                  O NOAEL FOR ANIMALS
MINIMAL RISK LEVEL FOR
CHRONIC EXTRAPOLATED
FROM INTERMEDIATE
EXPOSURE
                  Fig. L3. Leveb of significant exposure for 23,7,8-TCDD—oraL

-------
                                            Healch Effects Summary    15
              ACUTE
            (< 14 DAYS)

            LETHALITY
 INTERMEDIATE
 (15-364 DAYS)

TARGET ORGAN
                                                       CHRONIC
                                                     (2 365 DAYS)
  (ng/kg/day)

10.000 r
1000
 100
  10
                                                    QUANTITATIVE
                                                    DATA WERE
                                                    NOT AVAILABLE
                                 m (SKIN)
   1 L-
           • LOAEL     m MOUSE
                        h RABBIT
           Fig. 2.4. LeTda of significant exposure for

-------
 16    Section  2

 compounds  are normally dissolved.  Because some herbicides  and  some
 industrial chemicals  contain 2,3,7,8-TCDD as a contaminant,  the primary
 route of exposure  is  most  likely  to be dermal, although some oral and
 inhalation exposure probably also  occurs.

      Since chloroacne,  the only lesion definitively identified in humans
 as  resulting  from  2,3,7,8-TCDD exposure, can only be detected  in a few
 species, the  investigation of this effect has been limited. In hairless
 mice,  chloracne was produced after a single dose of 2,3,7,8-TCDD at
 70  MgAg (Greig 1984) ,  whereas a single dose of 70 MgAg (McConnell et
 al.  1978),  or a dose  of -0.01 MgAg/day in a 9-month feeding study in
 monkeys  (Allen et  al.  1977),  produced similar lesions on the face. These
 data are indicated in Figs.  2.1 and 2.3.

      The wasting syndrome  is  characterized by extreme loss of  body
 weight.  In acute studies,  this syndrome is associated with lethal doses.
 A dose-response relationship  for the wasting syndrome has been defined
 in  a 90-day study  in  guinea pigs (DeCaprio et al. 1986). Female Hartley
 guinea pigs were maintained on diets providing average 2,3,7,8-TCDD
 doses of 0, 0.12,  0.68, 4.86, and 31 ng/kg/day. The high dose
 represented a FEL; a  40% decrease in body weight and death occurred. The
 4.86-ng/kg (0.005  MgAg) dose represented a LOAEL, with a 13%  decrease
 in  body weight and no mortality. The 0.68 ng/kg (0.0007 MgAg) is a
 NOAEL. The  LOAEL and  NOAEL for intermediate exposure are indicated in
 Figs. 2.1  and 2.3.

      2,3,7,8-TCDD  is hepatotoxic in all species tested; however,  the
 severity of the lesions depends on the species studied. Although liver
 damage is  not as severe in the guinea pig, the most sensitive  species
 tested with regard to lethality, liver changes such as focal necrosis
 and hypertrophy have been  observed at very low doses by Turner and
 Collins  (1983). In this study, a small group of male and female guinea
 pigs was given a single dose  of 2,3,7,8-TCDD at 0.1,  0.5,  2.5. 12.5, or
 20  MgAg.  Effects  on  the liver occurred in all groups. The low dose was
 considered  a LOAEL (see Figs. 2.1 and 2.3),  but a NOAEL was not
 available.   A minimal  risk  for effects of acute oral exposure (see Fig.
 2.3) was calculated from this LOAEL, since it was the most sensitive end
point in acute studies. Similar liver damage was observed in guinea pigs
 in  the 90-day feeding study by DeCaprio et al.  (1986). The NOAEL was
0.68 ngAg  and the LOAEL was  4.86 ngAg,  the same as the NOAEL and LOAEL
 for the wasting syndrome (see Figs. 2.1 and 2.3). There are no chronic
 studies in  guinea  pigs; however, in chronic studies in rats, Kociba et
al.  (1978a,b) and  NTP (1982a) reported "toxic hepatitis" and
degenerative changes at the lowest dietary exposure that provided a dose
of  0.001 Mg of 2,3,7,8-TCDDAg/day in these 2-year studies. Again, only
a LOAEL, which is  plotted  in  Figs. 2.1 and 2.3, is available for this
end point after chronic exposure.

     The guinea pig also appears to be the most sensitive species to the
 immunotoxic effects of 2,3,7.8-TCDD. Vos et al. (1973) observed a
decrease in thymus weight,  total lymphocyte number, and total  leukocyte
number in groups of 10 guinea pigs given 2,3,7,8-TCDD weekly for 8 weeks
at doses of 0, 0.008, 0.04, or 0.2 MgAg-  The high dose was a  FEL and
also produced other toxic  effects including loss of body weight.  A LOAEL
of 0.04 MgAg and  a NOAEL  of  0.008 MgAg were defined. At the  LOAEL,

-------
                                             Health Effects Summary   17

body weight was comparable to controls. The LOAEL and NOAEL for
immunotoxicity are plotted on Fig. 2.1, but not on Fig. 2.3, because
liver toxicity and wasting syndrome are more sensitive end points of
intermediate oral exposure in guinea pigs. Immunotoxicity is not limited
to guinea pigs; Thigpen et al. (1975) reported that mice given 4 weekly
exposures to 2,3,7,8-TCDD at doses as low as 1 MgAg were more sensitive
to Salmonella-induced death. This dose caused no gross signs of toxicity
in animals not exposed to Salmonella.
     Developmental toxicity.  There have been no well-substantiated
reports of developmental toxicity in humans as a result of oral exposure
to 2,3,7,8-TCDD.
     2,3,7,8-TCDD produces anomalies in the fetus, including cleft
palate and hydronephrotic kidneys in mice and internal organ hemorrhage
in the rat. FELs in rats were reported to be 0.125 pgAg/day after
administration of the compound on days 6 through 15 of gestation
(Sparschu et al. 1971a,b), whereas the next lower dose tested, 0.03
MgAg, was a NOAEL (see Figs. 2.1 and 2.3). Doses of 1 MgAg caused
fetal death. Although developmental effects are also observed in mice,
this species appears less sensitive, with FELs of -1 ^gAg/day (when
administered during organogenesis) and a NOAEL of -0.3 MgAg (Neubert
and Dillman 1972) (see Figs.  2.1 and 2.3).
     Reproductive toxicity.   There have been no well-substantiated
reports of  reproductive toxicity in humans as a result of  oral exposure
alone to 2,3,7,8-TCDD.
     In studies with monkeys  maintained on a diet  for  7 months, which
provided 2,3,7,8-TCDD at levels of 0.0015 and 0.01 ^gAg/day,  there were
spontaneous abortions  in two-thirds of  the monkeys at  both  dose  levels
(Allen et al.  1979, Schantz  et  al. 1979). The 0.0015-Aig/kg/day level  is
indicated on Figs. 2.1 and  2.3. This study, which  reported  severe  frank
effects, indicates that monkeys may be  the most  sensitive species  with
regard to the  reproductive  toxicity of  2,3,7,8-TCDD. This study,
however, only  provided  FELs,  and  additional data were  not available for
determining a  NOAEL or  the  actual  relationship between species
sensitivity.
     Murray et al.  (1979)  conducted  a  three-generation reproductive
toxicity study in rats.  2,3.7,8-TCDD was  administered  in the diet  at
levels that provided  doses  of 0.001,  0.01,  and 0.1 /ig/kg/day.  The  high
dose  resulted  In decreased fetal  survival.  Murray et al.  (1979)
concluded  that the  0.01-dose represented a LOAEL (with effects observed
on litter  size and fetal and neonatal  survival)  and that the 0.001-dose
was considered to be  a NOAEL. Nisbet and Paxton (1982) reevaluated the
above  data, using different statistical methods, and concluded that the
lowest dose  tested produced dilated renal pelvises,  decreased fetal
weight,  and changes in the gestational index,  which indicated that
0.001  jigAg was « LOAEL.  As discussed in Sect.  4.3.4.2. EPA (1988b)
criticized the approach used by Nisbet and Paxton (1982) and concluded
 that 0.01  pgAg/day was che lowest effect level in the study by Murray
et al.  (1979)  that could be supported by the data, although further
 analysis of this study and the studies in monkeys (Allen et al.  1979;
 Schantz  et al. 1979)  may provide support for a LOAEL of 0.001 j*g/kg/aay.
As a dose  level of 0.0015 MgAg/day was associated with abortions in

-------
18   Section 2

monkeys (Allen et al." 1979), It is prudent to consider 0.001 MgAg/<*ay
as the LOAEL (see Figs. 2.1 and 2.3) from which to derive a minimal ris.
level for subchronic and chronic oral exposure (see Fig 2.3).
                  •   *
     Genotoxicity.  There have been no reports of genotoxicity in humans
as a result of oral exposure to 2,3,7,8-TCDD.

     2,3,7,8-TCDD has produced mostly negative responses in tests for
genotoxicity; however, there are a few positive responses, which may
suggest that 2,3,7,8-TCDD is genotoxic (see Sect. 4.3.5 on
genotoxicity). Some of the inconsistencies observed may be related to
experimental difficulties in testing 2,3,7,8-TCDD, such as the very low
solubility of this compound and the high toxicity in vivo (which limits
the quantity that can be tested), rather than to inherent biological
inactivity.
     Carcinogenicity.  There have been no reports of increased cancer
incidence in humans as a result of oral exposure to 2,3,7,8-TCDD.
     2,3,7,8-TCDD has been demonstrated to be an animal carcinogen in
both rats and mice in an NTP (1982a) bioassay, and in rats in a 2-year
bioassay by Kociba et al. (1978a,b). EPA (198Sa) used female rat data
from the Kociba et al. (1978a.b) study to derive a q.*. In this
derivation, the total incidences for tumors of the liver, lung, hard
palate, or nasal  turbinates, as reported by Kociba et al. (1978a,b),
were combined. These  data, along with a similar set of data  derived for
EPA by Squire on  the  reevaluation of the histologic section  from the
Kociba et al. (1978a,b) study, were used by EPA  (1985a) to derive a q^
The tumor incidences  reported by Kociba et al. (1978a,b)  for doses of
0, 0.001, 0.01, and 0.1 /*gAg/d«y were 9/85,  3/48, 18/48, and 34/40,
respectively, whereas the respective values reported by Squire were
16/85, 8/48, 27/48, and 34/40. The  q * thus calculated was 1.56 x 105
(jig/kg/day)"1. The dose of 0.01 pgAg/day, which  is associated with
increased tumor incidence in the Squire reevaluation, is  indicated on
Fig. 2.3. For cancer, EPA has estimated that  for  a population of 10,000
people exposed to 0.6 pgAg/day. the cancer  risk  is not likely to exceed
1/10,000, and similarly, that for a lesser exposure of 0.0006 pgAg/day
to 10,000,000 people,  the expected  cancer risk would not  exceed
1/10,000,000  (Fig.  2.3). EPA is  in  the process of reviewing  its  risk
assessment of 2,3,7,8-TCDD  (EPA  1988a).

2.2.1.3  Dermal
     Lethality and  decreased longevity.  There have been  no  reports  of
death  in humans as  a  result  of dermal  exposure to 2,3,7,8-TCDD.
     Schwetz  et al.  (1973)  reported a  dermal U>50 value  of 275 /igAg in
rabbits. As  in oral studies, there  was a  protracted length of time
between  application and death. No  other data were available. The U>50-is
plotted  on Figs.  2.2  and 2.4.
     Target  organ/systemic  tozicity.   Chloracne   ;  the only
substantiated effect in humans  produced by certa-.i compounds
contaminated with 2,3,7,8-TCDD.  As reviewed by Taylor (1979) and Suskind
 (1985),  these persistent,  deforming face  and uoper body lesions have
been recognized for many years  as  resulting from exposure to certain
halogenated aromatic compounds,  and it is believed that 2,3,7,8-TCDD is

-------
                                             Health Effects Summary   19

the most effective compound in producing this lesion.  There is no
information on determined levels of exposure to 2,3,7,8-TCDD needed to
produce chloracne in humans. By extrapolating dose-effect data obtained
from the ingestion of contaminated rice oil containing polychlorinated
biphenyls and polychlorinated dibenzofurans to dose-effect data for
2,3,7,8-TCDD, and taking into consideration the minimum toxic dose for
production of chloracne in nonhuman primates, Stevens (1981) estimated a
minimum toxic dose for 2,3,7,8-TCDD of 0.1 /igAg for humans.
     In addition, there are data that suggest that 2,3,7,8-TCDD is
hepatotoxic in humans. In populations exposed to herbicides and other
industrial chemicals contaminated with 2,3,7,8-TCDD, there have been
reports of increased serum levels of liver enzymes and the development
of porphyria cutanea tarda (EPA 198Sa).  In all studies, however,
exposure may have been to chemicals that also could cause liver damage,
and as pointed out by Jones and Che1sky (1986), the diagnosis of
porphyria cutanea tarda in some of the studies may be questionable. It
is thus difficult to assert that the presumed exposure to 2,3,7,8-TCDD
resulted in liver injury, and even if 2,3,7,8-TCDD induced liver damage,
there are no human data available that could provide dose-response
information. Similarly, data suggest that 2,3,7,8-TCDD might affect the
immune system in humans (Hoffman et al.  1986), but the same limitation
discussed with regard to hepatotoxicity applies to immunetoxicity.
     The only dermal animal data that provide quantitative information
on chloracne are provided by the study of Puhvel et al. (1982), in which
hairless mice given 0.1 pg of 2,3,7,8-TCDD per application three times
per week for 4 weeks developed dermal lesions that resembled some
features of chloracne in humans. Assuming that a mouse weighs 0.03 kg,
the dose is 1.4 /jgAg- This study, however, only used one dose which was
a PEL; hence, it does not provide the necessary information for defining
a dose-response relationship. The dose is plotted on Figs. 2.2 and 2.4
for intermediate target organ toxicity of dermal exposure.

     Developmental toxicity.  Studies of human populations exposed to
herbicides and other industrial chemicals contaminated with 2,3,7,8-TCDD
have suggested that 2,3,7,8-TCDD produces a variety of developmental
effects (Hanify et al. 1981, McQueen et al.  1977, Nelson et al. 1979,
Smith et al. 1982). After reviewing these studies, EPA (1985a,  1988b)
indicated that the data were not inconsistent with 2,3,7,8-TCDD
adversely affecting development, but as a result of the limitations of
the data, these studies could not prove an association with 2,3,7,8-TCDD
exposure and the observed effect. The major  limitations in  these human
studies were the concomitant exposure to other potentially  toxic
chemicals, the lack of any  specific quantitative data  on  the  extent of
exposure of  individuals within  the study group, and  the lack  of
statistical power of the studies.
     No animal studies were available on the developmental  toxicity of
2,3,7,8-TCDD following dermal exposure.
     Reproductive toxicity.  EPA  (1985a, 1988b) has  reviewed  human
reproductive toxicity studies of groups exposed to herbicides and other
industrial chemicals contaminated with 2,3,7,8-TCDD (Aldred 1978;
Bisanti et al. 1980; Bonaccorsi et al. 1978;  Department of  Health,  New
Zealand 1980; EPA 1979a; Field  and Kerr 1979;  McQueen  et  al.  1977;

-------
20   Seccion 2

Nelson et al. 1979; Reggiani 1980; Smith et al.  1982;  Thomas 1980).
These studies did not provide a scientifically valid indication that
2,3,7,8-TCDD adversely affects either male or female reproductive
performance, or that exposure to 2,3,7,8-TCDD is without effect.  The
limitations of the studies are similar to those discussed in the  section
above.

     No animal studies were available on the reproductive toxicity of
2,3,7,8-TCDD following dermal exposure.

     Carcinogenicity.  EPA (1985b, 1988b) and Hiremath et al.  (1986)
reviewed several epidemiology studies of humans exposed to herbicides
contaminated with 2,3,7,8-TCDD. A series of studies (Eriksson et  al.
1979, 1981; Hardell and Eriksson 1988; Hardell and Sandstrom 1979;
Hardell et al. 1980, 1981; Lynge 1985; Merlo and Puntoni 1986;  Puntoni
et al.  1986) reported an association between exposure  and soft tissue
sarcomas (of various sites) and lymphomas. Although many of these
studies had confounding factors, the problems with these studies  were
not sufficient to explain the highly significant risks of soft tissue
sarcoma in the exposed workers (EPA 1988b). EPA (1988b) also reviewed a
number of studies (Balarajan and Acheson 1984; Cantor  1982; Cook  et al.
1986; Fett et al. 1984; Kang et al. 1987; Kogan and Clapp 1985; Milham
1982; Smith et al. 1984; Woods et al. 1987; Zack and Suskind 1980) that
were considered to be consistent with or tended to support the findings
of soft tissue sarcoma in groups thought to be exposed to chemicals
contaminated with 2,3,7,8-TCDD. A number of studies found no association
between risk of soft tissue sarcoma and exposure to herbicides
contaminated with 2,3-,7,8-TCDD (Axelson et al. 1980; Fingerhut et al.
1984; Greenwald et al. 1984; Lathrop et al. 1984; Ott  et al. 1980;
Rlihimaki et al. 1982; Thiess et al. 1982; Wiklund and Holm 1986;
Uiklund et al. 1987; Wolfe et al. 1984,  1985). EPA (1988b) concluded
that the epidemiological data appear to provide limited evidence  that
exposure to phenoxyacetic acid herbicides and/or chlorophenols is
causally related to the risks of soft tissue sarcoma,  but none of the
data are sufficient to implicate 2,3,7,8-TCDD alone.

     There are some data from experimental animal studies regarding the
dermal carcinogenicity of 2,3,7,8-TCDD.  In Swiss mice, females, but not
males,  developed skin tumors following dermal application of
2,3,7,8-TCDD at 0.01 pg per application three times per week
(NTP 1982b). There has also been mixed evidence that 2,3,7,8-TCDD is a
tumor promoter. Poland et al. (1982) observed tumor-promoting activity
in hairless mice, but not in mice heterozygous for the hairless trait.
Similarly, Berry et al. (1978), Slaga and Nesnows (1985), and NTP
(1982b) have not been able to demonstrate promoting activity in CD-I,
Sencar, or Swiss-Webster mice.

2.2.2  Biological Monitoring as a Measure of Exposure and Effects

     Biological monitoring of body fluids provides qualitative
indications of exposure. With commonly available analytical techniques,
2,3,7,8-TCDD is not detected in body fluids, such as blood or urine,
although a recent method with parts-per-quadrillion sensitivity has
detected 2,3,7,8-TCDD in human serum (Patterson et al. 1987b).
Biomonitoring of adipose tissue provides qualitative and quantitative

-------
                                             Health Effects Summary   21

information on exposure, but the techniques are not commonly available.
Adipose tissue has been shown to be one of the primary storage sites for
2,3,7,8-TCDD, and tissue samples have been analyzed, although mixed
results have been obtained. As reported by Nygren et al. (1986),  Young
et al. (1983) failed to detect elevated levels of 2,3,7,8-TCDD in the
adipose tissues of Vietnam veterans exposed to Agent Orange, whereas
Gross et al.  (1984) detected increased levels in veterans who had been
exposed to high levels of Agent Orange. As reported by Nygren et al.
(1986), higher levels of 2,3,7,8-TCDD in adipose tissue have also been
reported in individuals exposed during a transformer fire accident in
Binghamton, New York, or the accident in Seveso, Italy. In another
study, 39 exposed individuals in Missouri had median adipose 2,3,7,8-
TCDD levels of 17 ppt, with a range of 2.8 to 750 ppt, whereas the
unexposed control group had a median level of 6.4 and a range of 1.4 to
20 ppt (Patterson et al. 1986). Although six of the subjects in the
exposed group had levels >5 times higher than the highest control, there
was also extensive overlap between the groups.
     EPA (1988c) estimated an upper limit value for the average
2,3,7,8-TCDD concentration in adipose tissue to be 6.72 ppt in the U.S.
population. From this adipose tissue burden and pharmacokinetic
considerations, it was estimated that the upper-bound 2,3,7,8-TCDD daily
intake ranges from 0.04 to 0.51 pgAg- However, the level of 2,3,7,8-
TCDD in the adipose tissue of certain individuals with no known history
of exposure to 2,3,7,8-TCDD generally are in the range of 5 to 18 ppt.
This would suggest a ubiquitous exposure to 2,3,7,8-TCDD, which makes it
difficult to assess the contribution to body burden from any particular
small additional exposure. A similar lack of correlation between
estimated exposure to 2,3,7,8-TCDD and sera levels of 2,3,7,8-TCDD was
reported in a preliminary  study in the MMWR (1987), in which Vietnam
veterans with military histories indicating exposure  to herbicides
containing 2,3,7,8-TCDD were compared to non-Vietnam veterans with
presumably no unusual exposure to 2,3,7,8-TCDD. In  these preliminary
results, at least, there was no difference  in  the range of  2,3,7,8-TCDD
levels (1 to 9 ppt of both groups based on  lipid weight) or the median
2,3,7,8-TCDD level (3.9 ppt for the presumably exposed  group  and  3.8  ppt
for'the nonexposed group). This lack of correlation may in  part be
attributed to the difficulty of identifying persons exposed to 2,3,7,8-
TCDD,  since  inclusion of nonexposed  individuals in  the  exposed
population will tend to make the two groups appear  similar.
     There are also  no  clear tests  for  the  effects  of exposure  to
2,3,7,8-TCDD. Chloracne  is the only  effect  that is  clearly  associated
with  exposure  to  chemicals contaminated with  2,3,7,8-TCDD;  however,
chloracne  is also caused by other halogenated aromatic compounds.  The
development  of chloracne  in an individual who may have been exposed to
2,3,7,8-TCDD would provide supportive  evidence that exposure  to  this
chemical had occurred.  The development of chloracne,  however, does not
indicate the extent  of exposure. Other signs  of toxicity observed in
animal studies  (i.e.,  liver damage,  effects on lipid metabolism,  and
types of circulating lipids) have  not been demonstrated in humans and
are not useful  in determining that exposure to 2,3,7,8-TCDD has
occurred.

-------
22   Section 2

2.2.3  Environmental Levels as Indicators  of Exposure and Effects

2.2.3.1  Levels found in the environment

     2,3,7,8-TCDD has been monitored in the  areas of extensive herbicide
use and areas contaminated with 2,3,7,8-TCDD through industrial
accidents; however, epidemiologic studies  of inhabitants of these areas
have lacked adequate exposure data that would permit the demonstration
of a clear association between exposure and  effects.  The biological
half-life of 2,3,7,8-TCDD in humans,  calculated to be 5 years by Poiger
and Schlatter (1986) and 7 years by the CDC  (1987),  would indicate that
repeated exposure to low levels of 2,3,7,8-TCDD could substantially
elevate the body burden of this compound to  a level equivalent to the
level obtained after a single exposure to  a  high level. Given the long
half-life of 2,3,7,8-TCDD, the total exposure history of an individual
has to be taken into account; hence,  the environmental levels during a
single particular exposure scenario may be misleading with regard to
either effects observed or levels of body  burden. The difficulties of
estimating safe environmental levels have  been discussed by Kimbrough et
al. (1984), who concluded that levels >1 ppb of 2.3,7.8-TCDD are of
concern in residential soil.

2.2.3.2  Human exposure potential

     The human exposure pathways to 2,3,7,8-TCDD have changed since the
late 1970s. Because 2,3,7,8-TCDD was a contaminant in herbicide
preparations containing 2,4,5-T, the manufacture, use, and disposal of
these herbicides were the primary sources  of exposure to 2,3,7,8-TCDD.
In 1979, EPA (1979) completely banned the  use of 2,4,5-T. With the
stoppage of production of 2,4,5-T and other  pesticide preparations
containing the contaminant 2,3,7,8-TCDD, both occupational and general
population exposure to 2,3,7,8-TCDD due to manufacture and use of the
herbicides ceased to exist. Presently, the important sources of
2,3,7,8-TCDD exposures to the general population are contaminated soil,
dumpsites, and municipal incinerators. With  the change of sources, the
exposure pathways have also changed over the years (i.e., dermal and
inhalation exposure from the manufacture and use of 2,4,5-T to ingestion
of foods obtained from contaminated sites).  Thus it is not possible to
state precisely which route of exposure to 2,3,7,8-TCDD is most relevant
to the human population. As with many human exposure scenarios, it is
anticipated that all routes of exposure occur, although there are few
data available to quantify the relative contribution of each route. An
environmental partitioning model has been used to estimate the levels of
2,3,7,8-TCDD in different environmental media (Travis and Hattemer-Frey
1987). From the estimated concentration levels, the average daily human
intake of 2,3,7,8-TCDD was predicted to be 98% from ingestion of food,
2% from air, and <0.01% from ingestion of drinking water. EPA (1988)
estimated the human exposure potential to 2,3,7,8-TCDD from a variety of
exposure scenarios resulting from contaminated soils, various land
disposal situations, and municipal waste incinerators. The highest
exposure was attributed to the food chain, i.e., from  ingestion of
contaminated fish, beef, dairy products, and other foods. Ingestion of
contaminated soil, especially by children with pica tendencies; dermal

-------
                                             Health Effects Summary   23

contact with contaminated  soil,  dust,  and sediment; and inhalation of
contaminated dust and vapor  further contribute to human exposure.

     Based on data on dermal and oral  absorption in animals, it is
anticipated that 2,3,7,8-TCDD adsorbed to soil will not be as
bioavailable as the  2,3,7,8-TCDD used  in experimental studies and
administered in oily vehicles. In  the  studies available, oral absorption
of 2,3,7,8-TCDD adsorbed to  soil was still substantial but only 50% of
that from corn oil (absorption of  SO to 80%) (Lucier et al. 1986,
McConnell et al. 1984, Umbreit et  al.  1986a). Bioavailability also
varies with the type of soil, as demonstrated by Umbriet et al. (1986b)
for New Jersey and Missouri  soils, where bioavailability from New Jersey
soils was less than  that from Missouri soils. The limited studies
available may not be representative of the variation in bioavailability,
since Poiger and Schlatter (1980)  demonstrated that strong binding
vehicles, such as activated carbon, can apparently reduce
bioavailability to zero, and Philippi  et al. (1981) and Huetter and
Philippi (1982) demonstrated that  the  strength of adsorption increases
with contact time in soil. Hence,  factors such as soil type and contact
time may affect the  bioavailability of 2,3,7,8-TCDD.

     It is likely that many of the same physical properties that result
in strong binding to soil, such  as extremely low water solubility and
planar configuration, also result  in very high bioconcentration factors.
Because of the high  lipophilicity  and  long half-life of 2,3,7,8-TCDD,
exposure through ingestion of fatty tissues of fish that inhabit
contaminated areas is anticipated  to be significant. In addition, as a
result of the lipophilic nature  of milk, secretion of milk can provide a
relatively efficient mechanism for decreasing the body burden of
2,3,7,8-TCDD in females. As discussed by Rappe et al. (1985), this
elimination of 2,3,7,8-TCDD through mother's milk can result in exposure
of the infant.  Since both milk and the fatty tissues of fish are
essentially providing an oily vehicle, it seems likely that these
sources would provide 2,3,7,8-TCDD in a form that is readily
bioavailable.

2.3  ADEQUACY OF DATABASE

2.3.1  Introduct ion

     Section 110 (3) of SARA directs the Administrator of ATSDR to
prepare a toxicological profile  for each of the 100 most significant
hazardous substances found at facilities on the CERCLA National
Priorities List. Each profile must include the following content:

    "(A)  An examination, summary, and interpretation of available
          toxicological information and epidemiologic evaluations on a
          hazardous  substance in order to ascertain the levels of
          significant human exposure for the substance and the
          associated acute, subacute, and chronic health effects.

     (B)  A determination of whether adequate information on the health
          effects of each substance is available or in the process of
          development to determine levels of exposure which present a
          significant risk to human health of acute, subacute, and
          chronic health effects.

-------
24   Section 2

     (C)  Where appropriate, an identification of toxicological testing
          needed to identify the types or levels of exposure that may
          present significant risk of adverse health effects in humans."
     This section'identifies gaps in current knowledge relevant to
developing levels of significant exposure for 2,3,7,8-TCDD. Such gaps
are identified for certain health effects end points (lethality,
system/target organ toxicity, developmental toxicity, reproductive
toxicity, and carcinogenicity) reviewed in Sect.  2.2 of this profile in
developing levels of significant exposure for 2,3,7,8-TCDD, and for
other areas such as human biological monitoring and mechanisms of
toxicity. The present section briefly summarizes the availability of
existing human and animal data, identifies data gaps, and summarizes
research in progress that may fill such gaps.

     Specific research programs for obtaining data needed to develop
levels of significant exposure for 2,3,7,8-TCDD will be developed by
ATSDR, NTP, and EPA in the future.

2.3.2  Health Effect End Points

2.3.2.1  Introduction and graphic summary

     The availability of data for health effects in humans and animals
is depicted on bar graphs in Figs. 2.5 and 2.6, respectively.
     The bars of full height indicate that there are data to meet at
least one of the following criteria:

 1.  For noncancer health end points, one or more studies are available
     that meet current scientific standards and are sufficient to define
     a range of toxicity from no-effect levels (NOAELs) to levels that
     cause effects (LOAELs or FELs).

 2.  For human carcinogenicity, a substance is classified as either a
     "known human carcinogen" or "probable human carcinogen" by both EPA
     and International Agency for Research on Cancer (IARC)
     (qualitative), and the data are sufficient to derive a cancer
     potency factor (quantitative).

 3.  For animal carcinogenicity, a substance causes a statistically
     signficant number of tumors in at least one species, and the data
     are sufficient to derive a cancer potency factor.

 4.  There are studies which show that the chemical does not cause this
     health effect via this exposure route.

     Bars of half height indicate that "some" information for the end
point exists but does not meet any of these criteria.

     The absence of a column indicates that no information exists for
that end point and route.

     Although adequacy of data is indicated in Fig. 2.5 for dermal
exposure only, the route of exposure in the available studies is not
clearly defined. Because of the nature of exposures to 2,3,7,8-TCDD,
both inhalation and oral exposure are likely to occur along with dermal
exposure; in some instances, exposure from these other routes will
contribute substantially to the body burden.

-------
                                             HUMAN  DATA
                                                                                                             V  SUFFICENT
                                                                                                             ^INFORMATION'
                                                                                                                    SOME
                                                                                                                INFORMATION
                                                                                                                     NO
                                                                                                                INFORMATION
                                                                                                 INHALATION
                                                                                            DERMAL
LETHALITY
ACUTE     INTERMEDIATE    CHRONIC   DEVELOPMENTAL  REPRODUCTIVE  CARCINOQENICITV
               	    /    TOXICITV       TOXICITV
                     SYSTEMIC TOXICITY
                                                                                                                                 3-
                                                                                                                                 o
                                                                                                                                 n
                                                                                                                                 to
   'Although data for human exposure were discussed lor the dermal route, inhalation and oral exposure also contribute to the total environmental exposure of
humans to 2.3,7,8 TCDD
   'Sufficient information exists to meet at least one of the criteria for cancer or noncancer end points.
                        hig. 2.5. Availability of information on health effects of 2,3,7,8-1 ( 1)1) (human data).

-------
LETHALITY
                                              ANIMAL  DATA
                                                                                                                                ro
                                                                                                                                §
                                                                                                                SUFFICIENT     *»
                                                                                                               INFORMATION*
                                                                                                            V     SOME

                                                                                                            /INFORMATION
                                                                                                          J
                                                                                                                    NO
                                                                                                               INFORMATION
                                                                                                    ORAL
                                                                                               INHALATION
                                                                                          DERMAL
               ACUTE     INTERMEDIATE
 CHRONIC   DEVELOPMENTAL  REPRODUCTIVE  CAHCINOOENICITY

	/    TOXICITY       TOXICITY
                     SYSTEMIC TOXICITY
                      'Sufficient information exists to meet at least one ot the criteria for cancer or noncancer end points.


                       Fig. 2.6. Availability of information on health effects of 2,3,7,8-TCDD (animal data).

-------
                                             Health Effects Summary   27

2.3.2.2  Descriptions of highlights of graphs

     Human.  Figure 2.5 indicates that there are very little human data
on the toxicological effects of exposure to 2.3,7,8-TCDD.  The data
obtained from human studies are considered to be from dermal exposures,
since most humans were exposed in contaminated areas considerably after
the application or initial release of the chemicals containing
2,3,7,8-TCDD. This long-term exposure would occur through contact of the
skin with soil and other articles contaminated with 2,3,7,8-TCDD.
Inhalation and oral exposures were also likely to occur through
inhalation of contaminated dust and ingestion of contaminated food and
dust. From reports of human exposure (acute accidental exposure as well
as repeated exposure), there is a clear indication that chemicals
containing 2,3,7,8-TCDD cause chloracne and there is some indication
that chemicals containing 2.3,7,8-TCDD are hepatotoxic; however, data
are insufficient to determine the levels of exposure required for
induction of these effects. Schecter et al. (1987) reported a calculated
effect level of 9.7 Mg for the body burden of 2,3,7,8-TCDD that resulted
in chloracne in some industrial workers; however, verification of this
value is not available. By extrapolating dose-effect data obtained from
the ingestion of contaminated rice oil containing polychlorinated
biphenyls and polychlorinated dibenzofurans to dose-effect data for
2,3,7,8-TCDD and taking into consideration the minimum toxic dose for
the production of chloracne in nonhuman primates, Stevens (1981)
estimated a minimum toxic dose for 2,3,7,8-TCDD of 0.1 MgAg f°r humans.
The epidemiologic data on the effects of 2,3.7,8-TCDD on fetal
development, human reproduction, immuno function, and the development of
cancer are not adequate to either prove or disprove an association.

     There are no additional data on the toxicity of 2,3,7,8-TCDD
following inhalation exposure or oral exposure.
     Animal.  Figure 2.6 indicates that there are considerably more data
on the toxicological effects of 2,3,7,8-TCDD in animals than in humans,
although the data are primarily limited to oral studies. For animal
data, it should be recognized that "adequate" may only apply to  the
specific species tested, since large species differences are observed
with 2,3,7,8-TCDD. As a result of these large differences  in species
sensitivity  (4 orders of magnitude for LD50 values), .complete
information  on a specific end point will be difficult  to obtain.  For  the
complete understanding of a toxicological  end point,  the most  sensitive
species must be determined and adequately  tested, or  it must be  clearly
demonstrated which animal species is most  representative of humans. The
lack of adequate data on species  sensitivity  is  a significant  gap in  the
toxicological data on 2,3,7,8-TCDD.
     Although dermal exposure  is  a potentially  significant route of
human exposure,  the only animal  data available  for  this  route  are a
single determination of a  lethal  dose  in  rabbits  (Schwetz  et  al.  1973)
and  a few  studies of  the effects  of  2,3,7,8-TCDD in the  two-stage
tumorigenesis assay  (EPA 1985a)  of  the  mouse.  Both  of these types of
studies provide  only  limited  data on the  dermal toxicity of
2,3,7,8-TCDD.

-------
28   Section 2

2.3.2.3  Summary of relevant ongoing research

     A review of Federal Research in Progress will  show that
investigations of the toxicological properties of 2,3,7,8-TCDD are very
active. This ongoing research is summarized in Table 2.1.  Also,  a very
important area of ongoing research is reviewed by Young and Kang (1985)
and Hanson (1987). This area consists of 15 ongoing epidemiology studies
that are being conducted for the U.S. government and coordinated by the
White House Agent Orange Working Group.  Subject areas include mortality
and morbidity, with particular concern for carcinogenic effects,
including the development of soft tissue sarcomas,  and anatomical birth
defects. A number of these studies are concerned with veterans exposed
to the herbicide Agent Orange in Vietnam.

2.3.3  Other Information Needed for Human Health Assessment

2.3.3.1  Pharmacokinetics and mechanisms of action

     Mechanisms of action.  The mechanism of 2,3,7,8-TCDD toxicity is
under extensive investigation. As reviewed by Roberts et al. (1985), it
has been proposed that 2,3,7,8-TCDD functions by a  receptor-mediated
mechanism. There is evidence that this mechanism is associated with many
of the toxicological end points of 2,3,7,8-TCDD, as indicated by the
segregation of the toxic properties of 2,3,7,8-TCDD with the Ah locus,
which is near the locus for the 2,3,7,8-TCDD receptor. Some toxicologic
end points, however, do not segregate with the Ah locus. It has been
shown in tissue cultures with human lymphoblastoid cells that there is a
large genetic variation in aryl hydrocarbon hydroxylase inducibility
(Nagayama et al. 1985), and the susceptibility of these cells to the
toxicity of 2,3,7,8-TCDD parallels the induction of this monooxygenase
system. In addition, the levels of the receptors in a particular species
do not necessarily correspond to species sensitivity. Thus, although a
receptor-mediated mechanism is well established, the complete
integration of this mechanism with the toxic action of 2,3,7,8-TCDD has
not been fully characterized.

     Other studies on the mechanisms of action of 2,3,7,8-TCDD  are also
being followed; for example, Rozman et al. (1985) have described results
of a study which  indicates that the  thyroid hormone thyroxine (T4) has
the ability to modulate the toxicity of 2,3,7,8-TCDD. Male Sprague-
Dawley rats were  divided into groups of normal rats, thyroidectomized
rats, and thyroidectomized rats that received T4 replacement  therapy.
Following administration of 2,3,7,8-TCDD, mean time to death  and percent
mortality at 90 days were similar between the normal rats and the
thyroidectomized  rats that received T&  (35 days and 89%, and  37 days and
80%, respectively). Thyroidectomized rats that did not receive  T4
survived longer  (mean time to death  of  63 days) and had lower mortality
at 90 days  (44%).  It was also demonstrated that the 2,3,7,8-TCDD
treatment resulted in a decrease  in  serum T4  levels in both normal  and
thyroidectomized  T4-supplemented  rats.  The modulation of  the  lethal
effects of  2,3,7,8-TCDD was considered  by the authors to be  related to
thyroid hormone modulation of energy metabolism. The participation  of
this mechanism  in the development of 2,3,7,8-TCDD  toxicity  needs  further
study.

-------
                                                  Table 2.1. Research In progress OB 2^,7,8-TCDD
Principal
investigator
Ami. S.D.
Ausl. J.B.
Bowman. R
Dubold. G J.
Fujimolo. J.M
Gasicwicz, T.A.
Gasiewicz, T A
Title
Toxic and anorectic effects of TCDD (rats, mice)
Fat tissue analysis for 2.3,7.8-telrachlorodibenzo-/>-dioxin (TCOD)
Kinetic*, reproductive, unmunofunclion, behavioral toxicily
studies (monkeys)
Oploacouslic detection of carcinogens
Effects of 2.3.7.8-lctrachlorodibcnzodioxin on hcpatobiliary
function in animals
Molecular toxicology of TCOD (rats, mice, guinea pigs, hamsters)
Chlorinated dibenzo-p-dioxms— mechanisms of toxicity
Performing
organization0
Michigan Stale University
VA Medical Center, San Antonio
University of Wisconsin Primate
Research Center
Brown University
VA Medical Center. Milwaukee
University of Rochester
University of Rochester
Sponsoring
organization"
NIEHS
VA
EPA
NIEHS
VA
NIEHS
NIEHS
                    (rats, mice, guinea pigs)
Gtertby. J F         Dioxin-cpithelial cell interactions—Mechanism and assay
Giesy, J.P.          Effects of toxic chemicals on aquatic systems
Gustafsson, J.        Structure and function of the TCDD receptor (rats)
Guslafsson, J        Binding and metabolism of toxic agents in the prostate (rats)
Holsapple, H.P      Immunotoxicology by chlorinated dibenzo-p-dioxins (mice)
Kurth. M.J.         Immunoassays for pyrrolizidme alkaloids and metabolites (rodents)
Miller. E.C          Biochemical studies in chemical carcmogenesis
Ncben. D W        Pharmacogenelics
Nelson. K           Receptor interaction and liver tumor promotion
Olson, J.R          Mechanism(s) for loxicily of chlorinated dibenzodioxins
Peterson. R E        Environmental pollutants and toxicology of the liver (rats, monkeys)
                    (rodents, humans)
Piper. W N          Toxicant deregulation of endocrine heme biosynthesis
                    (rats, hamsters, guinea pig)
N.Y Slate Department of Health
Michigan Slate University
Caroline Institute
Caroline Institute
Virginia Commonwealth University
University of California, Davis
University of Wisconsin
NICHD
NIEHS
SUNY at  Buffalo
University of Wisconsin
NIEHS & NYSDH
USDA
NIEHS
NCI
NIEHS
NIEHS
NCI
NICHD
NIEHS
NIEHS
NIEHS
ID
to
A
O
n
(a
to
University of Nebraska Medical Center     NIEHS
                                                                                                                                                                ro
                                                                                                                                                                vO

-------
                                                          Table 2.1 (coBliBoed)
Principal
investigator
Rice. R.H.
Russell. D.H.
Safe. S.H.
Safe. S.H.
Safe. S.H.
Shiremao, R B
Shivcnck, K.T.
Shiverick. KT
Silkworth, J.B
Whitlock, J.P.. Jr
Title
Keratinocyte envelopes — physiology and toxic mechanisms
(rau, rabbits, humans)
Mechanism(i) of TCDD toxicity (rats)
Mechanisms of dioxin toxicity (mice. rau. guinea pigs, avian)
Environmental toxicology of balogenated aromatic compounds
TCDD — effects of receptor modulators/antagonists
(mice, hamsters, guinea pigs)
Mechanisms of the transfer of sterols and glyccrides into cells
TCDD efTccU on steroid hormone synthesis in pregnancy (rau)
Steroid and xcnobiolic metabolism in the placenta (rats)
PCB unmunotoxicily (mice)
Carcinogen-metabolizing enzymes — action in variant cells (mice)
Performing
organization"
Harvard University
University of Arizona
Texas AAM University
Texas AAM University
Texas AAM University
University of Florida
University of Florida
University of Florida
N.Y. Slate Department of Health
Stanford University
Sponsoring
organization"
NIAMSD
NIEHS
NIEHS
USDA
NIEHS
USDA
NIEHS
NICHD
NIEHS, A NYSDH
NCI
"NCI - National Cancer Institute.
 NIAMSD - National Institute of Arthritis and Musculoskeletal and Skin Diseases
 NICHD - National Institute of Child Health and Human Development.
 NIEHS -  National Institute of Environmental Health Sciences
 NYSDH - New York State Department of Health
 USDA —  United Slates Department of Agriculture.
 VA — Veterans Administration.
CO
It
n
it
i-.
s
is»

-------
                                             Health Effects Summary   31

     In addition, mechanisms for individual end points, such as the
wasting syndrome and the development of cleft palate, have also been
investigated.  Although a number of mechanisms have been proposed for
such end points, there has yet to be a definitive understanding of the
underlining biochemical processes that result in the observed effects of
2,3,7,8-TCDD. Further investigation of these processes is necessary not
only to understand how 2,3,7,8-TCDD induces certain toxic end points,
but also how the different target organ responses relate to each other.
     Target organ/pharmacokinetic profiles.  There are few data to
indicate that target organs (such as liver and thyroid) contain
relatively higher levels of 2,3,7,8-TCDD than other, nontarget tissues.
In general, 2,3,7,8-TCDD appears to distribute to organs in proportion
to lipid content (Ryan et al. 1985b) rather than in regard to the
sensitivity of the organ to the toxic effect of 2,3,7,8-TCDD. Roberts et
al. (1985), however, reported that the distribution of the 2,3,7,8-TCDD
receptor may be a better indicator of target organ than the distribution
of 2,3,7,8-TCDD itself. Further work is needed to clarify this issue.
     Further research is also needed to determine the association
between a species' capability to metabolize 2,3,7,8-TCDD and the
sensitivity of that species to 2,3,7,8-TCDD-induced toxicity. Although
there are data suggesting that the ability to metabolize 2,3,7,8-TCDD is
important in determining species sensitivity to this chemical, the data
are not complete and a mechanism has not been proven.
     Ongoing research.  There are a relatively large number of studies
reported in Federal Research in Progress that are involved with the
toxicokinetics and the mechanisms of action of 2,3,7,8-TCDD. The list of
projects is too extensive for inclusion in this profile.

2.3.3.2  Monitoring human biological samples
     Adequate analytical methods are available to investigate 2,3,7,8-
TCDD levels in biological materials that are lipophilic and  thus
concentrate 2,3,7,8-TCDD. Biological fluids with high  lipid  content  for
which 2,3,7,8-TCDD quantification methods  in the low ppt range are
available are cow's milk and human breast milk (Sect.  7.2.4). Young
(1984) reported  that the methodology is not available  for detecting
2,3,7,8-TCDD in blood or tissues with low  lipid content, although  a
recent method reported  the detection of 2,3,7,8-TCDD  in parts-per-
quadrillion levels  in human  serum  (Patterson et al.  1987b).  The
development of more sensitive analytical methods is  necessary  for
environmental analysis, and  the  resulting  technology will be used  in
monitoring biological samples. At present,  the ability to monitor
2,3,7,8-TCDD in  human tissues appears to exceed the  ability  to  interpret
the toxicological significance of  the observed results.

2.3.3.3  Environmental  considerations
     Analytical  methods.   Concentrations of 2,3,7,8-TCDD  in  ambient  air
and drinking water  are  generally so  low  that  the existing  analytical
methodologies cannot measure the levels  in these  two media.

-------
32   Section 2

     Bioavailability. .The dependence of 2,3,7,8-TCDD bioavailability r
the matrix to which it is bound has been suggested by several
investigators. In guinea pigs, the bioavailability of 2,3,7,8-TCDD from
a contaminated soil in Newark, New Jersey,  was  reported to be 0.5% of
the administered dose, whereas a different  soil from Newark showed a
bioavailability of 21.3% (Umbrtet et al.  1986). From the studies of
McConnell et al. (1984), Umbriet et al.  (1986)  estimated a
bioavailability of about 85% in guinea pigs for soils from the Times
Beach area of Missouri. Similarly, the uptake of 2,3,7,8-TCDD from fly
ash to freshwater fish was found to depend  on the carbon content of the
fly ash (Kuehl et al. 1985). The bioavailability of 2.3.7.8-TCDD in male
Wistar rats was at least 10 times lower when the administered dose was
in the form of fly ash particle compared with dosage in oily vehicles
(Van den Berg et al. 1987). With oral dosage of contaminated Missouri
soil, Lucier et al. (1986) observed a dependence of bioavailability in
rats on its concentration in soil, the bioavailability being higher at
higher dosage. The dermal bioavailability in rats, however, was
approximately 1% of the administered dosage following 24 hours of
contact with skin, and the uptake was not influenced either by the
concentration of 2,3,7,8-TCDD in soil or the presence of oily vehicles
(Shu et al. 1987).  The insufficient data that  exist today indicate that
2,3,7,8-TCDD can bind strongly in soils with high organic carbon
content, thus decreasing its bioavailability. The available data also
indicate that bioavailability from soil may depend on its residence time
in soil (Shu et al. 1987). As it remains in soil, the fraction of the
irreversibly sorbed part increases and thus renders it less
bioavailable. Certain other factors (e.g.,  the  concentration of
2,3,7,8-TCDD in a medium, difference in species, presence of co-
contaminants in the dosage medium, lipid content of diet, and
interaction of 2,3,7,8-TCDD with host site) may alter the
bioavailability. Although fragmentary data on the dependence of
bioavailability on different parameters are available, no systematic
study has been made that attempts to explain the dependence of
bioavailability on the several possible parameters.
     Environmental fate and transport.  Some progress on the fate and
transport of 2,3,7,8-TCDD in environmental media has been made  in recent
years. Substantial gaps still exist in quantitative data regarding  its
biodegradability, photolysis, and volatility from environmental media.

     Interactions with other common co-contaminants.  There are no  data
to indicate that 2,3,7,8-TCDD will interact chemically with other
contaminants under environmental conditions. The presence of 2,3,7,8-
TCDD in both biological systems and the environment, and the lack of  any
reactive groups, would support the conclusion  that  2,3,7,8-TCDD will  not
easily react with other compounds.

     Ongoing  research.  Present research efforts  are focused primarily
on analytical methodology  to  develop more monitoring data. The  objective
of most of  this research  is to develop new  analytical methods  that  will
unequivocally identify  and  quantify very small  amounts  of  2,3,7,8-TCDD
in various  environmental media with a faster turnover  time. An example
of such an  ongoing  research is the investigation now being performed b**
Robens and  Zabik  (n.d.).  Substantial research  is  in progress  to
determine  the concentration of 2,3,7,8-TCDD in serum and human milk

-------
                                             Health Effects Summary   33

samples.  Groups of investigators in the Centers for Disease Control,
research organizations in Sweden, and at Rutgers University, the State
University of New York at Binghamton, and the Canadian Food Research
Division are involved in such efforts (Ryan 1987).

-------
                                                                      35
                 3.  CHEMICAL AND PHYSICAL INFORMATION

3.1  CHEMICAL IDENTITY

     The chemical identity of 2,3,7,8-tetrachlorodibenzo-p-dioxin,  to
be referred to as 2,3,7,8-TCDD throughout this document, is given in
Table 3.1.

3.2  PHYSICAL AND CHEMICAL PROPERTIES

     Selected physical and chemical properties of 2,3,7,8-TCDD are shown
in Table 3.2. 2,3,7,8-TCDD is stable toward heat, acids, and alkalies,
but begins to decompose at 500"C. The decomposition is virtually
complete within 21 s at 800°C. 2,3,7,8-TCDD is susceptible to photo-
degradation in the presence of ultraviolet light, particularly in the
presence of a hydrogen-donating solvent. Gamma radiation degrades
2,3,7,8-TCDD in organic solvents (EPA 198Sa).

-------
36    Section 3
                      Table 3.1.  Chemical identity of 2,3,7,8-TCDD
Chemical name
Trade name

Chemical formula
Wiswesser line notation


Chemical structure
 Identification Nos.
   CAS Registry No.
   NIOSH RTECS No.
   EPA Hazardous Waste No.
   OHM-TADS No.
   DOT/UN/NA/IMCO Shipping No.
   STCC No.
   Hazardous Substances Data Bank No.
   National Cancer Institute No.
2,3,7,8-tetrachlorodibenzo[b,e]( 1,4)-dioxin;
2,3,7,8-tetrachlorodibenzo-p-dioxin; dioxin; TCDBD;
2,3,7,8-TCDD; 2,3,7.8-tetrachlorodibenzodioxm;
2,3,7,8-tctrachlorodibenzo-l,4-dioxm (EPA 1985)

None. (The compound is not produced commercially.)
(EPA 1985b)
C,2H4CU02
TC666 BO IOJ EG FG LG MG or
TC666 BO IOJ DG EG LG MG (HSDB 1987)
                                        Cl
1746-01-6 (SANSS 1987)
HP3500000 (SANSS 1987)
Not assigned (HSDB 1987)
8300192 (SANSS 1987)
Not assigned (HSDB 1987
Not assigned (HSDB 1987)
4151 (HSDB 1987)
NCI-C03714 (SANSS 1987)

-------
                                             Chemical  and Physical Information    37
                          Table 3.2.  Physical properties of 2,3,7,8-TCDD
        Property
                  Value
      References
Molecular weight
Color
Physical state
Odor
Melting point
Boiling point
Autoigmtion temperature
Solubility
  Water (ng/L)
321.97
Colorless
Solid at room temperature
Unknown
305 °C
412.2°C (estimated)
NAfl

7.91 (20-22°C), 19.3 (22°C)
  Organic solvents (mg/L)   o-Dichlorobenzene,  1400; chlorobenzene, 720;
                           benzene, 570; chloroform, 370;
                           methanol,  10; acetone, 110
Density (g/mL)
Partition coefficients
Vapor pressure (mm Hg)

Henry's Law constant

Flash point
Refractive index
Flammability limits
Conversion factors
  Vapor
1.827 (estimated)
Log 1^:6.15-7.28
Log K^: 6.0-7.39
1.52 X 10-'(25°C)
1.4 X 10~9 (estimated at 25°C)
8.1  X 10~s atm-mj/mol (25°C) (estimated
from water solubility and vapor pressure)
6.4 X 10~* atm-m3/mole (estimated)
Unknown
Unknown
Unknown

1 ppb = 13.384 Mg/m3 at 20°C
EPA 1985a
EPA 1985a

Schroy et al. 1985, 1986
Schroy et al. 1985
Adams and Blame 1986,
Marple et al. 1986a
Schroy et al. 1985
Schroy et al. 1985
EPA 1985a.
Schroy et al. 1985,
Jackson et al. 1986,
Marple et al. 1986b
Schroy et al. 1986
Palansky et al. 1986
Podoll et al. 1986
    "Not available.

-------
                                                                      39
                         4.  TOXICOLOGICAL DATA
4.1  OVERVIEW
     Human and animal data indicate that 2,3,7,8-TCDD can be absorbed
following ingestion, and although bioavailability is affected by binding
to soils, the extent of absorption is only decreased by -50%. In
addition, animal data indicate that 2,3,7,8-TCDD may be absorbed well
through the skin depending on such variables as the dose or the material
to which 2,3,7,8-TCDD is adsorbed. Following absorption, 2,3,7,8-TCDD is
distributed to tissues in proportion to the lipid content. 2,3,7,8-TCDD
can cross the placenta with subsequent exposure of the fetus, and the
newborn can be exposed following redistribution of 2,3,7,8-TCDD during
lactation. Metabolism of 2,3,7,8-TCDD is currently thought to result
primarily in detoxification and relatively rapid removal of the
metabolites through excretion in the bile and urine. Unmetabolized
2,3,7,8-TCDD is also excreted through direct intestinal elimination in
feces as well as through lactation. There are species and strain
differences in both metabolism and elimination rates, with elimination
half-lives varying from 11 days in the hamster, which is relatively
resistant to 2,3,7,8-TCDD toxicity, to >1 year in the monkey, which is
sensitive to the toxicity of 2,3,7,8-TCDD. There is, however, no clear
correlation between toxicity and the elimination half-life for 2,3,7,8-
TCDD.
     Although humans have been exposed to 2,3,7,8-TCDD as a contaminant
of herbicides and industrial chemicals, there have been no reported
deaths from acute exposure. Lethal oral doses of 2,3,7,8-TCDD vary from
0.6 to 5,000 pg/kg for guinea pigs and hamsters, respectively, with
other species tested having LD50 values between these two extremes.
Subchronic LD50 values for cumulative (total) exposure during a 90-day
oral study with guinea pigs were essentially the same' as  those observed
after acute exposure. A 9-month feeding study of a small  number of
monkeys indicated that a dose of 2 to 3 MgAg resulted  in death. The
acute dermal LD50 value for 2,3,7,8-TCDD  in rabbits has been reported  to
be 275 MgAg: however, no other species have been tested, and rabbits
are only  intermediate in sensitivity to 2,3,7,8-TCDD in oral toxicity
studies.  Inhalation experiments have not  been conducted.
     The  only effect clearly demonstrated to be produced  in humans
following 2,3,7,8-TCDD exposure is chloracne. This  lesion is a systemic
toxic effect and not solely a dermal effect. Although animal models are
available to study chloracne, there are limitations  in  investigating
this toxicological end point. The  lesions produced  in animals are
different from the lesions in humans, and the expression  of  this lesion
may be limited to monkeys, specific strains of mice, and  rabbits
(observed only on the ears after  dermal application).

-------
40   Section 4

     In animals, the toxic effect of 2,3,7,8-TCDD exposure most often
observed is the wasting syndrome, in which the animals progressively
lose body weight prior to death, although no one organ is altered to
such an extent that it can be associated with the overt signs of
toxicity. Although this syndrome is observed in all species of chorduca
tested, it occurs predominantly only at doses that are lethal or near
lethal. The wasting syndrome has not been confirmed in humans, although
loss of weight has been included in the list of effects associated with
acute exposure to chemicals contaminated with 2,3,7,8-TCDD.
     Immunotoxicity has also been observed in a variety of animal
species and may be one of the most-sensitive effects of 2,3,7,8-TCDD.
Immunotoxicity, however, has not been directly related to 2,3,7,8-TCDD
exposure in humans.
     Hepatotoxicity has been observed in a variety of animal species,
and there is suggestive evidence that this effect also occurs in humans.
The human data are not completely clear, however, since mixed exposure
to other potentially hepatotoxic chemicals occurred. In addition,
species variations in induction of hepatotoxic effects indicate that the
two most sensitive species, guinea pigs and monkeys, develop relatively
less severe hepatic lesions than many other species.
     Other organ systems are also affected by 2,3,7,8-TCDD. There is
suggestive evidence in humans that the nervous system may be affected;
in animals, there are data on effects on the digestive system and the
kidney. These additional organ systems have not been investigated as
extensively as those described later. Also, they do not appear to be
prime  indicators of 2,3,7,8-TCDD toxicity or provide any special insight
into extrapolating effects observed in animals to those in man.

     Many of the toxic end points discussed have been observed to have
extensive species and strain differences in sensitivity. These
differences may be related to the receptor-mediated mechanism of
toxicity proposed for 2,3,7,8-TCDD. This mechanism follows the scheme of
2,3,7,8-TCDD binding to a soluble cytoplasmic receptor protein, which
subsequently migrates into the nucleus of the cell as a receptor-
2,3,7,8-TCDD complex. Once in the nucleus,  it is proposed  that both
transcription and translation of DNA may be affected. The  receptor
protein  is known genetically to  segregate with the Ah locus,  and many of
the strain differences  in toxicity observed in mice are explained by
this segregation. The receptor protein  is known  to be widely  dispersed
in different organ  systems and  in  different species,  although
quantitative levels differ. These  quantitative differences may help
explain  some of the species differences  in  sensitivity. The  receptor
mechanism cannot explain all the species and strain differences  in
2,3,7,8-TCDD toxicity,  because  some  toxic responses segregate with  the
Ah receptor while  others do not.

     The evidence  from  studies  of  human populations exposed  to
herbicides  and other  industrial  chemicals known  to be contaminated  with
2,3,7,8-TCDD  is inadequate  to demonstrate  that  2,3,7,8-TCDD  is  a human
developmental  toxicant. The data from  studies in laboratory  mice and
rats,  however,  clearly  demonstrate in  these species  that 2,3,7,8-TCDD
induces  a variety  of  developmental abnormalities.

-------
                                                 Toxicologies! Data   41

     The data from human populations exposed to herbicides and other
industrial chemicals contaminated with 2,3,7,8-TCDD are not adequate to
determine if 2,3,7,8-TCDD adversely affects reproductive health in
humans. Animal studies clearly demonstrate that 2,3,7,8-TCDD is
fetotoxic in several species, causing several effects including
spontaneous abortions. There is additional evidence that 2,3,7,8-TCDD
affects male hormone levels and the function of male reproductive
organs.
     Although 2,3,7,8-TCDD has not consistently produced positive
results in genotoxicity assays, there are positive results in diverse
bioassay systems which may suggest that 2,3,7,8-TCDD is a genotoxic
agent; however, the large numbers of negative results in assays usually
predictive of genotoxic agents suggest that 2,3,7,8-TCDD is not a
genotoxic agent.
     The human evidence that 2,3,7,8-TCDD is a carcinogen is
conflicting, with both positive associations and nonassociations
observed for the induction of cancer in cohorts exposed to herbicides
and other chlorinated chemicals known to be contaminated with 2,3,7,8-
TCDD. As a result, the human data only provide inadequate evidence that
2,3,7,8-TCDD is a human carcinogen. The animal data, however, provide
clear evidence that 2,3,7,8-TCDD is carcinogenic in animals.
     Exposure to 2,3,7,8-TCDD in the environment is never to 2,3,7,8-
TCDD alone but to materials such as incinerator fly ash or industrial
wastes, which contain 2,3,7,8-TCDD along with many other congeners of
PCDDs, as well as other potentially toxic materials. The EPA has
recognized the public and toxicological concerns resulting from exposure
to these compounds, as well as the gaps in available information with
which to evaluate the human health potential from exposure (EPA 1987).
In response to this problem, the EPA Chlorinated dibenzo-p-
dioxins/Chlorinated dibenzofurans Technical Panel of the Risk Assessment
Forum has developed and recommended an interim method for assisting in
estimating the risk from exposure to these chemical mixtures that can be
used until the data gaps are filled. This procedure generates the
"2,3,7,8-TCDD equivalence factors" (TEFs) of complex mixtures of
chlorinated dibenzo-p-dioxins and dibenzofurans based on congener- and
isomer-specific data. The TEFs presented in Table 4.1 are relative
values and are a means of relating toxicity data for other chlorinated
dibenzo-p-dioxins and dibenzofurans to an equivalent level of 2,3,7,8-
TCDD. The TEF for 2,3,7,8-TCDD is defined as unity, whereas  all other
TEFs are unity or less, thus reflecting the lower toxic potency of most
PCDD congeners. These relative values were developed using a tiered
approach to the evaluation of existing data. Definitive data on  the
human  carcinogenicity of a congener of 2,3,7,8-TCDD are the  most
appropriate data for establishing the TEF. If  this human data were not
available, animal carcinogenicity data would be used,  followed by  the
data on reproductive toxicity. Estimated exposure levels resulting  in
reproductive and carcinogenic effects are very similar  for  2,3,7,8-TCDD.
Finally, if none of the above data are available, the TEFs  are
determined by  the weight of  evidence  from  in vitro  tests with
particular weight placed on  data from tests evaluating  receptor  binding
interactions and  induction of  oxidative enzymes. The TEFs  thus  generated
can be used, assuming additivity of  the toxic  response,  for estimating

-------
42    Section
                    Table 4.1.  Recommended TEFs" for CDDs* and CDFsc
Compound
CDDs
EPA current
CDDs recommended values
MonoCDDs
DiCDDs
TriCDDs
2,3.7,8-TCDD
Other TCDDs
2,3.7,8-PeCDDs'
Other PeCDDs
2J,7,8-HxCDDs'
Other HxCDDs
2,3,7,8-HpCDDs'
Other HpCDDs
OCDD
0
0
0
I
0.01
0.5
0.005
0.04
0.0004
0.001
0.00001
0
Compound
CDFs
EPA current
CDFs recommended values
MonoCDFs
DiCDFs
TriCDFs
2,3.7,8-TCDFs
Other TCDFs
2,3,7,8-PeCDFs*
Other PeCDFs
2,3,7,8-HxCDFs*
Other HxCDFs
2,3,7,8-HpCDFs'
Other HpCDFs
OCDF
NR
NR
NR
0.1
0.001
0.1
0001
0.01
0.0001
0.001
0.00001
0
        TEFs = Toxicity equivalence factors.
        *CDDs = Chlorinated dibenzo-p-dioxins.
        cCDFs = Chlorinated dibenzofurans.
        rfNR = Not reported.
        'Any isomer that contains chlorine in the 2,3,7,8 positions.
        Source: EPA 1987a.

-------
                                                 ToxLcoLogical Data   43

the toxicity relative-to 2,3,7,8-TCDD of a mixture containing a known
distribution of congeners of 2,3,7,8-TCDD.

4.2  TOXICOKINETICS

4.2.1  Absorpt ion

4.2.1.1  Inhalation
     Pertinent data regarding the absorption of 2,3.7,8-TCDD by humans
or animals following inhalation exposure were not found in the available
literature.

4.2.1.2  Oral
     Human.   The absorption data available are from an elimination study
of 2,3,7,8-TCDD in which a male volunteer ingested 3H-2,3,7,8-TCDD in
corn oil at a dose of 1.14 ng/kg (Poiger and Schlatter 1986).
Measurements of cumulative elimination in the feces and urine, as well
as a determination of sequestering in fat via biopsy samples, indicated
that >87% of the dose was absorbed. Following absorption, the half-life
for elimination was calculated to be 2,120 days. CDC (1987) determined a
half-life of 2,595 days by measuring the decrease in serum levels of
2,3,7,8-TCDD in Vietnam veterans.
     Animal.  Studies reviewed in EPA (1985a) show that 2,3,7,8-TCDD is
generally well absorbed (50 to 80%) when administered to rats, guinea
pigs, or hamsters in a lipophilic vehicle by gavage  (Nolan et al. 1979,
Olson et al. 1980, Piper et ai. 1973). There appeared to be no change in
absorption rates with repeated dosing, but there was some decrease in
absorption at higher dose levels. Absorption of 2,3,7,8-TCDD when
administered in the diet was also between 50 and 60% (Fries and Marrow
1975).
     The vehicle in which 2,3,7,8-TCDD is administered has substantial
effects on its gastrointestinal absorption. As described  in EPA  (1985a),
Poiger and Schlatter (1980) observed a decrease in the absorption of
2,3,7,8-TCDD when administered in a soil  suspension  compared with a
solution in 50% ethanol, whereas no absorption occurred when  the
compound was administered as a suspension of activated carbon. Since
2,3,7,8-TCDD in the environment  is likely to be adsorbed  to  soil,
McConnell et al.  (1984) and Lucier et al. (1986) compared the  absorption
of 2,3,7,8-TCDD from contaminated  soil with  that  from 2,3,7,8-TCDD
administered in corn oil. As indicated by biological effects  and the
amount of 2,3,7,8-TCDD  in the  liver,  the  absorption  was -50%  less  from
soil  than from com oil. Umbreit et al.  (1986a)  showed that  2,3,7,8-
TCDD-contaminated soil  was less  toxic than an  equivalent  amount  of
2,3,7,8-TCDD, suggesting that  binding to  soil  had an influence on
bioavailability. Van den Berg  et al.  (1983,  1985,  1987) have studied the
bioavailability of polychlorinated dibenzodioxins  and dibenzofurans  from
fly  ash and determined  that bioavailability  was lowest on fly ash (0.4%
for  2,3,7,8-TCDD) as compared  to extracts of the same fly ash
administered  in an oily vehicle  (45%  for 2,3,7,8-TCDD).  Other effects on
the  observed bioavailability,  species  (rats,  hamsters, and guinea pigs),
and  specific congener may have resulted from differences in metabolism

-------
44   Section 4

or distribution, since 2,3,7,8-TCDD bioavailability was measured by
retention of the compounds in the liver.  These data indicate that
substantial absorption occurs from contaminated soil;  however,  soil type
and duration of contact, as suggested from the data that demonstrated
decreased extraction efficiency with increasing contact time between
soil and 2,3,7,8-TCDD (Huetter and Philippi 1982,  Philippi et al. 1981),
may substantially affect the absorption of 2,3,7,8-TCDD from soils
obtained from different contaminated sites.  Bioavailability appears to
be low from fly ash; however, studies have been very limited, and
factors such as source of the fly ash, moisture content,  or the co-
ingestion of lipophilic material may affect bioavailability.

4.2.1.3  Dermal

     Human.  Pertinent data regarding the absorption of 2,3,7,8-TCDD
following dermal exposure in humans were not found in the available
literature.

     Animal.  Data regarding dermal absorption in animals are limited.
2,3,7,8-TCDD is absorbed well through the skin, although as with oral
absorption the vehicle can substantially affect absorption. As discussed
in EPA (1985a), it was estimated that 40% of the dose was absorbed by
rats when the compound was applied in methanol, whereas application in
vaseline or polyethylene glycol resulted in 1.4 and 9.3% absorption,
respectively (Poiger and Schlatter 1980). Applying 2,3,7,8-TCDD as a
paste in soil or activated carbon resulted in poorer absorption than
observed in oral studies, with absorption of <2% and nondetectable,
respectively.

4.2.2  Distribution

4.2.2.1  Inhalation

     Pertinent data regarding the distribution of 2,3,7,8-TCDD following
inhalation exposure of humans and animals were not found in the
available literature.

4.2.2.2  Oral

     Human.  Poiger and Schlatter (1986) estimated that -90% of  the body
burden of 2,3,7,8-TCDD was sequestered in the fat after a volunteer
ingested 3H-2,3,7,8-TCDD in corn oil at a dose of 1.14 ng/kg. During
this study, which lasted 135 days, elevated radioactivity was detected
in the blood only during the initial 2 days following treatment. The
data would be consistent with the high bioconcentration potential of
2,3,7,8-TCDD in humans, as calculated by Geyer et al. (1986) from daily
intake assumptions, levels in human adipose tissue, and pharmacokinetic
models. Adipose tissue has been examined to determine if levels  of
2,3,7,8-TCDD in this reservoir correlated with exposure. Adipose tissues
of Vietnam veterans exposed to Agent Orange and humans occupationally
exposed to potential sources of 2,3,7,8-TCDD were reported  to have  up to
10 times the level of 2,3,7,8-TCDD in unexfosed control subjects
(Schecter et al. 1985, Gross et al. 1984); however, another  study by
Weerasinghe et al.  (1986) failed to detect any difference between  a
group of veterans and unexposed subjects. Although a clear  correlation

-------
                                                 Toxicologies! Data   45

with exposure was not demonstrated, it was apparent that adipose tissue
is a depot for 2,3,7,8-TCDD, and detectable levels were observed in
subjects with no known exposure. Ryan et al. (1985b) further examined
the distribution of 2,3,7,8-TCDD in two humans at autopsy. They
determined on a weight basis that 2,3,7,8-TCDD distributed in descending
order to fat (-6 ppt) and liver (-2 ppt), with levels in muscle and
kidney below detection; however, 2,3,7,8-TCDD levels compared on a per
lipid basis were similar between tissues. These data should be
interpreted with caution, since only two subjects were examined and one
of the subjects was suffering from fatty liver syndrome; therefore, the
data cannot be generalized to the entire population.

     Animal.  EPA (1985a) reviewed a number of early studies that
described the distribution of 2,3,7,8-TCDD in rats, mice, and guinea
pigs. In rats and mice, the liver contained the greatest amount of
2,3,7,8-TCDD, followed by the fat; however, in guinea pigs this was
reversed. Similar patterns were observed after intraperitoneal
administration. In studies of congenic mice which differ only at the
Ah locus, the distribution patterns were similar to those previously
described,  except that relatively higher levels of 2,3,7,8-TCDD were
reported in the livers of the responsive mice compared with the
nonresponsive strain (Gasiewicz et al. 1983a,b; Birnbaum 1986). Thus, it
appears that there is little potential for strain difference in the
distribution pattern of 2,3,7,8-TCDD. Lakshmanan et al. (1986) reported
that the tissue distribution of 2,3,7,8-TCDD is mediated through
transport in the lymph system in the rat, with initial high levels of
2,3,7,8-TCDD in the lymph decreasing rapidly with progressive
accumulation in the fat during the first 24 h. The half-life for
redistribution of 2,3,7,8-TCDD out of adipose tissue and liver was 7.6
and 5.3 weeks,  respectively.

     2,3,7,8-TCDD crosses the placenta and accumulates in the mouse
fetus after gavage administration (Weber and Birnbaum 1985). In this
study,  -0.5% of the dose was detected in the fetus. Similar results were
reported by Krowke (1986) in mice treated with 2,3,7,8-TCDD by
subcutaneous injection. Fecal distribution was not uniform, with 75% of
the 2,3,7,8-TCDD located in the fetal liver. In addition to in utero
exposure of the fetuses, Nau et al. (1986) reported that postnatal
exposure of the young occurred via the milk after a single
administration of 2,3,7,8-TCDD to pregnant mice between gestation days
14 and 17.  Again, the fetal and pup liver was the predominant storage
site.

4.2.2.3  Dermal
     Human.  Studies of humans exposed to herbicides or other industrial
compounds known to be contaminated with 2,3,7,8-TCDD were discussed in
Sect. 4.2.2.2 (oral distribution) for easier comparison with the only
available human experimental data. Routes of exposure in these studies
are not clearly defined; the most likely route is probably dermal,
although oral and inhalation exposure are also likely to occur.

     Animal.  No studies are available.

-------
46   Section 4

4.2.3  Metabolism

     Pertinent data regarding the metabolism of 2,3,7,8-TCDD following
inhalation or dermal exposure of humans or animals, or following oral
exposure of humans, were not found in the available literature. However,
data regarding the metabolism of 2,3,7,8-TCDD following oral exposure of
animals are available.

     2,3,7,8-TCDD is relatively slow to metabolize, but once metabolites
are formed they are rapidly excreted through the urine and bile as
glucuronide and sulfate conjugates (Olson et al. 1980, 1983, as reviewed
in EPA 198Sa). Recently, however, Olson (1986) reported that up to 28%
of the radioactivity in the tissues of guinea pigs treated with labeled
2,3,7.8-TCDD was associated with metabolites, indicating that under the
conditions used, the metabolites of 2,3,7,8-TCDD were not eliminated as
rapidly as previously believed. Sawahata et al.  (1982) identified 1-
hydroxy-2,3.7.8-tetrachlorodibenzo-p-dioxin and 8-hydroxy-2,3,7-
trichlorodibenzo-p-dioxin formed by insulation of 2,3,7,8-TCDD with
isolated rat hepatocytes, and Poiger et al. (1982) identified 2-
hydroxy-l,3,7,8-tetrachlorodibenzo-p-dioxin in dogs as major metabolites
of 2,3,7,8-TCDD. Other hydroxylated products were identified as minor
metabolites. Mason and Safe (1986a,b) synthezised some of these
metabolites and demonstrated that they had considerably less biological
activity than 2,3,7,8-TCDD, which supports the observation by Weber et
al. (1982) and Poiger et al. (1982) that metabolites extracted from the
bile of dogs treated with 2,3,7,8-TCDD were less toxic to rats and
guinea pigs, respectively, than equivalent amounts of 2,3,7,8-TCDD.

     Species and strain differences in the metabolism of 2,3,7,8-TCDD
have been investigated to determine if differences in metabolism could
account for the large observed difference in toxicity. Gasiewicz et al.
(1983b) reported that qualitative evaluations of the elution profiles of
the urinary and biliary metabolites of 2,3,7,8-TCDD were similar for
responsive and nonresponsive strains of mice. In dogs (in vivo) and rats
(in vitro), pretreatment with 2,3,7,8-TCDD resulted in a 100 and 320%
increase, respectively, in the rate of metabolism of a subsequent dose
of 2,3,7,8-TCDD, whereas in guinea pigs (in vitro), pretreatment with
2,3,7,8-TCDD had no effect on the rate of metabolism of a subsequent
dose of 2,3,7,8-TCDD (Olson and Wroblewski 1985, Poiger and Schlatter
1985,  Wroblewski and Olson 1985). These data suggest that some of the
differences in the toxicity of 2,3,7,8-TCDD may be related to the rate
of metabolism as well as qualitative and quantitative differences in the
metabolites formed.

4.2.4  Excretion

     Pertinent data regarding the excretion of 2,3,7,8-TCDD following
inhalation or dermal exposure of humans or animals were not found in the
available literature. However, data regarding the excretion of 2,3,7,8-
TCDD following oral exposure of humans and animals are available.

4.2.4.1  Human

     Poiger and Schlatter (1986) studied the elimination profile of
2,3,7,8-TCDD from a volunteer who ingested a single 1.14-ng/kg dose of
the tritiated chemical. Urinary levels of radioactivity were never above

-------
                                                 lexicological Data   47

background levels during the 135 days the individual was studied. Fecal
elimination was initially rapid, with 11% of the dose eliminated in the
first 3 days (speculated to be nonabsorbed material), whereas during
days 7 to 125, only-3.5% of the dose was eliminated. From these data,  a
half-life of 2,120 days in the body was calculated assuming first-order
kinetics.
     CDC (1987) has also estimated the half-life for elimination of
2,3,7,8-TCDD. From participants in the Air Force Ranch Hand study, blood
samples were obtained in 1982 from 50 subjects selected as having high
exposure to 2,3,7,8-TCDD. Serum obtained from this blood was used to
determine 2,3,7,8-TCDD on a lipid-weight basis. Similar determinations
were made in 1987. The final test population consisted of 36 subjects
that had 2.3,7,8-TCDD levels of greater than 10 ppt  (considered the
background level) at both testing times. The decrease in serum 2,3,7,8-
TCDD levels during the interval time was used to calculate the half-
life. The median half-life calculated was 7.1 years. For 31 of the
individuals, the range of half-life values was between 2.9 and 26.9
years, while the calculated half-life for 1 individual was 743 years.
Values could not be calculated  for 4 individuals because the 1987
2,3,7,8-TCDD level was greater  than the 1982 value.  The variation in
half-life calculations were likely the result of random variation in the
measurements of serum 2,3,7,8-TCDD levels. Considering the difference  in
experimental design,  the values determined by CDC  (1987) are very
similar  to those previously reported by Poiger and Schlatter  (1986).

4.2.4.2  Animal
     In  laboratory  animals, 2,3,7,8-TCDD and  its metabolites  are
eliminated predominantly  in the feces and in  the urine in an  apparent
first-order process.  There appear  to be no major differences  whether  the
material was administered by  the oral or intraperitoneal routes  (EPA
1985a).  Although metabolites  were  observed  in the  bile, unmetabolized
2,3,7,8-TCDD detected in  the  feces probably was derived from  direct
intestinal elimination, as demonstrated by Olson  (1986) in  guinea pigs.
Nau et al.  (1986) reported that elimination of 2,3,7,8-TCDD through
lactation  is  significant  in mice.  EPA  (1985a)  reported  that between 91
and 99%  of  the  excreted  2,3,7,8-TCDD-derived  radioactivity  was found in
the feces of  rats and guinea  pigs;  54  and  72% was  detected  in the feces
of mice, with  greater fecal excretion  in  the  responsive C57B1/6J strain;
and 59%  was  observed in  the feces  of hamsters.  Half-lives  for
elimination  also  varied with  species and  strain as follows  (in
decreasing  order):  guinea pigs  (22 to  30  days);  rats (17  to 31 days);
mice  (11 to  24 days,  11  days  in responsive  mice and 24  days in
nonresponsive  strains);  and hamsters (10  to 15 days).  Olson (1986)
reported even longer half-lives of >90 days in guinea pigs. Although
there  are  no adequate excretion data from monkeys, there  are indications
that  2,3,7,8-TCDD is very persistent in this  species,  with a possible
half-life  of >1 year (EPA 1985a).

-------
48   Section 4

4.3  TOXICITY

4.3.1  Lethality and Decreased Longevity

4.3.1.1  Inhalation

     Pertinent data regarding lethality and decreased longevity
following  inhalation exposure of humans or animals to 2,3,7,8-TCDD were
not found  in the available literature.

4.3.1.2  Oral

     Human.  No studies are available.

     Animal.  Results of extensive investigations of the acute lethality
of 2,3,7,8-TCDD indicate that this chemical is toxic at very low levels
in all species tested. Species differences in lethality cover 4 orders
of magnitude in dose level. LDso values, after gavage administration in
lipophilic solvents, were reported to range from 0.6 to 2.1 MgAg in
Hartley guinea pigs (Schwetz et al. 1973), 20 to 60 MgAg in rats, 100
to 600 MgAg in mice, and 1,000 to 5,000 MgAg in hamsters (EPA 1985a,
McConnell  1985).  Rhesus monkeys have also been studied, but the lowest
dose tested, 70 MgAg. was lethal (McConnell et al. 1978). Reproductive
toxicity studies indicate that female rhesus monkeys are very sensitive
to the lethal effects of 2,3,7,8-TCDD. Eight of 16 pregnant monkeys died
after treatment with a total dose of 1.0 MgAg over a 9-day period
during gestation (McNulty 1985); toxic effects, as indicated by
abortion, were observed at a total 2,3,7,8-TCDD dose of 0.2 MgAg-
Following acute administration, death is generally observed only after
an extended period of time that ranges from 5 to 40 days; this lag time
appears to be, to some extent, independent of dose after a threshold is
reached.

     Other factors besides species differences affect the acute toxicity
of 2,3,7,8-TCDD. These variables include the strain of animal tested,
with responsive C57BL/6J mice demonstrated to be twice as sensitive to
2,3,7,8-TCDD-induced lethality as DBA/2J nonresponsive strains
(Gasiewicz et al. 1983a,b), and differences in lethal dose (ranging from
165 to 320 /*gAg) reported for four strains of rats (Walden and Schiller
1985).  Strain differences have not been investigated in other species
but would be anticipated to occur. Also, the vehicle substantially
affects the acute lethality of 2,3,7,8-TCDD, probably by altering the
bioavailability of the compound. However, as discussed by Kaminsky et
al. (1985) and illustrated by Umbreit el al. (1985, 1986a,b), the
influence of matrix on bioavailability is complex, with factors such as
the properties of the matrix as well as duration of contact with the
matrix substantially altering the bioavailability. The complexity of
this issue is further illustrated by the protective effect of orally
administered activated charcoal on the lethality of subcutaneously or
intraperitoneally administered 2,3.7,8-TCDD (Manara et al. 1984),
suggesting that some of the effects are the result of processes other
than bioavailability.

     The lethal effects of subchronic exposure to 2,3,7,8-TCDD have
been studied by DeCaprio et al. (1986) in Hartley guinea pigs. Guinea
pigs were maintained on diets containing 2, 10, 76, or 430 ppt of

-------
                                                 lexicological Data   49

2,3,7,8-TCDD for 90 days. At 76 and 430 ppt, mortalities of 10 and 70%,
respectively, were observed, and an LD50 value of 0.8 pgAg for the
total consumption of 2,3,7,8-TCDD was calculated. This value is similar
to that observed -in the previously described study of the acute exposure
of male guinea pigs to 2,3,7,8-TCDD. Exposures of 0.61 ngAg/day
resulted in no deaths. In rhesus monkeys, however, Allen et al. (1977)
observed deaths in 5 of 8 animals maintained for 9 months on a diet that
contained 500 ppt of 2,3,7,8-TCDD. From food intake, the total dose
ingested was calculated to be 2 to 3 pgAg or 0.0093 to 0.014 MgAg/d*y.
4.3.1.3  Dermal
     Human.  No documented death has resulted from the acute systemic
toxicity of 2,3,7,8-TCDD following accidental exposure of humans.
     Animal.  There is only one study available regarding the lethality
of 2,3,7,8-TCDD by the dermal route. Schwetz et al. (1973) determined
that the acute LD50 of 2,3,7,8-TCDD in acetone was 275 /*gAg (range, 142
to 531 pgAg) in New Zealand white rabbits. As was observed after oral
administration, the time to death was protracted, with deaths observed
between 12 and 22 days after treatment.

4.3.2  Systemic/Target Organ Tozicity

4.3.2.1  Chloracne
     Inhalation.  Pertinent studies regarding the development of
chloracne by humans or animals following inhalation exposure were not
found in che available literature.
     Oral, human.  No studies are available.
     Oral, animal.  Greig  (1984) observed  typical skin lesions in male
and female hairless A2G-hr/+ mice 4 weeks  after a single  gavage
administration of 2,3,7,8-TCDD at 75 /igAg- The females appeared to be
more severely affected than the males. McConnell et al. (1978) observed
acne form eruptions in rhesus monkeys given a single dose  of 2,3,7,8-TCDD
at 70 MgAg- This dose was severely toxic, resulting  in many gross
effects and ultimately death. Longer-term  exposure of monkeys to
2,3,7,8-TCDD in the diet at 500 ppt (0.0094 to 0.014  pgAg/day) f°r
9 months also produced lesions that resembled chloracne (Allen et al.
1977). This dose was not a threshold for the formation of chloracne,
since the  lesions were severe and associated with  other dermal effects
such as subcutaneous edema and loss of facial hair. In addition, this
level of 2,3,7,8-TCDD resulted in the  deaths of  5  of  8 animals tested.
     Dermal, human.  Since the turn of the century, chloracne has been
observed in humans after a few days from the time  of  accidental exposure
to a variety of chlorinated aromatic compounds.  The prevalence of these
lesions has been reviewed  by Taylor  (1979) and Suskind  (1985). 2,3,7,8-
TCDD is known  to be one of the most potent compounds  in producing
chloracne; however, sufficient data on exposure  are not available to
define  the doses necessary to produce  this lesion. The  lesions usually
appear  on  the  face and upper  trunk  area. These lesions  can be very
disfiguring and can be persistent,  lasting many  years after exposure.
Mild cases of  chloracne generally result  in  no permanent  disfigurement

-------
50   Section 4

(Moses and Prioleau 1985). After the Seveso accident, children appeared
to develop chloracne more frequently than adults, but this may have beer.
related to greater exposure to contaminated soils during play rather
than to a greater sensitivity in the young (Suskind 1985, Taylor 1979).
     Other signs of toxicity have been reported in case studies of small
groups of people exposed to 2,3,7,8-TCDD.  These signs include aching
muscles, loss of appetite, weight loss, digestive disorders, easy
fatigability, insomnia, loss of libido, headache, neuropathy, sleep
disturbance, sensory changes, and uncharacteristic bouts of anger (Bauer
et al. 1961, Kimmig and Schulz 1957, Oliver 1975, Poland et al. 1971,
Schulz 1957). Many of these symptoms are commonly observed with acute
exposure to chemicals, and since exposure to 2,3,7,8-TCDD is a mixed
exposure, with 2,3,7,8-TCDD being only a minor component (on a
percentage basis),  it is difficult to state with certainty that these
symptoms are produced from 2,3,7,8-TCDD. The other chemicals that
provide concomitant exposure include not only the chlorinated compound
of which 2,3,7,8-TCDD is a contaminant, but, in many cases (such as with
herbicide use), petroleum solvents and other "inert ingredients" used in
the formulation of the final product. Epidemiologic studies have failed
to demonstrate an association between 2,3,7,8-TCDD exposure and any of
the above effects;  however, the epidemiclogic studies have many
limitations and may not have had the statistical power to detect any
effects that did not occur in a large segment of the exposed population.
     Dermal, animal.  Puhvel et al.  (1982) applied 0.1 /ig of 2,3,7,8-
TCDD to the skin of hairless Skh:HR-l mice 3 times/week for 4 weeks. Th
mice developed skin lesions which appeared on histologic examination to
resemble chloracne. Changes in the skin included hyperkeratinization,
hyperplasia, absences of sebaceous glands, and buildup of keratin into
dermal cysts. 2,3,7,8-TCDD was the most effective agent of the seven
tested in inducing this dermal response, with the next most effective
compound, 3,4,3',4'-tetrachlorobiphenyl, inducing a response at 200 pg
per application. The use of a single dose in this study precludes the
determination of a threshold dose.

     Toth et al. (1979) administered 2,3,7,8-TCDD to Swiss mice by
gavage for 1 year at doses of 0, 0.007, 0.7, or 7.0 pgAg/veek.
Amyloidosis was observed in the kidney, spleen, and liver, along with
dermatitis, in all treatment groups. The dermatitis had some
similarities to chloracne, although it is not clear if the etiology is
the same. If this dermatitis is similar to chloracne, then the LOAEL in
mice would be 0.007 ^g/kg/week.

     General discussion.  Chloracne is a persistent deformative skin
lesion that can be induced by single or multiple exposure to 2,3,7,8-
TCDD in humans. Chloracne is the first toxic effect usually observed in
humans exposed to chemicals contaminated with 2,3,7,8-TCDD and appears
to be a sensitive toxicological end point. It is believed that humans
can develop chloracne following exposure to 2,3.7,8-TCDD by any route.
but data are not available regarding the dose necessary to induce
chloracne in humans.

     Moses and Prioleau (1985) studied humans who had recovered from
chloracne and determined that histologic examination of the skin was
incapable of providing any indication of past exposure to 2,3,7,8-TCDD.
r

-------
                                                 lexicological Data   51

Only a few experimental animals develop chloracne upon exposure to
compounds that produce this lesion in humans; the limitation of
appropriate animal models has resulted in a poor understanding of this
lesion and little information on dose-response relationships. In vitro
studies by Greenlee et al. (1984) using human epidermal cultures
suggested that 2,3,7,8-TCDD may affect the receptors for epidermal
growth factors, which ultimately results in the typical dermal lesions
associated with 2,3,7,8-TCDD exposure.

4.3.2.2  Wasting syndrome
     Inhalation.  Pertinent data regarding wasting syndrome in humans or
animals following inhalation exposure to 2,3,7,8-TCDD were not found in
the available literature.
     Oral, human.  No studies are available.
     Oral, animal.  The wasting syndrome has been observed in all
species tested after administration of acute lethal doses of 2,3,7,8-
TCDD (EPA 1985a). As described in the above review, body weight will
decrease (or the rate of gain will be severely limited) after
administration of 2,3,7,8-TCDD, possibly in a biphasic pattern, until
death occurs 15 to 30 days after exposure. These changes in body weight
may be observed within 24 hours of administration of 2,3,7,8-TCDD
(Peterson et al. 1984). Studies with pair-fed rats suggest that the
wasting syndrome results from 2,3,7,8-TCDD-induced appetite suppression
rather than malabsorption or altered food energy utilization  (Kellin et
al, 1985; Potter et al. 1986; Rozman 1984; Seefeld et al. 1984a.b;
Seefeld and Peterson 1984). Recently, however, Lu et al. (1986)
demonstrated that total parenteral nutrition only partially protected
Hartley guinea pigs from loss of weight. Treated guinea pigs maintained
relatively stable body weight until a few days prior to death,
demonstrating that decreased food consumption accounts for most of  the
observed loss in body weight but that other mechanisms must  account  for
the final decrease in weight and ultimate death. Additional  studies  in
cold-adapted Sprague-Dawley rats, which maintain high levels  of food
intake after treatment with 2,3,7,8-TCDD, support  the observation that
decreased food consumption only partially accounts  for the wasting
syndrome, since the cold-adapted rats lost weight  twice as fast as  rats
treated at normal temperatures  (Rozman and Greim 1986).
     Rhesus monkeys and Hartley guinea pigs  also lose body weight after
prolonged exposure to diets containing 2,3,7,8-TCDD (Allen et al.  1977,
DeCaprio et al. 1986). Weight  loss was -20%  in  female monkeys ingesting
2,3.7,8-TCDD at -0.01 ng/kg/day  (the  only dose  level  studied)  for 9
months. Only eight monkeys were  tested and  five  died  at  this dose.
Guinea pigs  (10 males and 10  females) maintained for  90  days on diets
that provided  doses of 2,3,7,8-TCDD  of 0.0049 MgAg/day  *»*  a 15%
decrease  in body weight,  whereas guinea  pigs given doses  of  0.026
MgAg/day had  a 40% decrease  in body weight.  All  guinea pigs in the
high-dose group died or  were  killed  (when moribund) before  the end of
the study. No  effects were  observed  at  the  0.00061-MgAg/day level. It
appears  that even in long-term studies,  the severe loss  of body weight,
characteristic of exposure  to 2,3,7,8-TCDD,  Is  associated with dose
levels  that  are  lethal  to the animal.

-------
52   Section 4

     Dermal, human.  As discussed In the previous section on chloracne ,
case reports have indicated that weight loss was observed in some
individuals following acute exposure to 2 , 3 , 7 ,8-TCDD. The data are
insufficient to conclude that this reported weight loss was the result
of 2,3,7,8-TCDD exposure or that it is similar to the wasting syndrome
reported in laboratory animals. Epidemiologic studies have not detected
a condition similar to the wasting syndrome in populations with
potential exposure to 2,3,7,8-TCDD.

     Dermal, animal.  Pertinent data regarding the wasting syndrome in
animals exposed dermally to 2,3,7,8-TCDD were not found in the available
literature.

     General discussion.  The wasting syndrome is a characteristic sign
of 2,3,7,8-TCDD toxicity in experimental animals. This syndrome is
observed in both acute and longer-term studies and is most commonly
associated with lethal doses. The wasting syndrome has not been observed
in humans.

     Since the mechanism is not clearly understood, it remains to be
determined whether the severe body weight loss associated with 2,3,7,8-
TCDD toxicity is the cause of the subsequent death of the animal or an
effect that can be segregated from lethality. Mechanisms have been
proposed by Aust (1984) that suggest that the effect of 2,3,7,8-TCDD on
the thyroid results in activation of thyrotropin-releasing hormone,
which has an anorectic action, and, in conjunction with 2, 3, 7,8-TCDD-
induced vitamin A depletion, results in loss of body weight. Regardless
of the mechanism or length of exposure, the wasting syndrome appears tc
be an indicator of impending death rather than an early sign of
toxicity. No reports of abnormal weight change as a result of 2,3,7,8-
TCDD exposure in humans were found.

4.3.2.3  Hepatic effects

     Inhalation.  Pertinent data regarding hepatotoxicity in humans or
animals exposed by inhalation to 2,3,7,8-TCDD were not found in the
available literature.

     Oral, human.  No studies are available.

     Oral, animal.  The acute administration of 2,3,7,8-TCDD to rats and
mice results in toxic effects in the liver, whereas no severe changes in
the liver are observed upon acute administration of 2,3,7,8-TCDD to
guinea pigs and monkeys (EPA 198Sa) .  Lesions in rodents consist of
necrosis, proliferative changes, cellular membrane alterations, bile
duct proliferation, altered lipid metabolism, and excess amounts of
porphyrin. These liver effects have generally been observed following a
single exposure at relatively high doses of 5 to 200
     Turner and Collins (1983), however, did observe morphologic changes
in a small number of guinea pigs (groups of one male and four to six
females, strain not reported) given a single gavage dose of 2,3,7,8-TCDD
at 0.1, 0.5, 2.5, 12.5, or 20 Mg/kg- Changes including hypertrophy,
steatosis, focal necrosis, and hyalin-like bodies were reported in all
guinea pigs, although no indication of an association between dose and
increased severity or incidence was reported. Deaths were reported for
doses >0.5 and 12.5 jig/kg in males and females, respectively. In a

-------
                                                 Toxicological Data   S3

90-day feeding study with Hartley guinea pigs, similar mild liver
changes were observed in animals maintained on diets that provided doses
of 0.0049 /JgAg/day; liver changes were not observed at the next lower
dose of 0.00061 A*gAg/day (DeCaprio et al. 1986). Although chronic
studies are not available in guinea pigs, studies reviewed by EPA
(1985a) in rats (Kociba et al. 1979, NTP 1982a) and mice (NTP 1982a)
indicated that doses of -0.001 MgAg/day were LOAELs for liver effects
due to chronic oral exposure to 2,3,7,8-TCDD.
     Dermal, human.  Studies of the hepatotoxic effects of 2,3,7,8-TCDD
on humans have been conducted on populations exposed to herbicides and
other industrial chemicals contaminated with 2,3,7,8-TCDD. In these
studies, it is considered that the predominant route of exposure is
dermal, although some exposure by the oral and inhalation routes would
also be expected. In addition, all individuals studied were exposed to
multiple chemicals, which may have contributed to or been the cause of
the effects observed.
     Reports reviewed by EPA (1985a), May (1973), Holden (1979), Bogen
(1979), and Holmstedt (1980) have reported "liver dysfunction" as one of
the symptoms in workers and Vietnam veterans exposed to the herbicide
2,4,5-T contaminated with 2,3,7,8-TCDD. Since these were essentially
case reports, no firm association between exposure and effect could be
made. A more complete clinical study of the levels of serum liver enzyme
was conducted in children ages 6 to 10 who were potentially exposed to
2,3,7,8-TCDD in the Seveso accident (Mocarelli et al. 1986). The
children were examined yearly for 6 years following the accident. During
the initial 2 years, elevated serum levels of gamma-glutamyltransferase
(GGT) and alanine aminotransferase (ALT) were observed in both boys and
girls who resided in the most highly contaminated area. The increase was
slight, with a total incidence of 4.3% compared  to 3.6% in the control
group. Values returned to normal after 2 years.  Ideo et al. (1982)
reported elevated urinary D-glucaric acid in  the urine of children in
the Seveso area -2.5 years after the accident. They also noted that
there was a strong  correlation between glucaric  acid levels and the
activity of hepatic microsomal enzymes. However, no liver effects, as
indicated by serum  enzyme levels, were observed  by Falk et al.  (1984)  in
a pilot epidemiologic study of 122 persons exposed in Missouri to
chemicals contaminated with 2,3,7,8-TCDD.
     Liver involvement, as indicated by porphyria cutanea tarda, has
also been reported  to be associated with  2,3,7,8-TCDD exposure. The
major studies have  been reviewed by Jones and Chelsky  (1986), who
reported that in all cases the porphyria  cutanea tarda may not have been
definitively diagnosed or that the other  chemicals to which subjects
were exposed may have been the causative  agents. The pilot epidemiology
study by Falk et al. (1984) of the Missouri  accident failed to  detect
porphyria cutanea  tarda in a  population with known exposure.  The  results
of  Falk et al.  (1984) may be  confounded by misidentifying exposed
individuals. Patterson et al.  (in press)  explained  the wide variation in
2,3,7,8-TCDD levels in adipose tissues from  16 Missouri  residents who
reported exposure  to 2,3,7,8-TCDD in a questionnaire by  determining
through independent sources that  exposure at the site  in question was
unlikely in  the  individuals in whom measured adipose  tissue  levels of
2,3,7,8-TCDD were  low.

-------
54   Section 4

     Dermal, animal.  No studies are available.

     General discussion.  Hepatotoxic effects have been investigated iu
human populations exposed to chemicals contaminated with 2,3,7,8-TCDD
because of the known hepatotoxicity of 2,3,7,8-TCDD in rodents. Results
of these studies have generally been inconsistent with regard to the
detection of altered liver serum enzymes or porphyria cutanea tarda.
These inconsistencies may be related to the populations studied, the
level of contamination, or the bioavailability of the 2,3,7,8-TCDD under
the specific conditions of the contaminated site. These confounding
factors, along with the ubiquitous nature of and causative agents for
slight changes in liver function, have made confirmation of 2,3,7,8-
TCDD- induced adverse effects on the liver in humans impossible. In rats
and mice, 2,3,7,8-TCDD clearly produces adverse effects on the liver;
however, there are no studies available that investigated the dose-
response relationship. Thus, it is not known if initial changes in the
liver are a first toxic manifestation of 2,3,7,8-TCDD exposure or an
effect observed only after other toxic effects in other organ systems
have been manifested.

     Although Turner and Collins (1983) reported mild liver effects in
guinea pigs (a species generally resistant to 2,3,7,8-TCDD-induced liver
damage) at doses as low as 0.1 pgAg, the lack of association between
increasing dose and increasing severity of effect in this study makes it
difficult to determine a threshold dose. Observing similar liver changes
in a subchronic study in guinea pigs and in chronic studies in rats and
mice, however, supports the observation of the acute study that
hepatotoxicity is a sensitive indicator of 2,3,7,8-TCDD toxicity.

     Additionally, induction of hepatic microsomal enzymes has often
been associated with exposure of laboratory animals to 2,3,7,8-TCDD.
Early studies by Buu-Hoi et al. (1972) suggested that the toxicity of
2,3,7,8-TCDD was related to altered enzyme function. Later studies,
however, have demonstrated that 2,3,7,8-TCDD toxicity appears in many
cases to segregate with induction of enzyme activity rather than be the
cause of the toxic response. This has been demonstrated for DT-
diaphorase (Beatty and Neal 1976) and for mixed function oxidase (Greig
1979, Kociba and Schwetz 1982, Poland et al. 1979). In addition, the
toxicity of structurally related chlorinated dibenzo-p-dioxins
correlates well with the relative ability of these compounds to induce
enzyme activity (Poland and Glover 1973).

4.3.2.4  Immunotoxic ity

     Inhalation.  Pertinent data regarding immunotoxic effects of
2,3,7,8-TCDD following inhalation exposure of humans or animals were not
found in the available literature.

     Oral, human.  No studies are available.

     Oral, animal.  2.3,7,8-TCDD has been extensively investigated for
immunotoxicity in mice, rats, and guinea pigs, and as indicated in the
reviews by EPA (1985a, 1988b) and Knutsen (1984), this is a sensitive
end point of toxicity. Most studies were conducted with weekly exposures
for durations of between 4 and 8 weeks, and minimum effective doses
ranged from 1 pg/kg/week for mice to 5 pg/kg/week for rats, and

-------
                                                 lexicological Data   55

0.04 /jgAg/week guinea pigs. As the study by Vos et al. (1973)
indicates, the Hartley guinea pig may be the most sensitive species,
with a LOAEL of 0.04 /igAg/day and a NOAEL of 0.008 pg/kg/week. In
addition to species differences, strain differences in sensitivity to
the immunotoxic effects of 2,3,7,8-TCDD have been observed in mice, with
immunotoxic sensitivity segregating with the Ah locus. Segregation of
immunotoxicity has also been demonstrated in fetal thymus organ cultures
in vitro (Dencker et al. 1985). Thymus cultures from C57B1/6 mice, which
are Ah responsive, were very sensitive to the toxicity of 2,3,7,8-TCDD
(EC5Q of 10"10 Af) compared with no observed effects on thymus cultures
from the nonresponsive DBA/2J mouse at 3 x 10"8 M, the highest
concentration tested. Effects of 2,3,7,8-TCDD on the immune system
included decreases in thymus weight, sensitization to antigens
(bacterial antigens, skin grafts, and tumor cell development), serum
immunoglobins, and B but not T lymphocytes. Thigpen et al. (1975)
demonstrated that C57BL/6JFh mice that received as little as 1 pg/kg of
2,3,7,8-TCDD once a week for 4 weeks were more susceptible to death from
subsequent bacterial infection. This dose of 2,3,7,8-TCDD did not  result
in any gross signs of toxicity, suggesting that host susceptibility was
an early effect of 2,3,7,8-TCDD exposure. Immunotoxic effects have also
been reported in pups of Fischer rats and B6C3F1 mice following in utero
exposure and postnatal exposure through lactation  (Luster et al.  1982).
The effective doses on a maternal weight basis were approximately the
same as those that produced effects in adults. The study by Greenlee et
al. (1985), using thymic epithelium cell cultures, provides evidence
that 2,3,7,8-TCDD acts directly on the epithelium  cells by inhibiting
the maturation of the thyaocytes. Longer-term studies of  the effect of
2,3,7,8-TCDD on the immune system are not available.
     Dermal, human.  There  is  little  information on the  immunotoxic
effects of 2,3,7,8-TCDD  in humans exposed to herbicides  or other
chemicals contaminated with 2,3,7,8-TCDD. In a pilot  epidemiologic study
of 82 high-risk and 40  low-risk subjects from areas in Missouri where
2,3,7,8-TCDD exposure occurred, Stehr et al.  (1986) failed to  detect any
signs of  immunotoxicity.  In a  study of  154  exposed and 155 unexposed
persons in Gray Summit,  Missouri, Hoffman et al.  (1986)  reported  that
the exposed group had an increased  frequency of anergy (11.8%  vs  1.1%)
and relative anergy  (35.3% vs  11.8%). The exposed group  also  had  non-
statistically significant increased frequencies of abnormal T-cell
subset test results  (10.4%  vs  6.8%),  a  T4/T8  ratio of <1.0  (8.1%  vs
6.4%), and an abnormality in  the  functional T-cell test  results  (12.6%
vs 8.5%). Although  the  effects have not resulted  in  an excess of
clinical  illness  in the exposed group,  these  data suggest that exposure
to 2,3,7,8-TCDD may be  associated with  depressed  cell-mediated immunity.
In a  recent, more comprehensive review  of  these data, Knutsen et al.
(1987) indicated  that  confounding factors  in the  study design may have
affected  the  results.  The readers of the immunologic test results were
inconsistent with regard to recall antigens,  and psychological stress
may also  have  affected the results.  It  was concluded that further
studies were  necessary to confirm any effect of 2,3,7,8-TCDD on the
Immune system. Retesting of the subjects who were reported to be anergic
or  relatively anergic in the Hoffman et al. (1986) study revealed that
none  of  the  subjects were anergic and only one exposed and one
nonexposed subjects were relatively anergic (Evans et al. 1988).

-------
56   Section 6

     Dermal, animal.  No studies are available.

     General discussion.  There are abundant animal data that indicate
that immunetoxicity may be one of the most sensitive toxicologic end
points for 2,3,7,8-TCDD. There have been very limited investigations of
this end point in humans; thus, the importance of 2,3,7,8-TCDD-induced
immunotoxicity in humans cannot be evaluated. The time of onset, the
duration of the altered immune response, and the fact that evaluation of
altered immune response is a specialized analysis not routinely
performed in clinical settings will make the assessment of this end
point in humans difficult.

     The animal data show that 2,3,7,8-TCDD produces immunologic effects
in a number of species, and most of the studies have been concerned with
the investigation of alterations induced in the immune system. There are
less data available regarding dose-response relationships and species
and strain differences in sensitivity; these data would be of assistance
in evaluating response with regard to human health. Also, no data are
available on the immunotoxicity of 2,3,7,8-TCDD in monkeys, a species
that appears to be very sensitive to other toxic effects of 2,3,7,8-
TCDD.

4.3.3  Developmental Toxicity

4.3.3.1  Inhalation

     Pertinent data regarding the developmental toxicity of 2,3,7,8-TCDD
following inhalation exposure of humans or animals were not found in th
available literature.

4.3.3.2  Oral

     Human.  No studies are available.

     Animal.  2,3,7,8-TCDD has been extensively studied for
developmental toxicity, and these studies, reviewed in EPA (1985a,
1988b), indicate that 2,3,7,8-TCDD is teratogenic in mice and rats after
gavage administration. In mice, the most commonly observed developmental
effects were hydronephrotic kidney and cleft palate (Courtney 1976,
Moore et al. 1973, Neubert and Dillmann 1972, Smith et al. 1976). Both
of these anomalies have been observed at doses as low as 1 jig/kg
(Courtney 1976, Smith et al. 1976), with the kidney effects observed
after only a single exposure on day 10 of gestation.  In rats, gavage
administration of 2,3,7,8-TCDD during organogenesis at doses of -0.12S
to 0.25 pg/kg produced hemorrhage of internal organs and subcutaneous
edema (Sparschu et al. 1971a,b; Khera and Ruddick 1973).  No effects were
observed at 0.3 and 0.03 pgAg. respectively, in NMRI mice and Sprague-
Dawley rats. These teratogenic effects have been confirmed by a number
of studies in which 2.3,7,8-TCDD was administered subcutaneously
(studies summarized in EPA 1985b).

     It has been demonstrated that both genetic susceptibility and
concomitant exposure to other compounds affect the developmental
toxicity of 2,3,7,8-TCDD. Poland and Glover (1980) and Dencker and Pratt-
(1981) demonstrated genetic differences in the susceptibility of mice;
only responsive C57B1/6J mice developed the characteristic cleft palate

-------
                                                 Toxicological Data   57

and hydronephrotic kidneys after treatment. This indicates that
developmental toxicity, in addition to many other toxicological end
points of 2,3,7,8-TCDD, segregates with the Ah locus. Additionally, it
was demonstrated th'at simultaneous exposure to 2,3,7,8-TCDD and specific
polychlorinated biphenyls (Birnbaum et al. 1985), or to the hormones
hydrocortisone (Birnbaum et al. 1986) or thyroxine and triiodothyroxine
(Lamb et al. 1986), increases the sensitivity of mice to the
developmental effects of 2,3,7,8-TCDD. An additive effect was observed
by Weber et al. (1985) for simultaneous exposure to 2,3,7,8-TCDD and
2,3,7,8-tetrachlorodibenzofurans (TCDF).

4.3.3.3  Dermal
     Human.  The EPA (1985a, 1988b) evaluated several epidemiology
studies of human populations exposed to herbicides contaminated with
2,3,7,8-TCDD. It is assumed that the exposure in these studies was
predominantly dermal; however, some oral and inhalation exposure was
also likely. The studies reviewed were predominantly geographic
correlation studies that reported elevated incidence of birth defects,
including cleft palate, cleft lip, neural-tube defects, heart
abnormalities, hypospadias and epispadias, talipes, and cystic kidney
disease, as well as increases in stillbirths (EPA 1979a, Field and Kerr
1979, Hanify et al. 1981). Similar studies reviewed failed to
demonstrate a correlation between birth defects and possible exposure to
2,3,7,8-TCDD (Aldred 1978; Bisanti et al. 1980; Bonaccorsi et al. 1978;
Department of Health, New Zealand 1980; McQueen et al. 1977; Nelson et
al. 1979; Reggiani 1980; Smith et al. 1982; Thomas 1980). In addition, a
case control study of the offspring of Vietnam veterans potentially
exposed to 2,3,7,8-TCDD in Agent Orange detected increases in birth
defects that included spina bifida, cleft palate, and certain congenital
tumors (Erickson et al. 1984). When all types of defects were combined,
there was no elevated risk, and authors noted that the seemingly higher
risk for individual birth defects may have resulted from chance or other
unidentified risk factors. In a similar study, Townsend et al. (1982)
investigated the pregnancy outcome of wives of employees of Dow Chemical
Company who were potentially exposed  to 2,3,7,8-TCDD. When compared with
a control population, there was no statistical difference in pregnancy
outcome or several types of malformations. As a  result of the inherent
uncertainty in interpreting geographic correlation studies, the
concomitant exposure to other potentially active compounds, and the
review of similar studies that failed to  demonstrate any association,  it
was concluded that these investigations could neither prove nor disprove
the hypothesis that  2,3,7,8-TCDD was  a  teratogen in humans.

     Animal.  No studies are available.

4.3.3.4  General discussion
     The lack of a clearly defined exposed population, with adequate
quantitative data on levels and duration  of exposure,  and  the
confounding presence of exposure  to  other chemicals  that may  in
themselves be developmental toxicants,  have made results of epidemiology
data difficult to evaluate and inadequate for determining whether
2,3,7,8-TCDD  induces developmental defects in humans.  As with many
developmental  toxicants,  the  animal  data  indicated that  the  time  of

-------
58   Section 4

exposure was critical for Che induction of effects,  with treatment on
days 8 or 11 producing maximal effects and treatment after 13 days being
ineffective in mice (Neubert et al. 1973).
                 •   r
4.3.4  Reproductive Tozicity

4.3.4.1  Inhalation

     Pertinent data regarding the reproductive toxicity of 2,3,7,8-TCDD
in humans or animals following inhalation exposure were not found in the
available literature.

4.3.4.2  Oral

     Human.  No studies are available.

     Animal.  The fetotoxicity of 2,3,7,8-TCDD has been demonstrated
following short-term exposure in utero (rats and mice) as well as in a
multigeneration study (rats). Fetal death and vaginal bleeding have been
observed in developmental toxicity studies [reviewed by EPA (1985a,
1988b)] at doses of between 2 and 9 jig/kg/day. The three-generation
study of Murray et al. (1979) reported that dietary administration of
2,3,7,8-TCDD at 0.01 and 0.1 /igAg/day resulted in adverse effects on
litter size, fetal survival, and neonatal survival in Sprague-Dawley
rats. The 0.1-/ig/kg/day dose also resulted in a significant decrease in
fertility. Murray et al. (1979) considered the low dose of 0.001
/*gAg/day to be without substantial effects, since the only effect
observed was an increase in dilated renal pelvises in the Fl generation
This effect was not observed at statistically significant levels in any
other generation. The absence of effects at this lower dose level has
been questioned by Nisbet and Paxton (1982), who reevaluated the data
statistically and concluded that gestational index,  decreased fetal
weight, and the incidence of dilated renal pelvis were all increased at
both the 0.01- and 0.001-pg/kg/day doses. The analysis by Nisbet and
Paxton (1982) indicates that reproductive performance and the fetus are
very sensitive to the toxic properties of 2,3,7,8-TCDD.

     In this reevaluation, Nisbet and Paxton (1982)  applied a
statistical approach that included pooling of data across all
generations. However, since litters from different generations (as from
subsequent mating within a generation) are not the same, they have
different histories of exposure and each is tied to the effect of the
agent on its parental generation. Therefore, EPA (1988b) considered such
an approach of pooling of data across all generations to be biologically
inappropriate. According to EPA (1988b), the 0.01 pgAg/day dose in the
study by Murray et al. (1979) is the lowest effect level that can be
supported by the data. Further analysis of this study and of studies in
monkeys (Allen et al. 1979, Schantz et al. 1979), which indicate that
doses of 0.0015 /ig/kg/day result in abortions, may provide support for a
lower effect level of 0.001 pg/kg/day. Therefore, the 0.001 pgAg/day
dose, rather than the 0.01 pgAg/day dose, appears to be the LOAEL.

     The monkey, however, appears more sensitive to the toxicity of
2.3,7,8-TCDD than either rats or mice. McNulty (1984, 1985) described
the common occurrence of abortion  in rhesus monkeys treated with a to to.
of 1 MgAg of 2,3,7,8-TCDD over days 20 to 40 of gestation; the lowest

-------
                                                 lexicological Data   59

total dose tested, 0.2 /*g/kg, produced abortion in 1 of 4 test monkeys.
In fetuses examined, there was either only minimal indication (palatal
abnormalities) or no indication of developmental toxicity.  In an earlier
feeding study, groups of eight monkeys were maintained on diets
containing 50 or 500 ppt (the total dose ingested was 1.8 and 11.7 ^g,
respectively) of 2,3,7,8-TCDD for 7 months prior to pregnancy and during
pregnancy (Allen et al. 1979, Schantz et al. 1979). In both groups,
two-thirds of the pregnancies ended in abortion.
     As reported in EPA (1985a), chronic exposure of rats to 2,3,7,8-
TCDD in the diet results in a decrease in the weight of male
reproductive organs. It was suggested by Moore et al. (1985) that this
effect on the reproductive organs might be the result of decreases in
the plasma concentrations of androgens, which may account for the
reduced male Uistar rat reproductive performance following 2,3,7,8-TCDD
exposure that had been earlier observed by Khera and Ruddick (1973).
Levels of circulating androgens in 2,3,7,8-TCDD-treated male rats were
studied by Moore et al. (1985) and shown to be depressed as a result of
treatment. It was speculated that these depressed hormone levels may
participate in the reproductive dysfunction in males. Circulating
estradiol levels in pregnant females, however, appear not to be affected
by 2,3,7,8-TCDD exposure, although 2,3,7,8-TCDD does affect some
estrogen-metabolizing pathways when studied in vitro (Shiverick and
Muther 1983).

4.3.4.3  Dermal
     Human.  Epidemiology studies have been performed on groups of
individuals exposed to herbicides or industrial chemicals contaminated
with 2,3,7,8-TCDD. Although the dermal route  is considered  to be the
predominant route of exposure in these studies, some exposure by other
routes would also occur. These studies, which include those reviewed by
EPA  (1985a, 1988b) and Friedman (1984) (Aldred  1978; Bisanti et al.
1980; Bonaccorsi et al. 1978; Department of Health, New Zealand 1980;
EPA  1979a; Field and Kerr 1979; Hanify et  al. 1981; McQueen et al  1977;
Nelson et al. 1979; Reggiani 1980; Smith et al. 1982; Thomas  1980;
Tognoni and Bonaccorsi 1982; Townsend et al.  1982)  and that of Forsberg
and  Nordstrom (1985), were conducted in groups  of  male herbicide
application workers, chemical plant employees,  and soldiers exposed to
Agent Orange  in Vietnam, and to both males and  females exposed through
major industrial accidents or inappropriate disposal, which permitted
the  escape of large amounts of  chemical from  production plants. Studies
of these  groups have not clearly demonstrated that 2,3,7,8-TCDD produced
any  adverse effects on reproductive performance,  although  as  a result of
the  limitations of  the study, particularly with regard  to  the extent  of
exposure,  it  is not possible  to interpret  the results as  indicative of
the  absence of reproductive  toxicity for  2,3,7,8-TCDD  in humans.

     Animal.  No  studies are  available.

4.3.4.4   General  discussion
     The  evidence  that 2,3,7,8-TCDD  is a  reproductive  toxicant in humans
is limited by the  lack of  exposure data and the concomitant exposure  to
other chemicals  that  may have been biologically active.  The greatest

-------
60   Section 4

exposure to 2,3,7,8-TCDD occurred in male herbicide sprayers, soldiers,
and chemical plane workers, whereas the studies conducted have been
concerned mostly with fetotoxicity and spontaneous abortion. Although
the possibility of premating exposure in males resulting in fetotoxicity
and abortion is of concern, this end point is difficult to assess,
particularly when the exposure is temporally removed from the time of
conception. In addition, there is a lack of data on the functioning of
male reproductive organs during the time of potential exposure to
2,3,7,8-TCDD. There are no equivalent female populations studied that
have had extended periods of high exposure to chemicals contaminated
with 2,3,7,8-TCDD and only limited populations [females in the vicinity
of Seveso (Bisanti et al. 1980, Bonaccorsi et al. 1978, Reggiani 1980)
in the period immediately after the accident] from which to assess
effects on reproduction in females exposed while pregnant.

     Given the above limitations, the present epidemiology studies are
only consistent with observations in animals that 2,3,7,8-TCDD elicits
adverse effects on reproduction; they do not provide sufficient evidence
to prove that this chemical is a reproductive toxicant. Animal studies
indicate that 2,3,7,8-TCDD is a powerful reproductive toxicant; however,
differences in species sensitivity have been observed. The available
data indicate that the monkey may be the most sensitive species, but
tests in this species have been limited to rhesus monkeys, with only a
few animals studied at any dose level, which provides insufficient data
to fully evaluate the range and extent of potential toxic effects in
this species. In addition, the sensitivity of humans compared to
monkeys, rats, or mice can only be speculated.

4.3.5  Genotozic1ty

4.3.5.1  Human

     Although there have been no studies of human populations exposed to
2,3,7,8-TCDD alone, there are a number of studies on human populations
exposed to chemicals contaminated with 2,3,7,8-TCDD, as reviewed in EPA
(1985a). Czeizel and Kiraly (1976) reported that there was an increase
in chromosomal aberrations of peripheral lymphocytes from workers in an
East European 2,4,5-trichlorophenoxyethanol plant. Studies of soldiers
(spouses and children of soldiers) exposed to Agent Orange (Mulcahy
1980) and of individuals involved in the Seveso accident (DiLernia et
al. 1982, Kaye et al. 1985, Mottura et al. 1981, Reggiani 1980, Tenchini
et al. 1983) have, however, failed to detect chromosomal aberrations.
Some of the individuals in these studies had skin eruptions consistent
with exposure to 2,3,7,8-TCDD. The only chromosomal effect in the latter
studies was a decrease in satellite association (SA), which is evidence
of functional ribosomal genes, in 2,3,7,8-TCDD-exposed subjects
(DiLernia et al. 1982). The authors noted that similar decreases
observed after x-irradiation exposure may possibly represent damage to
the nucleolar organizing regions. All human data on chromosomal
aberrations were confounded by mixed exposure to other potentially
active compounds and the inability to determine quantitatively the
extent of exposure to 2,3,7,8-TCDD.

-------
                                                 Toxicologies! Data   61

4.3.5.2  Nonhuman

     A recent review by Girl  (1986) of the mutagenic and genotoxic
effect of 2.3,7,8-TCDD concluded that there is evidence for the
genotoxic activity of 2,3,7,8-TCDD, but additional testing would be
required to demonstrate this  activity with certainty. The results of
nonhuman genotoxicity studies are summarized in Tables 4.2 and 4.3. The
early positive results of Hussain et al. (1972) and Seller (1973) are
likely artifacts resulting  from extensive cell death and possibly from
impurities in the test material. In vitro cytogenic tests in yeast in
both the standard plate test  and the intrasanguineous host-mediated
assay (using CD-I mice) have  produced positive results (Bronzetti et al.
1983). In in vivo cytogenetic assays, Loprieno et al. (1982) observed
that the positive responses depended on sampling time, with negative
results obtained 24 h posttreatment and positive results 96 h posttreat-
ment. Although the toxic properties of 2,3,7,8-TCDD usually segregate
with the Ah locus, Meyne et al. (1985) observed negative responses in
vivo after administration of  2,3,7,8-TCDD at hepatotoxic levels to both
responsive, C57B1/6J, and nonresponsive, DBA/2J, mice when sampled at 24
h. There are also limited supportive data for the mutagenicity of
2,3,7,8-TCDD from the observation in vivo of the low-level binding of
2,3,7,8-TCDD to liver macromolecules (Poland and Glover 1979).

4.3.5.3  General discussion

     The nonhuman genotoxicity data on 2,3,7,8-TCDD are conflicting;
negative results were reported in many of the assay systems, and when
positive results were observed, the response was generally weak. These
conflicting data may result from technical difficulties in testing
2,3,7,8-TCDD rather than from a lack of biological activity. Testing
difficulties arise from the extreme insolubility of this compound and
the high toxicity observed  in some test systems, which would be
anticipated to result in a very narrow window for effective genotoxic
doses. As a result of the largely negative data from nonhuman
genotoxicity assays, some investigators, as discussed by Paustenbach et
al. (1986), have concluded  that 2,3,7,8-TCDD is not a genotoxic agent.
Sufficient data are not available to precisely define the toxicological
mechanism of 2,3,7,8-TCDD and resolve these differences of opinion.

     Human studies are primarily limited by the lack of data on the
extent of exposure. The only  indication of exposure in these studies was
the development of gross skin lesions, which does not provide a good
estimate of either the extent of exposure or the duration. In addition,
the time after exposure that  the cells were examined may not have been
optimal for observing cytogenetic effects. Because of the limitations of
the studies in humans, these  data cannot be used to demonstrate that
2,3,7,8-TCDD does not pose  a  genotoxic hazard.

4.3.6  Carcinogenicity

4.3.6.1  Inhalation

     Pertinent data regarding the carcinogenicity of 2,3,7,8-TCDD
following inhalation exposure of humans or animals were not found  in the
available literature.

-------
                               Table 4.2. Genoloxicily of 2,3,7,8-TCDD in vitro
End point Species (test system)
Gene mutation Salmonella lyphimurium
(reverse mutation)
S. lyphimurium
(reverse mutation)
Escherichia coli
Results
with activation/without activation
-/-
Not tested/ +
Not tested/ +
References
McCann 1978,
Gilbert et al. 1980.
Geigerand Ncal 1981,
Mortelmans et al. 1984
Hussain et al. 1972,
Seiler 1973
Hussain et al. 1972
'ection 4


Cytogenetic
(reverse mutation)
Saccharomyces cerevisiae
(reversion)
LSI78Y mouse lymphoma
cells (forward mutation)
5. cerevisiae
(gene conversion)
                                                         Not tested/+, and
                                                            not tested/ -
Bronzelliet al 1983


Rogers et al. 1982


Bronzettiet al. 1983


Cell transformation
S. cerevisiae
(host mediated)
Chinese hamster cells
(sister chromatid exchange)
Baby hamster kidney cells —
BHK
C3H/ 10 TI/2 cells
+ /NA"
Not tested/ -
Not tested/ +
Not tested/ -
Bronzelliet al. 1983
Toth et al. 1984
Hay 1982
Abernathy et al 1985
    "Not available.

-------
                                                          lexicological Data    63
                 Table 4.3. Geootoxicity of 2,3,7,8-TCDD in vivo
  End point
Species (test system)      Results
                                                             References
Gene mutation   Drosophila (sex-linked
                recessive lethal)

Cytogenetic      Drosophila (sister
                chromatid exchange)

                Drosophila
                (structural aberration)

                Rat
                (sister chromatid exchange)

                Rats • marrow cells
                (structural aberration)

                Rats - marrow cells
                (structural aberration)

                Mouse - marrow cells
                (structural aberration)

                Mouse - marrow cells
                (sister chromatid exchange)

                Mouse - marrow cells
                (structural aberration)

                Mouse - marrow cells
                (micronucleus)
                                   Zimmenng et al.  1985


                                   Zeiger 1983


                                   Zeiger 1983


                                   Lundgren et  al. 1986


                                   Green and Moreland 197S


                                   Green et al.  1977


                                   Loprienoet al. 1982


                                   Meyne et al. 198S


                                   Meyne et al. 1985


                                   Meyne et al. 1985

-------
 64    Section 4

 4.3.6.2  Oral

      Human.   No studies are  available.

      Animal.   A number of bioassays have been conducted, and all have
 demonstrated that 2,3,7,8-TCDD  is carcinogenic in animals via  the oral
 route.  The NTP (1982a) and Kociba et al. (1978a,b) studies are key
 bioassays of 2,3,7,8-TCDD. Although both studies used sufficient numbers
 of  animals and exposure durations, the study by Kociba et al.  (1978a,b)
 is  relevant  to human exposure scenarios because the compound was
 administered daily in the diet  rather than biweekly by gavage as in the
 NTP (1982a)  study.  Furthermore, the study by Kociba et al. (1978a,b) has
 proven  to be  the most sensitive indicator of the carcinogenic potency of
 2,3,7.8-TCDD.  The details of the Kociba et al. (1978a,b) study are
 summarized in Table 4.4.  Other  oral studies which support the conclusion
 that  2,3,7,8-TCDD is an animal  carcinogen are presented in Table 4.5.

 4.3.6.3  Dermal

      Human.   Although there are no known cohorts that have been exposed
 solely  to 2,3,7,8-TCDD,  a number of cohorts that have been exposed to
 herbicides or industrial  chemicals contaminated with 2,3,7,8-TCDD have
 been  studied.  The predominant exposure route for these cohorts was
 probably  dermal,  although some  inhalation and oral exposure was also
 likely. In addition,  2,3,7,8-TCDD was only a minor contaminant of these
 compounds, and any effects observed may have been either caused or
 potentiated by the contaminated compound itself.

      EPA  (1985b,  1988b) and Hiremath et al. (1986) reviewed several
 epidemiology  studies  of humans exposed to herbicides contaminated with
 2,3,7,8-TCDD.  The studies by Axelson et al. (1980) and Theiss and
 Frentzel-Beyme (1977)  found an association between exposure and stomach
 cancer. A series  of other studies (Eriksson et al. 1979, 1981-  Hardell
 and Sandstrom 1979;  Hardell et al. 1980,  1981; Lynge 1985; Puntoni et
 al. 1986; Merlo  and Puntoni 1986) reported an association between
 exposure  and  soft-tissue  sarcomas (of various sites) and lymphomas.  EPA
 (1988b) discussed criticisms of the studies by Hardell and Sandstrom
 (1979)  and Eriksson et  al. (1979, 1981).  These criticisms include recall
bias, unreliability of  the exposure data, information bias, observation
bias, and confounding factors.  EPA concluded that the problems with
 these studies  were  not  sufficient to explain the highly significant
 risks of  soft-tissue  sarcoma in the exposed workers. In addition,
Hardell and Eriksson (1988) conducted a case-referent study in which
 they controlled  for recall and observation bias and found a three-fold
 increased risk for  soft-tissue sarcoma for exposure to phenoxyacetic
acids.

     EPA  (1988b)  also reviewed several studies (Balarajan and Acheson
1984, Cantor  1982,  Cook et al.  1986,  Fett et al.  1984, Kang et al. 1987
Kogan and Clapp  1985, Milham 1982, Smith et al.  1984, Woods et al. 1987,
Zack and  Suskind  1980)  that were consistent with or tended to support
the findings of  soft-tissue sarcoma in groups thought to be exposed to
chemicals contaminated with 2,3,7,8-TCDD. In most of these studies,
however,  exposure  to  2,3,7,8-TCDD sufficient to produce a significant
change  in risk estimates could only be assumed.  Also, some of the

-------
                                                     Toxicologies! Data    65
         Table 4.4 Summary of the oral carcinogenicity bioassay of
                         Kociba et al. (1978a,b)
 Animal    Sex
 Dose tested
(Mg/kg/day)
Tumor type
                                                            Incidence
Sprague-    M      Control     Squamous cell carcinoma of      0/85
Dawley                         the tongue, adenoma of the
cats                            adrenal cortex, and squamous
                               cell carcinoma of the hard
                               palate

                   0.001       Squamous cell carcinoma of       I/50
                               the tongue

                   0.01        Squamous cell carcinoma of       1/50
                               the tongue
                               Adenoma of the adrenal          2/50
                               cortex

                   0.1         Squamous cell carcinoma of       3/50
                               the tongue
                               Adenoma of the adrenal          5/50
                               cortex
                               Squamous cell carcinoma of       4/50
                               the hard palate

            F      Control     Hepatocellular carcinoma        1/86

                   0.001       Hepatocellular carcinoma        0/50

                   0.01        Hepatocellular carcinoma        2/50
                               Squamous cell carcinoma of      1/50
                               the hard palate

                   0.1         Hepatocellular carcinoma        11 /49
                               Squamous cell carcinoma of      4/49
                               the hard palate
                               Squamous cell carcinoma of      7/49
                               the lung

-------
o\
Table
Method of
exposure
Diet


Gavage











(0
ft
o
n
t—
0
4.5. Other oral studies supporting the conclusion that 2,3,7,8-TCDD is an animal carcinogen 3
*»

Animal
Sprague-
Dawley
rats
Osborne-
Mendel rats
Osborne-
Mendel rats

B6C3F1 mice

B6C3FI mice


Swiss mice

Sex/
number
M/IO


M/50

F/50


M/50

F/50


M/44


Doses tested
0.01,0.005,
0.05,0.5, 1.0,
or 5 ppb
0.01,0.05, or
0.5 /ig/kg/week
0.01,005, or
0 5 /ig/kg/week

0.01,0.05, or
0.5 Mg/kg/week
0.01,0.05. or
0.5 Mg/kg/week

0.007, 0.7. or
7.0 jig/kg/week

Tumor type
Increase in total tumor
incidence

Follicular-cell adenomas and
carcinomas of the thyroid
Neoplaslic nodules and hepato-
cellular carcinomas of the
liver
Hepatocellular carcinomas

Hepatocellular carcinoma and
follicular-cell adenomas of
the thyroid
Hepatomas and hepatocellular
carcinomas

References
Van Miller
et al. I977a,b

NTP I982a

NTP I982a


NTP I982a

NTP I982a


Tolh et al
1979

-------
                                                 lexicological Data   67

investigators reported that their results did not support the studies by
Hardell and Sandstrom (1979) and Eriksson et al. (1979, 1981).
Nevertheless, risks of soft-tissue sarcoma, although not statistically
significant, wer.e detected in the studies. A number of studies found no
association between risk of soft-tissue sarcoma and exposure to
herbicides contaminated with 2,3,7,8-TCDD (Axelson et al. 1980;
Fingerhut et al. 1984; Greenwald et al. 1984; Lathrop et al.  1984; Ott
et al. 1980; Riihimaki et al. 1982; Thiess et al. 1982; Wiklund and Holm
1986; Wiklund et al. 1987; Wolfe et al. 1984, 1985). However, some of
these studies suffer from problems that include low power to detect a
significant risk, selection and survivorship bias, and insufficient
latency periods  (EPA 1988b).
     EPA (1988b) concluded that the epidemiological data provide limited
evidence that exposure to phenoxyacetic acid herbicides and/or
chlorophenols is causally related to the risks of soft-tissue sarcoma,
but none of the data are sufficient to implicate 2,3,7,8-TCDD alone.
     Animal.  There is only limited evidence that 2,3,7,8-TCDD produces
tumors in laboratory animals following dermal exposure. NTP  (1982b)
administered 2,3,7,8-TCDD alone to Swiss mice. Female mice, but not male
mice, had an increased incidence of fibrosarcomas in the integumentary
system.
     As reviewed by EPA (1985a, 1988a), there is conflicting evidence
that 2,3,7,8-TCDD acts as a tumor promoter when applied  to  the skin.
Berry et al. (1978) failed to detect any tumor-promoting activity by
2,3,7,8-TCDD in CD-I mice skin initiated with dimethyIbenzanthracene
(DMBA), and Slaga and Nesnow (1985) reported that unpublished data
indicated that 2,3,7,8-TCDD either had no promoting activity or very
weak promoting activity in Sencar mice skin. NTP  (1982b) also examined
the ability of 2,3,7,8-TCDD to act as a tumor promoter in Swiss Webster
mice, and animals treated with 2,3,7,8-TCDD alone had  similar  tumor
incidences to animals treated first with DMBA followed by treatment with
2,3,7,8-TCDD. NTP (1982b) concluded, however, that  "in the  DMBA-TCDD
experiment, failure to have included groups skin painted with  only  DMBA
precludes interpretation of these results." Poland  et  al.  (1982),
however, not only demonstrated promotion, but also  that  genetic
differences in CD-I mice affect the ability of  2,3,7,8-TCDD to act  as a
promoter. In HRS/J mice homozygous for the hairless trait,  promotion
with  2,3,7,8-TCDD after initiation with DMBA produced  as many skin
tumors (both incidence and multiplicity)  as promotion  with  the known
promotor TPA, whereas in mice heterozygous for  the  hairless trait  (and
wild  type), skin tumors could only be  promoted  by TPA  but  not 2,3,7,8-
TCDD.  Even  in the homozygous mice, 2,3,7,8-TCDD did not  produce  the
commonly observed hyperplasia associated  with promoters  such as  TPA,
suggesting  that  2,3,7,8-TCDD has  a different mechanism of  action.  Other
studies indicated that pretreatment with  2,3,7,8-TCDD  could block the
subsequent  DNA binding of known  carcinogens  and also prevent tumor
initiation  (Berry et  al.  1979, Cohen  et al.  1979).  Although not  a dermal
study, Pitot et  al.  (1980)  also  demonstrated promotion by  administering
a  single intragascric dose  of  the hepatocarcinogen diethylnitrosamine
(DEN)  followed by repeated  subcutaneous  injections  of  2,3,7,8-TCDD. No

-------
68   Section 4

hepatic tumors were observed in animals given DEN or 2,3,7,8-TCDD alone,
but the combined treatment resulted in the development of hepatocellulai
carcinomas.

4.3.6.4  General discussion

     The evidence from human epidemiology studies that 2.3,7,8-TCDD is
carcinogenic is difficult to assess because (1) exposure to 2,3,7,8-TCDD
is poorly documented and (2) exposure occurred to other potentially
active materials. The strongest evidence is from the induction of soft-
tissue sarcomas at various sites, and it has been questioned whether
combining tumor data from various sites is appropriate. EPA (1985b)
regards the human evidence for soft-tissue sarcomas and lymphomas as
"limited" (i.e., some evidence) from exposure to phenoxyacetic acid
herbicides and/or chlorophenols which have 2,3,7,8-TCDD impurities.
Thus, for 2,3,7,8-TCDD alone, the evidence is considered inadequate.
With regard to the observed increase in stomach cancer, the two groups
of workers studied were relatively small, and similar increases in
stomach tumors have not been reported in other more extensive studies.
     The animal data, however, clearly indicate that 2,3,7,8-TCDD is
carcinogenic, although there is some disagreement in the scientific and
international regulatory community as to whether 2,3,7,8-TCDD acts as a
complete carcinogen or as a carcinogen promotor (Shu et al. 1987). The.
rationale for describing 2,3,7,8-TCDD as a promotor is based on the poor
response of 2,3,7,8-TCDD in many short-term mutagenicity assays, the
lack of strong evidence for binding to DNA, and positive results in in
vivo promotion assays using the classic skin-painting technique (as
described above) and the study of Pitot et al. (1980) using the two-
stage model in rat liver. The classic feeding study bioassays, however,
would support the view that 2,3,7,8-TCDD is a complete carcinogen.

4.4  INTERACTIONS WITH OTHER CHEMICALS

     There are few data on the interactions of 2,3,7,8-TCDD with other
chemicals. As discussed in EPA (198Sa), 2,3,7,8-TCDD is a strong inducer
of microsomal enzymes; hence, prior exposure to 2,3,7,8-TCDD will alter
the race of metabolism and toxicity of many compounds that are either
detoxified or activated by this enzyme system. The observed inhibition
by 2,3,7,8-TCDD of the tumorigenic response of known tumorigens in  the
mouse skin bioassay  (Berry et al. 1979, Cohen et al. 1979) may be an
example of 2,3,7,8-TCDD-induced enzyme changes altering the metabolic
fate and toxicity of another compound.

     The only other  interactions that have been observed are the
additive effect of 2,3,7,8-TCDD and similar polychlorinated
dibenzofurans and polychlorinated biphenyls on the induction of cleft
palate in mice  (Weber et al. 1985, Birnbaum et al. 1985) and the
increased sensitivity of mice to 2,3,7,8-TCDD-induced cleft palate  with
the co-administration of the hormones thyroxine and triiodothyroxine
(Lamb et al. 1986). With regard  to environmental exposure, the relevance
of interactions of 2,3,7,8-TCDD  and the high levels of hormones used  in
the latter study is  unclear.

-------
                                                                      69
               5.  MANUFACTURE, IMPORT, USE, AND DISPOSAL

5.1  OVERVIEW
     2,3,7,8-TCDD is neither commercially manufactured nor imported into
the United States. It was produced inadvertently in small amounts as an
impurity during the manufacture of compounds for which 2,4,5-
trichlorophenol was a synthetic intermediate. At the present time,  its
only use is in chemical research. Several field-tested and untested
methods are available for the disposal/stabilization of 2,3,7,8-TCDD-
containing wastes. Some of the promising methods are incineration at
high temperature, oxidative destruction with the aid of a catalyst,
biodegradation by a fungus, photochemical destruction in the presence of
a hydrogen-donating substrate, and stabilization (in case of soils)
through the in situ addition of cementitious and asphaltic materials or
burial under a protective layer.

5.2  PRODUCTION
     2,3,7,8-TCDD is synthesized on a  laboratory scale primarily by two
processes: (1) condensation of dichlorocatechol with substituted
dichlorobenzer.es and (2) halogenation  of dibenzo-p-dioxin or its
dichloro-substituted derivative  (EPA 1985b). 2,3,7,8-TCDD is not
commercially manufactured in the United States but is produced as an
undesirable product during the manufacture  of compounds for which
2,4,5-trichlorophenol is a synthetic intermediate (see Sect. 6.2 on
Releases to the  Environment).

5.3  IMPORT
     2,3,7,8-TCDD is not imported  into the  United States  (EPA 1985b).

5.4  USE
     2,3,7,8-TCDD has been tested  for  flame-proofing polyesters  and  as a
control against  insects and wood-destroying fungi in Germany; however,
it has probably  never been commercially produced or used  for these
purposes. At present it is only  used as a  research chemical  (HSDB  1987).

5.5  DISPOSAL/STABILIZATION
     For  the destruction of  wastes and residues containing  2,3,7,8-TCDD,
 incineration at  a minimum  temperature  of  800 to 1,200'C  and a contact
 time of >30 s  was  found  to be  satisfactory. Polychlorophenols containing
 2,3,7,8-TCDD as  an  impurity  have been  destroyed by  dispersing or
dissolving the material  in water,  or a nonnucleophilic organic  solvent,
 and oxidizing  with  ruthenium tetraoxide  catalyst  at -70°C (HSDB 1987).
 Satisfactory  field-tested  methods  for  the disposal  of 2,3,7,8-TCDD-
 containing soils were  not  available until recently.  The  following

-------
70   Section 5

methods have either shown promise or are presently used for the
disposal/stabilization of 2,3,7,8-TCDD:  (1) stabilization through the L.
situ addition of cementitious and asphaltic materials;  (2) degradation
with Phanerochaete chrysosporium, a white rot fungus;  (3) a mobile
incineration system for the thermal destruction of 2,3,7,8-TCDD;  and (4)
ultraviolet photolysis in the presence of alkali polyethylene glycolate
reagents (des Rosiers 1986). A method (similar to method 4) for
photochemical destruction of 2,3,7,8-TCDD in the presence of olive oil
(a hydrogen donor) was used with soil from the area of  the Seveso
accident. A recent report discusses the  disposal/stabilization of
2,3,7,8-TCDD by a mobile poly(ethylene)glycol-potassium hydroxide
destruction unit (Rogers et al. 1987). Other methods for the
stabilization of contaminated soil that  are cost effective and produce
minimal dusting have been proposed (Hungerford 1988).  In these proposed
methods, stabilization is achieved either by placing a  protective layer
consisting of compacted soil with vegetation cover, asphalt, or concrete
over the contaminated soil or by inversion of the soil  horizon to
effectively bury the contaminated surfacial material.

-------
                                                                      71
                          6.  ENVIRONMENTAL FATE

6.1  OVERVIEW

     The important sources of 2,3,7,8-TCDD in the environment are
production and use of certain herbicides and chlorophenols,  incineration
of municipal and  industrial wastes, and improper disposal of chemical
wastes produced during the manufacture of 2,4,5-trichlorophenol, 2,4.5-
T, and related herbicides, hexachlorophene, and chlorinated benzenes.
The bleaching process in  the pulp and paper industry, and exhaust from
vehicles not equipped with catalytic converters are also important
sources of 2,3,7,8-TCDD.  The fate of 2,3,7,8-TCDD in the environment is
not clearly understood. It appears that particulate-bound 2,3,7,8-TCDD
in the air may undergo photolysis and may be removed by wet and dry
deposition. The half-life of atmospheric 2,3,7,8-TCDD is such that
2,3,7,8-TCDD can be transported long distances in the air, although this
transport will be of shorter distances compared with the transport of
higher chlorinated dibenzodioxins having longer half-lives.  The ultimate
sink of airborne 2,3,7,8-TCDD is sediments of surface waters. The two
processes that are likely to remove 2,3,7,8-TCDD from water and soils
are vaporization and photolysis. The estimated half-life of 2,3,7,8-TCCD
in surface water is >1 year, and the ultimate sink of aquatic 2,3,7,8-
TCDD is sediments. The bioconcentration factor of 2,3,7,8-TCDD in
rainbow trout (Sal/no gairdneri) is 39,000. 2,3,7,8-TCDD is immobile in
most soils, but horizontal movement of soil-bound 2,3,7,8-TCDD may occur
in runoff water during flooding. As observed in Seveso, Italy, minimal
vertical movement may occur in soils containing low organic matter. The
estimated half-life of 2,3,7,8-TCDD is 1 to 3 years on soil surfaces and
10 to 12 years in the interior of soils. Although not accumulated, the
level of 2,3,7,8-TCDD absorbed in parts of plants underground is of the
same order of magnitude as in soil, but the aerial parts of plants
contain 50% lower concentrations.

6.2  RELEASES TO THE ENVIRONMENT

     Although the following paragraphs discuss the sources of 2,3,7,8-
TCDD in the environment,  the sources responsible for its background
levels are not clear.

6.2.1  Production and Use of Certain Herbicides and Chlorophenols and
       Bleaching Process  in Pulp and Paper Industry

     The phenoxy herbicide 2,4,5-T produced prior to 1960 contained up
to 100 pg/g 2,3,7,8-TCDD. The level of 2,3,7,8-TCDD in commercial
2,4,5-T was reduced in recent years to <0.1 Mg/g. and most commercial
2,4,5-T available before  its banning contained <0.02 pg/g 2,3,7,8-TCDD.
Agent Orange, a 1:1 mixture of butyl esters of 2,4,5-T and 2,4-D
produced before 1970, contained 0.02 to 54 pg/g 2,3,7,8-TCDD.

-------
72   Section 6

Hexachlorophene, a germicide manufactured from trichlorophenol,
contained 0.2 to 0.5 ng/g 2,3,7,8-TCDD.  2,4,6-Trichloro-,  2,3,4.6-
tetrachloro-,  and pentachlorophenol were found to contain <0.1 A»g/g
other tetra isomers but no 2,3,7,8-TCDD. 2,3,7,8-TCDD was detected at a
concentration <1 ng/g (2,3,7,8-TCDD detection limit of 0.03 ng/g) in all
samples of sodium pentachlorophenate,  2,3,4,5-tetrachlorophenol,  and
hexachlorophene. It has been speculated  that catalytic dechlorination of
higher chlorinated dibenzodioxins in metal containers during pressure
treatment of wood may be an additional source of 2,3,7,8-TCDD in
pentachlorophenol and sodium pentachlorophenate-treated woods.
Therefore, pentachlorophenol-treated woods might well be one of the main
sources in the contribution of human body burden for 2,3,7,8-TCDD. A
sample of 2,4,5-trichlorophenol manufactured in 1969, on the other hand,
contained up to 6.2 pg/g 2,3,7,8-TCDD. Similarly, diphenyl ether
herbicides were found to contain other tetrachloro isomers but no
2,3,7,8-TCDD (EPA 1985b, Firestone et al. 1972, Hagenmaier 1986,  HSDB
1987, Rappe 1984, Weeren and Asshauer 1985).  From the analysis of
sediments of a western Lake Ontario site, Czuczwa and Hites (1986)
concluded that the likely source of tetrachlorodibenzo-p-dioxins was a
pentachlorophenol production facility. The analytical method used,
however, could not distinguish 2,3,7,8-TCDD from other tetra isomers.
Sludge from seven pulp and paper mills showed a 2,3,7,8-TCDD
concentration range from not detectable  (1 pg/g) to >400 pg/g (Kuehl et
al.  1987). The bleaching process used in this industry is the most
likely causative factor for the production of 2,3,7,8-TCDD and other
higher chlorinated dibenzodioxins.

6.2.2  Photochemical Reactions

     The photochemical reaction of phenoxy herbicides has been found to
produce polychlorinated dibenzo-p-dioxins through photodechlorination
and subsequent condensation reactions; however, this process does not
produce 2,3,7,8-TCDD (Rappe 1984). Lower substituted dibenzo-p-dioxins
are also formed during photodechlorination of higher chlorine-
substituted dibenzo-p-dioxins. Trace amounts of 2,3,7,8-TCDD were
observed from the photodechlorination of both 1,2,3,6,7,8-hexa- and
1,2,3,7,8,9-hexachlorodibenzo-p-dioxin (Buser 1979).

6.2.3  Thermal Reactions

     Small amounts of 2,3,7,8-TCDD have  been detected in the flue gases
from municipal incinerators.  From the experimentally determined
concentrations in flue gases of five municipal incinerators, the maximum
average concentration of 2,3,7,8-TCDD in ambient air at ground level was
estimated as 38 fg/g. Incineration of industrial wastes containing
2,4,5-T salts and esters, polychlorinated benzenes, and chlorophenoxy
ethers also produced 2,3,7,8-TCDD (Barnes 1983, Rappe 1984). Upon
analysis of sediments from Saginaw Bay,  Saginaw River, and the Great
Lakes, Czuczwa and Hites (1984,  1986)  concluded that the source of
tetrachlorodibenzo-p-dioxins was incineration, although the analytical
method used was unable to separate 2,3,7,8-TCDD from other tetra
isomers. Combustion of coal did not produce 2,3,7,8-TCDD at a detectio-
limit of 1.2 ng/kg (HSDB 1987),  but burning of woods did produce 0.65
pg/kg 2,3,7,8-TCDD (EPA 1985b).  Exhausts from automobiles powered with

-------
                                                 Environmental Face   73

leaded gasoline were reported to contain <0.05 to 0.3 ng 2,3,7,8-
TCDD/24.8 km, but no 2,3,7,8-TCDD was detected in exhausts of
automobiles powered with unleaded gasoline (Harklund et al.  1987).  It
was speculated that dichloroethane used as a scavenger was the source of
2,3,7,8-TCDD in exhaust from leaded gasoline. In automobiles powered
with unleaded gasoline and equipped with a catalytic converter, the
absence of dichloroethane may prevent formation of 2,3,7,8-TCDD and the
catalytic converter may additionally destroy any 2,3,7,8-TCDD that  may
be formed. Therefore, with the replacement of older automobiles,  this
source of 2,3,7,8-TCDD should be on the decline. Accidental fires
involving capacitors or transformers containing chlorobenzene will  also
release 2,3,7,8-TCDD to the environment. An example of such a
contamination is the transformer fire in the State Office Building  in
Binghamton, New York.

6.2.4  Improper Disposal of Chlorinated Chemical Wastes
     Improper disposal of certain chemical wastes produced during the
manufacture of 2,4,5-trichlorophenol, 2,4,5-T, and related herbicides,
hexachlorophene, and chlorinated benzenes, may be a source of 2,3,7,8-
TCDD in the environment. Examples of such improper disposal leading to
the contamination of the environment are the Love Canal, Niagara Falls,
New York, sites where 2,3,7,8-TCDD up to a level of 672 pg/kg was
detected. Similarly, several sites in the state of Missouri were
contaminated with up to 1,750 MgAg 2,3,7,8-TCDD (Tiernan et al. 1985).

6.3  ENVIRONMENTAL FATE
     The fate of 2,3,7,8-TCDD in air, water, and soil is not understood
with certainty. Although some experimental efforts have been directed in
recent years to elucidate its fate in different media, a substantial
data gap exists in this area. In air, 2,3,7,8-TCDD is likely to be
present partly in the gas phase and predominantly in the particle-sorbed
phase. The two important processes that may  remove 2,3,7,8-TCDD from the
atmosphere are photochemical degradation and wet deposition. The
photodegradation of gas phase 2,3,7,8-TCDD with an estimated half-life
of a few hours is expected to be much faster than particle-sorbed
chlorinated dibenzodioxins. Even an estimate of the atmospheric half-
life of 2,3,7,8-TCDD is not available. On the basis of photochemical
experiments with 2,3,7,8-TCDD coated on silica  gel, the half-life of
atmospheric particulate 2,3,7,8-TCDD may be  a few days. The  lifetime of
atmospheric 2,3,7,8-TCDD is such that it can be transported  long
distances  in the air, although  this transport will be of shorter
distances  compared with the transport of higher chlorinated
dibenzodioxins. The ultimate environmental sink of airborne  particulate
2,3,7,8-TCDD is likely  to be sediments  of surface waters  (Choudhry  and
Hutzinger  1982, Czuczva and Kites  1986, Eitzer  and Hites  1986, Miller et
al. 1987,  Podoll et al. 1986).
     The biodegradation of 2,3,7,8-TCDD in water  is probably slow.  The
two processes  that may be important  for the  removal of  2,3,7,8-TCDD are
volatility and photodegradation. Although the photolysis  of  2,3,7,8-TCDD
in hydrogen-donating solvents  is a fast process,  a  suspension of
2,3,7,8-TCDD in distilled water showed  no appreciable photodegradation.
In natural waters,  the  presence of small  amounts  of hydrogen-donating

-------
74   Section 6

substrate or the presence of photosensitizers may account for its
observed photodegradation; however, the photochemical degradabllity of
2,3,7.8-TCDD in water, as provided by model ecosystem studies (Matsumura
et al. 1983, Tsushimoto et al. 1982), has not provided definite evidence
through mass balance that the observed loss of 2,3,7,8-TCDD attributed
to photolysis was not due to its sorption on sediment and biota.  The
estimated half-lives for dissolved 2,3,7,8-TCDD in sunlight may range
from 118 hours in winter to 21 hours in clear near-surface water under
clear skies. The photodegradation is usually a dechlorination process
leading to the formation of tri- and dichlorinated dibenzo-p-dioxins.  In
sediment-containing lake water, the estimated half-life of 2,3,7,8-TCDD
is >1.S years. In lake water alone, the estimated half-life is >1 year.
The ultimate sink of aquatic 2,3,7,8-TCDD is the sediment. Recent flow-
through experiments with fathead minnows (Pimephales promelas) have
shown that the bioconcentration factor for 2,3,7,8-TCDD in this species
is 7,900 to 9,300 on a wet-weight basis (Adams et al. 1986, EPA 1985b,
Podoll et al 1986). Recently, the steady-state bioconcentration factor
for 2,3,7,8-TCDD in rainbow trout (Salmo gairdneri)  as extrapolated from
experimental data has been estimated to be 39,000 (Mehrle et al.  1988).
The lower values reported by earlier authors may be due to nonattainment
of steady-state concentration in fish tissues.

     2,3,7,8-TCDD is expected to be immobile in most soils by irrigation
and rainfalls. A downward movement of 10 cm in 12 years was observed
with soil from Eglin Air Force Base. Although 2,3,7,8-TCDD usually does
not leach through soil, leaching is possible in some instances from
soils of very low organic carbon content as a result of 2,3,7,8-TCDD
solvation with organic solvent or biotic mixing by earthworms or other
soil invertebrates. A white rot fungus (Phanerochaete chrysosporium") has
been shown to degrade 2,3,7,8-TCDD. This biodegradation does not occur
significantly in natural soils, probably because of the lack of this or
other degrading microorganisms. Both volatilization and photoreaction
may remove some 2,3,7,8-TCDD from soil surfaces. The photoreaction on
soil surfaces can be greatly enhanced by the presence of hydrogen-
donating substrates (e.g., olive oil or arachis oil) in soil. The
photoreaction will be insignificant beyond the surface soil layers. The
environmental half-life of 2,3,7,8-TCDD is highly dependent upon soil
characteristics, the mode of contamination, and climatological
conditions. The estimated half-life of 2,3,7,8-TCDD on soil surfaces is
1 to 3 years, but the half-life in the interior of soil may be 10 to 12
years (Bumpus et al. 1985, EPA 1985b, Freeman and Schroy 1986, HSDB
1987, Miller et al, 1987).

     2,3,7,8-TCDD present on leaves of plants as a result of spraying
herbicides will photolyze with a half-life of a few hours. The chemical
is absorbed by higher plants and is probably translocated, but it is not
accumulated. The absorption by underground parts may be at the same
level as soil, but the aerial part contains -50% lower concentrations
(Choudhry and Hutzinger 1982. Sacchi et al. 1986). Consumption of foods
derived from contaminated areas (e.g., near municipal incinerators),
could represent a significant dietary source. However, Insensee and
Jones (1971) and Wipf and Schmid (1983) observed no  significant plant
uptake in mature oats and soybeans, and edible parts of roots vegetable
contained much less 2,3,7,8-TCDD than the surrounding soil.

-------
                                                                      75
                    7.  POTENTIAL FOR HUMAN EXPOSURE
7.1  OVERVIEW
     The human exposure pathways to 2.3,7,8-TCDD have changed since the
late 1970s. Because 2,3,7,8-TCDD was a contaminant in herbicide
preparations containing 2,4,5-T, the manufacture, use, and disposal of
these herbicides were the primary sources of exposure to 2,3,7,8-TCDD.
In 1979, EPA (1979) completely banned the use of 2,4,5-T.  With the
stoppage of production of 2,4,5-T and other pesticide preparations
containing the contaminant 2,3,7,8-TCDD, both occupational and general
population exposure to 2,3,7,8-TCDD due to manufacture and use of the
herbicides ceased to exist. Presently, the important sources of
2,3,7,8-TCDD exposures to the general population are contaminated soil,
dump sites, and municipal incinerators. 2,3,7,8-TCDD has been found in
at least 28 of 1,177 sites on the National Priorities List (View 1989).
With the change of sources, the exposure pathways have also changed over
the years (i.e., dermal and inhalation exposure from the manufacture and
use of 2,4,5-T to ingestion of foods obtained from contaminated sites).

     There is a paucity of data on the level of 2.3.7.8-TCDD in ambient
air due to sampling and analytical difficulties associated with
quantification of very low levels of 2,3,7,8-TCDD. The ambient level of
2,3,7,8-TCDD in urban areas in West Germany and Sweden ranged from
<0.001-0.08 pg/m3. Although ambient air samples from several locations
in the United States have been collected for the quantification of
2,3,7,8-TCDD levels, results from these studies are not yet available.

     The concentration of total tetrachlorodibenzo-p-dioxin (not
2,3,7,8-TCDD alone) in ambient air in Bloomington, Indiana, was 18 to
92 fg/m3 (femtograms per cubic meter). Assuming that the 2,3,7,8-TCDD
isomer constitutes 5% of the total tetra isomers, the concentration of
2,3,7,8-TCDD in Bloomington air would be 0.9 to 4.6 fg/m3. The
concentration of 2,3,7,8-TCDD in the air around the stack of a municipal
incinerator has been estimated as 38 fg/m3. The accidental transformer
fire in Binghamton, New York, produced a much higher level of 2,3,7,8-
TCDD- -at 0.23 to 0.47 pg/m3. The concentration of 2,3,7,8-TCDD in  the
air surrounding a field after the application of Silvex containing
15 ppm 2,3,7,8-TCDD was 0.62 pg/m3. Other  than certain industrial
effluents and leachates from chemical dump sites, no 2,3,7,8-TCDD  has
ever been reported in drinking water. The  concentrations of 2,3,7,8-TCDD
in most uncontaminated soils are below  the detection  limits of
analytical methods. In urban soils, the level of 2,3,7,8-TCDD  is  in  the
range <0.0002 to 0.011 ng/g. Much higher levels have been detected in
soils contaminated by certain hazardous wastes, waste oils, and  spillage
of 2,4,5-trichlorophenol. A soil from Shenandoah Stables  in Missouri
contaminated by waste oil containing  2,3,7,8-TCDD had up  to  1,750  ng/g
of 2,3,7,8-TCDD. Fish samples collected from  lakes  and a  selected

-------
76   Section 7

Michigan river contained undetectable levels to 67 pg/g 2.3 . 7,8-TCDD. N
2,3,7,8-TCDD was detected in rice, soybeans, and crawfish in thfc United
States, or in Canadian chicken and pork samples. 2,3,7,8-TCDD at a
concentration range of 0.021-0.049 ng/kg has been detected in three of
six samples of cow's milk from Switzerland.  Although several other PCDDs
and PCDFs have been detected in some foods in Japan, no 2,3,7,8-TCDD has
been found in these samples at sub-ppt detection limits.  Since fat is
the chief contributor to the body burden of 2,3,7,8-TCDD in humans, it
has been analyzed by many investigators. Levels of 2,3,7,8-TCDD in
adipose tissue of exposed and control persons in Missouri have been
determined. The level of 2,3,7,8-TCDD in adipose tissue in certain
segments of the general population in the United States and Canada
ranges from undetectable to 20 pg/g, with a mean value of 5 to 7 pg/g.
Values as high as 99 pg/g were detected in an individual heavily exposed
during the spraying operation in South Vietnam. 2,3,7,8-TCDD has been
detected in human milk in the United States at 0.29 pg/g; in members of
the general population in Sweden at trace to 2.3 pg/g, with a mean value
of 0.6 pg/g; and in Germany at 1.3 to 3.3 pg/g, with a mean value of
I-9 Pg/g- 2,3,7,8-TCDD was also found in human milk in other European
countries. Although there are no data regarding exposure levels, workers
at sites of improper chemical waste disposal and the general population
residing near these sites may be susceptible to higher exposure to
2,3,7,8-TCDD. Breast-fed babies nursed by mothers residing near
improperly operated municipal incinerators may also be at higher risk.
The estimated daily human exposure to 2,3,7,8-TCDD, expressed as
picograms per kilogram of body weight (pgAg) ,  is 0.02 through
inhalation, 0.5 to 5 through the consumption of milk, and 20 through th
consumption of fish. The daily exposure of a 5-kg baby resulting from
the consumption of 850 mL of breast milk is 20 to 200 pg/kg.  From the
estimated body burden and the half-life of 2,3,7,8-TCDD in the human
body, the daily human intake of 2,3,7,8-TCDD has been estimated as
0.05 ng in one study and in the range of 0.003-0.036 ng in another.

7.2  LEVELS MONITORED OR ESTIMATED IN THE ENVIRONMENT

7.2.1  Air

     Due to sampling and analytical difficulties associated with
quantification of very low concentrations of 2,3,7,8-TCDD, very few data
are available for the levels of this compound in ambient air. The
concentration of total tetrachlorodibenzo-p-dioxin isomers in both vapor
and particulate phase in the ambient air in Bloomington. Indiana, was
approximately 18 to 92 fg/m3. The analytical method used could not
unequivocally identify 2,3,7,8-TCDD from other tetra isomers. Using the
measured concentration in flue gases from five municipal incinerators
and an air dispersion model, the maximum ambient concentration of
2,3,7,8-TCDD in the area around the stacks was estimated as 38 fg/m3.
Since municipal incinerators are one of the prime sources of atmospheric
2,3,7,8-TCDD, there are a vast number of publications concerning the
level of these compounds in the fly ashes of the incinerators; however,
most of the publications failed to distinguish 2,3,7,8-TCDD from its
isomers. The levels of 2,3,7,8-TCDD in the flue gas of European
incinerators are 0.05 to 1.3 ng/m3; in U.S. incinerators, the levels ai
a maximum of 3.5 ng/m3 (under normalized conditions of the emitted

-------
                                       Potent Lai for Human Exposure   77

gases) (Barnes 1983, Eitzer and Hites 1986, EPA 1985b, Marklund et al.
1986, Nottrodt and Ballschmiter 1986). The measured concentrations of
2,3,7,8-TCDD in four locations in Hamburg, West Germany,  were in the
range of 0.02-0.08 pg/m3 (Rappe and Kjeller 1987). The locations
consisted of an urban site, a traffic tunnel, a site downwind from a
municipal incinerator, and a site in the vicinity of a dump site and a
metal refinery. The levels of 2,3,7,8-TCDD in urban air in Sweden ranged
from 0.001-0.009 pg/m3 (Rappe and Kjeller 1987). Although ambient air
samples from several locations in the United States have been collected
for quantifying the levels of PCDDs and PCDFs, the results are not yet
available.
     Accidents involving certain transformer/capacitor fires can release
larger amounts of 2,3,7,8-TCDD in the air. The concentration of
2,3,7,8-TCDD in the air of the State Office Building  in Binghamton, New
York, following an accidental fire, was 0.23 to 0.47  pg/m3. Following an
accidental locomotive fire in Sweden, the concentration of 2,3,7,8-TCDD
was 50 pg/m3. The concentration of 2,3,7,8-TCDD in the air surrounding a
field after the application of Silvex containing  15 ppm 2,3,7,8-TCDD was
0.62 pg/m3, but the concentration dropped to a level  of 0.18 pg/m3 on
the second day (EPA 1985b, Rappe et al. 1985, Smith et al. 1986).

7.2.2  Water
     No report is available on the detection of 2,3,7,8-TCDD  in drinking
water, using methods with  detection limits  in  the pico-per-liter  range;
however,  2,3,7,8-TCDD has  been detected in  aqueous  industrial  effluents,
sediments, and leachates from hazardous waste  sites.  The  concentrations
of tetrachlorinated dibenzo-p-dioxins,  including  2,3,7,8-TCDD,  in
effluents from a  trichlorophenol manufacturing facility ranged from  none
detected  (detection limit,  10 to  30 pg/g)  to  100  pg/g. The discharged
wastewater effluent from Dow  into  the Tittabawassee River, in Michigan,
has been  reported to be approximately 15  pg/L.  The  leachate  samples  from
a waste disposal  site  in Jacksonville,  Arkansas,  had  a mean  2,3,7,8-TCDD
level of  14 ng/L. The  sump pump water from residences and leachates  from
the  Love  Canal area in New York contained 2,3,7,8-TCDD ranging from none
detected  to  1,560 ng/L. The concentrations of 2,3,7,8-TCDD in sediments
from storm sewers,  residential  sump  water,  and surface water around the
same  site were none detected  (detection limit,  10 to  100  pg/g) to 9,570
ng/g (EPA 1985b,  Lamparski et al.  1986, Tieman et aL. 1985).  The
concentration of  2,3,7,8-TCDD in the sludge of seven pulp and paper mill
wastewaters  in the  U.S. had concentration levels ranging from none
detectable (<1.0  pg/g) to  414 pg/g (Kuehl et al.  1987).

 7.2.3  Soil
      Concentrations of 2,3,7,8-TCDD in most soils with no obvious source
 of contamination are  below the detection limits of current analytical
 methods.  In urban soils,  the level of 2,3,7,8-TCDD is in the range of
 <0.0002 to 0.009  ng/g. In a national dioxin study, EPA sampled soils
 from 138 rural and 221 urban sites not associated with sources of
 2,3,7,8-TCDD. Only 17 of the rural and urban soils had detectable levels
 of 2,3,7,8-TCDD at a concentration range of 0.2-11.2 pg/g (TMN 1987).
 2,3,7,8-TCDD has been detected in samples that originated from certain
 industrial sites, waste disposal sites, and sites involved in accidental

-------
 78    Section 7

 spillage  of chemicals containing 2,3,7,8-TCDD. The levels of  2,3  7 8-
 TCDD in soils from different  locations are given  in Table 7.1.'it is
 apparent  from Table 7.1 that  the accidental or improper disposal of
 still-bottom residue -from the manufacture of 2,4,5-trichlorophenol
 (2,4,5-TCP)  may produce one of the highest levels of 2,3,7,8-TCDD in
 soils.

 7.2.4   Other

     There  are limited data that 2,3,7,8-TCDD does not bioaccumulate in
 crop plants  (Anonymous 1985).  Crops grown in soil contaminated with
 2,3,7,8-TCDD (up to 752 ppt)  after the Seveso accident contained only a
 few  parts per trillion of 2,3,7,8-TCDD in the aboveground portions.  The
 roots of  these plants,  however,  contained higher levels of 2,3,7,8-TCDD
 than the  surrounding soil, suggesting that a similar study using root
 crops would  demonstrate the contamination of the edible portion of the
 plant.  This  could represent a significant source of dietary intake in
 some areas.  However,  Wipf and Schmid (1983) reported that edible
 portions of  root vegetables contained much less 2,3,7,8-TCDD than the
 surrounding  soil.

     Since aquatic  organisms  bioconcentrate 2,3,7,8-TCDD, a few
 investigators  analyzed fish-eating birds as an indicator of possible
 pollution in the suspected water bodies.  A herring gull sample from Lake
 Huron contained 75  pg/g 2,3,7,8-TCDD. Similarly,  herring gull eggs
 collected from the  Great Lakes contained 12 to 101 pg/g 2,3,7,8-TCDD.
 Samples from Lake Ontario  and Saginaw Bay had the maximum levels of
 2,3,7,8-TCDD (Stalling  et  al.   1986); those from Lake Michigan and Lake
 Superior had the minimum levels of contamination (Buser and Rappe 1984).
 Fish samples  from the Great Lakes and selected Michigan rivers were
 shown to contain between undetectable amounts (detection limit  2 pg/g)
 and  67 pg/g  2,3,7,8-TCDD (Fehringer et al. 1985,  Niemann 1986, Ryan et
 al.   1984). Yellow perch samples from Woods Pond,  Massachusetts, were
 found to contain 26 pg/g 2,3,7,8-TCDD (Buser and Rappe 1984).  Fish
 obtained from  the Androscoggin River in Maine,  the Wisconsin River in
Wisconsin, and Rainy River in Minnesota,  all of which received
discharges from pulp and paper mills, contained 29,  125 and 185 pg/g of
 2,3,7,8-TCDD,  respectively (Kuehl et al.  1987).  Fish collected from
about 400 selected  and  random sites by EPA contained 2,3,7,8-TCDD at
 levels ranging from none detected «0.2 pg/g)  to 85 pg/g (TMN 1987).  No
 2,3,7,8-TCDD was found  in rice, soybean,  and crawfish samples from
Arkansas and Louisiana  at a detection limit of 10 pg/g, or in Canadian
chicken and pork samples at a detection limit of 2 to 4 pg/g (Ryan et
al.   1985a, Firestone et  al. 1985). As a result of the present day
bleaching processes used in the pulp and paper industry,  trace amounts
of 2,3,7,8-TCDD have been detected in paper products.  Preliminary
results indicate that 2,3,7,8-TCDD is present at 3 to 4 pg/g in certain
samples of paper towels  and 13 to 39 pg/g in samples of communication
papers, such as  stationery, computer papers,  and envelopes (TMN 1987).

     A model to estimate 2,3,7,8-TCDD intake by humans from inhalation
of air and ingestlon of drinking water and food predicted that the food
chain accounts  for  98% of the  dally human intake  of 2,3,7,8-TCDD (Travi
and Hattemer-Frey 1987). Connect and Webster (1987)  also concluded that
food could be  the predominant  source of 2,3,7,8-TCDD Intake by far,

-------
                      Table 7.1.  Levels of 2,3,7,8-TCDD in soil from different locations
            Site
       Sample history
TCDD concentration**
       (ng/g)
Love Canal, N.Y.
Jacksonville, Ariz.
Midland, Mich.
St. Louis, Mo.

Shenandoah Stables, Mo.
Soils outside the dump site
Waste disposal site
Inside DOW facility
Urban sample of no obvious
source of contamination
Contaminated by waste oil
 ND (0.001 -0020)
       ND-29
       0.01 -52
       0.12

       101-33,000
Timberlme Stables, Mo
Bliss Farm, Mo.
Bubbling Springs Ranch, Mo.
Minker Resident, Mo.
Times Beach, Mo
Urban areas, Umled Slates
New Jersey
New Jersey
Lansing, Mich
Gaylord. Miss
Detroit, Mich
Contaminated by waste oil
Contaminated by waste oil
Contaminated by waste oil
Contaminated by waste oil
Contaminated by waste oil
Urban samples of no obvious
source of contamination
Spillage of 2,4,5-TCP
still bottom
Scrap yard where used reactor
vessels were collected
Urban sample
Urban sample
Urban sample
30-42
382f
76-95
50'
44-317
<0.0002-0 009
26.000f
1,100'
ND(00007) 0003
ND(00002)
00021 00036
                                                                                          References
EPA I985b
tPA I985b
Neslncket al  1986
EPA I985b

Tiernan et al  1985,
Kimbrough el al
1977
Tiernan et al  1985
Tiernan et al  1985
Tiernan el al  1985
Tiernan el al  1985
Tiernan et al  1985
Nestnck et al  1986

Jackson el al  1986

Jackson et al  1986

Nolrick cl al  1986
Neslnck el al  1986
Neslnck el jl  1986
                                                                                                                            O
                                                                                                                            rt
                                                                                                                            n
                                                                                                                            »-.
                                                                                                                            Di
                                                                                                                           I
                                                                                                                            o
                                                                                                                            in
                                                                                                                            c

-------
                                                                                                                           CO
                                                                                                                           o
                                         Table 7.1 (continued)
Site
Chicago. III.
Akron, Ohio
Nashville, Tenn.
Pittsburgh, Pa.
Philadelphia, Pa.
Brooklyn, N.Y.
Arlington. Va.
Sample history
Urban sample
Urban sample
Urban sample
Urban sample
Urban sample
Urban sample
Urban sample
TCDD concentration"-*
(ng/g)
0.0042-0.0094
0.0063
0.0008
0.0026
0.0009
0.0026
ND(0.0003)
References
Nestnck et al. 1986
Nestrick et al. 1986
Nestrick et al. 1986
Nestrick et al. 1986
Nestrick et al. 1986
Nestrick et al. 1986
Nestrick et al. 1986
"ND = not detected.

^Values within parentheses are detection limits.

'Only one sample was analyzed.
                                                                                                                           Co
                                                                                                                           n
                                                                                                                           o
                                                                                                                           rt

-------
                                       Potential for Human Exposure   81

compared with inhalation intake. Actual data on measured levels of
2,3,7,8-TCDD in food, however, are limited. Fat specimens fron three of
seven steers that were confined to a fenced pasture sprayed with 2,4,5-T
herbicide contained 3-4 ngAg 2,3,7,8-TCDD (Korcher et al. 1978).  This
result indicates that beef obtained from cattle grazing in contaminated
pastures (for example, those near municipal incinerators) may contribute
to 2,3,7,8-TCDD ingestion from contaminated food. Six samples of cow's
milk from various locations in Switzerland were analyzed for PCDDs.
Although sub-ppt (ng/kg) levels of 2,3,7,8-TCDD-substituted hepta-CDDs
were found in all samples, only three contained 2,3,7,8-TCDD at a
0.021-0.049 ngAg range (detection limit <0.013 ngAg) (Rappe et al.
1987). The daily intake of PCDDs by residents of Japan through
consumption of food has been estimated by several investigators (Ogaki
et al. 1987, Ono et al. 1987, Takizawa and Muto 1987). None of these
investigators, however, reported the detection of 2,3,7,8-TCDD in any
food.
     Since adipose tissue appears to be the chief contributor to the
body burden of 2,3,7,8-TCDD, many investigators analyzed fat tissue from
both exposed and control populations. That fat has the highest burden is
confirmed by the tissue analysis of a woman who died 7 months after the
accident in Seveso, Italy. The  following levels of 2.3,7,8-TCDD (pg/g)
were found in different organs: fat, 1,840; pancreas, 1,040; liver, 150;
thyroid, 85; brain, 60; lung, 60; kidney, 40; and blood, 6. The levels
of 2,3,7,8-TCDD in adipose tissue in certain segments of the general
population of the United States and Canada ranged from undetectable to
20 pg/g, with a mean value of 5 to 7 pg/g. In a U.S. National Human
Adipose Tissue Survey  (Stanley  et al. 1986), 2,3,7,8-TCDD was detected
with a frequency of 76%. In Europe,  the range of 2,3,7,8-TCDD
concentration in the adipose  tissue  in segments  of the general
population is not detectable  to 9 pg/g, with a mean value of 3 pg/g.
Instances of higher levels in adipose tissue have been reported in
individuals exposed to this chemical either during spraying herbicides
containing 2,4,5-T or  during  accidental capacitor or  transformer  fires.
For example, the adipose  tissue of a few exposed individuals in the
State Office Building  fire in Binghamton,  New York, had  2,3,7,8-TCDD
concentrations  in the  range  11.6  to  28.3 pg/g. with a mean value  of
17.4 pg/g. The  adipose tissue of  a  few heavily  exposed  individuals
involved in spraying  operations in Vietnam had  2,3,7,8-TCDD  levels
ranging from undetectable  (detection limit,  3 pg/g)  to  99 pg/g, with  a
mean value of 37 pg/g; however, no  difference  in the  level of  2,3,7,8-
TCDD was found  in lightly exposed,  possibly  exposed,  and other Vietnam
veterans who  sought  medical  help  compared  with  the  control population
group  (EPA  1986a; Graham et  al. 1986;  Gross  et  al.  1984;  Nygren  et al.
1986;  Patterson et al. 1987c;  Schecter et  al.  1985,  1986;  Stanley et  al.
1986;  Weerasinghe et al.  1986;  Young 1984).  The median concentrations of
2,3,7,8-TCDD  (concentration  range in parentheses)  in control and exposed
populations  in  Missouri have been reported to be 6.4 pg/g (1.4 to
20.2  pg/g)  and  17 pg/g (2.8  to 750 pg/g),  respectively (Patterson et  al.
1986).  The  levels  of 2,3,7,8-TCDD in human serum have been reported by
researchers  from Centers for Disease Control (Patterson et al. 1987b,
Patterson  et  al.).  On a lipid-adjusted basis,  the levels of 2,3,7,8-TCDD
 in human serum of certain members of the general population was reported
 to range  from 4 pg/g to 7.6  pg/g. Determination of 2,3,7,8-TCDD levels

-------
82   Section 7

in blood may be used as an indicator of body burden.  The advantage of
this method is that it requires a noninvasive technique for withdrawing
sample specimens.
     Human breast milk has also been analyzed for 2,3,7,8-TCDD.  It has
been reported that human breast milk is the largest contributor  to the
body intake of 2,3,7,8-TCDD in breast-fed babies (Rappe et al.  1986,
Schecter and Gasiewicz 1987).  The following levels (the detection limits
are given in parentheses) of 2,3,7,8-TCDD have been determined in human
breast milk from different countries: United States,  none detected (0.1
to 6.0 pg/g) in mothers from 2,4,5-T-exposed areas and in control areas;
none detected (<0.2 pg/g) and 0.29 pg/g in two mothers' milk collected
in 1986 from fiihghamton, New York; Canada, 0.17 pg/g in a single pooled
sample of 200 mothers' milk collected in 1980-81; Seveso, Italy, 2.3  to
28.0 pg/g from mothers near accident area; South Vietnam, none detected
(0.5 pg/g) to 40 to 50 pg/g from mothers in sprayed areas; Sweden, trace
to 2.3 pg/g, with a mean of 0.6 pg/g; and Germany, 1.3 to 3.3 pg/g, with
a mean of 1.9 pg/g. 2,3,7,8-TCDD was also detected in human milk
obtained from Denmark, the Netherlands, and Yugoslavia (EPA 1985b, Heath
et al. 1986, Jensen 1987, Nygren et al. 1986, Patterson et al.  1986,
Rappe et al. 1986, Schecter et al. 1987, Young 1984). There is a large
unexplainable difference in the values of 2,3,7,8-TCDD concentrations in
milk from South Vietnamese mothers analyzed by two groups of
investigators (Schecter et al. 1987, Young 1984).

     According to a statement by CDC (1986), "Contamination of breast
milk with trace amounts of a variety of chlorinated compounds should not
discourage women from breastfeeding except under unusual circumstances
which have to be evaluated on an individual basis. . . .Substitutes useo
for human milk are not entirely free of these compounds and may have
high metal levels. .  .  .Avoiding breastfeeding would deprive the infant
of immunological protection and psychological benefits for the infant
and mother."
     Other human tissues, obtained from the autopsy of two subjects in
Canada, were analyzed for 2,3,7,8-TCDD with the following results:
liver, none detected to 2.5 pg/g; muscle, none detected; and kidney,
none detected. The detection limit in these determinations were in the
range of 1 to 4 pg/g  (Ryan et al. 1985b). No 2,3,7,8-TCDD was detected
in the blood of exposed workers following the accident in Binghamton,
New York, at a detection limit of 1  to 2 pg/g (Schecter et al.  1985).
Serum 2,3,7,8-TCDD levels in veterans who were heavily exposed  to Agent
Orange in Vietnam during 1967-1968 have been reported by  the Centers  for
Disease Control  to range between none detected  (0.0013 pg/g) and
25 Pg/g, with a median value of 3.8  pg/g. The same study  reported  serum
2,3,7,8-TCDD levels in  a group of non-Vietnam veterans to range between
none detected  (0.0013 pg/g) and 12 pg/g, with a median value of
3-9 Pg/g- Serum  levels  in a group occupationally  exposed  to 2,3,7,8-TCDD
prior to 1970, however, have been reported  to be  30-fold  higher in the
same study  (MMWR,  1987). Using an environmental partitioning model to
estimate the concentration levels of 2,3,7,8-TCDD in various
environmental media, Travis and Hattemer-Frey  (1987) estimated  the
following average  daily intake  (with the  per cent of the  total  intake in
parenthesis): air, 0.001 ng (2%); water,  6.5 x  10'6  ng (<0.01%);  and
food, 0.046 ng  (98%).  It  is likely  that  the secondary  source of

-------
                                       Potential for Human Exposure   83

2,3,7,8-TCDD in foods is atmospheric emissions. EPA (1988c) estimated
the human exposure to 2,3,7,8-TCDD from a variety of exposure scenarios
resulting from contaminated soils, various land disposal situations,  and
municipal waste incineration. The highest exposure was attributed to  the
food chain, i.e., ingestion of contaminated fish, beef, dairy products,
and other foods. Ingestion of contaminated soil, especially by children
with pica tendencies; dermal contact with contaminated soil, dust,  and
sediment; and inhalation of contaminated dust and vapor further
contribute to human exposure. EPA (1988c) estimated an upper limit value
for the average 2,3,7,8-TCDD concentration in adipose tissue to be 6.72
ppt in the U.S. population. From this adipose tissue burden and
pharmacokinetic considerations, it was estimated that the upper bound
2,3,7,8-TCDD daily intake ranges from 0.04 to 0.51 pg/kg. These
estimates apply to ambient exposure and not to exposure scenarios
related to accidents.

7.3  OCCUPATIONAL EXPOSURES
     Occupational exposures to 2,3.7,8-TCDD occured in the past during
the production and use of hexachlorophene, trichlorophenol, and
herbicides containing 2,4,5-T. The potential for heaviest exposure is
likely to occur during the step that is used to purify 2,4,5-
trichlorophenol from its contaminants, since these products contain much
higher levels of 2,3,7,8-TCDD than the purified products. Detection of
the highest level of 2,3,7,8-TCDD (1.1 jig/wipe) in wipe samples taken
from different trichlorophenol production and purification areas of a
production facility also confirms this (Ott et al. 1987). Few data on
the occupational exposure to 2,3,7,8-TCDD during the manufacture of
these chemicals are available (Rappe 1984). The indirect evidence of
occupational exposure to 2,3,7,8-TCDD is the significantly higher
adipose tissue levels of the compound in heavily exposed Vietnam
veterans and in certain workers at the State Office Building in
Binghamton, New York, following the transformer fire. The adipose
tissues of nine workers from a former trichlorophenol manufacturer in
Missouri showed a mean 2,3,7,8-TCDD level of 246 pg/g compared with a
mean value of 8.7 pg/g for nonexposed workers at the same site. The
lipid-adjusted mean 2,3,7,8-TCDD  level in serum of the same exposed
workers was 363 pg/g- compared with a value of 47.1 pg/g  for nonexposed
workers  (Patterson et al.  in press).

7.4  POPULATIONS AT HIGH RISK
     From the monitoring data discussed  in Sects. 7.2  and  7.3,  it  is
possible to predict the segments  of the  general population  and  of
occupational groups that may be exposed  to higher levels of 2,3,7,8-
TCDD. Among the occupational groups, workers involved  in the production
or use of trichlorophenol  or its  salts,  hexachlorophene, and 2,4,5-T  or
other herbicides containing  2,4,5-T have the potential  for  exposure  to
higher levels of 2,3,7,8-TCDD  than the general  population.  2,4,5-T and
2,4,5-trichlorophenol and  its  salts, however,  are no  longer manufactured
in the United States  (SRI  1987).  Workers in  the wood  treatment  industry
have the potential for 2,3.7,8-TCDD exposure due  to  the  possibility  of
2,3,7,8-TCDD formation as  a  result of catalytic dechlorination  of  higher
chlorinated dibenzodioxins during pressure treatment  of wood with

-------
84   Section 7

pentachlorophenol or sodium pentachlorophenate (Hagenmaier and Brunner
1987). Workers in pulp and paper mills also have the potential for
exposure to 2,3,7,8-TCDD due to the occurrence of 2,3,7,8-TCDD in
bleached Kraft paper-making processes (Amendela 1987, Clement et al.
1987, Kuehl et al.  1987). Since both flue gases and ashes from municipal
and industrial incinerators contain 2,3,7,8-TCDD, workers in this
profession are likely to be at higher risk of exposure to 2,3,7,8-TCDD.
Populations residing near municipal incinerators may also be subjected
to exposure. Workers at sites of improper chemical waste disposal (from
trichlorophenol, hexachlorophene,  2,4,5-T, and associated industries)
and the general population residing near those sites are potentially
exposed to 2,3,7,8-TCDD. Breast-fed babies nursed by mothers residing
near improperly operated municipal incinerators or other sources of
exposure are expected to receive 2,3,7,8-TCDD through the milk.
     Studies in humans have not demonstrated that there is a sensitive
subpopulation. Animal studies, however,  suggest that the fetus and
newborn infants may represent such a sensitive population. As discussed
in Sects. 4.3.3 and 4.3.4 on developmental and reproductive toxicity,
2,3,7,8-TCDD is a demonstrated teratogen in rats and mice and also
results in spontaneous abortions and fetal death in monkeys. Since these
effects occur at low doses, and in the case of teratologic effects at
doses that do not appear to adversely affect the mother, it is likely
that, at certain stages of fetal development, the fetus represents a
sensitive subgroup. Animal data also demonstrate that toxic levels of
2,3,7,8-TCDD can be ingested during nursing and that lactation is a
major route for elimination of 2,3,7,8-TCDD.

-------
                                                                       85
                          8.  ANALYTICAL METHODS

      Several methods are available for the analysis of 2,3.7,8-TCDD in
 different media. Some of the more recent methods are given in Tables 8 1
 and 8.2.  The statement of work for organic analysis in the EPA Contract
 Laboratory Program does not list any method for 2,3,7,8-TCDD analysis
 (EPA 1987b). The methods listed in these tables are not exhaustive but
 are illustrative of a few recent methods.  Methodologies for collecting
 samples before their analysis are important,  since the concentrations of
 2,3,7,8-TCDD in most samples are low.  This is particularly important for
 stack samples that exist both in the vapor and particulate phase.  The
 details of stack-sampling methods are available in Velzy (1986)   Ozvacic
 (1986), and Brenner (1986).

      The  accuracy of analysis has increased in recent years with the
 availability of stable isotope-labeled (37C1  and 13C) 2,3,7,8-TCDD for
 use as  an internal standard in mass spectral  analysis.  With a
 combination of one of several methods  available for sample cleanup,
 high-performance gas chromatography (GC)  (HRGC),  and high-resolution
 mass  spectrometry (MS) (HRMS),  unequivocal identification and
 quantification of 2,3,7,8-TCDD can be  performed at very low levels.
 Although  negative chemical ionization MS  (NCI/MS)  shows a higher
 sensitivity to all other polychlorinated  dibenzo-p-dioxins than  electron
 impact  MS  (EI/MS),  it has a lower sensitivity for  2,3,7,8-TCDD (Buser et
 al.  1985).  The analysis of fly ash samples poses a special challenge
 because of poor solvent extraction recovery and the difficulty in  the
 resolution of 2,3,7,8-TCDD from a large number of  congeners present  in
 these samples.  Best results were obtained  by  using digestion with  excess
 dilute HC1,  followed by freeze-drying  of  the  residue and hot extraction
 with  toluene.  The HC1 treatment opens  the  pore structure of fly  ash  to
 permit access to the solvent,  and freeze-drying removes water to improve
 material  transfer from the hydrophilic surface of  the fly ash to the
 solvent (Stieglitz  et al.   1986).  Emphasis has been placed on the
 analysis of adipose tissue and  mother's milk,  because these two  tissues
 may be  indicators of the  human  body burden for 2,3,7,8-TCDD.  The
 analysis of fish is also  important since some bottom feeders (e.g.,
 channel catfish and carp)  and those fish that feed upon bottom feeders
 may be indicators of 2,3,7,8-TCDD-polluted water (Ryan  et al.  1984a.
 Jensen 1987).

     Besides  the commonly used  analytical  methods,  other newly developed
 but yet generally untested methods are available for the analysis  of
 2,3,7,8-TCDD.  Some  of these methods are GC with a  polymeric liquid
 crystal capillary column  (Natkwadi and Karasek 1986), GC with matrix
 isolation Fourier transform infrared spectrometry  (Wurrey et al. 1986).
 and HRGC with microwave- induced plasma detection (Mohamad et al. 1986)!
More detailed descriptions  of the  analytical  methods  for 2,3,7,8-TCDD

-------
86   Section 8
can be found in Buser et al. (1985),  Tieman et al.  (1985),  Rappe
(1984), and EPA (1985b).
8.1  ENVIRONMENTAL MEDIA
8.1.1  Air. Water. Soil, and Food
     See Table 8.1.
8.2  BIOMEDICAL SAMPLES
8.2.1  Fluids/Ezudates and Tissues
     See Table 8.2.

-------
                                                 Table 8.1.  Analytical method* for emlromntiilal samples
Sample matrix
Ambient room air
following accidental
transformer fire
Sample preparation
Vapor phase sample collected
by silica gel, paniculate
sample collected on glass fiber
Analytical method"
HRGC/HRMS
Detection limn
0003 pg/m1
Accuracy0
131 ± 27% al
J- 10 pg/m1
References
Smith el al 1986
Ambient outdoor air
Stack emission
Fly ash from
municipal incinerator
Water
filler, solvent extracted, and
precleaned by alumina and carbon

Vapor and paniculate collected
on polyurethane foam and glass
fiber, precleaned by Floruit
and modified silica
Vapor and particle collected on
polyurethane and glass fiber,
precleaned by silica and alumina

Vapor and panicle collected on
XAD-2 and  glass fiber by modified
EPA method S; solvent extracted
and precleaned by silica, alumina,
and Biobead

Solvent extraction,  HPLC separa-
tion on normal-phase and reverse-
phase column

Solvent extraction,  precleaning
by two adsorbent columns,  and
further fractionalion on reverse-
and normal-phase HPLC

Solvent extraction,  precleaned on
silica and  alumina

Sample passed through glass fiber
filler and adsorbent cartridge,
solvent extracted and precleaned
by acid alumina, graphatued
carbon and alumina
HRGC/NICI/MS




HRGC/NIEC/MS-SIM



HRGC/MS





GC/MS or HROC/FID



GC/LRMS



HRGC/LRMS


GC/MS
01-0 2 pg/m1 (method
detection limit loo high
for TCDD determination)


NR (TCDD not separated
from other telra isomers)


NR
NR (TCDD not separated
from other telra isomers)


40 pg
                                                                                     NR
                                                                                                                    NA        Oehme el al  1986
                                                                                                                    NA        Eiucr and Hues 1986
                                                                                                                    NR        Hagenmaier el al 1986
NR        Tong el al 1984,
           Tong and Karasek 1986


NR        Lamparski and
           Ncstnck 1980
                               NR        Buttr and Rappe 1980


                             88% at       O'Kccrcci al  1986
                            6 5 pg/L
                                            b
                                            K-.
                                           V!
                                                                                                                                                               s-
                                                                                                                                                               o
                                                                                                                                                               Q.
                                                                                                                                                               in

-------
                                                               TaMe 8.1 (coBtiaoed)
   Sample matrix
Waslcwatcr
Sod
Fiib
       Sample preparation
                                                           Analytical method"
                                                                 Detection limit
Sample spiked with an internal
standard is solvent extracted, extract
cleaned by column chromatography
Solvent extraction. KOH wash, pie-
cleaned in alumina, revened-
phase HPLC. and carbon
Solvent extraction. KOH wash, pre-
cleaned in chemicaUy treated
silica, basic alumina
Homogenized fillet digested with
ethanobc KOH and solvent
extracted; extract cleaned by
silica gel-supported HjSO4 column
and HPLC
Homogenized Fillet solvent
extracted, partitioned with con-
centrated H,SO«, cleaned
by Florisil
Homogenate digested with concen-
trated HCI and solvent extracted.
extract cleaned by silica gel-
supported H,SO,. chemically
treated silica and alumina, and
reverse-phase HPLC
 Digested with alkali, solvent
extracted, washed with concen-
 trated H]SO,. and cleaned up
 by ute exclusion chromatography.
 normal-  and reverse-phase HPLC
HRGC/LRMS or HRMS   0 2 mg/l
(EPA method 613)
                                                         HRGC/LRMS
                                                         HRGC/LRMS
                                                         HRGC/MS
                                                         HKGC/HRMS
                                                         HRGC/MS/MS
                                                          HRGC/EC
                          3ng/g
                                                                                   NR
                          5-IOpg/g
                          2-10 pg/g
                          <«Pg
                           I2pg/g
                                                    Accuracy"
92 4% at
2 5 mg/l


   NR
                                                                                                                NR
                                                                                                          References
                                                                                                                           EPA 1982
Donnelly el al. 1986
                                                                                                                           Freeman et al 1986
                                                                                                                                     oo
                                                                                                                                     CO
                                                                                                                                                           o

                                                                                                                                                           00
                                                                                                                 NR       Fehrmger el al 1985
                                                                                                                 NR       Ryan el al  1984
                                                                                                                 NR        Clement el al 1986
 105% at      Niemann 1986
I8-4J

-------

Sample main*
Fish and herring gull

Sample preparation
Solvent extracted, cleaned up by
potoitium ulicale-silica gel.
Table 8.1 (condone
Analytical method"
HRGC/LRMS
d)
Detection limn
l-8pg/g

Accuracy'
NR

References
Stalling el al 1983.
Rappc 1984
Fun, egg. or sediment
Chicken and pork
carbon, and HjSO,-silica gel cesium
silicate and alumina

Added HCI and solvent extracted.
extract cleaned by gel permeation,
tnsodium phosphate, H]SO,,
alumina and carbon columns

Fat and liver solvent extracted,
partitioned with concentrated
HjSO4. cleaned by Flonsil and
reverse-phase HPLC
HRGC/EC/LRMS
HRGC/MS/MS
NR
2-4pg/g
                                                                                                                   72%        Lawrence el al  1985
                                                                                                                    NR        Ryan el al  I985a
   "HRGC - High-resolution gas chromalography, HRMS  - high-resolution mass apcclromelry. NICI/MS - negative ion chemical lomzalion mass speclromelry,
NIEC/MS - negative ion electron impact mass spectrometry. SIM  - selective ion monitoring. MS - mass ipectrometry. GC  - gas chromatography. FID - flame
lomzation detector; LRMS - low-resolution mass speclrometry. EC - electron capture detector; NR - not reported. NA - not applicable
                                                                                                                                                               I
                                                                                                                                                               O
                                                                                                                                                               to
                                                                                                                                                               3:
                                                                                                                                                               n

-------
                                                Table 8.2.  Aoalyl
                                                  «hoda for Mosaedical samples
  Sample niftlru
Adipose tissue
          Sample preparation
                     Detection limit
Analytical method"       (pg/g)
                     Accuracy/
                    % Recovery"
     References
Sapomficalion with hoi alkali, solvcbt
eitraclion, cleaned with concentrated
HjSO,. alumina
Extraction with potassium oxalate and
mixture of solvents, cleaned by gel

Acidic digestion, solvent extraction
multiple cleanups with adsorbents and
chemically modified adsorbents.
normal- and reverse-phase HPLC

Tissue solvent extracted and subjected
to eight different preparations
Cleaned through potassium silicate/
silica gel. carbon. H^./silica
and alumina
Washed with concentrated  H,SO4 and
passed through silica, chemically
treated silica, alumina, reverse- and
normal-phase HPLC
Digested in concentrated HCI and
cleaned up by silica/H]SO,. alumina.
carbon/celite
Saponified with alkali, solvent
extracted, and cleaned up by alumina.
charcoal/silica
Saponified with alkali, solvent
extracted, washed with concentrated
H,SO4. and chromalographed on silica
acid and alumina
Washed with H,SO, and chromalographed
on Florisil
Washed with H,SO4, chromatographcd on
alumina, charcoal/celite, alumina
                                                             HRGC/HRMSand   0 1-0.6. 05-6
                                                             LRGC/HRMS
                                                              HRGC/LRMS
                                                              LRGC/LRMS
                                                              HRGC/LRMS


                                                              HRGC/LRMS



                                                              HRGC/LRMS


                                                              HRGC/LRMS


                                                              HRGC/HRMS



                                                              HRGC/CIMS


                                                              HRGC/HRMS
5 (fat basis)
                                                                                       05
                                                                                        
-------
                                                              Table 8.2 (i
Sample matrix
Adipose luiue
Sample preparation
Paued through ulica/potaMium silicate,
Analytical method"
HRGC/NICI/MS
Detection limit
(Pg/g)

Accuracy/
% Recovery"
90%
References

Blood, liver, kidney
and muscle


Serum
•ilka/carbon, potassium silicate/
H]SO,/«lKa and alumina

Automated extraction and enrichment        HRGC/MS or             <2
apparatus consisting of solvent ex-           HRGC/HRMS
traction, cleanup by carbon and
silicate/silica gel

Homogenized sample solvent extracted.       HRGC/MS/MS           1-4
cleaned up with HjSO,, chromatography.
or Ftonsil

Solvent extraction, concentrated             Isoiopic                I 25 X 10'
sulfuric acid wash and cleanup by           dilution with
carbon and silicate/silica gel. followed        HRGC/HRMS
by silicate/H,SO./silica gel and
alumina columns
                                                                                                     >8S% at 24 pg/g
                                                                                                    (internal standard)
                                                                                                           NR
 69% at I 2 pg/g
(external standard)
                      Lapeza et al  1986.
                      Patterson et al  I987a
                      Ryan el al I985b.c
Patterson el al  I987b
   "HRGC — High-resolution gas chromatography. LRGC — low-resolution  gas  chromalography.  NICI/MS  —  negative :or. chemicai  lunnaiion mass
spectrometry. HRMS — high-rssoluticn mass  spectrumeiry,  MS — mass speclrometry. LRMS  —  low-resolution mass spcctromelry. CIMS  —  chemical
lomzation mats speciromelry; NR - not reported
                                                                                                                                                           n
                                                                                                                                                           to
                                                                                                                                                          3:
                                                                                                                                                          n
                                                                                                                                                          3-
                                                                                                                                                          o
                                                                                                                                                          CL
                                                                                                                                                          bi

-------
                                                                      93
                   9.  REGULATORY AND ADVISORY STATUS

9.1  INTERNATIONAL
     No World Health Organization standards were found.  The World Health
Organization advisory developed by IARC is presented below.

9.2  NATIONAL

9.2.1  Regulat ions
     The reportable quantity (RQ) for 2,3,7,8-TCDD is 1 Ib, which places
2,3,7,8-TCDD in category X (EPA 1985c). EPA (1985d) has listed under
RCRA wastes containing dioxins as acute hazardous wastes, defined as
"wastes that are so hazardous that they may, either through acute or
chronic exposure, cause or significantly contribute to an increase in
serious irreversible, or incapacitating reversible illness regardless of
how they are managed." In addition, EPA (1986b) has prohibited,
effective November 8, 1986, further land disposal of certain dioxin-
containing hazardous wastes.

9.2.2  Advisory Guidance

9.2.2.1  Air
     No health advisories  (HAs) for levels  of 2,3,7,8-TCDD in air were
encountered.

9.2.2.2  Water

AGENCY                                ADVISORY

EPA     Drinking water advisories:
        1-day HA--1.0 x  10'6 mg/L (child)
        10-day HA--1.0 x 10'7 mg/L (child)
        Long-term  HA--1.0  x  10'8  mg/L (child)
        Long-term  HA--3.5  x  10'8  mg/L (adult)
        Drinking water equivalent level--3.5  x  10'8  mg/L
        10'4  to  10'7 excess  cancer risk--2.2  x  10'8  to  2.2 x 10'11
          mg/L  (EPA  1986c)

EPA     Ambient water quality  criteria
        10'4  to  10'7 excess  cancer risk--1.3  x  10'9  to  1.3 x 10'1Z  mg/L
        (EPA  1984)

-------
94   Section  9

9.2.2.3   Food

AGENCY                                ADVISORY

FDA     Levels  in  fish:  No  serious health concerns--<25 ppt (EPA 1985b)

NIOSH   2.3,7.8-TCDD  is  regarded as a potential occupational carcinogen,
        and occupational exposure to 2,3,7,8-TCDD should be controlled
        to the  fullest extent feasible (NIOSH 1984).

9.2.3  Data Analysis

9.2.3.1   Reference doses (RfDs)

     EPA  (1985a) calculated a chronic oral RfD for 2,3,7,8-TCDD based on
the data  from a three-generation study in rats by Murray et al. (1979),
as reanalyzed by Nisbet  and Paxton (1982). In this study, rats were
exposed to diets containing 2,3,7,8-TCDD at levels that provided doses
of 0.001, 0.01, and 0.1  ^g/kg/day. The highest dose resulted in
decreased fetal survival; the middle dose resulted in effects on litter
size and  fetal and neonatal survival. The lowest dose resulted in
dilated renal pelvises,  decreased fetal weight, and changes in the
gestational index. Therefore, the dose of 0.001 ;*gAg/day is a LOAEL,
and the RfD was calculated  as follows:

        RfD - (0.001 j*gAg/day)/(100)(10) - 0.000001 MgAg/day ,

     where:   0.001 /igAg/day - LOAEL,
              100 - uncertainty factor for inter- and intraspecies
                   extrapolation,
              10 - uncertainty factor for use of a LOAEL.

9.2.3.2   Carcinogenic potency, q *

     EPA  (1985b) has classified 2,3,7,8-TCDD in Group B2 when 2,3,7,8-
TCDD is considered alone, and in Group Bl when 2,3,7,8-TCDD is
considered in association with phenoxyherbicides and/or chlorophenols.
Group B2  indicates chat  although evidence in humans is. inadequate, there
are sufficient animal carcinogenicity data to consider 2,3,7,8-TCDD a
probable human carcinogen.  Group Bl indicates that there are not only
sufficient animal data but  also limited human data to support the
consideration that 2,3,7,8-TCDD, in conjunction with phenoxyherbicides
and/or chlorophenols, is a  human carcinogen. IARC (1982) has classified
2,3,7,8-TCDD  in Group 2B, which is analogous to the EPA classification
of Group  B2.  NIOSH (1984) recommended that 2,3,7,8-TCDD be considered a
potential occupational carcinogen and that exposure be limited to the
fullest extent feasible.

     EPA  (198Sa) developed  a quantitative unit cancer risk estimate
based on  the  study by Kociba et al. (1978a,b) and a reexamination of the
histologic evidence from that study conducted by Squire for the EPA. The
calculations  were based  on  the increased incidence of tumors of the
lungs,  liver, hard palate,  and nasal turbinates in female rats
maintained on diets containing 2,3,7,8-TCDD for 2 years. The two

-------
                                     Regulatory and Advisory Status   95

different pathologic examinations produced differences in tumor
incidence and, hence, slightly different q^* values. The final value
derived, 1.56 x 105 (mgAg/day) "1, was an average based on separate
calculations of the q.* values. EPA is in the process of considering a
revision in the risk estimate for 2,3,7,8-TCDD (EPA 1988a) .

9.2.3.3  Carcinogenic potency, methods used by other agencies

     Both the Centers for Disease Control (CDC) (Kimbrough et al. 1984)
and the Food and Drug Administration (FDA 1983) have calculated a
virtual safe dose for 2,3,7,8-TCDD, which corresponds to an excess
cancer risk of 10'^. The FDA calculations were based on the Kociba et
al. (1978a,b) study, whereas the CDC calculations were based on the
Squire evaluation of the Kociba et al. (1978a,b) study. Thus, human
intake values that correspond to an estimated 10'6 risk derived by EPA,
CDC, and FDA, respectively, are 6.4, 27.6, and 57.2 fgAg/day. EPA is in
the process of considering a revision in the risk estimate for 2,3,7,8-
TCDD (EPA 1988a).

9.3  STATE
     Regulations and advisory guidance from the states were not
available.

-------
                                                                      97
                            10.  REFERENCES
Abernathy DJ,  Greenlee WF, Hubard JC, Boreiko CJ.  1985. 2,3,7,8-
Tetrachlorodibenzo-p-dioxin (TCDD) promotes Che transformation of
C3H/10T1/2 cells. CareinogenesIs 6:651-653.

Adams W J,  Blaine KM. 1986. A water solubility determination of 2,3,7,8-
TCDD. Chemosphere 15:1397-1400.

Adams W J,  DeGraeve GM, Sabourin TD, Conney JD, Mosher CM. 1986. Toxicity
and bioconcentration of 2,3,7,8-TCDD to fathead minnows (Pimephales
promelas).  Chemosphere 15:1503-1511.

Albro PW,  Crummett WB, Dupuy AE, Jr., Gross ML, Hanson, M. 1985.  Methods
for the quantitative determination of multiple, specific polychlorinated
dibenzo-p-dioxin and dibenzofuran isomers in human adipose tissue
in the parts-per-trillion range: An interlaboratory study. Anal Chem
57(13):2717-2725.

Aldred JE.  1978. Report of the Consultative Council on Congenital
Abnormalities in the Yarrom District. Minister of Health, Melbourne,
Victoria,  Australia (cited in EPA 1985a).

* Allen JR, Barsotti DA, Van Miller JP, Abrahamson LJ, Lalich JJ. 1977.
Morphological changes in monkeys consuming a diet containing low levels
of 2,3,7,8-tetrachlorodibenzo-p-dioxin. Food Cosmet Toxicol 15:401-410.

Allen JR,  Barsotti DA, Lambrecht LK, Miller JP. 1979. Reproductive
effects of halogenated aromatic hydrocarbons on nonhuman primates. Ann
NY Acad Sci 320:419-425 (cited in EPA 1985a).

Amendela GA. 1987. The occurrence and fate of PCDDs and PCDFs  in
bleached Kraft papermaking processes. In: Proceedings of the Seventh
International Symposium on Chlorinated Dioxins and Related Compounds.
Las Vegas, Nev: University of Nevada.

Anonymous. 1985. Dioxins  in the environment: No consensus on human
hazard. Chem Eng News, May 27, pp. 41-44.

Aust SD. 1984. On the mechanism of anorexia  and toxicity of TCDD and
related compounds. Banbury Rep. Vol  18. Iss  Biol Mech Dioxin Action,
pp.  309-319.
* Key Studies.

-------
98   Section 10

Axelson 0, Sundell L, Anderson K, Edling C,  Hogstedt C,  Kling H.  1980.
Herbicide exposure and tumor mortality: An updated epidemiologic
investigation on Swedish railroad workers.  Scand J Work Environ Health
6:73-79 (cited in SPA 1985a).

Balarajan R. Acheson ED. 1984. Soft tissue sarcoma in agriculture and
forestry workers. J Epidemiol Community Health 38:113-116.

Barnes DG. 1983. Chlorinated Dioxins Work Group. EPA, Washington, DC
12(4-5):645-655.

Barnes D, Bellini J, DeRosa C, et al. 1987.  Reference dose (RfD):
Description and use in health risk assessments.  Appendix A in Integrated
Risk Information System Supportive Documentation, Vol I. Washington, DC:
EPA, Office of Health and Environmental Assessment. EPA 600/8-86-032a.

Bauer H, Schulz KH, Spiegelberg A. 1961. Berufliche vergiftungen bei der
herstellung von chlorphenol-verbindunger. Arch Gewerbeath Gewerbehyg
18:538-555. (In German) (cited in EPA 1984).

Beatty PW, Neal RA. 1976. Evidence for a role for DT-Diaphorase
induction in the toxicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin.
J Pharmacol 18(2):211 (cited in EPA 1985a).

Berry DL, DiGiovanni J, Juchau MR, Bracken WM, Gleason GL, Slaga TJ.
1978. Lack of tumor-promoting ability of certain environmental chemicals
in a two-stage mouse skin tumorigenesis assay. Res Commun Chem Pathol
Pharmacol 20(1):101-108 (cited in EPA 1985a).

Berry DL. Slaga TJ, DiGiovanni J, Juchau MR. 1979. Studies with
chlorinated dibenzo-p-dioxins, polybrominated biphenyls and
polychlorinated biphenyls in a two-stage system of mouse skin
tumorigenesis: Potent anticarcinogenic effects.  Ann NY Acad Sci
320:405-414 (cited in EPA 1985a).

Birnbaum LS. 1986. Distribution and excretion of 2,3,7,8-
tetrachlorodibenzo-p-dioxin in congenic strains of mice which differ at
the Ah locus. Drug Metab Dispos 14(1):34-40.

Bimbaum LS, Weber H, Harris MW, Lamb IV, JC, McKinney JD. 1985. Toxic
interaction of specific polychlorinated biphenyls and 2,3,7,8-
tetrachlorodibenzo-p-dioxin: Increased incidence of cleft palate in
mice. Toxicol Appl Pharmacol 77:292-302.

Birnbaua LS, Harris MW, Miller CP, Pratt RM, Lamb IV, JC. 1986.
Synergistic interaction of 2,3,7,8-tetrachlorodibenzo-p-dioxin and
hydrocortisone  in the induction of cleft palate  in mice. Teratology
33:29-35.

Bisanti L, Bonetti F, Caramaschi F, et al.  1980. Experiences  from the
accident of Seveso. Acta Morphol Acad Sci Hung  28(1-2):139-157 (cited  in
EPA 1985a).

-------
                                                         References   99

Bogen G. 1979. Symptoms of Vietnam veterans exposed to Agent Orange.
JAMA 242(22):2391 (cited in EPA 1985a).

Bonaccorsi A, FaneHi R, Tognoni G. 1978. In the wake of Seveso. Ambio
7(5-6) :234-239 (cited in EPA 1985a).

Brenner KS.  1986. Pu-foam-plug technique and extractive co-distillation
(Bleidner apparatus), versatile tools  for stack emission sampling and
sample preparation. Chemosphere 15:1917-1922.

Bronzetti G, Zeiger E, Lee I, Suzuki K, Mailing HV. 1983. Mutagenicity
study of TCDD and ashes from urban incinerator "in vitro" and "in vivo"
using yeast  D7 strain. Chemosphere 12:549-553 (cited in EPA 1985a).

Bumpus JA, Tien M, Wright D, Aust SD.  1985. Oxidation of persistent
environmental pollutants by a white rot fungus. Science 228(4706):1434-
1436.

Buser HR. 1979. Formation and identification of tetra- and
pentachlorodibenzo-p-dioxins from photolysis of two isomeric
hexachlorodibenzo-p-dioxins. Chemosphere 4:251-257.

Buser HR, Rappe C. 1980. High-resolution gas chromatography of the 22
tetrachlorodibenzo-p-dioxin isomers. Anal Chem 52:2257-2262.

Buser HR, Rappe C. 1984. Isomer-specific separation of 2,3,7,8-
substituted  polychlorinated dibenzo-para-dioxins by high-resolution
gas-chromatography-mass-spectrometry,  Anal Chem 56(3):442-443.

Buser HR, Rappe C, Gergqvist PA. 1985. Analysis of polychlorinated
dibenzofurans, dioxins and related compounds in environmental samples.
Anal Chem 60:293-301.

Buu-Hoi NP,  Pham-Huu-Chanh, Sesque G,  Azum-Gelade MC, Saint-Ruf G. 1972.
Organs as targets of dioxin (2,3,7,8-tetrachlorodibenzo-p-dioxin)
intoxication. Naturwissenschaften 59(4):174-175 (cited in EPA 1985a).

Cantor KP. 1982. Fanning and mortality from non-Hodgkin's lymphoma: A
case-control study. Int J Cancer 29:239-247.

CDC (Centers for Disease Control). 1986. Centers for Disease Control
statement on breastfeeding. Vet Human  Toxicol 28:582-583.

CDC. 1987. Comparison of Serum Levels  of 2,3,7,8-TCDD with Indirect
Estimates of Agent Orange Exposure in  Vietnam Veterans. Final Report
Agent Orange Projects. Centers for Disease Control, Public Health
Service, U.S. Department of Health and Hunan Services, Atlanta, Ga.

Choudhry GG, Hutzinger 0. 1982. Photochemical formation and degradation
of polychlorinated dibenzofurans and dibenzo-p-dioxins. Res Rev 84:113-
161.

-------
100   Section 10

Clement RE, Bolelie B,. Taguchi, V. 1986.  Comparison of instrumental
methods for chlorinated dibenzo-p-dioxin (CDD)  determination:  Interim
results of a round-robin study involving GC-MS,  MS-MS and high
resolution MS. Chemosphere 15:1147-1156.

Clement RE, Tashero C, Reiner E, Hoilinger D.  1987.  Chlorinated
dibenzoflurans (CDFs) in effluents and sludges  from pulp and paper
mills. In: Proceedings of the Seventh International Symposium on
Chlorinated Dioxins and Related Compounds. Las  Vegas, Nev: University of
Nevada.

Cohen CM, Bracken WM, Iyer RP, Berry DL,  Selkirk JK, Slaga TJ.  1979.
Anticarcinogenic effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin on
benzo(a)pyrene and 7,12-dimethylbenz(a)anthracene tumor initiation and
its relationship to DNA binding. Cancer Res 39(10):4027-4033 (cited in
EPA 1985a)

Connett P, Webster T. 1987. An estimation of the relative human exposure
to 2,3,7,8-TCDD emissions via inhalation and ingestion of cow's milk.
Chemosphere 16:2079-2084.

Cook RR, Bond GG, Olson RA, Ott MG, Gondek MR.  1986. Evaluation of  the
mortality experience of workers exposed to the chlorinated dioxins.
Chemosphere 15:1769-1776.

Courtney KD. 1976. Mouse teratology studies with chlorodibenzo-p-
dioxins. Bull Environ Contain Toxicol 16(6):674-681  (cited in EPA 1985a)

Czeizel E, Kiraly J. 1976. Chromosome examinations  in workers producing
Klorinol and Buminol. In: L. Banki, ed. , The Development of a Pesticide
as a Complex Scientific Task. Medicina: Budapest. 239-256 (cited in EPA
1985a).

Czuczwa JM, Hites RA. 1984. Environmental  fate of combustion-generated
polychlorinated dioxins and furans. Environ Sci Technol 18(6):444-450.

Czuczwa JM, Hites RA. 1986. Airborne dioxins and dibenzofurans: Sources
and fates. Environ Sci Technol  20(2):195-200.

*  DeCaprio AP, McMartin DM, O'Keefe PW, Rej R, Silkworth  JB, Kaminsky
LS. 1986.  Subchronic oral  toxicity of  2,3,7,8-tetrachlorodibenzo-p-
dioxin in the guinea pig:  Comparisons  with a PCB-containing transformer
fluid pyrolysate.  Fundam Appl  Toxicol  6:454-463.

Dencker L, Pratt RM.  1981. Association between  the  presence of the Ah
receptor in  embryonic murine  tissues and  sensitivity to TCDD-induced
cleft palate. Teratogenesis Carcinog Mutagen 1:399-406.

Dencker L, Hassoun E, D'Argy  R,  Aim G.  1985. Fetal  thymus organ culture
as an in vitro  model for the  toxicity  of  2,3,7,8-TCDD and its  congeners.
Mol Pharmacol 27:133-140.

-------
                                                        References   101

Department of Health, New Zealand. 1980. Report to the Minister of
Health of an investigation  into allegations of an association between
human congenital defects and 2,4,5-T spraying in and around teKuiti.  New
Zealand Ned J 314-315 (cited in EPA 1985a).

des Rosiers PE. 1986. Methodologies for materials contaminated with
PCDDs and related compounds. Chemosphere 15:1513-1528.

DiLemia R, Crimaudo C, Pacchetti G. 1982. The study of X-rays and TCDD
effects on satellite associations may suggest a simple model for
application in environmental mutagenesis. Hum Genet 61(l):42-47 (cited
in EPA 1985a).

Donnelly JR, Vonnahme TL, Hedin CM, Niedenhut VJ. 1986. Evaluation of
RCRA method 8280 for analysis of dioxins and dibenzofurans.  Rappe C,
Choudhry G, Keith LH, eds. Chlorinated Dioxins and Dibenzofurans in
Perspective. Chelsea, Mich: Lewis Publishers, Inc., pp. 399-435.

Eitzer BD, Hites RA. 1986. Concentrations of dioxins and dibenzofurans
in the atmosphere. Int J Environ Anal Chem 27(3):215-230.

EPA (Environmental Protection Agency). 1979a. Report of Assessment of a
Field Investigation of Six-Year Spontaneous Abortion Rates in Three
Oregon Areas in Relation to Forest 2,4,5-T Spray Practice. Office of
Toxic Substances (cited in EPA 1985a).

EPA (Environmental Protection Agency). 1979b. 2,4,5-T and Silvex.
Introduction to suspensions and notice of intent to cavell.  Fed Regist
15874.

EPA (Environmental Protection Agency). 1982. Test Method: 2,3,7,8-
Tetrachlorodibenzo-p-Dioxin--Method 613. Cincinnati, Ohio: Environmental
Monitoring and Support Laboratory.

EPA (Environmental Protection Agency). 1984. Ambient Water Quality
Criteria Document for 2,3,7,8-Tetrachlorodibenzo-p-Dioxin. EPA-440/5-
84-007.

EPA (Environmental Protection Agency). 1985a. Drinking Water Criteria
Document for 2,3,7,8-Tetrachlorodibenzo-p-Dioxin. Cincinnati, Ohio:
Environmental Criteria and Assessment Office. EPA Report No. 600/X-84-
194-1.

EPA (Environmental Protection Agency). 1985b. Health Assessment Document
for Polychlorinated Dibenzo-p-Dioxins. Washington, DC: Office of Health
and Environmental Assessment. EPA Report No. 600/8-84-014.

EPA (Environmental Protection Agency). 1985c. Notification requirements:
Reportable quantity adjustments. Fed Regist 50:13457.

EPA (Environmental Protection Agency). 1985d. Hazardous waste management
system: Dioxin-containing wastes. Fed Regist 50:1978.

-------
102   Section 10

EPA (Environmental Protection Agency). 1986a.  Broad Scan Analysis of the
FY82 National Hunan Adipose Tissue Survey Specimens.  Vol. IV.
Polychlorinated Dibenzo-p-Dioxins (PCDD) and Polychlorinated
Dibenzofurans (PCDF). Washington, DC: Office of Toxic Substances. EPA
Report No. 560/5-86-038.

EPA (Environmental Protection Agency). 1986b.  Hazardous waste  management
system: Land disposal restrictions.  Fed Regist 51:40572.

EPA (Environmental Protection Agency). 1986c.  2,3,7,8-Tetrachloro-
dibenzo-p-dioxins. Health advisory.  Washington, DC: Office of  Drinking
Water. October 16, 1986. Draft.

EPA (Environmental Protection Agency). 1987a.  Interim Procedures for
Estimating Risks Associated with Exposures to Mixtures of Chlorinated
Dibenzo-p-Dioxins and Dibenzofurans (CDDs and CDFs).  Risk Assessment
Forum. Washington, DC. EPA Report No. 625/3-87/012.

EPA (Environmental Protection Agency). 1987b.  EPA Contract Laboratory
Program. Statement of work for organic analysis.  Washington, DC.

EPA (Environmental Protection Agency). 1988a.  A Cancer Risk-Specific
Dose Estimate for 2,3,7,8-TCDD. Washington, DC: Office of Research and
Development, Office of Health Effects Assessment. EPA Report No. 600/6-
88/007Aa.  Draft.

EPA (Environmental Protection Agency). 1988b.  A Cancer Risk-Specific
Dose Estimate for 2,3,7,8-TCDD. Appendices A through F. Washington, DC:
Office of Research and Development,  Office of Health Effects Assessment.
EPA Report No. 600/6-88/007Ab. Draft.

EPA (Environmental Protection Agency). 1988c.  Estimating Exposure to
2.3,7,8-TCDD. Washington, DC: Office of Research and Development, Office
of Health Effects Assessment. EPA Report No. 600/6-88-005A. Draft.

Erickson JD, Mulinare J, McClain SPW, et al. 1984.  Vietnam veterans:
Risks for fathering babies with birth defects. JAMA 252(7):903-912.

Eriksson M, Hardell L, Berg NO, Holler T. Axelson 0. 1979. Case-control
study on malignant mesenchymal tumors of the soft tissue and exposure to
chemical substances. Lakartidningen 76:3872-3875 (cited  in EPA  1985a).

Eriksson M, Hardell L, O'Berg N, Holier T, Axelson 0. 1981. Soft-tissue
sarcomas and exposure to chemical substances: A case-referent study. Br
J Ind Med 38:27-33 (cited in EPA 1985a).

Evans RG, Webb KG, Knutsen AP, et al. 1988. A medical follow-up  of the
health effects of long-term exposure to 2,3,7,8-TCDD. Arch Environ
Health (in press).

-------
                                                        References   103

Fairless BJ, Bates DI, Hudson J, Kleopfer RD, et al. 1987. Procedures
used to measure the amount of 2,3,7,8-tetrachlorodibenzo-p-dioxin in the
ambient air near a Superfund site cleanup operation. Environ Sci Technol
21:550-555.

Falk H, Stehr PA, Stein GF, et al.  1984. A pilot epidemiology study of
health effects due to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)
contamination in Missouri. Banbury Rep. Vol 18, Iss Biol Mech Dioxin
Action, pp. 447-460.

FDA (Food and Drug Administration). 1983. Statement by SA Miller,
Director Bureau of Foods, FDA. Testimony before the Subcommittee on
Natural Resources, Agriculture, Research and Environment, U.S. House of
Representatives, June 30 (cited in Hiremath et al. 1986).

Fehringer NV, Walter SM, Kozara RJ, Schneider LF. 1985. Survey of
2,3,7,8-tetrachlorodibenzo-para-dioxin in fish from the Great Lakes and
selected Michigan rivers. J Agric Food Chem 33(4):626-630.

Fett MJ, Dunn M, Adena MA, O'Toole BI, Forcier L. 1984. Australian
veterans health studies: The mortality report. Part I: A retrospective
cohort study of mortality among Australian national servicemen of the
Vietnam conflict era, and an executive summary of the mortality report.
Canberra, Australia: Australian Government Publishing Service (cited in
EPA 1988b).

Field B, Kerr C. 1979. Herbicide use and incidence of neural tube
defects. Lancet 1(8130):1341-1342 (cited in EPA 1985a).

Fingerhut MA, Halperin WE, Honchar  PA, Smith AB, Groth DH, Russell WO.
1984. An evaluation of reports of dioxin exposure and soft tissue
sarcoma pathology among chemical workers in the United States. Scand J
Work Environ Health 10:299-303.

Firestone D, Ress J, Brown NL, et al. 1972. Determination of
polychlorodibenzo-p-dioxins and related compounds in commercial
chlorophenols. J Assoc Off Anal Chem 55:85-92.

Firestone D, Niemann RA, Schneider  LF, Gridley JR, Brown DE. 1985.
Dioxin residues in fish and other foods. Abstracts of papers of  the
American Chemical Society 189:4.

Forsberg B, Nordstrom S. 1985. Miscarriages around a herbicide
manufacturing company in Sweden. Ambio 14(2):110-111.

Freeman RA. Schroy JM.  1986. Modeling the transport of 2,3,7,8-TCDD and
other low volatility chemicals in soils. Environ  Progress 5(l):28-33.

Freeman RA. Schroy JM. Hileman FD,  Noble RW.  1986. Environmental
mobility of 2,3,7,8-TCDD and comparison chemicals in a roadway  soil
matrix. Rappe C, Choudhary G, Keith LH, eds.  Chlorinated Dioxins  and
Dibenzofurans in Perspective. Chelsea, Mich:  Lewis Publishers,  Inc.,  pp.
171-183.

-------
104   Section 10

Friedman JH. 1984. Does Agent Orange cause birth defects? Teratology
29:193-221.

Fries GF, Marrow GS. 1975. Retention and excretion of 2,3,7,8-
tetrachlorodibenzo-p-dioxin by rats. J Agric Food Chem 23(2):265-269
(cited in EPA 1985a).

Fuerst P, Meemken HA,  Groebel W.  1986. Determination of polychlorinated
dibenzodioxins and dibenzofurans in human milk.  Chemosphere 15:1977-
1980.

Gasiewicz TA, Olson JR. Geiger LE,  Neal RA. 1983a. Absorption,
distribution and metabolism of 2,3,7,8-tetrachlorodibenzo-p-dioxin
(TCDD) in experimental animals. In: Human and Environmental Risk of
Chlorinated Dioxins and Related Compounds, Tucker RE, Young AL and Gray
AP, eds. New York: Plenum Press 495-525 (cited in EPA 1985a).

Gasiewicz TA, Geiger LE, Rucci G, Neal RA. 1983b. Distribution,
excretion, and metabolism of 2,3,7,8-tetrachlorodibenzo-p-dioxin in
C57BL/6J, DBA/2J, and B6D2F1/J mice. Drug Metab Dispos 11(5):397-403.

Geiger LE, Neal RA. 1981. Mutagenicity testing of 2,3,7,8-
tetrachlorodibenzo-p-dioxin in histidine auxotrophs of Salmonella
cyphimurium. Toxicol Appl Pharmacol 59(1):125-129 (cited in EPA 1985a).

Geyer HJ, Scheunert I, Fiser JG, Korte F. 1986. Bioconcentration
potential (BCP) of 2, 3,7,8-tetrachlorodibenzo-p-dioxin (2,3,7,8-TCDD)
terrestrial organisms including humans. Chemosphere 15:1495-1502.

Gilbert  P, Saint-Ruf G,  Poncelet F, Mercier M. 1980. Genetic effects of
chlorinated anilines and azobenzenes on Salmonella typhimurium. Arch
Environ  Contain Toxicol 9(5):533-541 (cited in EPA 1985a).

Giri AK. 1986. Mutagenic and genotoxic effect of 2,3,7,8-
tetrachlorodibenzo-p-dioxin, a review. Mutat Res 168:241-242.

Graham M, Hileman FD, Orth RG, Vendling JM, Wilson JV. 1986.
Chlorocarbons in  adipose tissue  from a Missouri population. Chemosphere
15:1595-1600.

Green S, Moreland FS.  1975. Cytogenetic evaluation of  several  dioxins  in
the  rat. Toxicol  Appl  Pharmacol  33:161  (cited in  EPA 1985a).

Green S, Moreland,  Sheu  C. 1977. Cytogenic effect of 2,3,7,8-
tetrachlorodibenzo-p-dioxin on rat bone marrow cells.  Washington.  DC:
Food and Drug Administration.  FDA  By-Lines 6:292  (cited  in EPA 1985a).

Greenlee WF, Osborne R,  Hudson LG,  Toxcano WA Jr. 1984.  Studies on the
mechanisms  of toxicity of TCDD to  human epidermis. Banbury Rep 18:365-
372.

-------
                                                        References   105

Greenlee WF, Dold  KM,  Irons RD, Osborne R.  1985. Evidence for direct
action of  2,3,7,8-TCDD on  thymic  epithelium. Toxicol Appl Pharmacol
79:112-120.

Greenwald  P, Kovasznay B,  Collins D, Therriault G. 1984. Sarcomas of
soft tissues after Vietnam service. J Natl  Cancer Inst 73:1105-1109.

Greig JB.  1979. The  toxicology of 2,3,7,8-tetrachlorodibenzo-p-dioxin
and its structural analogues. Ann Occup Hyg 22:411-420 (cited in EPA
1985a).

* Greig J. 1984. Differences between skin and liver toxicity of
2,3,7,8-tetrachlorodibenzo-p-dioxin in mice. Banbury Rep 18 (Biol Mech
Dioxin Action):391-397.

* Gross ML, Lay Jr,  JO, Lyon PA, et al. 1984. 2,3,7,8-
Tetrachlorodibenzo-p-dioxin levels in adipose tissue of Vietnam
veterans.  Environ  Res  33:261-268.

Hagenmaier H. 1986.  Determination of 2,3,7,8-TCDD in commercial
chlorophenols and  related  products. Fresenius Z Anal Chem 325:603-606.

Hagenmaier H, Brunner  H. 1987. Isomer specific analysis of
pentachlorophenol  and  sodium pentachlorophenate on 2,3,7,8-substituted
PCDD and PCDF at sub-ppb levels. Chemosphere 16:1759-1764.

Hagenmaier H, Kraft  M,  Jager V, Mayer U, Lutzke K. Siegel D. 1986.
Comparison of various  sampling methods for  PCDDs and PCDFs in stack gas.
Chemosphere 15:1187-1192.

Hanify JA, Metcalf P,  Nobbs CL, Uorsley RJ. 1981. Aerial spraying of
2,4,5-T and human  birth malformations: An epidemiological investigation.
Science 212:349-351  (cited in EPA 1985a).

Hanson DJ. 1987. Science failing to back up veteran concerns about Agent
Orange. Chem Eng News.  November 9. pp. 7-11,14.

Hardell L, Eriksson  M.  1988. The association between soft tissue
sarcomas and exposure  to phenoxyacetic acids, a new case referent study.
Cancer 62:652-656.

Hardell L, Eriksson  M,  Lenner P. 1980. Malignant lymphoma and exposure
to chemical substances, especially organic  solvents, chlorophenols and
phenoxy-acids. Lakartidningen 77:208-210 (cited in EPA 1985a).

Hardell L, Eriksson  M,  Lenner P, Lundgren E. 1981. Malignant lymphoma
and exposure to chemicals, especially organic solvents, chlorophenols
and phenoxy acids: A case-control study. Br J Cancer 43:169-176 (cited
in EPA 1985a).

Hardell L, Standstrom  A. 1979. Case-control study: Soft-tissue sarcomas
and exposure to phenoxyacetic acids or chlorophenols. Br J Cancer
39:711-717 (cited  in EPA 1985a).

-------
106   Section 10

Hay A. 1982. Toxicology of dioxins. In:  The Chemical Scythe:  Lessons of
2,4,5-T and Dioxin. New York, London:  Plenum Press,  41-47 (cited in EPA
1985a).

Heath RG, Harless RL, Gross ML, et al. 1986.  Determination of 2,3,7,8-
tetrachlorodibenzo-p-dioxin in human milk at the 0.1-10 parts-per-
trillion level: Method validation and survey results.  Anal Chem (USA)
58(2):463-468.

Henck JW, New MA, Kociba RJ, Rao KS. 1981. 2,3,7,8-Tetrachlorodibenzo-
p-dioxin: Acute oral toxicity in hamsters. Toxicol Appl Pharmacol
59:405-407  (cited in EPA 1985a).

Hiremath C, Bayliss D, Bayard S. 1986. Qualitative and quantitative
cancer risk assessment of 2,3,7,8-tetrachlorodibenzo-p-dioxin (2,3,7.8-
TCDD). Chemosphere 15:1815-1823.

Hoffman RE, Stehr-Green PA, Vebb KB, et al. 1986.  Health effects of
long-term exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. JAMA
255:2031-2038.

Holden C. 1979. Agent Orange furor continues to build. Science 205:77-
772 (cited  in EPA 1985a).

Holmstedt,  B. 1980. Prolegomena to Seveso, Ecclesiastes 1:18. Arch
Toxicol 44(4):211-230 (cited in EPA 1985a).

HSDB  (Hazardous Substances Data Bank). 1987. National Library of
Medicine. On-line printout. April 6,  1987.

Huetter R,  Philippi M. 1982. Studies  on microbial metabolism of TCDD
under laboratory conditions. Pergamon Ser  Environ Sci 5:87-93 (cited  in
EPA 1985a).

Hungerford  CM.  1988. Comments on behalf of Syntex Agribusiness Inc.,  in
response  to Draft Toxicological Profile for 2,3,7,8-Tetrachlorodibenzo-
p-dioxin. Feb.  11, 1988.

Hussain  S,  Ehrenberg L, Lofroth G,  Gejvail T. 1972. Mutagenic effects of
TCDD  on  bacterial  systems. Ambio 1:32-33  (cited in  EPA  1985a).

IARC  (International  Agency  for Research on Cancer). 1982. World Health
Organization-IARC  Monographs on the Evaluation of the Carcinogenic  Risk
of Chemicals  to Humans. Lyons,  France: WHO; IARC Vol. 1-29,  Suppl 4
238-243.

Ideo  G,  Bellati G, Bellobuono  A, Mocarelli P, Marocchi  A, Brambilla P.
1982. Increased urinary D-glucaric acid excretion by  children living in
an area  polluted with tetrachlorodibenzoparadioxin  (TCDD). Clinica
Chimica  Acta  120:273-283.

-------
                                                        References   107

Isenee AR, Jones GE. 1971. Absorption and translocation of root and
foliage applied 2,4-dichlorophenol, 2,7-dichlorodibenzo-p-dloxin, and
2,3,7,8-tetrachlorodibenzo-p-dioxin. J Agr Food Chem 19:1210-1214.

Jackson DR, Roulier MH, Grotten HM, Rust SW, Warner, JS. 1986.
Solubility of 2,3,7,8-TCDD in contaminated soils. In: Rappe C, Choudhary
G, Keith LH, eds. Chlorinated Dioxins and Dibenzofurans in Perspective.
Chilsea, Mich: Lewis Publishers, Inc.; pp. 185-200.

Jensen AA. 1987. Polychlorobiphenyls (PCBs), polychlorodibenzo-p-dioxins
(PCDDs) and polychlorodibenzofurans (PCDFs) in human milk,  blood and
adipose tissue. Sci Total Environ 64:259-93.

Jones RE, Che1sky M. 1986. Further discussion concerning porphyria
cutanea tarda and TCDD exposure. Arch Environ Health 41(12):100-103.

Kaminsky LS, DeCaprio AP, Gierthy JF, Silkworth JB, Tumasonis C. 1985.
The role of environmental matrices and experimental vehicles in
chlorinated dibenzodioxin and dibenzofuran toxicity. Chemosphere
14:685-695.

Kang HK, Enziger F, Breslin P, Feil M, Lee Y, Shepard B. 1987. Soft
tissue sarcoma and military service in Vietnam: A case comparison group
analysis of hospital patients. J Occup Med 28:1215-1218.

Kaye CI, Rao S, Simpson SJ, Rosenthal FS, Cohen MM. 1985. Evaluation of
chromosomal damage in males exposed to Agent Orange and their families.
J Craniofacial Genetics and Developmental Biology Supplement 1:259-265.

Kelling CK, Christian BJ, Inhorn SL, Peterson RE. 1985. Hypophagia-
induced weight loss in mice, rats and guinea pigs treated with 2,3,7,8-
tetrachlorodibenzo-p-dioxin. Fundam Appl Toxicol 5:700-712.

Khera KS, Ruddick JA. 1973. Polychlorodibenzo-p-dioxins: Perinatal
effects and the dominant lethal test in Vistar rats. Adv Chem Ser 12C:
70-84 (cited in EPA 1985a).

Kimbrough RD, Carter CD, Liddle JA, Cline RE. 1977. Epidemiology and
pathology of a tetrachlorodibenzodioxin poisoning episode.  Arch Environ
Health 32:77-86.

Kimbrough RD, Falk H, Stehr P, Fries G. 1984. Health implications of
2,3,7,8-tetrachlorodibenzodioxin (TCDD) contamination of residential
soil. J Toxicol Environ Health 14:47-93.

Kimmig SJ, Schulz KH. 1957. Berufliche akna (sog. chlorakne) durch
chloriette aromatische zyklische ather. Dermatologia 115:540-546. (In
German) (cited in EPA 1984).

Knutsen AP. 1984. Immunologic effects of TCDD exposure in humans. Bull
Environ Contain Toxicol 33:673-681.

-------
108   Section 10

Knutsen AP, Roodman ST, Evans RG, et al. 1987. Immune studies in
dioxin-exposed Missouri residents: Quail Run. Bull Environ Contain
Toxicol 39:481-489.

Kociba RJ, Schwetz BA. 1982. Toxicity of 2,3,7,8-tetrachlorodibenzo-p-
dioxin (TCDD). Drug Metab Rev 13:387-406 (cited in EPA 1985a).

* Kociba RJ, Keyes DG, Beyer JE, et al. 1978a. Results of a two-year
chronic toxicity and oncogenicity study of 2,3,7,8-tetrachlorodibenzo-
p-dioxin in rats. Toxicol Appl Pharmacol 46(2):279-303.

* Kociba RJ, Keyes DG, Beyer JE, Carreon RM. 1978b. Toxicologic studies
of 2,3,7,8-tetrachlorodibenzo-p-dioxin  (TCDD) in rats. Toxicol Occup Med
(De Toxicol Environ Sci) 4:281-287 (cited in EPA 1985a).

Kociba RJ, Keyes DG, Beyer JE, Carreon RM, Gehring PJ. 1979. Long-term
toxicologic studies of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in
laboratory animals. Ann NY Acad  Sci 320:397-404 (cited in EPA 1985a).

Kogan MD,  Clapp RV. 1985. Mortality among Vietnam veterans in
Massachusetts, 1972-1983. Massachusetts Office of the Commissioner of
Veterans Services, Agent Orange  Program, Massachusetts Department of
Public Health, Division of Health Statistics  and Research, Boston, Mass
(cited in  EPA 1988b).

Korcher CW, Mahle  NH.  Hummels RA, et  al.  1978. A search  for  the present
of 2,3,7,8-tetrachlorodibenzo-p-dioxin  in beef fat.  Bull Environ Contan
Toxicol 19:229-236.

Krovke R.  1986.  Studies on distribution and embryotoxicity of different
PCDD  and  PCDF in mice  and marmosets.  Chemosphere  15:2011-2022.

Kuehl DW,  Cook PM,  Batterman AP. 1985.  Studies on the bioavailability of
2,3,7,8-TCDD  from municipal incinerator fly ash to  freshwater  fish.
Chemosphere 14:871-872.

Keuhl DW,  Butterworth DC,  DeVita WM,  Sauer  CP,  1987. Environmental
contamination by polychlorinated dibenzo-p-dioxins and dibenzofurans
associated with pulp and paper mill discharge.  Biomed Environ Mass
 Spectrom 14:443-447.

 Lakshmanan MR, Campbell BS, Chirtel SJ, Ekarohita N, Ezekiel M.  1986.
 Studies on the mechanism of absorption and distribution of 2,3,7,8-
 tetrachlorodibenzo-p-dioxin in the rat. J Pharmacol Exp  Ther 239:673-
 677.

 Lamb IV.  JC,  Harris Mtf, McKinney JD, Birnbaum LS. 1986.  Effects of
 thyroid hormones on the induction of cleft palate by 2,3,7,8-
 tetrachlorodibenzop-dioxin (TCDD) in C57BL/6N mice. Toxicol Appl
 Pharmacol 84:115-124.

-------
                                                        References   109

Lamparski LL, Nestrick TJ. 1980. Determination of tetra-, hexa-,  hepta-
and octachlorodibenzo-p-dioxin isomers in particle samples at parts-
per-trillion range. Anal Chem 52:2045-2054.
                 •   »
Lamparski LL, Nestrick TJ, Frawley NN, Hummel RA, Kocher CU, et al.
1986. Perspectives of a large scale environmental study for chlorinated
dioxins: Water analysis. Chemosphere 15:1445-1452.

Langhorst ML, Shadoff LA. 1980. Determination of parts-per-trillion
concentrations of tetra-, hexa-, hepta-, and octachlorodibenzo-p-dioxins
in human milk samples. Anal Chem 52:2037-2044.

Lapeza CR, Jr., Patterson DG, Jr., Liddle JA. 1986.  Automated apparatus
for the extraction and enrichment of 2,3,7,8-tetrachlorodibenzo-p-dioxin
in human adipose tissue. Anal Chem 58(4):713-716.

Lathrop GD, Wolfe WH, Albanese RA, Moynahan PM. 1984. An epidemiological
investigation of health effects in Air Force personnel following
exposure to herbicides: Baseline morbidity study results. USAF School of
Aerospace Medicine, Brooks Air Force Base, Texas (cited in EPA 1988b).

Lawrence J, Onuska F, Wilinson R, Afgan BK. 1986. Methods in research:
Determination of dioxins in fish and sediment. Chemosphere 15:1085-1090.

Loprieno M, Sbrana I, Rusciano D, Lascialfari D, Lari T. 1982. In vivo
cytogenic studies on mice and rats exposed to 2,3,7,8-
tetrachlorodibenzo-p-dioxin (TCDD). In: Hutzinger 0, et al., eds.
Chlorinated Dioxins and Related Compounds: Impact on the Environment,
New York: Pergamon Press, pp. 419-428  (cited in EPA 1985a).

Lu CH, Baggs RB, Redmond D, Henary EC, Schecter A, Gasiewicz TA.  1986.
Toxicity and evidence for metabolic alterations in 2,3,7,8-
tetrachlorodibenzo-p-dioxin-treated guinea pigs fed by total parenteral
nutrition. Toxicol Appl Pharmacol 84:439-453.

* Lucier GW, Rumbaugh RC, McCoy Z, Hass R, Harvan D, Albro P. 1986.
Ingestion of soil contaminated with 2,3,7,8-tetrachlorodibenzo-p-dioxin
(TCDD) alters hepatic enzyme activities  in rats. Fundam Appl Toxicol
6:364-371.

Lundgren K, Andries M, Thompson C, Lucier GW. 1986. Dioxin treatment of
rats results in increased in vitro induction of sister chromatid
exchanges by o-naphthoflavone: An animal model for human exposure  to
halogenated aromatics. Toxicol Appl Pharmacol 85:189-195.

Luster MI, Dean JH, Boorman GA. 1982.  Altered immune functions in
rodents treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin, phorbol-12-
myristate-13-acetate, and benzo[a]pyrene. Banbury Rep. Vol 11, Iss
Environmental Factors in Human Growth  and Development, pp. 199-213.

Lynge E. 1985. A follow-up study of cancer incidence among workers in
manufacture of phenoxy herbicides in Denmark. Br J Cancer 52:259-270.

-------
110   Section 10

Hanara L, Coccia P, Crbci T. 1984. Prevention of TCDD toxicity in
laboratory rodents by addition of charcoal or cholic acids to chow.  Fooa
Chem Toxicol 22:815-818.

Marklund S, Kjeller LO, Hansson M, Tysklind M, Rappe C,  Ryan C,  Collazo
H, Doughterty R. 1986. Determination of PCDDs and PCDFs  in incineration
samples and pyrolytic products. In: Rappe C, Choudhary G,  Keith LH,  eds.
Chlorinated Dioxins and Dibenzofurans in Perspective. Chelsea, Mich:
Lewis Publishers, Inc. pp. 79-92.

Marklund S, Rappe, C, Tysklind, Egeback KE. 1987. Identification of
polychlorinated dibenzofurans and dioxins in exhausts from cars run on
leaded gasoline. Chemosphere (England) 16(l):29-36.

Marple L, Brunck R, Throop, L. 1986a. Water solubility of 2,3.7.8-
tetrachlorodibenzo-p-dioxin. Environ Sci Technol 20(2):180-182.

Marple, L, B. Berridge, L. Throop. 1986b. Measurement of the water-
octanol partition coefficient of  2,3,7,8-tetrachlorodibenzo-p-dioxin.
Environ Sci Technol 20(4):397-399.

Mason G, Safe S. 1986a. Synthesis, biologic and  toxic effects of  the
major 2,3,7,8-tetrachlorodibenzo-p-dioxin metabolites in the rat.
Toxicology 41:153-159.

Mason G, Safe S. 1986b. Synthesis, biologic and  toxic properties  of
2,3,7,8-tetrachlorodibenzo-p-dioxin. Chemosphere 15:2081-2083.

Matsumura  F, Quensen  J. Tsushimoto G. 1983. Microbial degradation of
TCDD in a  model  ecosystem.  In: Tucker RE,  et  al.,  eds. Human and
Environmental Risks of Chlorinated Dioxins  and Related Compounds. New
York: Plenum Press, pp. 191-219.

May G. 1973. Chloracne from the  accidental production of
tetrachlorodibenzodioxin.  Br J Ind Med  30(3):276-283 (cited in  EPA
1985a).

McCann J.  1978.  Unpublished study (cited in EPA  1985a).

McConnell  EE.  1985.  Comparative  toxicity of PCBs and related compounds
 in various species of animals. Environ  Health Perspect  60:29-33.

McConnell  EE.  Moore JA, Dalgard  DW.  1978.  Toxicity of 2,3,7,8-
 tetrachlorodibenzo-p-dioxin in rhesus monkeys (Hacaca mulatta)  following
 a single oral  dose.  Toxicol Appl Pharmacol 43:175-187 (cited in EPA
 1985a).

 * McConnell EE,  Lucier GU,  Rumbaugh RC, et al.  1984. Dioxin in soil:
 Bioavailability after ingestion  by rats and guinea pigs.  Science
 223:1077-1079.

 McNulty W. 1984. Fetotoxicity of 2,3,7.8-tetrachlorodibenzo-p-dioxin
 (TCDD)  for Rhesus Macaques (Hacaca mulatta). Am J  Primatol 6:41-47.

-------
                                                        References   111

McNulty W. 1985. Toxicity and fetotoxicity of TCDD, TCDF and PCB isomers
in Rhesus Macaques (Hacaca mulatea). Environ Health Perspect 60:77-88.

McQueen EG, Veale AMO, Alexander US, Bates MN. 1977. 2,4,5-T and Human
Birth Defects. Report of the Division of Public Health, New Zealand
Department of Health (cited in EPA  1985a).

Mehrle PM, Buckler DR, Little EE, et al. 1988. Toxicity and
bioconcentration of 2,3,7,8-tetrachlorodibenzodioxin and 2,3,7,8-
tetrachlorodibenzofuran in rainbow  trout. Environ Toxicol Chem 1:47-62.

Meyne J, Allison DC, Bose K, Jordan SU, Ridolpho PF, Smith J. 1985.
Hepatotoxic doses of dioxin do not  damage mouse bone marrow chromosomes.
Mutat Res 157:63-69.

Milham S, Jr. 1982. Herbicides, occupation, and cancer. The Lancet, pp.
1464-1465.

Miller GC, Hebert VR, Zepp RG. 1987. Chemistry and photochemistry of
low-volatility organic chemicals on environmental surfaces. Environ Sci
Technol 21:1164-1167.

MMWR (Morbidity and Mortality Weekly Report). 1987. Epidemiologic Notes
and Reports. Serum Dioxin in Vietnam-Era Veterans - Preliminary Report.
Centers for Disease Control Morbidity and Mortality Weekly Report
36(28):470-474.

Mocarelli P, Marcocchi A, Brambilla P, Gerthoux P, Young DS, Mantel N.
1986. Clinical laboratory manifestations of exposure to dioxin in
children. JAMA 256:2687-2695.

Mohamad AH, Zerezghi M, Caruso JA.  1986. Determination of
polychlorinated dibenzo-p-dioxins using capillary gas chromatography
with microwave-induced plasma detection. Anal Chem 58(2):469-471.

Moore JA, Gupta BN, Zinkl JG, Yos JG. 1973. Postnatal effects of
maternal exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Environ
Health Perspect 5:81-85  (cited in EPA 1985a).

Moore RW, Potter CL. Theobald HM, Robinson JA, Peterson RE.  1985.
Androgenic deficiency in male rats  treated with 2,3,7,8-
tetrachlorodibenzo-p-dioxin. Toxicol Appl Pharmacol 79:99-111.

Mortelmans K, Haworth S, Speck W, Zeiger E. 1984. Mutagenicity testing
of Agent Orange and related compounds. Toxicol Appl Pharmacol 75:137-
146.

Moses M, Prloleau PC. 1985. Cutaneous histologic  findings  in chemical
workers with and without chloracne  with past  exposure  to 2,3,7,8-
tetrachlorodibenzo-p-dioxin. J Am Acad Dernatol 12(3):497-506.

-------
112   Section 10

Mottura A, Zet G, Nuzzo F, et al. 1981. Evaluation of results of
chromosome analyses on lymphocytes of TCDD exposed subjects after the
Seveso accident. Hutat Res 85(4):238-239 (cited in EPA 1985a).

Mulcahy MT. 1980. Chromosome aberrations and "Agent Orange." Med J Aust
2(10):573-574 (cited in EPA 1985a).

* Murray FJ, Smith FA, Nitschke KD,  Humiston CG,  Kociba RJ, Schwetz BA.
1979. Three-generation reproduction study of rats given 2,3,7,8-
tetrachlorodibenzo-p-dioxin (TCDD) in the diet.  Toxicol Appl Pharmacol
50:241-251.

Naikwadi KP, Karasek FW. 1986. Gas chromatographic separation of
2,3,7,8-tetrachlorodibenzo-p-dioxin from polychlorinated biphenyls and
tetrachlorodibenzo-p-dioxin isomers using a polymeric liquid crystal
capillary column. J Chromatogr 369(1):203-207.

Nagayama J, Klyohara C, Masuda Y. Kuratsune M.  1985. Genetically
mediated induction of aryl hydrocarbon hydroxylase activity in human
lymphoblastoid cells by polychlorinated dibenzofuran isomers and
2,3,7,8-TCDD. Arch Toxicol 56:230-235.

Nau H, Bass R, Neubert D. 1986. Transfer of 2,3,7,8-tetrachlorodibenzo-
p-dioxin (TCDD) via placenta and milk,  and postnatal toxicity in the
mouse. Arch Toxicol 59:36-40.

Nelson CJ, Holson JF, Green HG, Gaylor DW. 1979.  Retrospective study of
the relationship between agricultural use of 2,4,5,-T and cleft palate
occurrence in Arkansas. Teratology 19:377-384 (cited in EPA 1985a).

Nestrick TJ, Lamparski LL, Frawley NN,  Hummel RA, Kocher CV, et al.
1986. Perspectives of a large scale environmental survey for chlorinated
dioxins. Overview and soil data. Chemosphere 15:1453-1460.

* Neubert D, Dillman I. 1972. Embryotoxic effects in mice treated with
2,4.5-trichlorophenoxyacetic acid and 2,3,7,8-tetrachlorodibenzo-p-
dioxin. Arch Pharmacol 272(3):243-264.

Neubert D, Zens  P, Rothenwallner A, Merker HJ. 1973. A survey of the
embryotoxic effects of TCDD in mammalian species. Environ Health
Perspect 5:67-79  (cited in EPA 1985a).

Niemann RA. 1986. Surrogate-assisted determination  of 2,3,7.8-
tetrachlorodibenzo-para-dioxin in fish by electron-capture  capillary
gas-chromatography. J Assoc Off  Anal Chem 69(6):976-980.

NIOSH (National  Institute of Occupational Safety and Health). 1984.
2,3,7,8-Tetrachlorodibenzo-p-Dioxin. Current Intelligence  Bulletin 40
U.S.  Department  of Health and Human  Services, Public Health Service,
Centers for Disease Control, National  Institute  for Occupational  Safety
and Health.

-------
                                                        References   113

* Nisbec ICT, Paxton MB. 1982. Statistical aspects of three-generation
studies of the reproductive toxicity of TCDD and 2,4,5-T. Am Stat
36(3):290-298.

Nolan RJ, Smith FA, Hefner JG. 1979. Elimination and tissue distribution
of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCOD) in female guinea pigs
following a single oral dose. Toxicol Appl Pharmacol 48(1):A162 (cited
in EPA 1985a).

Nottrodt IA, Ballschmiter K. 1986. Causes for and reduction strategies
against emissions of PCDD/PCDF from waste incineration plants--
interpretations of recent measurements. Chemosphere 15:1225-1237.

* NTP (National Toxicology Program). 1982a. Bioassay of 2,3,7,8-
Tetrachlorodibenzo-p-Dioxin for Possible Careinogenieity (Gavage Study).
DHHS Publ No (NIH) 82-1765. Bethesda, Md: Carcinogenesis Testing
Program, National Cancer Institute, National Institute of Health;
Research Triangle Park, N.C.: National Toxicology Program.

NTP (National Toxicology Program). 1982b. Bioassay of 2,3,7,8-
Tetrachlorodibenzo-p-Dioxin for Possible Carcinogenicity (Dermal Study).
DHHS Publ No (NIH) 82-1757. Bethesda, Md: Carcinogenesis Testing
Program, National Cancer Institute, National Institute of Health;
Research Triangle Park, N.C.: National Toxicology Program.

Nygren M, Rappe C, Lindstrom G, Hansson M, Bergqvist PA, et al. 1986.
Identification of 2,3,7,8-substituted polychlorinated dioxins and
dibenzofurans in environmental and human samples. In: Rappe C, Choudhary
G, Keith LH, eds. Chlorinated Dioxins and Dibenzofurans in Perspective.
Chelsea, Mich: Lewis Publishers, Inc, pp. 15-24.

Oehme M, Mano S, Mikalsen A, Kirschmer P. 1986. Quantitative method for
the determination of femtogram amounts of polychlorinated dibenzo p-
dioxins and dibenzofurans in outdoor air. Chemosphere 15(5):607-617.

Ogaki J, Takayama K, Miyata H, Kashimoto T. 1987. Levels of PCDDs and
PCDFs in human tissues and various foodstuffs in Japan. Chemosphere
16:2047-2056.

O'Keefe P, Meyer C, Smith R, Hilker D, Aldons K. Wilson L. 1986.
Reverse-phase adsorbent cartridge for trapping dioxins in drinking
water. Chemosphere 15:1127-1134.

Oliver RM. 1975. Toxic effects of 2,3l7,8-tetrachlorodibenzo-l,4-dioxin
in laboratory workers. Br J Ind Med 32(l):49-53 (cited in EPA 1984).

Olson JR. 1986. Metabolism and disposition of 2,3,7,8-
tetrachlorodibenzo-p-dioxin in guinea pigs. Toxicol Appl Pharmacol
85:263-273.

Olson JR, Wroblewski VJ. 1985. Metabolism of 2,3,7,8-
tetrachlorodibenzo-p-dioxin (TCDD) in isolated hepatocytes from guinea
pigs and rats. Chemosphere 14:979-982.

-------
114   Section 10

Olson JR, Gasiewicz TA, Neal, RA Neal.  1980.  Tissue distribution,
excretion, and metabolism of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)
in the Golden Syrian hamster. Toxicol Appl Pharmacol 56:78-85 (cited in
EPA 1985a).

Olson JR, Gasiewicz TA, Geiger LE, Neal RA.  1983.  The metabolism of
2,3,7,8-tetrachlorodibenzo-p-dioxin in mammalian systems.  In: Coulston
R,  and Pocchiari F, eds. Accidental Exposure  to Dioxins:  Human Health
Aspects.  New York: Academic Press, pp. 81-100 (cited in EPA 1985a).

Ono M, Kashima Y, Wakimoto T, Taksukawa R. 1987.  Daily intake of PCDDs
and PCDFs by Japanese through food. Chemosphere 16:1823-1828.

Ott KG, Holder BB, Olson RO. 1980. A mortality analysis of employees
engaged in the manufacture of 2,4,5-trichlorophenoxyacetic acid. J Occup
Med 22(1):47-50 (cited in EPA 1985a).

Ott HG, Olson RA, Cook RR. 1987. Cohort mortality study of chemical
workers with potential exposure to the higher chlorinated dioxins. J
Occup Med 29(5):422-429.

Ozvacic V. 1986. A review of stack sampling methodology for PCDDs/
PCDFs. Chemosphere 15:1173-1178.

Palansky J, Kapila S, Manahan SE, Venders AF, Halhotra RK, Clevenger TE.
1986. Studies on vapor phase transport and role of dispersing medium on
mobility of 2,3,7,8-TCDD in soil. Chemosphere 15:1389-1396.

Patterson DG, Hoffman RE, Needham LL, et al.  1986. 2,3,7,8-
Tetrachlorodibenzo-p-dioxin levels in adipose tissue of exposed and
control persons in Missouri. JAMA 256:2683-2686.

Patterson DG, Jr., Holler JS, Belson UT, Boozer EL, Lapeza CR, Needham
LL. 1987a. Determination of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)
in human adipose tissue on whole-weight and lipid basis. Chemosphere
16:935-936.

Patterson DG, Jr., Hampton L, Lapeza CR, Jr., et al. 1987b. High-
resolution gas chromatographic/high-resolution mass spectrometric
analysis of human  serum on a whole-weight and lipid basis  for 2,3,7,8-
tetrachlorodibenzo-p-dioxin. Anal Chem 59:2000-2005.

Patterson DG, Holler JS, Lapeza CR, Jr., et al. 1987c. High-resolution
gas chromatographic/high-resolution mass spectrometric analysis of human
adipose  tissue for 2,3,7,8-tetrachlorodibenzo-p-dioxin. Anal Chem
58(4):705-713.

Patterson DG, Fingerhut MA, Roberts DR, et al. Levels of polychlorinated
dibenzo-p-dioxins  (PCDDs) and dibenzofurans  (PCDFs)  in workers  exposed
to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Submitted  for publication to  Am
J Ind Med.

-------
                                                        References   115

Paustenbach, DJ, Shu HP, Murray FJ. 1986. A critical examination of
assumptions used in risk assessments of dioxin contaminated soil. Regul
Toxicol Pharmacol 6:284-307.

Peterson RE, Seefeld MD, Christian BJ, Potter CL, (Celling CK, Keesey RE.
1984. The wasting syndrome in 2,3,7,8-tetrachlorodibenzo-p-dioxin
toxicity: Basic features and their interpretation. In: Poland A,
Kimbrough RD, eds. Banbury Rep 18. Cold Spring Harbor Laboratory; 291-
308.

Philippi M, Krasnobage V, Zeyer J, Huetter R. 1981. Fate of 2,3,7,8-
tetrachlorodibenzo-p-dioxin (TCOD) in microbial cultures and soil under
laboratory conditions. FEMS Symp 12:2210-2233.

Piper WN. Rose RQ, Gehring PJ. 1973. Excretion and tissue distribution
of 2,3,7,8-tetrachlorodibenzo-p-dioxin in the rat. Environ Health
Perspect 5:241-244 (cited in EPA 1985a).

Pitot HC, Goldsworthy T, Poland H. 1980.  Promotion by 2,3,7,8-
tetrachlorodibenzo-p-dioxin of hepatocarcinogenesis from
diethylnitrosamine. Cancer Res 40:3616-3620 (cited in EPA 198Sa).

Podoll RT, Jaber HM, Mill T. 1986. Tetrachlorodibenzodioxin: Rates of
volatilization and photolysis in the environment. Environ Sci Technol
20:490-492.

Poiger H, Schlatter C. 1980. Influence of solvents and adsorbents on
dermal and intestinal absorption of TCDD. Food Cosmet Toxicol
18(S):477-481 (cited in EPA 1985a).

Poiger H, Schlatter C. 1985. Influence of phenobarbital and TCDD on the
hepatic metabolism of TCDD in the dog. Experientia 41:376-378.

* Poiger H, Schlatter C. 1986. Pharmacokinetics of 2,3,7,8-TCDD  in man.
Chemosphere 15:9-12.

Poiger H, Weber H, Schlatter CH. 1982. Special aspects of metabolism and
kinetics of TCDD in dogs and rats. Assessment of toxicity of TCDD-
metabolite(s) in guinea pigs. In: Hutzinger 0, Frei RW, Merian E,
Pocchiari F, eds. Chlorinated Dioxins and Related Compounds. Impact on
the Environment. New York: Pergamon Press, pp. 317-325 (cited in EPA
1985a).

Poland A, Glover E. 1973. Chlorinated dibenzo-p-dioxins: Potent  inducers
of gamma-aminolevulinic acid synthetase and aryl hydrocarbon
hydroxylase. II. A study of the structure activity relationship. Mol
Pharmacol 9:736 (cited in EPA 1985a).

Poland A, Glover E. 1979. An estimate of the maximum  in vivo covalent
binding of 2,3,7,8-tetrachlorodibenzo-p-dioxin to rat liver protein,
ribosomal RNA and DNA. Cancer Res 39(9):3341-3344  (cited  in EPA  1985a).

-------
116   Section 10

Poland A, Glover E. 1980. 2,3,7,8-Tetrachlorodibenzo-p-dioxin
segregation of toxicity with the Ah locus.  Hoi Pharmacol 17(l):86-94.

Poland A, Knutsort JC-. 1982. 2,3, 7 ,8-Tetrachlorodibenzo-p-dioxin and
related halogenated aromatic hydrocarbons:  Examination of the mechanism
of toxicity. Ann Rev Pharmacol Toxicol 22:517-554 (cited in EPA 1985a).

Poland AP, Smith D, Hotter G, Fossick P.  1971. A health survey of
workers in a 2,4-D and 2,4,5-T plant with special attention to
chloracne, porphyria cutanea tarda, and psychologic parameters. Arch
Environ Health 22:316-327 (cited in EPA 1984).

Poland A, Greenlee WF, Kende AS.  1979. Studies on the mechanisms of
action of the chlorinated dibenzo-p-dioxins and related compounds.  Ann
NY Acad Sci 320:214-230  (cited in EPA 1985a).

Poland A, Palen D, Glover E. 1982.  Tumour promotion by TCDD in skin of
HRS/J mice. Nature 300(5889):271-273 (cited in EPA 1985a).

Potter CL, Henahan LA, Peterson RE. 1986. Relationship of alterations in
energy metabolism in rats treated with 2,3,7,8-Tetrachlorodibenzo-p-
dioxin. Fundam Appl Toxicol 6:89-97.

* Puhvel SH, Sakamoto M, Ertl DC, Reisner RH.  1982. Hairless mice as
models for chloracne: A  study of cutaneous changes induced by topical
application of established chloracnegens. Toxicol Appl Pharmacol
64:492-503.

Puntoni R, Herlo F, Fini A, Heazza L, Santi L. 1986. Soft tissue
sarcomas in Seveso. Lancet 2:525.

Rappe C. 1984. Analysis  of polychlorinated dioxins and furans. All 75
PCDDs and 135 PCDFs can  be identified by isomer-specific techniques.
Environ Sci Technol  18(3):78A-90A.

Rappe C, Nygren H, S. Harklund, et al. 1985. Assessment of human
exposure to polychlorinated  dibenzofurans and dioxins. Environ Health
Perspect 60:303-304.

Rappe C, Harklund  S, Kjeller LO, Tysklind H.  1986. PCDDs and PCDFs  in
emissions from various  incinerators. Chemosphere  15:1213-1218.

Rappe C, Kjeller L-0. 1987.  PDDs and PCDFs  in environmental  samples,
air, particulates, sediments and soil. Chemosphere 16:1775-1780.

Rappe C, Nygren M, Lindstrom G, et al. 1987.  Polychlorinated
dibenzofurans and  dibenzo-p-dioxins and  other chlorinated  contaminants
in cow milk from various locations in Switzerland. Environ Sci Technol
21:964-970.

Regglanl G.  1980.  Acute human exposure Co TCDD  in Seveso,  Italy. J
Toxicol  Environ Health  6(l):27-43  (cited in EPA 1985a).

-------
                                                        References   117

Rilhimaki V,  Sisko A,  Hernberg S.  1982. Mortality of 2,4-
dichlorophenoxyacetic  acid and 2,4,5-trichlorophenoxyacetic acid
herbicide applicators  in  Finland.  Scand J Work Environ Health 8:37-42
(cited  in EPA 1965a>.

Robens  JF, Zabik M. n.d.  Triple Quadrapole Mass Spectrometry and RM Temp
Phosphoresence-Dioxin  Residue.  Michigan State University.

* Roberts EA,  Shear NH, Okey AB, Manchester DK. 1985. The Ah receptor
and dioxin toxicity: From rodent to human tissues. Chemosphere
14:661-674.

Rogers  AM, Anderson ME, Back KG. 1982. Mutagenicity of 2,3,7,8-
tetrachlorodibenzo-p-dioxin and perfluoro-n-decanoic acid in LS178y
mouse lymphoma cells.  Mutat Res 105:445-449 (cited in EPA 1985a).

Rogers  CJ, Kernel A, Peterson RL.  1987. Mobile KPEG destruction unit for
PCBs, dioxins, and furans  in contaminated wastes. Presented at the 13th
Annual  Research Symposium:  Land Disposal, Remedial Action. Incineration
and Treatment  of Hazardous  Wastes, Cincinnati, Ohio. May 6-8, 1987.

Rondorf B. 1986. Thermal  properties of dioxins, furans and related
compounds. Chemosphere 15:1325-1332.

Rozman K. 1984. Separation  of wasting syndrome and lethality caused by
2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Lett 22:279-285.

Rozman K, Grelm H. 1986. Toxicity  of 2,3,7,8-tetrachlorodibenzo-p-dioxin
in cold-adapted rats.  Arch  Toxicol 59:211-215.

Rozman K, Rozman T, Scheufler E, Pazdemik T, Greim H. 1985. Thyroid
hormones modulate the  toxicity  of  2,3,7,8-TCDD. J Toxicol Environ Health
16:481-491.

Ryan JJ. 1987. Food Research Division, Health Protection Branch, Ottawa,
Canada.  Personal telephone  communication to DK Basu, Syracuse Research
Corporation,  N.Y., September 2, 1987.

Ryan JJ, Lau PY, Pilon JC,  et al.  1984. Incidence and levels of
2,3,7,8-tetrachlorodibenzo-p-dioxin in Lake Ontario commercial fish.
Environ Sci Techno1 18(9):719-721.

Ryan JJ, Lizotte R, Sakuma  T, Mori B. 1985a. Chlorinated dibenzo-para-
dioxins, chlorinated dibenzofurans and pentachlorophenol in Canadian
chicken and pork samples. J Agric  Food Chem 33(6):1021-1026.

Ryan JJ, Schecter A, Lizzote R, Sun WF, Miller L. 1985b. Tissue
distribution of dioxins and furans in humans from the general
population.  Chemosphere 14:929-932.

Ryan JJ, Lizotta R, Lau BPY. 1985c. Chlorinated dibenzo-p-dioxins and
chlorinated dibenzofurans in Canadian human adipose tissue. Chemosphere
14:697-706.

-------
118   Section 10

Sacchi GA, Vigano P. Fortunati G, Cocucci SM.  1986.  Accumulation of
2,3,7,8-tetrachlorodibenzo-p-dioxin from soil  and nutrient solution by
bean and maize plants. Experientia 42(5):586-588.

SANSS (Structure and Nomenclature Search System).  1987.  On-line
printout. April 3, 1987.

Sawahata T, Olson JR. Neal RA. 1982. Identification of metabolites of
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) formed on incubation with
isolated rat hepatocytes. Biochem Biophys Res  Commun 105(1):341-346
(cited in EPA 1985a).

Schantz SL, Barsotti DA, Allen JR. 1979. Toxicological effects produced
in nonhuman primates chronically exposed to fifty parts-per-trillion
2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Toxicol Appl Pharmacol
48(2):A180 (cited in EPA 1988b).

Schecter A, Gasiewicz TA. 1987. Health hazard assessment of chlorinated
dioxins and dibenzofurans contained in human milk. Chemosphere 16:2147-
2154.

Schecter A, Tieman T, Schaffner F. 1985. Patient fat biopsies for
chemical analysis and liver biopsies for ultrastructural
characterization after exposure to polychlorinated dioxins, furans and
PCBs. Environ Health Perspect 60:241-254.

Schecter A, Ryan JJ, Gitlitz G. 1986. Chlorinated dioxin and
dibenzofurans levels in human adipose tissues from exposed and control
populations. In: Rappe C, Choudhary G, Keith LH, eds. Chlorinated
Dioxins and Dibenzofurans in Perspective. Chilson, Mich: Lewis
Publishers Inc., pp. 51-56.

Schecter A, Ryan JJ, Constable JD.  1987. Polychlorinated dibenzo-p-
dioxin and polychlorinated dibenzofuran levels  in human breast milk  from
Vietnam compared with cow's milk and human breast milk from the North
American continent.  Chemosphere  16:2003-2016.

Schroy JM, Hileman  FD,  Cheng  SC.  1985.  Physical/chemical properties  of
2,3,7,8-TCDD. Chemosphere 14:877-880.

Schroy JM, Hileman  FD,  Cheng  SS.  1986.  Physical/chemical properties  of
2,3,7,8-tetrachlorodibenzo-p-dioxin.  In:  Bahner RC,  Hansen DJ,  eds.
Aquatic  Toxicology  and  Hazard Assessment:  Eighth  Symposium.
Philadelphia, Pa: ASTM;  ASTM-STP 891, pp.  409-421.

Schulz KH. 1957.  Klinishce und experimentelle untersuchungen  zur
atiologie  der chloracne. Arch Klin Exp  Dermatol 206:589-596.  (In  German)
 (cited  in EPA 1984).

*  Schwetz  BA, Norris JM,  Sparschu GL, et  al.  1973.  Toxicology of
chlorinated  dibenzo-p-dioxlns. Environ  Health Perspect  5:87-99.

-------
                                                        References   119

Seefeld MS, Peterson RE.  1984. Digestible energy and efficiency of feed
utilization in rats treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin.
Toxicol Appl Pharmacol  74:214-222.

Seefeld MS, Corbett SW, Keesey RE, Peterson RE. 1984a. Characterization
of the wasting syndrome in rats treated with 2,3,7,8-
tetrachlorodibenzo-p-dioxin. Toxicol Appl Pharmacol 73:311-322.

Seefeld MS, Keesey RE, Peterson RE. 1984b. Body weight regulation in
rats treated with 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Appl
Pharmacol 76:526-536.

Seiler JP. 1973. A survey on the mutagenicity of various pesticides.
Experientia 29:622-623  (cited in EPA 1985a).

Shiverick KT, Muther TF. 1983. 2,3,7,8-Tetrachlorodibenzo-p-dioxin
(TCDD) effects on hepatic microsomal steroid metabolism and serum
estradiol of pregnant rats. Biochem Pharmacol 32:991-995.

Shu HP, Paustenbach DJ, Murray FJ. 1987. A critical evaluation of the
use of mutagenesis, carcinogenesis, and tumor promotion data in a cancer
risk assessment of 2,3,7,8-tetrachlorodibenzo-p-dioxin. Regul Toxicol
Pharmacol 7:57-88.

Shu H, Teitelbaum P, Webb AS, et al. 1988. Bioavailability of soil-bound
TCDD: Dermal bioavailability in the rat. Fundam Appl Toxicol 10:335-343.

Slaga TJ, Nesnow S. 1985. Sencar mouse skin cumorigenesis. In: Handbook
of Carcinogen Testing. Millmen HA, Veisburger EK, eds. Park Ridge, N.J,
Noyes Publ, pp. 230-250.

Smith AH, Fisher DO, Dip NP, Chapman CJ. 1982. Congenital defects and
miscarriages among New Zealand 2,4,5-T sprayers. Arch Environ Health
37:197-200 (cited in EPA 1985a).

Smith AH, Pearce NE, Fisher DO. Giles HJ, Teague CA, Howard JK. 1984.
Soft tissue sarcoma and exposure to phenoxyherbicides and chlorophenols
in New Zealand. J Natl Cancer Inst 73:1111-1117.

Smith FA, Schwetz BA, Nitschke KD. 1976. Teratogenicity of 2,3,7,8-
tetrachlorodibenzo-p-dioxin in CF-1 mice. Toxicol Appl Pharmacol
38(3):517-523 (cited in EPA 1985a).

Smith RM, O'Keefe PW, Hilker DR, Aldous KM. 1986. Determination of
picogram per cubic meter concentrations of tetrachlorinated and
pentachlorinated dibenzofurans and dibenzo-para-dioxins in indoor air  by
high-resolution gas chromatography high-resolution mass spectrometry.
Anal Chem 58(12):2414-2420.

Sparschu GL, Jr. , Dunn FL, Jr. , Row VK, Jr. 1971a. Study of the
teratogenicity of 2,3,7,8-tetrachlorodibenzo-p-dioxin in the rat. Food
Cosmet Toxicol 9:405-412  (cited in EPA 1985a).

-------
120   Section 10

Sparschu GL, Dun FL, Lisowe RW, Rowe VK.  1971b.  Effects of high levels
of 2,4,5-trichlorophenoxyacetic acid on fetal development in the rat.
Food Cosmet Toxicol 9(40):527-530 (cited in EPA 1985a).

SRI (Stanford Research Institute).  1987.  1987 Directory of Chemical
Producers. United States of America, SRI  International,  Menlo Park
Calif.

Stalling DL, Smith LM, Petty JD, Hogan JW,  Johnson JL et al. 1983.
Residues of polychlorinated dibenzo-p-dioxins and dibenzofurans in
Laurentar Great Lakes fish. In: Human and Environmental Risks of
Chlorinated Dioxins and Related Compounds.  Tucker E,  Young AL,  Gray AP,
eds. New York: Plenum Publishing Corp, pp.  221-240.

Stalling DL, Peterman PH, Smith LM,  Norstrom RJ,  Simon M.  1986. Use of
pattern recognition in the evaluation of PCDD and PCDF residue data from
GC/MS analysis. Chemosphere 15:1435-1443.

Stanley JS, Boggess KE, Onstot J, Sack TM,  Remmers JC, et al. 1986.
PCDDs and PCDFs in human adipose tissue from the  EPA TY82 NHATS
repository. Chemosphere 15:1605-1612.

Stehr PA, Stein G, Falk H,  et al. 1986. A pilot epidemiologic study of
possible health effects associated with 2,3,7,8-tetrachlorodlbenzo-p-
dioxin contaminations in Missouri.  Arch Environ Health 41(l):16-22.

Stevens KM. 1981. Agent Orange toxlcity:  A quantitative perspective. H.
Toxicol 1:31-39.

Stieglitz L, Zwick G, Roth W. 1986.  Investigation of different treatment
techniques for PCDD/PCDF in fly ash. Chemosphere  15:1135-1140.

Suskind RR. 1985. Chloracne, "the hallmark of dioxin intoxication."
Scand J Work Environ Health 11:165-171.

Takizawa Y, Muto H. 1987. PCDDs and PCDFs carried to the human body from
the diet. Chemosphere 16:1971-1975.

Taylor JS. 1979. Environmental chloracne: Update  and overview.  Ann NY
Acad Sci 320:295-307.

Tenchini ML, Crimaudo C, Pacchetti G, Mottura A,  Agosti S, DeCarli L.
1983. A comparative cytogenetic study on cases of induced abortions in
TCDD-exposed and nonexposed women.  Environ Mutagen 5:73-85.

Thiess AM, Frentzel-Beyme R. 1977.  Mortality study of persons exposed to
dioxin following an accident which occurred in the BASF on 13, November,
1953. Working Papers, Joint NIEHS/IARC Working Group Report. Lyon,
France, June (cited in EPA 1985a).

Thiess AM, Frentzel-Beyme,  Link R.  1982.  Mortality study of persons
exposed to dioxin in a trichlorophenol-process accident that occurred
the BASFAG on November 17,  1953. Am J Ind Med 3:179-189.

-------
                                                        References   121

Thigpen JE. Faith RE, McConnell  EE, Moore JA. 1975. Increased
susceptibility of bacterial  infection as a sequela of exposure to
2,3,7,8-tetrachlorodibenzo-p-dioxin. Infect Immun 12(6):1319-1324.

Thomas HF. 1980. Internal memo to P. Cohn, Office of Toxic Substances,
EPA, Washington, DC  (cited in EPA 1985a).

Tiernan TO, Taylor ML, Garrett JH, et al. 1985. Sources and fate of
polychlorinated dibenzodioxins,  dibenzofurans and related compounds in
human environments.  Environ  Health Perspect 59:145-158.

TMN (Toxic Materials News).  1987. Survey finds little dioxin at control
sites. Industry finds trace  amounts in paper products and highest levels
at pesticide plants. Sept. 30, p. 301.

Tognoni G, Bonaccorsi A. 1982. Epidemiological problems with TCDD (a
critical view). Drug Metab Rev 13:447-469.

Tong HY, Karasek, FW. 1986.  Comparison of quantitation of
polychlorinated dibenzodioxins and polychlorinated dibenzofurans in
complex environmental samples by high resolution gas chromotography with
flame ionization, electron capture and mass spectrometric detection.
Chemosphere 15:1141.1146.

Tong HY, Shore DL, Karasek FW. 1984. Isolation of polychlorinated
dibenzodioxins and polychlorinated dibenzofurans for a complex organic
mixture by two-step liquid chromatographic fractionation for
quantitative analysis. Anal  Chem 56:2442-2447.

Toth K, Somfai-Relle S, Sugar J, Bence J. 1979. Carcinogenicity testing
of herbicide 2,4,5-trichlorophenoxyethanol containing dioxin and of pure
dioxin in Swiss mice. Nature 278(5704):548-549 (cited in EPA 1985a).

Toth K, Olah E, Somfair-Relle S, Sugar J. 1984. Effect of herbicide
2,4,5-trichlorophenoxyethanol (TCPE) containing dioxin on mutation and
induction of sister chromatid exchanges. Carcinogenesis 5:1725-1728
(cited in Girl 1986).

Townsend JC, Bodner KM, Van  Peenen PFD, Olson RD, Cook RR. 1982. Survey
of reproductive events of wives  of employees exposed to chlorinated
dioxins. An J Epidem 115:695-713.

Travis CC, Hattemer-Frey HA. 1987. Human exposure to 2,3,7,8-TCDD.
Chemosphere 16:2331-2342.

Tsushimoto G, Matsumura F, Sago  R. 1982. Fate of 2,3,7,8-TCDD in an
outdoor pond and in model aquatic ecosystems. Environ Toxicol Chem
1:61-68.

* Turner JN, Collins ON. 1983. Liver morphology in guinea pigs
administered either pyrolysis products of a polychlorinated biphenyl
transformer fluid or 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol Appl
Pharmacol 67:417-429.

-------
122   Section 10

Umbreit TH, Patel D, Gallo HA. 1985. Acute toxicity of TCDD contaminat
soil from an industrial site. Chemosphere 14:945-947.

Umbreit TH, Hesse EJ, Gallo MA. 1986a. Unavailability of dioxin in soil
from a 2,4,5-T manufacturing site. Science 232:497-499.

Umbreit TH. Hesse EJ, Gallo MA. 1986b. Comparative toxicity of TCDD
contaminated soil from Times Beach, Missouri, and Newark, New Jersey.
Chemosphere 15:2121-2124.

Van den Berg M, Olie K, Hutzinger 0. 1983. Uptake and selection in rats
of orally administered chlorinated dioxins and dibenzofurans from fly-
ash and fly-ash extract. Chemosphere 12:537-544.

Van den Berg M, de  Vroom E, van Greevenbroek M, Olie K, Hutzinger 0.
1985. Bioavailability of PCDDs and PCDFs absorbed on fly ash in rat,
guinea pig and Syrian golden hamster. Chemosphere 14:865-869.

Van den Berg M, Sinke M, Wever H. 1987. Vehicle dependent
bioavailability of  polychlorinated dibenzo-p-dioxins (PCDDs) and
dibenzofurans  (PCDFs) in the rat. Chemosphere 16:1193-1203.

Van Miller JP, Lalich JJ,  Allen JR.  1977a. Increased incidence of
neoplasms  in rats exposed  to low  levels of 2,3,7,8-tetrachlorodibenzo-
p-dioxin.  Chemosphere 6(10):625-632  (cited in EPA 1985a).

Van Miller JP, Lalich JJ,  Allen JR.  1977b. Increased incidence of
neoplasms  in rats exposed  to  low  levels of 2,3,7,8-tetrachlorodibenzo-
p-dioxin.  Chemosphere 6(9):537-544  (cited in EPA 1985a).

Velzy CO.  1986. ASME standard  sampling and analysis  methods  for
dioxins/furans. Chemosphere  15:1179-1185.

View Data  Base. 1989. Agency for  Toxic Substances and  Disease Registry
 (ATSDR), Atlanta, Georgia: Office of External Affairs,  Exposure  and
Disease Registry  Branch,  February 1989.

* Vos JG,  Moore,  JA, Zinkl JG.  1973.  Effect  of  2,3,7,8-
 tetrachlorodibenzo-p-dioxin on the immune system of  laboratory  animals.
 Environ Health Perspect 5:149-162.

 Walden R,  Schiller CM.  1985.  Comparative  toxicity  of 2,3,7,8-
 tetrachlorodibenzo-p-dioxin (TCDD) in four  (sub)strains of adult male
 rats.  Toxicol Appl Pharmacol 77:490-495.

 Weber H,  Bimbaum LS.  1985. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD)
 and 2,3,7,8-tetrachlorodibenzofuran (TCDF)  in pregnant C57BL/6N mice:
 Distribution to the embryo and excretion. Arch Toxicol 57:159-162.

 Weber H,  Poiger H,  Schlatter C.  1982. Acute oral toxicity of TCDD-
 metabolites in male guinea pigs.  Toxicol Lett 14:117-122.

-------
                                                        References   123

Weber H, Harris MW, Haseman JK, Birnbaun LS. 1985. Teratogenic potency
of TCDD, TCDF, and TCDD-TCDF combinations in C57BL/6N mice. Toxicol Lett
26:159-167.

Veerasinghe NCA, Schecter AJ, Pan JC, et al. 1986. Levels of 2,3,7,8-
tetrachlorodibenzo-p-dioxin (2,3,7,8-TCDD) in adipose tissue of Vietnam
veterans seeking medical assistance. Chemosphere 15:1787-1794.

Weeren RD, Asshauer J. 1985. Problems and results of trace analysis of
2,3,7,8-tetrachlorodibenzo-p-dioxin in 2,4,5-trichlorophenoxyacetic acid
and its esters. J Assoc Off Anal Chem 68(5):917-921.

Wiklund K, Dich J, Holm LE. 1987. Risk of malignant lymphoma in Swedish
pesticide appliers. Br J Cancer 56:505-508.

Viklund K, Holm LE. 1986. Soft tissue sarcoma risk in Swedish
agricultural and forestry workers. J Natl Cancer Inst 76:229-234.

Wifp HK, Schmid J. 1983. Seveso--an environmental assessment. In: Tucker
RE, ed. Human and Environmental Risk of Chlorinated Dioxins and Related
Compounds. New York: Plenum Publishing Corp, pp. 255-274.

Wolfe WH, Lathrop GD, Albanese RA. Moynahan PM. 1985. An epidemiologic
investigation of health effects in Air Force personnel following
exposure to herbicides and associated dioxins. Chemosphere 14:707-716.

Wolfe WH, Lathrop GD, Albanese RA, Moynahan PM. 1984. Chemosphere
14:707-716 (cited in Hiremath et al. 1986).

Woods JS. Polissar L, Severson RK, Heuser LS. Kulander BG. 1989. Soft
tissue sarcoma and non-Hodgkin's lymphoma in relation to
phenoxyherbicide and chlorinated phenol exposure in western Washington.
J Natl Cancer Inst 78:889-910.

Wroblewski VJ, Olson JR. 1985. Hepatic metabolism of 2,3,7,8-
tetrachlorodibenzo-p-dioxin (TCDD) in the rat and guinea pig. Toxicol
Appl Pharmacol 81:231-240.
                                                     \
Wurrey CJ, Bourne S, Kleopfer RD. 1986. Application of gas
chromatography/matrix isolation/Fourier transform infrared spectrometry
to dloxin determinations. Anal Chem 58(2):482-83.

Young AL. 1984. Determination and measurement of human exposure to the
dibenzo-p-dioxins. Bull Environ Contain Toxicol 33(6) :702-709.

Young AL, Kang HK. 1985. Status and results of federal epidemiologic
studies of populations exposed to TCDD. Chemosphere 14:779-790.

Young AL, Kang HK, Shepard BN. 1983. Chlorinated dioxins as herbicide
contaminants. Environ Sci Technol 17:530A-540A (cited in EPA 1985a).

-------
124   Section 10

Zack JA, Susklnd RR. 1980. The mortality experience of workers exposed
to tetrachlorodlbenzodloxln in a trichlorophenol process accident. J
Occup Med 22(1):11-14 (cited in EPA 1985a).

Zeiger E. 1983. Memorandum from Dr. Zeiger to Dr. E.E. McConnell on the
results of test performed for the Environmental Mutageneis Development
Program. NTP, NIEHS (cited in EPA 1985a).

Zimnering S, Mason JM, Valencia R, Woodruff RC. 1985. Chemical
mutagenesis testing in Drosophila. II. Results of 20 coded compounds
tested for the National Toxicology Program.  Environ Mutagen 7:87-100.

-------
                                                                     125
                             11.  GLOSSARY

Acute Exposure--Exposure Co a chemical for a duration of 14 days or
less, as specified in the Toxicological Profiles.

Bioconcentration Factor (BCF)--The quotient of the concentration of a
chemical in aquatic organisms at a specific time or during a discrete
time period of exposure divided by the concentration in the surrounding
water at the same time or during the same time period.

Carcinogen--A chemical capable of inducing cancer.

Ceiling value (CL)--A concentration of a substance that should not be
exceeded, even instantaneously.  .

Chronic Exposure--Exposure to a chemical for 365 days or more, as
specified in the Toxicological Profiles.

Developmental Toxicity--The occurrence of adverse effects on the
developing organism that may result from exposure to a chemical prior to
conception (either parent), during prenatal development, or postnataliy
to the time of sexual maturation. Adverse developmental effects may be
detected at any point in the life span of the organism.

Embryotoxlcity and Fetotoxicity--Any toxic effect on the conceptus as a
result of prenatal exposure to a chemical; the distinguishing feature
between the two terms is the stage of development during which the
insult occurred. The terms, as used here, include malformations and
variations, altered growth, and in utero death.

Frank Effect Level (FEL)--That level of exposure which produces a
statistically or biologically significant increase in frequency or
severity of unmistakable adverse effects, such as irreversible
functional impairment or mortality, in an exposed population when
compared with its appropriate control.

EPA Health Advisory--An estimate of acceptable drinking water levels for
a chemical substance based on health effects information. A health
advisory is not a legally enforceable federal standard, but serves as
technical guidance to assist federal, state, and local officials.

Immediately Dangerous to Life or Health (IDLH)--The maximum
environmental concentration of a contaminant from which one could escape
within 30 min without any escape-impairing symptoms or irreversible
health effects.

-------
126   Section 11

Intermediate Exposure--Exposure to a chemical for a duration of 15-364
days, as specified in the Toxicological Profiles.

Imnunologic Tozi'city--The occurrence of adverse  effects on the immune
system that may result from exposure to environmental agents such as
chemicals.

In vitro--Isolated from the living organism and  artificially maintained,
as in a test tube.

In vivo--Occurring within the living organism.

Key Study--An animal or human toxicological study that best illustrates
the nature of the adverse effects produced and the doses associated with
those effects.

Lethal Concentration(LO) (LCLO)--The lowest concentration of a chemical
in air which has been reported to have caused death in humans or
animals.

Lethal Concentration(SO) (LCso)--A calculated concentration of a
chemical in air to which exposure for a specific length of time is
expected to cause death in 50% of a defined experimental animal
population.

Lethal Dose(LO) (LDLO)--The lowest dose of a chemical introduced by a
route other than inhalation that is expected to have caused death in
humans or animals.

Lethal Dose(SQ) (LDSO)--The dose of a chemical which has been calculated
to cause death in 50% of a defined experimental animal population.

Lovest-Observed-Adverse-Effect Level (LOAEL)--The lowest dose of
chemical in a study or group of studies which produces statistically or
biologically significant increases in frequency or severity of adverse
effects between the exposed population and its appropriate control.

Lovest-Observed-Effect Level (LOEL)--The lowest dose of chemical in a
study or group of studies which produces statistically or biologically
significant increases in frequency or severity of effects between the
exposed population and its appropriate control.

Malformations--Permanent structural changes that may adversely affect
survival, development, or function.

Minimal Risk Level--An estimate of daily human exposure to a chemical
that is likely to be without an appreciable risk of deleterious effects
(noncancerous) over a specified duration of exposure.

Mutagen--A substance that causes mutations. A mutation  is a change  in
the genetic material in a body cell. Mutations can lead to birth
defects, miscarriages, or cancer.

-------
                                                          Glossary   127

Neurotoxicity--The occurrence of adverse effects on the nervous system
following exposure to a chemical.

No-Observed-Adverse-Effect Level (NOAEL)--That dose of chemical at which
there are no statistically or biologically significant increases in
frequency or severity of adverse effects seen between the exposed
population and its appropriate control. Effects may be produced at this
dose, but they are not considered to be adverse.

No-Observed-Effect Level (NOEL)--That dose of chemical at which there
are no statistically or biologically significant increases in frequency
or severity of effects seen between the exposed population and its
appropriate control.

Permissible Exposure Limit (PEL)--An allowable exposure level in
workplace air averaged over an 8-h shift.

q.*--The upper-bound estimate of the low-dose slope of the dose-response
curve as determined by the multistage procedure. The q.* can be used to
calculate an estimate of carcinogenic potency, the incremental excess
cancer risk per unit of exposure (usually pg/L for water, mg/kg/day for
food, and pg/m3 for air).

Reference Dose (RfD)--An estimate (with uncertainty spanning perhaps an
order of magnitude) of the daily exposure of the human population to a
potential hazard that is likely to be without risk of deleterious
effects during a lifetime. The RfD is operationally derived from the
NOAEL (from animal and human studies) by a consistent application of
uncertainty factors that reflect various types of data used to estimate
RfDs and an additional modifying factor, which is based on a
professional judgment of the entire database on the chemical. The RfDs
are not applicable to nonthreshold effects such as cancer.

Reportable Quantity (RQ)--The quantity of a hazardous substance that is
considered reportable under CERCLA. Reportable quantities are: (1) 1 Ib
or greater or (2) for selected substances, an amount established by
regulation either under CERCLA or under Sect. 311 of the Clean Water
Act. Quantities are measured over a 24-h period.

Reproductive Toxicity--The occurrence of adverse effects on the
reproductive system that may result from exposure to a chemical. The
toxicity may be directed to the reproductive organs and/or the related
endocrine system. The manifestation of such toxicity may be noted as
alterations in sexual behavior, fertility, pregnancy outcomes, or
modifications in other functions that are dependent on the integrity of
this systea.

Short-Tern Exposure Limit  (STEL)--The maximum concentration to which
workers can be exposed for up to 15 min continually. No more  than four
excursions are allowed per day, and there must be at least 60 min
between exposure periods. The daily TLV-TWA may not be exceeded.

-------
128   Section 11

Target Organ Toxlclty--This term covers a broad range of adverse effect'
on target organs or physiological systems (e.g., renal, cardiovascular)
extending from those arising through a single limited exposure to those
assumed over a lifetime of exposure to a chemical.

Teratogen--A chemical that causes structural defects that affect the
development of an organism.

Threshold Limit Value (TLV)--A concentration of a substance to which
most workers can be exposed without adverse effect. The TLV may be
expressed as a TWA, as a STEL, or as a CL.

Time-weighted Average (TWA)--An allowable exposure concentration
averaged over a normal 8-h workday or 40-h workweek.

Uncertainty Factor  (UF)--A factor used in operationally deriving the RfD
from experimental data. UFs are intended to account for (1) the
variation in sensitivity among the members of the human population,
(2) the uncertainty in extrapolating animal data to the case of humans.
(3) the uncertainty in extrapolating from data obtained in a study that
is of less than lifetime exposure, and (4) the uncertainty in using
LOAEL data rather than NOAEL data. Usually each of these factors is set
equal to 10.

-------
                                                                     129
                         APPENDIX:  PEER REVIEW

     A peer review panel was assembled for 2,3,7,8 tetrachlorodibenzo-
p-dioxin (TCDD). The panel consisted of the following members:
Dr. Herbert Cornish, University of Michigan; Dr. Shane Que Hee,
University of Cincinnati Medical Center; and Dr. James Olson, State
University of New York at Buffalo, School of Medicine.  These experts
collectively have knowledge of 2,3,7,8-TCDD's physical and chemical
properties, toxicokinetics, key health end points, mechanisms of action,
human and animal exposure, and quantification of risk to humans. All
reviewers were selected in conformity with the conditions for peer
review specified in the Superfund Amendments and Reauthorization Act of
1986, Section 110.

     A joint panel of scientists from ATSDR and EPA has reviewed the
peer reviewers' comments and determined which comments will be included
in the profile. A listing of the peer reviewers' comments not
incorporated in the profile, with a brief explanation of the rationale
for their exclusion, exists as part of the administrative record for
this compound. A list of databases reviewed and a list of unpublished
documents cited are also included in the administrative record.

     The citation of the peer review panel should not be understood to
imply their approval of the profile's final content. The responsibility
for the content of this profile lies with the Agency for Toxic
Substances and Disease Registry.

-------